WO2013163466A1 - Inhibitors of hepatitis c virus - Google Patents

Inhibitors of hepatitis c virus Download PDF

Info

Publication number
WO2013163466A1
WO2013163466A1 PCT/US2013/038275 US2013038275W WO2013163466A1 WO 2013163466 A1 WO2013163466 A1 WO 2013163466A1 US 2013038275 W US2013038275 W US 2013038275W WO 2013163466 A1 WO2013163466 A1 WO 2013163466A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
mmol
esi
added
residue
Prior art date
Application number
PCT/US2013/038275
Other languages
French (fr)
Inventor
Min Zhong
Leping Li
Original Assignee
Presidio Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US13/456,181 external-priority patent/US9085587B2/en
Application filed by Presidio Pharmaceuticals, Inc. filed Critical Presidio Pharmaceuticals, Inc.
Publication of WO2013163466A1 publication Critical patent/WO2013163466A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/12Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • C07D498/14Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • the invention relates to compounds useful for inhibiting hepatitis C virus (“HCV”) replication, particularly functions of the non-structural 5B (“NS5B”) protein of HCV.
  • HCV hepatitis C virus
  • HCV is a single-stranded RN A virus that is a member of the Flaviviridae family.
  • the virus shows extensive genetic heterogeneity as there are currently seven identified genotypes and more than 50 identified subtypes.
  • viral RNA is translated into a polyprotein that is cleaved into ten individual proteins.
  • structural proteins the core (C) protein and the envelope glycoproteins, El and E2, p7, an integral membrane protein, follows El and E2.
  • C core
  • E2 envelope glycoproteins
  • p7 an integral membrane protein
  • NS5B is the RNA polymerase or replicase of the virus and is responsible for replication of both positive and negative-strand genomic RNA during the viral replicative cycle. NS5B plays an essential and critical role in viral replication, and a functional NS5B replicase is required for HCV replication and infection. Thus, inhibition of NS5B RNA-dependent polymerase activity is believed to be an effective way of treating HCV infection.
  • HCV infection is a serious health issue. It is estimated that 170 million people worldwide are chronically infected with HCV. HCV infection can lead to chronic hepatitis, cirrhosis, liver failure and hepatocellular carcinoma. Chronic HCV infection is thus a major worldwide cause of liver-related premature mortality.
  • the present disclosure describes a class of heterocyclic compounds targeting HCV NS5B polymerase and methods of their use to treat HCV infection in humans.
  • L 1 , L 2 and-NCSQzR 12 together with the attached carbons of the aromatic ring to form a 5-12 member ring, containing 1-4 heteroatoms of N, O, S, P and/or Si;
  • L'or L 2 is independently selected from the group consisting of a bond, -0-, -C(R I5 R 16 )-, -NR 3 -, - S(O » -P(0 , -Si(R 4 R 5 K -C(O)-, -C(0)0-, and substituted alkyl, alkenyl, alkoxy, cycloalkyl, cycloalkoxy, heterocycle, aryl, heteroaryl, amide, carbamate, urea, and sulfonamide;
  • n 0, 1, or 2;
  • R' is selected from hydrogen, -(CH 2 CHzO)o-8-Me, C 2 .24 alkyl optionally containing 0-6 heteroatoms of O, NR N and/or S, and 3-8 cycloalkyl optionally containing 0-2 heteroatora of O, NR N , and/or S;
  • R 3 is selected from the group of hydrogen, alkylcarbonyl, cycloalkylcarbonyl, alkoxylcarbonyl, cycloalkoxycarbonyl, alkylsulfonyl and cycloalkylsulfonyl;
  • R 4 and R 5 are independently methyl, ethyl, or cyclopropyl;
  • R 12 is independently C]. 3 alkyl, cyclopropyl, -OMe, or -NHMe;
  • R 15 , R 16 is independently hydrogen, hydroxyl, azide, C 2 - alkeny!, CM alkyl, cyclopropyl, C alkoxy, or cyclopropoxy or R 15 and R 16 together are a carbonyl or C alkenylidene or R 15 and R 16 joined together with the attached carbon are 3-6 member ring optionally containing 0-3 heteroatoms of O, NR N and or S; and
  • R ,7 is F, Cl or CN.
  • the compound of this embodiment may have an inhibitory activity with respect to HCV,
  • R v is selected from hydrogen, -NR'R", alkoxyls, alkyls, cycloalkyls, aryls, heteroaryls, and aralkyl;
  • R w and R* are indenpendrntry selected from hydrogen, -C(0)H, -C(0)R Hi , -C(0)OR iv -, - C ⁇ NR'H", alkoxyls, alkyls, cycloalkyls, aryls, heteroaryls, and aralkyl; optionally, R v and R together form a C4.8 member ring;
  • R y and R z are independently selected from hydrogen, alkoxyls, alkyls, cycloalkyls, aryls, heteroaryls, and aralkyl; optionally, R y and R z together form a 3-8 member ring;
  • R' and R" are indenpendently selected from hydrogen, -Me, -Et, c-Pr-, c-Butyl;
  • R" 1 is selected from alkyoxyl, alkyls, cycloalkyls, aryls, heteroaryls, and aralkyl;
  • R lv is selected from alkyoxyl, alkyls, cycloalkyls, aryls, heteroaryls, and aralkyl; and R v and R VI are indenpendently selected from hydrogen, aUcyoxyl, alkyls, cycloalkyls, aryls, heteroaryls, and aralkyl.
  • the compound has the structural formula:
  • X' is selected from and
  • R' 3 is selected from -CH 3 and -OCH 3 .
  • the compound may be selected from the group consisting of B243, B244, B245, B246, B248, B249, B252, B253, B255, B256, B257, B258, B259, B260, B261, B262,B263, B264, B265, B266, B267, B268, B269, B270, B271, B272, B273, B274, B275, B276, B277, B278, B279, B280, B281, B282, B283, B284, B285, B286, B287, B288, B289, B290, B291, B293, B294, B295, B296, B297, B298, B299, B300, B301, B302, B303, B304, B305, B306, B307, B308, B309, B310, B311, B312, B313, B314, B315, B316, B317, B318, B319, B320, B321, B322,
  • R'i pa a-F
  • R u -0- is as above
  • R ⁇ is -C3 ⁇ 4.
  • the compound in this group may be selected from the group consisting of B243, B244, B245, B246, B248, B249, B252, B253, B255, B256, B257, B258, B259, B260, B261, B262,B263, B264, B265, B266, B267, B268, B269, B270, B271, B272, B273, B274, B275, B276, B277, B278, B279, B280, B281, B282, B285, B286, B289, B300, B302, B306, B307, B308, B309, B310, B311, B314, B316, B317, B318, B319, B320, B333, B335, B336, B337, B338, B343, B344, B345, B346, B347, B348, B349, B350, B351, B352, B353, B354, B373, B378, B379, B38
  • R'i is selected from -H, -F, -CI, -Br, -I,
  • X' is selected and R' 3 is selected from -CH 3 and -OCH3.
  • Specific compounds in this group (Group 2) are selected from the group consisting of B283, B284, B287, B288, B290, B291, B293, B422, B423, B424, B426, B432, B443, B446, B447, B455, B456, B459, B463, B464, B466, B467, B470, B478, B479, B487, B488, B489, B491, B492, B493, B494, B495, B496, B497, B498, B499, B500, B501, B502, B503, B504, B505, B506, B507, B508, B509, B510, B511, B512, B513, B514, B515, B516, B517, B518, B5
  • R'3 is selected from -C3 ⁇ 4 and -OCH3.
  • Specific compounds in this group are selected from the group consisting of B294, B295, B296, B297, B298, B299, B301, B303, B304, B305, B312, B313, B315, B321, B322, B323, B326, B327, B328, B329, B330, B334, B339, B340, B341, B342, B355, B356, B357, B358, B360, B361, B362, B363, B364, B365, B366, B367, B368, B369, B370, B371, B372, B374, B375, B376, B377, B380, B382, B387, B393, B396, B398, B403, B406, B407, B408, B410, B411, B412, B413, B414, B415, B416, B417, B418, B419, B420, B421, B425, B427, B428, B429
  • R' l is selected from para-F
  • R' 2 is selected from -OH and -S0 2 Me; and R' 3 is selected from -CH 3 and -OCH 3 , as exemplified by B324, B325, B331, and B332.
  • the compound of the invention has the structural formula:
  • R' i is selected from para-F ;
  • R' 2 is selected from -OH and -S0 2 Me; and R * 3 is selected from -CH 3 and -OCH3, as exemplified by compound B359.
  • a pharmaceutical composition comprising a compound of any one of compounds above, or its pharmaceutically acceptable salts, together with one or more pharmaceutically acceptable excipients or vehicles, and optionally other therapeutic and/or prophylactic ingredients.
  • composition may further includes additional one, two or three anti-HCV agent(s) selected from the group consisting of interferon-alpha, ribavirin, cyclosporine derivatives, HCV NS3 protease inhibitors, HCV NS4B inhibitors, HCV NS5 A inhibitors, HCV NS5B polymerase inhibitors, and p7 inhibitors.
  • anti-HCV agent(s) selected from the group consisting of interferon-alpha, ribavirin, cyclosporine derivatives, HCV NS3 protease inhibitors, HCV NS4B inhibitors, HCV NS5 A inhibitors, HCV NS5B polymerase inhibitors, and p7 inhibitors.
  • the invention includes a method of treating HCV infection in a subject comprising administering to the subject, a pharmaceutically acceptable dose of the compound above, and continuing the administering until a selected reduction in the subject's HCV titer is achieved
  • the method may use a compound selected from the group consisting of B243, B244, B245, B246, B248, B249, B252, B253, B255, B256, B257, B258, B259, B260, B261, B262,B263, B264, B265, B266, B267, B268, B269, B270, B271, B272, B273, B274, B275, B276, B277, B278, B279, B280, B281, B282, B283, B284, B285, B286, B287, B288, B289, B290, B291, B293, B294, B295, B296, B297, B298, B299, B300, B301, B302, B303, B304, B305, B306, B307, B308, B309, B310, B311, B312, B313, B314, B315, B316, B317, B 18, B319, B320, B321, B
  • the compound is selected from the group (Group 1 ) consisting of B243, B244, B245, B246, B248, B249, B252, B253, B255, B256, B257, B258, B259, B260, B261, B262.B263, B264, B265, B266, B267, B268, B269, B270, B271, B272, B273, B274, B275, B276, B277, B278, B279, B280, B281, B282, B285, B286, B289, B300, B302, B306, B307, B308, B309, B310, B311, B314, B316, B317, B318, B319, B320, B333, B335, B336, B337, B338, B343, B344, B345, B346, B347, B348, B349, B350, B351, B352, B353, B354, B373, B
  • the compound is selected from the group (Group 2) consisting of B283, B284, B287, B288, B290, B291, B293, B422, B423, B424, B426, B432, B443, B446, B447, B455, B456, B459, B463, B464, B466, B467, B470, B478, B479, B487, B488, B489, B491, B492, B493, B494, B495, B496, B497, B498, B499, B500, B501, B502, B503, B504, B505, B506, B507, B508, B509, B510, B511, B512, B513.
  • Group 2 consisting of B283, B284, B287, B288, B290, B291, B293, B422, B423, B424, B426, B432, B443, B446, B447, B455, B456, B459, B463, B464, B466, B467,
  • the compound is selected from the group (Group 3) consisting of B294, B295, B296, B297, B298, B299, B301, B303, B304, B305, B312, B313, B315, B321, B322, B323, B326, B327, B328, B329, B330, B334, B339, B340, B341, B342, B355, B356, B357, B358, B360, B361, B362, B363, B364, B365, B366, B367, B368, B369, B370, B371, B372, B374, B375, B376, B377, B380, B382, B387, B393, B396, B398, B403, B406, B407, B408, B410, B411, B412, B413, B414, B415, B416, B417, B418, B419, B420, B421, B425, B427, B
  • the invention is intended to include all isotopically labeled analogs of the compounds of the invention.
  • Isotopes include those atoms having the same atomic number but different mass.
  • isotopes of hydrogen include 2 H(D) and 3 H(T) and isotopes of carbon include ,3 C and 14 C.
  • Isotopically labeled compounds of the invention can be prepared according to methods commonly known in the art. Such compounds may have various potential uses as, but not limited to, standards and reagents in determining biological/pharmacological activities. For those stable isotopically labeled compounds of the invention, they may have the potential to favorably modulate biological, pharmacological, or pharmacokinetic properties.
  • alkanoyl as used herein contemplates a carbonyl group with a lower alkyl group as a substituent.
  • alkenyl as used herein contemplates substituted or unsubstituted, straight and branched chain alkene radicals, including both the E- and Z-forms, containing from two to eight carbon atoms.
  • the alkenyl group may be optionally substituted with one or more substituents selected from the group consisting ofhalogen, -CN, -N(1 ⁇ 4, CC3 ⁇ 4R, C(0)R, -O-R, -N(R N ) 2 , - N(R N )C(0)R, -N(R N )S(0) 2 R, -SR, -C(0)N(R N ) 2 , -OC(0)R,
  • alkoxycarbonyl contemplates a carbonyl group with an alkoxy group as a substituent.
  • alkyl contemplates substituted or unsubstituted, straight and branched chain alkyl radicals containing from one to fifteen carbon atoms.
  • lower alkyP contemplates both straight and branched chain alkyl radicals containing from one to six carbon atoms and includes methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert- butyl and the like.
  • the alkyl group may be optionally substituted with one or more substituents selected from halogen, -CN, -N0 2 , -C(0) 2 R, -C(0)R, -O-R, -N(R N ) 2 , -N(R N )C(0)R, - N(R N )S(0)2R, -SR, -C(0)N(R N ) 2 , -OC(0)R, -OC(0)N(R N ) 2 , -SOR, -S0 2 R, -SO3R, -S(0)2N(R N ) 2i phosphate, phosphonate, cycloalkyl, cycloalkenyl, aryl and heteroaryl.
  • substituents selected from halogen, -CN, -N0 2 , -C(0) 2 R, -C(0)R, -O-R, -N(R N ) 2 , -N(R N )C(0)R,
  • alkylene alkenylene and alkynylene
  • alkynylene refers to the groups “alkyl,” “alkenyl” and “alkynyl” respectively, when they are divalent, ie, attached to two atoms.
  • alkylsulfonyl as used herein contemplates a sulfonyl group which has a lower alkyl group as a substituent.
  • alkynyl as used herein contemplates substituted or unsubstituted, straight and branched carbon chain containing from two to eight carbon atoms and having at least one carbon- carbo triple bond.
  • alkynyl includes, for example ethynyl,
  • alkynyl group may be optionally substituted with one or more substituents selected from halo, -CN, -N0 2 , -C0 2 R, - C(0)R, -O-R, -N(R N ) 2 , -N(R N )C(0)R, -N(R N )S(0) 2 R, -SR, -C(0)N(R N ) 2 , -OC(0)R,
  • amino as used herein contemplates a group of the structure -NR N 2 .
  • D or the L configuration includes but is not limited to the twenty "standard” amino acids: isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan, valine, alanine, asparagine, aspartate, cysteine, glutamate, glutamtne, glycine, proline, serine, tyrosine, arginine and histidine.
  • the present invention also includes, without limitation, D-configuration amino acids, beta-amino acids, amino acids having side chains as well as all non-natural amino acids known to one skilled in the art.
  • aralkyl as used herein contemplates a lower alkyl group which has a substituent on an aromatic group, which aromatic group may be substituted or unsubstituted.
  • the aralkyl group may be optionally substituted with one or more substituents selected from halogen, -CN, -NO2, -CO2R, -C(0)R, -O-R, -N(R N )2, -N(R N )C(0)R, -N(R N )S(0) 2 R, -SR,
  • aryl as used herein contemplates substituted or unsubstituted single-ring and multiple aromatic groups (for example, phenyl, pyridyl and pyrazole, etc.) and polycyclic ring systems (naphthyl and quinolinyl, etc.).
  • the polycyclic rings may have two or more rings in which two atoms are common to two adjoining rings (the rings are "fused") wherein at least one of the rings is aromatic, e.g., the other rings can be cycloalkyls, cycloalkenyls, aryl, heterocycles and/or heteroaryls.
  • the aryl group may be optionally substituted with one or more substituents selected from halogen, alkyl, -CN, -NO2, - CC3 ⁇ 4R, -C(0)R, -O-R, -N(R N ) 2 , -N(R N )C(0)R, -N(R N )S(0) 2 R, -SR, -C(0)N(R N ) 2 , -OC(0)R, - OC(0)N(R N ) 2 , -SOR, -S0 2 R, -SO3R, -SiO ⁇ N ⁇ , -SiR 3 , -P(0)R, phosphate, phosphonate, cycloalkyl, cycloalkenyl, aryl and heteroaryl.
  • substituents selected from halogen, alkyl, -CN, -NO2, - CC3 ⁇ 4R, -C(0)R, -O-R, -N(R N
  • arylsulfonyl as used herein contemplates a sulfonyl group which has as a substituent an aryl group.
  • the term is meant to include, without limitation, monovalent as well as multiply valent aryls (eg, divalent aryls).
  • Carboxyl as used herein contemplates a group of the structure [055]
  • cycloalkyt as used herein contemplates substituted or unsubstituted cyclic alkyl radicals containing from three to twelve carbon atoms and includes cyclopropyl, cyclopentyl, cyclohexyl and the like.
  • cycloalkyl also includes polycyclic systems having two rings in which two or more atoms are common to two adjoining rings (the rings are "fused”).
  • the cycloalkyl group may be optionally substituted with one or more substituents selected from halo, -CN, -NO 2 , -CO2R, -C(0)R, -O-R, -N( N > 2 , -N(R N )C(0)R, -N(R N )S(0) 2 R, -SR, -C(0)N(R N >2, - OC(0)R, -OC(0)N(R N ) 2 , -SOR, -S0 2 R, -S(0) 2 N(R N ) 2 , phosphate, phosphonate, alkyl,
  • cycloalkenyl as used herein contemplates substituted or unsubstituted cyclic alkenyl radicals containing from four to twelve carbon atoms in which there is at least one double bond between two of the ring carbons and includes cyclopentenyl, cyclohexenyl and the like.
  • cycloalkenyl also includes polycyclic systems having two rings in which two or more atoms are common to two adjoining rings (the rings are "fused").
  • the cycloalkenyl group may be optionally substituted with one or more substituents selected from halo, -CN, -NO 2 , -C0 2 R, - C(0)R, -O-R, -N(R N ) 2 , -N(R N )C(0)R, -N(R N )S(0) 2 R, -SR, -C(0)N(R N ) 2 , -OC(0)R, - OC(0)N(R N ) 2 , -SOR, -S0 2 R, -S(0) 2 N(R N ) 2 , phosphate, phosphonate, alkyl, cycloalkenyl, aryl and heteroaryl.
  • substituents selected from halo, -CN, -NO 2 , -C0 2 R, - C(0)R, -O-R, -N(R N ) 2 , -N(R N )C(0)R, -N(R N
  • halo or halogen as used herein includes fluorine, chlorine, bromine and iodine.
  • heteroalkyl as used herein contemplates an alkyl with one or more
  • heteroatom particularly within a ring system, refers to N, O and S.
  • heterocyclic group contemplates substituted or unsubstituted aromatic and non-aromatic cyclic radicals having at least one heteroatom as a ring member.
  • Preferred heterocyclic groups are those containing five or six ring atoms which includes at least one hetero atom and includes cyclic amines such as morpholtno, piperidino, pyrrolidino and the like and cyclic ethers, such as tetrahydrofuran, tetrahydropyran and the like.
  • Aromatic heterocyclic groups also termed "heteroaryl” groups, contemplates single-ring hetero-aromatic groups that may include from one to three heteroatoms, for example, pyrrole, furan, thiophene, imidazole, oxazole, thiazole, triazole, pyrazole, oxodiazole, thiadiazole, pyridine, pyrazine, pyridazine, pyrimidine and the like.
  • heteroaryl also includes polycyclic hetero-aromatic systems having two or more rings in which two or more atoms are common to two adjoining rings (the rings are "fused") wherein at least one of the rings is a heteroaryl, e.g., the other rings can be cycloalkyls, cycloalkenyls, aryl, heterocycles and/or heteroaryls.
  • polycyclic heteroaromatic systems examples include quinoline, isoquinoline, cinnoline, tetrahydroisoquinoline, quinoxaline, quinazoline, benzimidazole, benzofuran, benzothiophene, benzoxazole, benzothiazole, indazole, purine, benzotriazole, pyrrolepyridine, pyrrazolopyridine and the like.
  • the heterocyclic group may be optionally substituted with one or more substituents selected from the group consisting of halo, alkyl, -CN, -NO 2 , -CO2R, -C(0)R, - O-R, -N(R N ) 2 , -N(R N )C(0)R, -N(R N )S(0) 2 R, -SR, -C(0)N(R N >2, -OC(0)R, -OC(0)N(R N ) 2 , -SOR, -SO2R, -SO3R, -S(0) 2 N(R N ) 2 , -S1R3, -P(0)R, phosphate, phosphonate, cycloalkyl, cycloalkenyl, aryl and heteroaryl.
  • substituents selected from the group consisting of halo, alkyl, -CN, -NO 2 , -CO2R, -C(0)R, - O-R,
  • pharmaceutically acceptable or ''pharmacologically acceptable is meant a material which is not biologically or otherwise undesirable, i.e., the material may be administered to an individual without causing any undesirable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained.
  • “Pharmaceutically acceptable salt” refers to a salt of a compound of the invention which is made with counterions understood in the art to be generally acceptable for pharmaceutical uses and which possesses the desired pharmacological activity of the parent compound.
  • Such salts include: (1) acid addition salts, formed with inorganic acids such as hydrochloric acid,
  • hydrobromic acid sulfuric acid, nitric acid, phosphoric acid and the like; or formed with organic acids such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, rumaric acid, tartaric acid, citric acid, benzoic acid, 3 -(4-hydroxybenzoyl) benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1 ,2-ethane-disulfonic acid,
  • 2-naphthalenesulfonic acid 4-toluenesulfonic acid, camphorsulfonic acid,
  • salts of amino acids such as arginates and the like, and salts of organic acids like glucurmic or galactunoric acids and the like ⁇ see, e.g., Berge etal., 1977, J. Pharm. Sci. 66:1-19).
  • salts and “hydrates” refers to the hydrated forms of the compound that would favorably affect the physical or pharmacokinetic properties of the compound, such as solubility, palatability, absorption, distribution, metabolism and excretion.
  • Other factors, more practical in nature, which those skilled in the art may take into account in the selection include the cost of the raw materials, ease of crystallization, yield, stability, solubility, hygroscopicity, flowability and manufacturability of the resulting bulk drug.
  • sulfonamide as used herein contemplates a group having the structure
  • R" is selected from the group consisting of hydrogen, Ci-Cio alkyl, Cj-
  • Substituted sulfonyl as used herein contemplates a group having the structure
  • alkylsulfonyl including, but not limited to alkylsulfonyl and arylsulfonyl.
  • thiocarbonyl means a carbonyl wherein an oxygen atom has been replaced with a sulfur.
  • Each R is independently selected from hydrogen, -OH, -CN, -N0 2 , halogen, d to C )2 alkyl, C
  • Each R N is independendy selected from the group consisting of hydrogen, -OH, Ci to Cu alkyl, Cj toC heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycle, aryl, heteroaryl, aralkyl, alkoxy, alkoxycarbonyl, alkanoyl, carbamoyl, substituted sulfonyl, sulfonate and sulfonamide.
  • Two R N may be taken together with C, O, N or S to which they are attached to form a five to seven membered ring which may optionally contain a further heteroatom.
  • the compounds of the present invention may be used to inhibit or reduce the activity of HCV, particularly HCV's NS5B protein.
  • inhibition and reduction of activity of the NS5B protein refers to a lower level of the measured activity relative to a control experiment in which the cells or the subjects are not treated with the test compound.
  • the inhibition or reduction in the measured activity is at least a 10% reduction or inhibition.
  • reduction or inhibition of the measured activity of at least 20%, 50%, 75%, 90% or 100%, or any number in between, may be preferred for particular applications.
  • L'or L 2 is independently selected from the group consisting of a bond, -0-, -C(R 15 R I6 > -NR 3 -, - S(0) admir-, -P(O)-, -SifR ⁇ 5 )-, -C(O)-, -C(0)0-, and substituted alkyl, alkenyl, alkoxy, cycloalkyl, cycloalkoxy, heterocycle, aryl, heteroaryl, amide, carbamate, urea, and sulfonamide;
  • R* is selected from hydrogen, -(CH2CH 2 0)o ⁇ -Me, C 2 -24 alkyl optionally containing 0-6 heteroatoms of O, NR N and/or S, and 3-8 cycloalkyl optionally containing 0-2 heteroatom of O, NR N , and/or S;
  • R 2 is an aryl or heteroaryl having one or more R i7 substituents
  • R 3 is selected from the group of hydrogen, alkylcarbonyl, cycloalkylcarbonyl, alkoxylcarbonyl, cycloalkoxycarbonyl, alkylsulfonyl and cycloalkylsulfonyl;
  • R 4 and R 5 are independently methyl, ethyl, or cyclopropyl
  • R 12 is independently C1.3 alkyl, cyclopropyl, -OMe, or -NHMe;
  • R 15 , R 16 is independently hydrogen, hydroxyl, azide, C2-4 alkenyl, C2- 4 alkynyl, C alkyl, cyclopropyl, CM alkoxy, or cyclopropoxy or R 13 and R 16 together are a carbonyl or C
  • alkenylidene or R 15 and R 16 joined together with the attached carbon are 3-6 member ring optionally containing 0-3 heteroatoms of O, NR N and/or S;
  • R 17 is F, Cl orCN.
  • the compound of this aspect may have an inhibitory activity with respect to HCV, as measured by the concentration of the compound effective to produce a half-maximal inhibition of HCVlb replication (EC 50 ) in a lb_Huh-Luc Neo-ET cell line in culture, of lOO nM or less.
  • R u is selected from hydrogen, -C(0)H, -C(0)R in , -C(0)OR iv , -CiONR'R , alkyls, cycloalkyls, aryls, heteroaryls, and aralkyl;
  • R v is selected from hydrogen, -NR'R", alkoxyls, alkyls, cycloalkyls, aryls, heteroaryls, and aralkyl;
  • R w and R* are independrntly selected from hydrogen, -C(0)H, -C(0)R Ui , -C(0)OR iv -, - C(0)NR v R vi , alkoxyls, alkyls, cycloalkyls, aryls, heteroaryls, and aralkyl; optionally, R v and R w together form a C4.8 member ring;
  • R y and R z are independently selected from hydrogen, alkoxyls, alkyls, cycloalkyls, aryls, heteroaryls, and aralkyl; optionally, R y and R x together form a 3-8 member ring;
  • R' and R u are indenpendently selected from hydrogen, -Me, -Et, oPr-, c-Butyl;
  • R'" is selected from alkyoxyl, alkyls, cycloalkyls, aryls, heteroaryls, and aralkyl;
  • R 1V is selected from alkyoxyl, alkyls, cycloalkyls, aryls, heteroaryls, and aralkyl;
  • R v and R VI are indenpendently selected from hydrogen, alkyoxyl, alkyls, cycloalkyls, aryls, heteroaryls, and aralkyl.
  • the compound of this aspect may have the structure in which R 12 0 is one of
  • R 1 in mis aspect may be
  • R 2 in this aspect may be a phenyl substituted with one or more R 17 substituents.
  • R 2 in this aspect may be a 4-phenoxyphenyl and the phenoxy group is substituted with one or mor 17 substituents.
  • the compound may have the particular sterochemical structure shown below:
  • the compound in this group may be selected from the group consisting of B243, B244, B245, B246, B248, B249, B252, B253, B255, B256, B257, B258, B259, B260, B261, B262.B263, B264, B265, B266, B267, B268, B269, B270, B271, B272, B273, B274, B275, B276, B277, B278, B279, B280, B281, B282, B285, B286, B289, B300, B302, B306, B307, B308, B309, B310, B311, B314, B316, B317, B318, B319, B320, B333, B335, B336, B337, B338, B343, B344, B345, B346, B347, B348, B349, B350, B351, B352, B353, B354, B373, B378, B379, B38
  • R' I is selected from -H, -F, -CI, -Br, -I,
  • n 0, 1 , 2
  • X' is selected and « N-0(CH 2 )j-23CH 3 , and
  • R' 3 is selected from -C3 ⁇ 4 and -OCH3.
  • Specific compounds in this group are selected from the group consisting of B283, B284, B287, B288, B290, B291, B293, B422, B423, B424, B426, B432, B443, B446, B447, B455, B456, B459, B463, B464, B466, B467, B470, B478, B479, B487, B488, B489, B491, B492, B493, B494, B495, B496, B497, B498, B499, B500, B501, B502, B503, B504, B505, B506, B507, B508, B509, B510, B511, B512, B513, B514, B515, B516, B517, B518, B519, B520, B521, B522, B523, B524, B525, B526, B527, B528, B529, B530, B531, B532, B533, B534, B535, B
  • Specific compounds in this group are selected from the group consisting of B294, B295, B296, B297, B298, B299, B301, B303, B304, B305, B312, B313, B315, B321, B322, B323, B326, B327, B328, B329, B330, B334, B339, B340, B341, B342, B355, B356, B357, B358, B360, B361, B362, B363, B364, B365, B366, B367, B368, B369, B370, B371, B372, B374, B375, B376, B377, B380, B382, B387, B393, B396, B398, B403, B406, B407, B408, B410, B411, B412, B413, B414, B415, B416, B417, B418, B419, B420, B421, B425, B427, B428, B429
  • the compound of the invention has the structural formula:
  • R'2 is selected from -OH and -SOaMe; and R'j is selected from -CH3 and -OCH3, as exemplified by compound B359.
  • This compound can be prepared in accordance with Scheme 39 detailed below.
  • the compounds of the invention may be prepared by a variety of synthetic routes, samples of which are illustrated in the synthetic schemes outlined below.
  • the synthesis starts with constructing the central scaffolds such as benzofuran, benzothiophene, imidazopyridine or pyrazolopyridine by employing various synthetic techniques known to those skilled in the art. (e.g. in Heterocyclic Chemistry, J. A. Joule and K. Mills, J Wiley and Sons, 2010.) .
  • further functional group manipulations including but not limited to chain elongation, amidation, esterification, cyclization are performed as necessary to lead to the target molecules.
  • the central cores may preferably be introduced toward the end of the synthesis.
  • protection-deprotection and, in some cases, orthogonal protection-deprotection strategies are required to accomplish the desired transformation. More comprehensive descriptions of these synthetic methodologies and techniques can be in found in these and other references:
  • PPTs Pyridinium / oluenesulfonate
  • Reagents and solvents used below can be obtained from commercial sources such as Aldrich Chemical Co. (Milwaukee, Wisconsin, USA).
  • ⁇ NMR spectra were recorded on a Broker 400 MHz or 500 MHz NMR spectrometer. Significant peaks are tabulated in the order: multiplicity (s, singlet; d, doublet; t, triplet; q, quartet; m, multiplet; br s, broad singlet), coupling constants) in Hertz (Hz) and number of protons.
  • Electrospray spray ionization (ESI) mass spectrometry analysis was conducted on a Hewlett-Packard 1100 MSD electrospray mass spectrometer using the HP 1 100 HPLC for sample delivery.
  • Mass spectrometry results are reported as the ratio of mass over charge, followed by the relative abundance of each ion (in parentheses) or a single m z value for the M + H (or, as noted, M - H) ion containing the most common atomic isotopes. Isotope patterns correspond to the expected formula in all cases. Normally the analyte was dissolved in methanol at 0.1 mg mL and 5 microliter was infused with the delivery solvent into the mass spectrometer, which scanned from 100 to 1500 daltons. All compounds could be analyzed in the positive ESI mode, using an acetonitrile/water gradient (10% ⁇ 90%) acetonitrile in water with 0.1% formic acid as delivery solvent. The compounds provided below could also be analyzed in the negative ESI mode, using 2 mM N3 ⁇ 4OAc in
  • the compound of the invention includes a compound having the structural formula:
  • R' l is selected from para-
  • R * 2 is selected from -OH and -S0 2 Me; and R'3 is selected from -CH ⁇ and -OCH3, as exemplified by B324, B325, B331, and B332.
  • This compound can be synthesized in accordance with the scheme 40 below. 0102]
  • the compound of the invention has the structural formula:
  • R' I is selected from para- ⁇ ;
  • R' 2 is selected from -OH and -S0 2 Me; and R'3 is selected from -CH 3 and -OCH3, as exemplified by compound B359.
  • Step 1 Refer to Scheme 4. To a solution of compound 3-1 (prepared by following the procedures described in WO200759421 with some modifications) (4.00 g, 10.3 mmol) in CH 2 C1 2 (30 mL) was added BC1 3 (I N in CH 2 C1 2 , 20.6 mmol) at 0 °C. After stirring at rt for 1 hr, the reaction mixture was added ice water (100 mL). The mixture was extracted with CH 2 C1 2 (800 mL x 2) and the combined organic extracts were washed with water and brine and dried with anhydrous Na 2 SO_ ⁇ . The solvent was removed and the residue was dried in vacuo to give crude compound 4-1 (3.4 g, 96% yield) as a yellow solid. LC-MS (ESI): mlz 346 [M + H] + .
  • Step 2 To a solution of compound 4-1 (3.4 g, 9.8 mmol) in CH 2 C1 2 (100 mL) were added DMAP (120 mg, 0.980 mmol) and D1EA (1.52 g, 11.8 mmol), followed Tf z O (3.20 g, 1 1.3 mmol) at 0 °C. After stirring at 0 °C for 2 hrs, the reaction mixture was added ice water (100 mL). The organic layer was separated, washed with water and brine, and dried with anhydrous Na 2 S0 4 . The solvent was removed and the residue was dried in vacuo to give crude compound 4-2 (4.6 g, quantitative yield) as a yellow solid. LC-MS (ESI): mlz 478 [M + H] + .
  • Step 5 To a solution of compound 4-5 (1.80 g, 4.85 mmol) in CH 2 C1 2 (50 mL) was added DMAP (6 mg) and anhydrous pyridine (3.07 g, 38.8 mmol), followed by MsCl (1.60 g, 14.5 mmol) at 0 °C. After stirring at rt for 2 hrs, the reaction mixture was added ice water (50 mL).
  • Step 6 To a suspension of NaH (0.21 g, 60% in mineral oil, 5.31 mmol) in anhydrous THF (160 mL) was added a solution of compound 4-6 (1.40 g, 2.65 mmol) in anhydrous THF (40 mL) at 0 °C. After stirring at rt for 2 hrs, the reaction mixture was added sat. aq. NH4CI (10 mL). The resulting mixture was concentrated and the residue was diluted with EtOAc (100 mL). The mixture was washed with water and brine and dried with anhydrous NaiSC The solvent was removed and the residue was purified by silica gel column chromatography (Petroleum).
  • Step 7 To a solution of compound 4-7 (50 mg, 0.12 mmol) in MeOH THF (2 mL/4 mL) was added LiOH (2.0 N, 0.46 mmol). The resulting mixture was stirred at 70 °C for 2 hrs, cooled to rt and acidified with 1 N aq. HC1 (5 mL). Subsequently, the suspension was filtered and the solid was washed with waster and dried in vacuo to give crude compound 4-8 (46 mg, 95% yield) as a white solid, which was used directly for the next step without further purification.
  • Step 1 Refer to Scheme 8. To a stirred solution of compound 4-2 (9.00 g, 18.9 mmol) in DMF (100 mL) were added Et 3 N (7.84 mL, 56.6 mmol), Pd(OAc) 2 (212 mg, 0.94 mmol), dppp (469 mg, 1.13 mmol) and butyl vinyl ether (12.1 mL, 94.4 mmol) under an atmosphere of Ar. After stirring at 100 °C for 2 hrs, the reaction mixture was concentrated. The residue was diluted with EtOAc (250 mL) and the resulting mixture was washed with water (100 mL x 3) and dried with anhydrous Na2SC>4.
  • Step 2 A solution of compound 8-1 (3.90 g, 9.13 mmol) in THF (60 mL) was added 1 N aq. HC1 (10 mL) at rt. After stirring at rt for 15 min, the reaction mixture was concentrated and the residue was diluted with DCM (100 mL). The resulting mixture was washed with brine and dried with anhydrous Na 2 S04. The solvent was removed and the residue was dried in vacuo to give crude compound 8-2 (3.27 g, 96% yield) as a yellow solid, which was used for the next step without further purification.
  • Step 3 To a stirred solution of compound 8-2 (2.00 g, 5.38 mmol) in EtOAc (50 mL) was added SnCl 2 2H 2 0 (3.47 g, 16.2 mmol). After stirring at 80 °C for 1 hr, the reaction mixture was added sat. aq. NaHC0 3 (50 mL) and the resulting mixture was stirred at rt for 30 min.
  • Step 4 To a stirred solution of compound 8-3 (900 mg, 2.64 mmol) in anhydrous pyridine (15 mL) was added MsCl (0.25 mL, 3.17 mmol) at 0 °C. After stirring at rt for 1 hr, the reaction mixture was diluted with EtOAc (100 mL) and the resulting mixture was washed with 2 N aq. HC1 (20 mL x 2) and H 2 0 (50 mL x 3) and dried with anhydrous Na 2 S0 4 . The solvent was removed and the residue was purified by silica gel column chromatography (Petroleum
  • Step 5 To a solution of compound 8-4 (380 mg, 0.91 mmol) in MeOH (10 mL) and THF (10 mL) was added NaBH 4 ( 172 mg, 4.54 mmol) in several portions at 0 °C. After stirring at 0 °C for 15 min, the reaction was quenched by adding acetone (1 mL). The mixture was concentrated and the residue was diluted with EtOAc (100 mL). The resulting mixture was washed with 2 N aq. HC1 (20 mL) and H 2 0 (50 mL x 3) and dried with anhydrous Na 2 S0 4 . The solvent was removed
  • Step 7 To a solution of compound 8-7 (40 mg, 0.09 mmol) in MeOH THF (2 mL/4 mL) was added 2.0 N aq. LiOH (0.18 mmol, 0.36 mmol). After stirring at 75 °C for 3 hrs, the reaction mixture was cooled to 0 °C and acidified with 2N aq. HC1 adjust pH value to 5 ⁇ 6. Subsequently, the suspension was filtered and the solid was washed with water and dried in vacuo to give compound 8-8 (38 mg, 97% yield) as a white solid, which was used for the next step without further purification.
  • Step 8 To a solution of compound 8-8 (40 mg, 0.10 mmol) in DMF (3 mL) was added HATU (43 mg, 0.12 mmol). The resulting mixture was stirred at rt for 60 min and DIE A (0.16 mL, 0.95 mmol) and MeNH 2 HCl (20 mg, 0.29 mmol) were added. After stirring at rt for 15 min, the reaction mixture was added into water (30 mL). The suspension was filtered and the solid was washed with water and dried in vacuo. The residue was dissolved in DCM (1.5 mL) and the solution was added into hexane (40 mL).
  • MeOH/liquid CO2 10/90 (v V), flow rate: 60 g/min and back pressure: 100 bar).
  • Step 1 Refer to Scheme 15. To a solution of NaH (80 g, 60% mineral oil dispersion, 2 mol) in toluene (1.2 L) was added diethyl carbonate (295 g, 2.50 mol) at 0 °C. After stirring at rt for 2 hrs, the mixture was added drop wise to a solution of compound 15-1 (99 g, 0.50 mol) in toluene (400 mL) at reflux. After refluxing overnight, the reaction mixture was cooled to rt and sequentially treated with HOAc (140 mL) and aq. HC1 (2 M, 864 mL).
  • Step 3 To a solution of compound 15-3 (26 g, 72 mmol) in NMP (200 mL) was added Cs 2 C0 3 (47.0 g, 144 mmol). After stirring at rt for 20 min, 2-bromopropane (20.0 ml, 216 mmol) was added. The resulting mixture was stirred at 80 °C for 4 hrs, then diluted with ammonia and agitated for 30 min. The mixture was diluted with water (200 mL) and the aqueous phase was extracted with EtOAc (150 mL x 3). The combined organic extracts were washed with water (200 mL x 3) and dried with anhydrous Na 2 S0 4 . The solvent was removed and the residue was dried in vacuo to give compound 15-4 (27.5 g, 95% yield) as a colorless oil.
  • Step 5 A mixture of compound 15-5 (5.0 g, 1 1.2 mmol), 4-fluorophenol (1.7 g, 14.5 mmol), Pd(OAc) 2 (250 mg, 1.12 mmol), -BuXphose (380 mg, 0.9 mmol) and K 3 PO 4 (4.8 g, 22.4 mmol) in toluene (50 mL) was stirred at 100°C under an atmosphere of Ar and monitored by LC- MS. After 2 hrs, the reaction mixture was concentrated and the residue was diluted with water (100 mL).
  • Step 9 To a solution of compound 15-9 (1.4 g, 3.4 mmol) in EtOAc (50 mL) was added SnCl 2 2H 2 0 (2.8 g, 13.6 mmol) at rt and the resulting mixture was stirred at 80 °C for 1 hr. The mixture was cooled to rt and d its pH value was adjusted to 8 ⁇ 9 by adding saturated aq.
  • Step 10 To a solution of compound 15-10 (1.1 g, 2.5 mmol) in anhydrous pyridine (20 mL) was added MsCl ( 1.8 mL) at 0 °C. After the mixture was stirred at 30 °C for 2 hrs, LC-MS analysis indicated that the reaction went to completion. The mixture was diluted with water (100 mL) and EtOAc (50 mL). The aqueous phase was extracted with EtOAc (50 mL x 3). The combined organic extracts were washed with sat. aq. NH 4 C1 (50 mL x 3) and brine (50 mL) and dried with anhydrous Na 2 S0 4 . The solvent was removed and the residue was dried in vacuo to give crude compound 15-11 (1.1 g, 90% yield) as a yellow solid. LC-MS (ESI): mlz 512.1 [M + H] + .
  • Step 11 To a solution of compound 15-11 (1.1 g, 2.1 mmol) in THF (30 mL) was added aBH 4 (560 mg, 14.7 mmol) in portions at 0 °C. After stirring at 0 °C for 30 min, LC-MS analysis indicated that the reaction went to completion and acetone (2 mL) was added to quench excess amount of NaB3 ⁇ 4. The mixture was concentrated and the residue was diluted with EtOAc (100 mL). The resulting mixture was washed with water (50 mL x 3) and brine (50 mL) and dried with anhydrous Na 2 SO.t. The solvent was removed and the residue was dried in vacuo to give compound 15-12 (0.9 g, 90% yield) as a yellow solid. LC-MS (ESI): mlz 514.1 [M + H] + .
  • Step 12 To a solution of compound 15-12 (0.9g, 1.7 mmol) in anhydrous DCM (30 mL) was added NaH (60% in paraoil, 200 mg, 5 mmol) at 0 °C, followed by compound 8-6 ( 1.1 g, 2.55 mmol). After stirring at 0 °C for 3 hrs and at it for 12 hrs, the reaction was quenched by adding saturated aq. NH 4 C1 (10 mL). The resulting mixture was extracted with DCM (30 mL x 3) and the combined organic extracts were washed with brine (50 mL) and dried with anhydrous Na 2 S0 4 . The solvent was removed and the residue was purified by silica gel column
  • Step 13 To a solution of compound 15-13 (520 mg, 0.96 mmol) in MeOH/THF (4 mL/8 mL) was added aq. LiOH (2.0 M, 2 mL) at rt. After stirring at 80 °C for 12 hrs, the reaction mixture was cooled to rt and adjusted its pH value to 2 ⁇ 3 by adding aq. HC1 (2.0 M). The organic solvent was removed and the residue was diluted with EtOAc (50 mL). The organic layer was isolated, washed with brine (25 mL) and dried with anhydrous Na2SC>4. The solvent was removed and the residue was dried in vacuo to give crude compound 15-14 (442 mg, 90% yield) as a white solid.
  • Step 14 Compound 15-14 (442 mg, 0.86 mmol) was dissolved in DMF (5 mL), followed by addition of HATU (450 mg, 1.17 mmol). After stirring at rt for I hr, the reaction mixture was added DIPEA (503 mg, 3.9 mmol) and MeNH 2 HCl (157 mg, 2.34 mmol). The resulting mixture was stirred at rt for another 1 hr before being concentrated The residue was diluted with water (25 mL) and EtOAc (50 mL). The organic layer was isolated, washed with brine (25 mL), and dried with anhydrous Na 2 S0 .
  • compound 28-5b can be obtained using compound 28-7b as the starting material described in Scheme 28.
  • MeOH/liquid CO2 30/70 (v/v); CO 2 flow rate: 2.1 g min and co-solvent flow rate: 0.9 g min; back pressure: 150 bar).
  • compound 28-13b can be obtained using compound 28-1 lb as th
  • Step 1 Refer to Scheme 29. To a solution of compound 4-2 (9.0 g, 18.9 mmol) in DME (200 mL) and H 2 0 (400 mL) were added 2 C0 3 (7.8 g, 56.6 mmol), Pd(dppf)Cl 2 (1.5 g, 1.9 mmol) and 4,4,5,5-tetramethyl-2-vinyl- 1 ,3,2-dioxaborolane (4.4 g, 28.3 mmol). After stirring at 60 °C for 2 hrs under an atmosphere of Ar, the reaction mixture was concentrated and the residue was partitioned between water (150 mL) and EtOAc (150 mL).
  • Step 2 To a solution of compound 29-1 (1.2 g, 3.4 mmol) in THF (50 mL), EtOH (20 mL) and HOAc (40 mL) was slowly added Zn ( 1.3 g, 20.1 mmol) at 0 °C. After stirring at rt for 2 hrs, the reaction mixture was filtered and the filtrate was concentrated. The residue was partitioned between water (80 mL) and EtOAc (80 mL) and the organic layer was extracted with EtOAc (60 mL x 3). The organic extracts were combined and washed with water (80 mL x 2), sat.
  • Step 4 To a solution of compound 29-3 (700 mg, 1.7 mmol) in DMF (30 mL) were added K2C(1 ⁇ 4 (719 mg, 5.2 mmol) and I (144 mg, 0.87 mmol), followed by 2-bromobenzyl chloride (534 mg, 2.6 mmol). After stirring at 70 °C for 3 hrs, the reaction mixture was concentrated and the residue was partitioned between water (50 mL) and EtOAc (50 mL). The aqueous layer was extracted with EtOAc (3 x 40 mL) and the combined organic extracts were washed with water (80 mL x 3) and brine (50 mL) and dried over anhydrous NaaSO-j.
  • Step 6 To a solution of compound 29-5 (150 mg, 0.31 mmol) in EtOH (30 mL) was added 5% Pd/C (w w, 200 mg). After stirring at 50 °C for several hours under an atmosphere of 3 ⁇ 4, the reaction mixture was filtered through a pad of Celite ® 545. The filtered cake was washed with EtOH (15 mL x 2). The filtrate was concentrated and the residue was dried in vacuo to give crude compound 29-6 (149 mg, 99% yield) as a yellow solid.
  • Step 7 Following the same procedure as that for for the preparation of compound 1-16 described in Scheme 1 and replacing compound 1-14 with 29-6, compound 29-7 was obtained (130 mg, 90% yield) as a pale brown solid.
  • Step 3 To a stirred solution of compound 34a-l (94.0 mg, 0.20 mmol) in DCE (20 mL) at 85°C was added TsOH (94 mg, 0.49 mmol). After refluxing overnight, the reaction mixture was concentrated. The residue was diluted with DCM (50 mL) and the resulting mixture was washed with sat. aq. NaHCCb (50 mL) and brine (50 mL), and dried over anhydrous Na2S(> .
  • Step 1 Refer to Scheme 34b. To a stirred solution of compound 34b-l (3.0 g, 17.2 mmol) in dry THF (20 mL) was added L1AIH 4 (1.3 g, 34.4mmol) in portions at 0 °C. After stirring at rt overnight, the reaction was quenched by slowly adding isopropanol (10 mL) at 0 °C. The suspension was filtered through Celite ® 545 and the filtrate was concentrated. The residue was dried in vacuo to give crude compound 34b-2 (2.0 g) as a white solid, which was used directly for the next step without further purification.
  • L1AIH 4 1.3 g, 34.4mmol
  • (5S.7S) 36j-3a and (5S,7 ?)-36j-3b were obtained as white solid.
  • (5S,7S)-36j-3a LC-MS (ESI): miz 608.1 [M + H] + ; 1 H NMR (500 MHz, ⁇ /-DMSO): 6 8.51 (q, J - 4.0 Hz, I H), 8.43 (d, j - 5.0
  • 39-12b-2 4-F-PhO-.
  • R' 39.13b-2 R s4-F-PhO-.
  • Step 1 Refer to Scheme 39a. To a stirred solution of compound 39a- 1 (5.1 g, 17 mmol) (prepared by following the procedure descried in WO200759421 with some modifications) in CH 3 CN (300 mL) was added Selectfluor ® (7.25 g, 20.4 mmol) portion-wise at rt. After stirring at rt overnight, the reaction mixture was concentrated. The residue was added water (50 mL) and the resulting mixture was extracted with EtOAc (100 mL x 3). The combined organic extracts were washed with brine, dried over anhydrous Na2SC>4 and concentrated.
  • Step 2 To a stirred solution of H 0 3 (5 mL) in DCM (15 mL) at 0°C was added a solution of compound 39a-2 (954 mg, 3 mmol) in DCM (15 mL) dropwise (Caution: the reaction mixture must be kept around 0 °C). After stirring at 0 °C for 1 hr, the reaction mixture was slowly warmed to rt and diluted with ice-water (15 mL). The organic phase was washed with 3 ⁇ 40 (25 mL x 3), dried over anhydrous a 2 S0 4 , and concentrated. The residue was dried in vacuo to give crude compound 39a-3 (850 mg) as a dark yellow solid, which was used for the next step without further purification.
  • Step 3 To a stirred solution of crude compound 39a-3 ( 1.24 g, about 3.4 mmol) in DCM (50 mL) was added DIPEA (1.7mL, 10.2 mmol) and DMAP (5 mg, 0.34 mmol), followed by Tf 2 0 (0.86 mL, 5.1 mmol) at 0 °C. After stirring at rt for 5 hrs, the reaction mixture was diluted with water (50 mL) and DCM (50 mL). The organic phase was washed with brine (50 mL), dried over anhydrous Na2S0 4 . and concentrated.
  • Step 4 To a solution of the compound 39a-4 (1.5 g, 3.0 mmol) in DME (50 mL) and 2 M Na 2 C0 3 solution (4 mL) was added Pd(PPh 3 ) 2 Cl2 (210 mg, 0.3 mmol), the resulting mixture was saturated with N2. Next, vinylboronic acid pinacol ester (0.6 mL, 4.5 mmol) was added and the resulting mixture was stirred at 80 °C for 4 hrs under an atmosphere of N 2 . Subsequently, the mixture was concentrated and the residue was added water (50 mL) and EtOAc (50 mL). The aqueous phase was extracted with EtOAc (25 mL x 3).
  • Step 5 To a stirred solution of the compound 39a-5 (1.0 g, 2.7 mmol) in EtOAc (30 mL) was added SnC 2 -2H20 ( 1.22 g, 5.4 mmol) portion-wise. After refluxing for 1 hr under an atmosphere of N 2 , the reaction mixture was cooled to rt and slowly added saturated aq. NaHC0 3 solution (20 mL). The mixture was filtered through Celite ® 545 and the filtered cake was washed with EtOAc (25 mL x 3). The organic phase of the filtrate was dried over anhydrous Na 2 S0 4 and concentrated.
  • Step 7 Following the same procedure as that used for preparing compound (55,75)-34-2a shown in Scheme 34a and replacing compound 34a-2 with 39a-7, compound 39a-8 (35 mg, 25% yield) was obtained as a white solid.
  • Step 8 Following the same procedure as that used for preparing compound 1-15 shown in Scheme 1 (WO2012/058125) and replacing compound 1-14 with 39a-8, compound 39a-9 was obtained as a light solid.
  • Step 9 Following the same procedure as that used for preparing compound 1-16 shown in Scheme 1 (WO2012/058125) and replacing compound 1-15 with 39a-9, compound 39-12a-l was obtained as a white solid.
  • Step 1 Refer to Scheme 40. To a stirred solution of 6-amino-pyridin-2-ol (5g, 45.5 mmol) in acetic acid (50 mL) was added a solution of N-iodosuccinimide (20.45 g, 90.9 mmol) in acetic acid (90 mL). After stirring at rt for lhr, the resulting precipitate was collected by filtration, washed with acetic acid, and dried in vacuo to give compound 40-2 (12.5g, 84% yield) as yellow solid.
  • Step 2 A solution of compound 40-2 (10 g, 27.6 mmol) in Ac 2 0 (50 mL) was stirred at 90 °C for 1 hr and then concentrated. The residue was carefully added saturated aq. NaHCOj solution (100 mL) at 0 °C and the resulting mixture was extracted with DCM (150 mL x 3). The organic extracts were combined, dried over anhydrous Na 2 S0 4 , and concentrated. The residue was dried in vacuo to give compound 40-3 ( 15 g, 68 % yield) as a light yellow solid. LC-MS (ESI): m/z 510.5 [M + H] + . (0292] Step 3.
  • Step 5 To a stirred solution of compound 40-5a (5 g, 14.1 mol) in anhydrous THF (50 mL) was added 1 M TBAF in THF (14.1 mL). After refluxing for 16 hrs, the reaction mixture was concentrated. The residue was purified by silica gel column chromatography
  • Step 6 To a stirred solution of compound 40-6a (5.0 g, 14 mmol) and anhydrous Et 3 N ( 11.6 mL, 85 mmol) in dry DCM (200 mL) at 0 °C was added MsCl (4.3 mL, 56 mmol) dropwise. After stirring at rt for 16 hrs, the reaction mixture was concentrated and the residue was diluted with water (100 mL). The mixture was extracted with EtOAc (100 mL x3). The organic extracts were combined, dried with anhydrous Na 2 SC>4, and concentrated. The residue was dried in vacuo and then dissolved in MeOH (150 mL), followed by adding LiOH ( 1 M aq. solution, 25 mL).
  • Step 7 To a stirred solution of the compound 40-7a (2.5 g, 5.8 mmol) in DME (30 mL) and 2 M aq. Na 2 C0 3 solution (7.5 mL) was added Pd(PPh 3 ) 2 Cl 2 (407 mg, 0.58 mmol), followed by vinylboronic acid pinacol ester (1.13 g, 8.7 mmol) under an atmosphere of N 2 . After stirring at 80 °C for 4 hrs, the reaction mixture was concentrated and the residue was poured into water ( 100 mL). The mixture was extracted with EtOAc (50 mL x 3). The extracts were combined, washed with brine, dried with anhydrous Na 2 S0 4 , and concentrated. The residue was purified by silica gel column chromatography (DCM) to give compound 40-8a (900 mg, 47% yield) as a yellow solid.
  • Step 11 To a solution of compound 40- 11 a ( 100 mg, 0.19 mmol) and Et 3 N (0.13 mL, 0.94 mmol) in DMF (3 mL) and MeOH (3 mL) was added ( 13 mg, 0.019 mmol) under an atmosphere of N 2 . Subsequently, the reaction mixture was saturated with CO and stirred at 60 °C for 24 hrs under an atmosphere of CO. The reaction mixture was concentrated and the residue was diluted with water (25 mL). The mixture was extracted with EtOAc (25 mL x 4). The organic extracts were combined, washed with brine, dried with anhydrous a 2 S0 4 , and concentrated.
  • Step 1 Refer to Scheme 42a. To a stirred solution of sodium propane-2-thiolate (/-PrSNa) (120 mg, 1.22 mmol) in anhydrous DMF (8 mL) was added I (267 mg, 1.62 mmol) at rt under an atmosphere of N 2 . After stirring at rt for 30 min, compound 36e-l (500 mg, 0.81 mmol) was added in small portions. The resulting mixture was stirred at rt overnight and then diluted with ice water. The suspension was filtered and the solid was washed with water and dried in vacuo to give crude compound 42a-l (450 mg, 67% purity based on LC-MS), which was used for the next step without further purification.
  • Step 2 To a stirred solution of compound 42a-l (450 mg, 0.81 mmol) in acetone (50 mL) was added hydrogen peroxide (20 mL, 30 wt. % in H 2 Q at rt. After stirring at 60 °C overnight, the reaction mixture was cooled to rt and quenched with saturated aq. Na 2 S0 3 solution (20 mL). The mixture was concentrated to remove acetone and the resulting suspension was filtered. The solid was washed with water and purified by preparative HPLC to give compound 42a-2 (110 mg, 25% yield) as a white solid.
  • icai activity of the compounds of the invention was determined using an HCV replicon assay.
  • the lb_Huh-Luc/Neo-ET cell line persistently expressing a bicistronic genotype lb replicon in Huh 7 cells was obtained from ReBLikon GMBH. This cell line was used to test compound inhibition using luciferase enzyme activity readout as a measurement of compound inhibition of replicon levels.
  • Biological activity of the compounds of the invention was determined using an HCV replicon assay.
  • the 1 b_Huh-Luc Neo-ET cell line persistently expressing a bicistronic genotype lb replicon in Huh 7 cells was obtained from ReBLikon GMBH. This cell line was used to test compound inhibition using luciferase enzyme activity readout as a measurement of compound inhibition of replicon levels.
  • Compounds of the invention can inhibit multiple genotypes of HCV selected but not limited to la, l b, 2a, 3a, and 4a.
  • the ECs 0 s are measured in the corresponding replicon assays that are similar to HCV lb replicon assay as described above.
  • Exemplary compounds of the disclosed invention are illustrated in the Tables attached as Appendix A and Appendix B.
  • Apendix A shows inhibitory activity of the compound against HCV lb.
  • the biological activity against HCV lb is indicated as being *, **, ***, or *** ⁇ , which corresponds to EC*) ranges of EC50 > 1000 nM, 100 nM ⁇ EC 50 ⁇ 1000 nM, 10 nM ⁇ ECso ⁇ 100 nM, or EC50 ⁇ 10 nM, respectively.
  • Appendix A shows structures of compounds of the invention identified by ID NOD B243- B577, and ECJO values determined for the compounds. Of these, all of compounds B243-B282, B285-B289, B292, B294-B426, B429-B438, B440-B443, B445-B469, B471-B540, B542-B549, B551 -B552, B554-B556, B558-B559, B561 -B575 had a measured activity of either 10 nM ⁇ EC50 ⁇ 100 nM, or EC50 ⁇ 10 nM.
  • the invention includes a pharmaceutical composition comprising one or more of the compounds named above, together with one or more pharmaceutically acceptable excipients or vehicles, and optionally other therapeutic and/or prophylactic ingredients.
  • the composition may further include one, two or three anti-HCV agent(s) selected from the group consisting of interferon-alpha, ribavirin, cyclosporine derivatives, NS3 protease inhibitors, NS4B inhibitors, NS5A inhibitors, NS5B polymerase inhibitors, and p7 inhibitors.
  • the compounds described herein may be formulated in a pharmaceutical composition
  • a pharmaceutical composition comprising one or more compounds, optionally together with one or more pharmaceutically acceptable excipients or vehicles, and optionally other therapeutic and/or prophylactic ingredients.
  • excipients are known to those of skill in the art.
  • Pharmaceutically acceptable salts can be used in the compositions of the present invention and include, for example, mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates and the like; and the salts of organic acids such as acetates, propionates, malonates, benzoates and the like.
  • mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates and the like
  • organic acids such as acetates, propionates, malonates, benzoates and the like.
  • the pharmaceutical compositions may be in the form of solid, semi-solid or liquid dosage forms, such as, for example, tablets, suppositories, pills, capsules, powders, liquids, suspensions, creams, ointments, lotions or the like, preferably in unit dosage form suitable for single administration of a precise dosage.
  • the compositions will include an effective amount of the selected drug in combination with a pharmaceutically acceptable carrier and, in addition, may include other pharmaceutical agents, adjuvants, diluents, buffers, etc.
  • the compound comprised within the pharmaceutical composition includes isomers, racemic or non-racemic mixtures of isomers, or pharmaceutically acceptable salts or solvates thereof, together with one or more pharmaceutically acceptable carriers and optionally other therapeutic and/or prophylactic ingredients.
  • conventional nontoxic solid carriers include, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talc, cellulose, glucose, sucrose, magnesium carbonate and the like.
  • the composition will generally take the form of a tablet, capsule, a softgel capsule nonaqueous solution, suspension or syrup. Tablets and capsules are preferred oral administration forms. Tablets and capsules for oral use will generally include one or more commonly used carriers such as lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. When liquid suspensions are used, the active agent may be combined with emulsifying and suspending agents. If desired, flavoring, coloring and/or sweetening agents may be added as well. Other optional components for incorporation into an oral formulation herein include, but are not limited to, preservatives, suspending agents, thickening agents and the like.
  • ⁇ 03521 In yet another aspect, provided herein is the use of the compounds of the invention in the manufacture of a medicament, e.g., for the treatment of hepatitis C.
  • a method of treating hepatitis C comprises administering to a subject in need thereof, a therapeutically effective amount of a compound of the invention, optionally contained in a pharmaceutical composition.
  • a pharmaceutically or therapeutically effective amount of the composition will be delivered to the subject.
  • the precise effective amount will vary from subject to subject and will depend upon the species, age, the subject's size and health, the nature and extent of the condition being treated, recommendations of the treating physician, and the therapeutics or combination of therapeutics selected for administration. Thus, the effective amount for a given situation can be determined and optimized by routine experimentation.
  • the subject may be administered as many doses as is required to reduce and/or alleviate the signs, symptoms or causes of the disorder in question, or bring about any other desired alteration of a biological system.
  • One of ordinary skill in the art of treating such diseases will be able, without undue experimentation and in reliance upon personal knowledge and the disclosure of this application, to ascertain a therapeutically effective amount of the compounds of this invention for a given disease.
  • a treatment regimen using one or more of the compounds provided herein, optionally in combination with one or more anti-HCV agents, will generally involve administering a therapeutically effective dose of the compound, which can readily be determined by one skilled in the art taken into account variables as previously described.
  • a typical dose of a compound as provided herein will generally range from about 10 mg to 1000 mg a day.
  • Representative dosage ranges include from about 10 mg to 900 mg a day, or from about 30 mg to about 700 mg a day, from about 50 mg to about 600 mg a day.
  • a therapeutically effective dosage amount will typically range from 0.1 mg kg body weight to about 30 mg/kg body weight, or from about 0.1 mg kg body weight to about 15 mg kg body weight.
  • the compound may be administered once or multiple times (two to three times) daily, once a week, twice a week, three times a week, etc., over a time course effective to achieve a positive virologic response.
  • Representative courses of treatment include but are not limited to 10 weeks, 12 weeks, 24 weeks, 36 weeks, 48 weeks, and the like.
  • a positive virologic response generally refers to undetectable HCV RNA in serum as measured by PCR and a biochemical response (normalization of aminotransferase levels).
  • post-treatment biopsies are generally less preferable to an assessment based upon a virologic response.
  • the treatment compound may be one or more of the compounds of B243, B244, B245, B246, B248, B249, B252, B253, B255, B256, B257, B258, B259, B260, B261, B262,B263, B264, B265, B266, B267, B268, B269, B270, B271, B272, B273, B274, B275, B276, B277, B278, B279, B280, B281, B282, B283, B284, B285, B286, B287, B288, B289, B290, B291, B293, B294, B295, B296, B297, B298, B299, B300, B301, B302, B303, B304, B305, B306, B307, B308, B309, B310, B31 1, B312, B313, B314, B315, B316, B317, B318, B319, B320, B321, B322,
  • the administering may be by oral administration. Also disclosed is a compound as named above for use in treating HCV infection in an infected subject.
  • treatment by administering a compound as provided herein is effective to result in at least a 2 log reduction in HCV RNA levels over the course of treatment.
  • HCV RNA will still remain positive (detectable) after a course of treatment, although ideally, treatment is effective to achieve undetectable HCV RNA after completing a round of therapy.
  • Subjects may also be assessed following completion of therapy to confirm maintenance of a positive and sustained virologic response (undetectable HCV RNA levels).
  • follow-up assessments may be carried out over the course of months or even years following treatment.
  • J0357[ The compounds of the present invention and their isomeric forms and pharmaceutically acceptable salts thereof are useful in treating and preventing HCV infection alone or when used in combination with other compounds targeting viral or cellular elements or functions involved in the HCV life cycle.
  • Classes of compounds useful in the invention may include, without limitation, all classes of HCV an ivirals, both direct-acting and indirect-acting ('cell-targeted' inhibitors of HCV replication).
  • mechanistic classes of agents that may be useful when combined with the compounds of the present invention include, for example, nucleoside and non-nucleoside inhibitors of the HCV protease inhibitors, helicase inhibitors, NS5A inhibitors, NS4B inhibitors and medicinal agents that functionally inhibit the internal ribosomal entry site (IRES), other NS5B inhibitors and other medicaments that inhibit HCV cell attachment or virus entry, HCV RNA translation, HCV RNA transcription, replication or HCV maturation, assembly or virus release.
  • IRS internal ribosomal entry site
  • telaprevir VX- 950
  • boceprevir SCH-503034
  • narlaprevir SCH-900518
  • ITMN-191 R-7227
  • TMC-435350 a.k.a.
  • nucleosidic HCV polymerase (replicase) inhibitors useful in the invention include, but are not limited to, ALS2200, R7128, PSI-7851, PS1-7977 (GS-7977), PSI-938, PSI-879, PSI-6130, IDX-184, IDX-102, ⁇ -189, R1479, R1626, UNX-08189, and various other nucleoside and nucleo
  • NS5A inhibitors useful in the invention include, but are not limited to, ⁇ -46 , PPI-668, BMS-790052, BMS-824393, GS-5885, EDP-239, ACH-2928, AZD-7295, IDX-719, IDX-380, ABT-267, GS -2336805, CF-102, A-831 and ⁇ -9916.
  • Non-nucleosidic HCV polymerase (replicase) inhibitors useful in the invention include, but are not limited to, VCH-759, VCH-916, VCH-222 (VX-222), ANA-598, M -3281, ABT-333, ABT- 072, PF-00868554 (filibuvir), BI-207127, GS-9190, A-837093, GS -625433, J T-109, GL- 59728 and GL-60667.
  • HCV P7 inhibitors useful in the present invention include BIT-225 and other P7 inhibitors, as well as HCV NS4B inhibitors including but not limited to histamine agents that antagonize HCV NS4B function.
  • NS5B inhibitors of the present invention may be used in combination with cyclophyllin and immunophyllin antagonists (eg, without limitation, DEBIO compounds, NM- 81 1, SCY-635, EP-CyP282, as well as cyclosporine and its derivatives), kinase inhibitors, inhibitors of heat shock proteins (e.g., HSP90 and HSP70), other immunomodulatory agents that may include, without limitation, interferons (-alpha, -beta, -omega, -gamma, -lambda or synthetic) such as Intron ATM, Roferon-ATM, Canferon-A300TM, AdvaferonTM, InfergenTM, HumoferonTM, Sumiferon MPTM, AlfaferoneTM, IFN- ⁇ TM, FeronTM and the like; polyethylene glycol derivatized (pegylated) interferon compounds, such as PEG interferon-a-2a (Pegas
  • any of the above- described methods involving administering an NS5B inhibitor, a Type I interferon receptor agonist (e.g., an IFN-a) and a Type II interferon receptor agonist (e.g., an IFN- ⁇ ) can be augmented by administration of an effective amount of a TNF- antagonist
  • a Type I interferon receptor agonist e.g., an IFN-a
  • a Type II interferon receptor agonist e.g., an IFN- ⁇
  • TNF- antagonists that are suitable for use in such combination therapies include ENBRELTM, REMICADETM and HUMI ATM.
  • J0360J NS5B inhibitors of the present invention may also be used with alternative forms of interferons and pegylated interferons, ribavirin or its analogs (e.g., tarabavarin, levoviron), microRNA, small interfering RNA compounds (e.g., S1RPLEX-140-N and the like) and microR A agents (such as micro-RNA-122), nucleotide or nucleoside analogs, multifunction inhibitors such as nitazoxanide, immunoglobulins, hepatoprotectants, anti-inflammatory agents and other direct and indirect inhibitors of HCV replication.
  • ribavirin or its analogs e.g., tarabavarin, levoviron
  • microRNA small interfering RNA compounds
  • microR A agents such as micro-RNA-122
  • nucleotide or nucleoside analogs such as multifunction inhibitors such as nitazoxanide, immunoglobulins, hepatoprotect
  • Inhibitors of other targets in the HCV life cycle include NS3 helicase inhibitors; NS4A co- factor inhibitors; and sense oligonucleotide inhibitors, such as ISIS- 14803, ISlS-11, AVI-4065 and the like; vector-encoded short hairpin RNA (shRNA); HCV specific ribozymes such as heptazyme, RPI-13919 and the like; entry inhibitors such as HepeX-C, HuMax-HepC and the like; alpha glucosidase inhibitors such as celgosivir, UT-231B and the like; KPE-02003002 and BIVN 401 and IMPDH inhibitors.
  • NS3 helicase inhibitors such as ISIS- 14803, ISlS-11, AVI-4065 and the like
  • shRNA vector-encoded short hairpin RNA
  • HCV specific ribozymes such as heptazyme, RPI-13919 and the like
  • entry inhibitors
  • HCV inhibitor compounds include those disclosed in the following publications: U.S. Pat. No. 5,807,876; U.S. Pat. No. 6,498,178; U.S. Pat. No. 6,344,465; U.S. Pat. No. 6,054,472; U.S. Pat. No. 7,759,495; U.S. Pat. No. 7,704,992; U.S. Pat. No.
  • combinations of, for example, ribavirin a NS3 protease inhibitor, a replicase inhibitor and interferon may be administered as multiple combination therapy with at least one of the compounds of the present invention.
  • the present invention is not limited to the
  • combination therapies of the present invention include any chemically compatible combination of a compound of this inventive group with other compounds of the inventive group or other compounds outside of the inventive group, as long as the combination does not eliminate the anti-viral activity of the compound of this inventive group or the anti-viral activity of the pharmaceutical composition itself
  • Combination therapy can be sequential, that is treatment with one agent first and then a second agent (for example, where each treatment comprises a different compound of the invention or where one treatment comprises a compound of the invention and the other comprises one or more biologically active agents) or it can be treatment with both agents at the same time (concurrently).
  • Sequential therapy can include a reasonable time after the completion of the first therapy before beginning the second therapy. Treatment with both agents at the same time can be in the same daily dose or in separate doses.
  • the dosages for both concurrent and sequential combination therapy will depend on absorption, distribution, metabolism and excretion rates of the components of the combination therapy as well as other factors known to one of skill in the art. Dosage values will also vary with the severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens and schedules may be adjusted over time according to the individual's need.

Abstract

A class of compounds that inhibit Hepatitis C Virus (HCV) is disclosed, along with compositions containing the compound, and methods of using the composition for treating individuals infected with HCV. The invention is intended to include all isotopically labeled analogs of the compounds of the invention. Isotopes include those atoms having the same atomic number but different mass.

Description

INHIBITORS OF HEPATITIS C VIRUS
Field
[001] The invention relates to compounds useful for inhibiting hepatitis C virus ("HCV") replication, particularly functions of the non-structural 5B ("NS5B") protein of HCV.
Background
[002] HCV is a single-stranded RN A virus that is a member of the Flaviviridae family. The virus shows extensive genetic heterogeneity as there are currently seven identified genotypes and more than 50 identified subtypes. In HCV infected cells, viral RNA is translated into a polyprotein that is cleaved into ten individual proteins. At the amino terminus are structural proteins: the core (C) protein and the envelope glycoproteins, El and E2, p7, an integral membrane protein, follows El and E2. Additionally, there are six non-structural proteins, NS2, NS3, NS4A, NS4B, NS5A and NS5B, which play functional roles in the HCV life cycle, (see, for example, B.D. Lindenbach and CM. Rice, Nature. 436:933-938, 2005). NS5B is the RNA polymerase or replicase of the virus and is responsible for replication of both positive and negative-strand genomic RNA during the viral replicative cycle. NS5B plays an essential and critical role in viral replication, and a functional NS5B replicase is required for HCV replication and infection. Thus, inhibition of NS5B RNA-dependent polymerase activity is believed to be an effective way of treating HCV infection.
[003] Infection by HCV is a serious health issue. It is estimated that 170 million people worldwide are chronically infected with HCV. HCV infection can lead to chronic hepatitis, cirrhosis, liver failure and hepatocellular carcinoma. Chronic HCV infection is thus a major worldwide cause of liver-related premature mortality.
[004 [ The current standard of care treatment regimen for HCV infection involves interferon- alpha, alone, or in combination with ribavirin and a protease inhibtor. The treatment is cumbersome and sometimes has debilitating and severe side effects and many patients do not durably respond to treatment. New and effective methods of treating HCV infection are urgently needed.
i Summary
[005] Essential functions of the NS5B protein in HCV replication make it an attractive intervention target for treating HCV infection. The present disclosure describes a class of compounds targeting the NS5B protein and methods of their use to treat HCV infection in humans.
[006] More particularly, the present disclosure describes a class of heterocyclic compounds targeting HCV NS5B polymerase and methods of their use to treat HCV infection in humans.
[007 [ In a first aspect of the invention is a compound that has the structure:
Figure imgf000003_0001
wherein,
L1, L2 and-NCSQzR12)- together with the attached carbons of the aromatic ring to form a 5-12 member ring, containing 1-4 heteroatoms of N, O, S, P and/or Si;
L'or L2 is independently selected from the group consisting of a bond, -0-, -C(RI5R16)-, -NR3-, - S(O » -P(0 , -Si(R4R5K -C(O)-, -C(0)0-, and substituted alkyl, alkenyl, alkoxy, cycloalkyl, cycloalkoxy, heterocycle, aryl, heteroaryl, amide, carbamate, urea, and sulfonamide;
n is 0, 1, or 2; and
R1 is selected from hydrogen, halide, -CF3, -CN, -C(0)H, -C(0)OR6, -C(0)NHR7, - 7, -C(=NOR')NHR7, -C(CF3)NHR8, -C(CN)NHR9, -S(0)2NHR10, -
Figure imgf000003_0002
R' is selected from hydrogen, -(CH2CHzO)o-8-Me, C2.24 alkyl optionally containing 0-6 heteroatoms of O, NRN and/or S, and 3-8 cycloalkyl optionally containing 0-2 heteroatora of O, NRN, and/or S;
R2 is an aryl or heteroaryl having one or more R17 substituents;
R3 is selected from the group of hydrogen, alkylcarbonyl, cycloalkylcarbonyl, alkoxylcarbonyl, cycloalkoxycarbonyl, alkylsulfonyl and cycloalkylsulfonyl; R4 and R5 are independently methyl, ethyl, or cyclopropyl;
R12 is independently C].3 alkyl, cyclopropyl, -OMe, or -NHMe;
R15, R16 is independently hydrogen, hydroxyl, azide, C2- alkeny!,
Figure imgf000004_0001
CM alkyl, cyclopropyl, C alkoxy, or cyclopropoxy or R15 and R16 together are a carbonyl or C alkenylidene or R15 and R16 joined together with the attached carbon are 3-6 member ring optionally containing 0-3 heteroatoms of O, NRN and or S; and
R,7 is F, Cl or CN.
(008] The compound of this embodiment may have an inhibitory activity with respect to HCV,
Figure imgf000004_0002
Rv is selected from hydrogen, -NR'R", alkoxyls, alkyls, cycloalkyls, aryls, heteroaryls, and aralkyl;
Rw and R* are indenpendrntry selected from hydrogen, -C(0)H, -C(0)RHi, -C(0)ORiv-, - C^NR'H", alkoxyls, alkyls, cycloalkyls, aryls, heteroaryls, and aralkyl; optionally, Rv and R together form a C4.8 member ring;
Ry and Rz are independently selected from hydrogen, alkoxyls, alkyls, cycloalkyls, aryls, heteroaryls, and aralkyl; optionally, Ry and Rz together form a 3-8 member ring;
R' and R" are indenpendently selected from hydrogen, -Me, -Et, c-Pr-, c-Butyl;
R"1 is selected from alkyoxyl, alkyls, cycloalkyls, aryls, heteroaryls, and aralkyl;
Rlv is selected from alkyoxyl, alkyls, cycloalkyls, aryls, heteroaryls, and aralkyl; and Rv and RVI are indenpendently selected from hydrogen, aUcyoxyl, alkyls, cycloalkyls, aryls, heteroaryls, and aralkyl.
[010] In one general embodiment, the compound has the structural formula:
Figure imgf000005_0001
Figure imgf000006_0001
Figure imgf000007_0001
Figure imgf000008_0001
Figure imgf000009_0001
ο-ά&F #o o#°T#°,o#o
Figure imgf000010_0001
Figure imgf000010_0002
Figure imgf000010_0003
o
Figure imgf000011_0001
X' is selected from
Figure imgf000011_0002
and
R'3 is selected from -CH3 and -OCH3.
[011[ In a more specific embodiment of the compound above, is para-F, R'2is as above, X' is =0, and R'3 is -CH3.
[012] The compound may be selected from the group consisting of B243, B244, B245, B246, B248, B249, B252, B253, B255, B256, B257, B258, B259, B260, B261, B262,B263, B264, B265, B266, B267, B268, B269, B270, B271, B272, B273, B274, B275, B276, B277, B278, B279, B280, B281, B282, B283, B284, B285, B286, B287, B288, B289, B290, B291, B293, B294, B295, B296, B297, B298, B299, B300, B301, B302, B303, B304, B305, B306, B307, B308, B309, B310, B311, B312, B313, B314, B315, B316, B317, B318, B319, B320, B321, B322, B323, B326, B327, B328, B329, B330, B333, B334, B335, B336, B337, B338, B339, B340, B341, B342, B343, B344, B345, B346, B347, B348, B349, B350, B351, B352, B353, B354, B355, B356, B357, B358, B360, B36i, B362, B363, B364, B365, B366, B367, B368, B369, B370, B371, B372, B373, B374, B375, B376, B377, B378, B379, B380, B381, B382, B383, B384, B385, B386, B387, B388, B389, B390, B391, B392, B393, B394, B395, B396, B397, B398, B399, B400, B401, B402, B403, B404, B405, B406, B407, B408, B409, B410, B411, B412, B413, B414, B415, B416, B417, B418, B419, B420, B421, B422, B423, B424, B425, B426, B427, B428, B429, B430, B431, B432, B433, B434, B435, B436, B437, B438, B439, B440, B441, B442, B443, B444, B446, B447, B448, B449, B450, B451, B452, B453, B454, B455, B456, B457, B458, B459, B460, B461, B462, B463, B464, B465, B466, B467, B468, B469, B470, B471, B472, B473, B474, B475, B476, B477, B478, B479, B480, B481, B482, B483, B484, B485, B486, B487, B488, B489, B490, B491, B492, B493, B494, B495, B496, B497, B498, B499, B500, B501, B502, B503, B504, B505, B506, B507, B508, B509, B510, B511, B512, B513, B514, B515, B516, B517, B518, B51 , B520, B521, B522, B523, B524, B525, B526, B527, B528, B529, B530, B531, B532, B533, B534, B535, B536, B537, B538, B539, B540, B541, B542, B543, B544, B545, B546, B547, B548, B549, B550, B551, B552,
B563, B564, B565, B566, B567, B568, and B577.
hemical structure shown below:
Figure imgf000012_0001
[014] In one general embodiment, indicted as Group 1 herein, the compound has have the structural formula:
Figure imgf000012_0002
wherein:
Figure imgf000012_0003
i t
Figure imgf000013_0001

Figure imgf000014_0001
Χ' is selected from =0, =N-OH,
Figure imgf000014_0002
and «N-0(CH2)i-23CH3, and R'3 is selected firom -CH3 and -OCH3. [01 SJ The above compound may have the same structural formula, wherein R'i is pa a-F, Ru-0- is as above, and X* is =0, and R^ is -C¾.
[016] The compound in this group may be selected from the group consisting of B243, B244, B245, B246, B248, B249, B252, B253, B255, B256, B257, B258, B259, B260, B261, B262,B263, B264, B265, B266, B267, B268, B269, B270, B271, B272, B273, B274, B275, B276, B277, B278, B279, B280, B281, B282, B285, B286, B289, B300, B302, B306, B307, B308, B309, B310, B311, B314, B316, B317, B318, B319, B320, B333, B335, B336, B337, B338, B343, B344, B345, B346, B347, B348, B349, B350, B351, B352, B353, B354, B373, B378, B379, B381, B383, B384, B385, B386, B388, B389, B390, B391, B392, B394, B395, B397, B399, B400, B401, B402, B404, B405, and B409.
cated herein as Group 2, the compound has the
Figure imgf000015_0001
wherein:
R'i is selected from -H, -F, -CI, -Br, -I,
[018J Rv-S(OV, wherein n = 0, 1, 2, is selected from
Figure imgf000015_0002
o#o θ HO^#O - #o ~ D o -o#,
Figure imgf000016_0001
IS
Figure imgf000017_0001
*nr"£. °cr¾, T¾, 0"¾, ° T¾
Figure imgf000017_0002
X' is selected
Figure imgf000017_0003
and R'3 is selected from -CH3 and -OCH3.
[019J The compound in this group may have the same structure where R'i is para-F, Rv-S(0)„-, are as above, X' is =0, and R'3 is -CH3. [020J Specific compounds in this group (Group 2) are selected from the group consisting of B283, B284, B287, B288, B290, B291, B293, B422, B423, B424, B426, B432, B443, B446, B447, B455, B456, B459, B463, B464, B466, B467, B470, B478, B479, B487, B488, B489, B491, B492, B493, B494, B495, B496, B497, B498, B499, B500, B501, B502, B503, B504, B505, B506, B507, B508, B509, B510, B511, B512, B513, B514, B515, B516, B517, B518, B519, B520, B521, B522, B523, B524, B525, B526, B527, B528, B529, B530, B531, B532, B533, B534, B535, B536, B537, B538, B539, B540, B541, B542, B543, B544, B545, B546, B547, B548, B549, B550, B551, B552, B553, B554, B555, B556, B557, B558, B561, B562, B563, B564, B565, B566, B567, B568, B569, B570, B571, B572, B573, B574, B575, B576, and B577.
[021] In another general embodiment, indicated herein as Group 3, the compound of the invention has the structural formula:
Figure imgf000018_0001
0*§ CH3 wherein:
Figure imgf000018_0002
Figure imgf000019_0001
Figure imgf000020_0001
X' is selected
Figure imgf000020_0002
and
R'3 is selected from -C¾ and -OCH3.
[023] The compound in this group may have the same structural formula wherein: R'i is para-Έ, R R*N- is selected as above, X' is =0, and R'3 is -C¾.
[024J Specific compounds in this group (Group 3) are selected from the group consisting of B294, B295, B296, B297, B298, B299, B301, B303, B304, B305, B312, B313, B315, B321, B322, B323, B326, B327, B328, B329, B330, B334, B339, B340, B341, B342, B355, B356, B357, B358, B360, B361, B362, B363, B364, B365, B366, B367, B368, B369, B370, B371, B372, B374, B375, B376, B377, B380, B382, B387, B393, B396, B398, B403, B406, B407, B408, B410, B411, B412, B413, B414, B415, B416, B417, B418, B419, B420, B421, B425, B427, B428, B429, B430, B431, B433, B434, B435, B436, B437, B438, B439, B440, B441, B442, B444, B448, B449, B450, B451, B452, B453, B454, B457, B458, B460, B461, B462, B465, B468, B471, B472, B473, B474, B475, B476, B477, B480, B481, B482, B483, B484, B485, B486, and B490.
of the invention has the structural formula:
Figure imgf000021_0001
wherein:
R'l is selected from para-F
Figure imgf000021_0002
R'2 is selected from -OH and -S02Me; and R'3 is selected from -CH3 and -OCH3, as exemplified by B324, B325, B331, and B332.
[026] In still another aspect, the compound of the invention has the structural formula:
Figure imgf000021_0003
wherein:
R' i is selected from para-F
Figure imgf000021_0004
;
R'2 is selected from -OH and -S02Me; and R* 3 is selected from -CH3 and -OCH3, as exemplified by compound B359. [027] Also forming part of the invention is a pharmaceutical composition comprising a compound of any one of compounds above, or its pharmaceutically acceptable salts, together with one or more pharmaceutically acceptable excipients or vehicles, and optionally other therapeutic and/or prophylactic ingredients. The composition may further includes additional one, two or three anti-HCV agent(s) selected from the group consisting of interferon-alpha, ribavirin, cyclosporine derivatives, HCV NS3 protease inhibitors, HCV NS4B inhibitors, HCV NS5 A inhibitors, HCV NS5B polymerase inhibitors, and p7 inhibitors.
In still another aspect, the invention includes a method of treating HCV infection in a subject comprising administering to the subject, a pharmaceutically acceptable dose of the compound above, and continuing the administering until a selected reduction in the subject's HCV titer is achieved
[028] The method may use a compound selected from the group consisting of B243, B244, B245, B246, B248, B249, B252, B253, B255, B256, B257, B258, B259, B260, B261, B262,B263, B264, B265, B266, B267, B268, B269, B270, B271, B272, B273, B274, B275, B276, B277, B278, B279, B280, B281, B282, B283, B284, B285, B286, B287, B288, B289, B290, B291, B293, B294, B295, B296, B297, B298, B299, B300, B301, B302, B303, B304, B305, B306, B307, B308, B309, B310, B311, B312, B313, B314, B315, B316, B317, B 18, B319, B320, B321, B322, B323, B326, B327, B328, B329, B330, B333, B334, B335, B336, B337, B338, B339, B340, B341, B342, B343, B344, B345, B346, B347, B348, B349, B350, B351, B352, B353, B354, B355, B356, B357, B358, B360, B361, B362, B363, B364, B365, B366, B367, B368, B369, B370, B371, B372, B373, B374, B375, B376, B377, B378, B379, B380, B381, B382, B383, B384, B385, B386, B387, B388, B389, B390, B391, B392, B393, B394, B395, B396, B397, B398, B399, B400, B401, B402, B403, B404, B405, B406, B407, B408, B409, B410, B411, B412, B413, B414, B415, B416, B417, B418, B419, B420, B421, B422, B423, B424, B425, B426, B427, B428, B429, B430, B431. B432, B433, B434, B435, B436, B437, B438, B439, B440, B441, B442, B443, B444, B446, B447, B448, B449, B450, B451, B452, B453, B454, B455, B456, B457, B458, B459, B460, B461, B462, B463, B464, B465, B466, B467, B468, B469, B470, B471, B472, B473, B474, B475, B476, B477, B478, B479, B480, B481, B482, B483, B484, B485, B486, B487, B488, B489, B490, B491, B492, B493, B494, B495, B496, B497, B498, B499, B500, B501, B502, B503, B504, B505, B506, B507, B508, B509, B510, B511, B512, B513, B514, B515, B516, B517, B518, B519, B520, B521, B522, B523, B524, B525, B526, B527, B528, B529, B530, B531, B532, B533, B534, B535, B536, B537, B538, B539, B540, B541, B542, B543, B544, B545, B546, B547, B548, B549, B550, B551, B552, B553, B554, B555, B556, B557, B558, B561, B562, B563, B564, B565, B566, B567, B568, B569, B570, B571, B572, B573, B574, B575, B576, and B577.
[029] In one embodiment of the method, the compound is selected from the group (Group 1 ) consisting of B243, B244, B245, B246, B248, B249, B252, B253, B255, B256, B257, B258, B259, B260, B261, B262.B263, B264, B265, B266, B267, B268, B269, B270, B271, B272, B273, B274, B275, B276, B277, B278, B279, B280, B281, B282, B285, B286, B289, B300, B302, B306, B307, B308, B309, B310, B311, B314, B316, B317, B318, B319, B320, B333, B335, B336, B337, B338, B343, B344, B345, B346, B347, B348, B349, B350, B351, B352, B353, B354, B373, B378, B379, B381, B383, B384, B385, B386, B388, B389, B390, B391, B392, B394, B395, B397, B399, B400, B401, B402, B404, B405, and B409.
[030] In another embodiment of the method, the compound is selected from the group (Group 2) consisting of B283, B284, B287, B288, B290, B291, B293, B422, B423, B424, B426, B432, B443, B446, B447, B455, B456, B459, B463, B464, B466, B467, B470, B478, B479, B487, B488, B489, B491, B492, B493, B494, B495, B496, B497, B498, B499, B500, B501, B502, B503, B504, B505, B506, B507, B508, B509, B510, B511, B512, B513. B514, B515, B516, B517, B518, B519, B520, B521, B522, B523, B524, B525, B526, B527, B528, B529, B530, B531, B532, B533, B534, B535, B536, B537, B538, B539, B540, B541, B542, B543, B544, B545, B546, B547, B548, B549, B550, B551, B552, B553, B554, B555, B556, B557, B558, B561, B562, B563, B564, B565, B566, B567, B568, B569, B570, B571, B572, B573, B574, B575, B576, and B577.
[031] In still another embodiment of the method, the compound is selected from the group (Group 3) consisting of B294, B295, B296, B297, B298, B299, B301, B303, B304, B305, B312, B313, B315, B321, B322, B323, B326, B327, B328, B329, B330, B334, B339, B340, B341, B342, B355, B356, B357, B358, B360, B361, B362, B363, B364, B365, B366, B367, B368, B369, B370, B371, B372, B374, B375, B376, B377, B380, B382, B387, B393, B396, B398, B403, B406, B407, B408, B410, B411, B412, B413, B414, B415, B416, B417, B418, B419, B420, B421, B425, B427, B428, B429, B430, B431, B433, B434, B435, B436, B437, B438, B439, B440, B441, B442, B444, B448, B449, B450, B451, B452, B453, B454, B457, B458, B460, B461, B462, B465, B468, B471, B472, B473, B474, B475, B476, B477, B480, B481, B482, B483, B484, B485, B486, and B490.
[032] The method may be practiced by administering the compound orally.
[033] Also disclosed is one or more of the above compounds for use in treating HCV infection in an infected subject.
[034] Further disclosed is the use of a compound as set forth herein, such as one of the compounds identified by ID NOS, in the preparation of a medicament for the treatment of HCV in an HCV-infected subject
[035] Some of the compounds of the invention possess chiral carbons. The invention included all stereoisomeric forms, including enantiomers and diastereomers as well as mixtures of stereoisomers such as racemates. The stereoisomers or their precursors can be either
asymmetrically synthesized or obtained by separations of the racemates according to methods commonly known in the art.
[036] The invention is intended to include all isotopically labeled analogs of the compounds of the invention. Isotopes include those atoms having the same atomic number but different mass. For example, isotopes of hydrogen include 2H(D) and 3H(T) and isotopes of carbon include ,3C and 14C. Isotopically labeled compounds of the invention can be prepared according to methods commonly known in the art. Such compounds may have various potential uses as, but not limited to, standards and reagents in determining biological/pharmacological activities. For those stable isotopically labeled compounds of the invention, they may have the potential to favorably modulate biological, pharmacological, or pharmacokinetic properties.
[037] Additional embodiments of the compounds, compositions, methods, uses and the like will be apparent from the following description, examples, and claims. As can be appreciated from the foregoing and following description, each and every feature described herein, and each and every combination of two or more of such features, is included within the scope of the present disclosure provided that the features included in such a combination are not mutually inconsistent. In addition, any feature or combination of features may be specifically excluded from any embodiment of the present invention. Additional aspects and advantages of the present invention are set forth in the following description, particularly when considered in conjunction with the accompanying examples.
Detailed Description
[038] Unless otherwise stated, the following terms used in this application, including the specification and claims, have the definitions given below. It must be noted that, as used in the specification and the appended claims, the singular forms "a," "an" and "the" include plural referents unless the context clearly dictates otherwise. Definition of standard chemistry terms may be found in reference works, including Carey and Sundberg (2007) "Advanced Organic Chemistry 5th Ed." Vols. A and B, Springer Science+Business Media LLC, New York. The practice of the present invention will employ, unless otherwise indicated, conventional methods of synthetic organic chemistry, mass spectroscopy, preparative and analytical methods of chromatography, protein chemistry, biochemistry, recombinant DNA techniques and
pharmacology.
[039] The term "alkanoyl" as used herein contemplates a carbonyl group with a lower alkyl group as a substituent.
[040] The term "alkenyl" as used herein contemplates substituted or unsubstituted, straight and branched chain alkene radicals, including both the E- and Z-forms, containing from two to eight carbon atoms. The alkenyl group may be optionally substituted with one or more substituents selected from the group consisting ofhalogen, -CN, -N(¼, CC¾R, C(0)R, -O-R, -N(RN)2, - N(RN)C(0)R, -N(RN)S(0)2R, -SR, -C(0)N(RN)2, -OC(0)R,
-OC(0)N(RN) , S(0)R, S02R, -S03R, -S(0)2N(RN)2, phosphate, phosphonate, cycloalkyl, cycloalkenyl, aryl and heteroaryl.
[041] The term "alkoxy" as used herein contemplates an oxygen with a lower alkyl group as a substituent and includes methoxy, ethoxy, butoxy, trifluromethoxy and the like. It also includes divalent substituents linked to two separated oxygen atoms such as, without limitation, -0-CF2-0-, -0-(CH2)M-0-(CH2CH2-0)M- and -(0-CH2C¾-0)M-.
[042] The term "alkoxycarbonyl" as used herein contemplates a carbonyl group with an alkoxy group as a substituent. {043] The term "alkyl" as used herein contemplates substituted or unsubstituted, straight and branched chain alkyl radicals containing from one to fifteen carbon atoms. The term "lower alkyP as used herein contemplates both straight and branched chain alkyl radicals containing from one to six carbon atoms and includes methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert- butyl and the like. The alkyl group may be optionally substituted with one or more substituents selected from halogen, -CN, -N02, -C(0)2R, -C(0)R, -O-R, -N(RN)2, -N(RN)C(0)R, - N(RN)S(0)2R, -SR, -C(0)N(RN)2, -OC(0)R, -OC(0)N(RN)2, -SOR, -S02R, -SO3R, -S(0)2N(RN)2i phosphate, phosphonate, cycloalkyl, cycloalkenyl, aryl and heteroaryl.
[044] The term "alkylene," "alkenylene" and "alkynylene" as used herein refers to the groups "alkyl," "alkenyl" and "alkynyl" respectively, when they are divalent, ie, attached to two atoms. 1045] The term "alkylsulfonyl" as used herein contemplates a sulfonyl group which has a lower alkyl group as a substituent.
[046] The term "alkynyl" as used herein contemplates substituted or unsubstituted, straight and branched carbon chain containing from two to eight carbon atoms and having at least one carbon- carbo triple bond. The term alkynyl includes, for example ethynyl,
1-propynyl, 2- propynyl, 1-butynyl, 3-methyl- 1 -butynyl and the like. The alkynyl group may be optionally substituted with one or more substituents selected from halo, -CN, -N02, -C02R, - C(0)R, -O-R, -N(RN)2, -N(RN)C(0)R, -N(RN)S(0)2R, -SR, -C(0)N(RN)2, -OC(0)R,
-OC(0)N(RN)2, -SOR, -S02R, -SO3R, -S(0)2N(RN)2, phosphate, phosphonate, cycloalkyl, cycloalkenyl, aryl and heteroaryl.
[047] The term "amino" as used herein contemplates a group of the structure -NRN 2.
of the structure
Figure imgf000026_0001
D or the L configuration and includes but is not limited to the twenty "standard" amino acids: isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan, valine, alanine, asparagine, aspartate, cysteine, glutamate, glutamtne, glycine, proline, serine, tyrosine, arginine and histidine. The present invention also includes, without limitation, D-configuration amino acids, beta-amino acids, amino acids having side chains as well as all non-natural amino acids known to one skilled in the art.
[049] The term "aralkyl" as used herein contemplates a lower alkyl group which has a substituent on an aromatic group, which aromatic group may be substituted or unsubstituted. The aralkyl group may be optionally substituted with one or more substituents selected from halogen, -CN, -NO2, -CO2R, -C(0)R, -O-R, -N(RN)2, -N(RN)C(0)R, -N(RN)S(0)2R, -SR,
-C(0)N(RN)2, -OC(0)R, -OC(0)N(RN)2, -SOR, -S02R, -SO3R, -S(0)2N(RN)2, phosphate, phosphonate, cyctoalkyl, cycloalkenyl, aryl and heteroaryl.
[050] The terms "aryl," "aromatic group" or "aromatic ring" as used herein contemplates substituted or unsubstituted single-ring and multiple aromatic groups (for example, phenyl, pyridyl and pyrazole, etc.) and polycyclic ring systems (naphthyl and quinolinyl, etc.). The polycyclic rings may have two or more rings in which two atoms are common to two adjoining rings (the rings are "fused") wherein at least one of the rings is aromatic, e.g., the other rings can be cycloalkyls, cycloalkenyls, aryl, heterocycles and/or heteroaryls. The aryl group may be optionally substituted with one or more substituents selected from halogen, alkyl, -CN, -NO2, - CC¾R, -C(0)R, -O-R, -N(RN)2, -N(RN)C(0)R, -N(RN)S(0)2R, -SR, -C(0)N(RN)2, -OC(0)R, - OC(0)N(RN)2, -SOR, -S02R, -SO3R, -SiO^N^ , -SiR3, -P(0)R, phosphate, phosphonate, cycloalkyl, cycloalkenyl, aryl and heteroaryl.
[051] The term "arylsulfonyl" as used herein contemplates a sulfonyl group which has as a substituent an aryl group. The term is meant to include, without limitation, monovalent as well as multiply valent aryls (eg, divalent aryls).
"carbamoyl" as used herein contemplates a group of the structure
Figure imgf000027_0001
[053] The term "carbonyl" as used herein contemplates a group of the structure
Figure imgf000027_0002
"carboxyl" as used herein contemplates a group of the structure
Figure imgf000027_0003
[055] The term "cycloalkyt" as used herein contemplates substituted or unsubstituted cyclic alkyl radicals containing from three to twelve carbon atoms and includes cyclopropyl, cyclopentyl, cyclohexyl and the like. The term "cycloalkyl" also includes polycyclic systems having two rings in which two or more atoms are common to two adjoining rings (the rings are "fused"). The cycloalkyl group may be optionally substituted with one or more substituents selected from halo, -CN, -NO2, -CO2R, -C(0)R, -O-R, -N( N>2, -N(RN)C(0)R, -N(RN)S(0)2R, -SR, -C(0)N(RN>2, - OC(0)R, -OC(0)N(RN)2, -SOR, -S02R, -S(0)2N(RN)2, phosphate, phosphonate, alkyl,
cycloalkenyl, aryl and heteroaryl.
[056] The term "cycloalkenyl" as used herein contemplates substituted or unsubstituted cyclic alkenyl radicals containing from four to twelve carbon atoms in which there is at least one double bond between two of the ring carbons and includes cyclopentenyl, cyclohexenyl and the like. The term "cycloalkenyl" also includes polycyclic systems having two rings in which two or more atoms are common to two adjoining rings (the rings are "fused"). The cycloalkenyl group may be optionally substituted with one or more substituents selected from halo, -CN, -NO2, -C02R, - C(0)R, -O-R, -N(RN)2, -N(RN)C(0)R, -N(RN)S(0)2R, -SR, -C(0)N(RN)2, -OC(0)R, - OC(0)N(RN)2, -SOR, -S02R, -S(0)2N(RN)2, phosphate, phosphonate, alkyl, cycloalkenyl, aryl and heteroaryl.
[057] The term "halo" or "halogen" as used herein includes fluorine, chlorine, bromine and iodine.
[058] The term "heteroalkyl" as used herein contemplates an alkyl with one or more
heteroatoms.
[059] The term "heteroatom", particularly within a ring system, refers to N, O and S.
[060] The term "heterocyclic group," "heterocycle" or "heterocyclic ring" as used herein contemplates substituted or unsubstituted aromatic and non-aromatic cyclic radicals having at least one heteroatom as a ring member. Preferred heterocyclic groups are those containing five or six ring atoms which includes at least one hetero atom and includes cyclic amines such as morpholtno, piperidino, pyrrolidino and the like and cyclic ethers, such as tetrahydrofuran, tetrahydropyran and the like. Aromatic heterocyclic groups, also termed "heteroaryl" groups, contemplates single-ring hetero-aromatic groups that may include from one to three heteroatoms, for example, pyrrole, furan, thiophene, imidazole, oxazole, thiazole, triazole, pyrazole, oxodiazole, thiadiazole, pyridine, pyrazine, pyridazine, pyrimidine and the like. The term heteroaryl also includes polycyclic hetero-aromatic systems having two or more rings in which two or more atoms are common to two adjoining rings (the rings are "fused") wherein at least one of the rings is a heteroaryl, e.g., the other rings can be cycloalkyls, cycloalkenyls, aryl, heterocycles and/or heteroaryls. Examples of polycyclic heteroaromatic systems include quinoline, isoquinoline, cinnoline, tetrahydroisoquinoline, quinoxaline, quinazoline, benzimidazole, benzofuran, benzothiophene, benzoxazole, benzothiazole, indazole, purine, benzotriazole, pyrrolepyridine, pyrrazolopyridine and the like. The heterocyclic group may be optionally substituted with one or more substituents selected from the group consisting of halo, alkyl, -CN, -NO2, -CO2R, -C(0)R, - O-R, -N(RN)2, -N(RN)C(0)R, -N(RN)S(0)2R, -SR, -C(0)N(RN>2, -OC(0)R, -OC(0)N(RN)2, -SOR, -SO2R, -SO3R, -S(0)2N(RN)2, -S1R3, -P(0)R, phosphate, phosphonate, cycloalkyl, cycloalkenyl, aryl and heteroaryl.
[061] The term "oxo" as used herein contemplates an oxygen attached with a double bond.
[062] By "pharmaceutically acceptable" or ''pharmacologically acceptable" is meant a material which is not biologically or otherwise undesirable, i.e., the material may be administered to an individual without causing any undesirable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained.
"Pharmaceutically acceptable salt" refers to a salt of a compound of the invention which is made with counterions understood in the art to be generally acceptable for pharmaceutical uses and which possesses the desired pharmacological activity of the parent compound. Such salts include: (1) acid addition salts, formed with inorganic acids such as hydrochloric acid,
hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like; or formed with organic acids such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, rumaric acid, tartaric acid, citric acid, benzoic acid, 3 -(4-hydroxybenzoyl) benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1 ,2-ethane-disulfonic acid,
2-hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid,
2-naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid,
4-methylbicyclo-oct-2-ene- 1 -carboxylic acid, glucoheptonic acid, 3 -pheny .propionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, bydroxynaphtfaoic acid, salicylic acid, stearic acid, muconic acid and the like; or (2) salts formed when an acidic proton present in the parent compound is replaced by a metal ion, e.g., an alkali metal ion, an alkaline earth ion or an aluminum ion; or coordinates with an organic base such as ethanolamine, diethanolamine, triethanolamine, N-methylglucamine, morpholine, piperidine, dimethylamine, diethylamine and the like. Also included are salts of amino acids such as arginates and the like, and salts of organic acids like glucurmic or galactunoric acids and the like {see, e.g., Berge etal., 1977, J. Pharm. Sci. 66:1-19).
[063] The terms "phosphate" and "phosphonate" as used herein refer to the moieties having the following structures, respectively:
Figure imgf000030_0001
[064] The terms "salts" and "hydrates" refers to the hydrated forms of the compound that would favorably affect the physical or pharmacokinetic properties of the compound, such as solubility, palatability, absorption, distribution, metabolism and excretion. Other factors, more practical in nature, which those skilled in the art may take into account in the selection include the cost of the raw materials, ease of crystallization, yield, stability, solubility, hygroscopicity, flowability and manufacturability of the resulting bulk drug.
sulfonamide as used herein contemplates a group having the structure
Figure imgf000030_0002
"sulfonate" as used herein contemplates a group having the structure
Figure imgf000030_0003
wherein R" is selected from the group consisting of hydrogen, Ci-Cio alkyl, Cj-
Cio alkenyl, C2-C10 alkynyl, Ci-C!0 aikanoyl, or C|-Cjo alkoxycarbonyl. [067 [ The term "sulfonyl" as used herein contemplates a group having the structure
Figure imgf000031_0001
"Substituted sulfonyl" as used herein contemplates a group having the structure
Figure imgf000031_0002
including, but not limited to alkylsulfonyl and arylsulfonyl.
[069] The term "thiocarbonyl," as used herein, means a carbonyl wherein an oxygen atom has been replaced with a sulfur.
[070[ Each R is independently selected from hydrogen, -OH, -CN, -N02, halogen, d to C)2 alkyl, C| to heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycle, aryl, heteroaryl, aralkyl, alkoxy, alkoxycarbonyl, alkanoyl, carbamoyl, substituted sulfonyl, sulfonate, sulfonamide, amino, and oxo.
[071] Each RN is independendy selected from the group consisting of hydrogen, -OH, Ci to Cu alkyl, Cj toC heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycle, aryl, heteroaryl, aralkyl, alkoxy, alkoxycarbonyl, alkanoyl, carbamoyl, substituted sulfonyl, sulfonate and sulfonamide. Two RN may be taken together with C, O, N or S to which they are attached to form a five to seven membered ring which may optionally contain a further heteroatom.
[072 [ The compounds of the present invention may be used to inhibit or reduce the activity of HCV, particularly HCV's NS5B protein. In these contexts, inhibition and reduction of activity of the NS5B protein refers to a lower level of the measured activity relative to a control experiment in which the cells or the subjects are not treated with the test compound. In particular aspects, the inhibition or reduction in the measured activity is at least a 10% reduction or inhibition. One of skill in the art will appreciate that reduction or inhibition of the measured activity of at least 20%, 50%, 75%, 90% or 100%, or any number in between, may be preferred for particular applications.
I. Compounds of the Invention
[073] In a first aspect of the invention is a compound that has the structure:
Figure imgf000032_0001
wherein,
L1, L2 and-N(S02R12)- together with the attached carbons of the aromatic ring to form a 5-12 member ring, containing 1-4 heteroatoms of N, O, S, P and/or Si;
L'or L2 is independently selected from the group consisting of a bond, -0-, -C(R15RI6 > -NR3-, - S(0)„-, -P(O)-, -SifR^5)-, -C(O)-, -C(0)0-, and substituted alkyl, alkenyl, alkoxy, cycloalkyl, cycloalkoxy, heterocycle, aryl, heteroaryl, amide, carbamate, urea, and sulfonamide;
is 0, 1, or 2; and
R1 is selected from hydrogen, faalide, -CF3, -CN, -C(0)H, -C(0)OR6, -C(0)NHR7, - 7, -C(=NOR')NHR7, -C(CF3)NHR8, -C(CN)NHR9, -S(0)2NHR10, -
Figure imgf000032_0002
R* is selected from hydrogen, -(CH2CH20)o^-Me, C2-24 alkyl optionally containing 0-6 heteroatoms of O, NRN and/or S, and 3-8 cycloalkyl optionally containing 0-2 heteroatom of O, NRN, and/or S;
R2 is an aryl or heteroaryl having one or more Ri7 substituents;
R3 is selected from the group of hydrogen, alkylcarbonyl, cycloalkylcarbonyl, alkoxylcarbonyl, cycloalkoxycarbonyl, alkylsulfonyl and cycloalkylsulfonyl;
R4 and R5 are independently methyl, ethyl, or cyclopropyl;
R12 is independently C1.3 alkyl, cyclopropyl, -OMe, or -NHMe;
R15, R16 is independently hydrogen, hydroxyl, azide, C2-4 alkenyl, C2-4alkynyl, C alkyl, cyclopropyl, CM alkoxy, or cyclopropoxy or R13 and R16 together are a carbonyl or C
alkenylidene or R15 and R16 joined together with the attached carbon are 3-6 member ring optionally containing 0-3 heteroatoms of O, NRN and/or S; and
R17 is F, Cl orCN. [074] The compound of this aspect may have an inhibitory activity with respect to HCV, as measured by the concentration of the compound effective to produce a half-maximal inhibition of HCVlb replication (EC50) in a lb_Huh-Luc Neo-ET cell line in culture, of lOO nM or less.
Figure imgf000033_0001
Ru is selected from hydrogen, -C(0)H, -C(0)Rin, -C(0)ORiv, -CiONR'R , alkyls, cycloalkyls, aryls, heteroaryls, and aralkyl;
Rv is selected from hydrogen, -NR'R", alkoxyls, alkyls, cycloalkyls, aryls, heteroaryls, and aralkyl;
Rw and R* are independrntly selected from hydrogen, -C(0)H, -C(0)RUi, -C(0)ORiv-, - C(0)NRvRvi, alkoxyls, alkyls, cycloalkyls, aryls, heteroaryls, and aralkyl; optionally, Rv and Rw together form a C4.8 member ring;
Ry and Rz are independently selected from hydrogen, alkoxyls, alkyls, cycloalkyls, aryls, heteroaryls, and aralkyl; optionally, Ry and Rx together form a 3-8 member ring;
R' and Ru are indenpendently selected from hydrogen, -Me, -Et, oPr-, c-Butyl;
R'" is selected from alkyoxyl, alkyls, cycloalkyls, aryls, heteroaryls, and aralkyl;
R1V is selected from alkyoxyl, alkyls, cycloalkyls, aryls, heteroaryls, and aralkyl; and
Rv and RVI are indenpendently selected from hydrogen, alkyoxyl, alkyls, cycloalkyls, aryls, heteroaryls, and aralkyl.
Figure imgf000034_0001
33
Figure imgf000035_0001

Figure imgf000036_0001

Figure imgf000037_0001
Figure imgf000038_0001
Figure imgf000039_0001
[077] The compound of this aspect may have the structure in which R120 is one of
Figure imgf000039_0002
Figure imgf000040_0001

Figure imgf000041_0001
40
Figure imgf000042_0001
41
Figure imgf000043_0001
42
Figure imgf000044_0001
Figure imgf000045_0001
R1 in mis aspect may be
[0811 R2 in this aspect may be a phenyl substituted with one or more R17 substituents.
[082] R2 in this aspect may be a 4-phenoxyphenyl and the phenoxy group is substituted with one or mor 17 substituents.
[083]
Figure imgf000045_0002
[084] The compound may have the particular sterochemical structure shown below:
Figure imgf000046_0001
(085 [ In one general embodiment, indicted as Group 1 herein, the compound has have the structural formula:
Figure imgf000046_0002
wherein:
selected from -H, -F, -CI, -Br, -I, and
Figure imgf000046_0003
Figure imgf000046_0004
Figure imgf000047_0001

Figure imgf000048_0001
[086] The above compound may have the same structural formula, wherein R' 1 is para-Έ, Ru-0- is as above, and X' is =0, and R'3 is -CH3.
[087] The compound in this group may be selected from the group consisting of B243, B244, B245, B246, B248, B249, B252, B253, B255, B256, B257, B258, B259, B260, B261, B262.B263, B264, B265, B266, B267, B268, B269, B270, B271, B272, B273, B274, B275, B276, B277, B278, B279, B280, B281, B282, B285, B286, B289, B300, B302, B306, B307, B308, B309, B310, B311, B314, B316, B317, B318, B319, B320, B333, B335, B336, B337, B338, B343, B344, B345, B346, B347, B348, B349, B350, B351, B352, B353, B354, B373, B378, B379, B381, B383, B384, B385, B386, B388, B389, B390, B391, B392, B394, B395, B397, B399, B400, B401, B402, B404, B405, and B409.
[088] In another geneal embodiment, indicated herein as Group 2, the compound has the structural formula:
Figure imgf000049_0001
wherein:
R'I is selected from -H, -F, -CI, -Br, -I,
Figure imgf000049_0002
H3C-S-:
v-S(0)„-, wherein n = 0, 1 , 2, is selected from n n / n
Figure imgf000049_0003
Figure imgf000050_0001

Figure imgf000051_0001
X' is selected
Figure imgf000051_0002
and «N-0(CH2)j-23CH3, and
R'3 is selected from -C¾ and -OCH3.
[090] The compound in Ms group may have the same structure where R'i is para-V, Rv-S(0)a-, are as above, X' is =0, and R'3 is -CH3.
[091[ Specific compounds in this group (Group 2) are selected from the group consisting of B283, B284, B287, B288, B290, B291, B293, B422, B423, B424, B426, B432, B443, B446, B447, B455, B456, B459, B463, B464, B466, B467, B470, B478, B479, B487, B488, B489, B491, B492, B493, B494, B495, B496, B497, B498, B499, B500, B501, B502, B503, B504, B505, B506, B507, B508, B509, B510, B511, B512, B513, B514, B515, B516, B517, B518, B519, B520, B521, B522, B523, B524, B525, B526, B527, B528, B529, B530, B531, B532, B533, B534, B535, B536, B537, B538, B539, B540, B541, B542, B543, B544, B545, B546, B547, B548, B549, B550, B551, B552, B553, B554, B555, B556, B557, B558, B561, B562, B563, B564, B565, B566, B567, B568, B569, B570, B571, B572, B573, B574, B575, B576, and B577.
[092] In another general embodiment, indicated herein as Group 3, the compound of the invention has the structural formula:
Figure imgf000052_0001
51
Figure imgf000053_0001
Figure imgf000054_0001
X' is selected from=0, =N-OH, =N-0(CH2CH20)o-8CH3, and =N-0(CH2)|.23CH3, and
R*3 is selected from -C¾ and -OCH3.
[094] The compound in this group may have the same structural formula wherein: R' 1 is para-V, RXR* - is selected as above, X' is =0, and R'3 is -CH¾.
[095] Specific compounds in this group (Group 3) are selected from the group consisting of B294, B295, B296, B297, B298, B299, B301, B303, B304, B305, B312, B313, B315, B321, B322, B323, B326, B327, B328, B329, B330, B334, B339, B340, B341, B342, B355, B356, B357, B358, B360, B361, B362, B363, B364, B365, B366, B367, B368, B369, B370, B371, B372, B374, B375, B376, B377, B380, B382, B387, B393, B396, B398, B403, B406, B407, B408, B410, B411, B412, B413, B414, B415, B416, B417, B418, B419, B420, B421, B425, B427, B428, B429, B430, B431 , B433, B434, B435, B436, B437, B438, B439, B440, B441, B442, B444, B448, B449, B450, B451, B452, B453, B454, B457, B458, B460, B46I, B462, B465, B468, B471, B472, B473, B474, B475, B476, B477, B480, B481, B482, B483, B484, B485, B486, and B490.
[096] The compounds above can be prepared in accordance with the synthetic schemes detailed below,
[097] In another aspect the compound of the invention has the structural formula:
S3 ,
Figure imgf000055_0001
wherein:
R'i is selected
Figure imgf000055_0002
R*2 is selected from -OH and -SOaMe; and R'3 is selected from -CH3 and -OCH3, as exemplified by B324, B325, B331, and B332. This compound can be prepared in accordance with Scheme 40 detailed below.
098] In still another aspect, the compound of the invention has the structural formula:
Figure imgf000055_0003
wherein:
Figure imgf000055_0004
R'2 is selected from -OH and -SOaMe; and R'j is selected from -CH3 and -OCH3, as exemplified by compound B359. This compound can be prepared in accordance with Scheme 39 detailed below.
II. Synthesis of Compounds of the Invention
]099] The compounds of the invention may be prepared by a variety of synthetic routes, samples of which are illustrated in the synthetic schemes outlined below. In general, the synthesis starts with constructing the central scaffolds such as benzofuran, benzothiophene, imidazopyridine or pyrazolopyridine by employing various synthetic techniques known to those skilled in the art. (e.g. in Heterocyclic Chemistry, J. A. Joule and K. Mills, J Wiley and Sons, 2010.) . Once the properly substituted cores are made, further functional group manipulations including but not limited to chain elongation, amidation, esterification, cyclization are performed as necessary to lead to the target molecules. When being allowed chemically and in some cases necessary, the central cores may preferably be introduced toward the end of the synthesis. Often, protection-deprotection and, in some cases, orthogonal protection-deprotection strategies are required to accomplish the desired transformation. More comprehensive descriptions of these synthetic methodologies and techniques can be in found in these and other references: Comprehensive Organic
Transformations, R. C. Larock Ed., Wiley-RCH, 1999. Protective Groups in Organic Synthesis, T. W. Greene and P. G. M. Wuts, 3ri ed. J Willey and Sons, 1999.
[0100] The following abbreviations are used throughout this application:
ACN Acetonitrile
AcOH Acetic acid
aq Aqueous
Boc terr-Butoxycarbonyl
Bu Butyl
Cbz Benzoxylcarbonoyl
Coned. Concentrated
GDI 1 , 1 '-Carbonyldiimidazole
Figure imgf000056_0001
DCM Dichloromethane
DffiA (DIPEA) Diisopropylethylamine
DMA MW-Dimethylacetamide
DMAP N^V-dimethyI-4-aminopyridine
DME 1 ,2-Dimethoxyethane
DMF ACiV-Dimethylformamide
DMSO Dimethylsulfoxide
dppf 1 , 1 '-Bis(diphenylphosphino)ferrocene
DCI l-Ethyl-3-[3-(dimethyIamino) propyl]carbodiimide hydrochloride ECso Effective concentration to produce 50% of the maximal effect
ESI Electrospray Ionization
EtjN, TEA Triethylamine
EtOAc, EtAc Ethyl acetate
EtOH Ethanol
g Gram(s)
h or hr Hour(s)
HATU 2-(7-Aza- lH-benzotriazole- 1 -yl 1 , 1 ,3.3-tetramethyluronium hexafluorophosphate
Hex Hexanes
HOBt 1-Hydroxybenzotriazole
HPLC High performance liquid chromatography
IC50 The concentration of an inhibitor that causes a 50 % reduction in a measured activity
LC-MS Liquid Chromatography-Mass Spectrometry
μΜ Micromolar(s)
Mel Methyl Iodide
MeOH Methanol
min Minute(s)
mM Millimolar(s)
mmol Millimole(s)
MoNP 2-Methoxy-2-(l-naphthyl)propionic acid
Ms Mesyl, Methylsulfonyl, Methanesulfonyl
MSH 0-(Mesitylsulfonyl)hydroxyamine
mw Microwave
NBS W-Bromosuccinimide
NIS iV-Iodosuccinimide
nM Nanomoiar(s)
NMO V-MethyIraorpholine-iV-oxide
NMP iV-Methylpyrrolidinone NMR Nuclear magnetic resonance
PE Petroleum ether
PPA Polyphosphoric Acid
PPh3 Triphenylphosphine
PPTs Pyridinium / oluenesulfonate
Py. Pyr Pyridine
rt Room temperature
Sat. Saturated
TBAF Tetra-n-butylammonium fluoride
TEA Triethylamine
TfOH Trifluoromethanesulfonic acid
TFA Trifluoroacetic acid
THF Tetrahydrofuran
TLC Thin Layer Chromatography
TMSOTf Trimethylsilyl trifluoromethanesulfonate
tR Retention time
Ts Tosyl, Methylphenylsulfonyl
TsOH Tosylic acid, -Methylphenylsulfonic acid
w/w Weight/weight
v/v Volume volume
[0101] Reagents and solvents used below can be obtained from commercial sources such as Aldrich Chemical Co. (Milwaukee, Wisconsin, USA). Ή NMR spectra were recorded on a Broker 400 MHz or 500 MHz NMR spectrometer. Significant peaks are tabulated in the order: multiplicity (s, singlet; d, doublet; t, triplet; q, quartet; m, multiplet; br s, broad singlet), coupling constants) in Hertz (Hz) and number of protons. Electrospray spray ionization (ESI) mass spectrometry analysis was conducted on a Hewlett-Packard 1100 MSD electrospray mass spectrometer using the HP 1 100 HPLC for sample delivery. Mass spectrometry results are reported as the ratio of mass over charge, followed by the relative abundance of each ion (in parentheses) or a single m z value for the M + H (or, as noted, M - H) ion containing the most common atomic isotopes. Isotope patterns correspond to the expected formula in all cases. Normally the analyte was dissolved in methanol at 0.1 mg mL and 5 microliter was infused with the delivery solvent into the mass spectrometer, which scanned from 100 to 1500 daltons. All compounds could be analyzed in the positive ESI mode, using an acetonitrile/water gradient (10% ~ 90%) acetonitrile in water with 0.1% formic acid as delivery solvent. The compounds provided below could also be analyzed in the negative ESI mode, using 2 mM N¾OAc in
acetonitrile/water as delivery solvent. Enantiomeric purity was determined using a Hewlett- Packard Series 1050 system equipped with a chiral HLPC column (ChiraiPak AD, 4.6 mm x 150 mm) and isocratic etution using isopropanol/hexane = 5/95 (v/v) as mobile phase.
Figure imgf000060_0001

Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
These compounds may be synthesized in accordance with the synthetic schemes disclosed below.
In another aspect, the compound of the invention includes a compound having the structural formula:
Figure imgf000069_0002
wherein:
R'l is selected from para-
Figure imgf000069_0003
R* 2 is selected from -OH and -S02Me; and R'3 is selected from -CH^ and -OCH3, as exemplified by B324, B325, B331, and B332. This compound can be synthesized in accordance with the scheme 40 below. 0102] In another aspect, the compound of the invention has the structural formula:
Figure imgf000070_0001
wherein:
R' I is selected from para-Έ
Figure imgf000070_0002
;
R'2 is selected from -OH and -S02Me; and R'3 is selected from -CH3 and -OCH3, as exemplified by compound B359.
This compound can be synthesized in accordance with Scheme 39 below.
(0118] The following examples illustrate the preparation and antiviral evaluation of compounds within the scope of the invention. These examples and preparations which follow are provided to enable those skilled in the art to more clearly understand and to practice the present invention. They should not be considered as limiting the scope of the invention, but merely as being illustrative and representative thereof.
Scheme 4
Figure imgf000070_0003
[0119] Step 1. Refer to Scheme 4. To a solution of compound 3-1 (prepared by following the procedures described in WO200759421 with some modifications) (4.00 g, 10.3 mmol) in CH2C12 (30 mL) was added BC13 (I N in CH2C12, 20.6 mmol) at 0 °C. After stirring at rt for 1 hr, the reaction mixture was added ice water (100 mL). The mixture was extracted with CH2C12 (800 mL x 2) and the combined organic extracts were washed with water and brine and dried with anhydrous Na2SO_}. The solvent was removed and the residue was dried in vacuo to give crude compound 4-1 (3.4 g, 96% yield) as a yellow solid. LC-MS (ESI): mlz 346 [M + H]+.
10120] Step 2. To a solution of compound 4-1 (3.4 g, 9.8 mmol) in CH2C12 (100 mL) were added DMAP (120 mg, 0.980 mmol) and D1EA (1.52 g, 11.8 mmol), followed TfzO (3.20 g, 1 1.3 mmol) at 0 °C. After stirring at 0 °C for 2 hrs, the reaction mixture was added ice water (100 mL). The organic layer was separated, washed with water and brine, and dried with anhydrous Na2S04. The solvent was removed and the residue was dried in vacuo to give crude compound 4-2 (4.6 g, quantitative yield) as a yellow solid. LC-MS (ESI): mlz 478 [M + H]+.
10121 [ Step 3. To a solution of 4-2 (2.0 g, 4.2 mmol) in 20 mL DMF was added 4-3 (0.44 g, 6.3 mmol), Cul (0.16 g, 0.84 mmol), Pd(PPh3)2Cl2 (0.29 g, 0.42 mmol) and Et3N (20 mL). The resulting mixture was flushed with Ar, stirred at rt for 1 hr and poured into ice water ( 100 mL). The mixture was extracted with EtOAc (50 mL x 5) and the combined organic extracts were washed with water and dried with anhydrous Na2S04. The solvent was removed and the residue was purified by silica gel column chromatography (Petroleum ether/acetone = 4/1 (v/v) to 3/2 (v/v)) to give compound 4-4 (1.10 g, 69% yield) as a gray solid. LC-MS (ESI): mlz 398 [M + H]+. 10122] Step 4.To a solution of compound 4-4 (2.00 g, 5.03 mmol) in EtOAc (150 mL) was added 10% Pd C (2.0 g). The resulting mixture was flushed with H2 and stirred at rt for 1.5 hrs.
Subsequently, the reaction mixture was filtered through Celite®545 and the filtrate was concentrated and dried in vacuo to give compound 4-5 (1.8 g, 97% yield). LC-MS (ESI): mlz 372 [M+H ; Ή NMR (500 MHz, CDC13): δ 7.98 - 8.01 (m, 2H), 7.70 (s, 1H), 7.12 - 7.16 (m, 2H), 6.82 (s, 1H), 4.39 (dd, J, = 14.5 Hz, J2 = 7 Hz, 2H), 3.73 (t, J= 6 Hz, 3H), 2.66 (t, J= 7.5 Hz, 2H), 1.69 - 1.80 (m, 4H), 1.40 (t, J= 7 Hz, 3H) ppm.
10123] Step 5. To a solution of compound 4-5 (1.80 g, 4.85 mmol) in CH2C12 (50 mL) was added DMAP (6 mg) and anhydrous pyridine (3.07 g, 38.8 mmol), followed by MsCl (1.60 g, 14.5 mmol) at 0 °C. After stirring at rt for 2 hrs, the reaction mixture was added ice water (50 mL). The organic layer was separated, washed with water and brine and dried with anhydrous NajSO-j, The solvent was removed and the residue was purified by silica gel column chromatography (Petroleum ether/acetone = 5/1 (v/v)) to give compound 4-6 (1.4 g, 55% yield) as a yellow solid. LC-MS (ESI): mlz 449 [M - Ms + H]+; lH NMR (500 MHz, CDC13): δ 8.02 - 8.05(m, 2H), 7.90 (s, l H), 7.71 (s, 1H), 7.16 - 7.19 (m, 2H), 6.61 (s, 1H), 4.42 (dd, Js= 14 Hz, J2 = 7.0 Hz, 2H), 4.34 (t, J= 5.5 Hz, 2H), 3.04 - 3.08 (m, 6H), 2.83 (t, J= 8.0 Hz, 2H), 1.81 - 1.92 (m, 4H), 1.41 (t, J- 7.0 Hz, 3H) ppm.
[0124] Step 6. To a suspension of NaH (0.21 g, 60% in mineral oil, 5.31 mmol) in anhydrous THF (160 mL) was added a solution of compound 4-6 (1.40 g, 2.65 mmol) in anhydrous THF (40 mL) at 0 °C. After stirring at rt for 2 hrs, the reaction mixture was added sat. aq. NH4CI (10 mL). The resulting mixture was concentrated and the residue was diluted with EtOAc (100 mL). The mixture was washed with water and brine and dried with anhydrous NaiSC The solvent was removed and the residue was purified by silica gel column chromatography (Petroleum
ether/acetone = 5/ 1 (v/v)) to give compound 4-7 (1.1 g, 96% yield) as a yellow solid. LC-MS (ESI): mlz 432 [M + H]+; H NMR (500 MHz, CDCI3): δ 8.02 - 8.06 (m, 2H), 7.90 (s, 1H), 7.60 (s, 1H), 7.15 - 7.20 (m, 2H), 4.42 (dd,J/ = 14 Hz, J2 = 6.5 Hz, 2H), 3.69 (t, J= 6.0 Hz, 2H), 3.07 (s, 3H), 3.02 (t, J = 6.0 Hz, 2H), 1.94 (dd, J, = 11 Hz, J2 = 5.5 Hz, 2H), 1.77 (br s, 2H), 1.40 - 1.43 (m, 3H) ppm.
10125] Step 7. To a solution of compound 4-7 (50 mg, 0.12 mmol) in MeOH THF (2 mL/4 mL) was added LiOH (2.0 N, 0.46 mmol). The resulting mixture was stirred at 70 °C for 2 hrs, cooled to rt and acidified with 1 N aq. HC1 (5 mL). Subsequently, the suspension was filtered and the solid was washed with waster and dried in vacuo to give crude compound 4-8 (46 mg, 95% yield) as a white solid, which was used directly for the next step without further purification. LC-MS (ESI): mlz 404 [M + H]+.
|0126] Step 8. To a solution of compound 4-8 (46 mg, 0.12 mmol) in DMF (2 mL) was added HATU (54 mg, 0.14 mmol). The resulting mixture was stirred at rt for 30 min and added D1EA (154 mg, 1.20 mmol) and MeNH2 HC1 (41 mg, 0.60 mmol). After stirring at rt for 20 min, the reaction mixture was poured into ice water (50 mL). The suspension was filtered and the solid was purified by silica gel column chromatography (Petroleum ether/acetone = 3/1 (v/v)) to give compound 4-9 (30 mg, 61% yield) as a white solid. LC-MS (ESI): mlz M [M + H]+; 1H NMR (500 MHz, CDCI3): 5 7.89 - 7.92 (m, 2H), 7.68 (s, 1H), 7.59 (s, 1 H), 7.19 (t, J= 9.0 Hz, 2H), 5.80 (d, J= 4.0 Hz, 1H), 3.69 (<L J= 6.0 Hz, 2H), 3.06 (s, 3H), 2.98-3.03 (m, 5H), 1.93 (dd, J/= 11 Hz, J2 = 5.5 Hz, 2H), 1.75 (d, J= 2.5 Hz, 2H) ppm.
|0127] Synthesis of compound 4-10. A mixture of compound 4-8 (50 mg, 0.12 mmol) in SOCl2 (1.5 mL) was stirred at 80 °C for 2 hrs. The solvent was removed and the residue dried in vacuo to give the crude acid chloride, which was used for the next step without further purification. Subsequently, the crude acid chloride was dissolved in anhydrous pyridine (1.5 mL), followed by 0-methylhydroxylamine hydrochloride ( 124 mg, 0.490 mmol). After stirring at 100 °C for 1 5 hrs, the reaction mixture was concentrated and the residue was purified by preparative HPLC to give compound 4-10 (20 mg, 37% yield) as a white powder. LC-MS (ESI): mlz 433 [M + H ; Ή NMR (500 MHz, CDCI3): δ 8.31 (s, 1H), 7.90 - 7.93 (m, 2H), 7.66 (s, 1 H), 7.20 (t, J= 8.5 Hz, 2H), 3.85 (s, 3H), 3.03 (s, 3H), 2.97 - 3.00 (m, 2H), 1.93 - 1.96 (m, 2H), 1.69 - 1.76 (m, 2H) ppm.
Figure imgf000073_0001
[0128] Step 1. Refer to Scheme 8. To a stirred solution of compound 4-2 (9.00 g, 18.9 mmol) in DMF (100 mL) were added Et3N (7.84 mL, 56.6 mmol), Pd(OAc)2 (212 mg, 0.94 mmol), dppp (469 mg, 1.13 mmol) and butyl vinyl ether (12.1 mL, 94.4 mmol) under an atmosphere of Ar. After stirring at 100 °C for 2 hrs, the reaction mixture was concentrated. The residue was diluted with EtOAc (250 mL) and the resulting mixture was washed with water (100 mL x 3) and dried with anhydrous Na2SC>4. The solvent was removed and the residue was purified by silica gel column chromatography (Petroleum ether EtOAc = 16/1 (v/v)) to give compound 8-1 (3.9 g, 48% yield) as a yellow solid. LC-MS (ESI): mlz 427 [M + H)+.
[0129] Step 2. A solution of compound 8-1 (3.90 g, 9.13 mmol) in THF (60 mL) was added 1 N aq. HC1 (10 mL) at rt. After stirring at rt for 15 min, the reaction mixture was concentrated and the residue was diluted with DCM (100 mL). The resulting mixture was washed with brine and dried with anhydrous Na2S04. The solvent was removed and the residue was dried in vacuo to give crude compound 8-2 (3.27 g, 96% yield) as a yellow solid, which was used for the next step without further purification. LC-MS (ESI): mlz 372 [M + H]+.
[0130] Step 3. To a stirred solution of compound 8-2 (2.00 g, 5.38 mmol) in EtOAc (50 mL) was added SnCl22H20 (3.47 g, 16.2 mmol). After stirring at 80 °C for 1 hr, the reaction mixture was added sat. aq. NaHC03 (50 mL) and the resulting mixture was stirred at rt for 30 min.
Subsequently, the mixture was filtered through Celite®545 and the the filtered cake was washed with EtOAc (50 mL x 3). The organic layer of the filtrate was washed with brine and dried with anhydrous Na2S04. The solvent was removed and the residue was dried in vacuo to give crude compound 8-3 (1.8 g, 98% yield) as a brown solid, which was used for the next step without further purification. LC-MS (ESI): mlz 342 [M + H]+.
10131] Step 4. To a stirred solution of compound 8-3 (900 mg, 2.64 mmol) in anhydrous pyridine (15 mL) was added MsCl (0.25 mL, 3.17 mmol) at 0 °C. After stirring at rt for 1 hr, the reaction mixture was diluted with EtOAc (100 mL) and the resulting mixture was washed with 2 N aq. HC1 (20 mL x 2) and H20 (50 mL x 3) and dried with anhydrous Na2S04. The solvent was removed and the residue was purified by silica gel column chromatography (Petroleum
ether DCM/EtOAc = 8/4/1 (v/v)) to compound 8-4 (520 mg, 47% yield) as a yellow solid. LC-MS (ESI): m/2442 [M + Naf.
[0132] Step 5. To a solution of compound 8-4 (380 mg, 0.91 mmol) in MeOH (10 mL) and THF (10 mL) was added NaBH4( 172 mg, 4.54 mmol) in several portions at 0 °C. After stirring at 0 °C for 15 min, the reaction was quenched by adding acetone (1 mL). The mixture was concentrated and the residue was diluted with EtOAc (100 mL). The resulting mixture was washed with 2 N aq. HC1 (20 mL) and H20 (50 mL x 3) and dried with anhydrous Na2S04. The solvent was removed
13 and the residue was dried in vacuo to give crude compound 8-5 (240 mg, 63% yield), which was used for the next step without further purification. LC-MS (ESI): mlz 444 [M + Na]+.
[0133J Step 6. To a stirring solution of compound 8-5 (50 mg, 0.12 mmol) in THF (15 mL) was added NaH (24 mg, 0.6 mmol) at 0 °C under an atmosphere of Ar. After stirring at rt for 15 min, the mixture was added compound 8-6 (106 mg, 0.24 mmol) (prepared following the procedure described in Angew. Chem. Intl. Ed. 2008, 47, 3784) at 0 °C and the resulting mixture was stirred at 0 °C for 3 hrs and rt overnight. Subsequently, saturated aq. NH4CI (10 mL) was added to quench the reaction and the mixture was concentrated. The residue was diluted with EtOAc (50 mL) and the mixture was washed with brine ( 10 mL) and dried with anhydrous Na2SC«4. The solvent was removed and the residue was purified by silica gel column chromatography
(Petroleum ether/Acetone = 4/1 (v/v)) to give compound 8-7 (30 mg, 56% yield) as a white solid. LC-MS (ESI): mlz 448 [M + H]+.
[0134] Step 7. To a solution of compound 8-7 (40 mg, 0.09 mmol) in MeOH THF (2 mL/4 mL) was added 2.0 N aq. LiOH (0.18 mmol, 0.36 mmol). After stirring at 75 °C for 3 hrs, the reaction mixture was cooled to 0 °C and acidified with 2N aq. HC1 adjust pH value to 5~6. Subsequently, the suspension was filtered and the solid was washed with water and dried in vacuo to give compound 8-8 (38 mg, 97% yield) as a white solid, which was used for the next step without further purification. LC-MS (ESI): mlz 442 [M + Na .
[0135] Step 8. To a solution of compound 8-8 (40 mg, 0.10 mmol) in DMF (3 mL) was added HATU (43 mg, 0.12 mmol). The resulting mixture was stirred at rt for 60 min and DIE A (0.16 mL, 0.95 mmol) and MeNH2 HCl (20 mg, 0.29 mmol) were added. After stirring at rt for 15 min, the reaction mixture was added into water (30 mL). The suspension was filtered and the solid was washed with water and dried in vacuo. The residue was dissolved in DCM (1.5 mL) and the solution was added into hexane (40 mL). The resulting suspension was filtered and the solid was dried in vacuo to give compound 8-9 (23 mg, 56% yield). LC-MS (ESI): mlz 433 [M+H]+; Ή NMR (500 MHz, CDCI3): δ 7.88 - 7.91 (m, 3H), 7.62 (s, 1 H), 7.20 (t,J= 8.5 Hz, 2H), 5.80 (br s, 1 H), 4.96 (q, J= 6.5 Hz, 1H), 4.15 - 4.18 (m, 1 H), 4.02 - 4.09 (m, 2H), 3.29 - 3.34 (m, 1H), 3.15 (s, 3H), 3.01 (d, J= 5.0 Hz, 3H), 1.74 (d, J= 6.5 Hz, 3H) ppm. Compound 8-9 was separated into a pair of enantiomers: enantiomer 8-9 A (tR = 3.34 min) and enantiomer 8-9_B (.R = 3.89 min) detected by UV absorption at 214 run on a Daicel CHIRALPAK AS-H column (eluent:
MeOH/liquid CO2 = 10/90 (v V), flow rate: 60 g/min and back pressure: 100 bar).
Figure imgf000076_0001
[0136] Step 1. Refer to Scheme 15. To a solution of NaH (80 g, 60% mineral oil dispersion, 2 mol) in toluene (1.2 L) was added diethyl carbonate (295 g, 2.50 mol) at 0 °C. After stirring at rt for 2 hrs, the mixture was added drop wise to a solution of compound 15-1 (99 g, 0.50 mol) in toluene (400 mL) at reflux. After refluxing overnight, the reaction mixture was cooled to rt and sequentially treated with HOAc (140 mL) and aq. HC1 (2 M, 864 mL). The resulting mixture was extracted with EtOAc (400 mL x 3) and the combined organic extracts were washed with water (500 mL x 4) and brine (200 mL x 2) and dried with anhydrous Na2SO.i. The solvent was removed and the residue was dried in vacuo to give compound 15-2 (122 g, 90% yield) as an oil. LC-MS (ESI): m/z 271.0 [M + H]'
[0137 J Step 2. To a solution of compound 15-2 (100 g, 369 mmol) in DMF (70 mL) was added /?-benzoquinone (40 g, 369 mmol), followed by ZnCl2 (50 g, 369 mmol) in portions at rt. After stirring at 105 °C for 3.5 hrs, the reaction mixture was partitioned between water (800 mL) and EtOAc (800 mL) and filtered. The aqueous phase was extracted with EtOAc (500 mL x 2). The combined organic extracts were washed with water (1000 mL x 2) and dried with anhydrous Na2S04. The solvent was removed and the residue was purified by silica gel column
chromatography (Petroleum ether/Acetone = 10/l(v/v)) to give compound 15-3 (26 g, 20% yield) as a yellow solid. LC-MS (ESI): m/z 361.0 [M + H]+.
[0138] Step 3. To a solution of compound 15-3 (26 g, 72 mmol) in NMP (200 mL) was added Cs2C03 (47.0 g, 144 mmol). After stirring at rt for 20 min, 2-bromopropane (20.0 ml, 216 mmol) was added. The resulting mixture was stirred at 80 °C for 4 hrs, then diluted with ammonia and agitated for 30 min. The mixture was diluted with water (200 mL) and the aqueous phase was extracted with EtOAc (150 mL x 3). The combined organic extracts were washed with water (200 mL x 3) and dried with anhydrous Na2S04. The solvent was removed and the residue was dried in vacuo to give compound 15-4 (27.5 g, 95% yield) as a colorless oil. LC-MS (ESI): m/z 403.0 [M
|0139[ Step 4. To a solution of HN03 (cone. 66 mL, 0.89 mol) and CH2C12 (300 mL) a solution of compound 15-4 was added drop wise (27.5 g, 68.2 mmol) in CH2C12 (120 mL) at 0 °C over 1 nr. After stirring at rt for 30 min, the reaction mixture was diluted with water (200 mL) and extracted with DCM (150 mL x 3). The combined organic extracts were washed with water (200 mL x 3) and dried with anhydrous Na2S0 . The solvent was removed and the residue was re- crystallized in methyl /-butyl ether (MTBE) to give compound 15-5 (24.6 g, 80% yield) as a pale yellow solid. LC-MS (ESI): m/z 448.0 [M + H]+; 1H NMR (500 MHz, CDC13): δ 7.92 (d, J= 8.5 Hz, 2H), 7.87 (m, 2H), 7.76 (s, 1H), 7.65 (d, J = 8.5 Hz, 2H), 4.69 - 4.74 (m, 1H), 4.40 - 4.44 (m, 2H), 1.54 (s, 6H), 1.41 - 1.45 (t, 3H) ppm.
|0140] Step 5. A mixture of compound 15-5 (5.0 g, 1 1.2 mmol), 4-fluorophenol (1.7 g, 14.5 mmol), Pd(OAc)2 (250 mg, 1.12 mmol), -BuXphose (380 mg, 0.9 mmol) and K3PO4 (4.8 g, 22.4 mmol) in toluene (50 mL) was stirred at 100°C under an atmosphere of Ar and monitored by LC- MS. After 2 hrs, the reaction mixture was concentrated and the residue was diluted with water (100 mL). The mixture was extracted with EtOAc ( 100 mL x 3) and the combined organic extracts were washed with water (100 mL x 2) and dried with anhydrous Na2S0 . The solvent was removed and the residue was dried in vacuo to give crude compound 15-6 (4.8 g, 90% yield) as a yellow powder. LC-MS (ESI): m/z 480.1 [M + H]+. [0141| Step 6. To a solution of compound 15-6 (2.0 g, 4.2 mmol) in DCM (30 mL) drop wise was added BCU (8.4 mL, 8.4 mmol) at -78 °C. After stirring at -40 °C for 1 hr, the reaction was quenched by adding sat. aq. NH4CI (20 mL). The resulting mixture was extracted with DCM (50 mL x 2) and the combined organic extracts were washed with water (50 mL x 3) and brine (50 mL) and dried with anhydrous Na2S04. The solvent was removed and the residue was dried in vacuo to give crude compound 15-7 (1.6 g, 90% yield) as a yellow solid. LC-MS (ESI): /z 438.1 [M + H]+.
|0142[ Step 7. To a solution of compound 15-7 (1.6 g, 3.9 mmol) and DMAP (24 mg, 0.2 mmol) in DCM (30 mL) at 0 °C was added Et3N (790 mg, 7.8 mmol), followed by Tf20 (1.6 g, 5.82 mmol). After stirring at rt for 1 hr, LC-MS analysis indicated that the reaction went completion. The mixture was diluted with DCM (100 mL), washed water (50 mL x 3) and brine (50 mL) and dried with anhydrous Na2S04. The solvent was removed and the residue was dried in vacuo to give crude compound 15-8 (1.8 g, 94% yield) as a yellow solid. LC-MS (ESI): m/z 570.0 [M +
H]+.
[01431 Step 8. To a solution of compound 15-8 (1.8 g, 3.2 mmol) in CH3CN (50 mL) was added NaOAc (1.6 g, 16 mmol.), dppf (180 mg, 0.32 mmol), and Pd(OAc)2 (150 mg, 0.64 mmol), and the resulting mixture was saturated with Ar. After l-(vinyloxy)butane (1.6 g, 16 mmol) was added, the mixture was stirred at 100 °C for 2 hrs under an atmosphere of Ar. Subsequently, the reaction mixture was cooled to rt, concentrated, and diluted with EtOAc (100 mL). The resulting mixture was washed with water (50 mL x 2) and brine (100 mL) and dried with anhydrous Na2S04. The solvent was removed and the residue was dissolved in THF (50 mL) and aq. HC1 (2 N, 12 mL). After refluxing for 1 hr, the mixture was cooled to rt and concentrated to remove most of the organic solvent. The resulting mixture was extracted with EtOAc (50 mL x 2). The combined organic extracts were washed with water (50 mL x 3) and brine (50 mL) and dried with anhydrous Na2S04. The solvent was removed and the residue was dried in vacuo to give crude compound 15-9 (1.4 g, 98% yield). LC-MS (ESI): m/z 464.1 [M + Hf.
[0144[ Step 9. To a solution of compound 15-9 (1.4 g, 3.4 mmol) in EtOAc (50 mL) was added SnCl22H20 (2.8 g, 13.6 mmol) at rt and the resulting mixture was stirred at 80 °C for 1 hr. The mixture was cooled to rt and d its pH value was adjusted to 8 ~ 9 by adding saturated aq.
NaHC03. The mixture was filtered and the filtrate was extracted with EtOAc (50 mL x 2). The combined organic extracts were washed with water (50 mL x 3) and brine (50 mL) and dried with anhydrous Na2S04. The solvent was removed and the residue was dried in vacuo to give crude compound 15-10 (1.1 g, 85% yield) as a yellow solid. LC-MS (ESI): mlz 434.1 [M + H]+.
[0145] Step 10. To a solution of compound 15-10 (1.1 g, 2.5 mmol) in anhydrous pyridine (20 mL) was added MsCl ( 1.8 mL) at 0 °C. After the mixture was stirred at 30 °C for 2 hrs, LC-MS analysis indicated that the reaction went to completion. The mixture was diluted with water (100 mL) and EtOAc (50 mL). The aqueous phase was extracted with EtOAc (50 mL x 3). The combined organic extracts were washed with sat. aq. NH4C1 (50 mL x 3) and brine (50 mL) and dried with anhydrous Na2S04. The solvent was removed and the residue was dried in vacuo to give crude compound 15-11 (1.1 g, 90% yield) as a yellow solid. LC-MS (ESI): mlz 512.1 [M + H]+.
[0146] Step 11. To a solution of compound 15-11 (1.1 g, 2.1 mmol) in THF (30 mL) was added aBH4 (560 mg, 14.7 mmol) in portions at 0 °C. After stirring at 0 °C for 30 min, LC-MS analysis indicated that the reaction went to completion and acetone (2 mL) was added to quench excess amount of NaB¾. The mixture was concentrated and the residue was diluted with EtOAc (100 mL). The resulting mixture was washed with water (50 mL x 3) and brine (50 mL) and dried with anhydrous Na2SO.t. The solvent was removed and the residue was dried in vacuo to give compound 15-12 (0.9 g, 90% yield) as a yellow solid. LC-MS (ESI): mlz 514.1 [M + H]+.
[0147] Step 12. To a solution of compound 15-12 (0.9g, 1.7 mmol) in anhydrous DCM (30 mL) was added NaH (60% in paraoil, 200 mg, 5 mmol) at 0 °C, followed by compound 8-6 ( 1.1 g, 2.55 mmol). After stirring at 0 °C for 3 hrs and at it for 12 hrs, the reaction was quenched by adding saturated aq. NH4C1 (10 mL). The resulting mixture was extracted with DCM (30 mL x 3) and the combined organic extracts were washed with brine (50 mL) and dried with anhydrous Na2S04. The solvent was removed and the residue was purified by silica gel column
chromatography (Petroleum ether/EtOAc = 67l(v/v)) to give compound 15-13 (520 mg, 55% yield) as a white solid. LC-MS (ESI): mlz 540.1 [M + H]+.
[0148] Step 13. To a solution of compound 15-13 (520 mg, 0.96 mmol) in MeOH/THF (4 mL/8 mL) was added aq. LiOH (2.0 M, 2 mL) at rt. After stirring at 80 °C for 12 hrs, the reaction mixture was cooled to rt and adjusted its pH value to 2 ~ 3 by adding aq. HC1 (2.0 M). The organic solvent was removed and the residue was diluted with EtOAc (50 mL). The organic layer was isolated, washed with brine (25 mL) and dried with anhydrous Na2SC>4. The solvent was removed and the residue was dried in vacuo to give crude compound 15-14 (442 mg, 90% yield) as a white solid. LC-MS: (ESI): mlz 512.1 [M + H]+.
[0149] Step 14. Compound 15-14 (442 mg, 0.86 mmol) was dissolved in DMF (5 mL), followed by addition of HATU (450 mg, 1.17 mmol). After stirring at rt for I hr, the reaction mixture was added DIPEA (503 mg, 3.9 mmol) and MeNH2 HCl (157 mg, 2.34 mmol). The resulting mixture was stirred at rt for another 1 hr before being concentrated The residue was diluted with water (25 mL) and EtOAc (50 mL). The organic layer was isolated, washed with brine (25 mL), and dried with anhydrous Na2S0 . The solvent was removed and the residue was purified by silica gel column chromatography (Petroleum ether/EtOAc = 5/1 (v V)) to give compound 15-15 (360 mg, 80% yield) as a white solid. LC-MS (ESI): mlz 525.1 [M + Hf ; Ή NMR (500 MHz, CDC13): δ 7.89 (s, I H), 7.82 - 7.84 (m, 2H), 7.26 (s, I H), 7.04 - 7.12 (m, 6H), 5.85 (m, lH), 4.94 - 4.98 (m, I H), 4.14 - 4.18 (m, 1 H), 4.02 - 4.06 (m, 2H), 3.31 (m, 1 H), 3.15 (d, J = 8.5 Hz, 3H), 3.00 (<L J = 5.0 Hz, 3H), 2.07 - 1.73 (d, J = 6.5 Hz, 3H) ppm. Compound 15-15 was separated into a pair of enantiomers: enantiomer 15-15a (tR = 3.66 min) and enantiomer 15-15b (fe = 4.25 min) detected by UV absorption at 214 nm on a 4.6 mm x 250 mm x 5 μιη Daicel CHIRALPAK AS-H column (column temperature: 39.7 °C; eluent: MeOH/liquid CO2 = 20/80 (vV); CO2 flow rate: 2.4 g min and co-solvent flow rate: 0.6 g min; front pressure: 198 bar and back pressure: 150 bar).
Scheme 28
Figure imgf000081_0001
[0150] Synthesis of compound 28-3a. Refer to Scheme 28. Following the same procedure as that for the praparation of compound 27-3a from 15-11 described in Scheme 27 and replacing 3- bromo-2-methylpropene with allyl bromide, compound 28-3a was obtained as a white solid. LC- MS (ESI): m/z 732.1 [M + Naf.
[0151] Synthesis of compound 28-5a. Following the same procedure as that for the praparation of compound 27-5a from 27-3 a described in Scheme 27 and replacing compound 27-3a with 28- 3a, compound 28-5a was obtained as a white solid. LC-MS (ESI): m/z 539.2 [M + H]+; Ή NMR (500 MHz, CDCb): δ 7.89 and 7.78 (s, s, lH), 7.83 (dd, J, = 6.5 Hz, J2 = 8.5 Hz, 2H), 7.61 and 7.58 (br s, s, 1 H), 7.04 - 7.12 (m, 6H), 5.83 (d, J = 4.0 Hz, IH), 5.18 and 4.98 (dd, dd, J/ = 13.5 Hz, J2 = 6.5 Hz, 1H), 4.13 - 4.16 and 3.78 - 3.81 (m, m, 2H), 3.15 and 3.12 (s, s, 3H), 3.12 and 2.85 (m, m, 1 H), 3.00 (d, J = 5.0 Hz, 3H), 1.73 (t, J= 6.5 Hz, 3H), 1.23 and 1.17 (d, d, J= 6.5 Hz, 3H) ppm. Alternatively, compound 28-5a can be obtained using compound 28-7a as the starting material described in Scheme 28.
[0152] Synthesis of compound 28-5b. Following the same procedure as that for the preparation of compound 28-5a described in Scheme 27 and replacing compound 27-3a with 28-3b,
compound 28-5b was obtained as a white soUd. LC-MS (ESI): m/z 447.1 [M + H ; Ή NMR (500 MHz, CDCb): δ 7.89 - 7.92 (m, 2H), 7.87 (s, 1H), 7.59 (s, 1H), 7.20 (t, J= 8.5 Hz, 2H),
5.78 (m, 1H), 4.98 (m, 1H), 4.14 (m, 2H), 3.15 and 3.12 (s, s, 3H), 3.01 (d, J = 4.5 Hz, 3H), 2.83 (m, 1 H), 1.73 and 1.71 (d, d, J = 7.0 Hz, 3H), 1.23 and 1.18 (d, d, J = 6.5 Hz, 3H) ppm.
Alternatively, compound 28-5b can be obtained using compound 28-7b as the starting material described in Scheme 28.
[0153] Synthesis of compound 8-6a. To a solution of compound 28-3a (2.1 g, 3.0 mmol) in DCM (200 mL) was added m-CPBA (563 mg, 3.3 mmol) at 0 °C. After stirring at 0°C for 30 min, the reaction mixture was washed with saturated aq. NaHCCb solution and water. The organic layer was dried over anhydrous Na2S04 and concentrated to dryness to give crude compound 28- 6a as a red solid, which was used in the next step without further purification. LC-MS (ESI): m/z 748.1 [M + Naf.
[0154] Synthesis of compound 28-7a. Compound 28-6a (375 mg, 0.52 mmol) was dissolved in dry toluene (300 mL) and the resulting solution was added DBU (4.2 mL, 28.1 mmol) at 0 °C. After stirring at 100 °C for 45 min under an atmosphere of N2, the reaction mixture was concentrated and the residue was purified by silica gel column chromatography (PE/EtOAc = 5/1 to 3/1 (v/v)) to give compound 28-7a (248 mg, 87% yield) as an off white solid. LC-MS (ESI): m/z 552.1 [M + H]+; Ή NMR (500 MHz, d*-DMSO): δ 8.05 (d, J= 9.0 Hz, 2H), 8.00 (s, 1H),
7.79 (s, IH), 7.31 (m, 2H), 7.21 - 7.23 (m, 2H), 7.12 (d, J = 9.0 Hz, 2H), 5.46 (q, J= 6.0 Hz, 1H), 4.67 (d, J = 14.0 Hz, 1H), 4.46 (s, 1H), 4.40 (s, IH), 4.36 (q, J= 7.0 Hz, 2H), 4.20 (d, J= 14.0 Hz, 1H), 3.24 (s, 3H), 1.72 (d, J= 6.0 Hz, 3H), 1.36 (t, J= 7.0 Hz, 3H) ppm.
[0155] Synthesis of compound 28-8a. Under an atmosphere of N2, ZnEt2 (IM in hexane, 7.40 mL, 7.40 mmol) was added to dry DCM (20 mL) at -78 °C, followed by CH2I2 (1.2 mL, 14.8 mmol) over 10 tnin. The resulting mixture was stirred at -78 °C for 30 min and then at -10 °C for 30 min. The mixture was cooled to -78 °C and a solution ofTFA (137 μί, 1.9 mmol) in DCM (I mL) was added dropwise. After stirring at -78 °C for 30 min, the mixture was added dropwise a solution of compound 28-7a (340 mg, 0.62 mmol) in DCM (2 mL) at -78 °C. The resulting mixture was stirred at -78 °C for 10 min, 0 °C for 1 hr, and 25 °C for 4 hrs. Subsequently, saturated aq. NH4CI solution (10 mL) was added and the mixture was concentrated. The residue was extracted with DCM (20 mL x 3). The combined organic extracts were washed with saturated aq. NaHC03 solution and water and dried over anhydrous Na2S04. The solvent was removed and the residue was purified by silica gel column chromatography (PE EtOAc = 10/1 to 4/1 (vA>)) to give compound 28-8a (230 mg, 66% yield) as a white solid. LC-MS (ESI): m/z 566.2 [M + Hf; Ή NMR (500 MHz,
Figure imgf000083_0001
δ 8.06 - 8.08 (m, 2H), 8.01 (s, IH), 7.88 (s, IH), 7.30 - 7.34 (m, 2H), 7.21 - 7.24 (m, 2H), 7.12 - 7.14 (m, 2H), 5.09 (q, J = 6.0 Hz, IH), 4.36 (q, J = 7.5 Hz, 2H), 3.69 (d, J= 15.0 Hz, IH), 3.57 (d, J= 15.0 Hz, IH), 3.32 (s, 3H), 1.57 (d, J = 6.0 Hz, 3H), 1.35 (t, J= 7.5 Hz, 3H), 0.71 - 0.96 (m, 4H) ppm.
[0156] Synthesis of compound 28-9a. Following the same procedure as that for the praparation of 1-16 described in Scheme 1 (W02012/058125) and replacing compound 1-14 with 28-8a, compound 28-9a was obtained as a white solid. LC-MS (ESI): m/z 551.2 [M+Hf ; Ή NMR (500
MHz, i^-DMSO): δ 8.50 (d, J= 4.5 Hz, IH), 7.92 (d, J= 9 Hz, 2H), 7.83 (s, IH), 7.56 (s, IH),
7.28 - 7.32 (m, 2H), 7.17 - 7.20 (m, 2H), 7.12 (d, J= 8.5 Hz, 2H), 5.06 (dd, J, = 12.5 Hz, J2 =
6.5 Hz, IH), 3.67 (d, J = 15.5 Hz, IH), 3.56 (d, J= 14.5 Hz, IH), 3.31 (s, 3H), 2.84 (d, J= 4.5 Hz,
3H), 1.54 (d, J= 6 Hz, 3H), 0.70 - 0.93 (m, 4H) ppm. Compound 28-9a was separated into a pair of enantiomers: enantiomer 28-9a_A (t¾ = 4.13 min) and enantiomer 28-9a_B (tR = 5.05 min) detected by UV absorption at 214 nm on a 4.6 mm x 250 mm x 5 μπι Regis ( ?^? Whelk-ol column (column temperature: 39.3 °C; eluent: MeOHAiquid CO2 = 50/50 (v/v); CO2 flow rate: 1.5 g/min and co-solvent flow rate: 1.5 g/min; front pressure: 218 bar and back pressure: 152 bar).
[0157] Synthesis of compound 28-9b. Following the same procedure as that for the praparation of compound 28-9a described in Scheme 28 and replacing compound 15-11 with 8-4, compound
28-9b was obtained as a white solid. LC-MS (ESI): m/z 459.1 [M + Hf; Ή NMR (500 MHz, t -
DMSO): δ 8.53 (m, IH), 7.97 (dd, J, = 5.5 Hz, J2 = 8.7 Hz, 2H), 7.85 (s, IH), 7.58 (s, IH), 7.41 (t,
J = 8.5 Hz, 2H), 5.06 (q, J = 6.5 Hz, IH), 3.68 (d, J = 14.5 Hz, IH), 3.57 (<L J= 14.5 Hz, IH), 3.32 (s, 3H), 2.84 (d, J= 4.5 Hz, 3H), 1.54 (d, J= 6.5 Hz, 3H), 0.93 (m, 1H), 0.84 - 0.86 (m, 2H), 0.70 (m, 1H) ppm. Compound 28-9b was separated into a pair of enantiomers: enantiomer 28- 9b_A (tR = 4.36 min) and enantiomer 28-9b_B (tR = 6.09 min) detected by UV absorption at 214 nm on a 4.6 mm x 250 mm 5 μιη ChiralPak® AD-H column (column temperature: 39.8 °C;
eluent: MeOH/liquid CO2 = 30/70 (v/v); CO2 flow rate: 2.1 g min and co-solvent flow rate: 0.9 g min; back pressure: 150 bar).
[0158] Synthesis of compound 28-10a. To a solution of compound 28-7a (680 mg, 1.2 mmol) in THF (10 mL) was added BH3 THF (7.4 mL, 7.4 mmol) at 0°C. After stirring at rt for 3 hrs, the reaction mixture was added 3 N aq. NaOH (7 mL) at 0°C, followed by 30% aq. H2O2 (7 mL). The reaction mixture was stirred at rt overnight and the added iced water (30 mL). The mixture was extracted with EtOAc (25 mL x 2). The combined organic extracts were washed with water (20 mL x 2), dried over anhydrous Na2SC>4, and concentrated. The residue was purified by silica gel column chromatography (DCM/Acetone = 50/1 (v/v)) to give compound 28-10a (560 mg, 80% yield) as a white solid. LC-MS (ESI): mlz 592.2 [M + Naf.
[0159] Synthesis of compound 28-10b. Following the same procedure as that for the praparation of compound 28-10a and replacing compound 28-7a with 28-7b, compound 28-10b was obtained as a white solid. LC-MS (ESI): mlz 500.1 [M + Naf.
[0160] Synthesis of compound 28-1 la. Following the same procedure as that for the praparation of compound 1-16 described in Scheme 1 and replacing compound 1-14 with 28-10a, compound 28-1 la was obtained as a white solid. LC-MS (ESI): mlz 555.2 [M + H ; H NMR (500 MHz, <?- DMSO): δ 8.42 - 8.50 (m, IH), 7.90 - 7.93 (m, 2H), 7.82 and 7.77 (s, s, 1H), 7.56 and 7.54 (s, s, 1H), 7.27 - 7.31 (m, 2H), 7.17 -7.19 (m, 2H), 7.1 1 (d, J= 8.5 Hz, 2H), 5.17 and 4.88 (m, m, IH), 4.78 (m, IH), 4.18 and 3.99 (m, m, IH), 3.93 (m, IH), 3.45 (m, IH), 3.47 (m, IH), 3.38 and 3.36 (s, s, 3H), 2.84 (<L J = 4.5 Hz, 3H), 2.83 (m, IH), 1.62 and 1.60 (d, d, J= 6.5 Hz, 3H) ppm.
[0161] Synthesis of compound 28-1 lb. Following the same procedure as that for the praparation of compound 28-1 la described in Scheme 28 and replacing compound 28-10a with 28-10b, compound 28-1 lb was obtained as a white solid. LC-MS (ESI): mlz 463.1 [M + H]+; lH NMR (500 MHz, i -DMSO): δ 8.52 (m, IH), 7.95 - 7.98 (m, 2H), 7.83 and 7.79 (s, s, IH), 7.58 and 7.57 (s, s, IH), 7.40 (t, J= 9.0 Hz, 2H), 4.88 (q, J = 6.5 Hz, IH), 4.78 (t, J= 5.5 Hz, IH), 4.19 (d,
S3 J= 15 Hz, 1H), 3.92 (m, 1H), 3.43 - 3.48 (m, 1H), 3.39 (s, 3H), 3.26 - 3.31 (m, 1H), 2.84 (<L J= 4.5 Hz, 3H), 2.83 (m, 1H), 1.62 and 1.60 (d, d, J= 6.0 Hz, 3H) ppm.
[0162] Synthesis of compound 28-12a. To a solution of compound 28-10a (200 mg, 0.35 mmol), DMAP (21 mg, 0.18 mmol) and Et3N (0.15 mL, 1.1 mmol) in CH2C12 (5 mL) was added TsCl ( 100 mg, 0.53 mmol) at 0 °C. After stirring at rt for 2 hrs, the reaction mixture was added ice water (10 mL) and DCM (25 mL). The organic layer was washed with saturated aq. NaHCOi (10 mL x 2), water (10 mL x 2) and brine ( 10 mL), dried over anhydrous NajSO-j, and concentrated. The residue was purified by silica gel column chromatography (PE EtOAc = 6/1 to 2/1 (v/v)) to give the tosylate as a white solid (230 mg, 91% yield). LC-MS (ESI): mlz 746.2 [M+Na] +.
Subsequently, a mixture of the tosylate (140 mg, 0.19 mmol), MeSC^Na (59 mg, 0.58 mmol) and KI (964 mg, 0.581 mmol) in DMF (2 mL) was stirred at 120 °C for 2 hrs. The mixture was then poured into water (15 mL). The resulting precipitate was filtered and the white was washed with water (15 mL x 3) and dried in vacuo to give compound 28-12a (100 mg, 82% yield). LC-MS (ESI); mlz 654.1 [M + Naf.
[0163] Synthesis of compound 28-12b. Following the same procedure as that for the praparation of compound 28-12a described in Scheme 28 and replacing compound 28-10a with 28-10b, compound 28-12b was obtained as a white solid. LC-MS (ESI): mlz 562.1 [M + Hf.
[0164] Synthesis of compound 28-13a. Following the same procedure as that for the praparation of compound 1-16 described in Scheme 1 and replacing compound 1-14 with 28-12a, compound 28-13a was obtained as a white solid. LC-MS (ESI): mlz 617.1 [M + H]+; Ή NMR (500 MHz, CDCb): δ 7.94 (s, IH), 7.81 (dd, J, = 2.5 Ez, J2 = 8.5 Hz, 2H), 7.61 (s, 1H), 7.05 - 7.13 (m, 6H), 5.82 (m, IH), 5.09 (q, J= 8.5 Hz, 1H), 4.59 (t, J= 1 1.5 Hz, 1H), 4.24 (d, J= 18.5 Hz, IH), 3.16 (s, 3H), 3.05 - 3.13 (m, 3H), 3.07 (s, 3H), 2.98 (d, J= 6.0 Hz, 3H), 1.78 (d, J= 8.5 Hz, 3H) ppm. Alternatively, compound 28-13a can be obtained using compound 28-1 la as the starting material as described in Scheme 28.
[0165] Synthesis of compound 28-13b. Following the same procedure as that for the praparation of compound 28-13a described in Scheme 28 and replacing compound 28-12a with 28-12b, compound 28-13b was obtained as a white solid. LC-MS (ESI): mlz 525.1 [M + Hf ; Ή NMR (500 MHz, tZ-DMSO): δ 8.52 (m, IH), 7.95 - 7.99 (m, 2H), 7.87 (s, IH), 7.61 (s, IH), 7.40 (t, J = 8.5 Hz, 2H), 4.98 (q, J = 6.0 Hz, 1 H), 4.39 (t, J = 9.0 Hz, IH), 4.20 (d, J= 14.0 Hz, 1H), 3.37 (s, 3H), 3.29 - 3.32 (m, 2H), 3.04 (s, 3H), 2.89 (m, 1H), 2.85 (d, J =4.5 Hz, 3H), 1.66 (d, J=
3H) ppm. Alternatively, compound 28-13b can be obtained using compound 28-1 lb as th
Figure imgf000086_0001
[0166] Step 1. Refer to Scheme 29. To a solution of compound 4-2 (9.0 g, 18.9 mmol) in DME (200 mL) and H20 (400 mL) were added 2C03 (7.8 g, 56.6 mmol), Pd(dppf)Cl2 (1.5 g, 1.9 mmol) and 4,4,5,5-tetramethyl-2-vinyl- 1 ,3,2-dioxaborolane (4.4 g, 28.3 mmol). After stirring at 60 °C for 2 hrs under an atmosphere of Ar, the reaction mixture was concentrated and the residue was partitioned between water (150 mL) and EtOAc (150 mL). The aqueous phase was extracted with EtOAc ( 100 mL x 3) and the combined organic extracts were washed with water ( 100 mL x 3) and brine (100 mL) and dried over anhydrous Na2S04. The solvent was removed and the residue was purified by silica gel column chromatography (PE/EtOAc = 15/1 (vV)) to give compound 29-1 (5.0 g, 75% yield) as a yellow solid. LCMS (ESI): m/z 356.1 [M + H ,
[0167] Step 2. To a solution of compound 29-1 (1.2 g, 3.4 mmol) in THF (50 mL), EtOH (20 mL) and HOAc (40 mL) was slowly added Zn ( 1.3 g, 20.1 mmol) at 0 °C. After stirring at rt for 2 hrs, the reaction mixture was filtered and the filtrate was concentrated. The residue was partitioned between water (80 mL) and EtOAc (80 mL) and the organic layer was extracted with EtOAc (60 mL x 3). The organic extracts were combined and washed with water (80 mL x 2), sat. aq, NaHC03 (80 mL) and brine (80 mL) and dried over anhydrous Na2S04. The solvent was concentrated and the residue was dried in vacuo to give crude compound 29-2 (1.0 g, 91% yield) as a yellow solid. LC-MS (ESI): m/z 326.1 [M + H]+. [0168] Step 3. To a solution of compound 29-2 (1.0 g, 3.1 mmol) in anhydrous pyridine (5 mL) was treated with DMAP (20 mg), followed by a solution ofMsCl (I. I g, 9.2 mmol) in DCM (3 mL) at 0 °C. After stirring at rt for 3 hrs, the reaction mixture was concentrated and the residue was partitioned between water (20 mL) and EtOAc (20 mL). The aqueous layer was extracted with DCM (30 mL x 3) and the combined organic extracts were washed with water (60 mL x 2) and brine (60 mL) and dried over anhydrous Na2S04. The solvent was removed and the residue was dried in vacuo to give crude compound 29-3 (1.1 g, 88% yield) as a yellow solid. LC-MS (ESI): m/z 404.1 [M + Hf.
[0169] Step 4. To a solution of compound 29-3 (700 mg, 1.7 mmol) in DMF (30 mL) were added K2C(¼ (719 mg, 5.2 mmol) and I (144 mg, 0.87 mmol), followed by 2-bromobenzyl chloride (534 mg, 2.6 mmol). After stirring at 70 °C for 3 hrs, the reaction mixture was concentrated and the residue was partitioned between water (50 mL) and EtOAc (50 mL). The aqueous layer was extracted with EtOAc (3 x 40 mL) and the combined organic extracts were washed with water (80 mL x 3) and brine (50 mL) and dried over anhydrous NaaSO-j. The solvent was removed and the residue was purified by silica gel column chromatography (Petroleum ether EtOAc = 15/1 (vV)) to give compound 29-4 (800 mg, 87% yield) as a yellow solid. LC-MS (ESI): m/z 574.0 [M + H]+.
[0170] Step 5. To a solution of compound 29-4 (770 mg, 1.34 mmol) in CH3CN (25 ml) were added Et3N (4.6 mL), and Pd(PPh3)4(l.55 g, 1.34 mmol). After stirring at 80 °C for several hours under an atmosphere of Ar, the reaction mixture was concentrated and the residue was purified by silica gel column chromatography (Petroleum ether/EtOAc = 10/1 (vV)) to give compound 29-5 (260 mg, 40% yield) as a yellow solid. LC-MS (ESI): m/z 492.1 [M + H]+.
[0171] Step 6. To a solution of compound 29-5 (150 mg, 0.31 mmol) in EtOH (30 mL) was added 5% Pd/C (w w, 200 mg). After stirring at 50 °C for several hours under an atmosphere of ¾, the reaction mixture was filtered through a pad of Celite®545. The filtered cake was washed with EtOH (15 mL x 2). The filtrate was concentrated and the residue was dried in vacuo to give crude compound 29-6 (149 mg, 99% yield) as a yellow solid. LC-MS (ESI): m/z 494.1 [M + H]+.
[0172] Step 7. Following the same procedure as that for for the preparation of compound 1-16 described in Scheme 1 and replacing compound 1-14 with 29-6, compound 29-7 was obtained (130 mg, 90% yield) as a pale brown solid. LC-MS (ESI): m/z 479.1 [M + Hf; 'H NMR (500 MHz, CDC13): δ 7.85 - 7.88 (m, 2H), 7.78 (s, 1H), 7.71 (s, IH), 7.30 (d, 2H), 7.15 - 7.24 (m, 5H), 5.76 (ore, 2H), 5.12 (d, J= 16 Hz, 2H), 4.95 (d, J= 16 Hz, 5H), 4.57 - 4.62 (dd, J, = 15 Hz, J2 = 7 Hz, 1H), 2.72 (d, J = 5 Hz, 3H), 2.73 (s, 3H), 1.80 (d, J = 8 Hz, 3H) ppm. Compound 29-7 was separated into a pair of enantiomers: enantiomer 29-7_A (tR = 4.16 min) and enantiomer 29-7 B (tR = 6.05 min) detected by LTV absorption at 214 nm on a 4.6 mm x 250 mm 5 μηι ChiralPak® OD-H column (column temperature: 40.4 °C; eluent: MeOH/liquid C02 = 30/70 (v/v); CO2 flow rate: 2.1 g/min and co-solvent flow rate: 0.9 g/min; front pressure: 205 bar and back pressure: 148 bar).
Figure imgf000088_0001
B R = F 30-2· R » F
b R - 4-F-PhO- 30-2b R« 4-F-PhO-
(0173] Chiral separation of compound 8-5. Compound 8-5 (3.8 g) was separated into a pair of enantiomers: (Λ)-8-5 (tR = 2.61 min, 1.6 g, 84% yield) and (S)-8-5 (tR = 3.14 min, 1.6 g, 84% yield) detected by UV absorption at 214 nm on a 4.6 mm x 250 mm x 5 μπι ChiralPak® AD-H column (column temperature: 40.2 °C; eluent: MeOH (0.1% DEA)/liquid C02 = 30/70 (v/v); C02 flow rate: 2.1 g/min and co-solvent flow rate: 0.9 g/min; front pressure: 206 bar and back pressure: 149 bar).
[0174] Chiral separation of compound 15-12. Using the same prep-chiral HPLC condition as that used for separating compound 8-5, Compound 15-12 (5.6 g) was separated into a pair of enantiomers: (fl)-15-12 (tR = 5.71 min, 1.1 g, 39% yield) and (S)- 15-12 (tR = 6.58 min, 1.0 g, 36% yield).
Figure imgf000089_0001
[0175] Synthesis of compound 30-1 a. To a solution of the enantiomer came out first from the chiral separation of compound 8-5 (tR = 2.61 min) (30 mg, 0.07 mmol) and (Λ)-ΜαΝΡ (18.4 mg, 0.08 ramol) in CH2Cl2 (2 mL) was added DCC (72.1 mg, 0.35 mmol), followed by DMAP (17.1 mg, 0.14 mmol). After stirring at rt for 20 hrs, the reaction mixture was concentrated and the residue was diluted with EtOAc (45 mL). The solution was washed with water (20 mL) and brine (20 mL), dried with anhydrous Na2S0 , and concentrated. The residue was purified by prep- HPLC to give compound 30-la (15 mg) as a white powder. LC-MS: (ESI) miz 656.2 [M + Naf; Ή NMR (500 MHz, CDC13): 6 8.1 1 - 8.13 (m, 2H), 7.74 - 7.77 (m, 2H), 7.60 (<L J = 7.0 Hz, 2H). 7.47 (t, J= 8.0 Hz, 1H), 7.45 (s, 1H), 7.22 (t, J= 9.0 Hz, 2H ), 7.08 (s, 1H), 7.00 (t, J = 7.0 Hz, 1H), 6.76 (t, J= 8.0 Hz, 1H), 6.07 (q, J= 7.0 Hz, 1H), 4.23 - 4.36 (m, 2H), 3.05 (s, 3H), 2.98 (s,
= 7.0 Hz, 3H), 1.27 (t, J = 7.0 Hz, 3H) ppm.
Figure imgf000089_0002
[0176] Synthesis of compound 30-2a. Following the same procedure as that used for preparing compound 30-la and replacing (Λ)-ΜαΝΡ with (S)-MaNP, compound 30-2a was obtained. LC- MS: (ESI) mlz 656.2 [M + Naf;Ή NMR (500 MHz, CDC13): δ 8.06 - 8.09 (m, 2H), 7.95 (d, J= 8.5 Hz, lH), 7.87 (s, 1H), 7.82 (d, J= 8.0 Hz, 1H), 7.78 (d, J= 8.0 Hz, 1H), 7.73 (s, IH), 7.67 (s, 1H), 7.64 (d, J = 7.0 Hz, 1H), 7.47 (t, J= 8.0 Hz, 1H), 7.28 (t, J= 7.5 Hz, 1H), 7.20 (t, J = 7.5 Hz, 2H), 7.06 (t, J = 7.5 Hz, IH), 6.12 (q, J= 6.5 Hz, 1H), 4.35 - 4.41 (m, 2H), 3.12 (s, 3H), 3.00 (s,
= 6.5 Hz, 3H), 1.27 (t, J = 7.0 Hz, 3H) ppm.
Figure imgf000089_0003
[0177J Synthesis of compound 30-lb. Following the same procedure as that used for preparing compound 30-la and using the enantiomer came out first from the chiral separation of compound 15-12 (tR = 5.71 min), compound 30-lb was obtained. LC-MS: (ESI) miz 748.2 [M + Naf; Ή NMR (500 MHz, CD3C1): δ 8.08 - 8.10 (m, 2H), 7.06 - 7.1 1 (m, 7H), 6.99 (t, 7= 7.0 Hz, 1H), 6.75 (dt, J, = 1.0 Hz, J2 = 8.0 Hz, 1H), 6.06 (q, J= 7.0 Hz, 1H), 4.24 -4.35 (m, 2H), 3.05 (s, 3H),
J= 7.0 Hz, 3H), 1.27 (t, J= 7.0 Hz, 3H) ppm.
Figure imgf000090_0001
10178) Synthesis of compound 30-2b. Following the same procedure as that used for preparing compound 30-lb and replacing (Λ)-ΜαΝΡ with (5)-ΜαΝΡ, compound 30-2b was obtained. LC- MS: (ESI) miz 748.2 [M + Naf; Ή NMR (500 MHz, CD3C1): 5 8.06 (d, J= 7.0 Hz, 2H), 7.93 (d, J= 8.5 Hz, 1H), 7.85 (s, 1H , 7.83 (d, J= 8.0 Hz, 1H), 7.78 (d, J= 8.0 Hz, 1H), 7.69 (s, 1H), 7.63 - 7.66 (m, 2H), 7.46 (t, J= 8.0 Hz, 1H), 7.29 (t, J= 8.0 Hz, 1H), 7.04 - 7.10 (m, 7H), 6.10 (q, J= 7.0 Hz, 1H), 4.36 - 4.39 (m, 2H), 3.13 (s, 3H), 2.99 (s, 3H), 1.91 (s, 3H), 1.48 (d, J= 7.0 Hz, 3H),
= 7.0 Hz, 3H) ppm.
Figure imgf000090_0002
101791 Determination of chiraliry. Refer to Scheme 31. Based on the general rule of chemical shfits of a pair of diastereomeric esters derived from an alcohol with (Λ)-ΜαΝΡ and (5)-MaNP, the chiraliry of the benzylic carbon in the enantiomer came out first (t¾ = 2.61 min) from the chiral separation of compound 8-5 was determined as R. Accordingly, the chirality of the benzylic carbon in the enantiomer came out first (tR = 5.71 min) from the chiral separation of compound 15-12 was determined as R.
Figure imgf000091_0001
{01801 Synthesis of compound (J?)-8-5. Refer to scheme 32. A 25 mL flask was charged with triethylamine (76 mg, 0.75 mmol, 7 eq.) in an ice bath, followed by adding formic acid (35 mg, 0.75 mmol, 7 eq.) dropwise. After stirring at rt for 20 min, a solution of compound 8-4 (45 mg, 0.107 mmol, 1 equiv.) in DMF (6 mL) and RuCl[(R,i.)-Tsdpen](p-cymene) (1.6 mg, 0.0029 mmol 0.024 equiv.) were added. The resulting dark red reaction mixture was stirred at 40 °C overnight and then concentrated. The residue was purified by silica column chromatography (EtOAc/PE = 1 :1 (v/v)) to give compound (Jf)-8-5 (30 mg, 66% yield, 95.5%?<?) as a white solid. The absolute configuration of the sample (tR = 2.59 min) was determined as R by taking chiral HPLC along with compound 8-5 following chiral HPLC the condition in Scheme 30.
101811 Synthesis of compound (S)-8-5. Following the same procedure as described for the preparation of compound (Jf)-8-5 and replacing RuCl[(i?^?)-Tsdpen](p-cymene) with RuCl[(S,S)- Tsdpen](p-cymene), compound (S)-8-5 (28 mg, 66% yield, 94.0%ee, tR = 3,12 min, S
configuration) was obtained from compound 8-4 (42 mg, 0.1 mmol).
]0182| Synthesis of compound (Jf)-15-12. Following the same procedure as described for the preparation of compound (R)S-5 and replacing compound 8-4 with compound 15-11 (50 mg, 0.1 mmol), compound (S)-8-12 (38 mg, 75% yield, 95.9%ee, tR = 5.76 min, R configuration).
101831 Synthesis of compound (S)-15-12. Following the same procedure as described for the preparation of compound ( ?)-15-12 and replacing RuCl[(i?,/?)-Tsdpen](p-cymene) with
RuCl[(.?^S)-Tsdpen](p-cymene), compound (5)-15-12 (70 mg, 70% yield, 96.6% ee, tR = 6.70 min, S configuration) was obtained from compound 15-11 (100 mg, 0.2 mmol). 10184] Synthesis of compound (Λ)-8-9. Using compound (J?)-8-5 as the starting material and following the same procedure for the preparation of compound 8-9 described in Scheme 8, compound (J?)-8-9 was obtained. Chiral HPLC analysis determined that compound (Ji)-8-9 and enantiomer 8-9_A obtained from chiral separation of compound 8-9 are identical.
{01851 Synthesis of compound (J?)- 15- 15. Using compound (Λ)-15-12 as the starting material and following the same procedure for the preparation of compound 15-15 described in Scheme 15, compound (Λ)-15-15 was obtained. Chiral HPLC analysis determined that compound (Λ)-15-15 and enantiomer 15~15_A obtained from chiral separation of compound 15-15 are identical.
10186) Syntheses of diastereomers of compounds 28-1 la and -13a. Using either compounds (Jf)-15-12 or 0-0-15-12 as the starting material and following the procedure for the preparation of compounds 28-1 la and -13a described in Scheme 28, those diastereomers of compounds 28-1 la and -13a were obtained, respectively. The absolute configurations of those diastereomers were determined by 2D-COSY and NOESY spectra.
]0187I Syntheses of diastereomers of compounds 28-1 lb and -13b. Using either compounds (Λ)-8-5 or (-5)-8-5 as the starting material and following the procedure for the preparation of compounds 28-llb and -13b described in Scheme 28, those diastereomers of compounds 28-1 lb and -13b were obtained, respectively. The absolute configurations of those diastereomers were determined by 2D-COSY and NOESY spectra.
Scheme 34
Figure imgf000092_0001
<5S,7S)-3 -2a R - F (SS.7R> 4-2a R = F
(5S.7S)-3 -2b R - 4-F-PhO- (SS m-3**b R - 4JF-PHQ-
|0188| Step 1. Refer to Scheme 34. To a solution of compound (5)-15-12 (5.13 g, lO mmol) in
DMF (40 mL) was added K2C03 (2.07 g, 15 mmol), the resulting mixture was stirred at rt for 30 min. Subsequently, a solution of racemic 2-(bromomethyl)oxirane (1.23 mL, 15 mmol) in DMF
(10 mL) was dropwise added to the mixture. After stirring at 60 °C overnight, the reaction mixture was concentrated. The residue was diluted with H20 (100 mL) and EtOAc (150 mL). The aqueous layer was extracted with EtOAc (150 mL x 2). The combined organic extracts were washed with H2O (100 mL x 2) and brine (100 mL), dried over anhydrous a2S >4, filtrated and concentrated. The residue was purified by silica gel column chromatography (Petroleum ether/EtOAc = 1/1 (vV)) to give compound 3 -lb (4.8 g, 84% yield) as a white solid. LC-MS
Figure imgf000093_0001
101891 Step 2. To a solution ofTsOH (91 mg, 0.53 mmol) in DCM (10 mL) at rt was added compound 34-lb (150 mg, 0.26 mmol). After stirring at rt overnight, the reaction mixture was concentrated and the residue was dissolved in DCM (50 mL). The mixture was washed with sat. aq. NaHCC>3 (25 mL x 2) and brine (25 mL), dried over anhydrous Na2S04, filtered, and concentrated. The residue was purified by silica gel column chromatography (Petroleum ether/EtOAc = 1/1 (v v)) to give a mixture of compounds (55,75>34-2b and (55,7 ?)-34-2b (120 mg, 81% yield) as white solid in a ratio of 1/1 as determined by comparing the integrations of the benzylic carbons at 5.08 ppm for compound (5S,7S)-34-2b and 5.20 ppm for compound (5S,7R)- 34-2b, respectively. LC-MS (ESI): mlz 570.1 [M + H]+. Compound (5S,7S)-34-2b: Ή NMR (500 MHz, CDCI3): 5 8.14 (s, 1H), 8.04 (m, 2H), 7.58 (s, 1H), 7.03 - 7.1 1 (m, 6H), 5.08 (q, J= 6.5 Hz, 1H), 4.42 (q, J= 7.0 Hz, 2H), 4.13 - 4.19 (m, 2H), 3.73 (dd, J, = 3.5 Hz, J2 = 11.5 Hz, 1H), 3.53 (dd, Jj = 6.0 Hz, J. = 11.8 Hz, 1H), 3.18 (s, 1H), 3.12 (m, 1H), 1.92 (bs, 1H), 1.77 (d, J= 7.0 Hz, 3H), 1.44 (t, J= 6.5 Hz, 3H) ppm. Compound (5S,7/?)-34-2b: Ή NMR (500 MHz, CDC13): δ 7.94 - 7.97 (m, 3H), 7.52 (s, 1H), 6.96 - 7.03 (m, 6H), 5.20 (q, J= 6.5 Hz, 1H), 4.35 (q, J= 7.0 Hz, 2H), 3.94 (brm, 1H), 3.71 (dd, J, =4.5 Hz, J2 = 1 1.8 Hz, 1H), 3.58 - 3.66 (m, 4H), 3.08 (s, 3H), 1.92 (bs, 1H), 1.67 (d, J= 7.0 Hz, 3H), 1.36 (t, J= 7.0 Hz, 3H) ppm. Further chiral HPLC analyses showed that although better ratios were achieved when performing the cyclization at elevated temperatures, racemization of the chirality at the benzylic carbon was observed and a racemic synlanti mixture of the cyclized products was obtained.
|0190| Comparison of the stereoselectivity of the acid catalyzed epoxide-opening-ring-formation step
Temperature
Starting
Acid Solvent and reaction ratio of compound 34-2 material
time
34-la TsOH (0.3 eq.) DCM 0 °C, 1.0/3.0 ((5S,75)-34-2a (55,7/?)-34-2a)
overnight 34-la TsOH (2.0 eq.) DCM rt, overnight 1.0/11.0 ((55,75)-■34-2a (55,7*)-34-2a)
34-la TsOH (2.0 eq.) DCE 60 °C, 4 hr 4.0/11.0 ((±)-syn- 34-2a/(±)-a/ift-34-2a)
34-lb TsOH (0.3 eq.) DCM 0 °C, 1.0/3 Ϊ.0 ((55,75)· ■34-2b/(55,7i?)-34-2b) overnight
34-lb TsOH (2.0 eq.) DCM rt, overnight 1.0/1 1.0 ((55,75)· ■34-2b/(55,7i¾)-34-2b)
34-lb TsOH (2.0 eq.) DCE 60 °C, 4 hr 6.5/1 1.0 i(± -syn-■34-2b/(±)- n/ -34-2b)
34-lb TsOH (2.0 eq.) Toluene 100 °C, 4 hr 7.5/11.0 {(±)sytt- 34-2b/(±)-an/i-34-2b)
34-lb TsOH (2.0 eq.) DMF 150 °C, 2 for 3.0/210 ((±)-syn- 34-2b/(±)-an/ -34-2b)
Figure imgf000094_0001
(5S,7S)-3 -3a
|0191J Step 1. Refer to Scheme 34a. To a stirred solution of compound 8-5 (15.0 g, 35.6 mmol) in toluene (250 mL) at 70 °C was added PPTs (3.56 g, 14.2 mol). After refluxing overnight, the reaction mixture was concentrated and the residue was dissolved in DCM (200 mL). The resulting mixture was washed with sat, aq. NaHC03 (300 mL) and brine (200 mL), and dried over anhydrous Na2SC> . The solvent was removed and the residue was purified by silica gel column chromatography (Petroleum ether/Acetone = 10/1 (vV)) to give compound 29-3 (10.0 g, 70% yield) as a pale yellow solid. LC-MS (ESI): mlz 404.1 [M + Hf; Ή NMR (500 MHz, CDC13): δ 8.14 (s, 1H), 8.07 - 8.04 (m, 2H), 7.72 (s, 1H), 7.18 ( = 8.5 Hz, 2H), 6.97 (dd, Jt = 11 Hz, J2 = 17 Ηζ,ΙΗ), 6.60 (s, 1H), 5.78 (d, J = 17 Hz, 1H), 5.53 (d, J= 11 Hz, 1H), 4.42 (q, J= 7.0 Hz, 2H), 3.02 (s, 3H), 1.42 (t, J= 7.0 Hz, 3H) ppm.
101921 Step 2. To a solution of compound 29-3 (100 mg, 0.25 mmol) in DMF (2 mL) were added K2CO3 (103 mg, 0.75 mmol) and (J?)-3-chloropropane-l,2-diol (82 mg, 0.744 mmol). After stirring at 110°C overnight, the reaction mixture was cooled to rt and poured into water (50 mL). The suspension was filtered; the solid was washed with water (20 mL), dried, and purified by silica gel column chromatography (DCM / MeOH = 100/1 (v V)) to give compound 34a-l (100 mg, 84% yield) as a white solid. LC-MS (ESI): mlz 500.1 [M + Na]+.
10193] Step 3. To a stirred solution of compound 34a-l (94.0 mg, 0.20 mmol) in DCE (20 mL) at 85°C was added TsOH (94 mg, 0.49 mmol). After refluxing overnight, the reaction mixture was concentrated. The residue was diluted with DCM (50 mL) and the resulting mixture was washed with sat. aq. NaHCCb (50 mL) and brine (50 mL), and dried over anhydrous Na2S(> . The solvent was removed and the residue was purified by silica gel column chromatography (Petroleum ether/EtOAc = 2/1 (v V)) to give a mixture of compounds (55,75 34-2a and (5/?,7S)-34-2a with a ratio of 7.8/1 (75 mg, 80% yield) as a white solid. LC-MS (ESI): mlz 500.1 [M + Naf.
10194] Synthesis of compound 34a-2. Method A. To a solution of compound 29-3 (1.0 g, 2.5 mmol) in DMF (25 mL) were added K2C03 (1.04 g, 7.5 mmol) and ( ?)-(2,2-dimethyl-l,3- dioxolan-4-yl)methyl 4-methylbenzenesulfonate (1.42 g, 5.0 mmol). After stirring at 90°C for 48 hr, the reaction mixture was cooled, diluted with EtOAc (100 mL), and filtered through
Celite®545. The filtered cake was washed with EtOAc (100 mL). The filtrate was concentrated and the residue was diluted with EtOAc (100 mL). The mixture was washed with water (50 mL x 2) and brine (50 mL), and dried with anhydrous Na2SC>4. The solvent was removed and the residue was purified by silica gel column chromatography (Petroleum ether/EtOAc = 5/1 (v/V)) to give compound 34a-2 (0.95 g, 74% yield) as a yellow solid. LC-MS (ESI): m/∑ 540.2 [M + Naf. |0195| Synthesis of compound 34a-2. Method B. To a solution of compound 29-3 (1.2 g, 3.0 mmol) in dry toluene (20 mL) were added n-BujP (1.4 mL, 6 mmol) and DIAD (1.2 mL, 6 mmol) under an atmosphere of N2. After stirring at 0 °C for 30 min, the reaction mixture was added a solution of (5)-(2,2-dimethyl-l,3-dioxolan-4-yl)methanol (0.56 mL, 4.5 mmol) in toluene (5 mL). The resulting reaction mixture was stirred at 90 °C overnight and then concentrated. The residue was diluted with DCM (200 mL), washed with water (50 mL x 2) and brine (50 mL), and dried over anhydrous Na2SC>4. The solvent was removed and the residue was purified by silica gel column chromatography (Petroleum ether/EtOAc = 4/1 (v/v)) to give compound 34a-2 (1.3 g, 83% yield) as a yellow solid. LC-MS (ESI): m/z 540.2 [M + Naf.
10196] Synthesis of compound (5S,75)-34-2a from compound 34a-2. To a stirred solution of compound 34a-2 (0.95 g, 1.8 mmol) in DCE (40 mL) at 85°C was added TsOH (0.86 g, 4.5 mmol). After reflux ing overnight, the reaction mixture was concentrated. The residue was diluted with DCM (200 mL) and the resulting mixture was washed with sat. aq. NaHC03 (50 mL x 2) and brine (50 mL), and dried over anhydrous Na2SC>4. The solvent was removed and the residue was purified by silica gel column chromatography (Petroleum ether/EtOAc = 2/1 (v/v)) to give a mixture of compounds (5S,7S)-34-2a and (5«,7S)-34-2a with a ratio of 7.8/1 (0.64 g, 74% yield) as a white solid. LC-MS (ESI): m/z 500.1 [M + Naf. The mixture was dissolved in DCE at reflux and slowly cooled to rt. The resulting suspension was filtered. The solid was washed with cold DCE and dried in vacuo to give compound (55,75)-34-2a with a diastereomeric purity of > 99% in 75% yield.
Figure imgf000096_0001
101971 Step 1. Refer to Scheme 34b. To a stirred solution of compound 34b-l (3.0 g, 17.2 mmol) in dry THF (20 mL) was added L1AIH4 (1.3 g, 34.4mmol) in portions at 0 °C. After stirring at rt overnight, the reaction was quenched by slowly adding isopropanol (10 mL) at 0 °C. The suspension was filtered through Celite®545 and the filtrate was concentrated. The residue was dried in vacuo to give crude compound 34b-2 (2.0 g) as a white solid, which was used directly for the next step without further purification. LC-MS (ESI): miz 147.1 [M + H]+, ]H NMR (500 MHz, CDClj): 54.04 - 4.01 (m, 1H), 3.82 (dd, J/ = 4.5 Hz, J2= 14.0 Hz, 1H), 3.67 - 3.64 (m, 1H), 3.61 (dd, J/ = 4.5 Hz, J 2= 14.0 Hz, 1H), 2.17 (br, 1H), 1.43 (s,3H), 1.41 (s, 3H), 1.30 (d, J= 6.0 Hz, 3H) ppm.
|0198| Step 2. To a stirred solution of compound 34b-2 (2.0 g, 13.6 mmol) in dry DCM (20 mL) and pyridine (5 mL) was added TsCl (3.8 g, 20.5mmol) in portions at 0 °C. After stirring at rt overnight, the reaction mixture was added water (20 mL). The mixture was extracted with DCM (50 mL x 3) and the combined organic extracts were washed with saturated aq. NH4CI and brine, and dried over anhydrous Na2SC>4. The solvent was removed and the residue was purified by silica gel column chromatography (Petroleum ether/EtOAc = 10/1 (v/v)) to give compound 34b-3 (3.4 g, 85% yield) as a white solid. LC-MS (ESI): miz 323.1 [M + Naf; Ή NMR (500 MHz, CDCI3): δ 7.80 (d, J = 8.0Hz, 2H), 7.35 (d, J= 8.5Hz, 2H), 4.10 -4.08 (m, 2H), 3.93 - 3.92 (m, 1H), 3.71 - 3.69 (m, 1H), 2.46 (s, 3H), 1.38 (s, 3H), 1.31 (s, 3H), 1.28 (d, J= 6.0 Hz, 3H) ppm. (0199| Step 3. To a stirred solution of compound 34b-3 (600 mg, 2.0 mmol) and 29-3 (1.0 g, 2.5 mmol) in dry DMF (4 mL) was added K2C03 (0.7 g, 5 mmol) at rt. After stirring at 100 °C under an atmosphere of Ar for 18 hr, the reaction mixture was cooled to rt and added water (40 mL). The suspension was filtered and the solid was washed with water and further purified by silica gel column chromatography (Petroleum ether/EtOAc = 10/1 (v/v)) to give compound 34b-4 (350 mg, 32% yield) as a white solid. LC-MS (ESI): miz 554.2 [M + Na] +.
10200} Step 4. To a refluxed solution of compound 34Ϊ (350 mg, 0.66 mmol) in dry DCE (4 mL) was slowly added /?-TsOH (113 mg, 0.66 mmol). After stirring was stirred at reflux for another 30 min, the reaction mixture was concentrated. The residue was added water (25 mL) and the suspension was filtered. The solid was washed with water and dried in vacuo to give a mixture (270 mg, 83% yield) of two diastereomers with a ratio of 3.5/1 (the desired diastereomer as a major product) based on chiral HPLC analysis. This mixture was separated by chiral HPLC to give compound 34b-5 (70 mg, 26% yield) as a white solid. LC-MS (ESI): miz 514.1 [M + Naf; Ή NMR (500 MHz, CDCI3): S 8.14 (s, 1H), 8.07 (dd, J/ = 6.5 Hz, J 2= 1 1.5 Hz, 2H), 7.19 (t, J=10.5 Hz, IH), 5.06 (q, J= 6.0 Hz, IH), 4.42 (q, J= 9.0 Hz, 2H), 4.19 (d, J= 17.0 Hz, 1H), 3.82 - 3.80 (m, IH), 3.65 - 3.59 (m, IH), 3.18 (s, 3H), 3.15 - 3.10 (m, IH), 2.30 (br, IH), 1.78 (d, J = 8.0 Hz, 3H), 1.43 (t, J= 9.5 Hz, 3H), 1.28 (d, J= 8.0 Hz, 3H) ppm.
[02011 Step 5. Following the same procedure used for preparing compound 4-9 shown in Scheme 4 and replacing compound 4-7 with 34b-5, compound 34b-7 was obtained as a white solid. LC- MS (ESI): mlz 499.1 [M + Na]+; 'H NMR (500 MHz, i/-DMSO): δ 8.54 (q, J= 4.5 Hz, IH), 7.97 (dd, J, = 9.0 Hz, J2= 5.5 Hz, 2H), 7.83 (s, IH), 7.58 (s, IH), 7.40 (t, J = 9.0 Hz, 2H), 4.86 (q, J= 7.0 Hz, IH), 4.62 (d, J= 5.0 Hz, IH), 4.22 (d, J = 10.0 Hz, IH), 3.80- 3.75 (m, IH), 3.68 - 3.65 (m, lH), 3.36 (s, 3H), 2.88 - 2,84 (m, IH), 2.84 (d, 7= 5.0 Hz, 3H), 1.63 (d, J= 6.0 Hz, 3H), 1.04 (d, J= 6.0 Hz, 3H) ppm.
Scheme 3S
Figure imgf000098_0001
10202] Synthesis of compound (55,75)-35-lb. Refer to Scheme 35. To a solution of compound (55,75)-34-3b (120 mg, 0.22 mmol) and DMAP (159 mg, 1.3 mmol) in DCM (2 mL) was added diphenyl phosphorochloridate (291 mg, 1.08 mmol) at 0 °C under an atmosphere of Ar. After stirring rt overnight, the reaction mixture was added ice water (10 mL) and DCM (10 mL). The organic layer was washed with saturated aq. NaHCQj (10 mL x 3) and water (10 mL x 3), dried over anhydrous Na2SC>4, filtered and concentrated. The residue was dried in vacuo to give crude compound (5S,7S)-35-lb (120 mg, 71% yield) as a white solid. LC-MS (ESI): miz 787.2 [M + H]+.
102031 Synthesis of compound (5S,7S)-35-2b. To a solution of compound (5S,7S)-35-lb (120 mg, 0.15 mmol) in THF (5 mL) was added PtC (50 mg). The resulting mixture was flushed with ¾ and stirred at rt overnight. Subsequently, the mixture was diluted with THF (25 mL) and filtered through Celite®545. The filtrate was concentrated and the residue was diluted with water (25 mL). The suspension was filtered; the solid was washed with water (10 mL) and C¾CN (10 mL) and dried in vacuo to give compound (55,75)-35-2b (50 mg, 52% yield) as a white solid. LC-MS (ESI): miz 635.2 [M + Hf; lU NMR (500 MHz, ^-DMSO): S 8.50 (d, J= 4.5 Hz, 1 H), 7.91 (d, J= 8.5 Hz, 2H),7.81 (s, 1H), 7.56 (s, 1H), 7.29 (d, J = 8.5 Hz, 2H), 7.15 - 7.18 (m, 2H), 7.10 (d, J= 8.5 Hz, 2H), 4.89 (d, J- 5.5 Hz, 1H), 4.10- 4.16 (m, 1H), 4.03 (br, lH), 3.77 (br, 1H), 3.58 - 3.60 (m, 1 H), 3.47 (br, 2H), 3.36 (s, 3H), 2.89 (br, 1H), 2.83 (d, J= 4.5 Hz, 3H), 1.60 (d, J= 5.5 Hz, 3H) ppm.
] 02041 Synthesis of compound (55,75 35-3b. To a solution of N, N-dimethylglycine (45 mg, 0.43 mmol), DCC ( 149 mg, 0.72 mmol) and compound (55,75)-34-3b (80 mg, 0.14 mmol) in CH2C12 (2 mL) was added DMAP (89 mg, 0.72 mmol) at rt. After stirring at rt overnight, the reaction mixture was filtered and the filtrated was concentrated. The residue was purified by preparative HPLC and product was converted to its HCl salt to give compound (5S,7S)-35-3b (50 mg, 54% yield) as a white solid. LC-MS (ESI): miz 640.2 [M + Hf; !H NMR (500 MHz, a6- DMSO): δ 10.32 (br, 1H), 8.51 (d, J= 5.0 Hz, 1H), 7.91 (d, J= 9.0 Hz, 2H),7.86 (s, 1H), 7.57 (s, 1H), 7.30 (t, J= 8.5 Hz, 2H), 7.17 - 7.20 (m, 2H), 7.12 (d, J= 9.0 Hz, 2H), 4.92 (q, J= 6.5 Hz, 1H), 4.19 -4.26 (m, 6H), 3.04 (s, 1H), 3.00 (br, 1 H), 2.83 (s, 9H), 1.64 (d, J= 6.0 Hz, 3H) ppm. J0205I Synthesis of compound (5S,7S)-35-4c. To a solution of /V-Boc-L- Alanine (68 mg, 0.36 mmol), DCC (149 mg, 0.72 mmol) and compound (55,75 -34-3b (100 mg, 0.18 mmol) in CH2C12 (2 mL) was added DMAP (89 mg, 0.72 mmol) at rt. After stirring at rt overnight, the reaction mixture was filtered; the filtrate was concentrated and the residue was dried in vacuo to give crude compound (5S,7S)-35-4c (105 mg, 80% yield) as a white solid. LC-MS (ESI): miz 626.1 [M - Boc + 2H .
102061 Synthesis of compound (5S,7S)-35-5c. A mixture of compound (5S,7S)-35-4c (100 mg, 0.14 mmol) in 4.0 N HCl in 1 ,4-dioxane (3 mL) was stirred at rt for 2 hr. The solvent was removed and the residue was purified by prep-HPLC to give compound (55,75)-35-5c (50 mg, 58% yield) as a white solid. LC-MS (ESI): m/z 626.1 [M + H] +; Ή NMR (500 MHz, t -DMSO): δ 8.48 - 8.51 (m, 1H), 8.40 (s, 2H), 7.90 (d, J = 8.5 Hz, 2H), 7.81 (s, 1H), 7.56 (s, 1H), 7.31 (t, J = 8.5 Hz, 2H), 7.16 - 7.20 (m, 2H), 7.12 (d, 7= 9.0 Hz, 2H), 4.91 (q, 7= 6.5 Hz, 1H), 4.09 - 4.26 (m, 5H), 4.00 (s, 3H), 2.95 (br, 1H), 2.83 (d, 7 = 6.5 Hz, 1H), 1.61 (d, 7 = 4.5 Hz, 3H), 1.40 (d, 7 = 7.0 Hz, 3H) ppm.
[0207] Synthesis of compound (5S,7S)-35-5d. Following the same procedure as that used for preparing compound (55,75)-35-5c and replacing N-Boc-L-Alanine with N-Boc-L-Valine, compound (5S,75)-35-5d was obtained as a white solid in 69% yield. LC-MS (ESI): m/z 654.2 [M + Hf; Ή NMR (500 MHz,
Figure imgf000100_0001
δ 8.49 - 8.5 l(m, 1H), 8.08 (br, 2H), 7.91 (cL 7 = 9.0 Hz, 2H), 7.86 (s, 1H), 7.57 (s, 1H), 7.30 (t, 7= 9.0 Hz, 2H), 7.17 - 7.20 (m, 2H), 7.12 (d, 7 = 9.0 Hz, 2H), 4.93 (d, 7= 6.5 Hz, 1H), 4.15 - 4.20 (m, 4H), 3.87 (s, 1 H), 3.36 (s, 3H), 2.95 (br, 1H), 2.84 (d, 7 = 4.0 Hz, 3H), 2.13 - 2.16 (m, 1H), 2.16 (d, 7= 6.5 Hz, 3H), 1.00 (d, 7= 6.5 Hz, 3H), 0.95 (d, 7= 6.5 Hz, 3H) ppm.
[0208] Synthesis of analogs of compound (55,7£)-35-2, -3, and -5. Following the same synthetic strategy described in Scheme 35, the following analogs were readily obtained.
Figure imgf000100_0002
Figure imgf000101_0001
3H), 1.41 (d,J=7Hz,3H)
10.43 (br, IH), 8.53 (q,J=4.5 Hz, IH), 7.96 (t, J = 4.5 Hz, 2H), 7.87 (s, IH), 7.60 (s, IH), 7.41 (t,J=9.0 Hz, 2H), 4.93 (q,J = r 548.2
6.5 Hz, IH), 4.18-4.26 (m, 6H), 3.41 (s, 3H), 2.97 (t,J= 9.0 Hz, IH), 2.80-2.85 (m , 9H), 2.54 (s, IH), 1.64 (d,J=6.5Hz,3H)
8.54 (q, J = 4.0 Hz, IH), 8.38 (br, 3H), 7.95 (q,J=5.5 Hz, 2H), 7.87 (s, IH), 7.60 (s, IH), 7.41 (t, J =8.5 Hz, 2H), 4.92 (q,
0.
520.1 J- 6.0 Hz, IH), 4.17-4.24 (m,
BocHN—'
4H),3.82 (d,J=5.5 Hz, 2H), 3.41 (s, 3H), 2.98 (t,J= 12.5 Hz, IH), 2.84 (d, J =4.5 Hz, 3H), 1.65 (d, J =6.5 Hz, 3H)
8.54 (q, J =3.5 Hz, IH), 7.88 (d, J=8.5Hz,2H), 7.80 (s, IH), 7.61 (s, IH), 7.30 (t,J=9.0 Hz, 2H), 7.17- 7.19 (m, 2H), 7.13
VOH 612.2 (d, J= 9.0 Hz, 2H), 4.93 (q, J = BocHN-'
o P 6.5 Hz, IH), 4.24-4.30 (m, 2H),
3.76 (s, 2H), 3.36 (s, 3H), 3.00- 3.03 (m, 1H),2.86 (d,J=4.0 Hz, 3H), 1.65 (d,J = 6.5 Hz, 3H)
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000105_0001
Figure imgf000106_0001
Figure imgf000107_0001
J= 6.5 Hz, 3H)
8.62 (bra, 3H), 8.55 (q, J =4.5 Hz, IH), 7.97 (dd, J = 9.0 Hz, J2 = 5.0 Hz, 2H), 7.89 (s, IH), 7.59 (s, lH),7.42(t,J=9.0 Hz, 2H), 7.25 - 7.31 (m, 5H), 4.89 (q,J= 6.0 Hz, IH), 4.28 (t,J = 6.5 Hz, lH),4.16(d,J = 10.0 Hz, 2H),
610.2 4.08 (dd,J; = 12.0 Hz, J2= 6.5
Hz, 2H),3.92(d,J = 15.0 Hz, 1H),3.41 (s, 3H),3.19(dd,J,= 14.0 Hz, J2 = 5.5 Hz, 1H),3.50 (dd, J/ = 14.0 Hz, J; = 7.5 Hz, IH), 2.85 (d,J=5.0 Hz, 3H), 2.85 (m,lH), 1.63 (d,J=6.5 Hz,3H)
8.53 (q,J = 4.5 Hz, IH), 8.27 (brs, 2H), 7.95 (dd, J, = 8.8 Hz, J2 = 5.0Hz,2H), 7.88 (s, IH), 7.59 (s, 1H),7.41 (t,J = 9.0 Hz, 2H), 6.61 (d,J=4.5 Hz, IH), „NHBoc
~OH 564.2 4.91 (q,J=6.0 Hz, IH), 4.17- 4.25 (m, 4H), 4.04 (m, IH), 3.95 (m, 1H),3.40 (s,3H), 2.98 (m, lH),2.84(d,J=4.5 Hz,3H), 1.63 (d, J = 7.0 Hz, 3H), 1.20 (d, J= 6.5 Hz, 3H)
im 8.53 (q, 7 = 4.5 Hz, IH), 8.46
(brs, 2H), 7.94 - 7.97 (m, 2H), 7.88 (s, IH), 7.59 (s, IH), 7.41 (t,J=9.0Hz,2H),5.60 (brs,
550.2 IH), 4.92 (q,J=6.0 Hz, IH),
Figure imgf000109_0001
4.13 -4.24 (m5H), 3.79 (m, 2H), 3.41 (s, 3H), 2.98 (m, IH), 2.84 (d, J= 4.5 Hz, 3H), 1.64 (d, J =6.5 Hz, 3H)
8.54 (m,4H), 7.96 (dd,Jj =9.0 Hz,y2 = 5.0 Hz, 2H), 7.89 (s, IH), 7.60 (s, lH),7.42(t,J=9.0 Hz, 2H), 4.92 (q,J=6.0Hz, IH), 4.36 (d,J= 11.5 Hz, IH),
\ NHBoc
562.2 4.20-4.22 (m, 2H),3.91 -4.12
(m, 5H), 3.42 (s, 3H), 2.98 (m, IH), 2.85 (d,J=4.5 Hz, 3H), 2.18(m, IH), 1.63 (d, J =6.5 Hz,3H), 0.98 (d, J = 7.0 Hz, 3H), 0.96 (d,J=7.0 Hz, 3H)
8.61 (brs,3H), 8.54 (m, IH), 7.97 (dd, J; = 8.5 Hz,J2=5.5 Hz,2H), 7.88 (s, 1H),7.60 (s, HBoc lH),7.42(t,J=8.0 Hz), 2H),
564.2 4.94 (m, IH), 4.17-4.36 (m,
-" "°"
5H), 3.74 (m, 2H), 3.55 (s, 3H), 3.41 (s, 3H), 2.97 (m, IH), 2.85 (d,J=5.0 Hz, 3H), 1.65 (d,J= 6.5 Hz)
Figure imgf000110_0001
|0209| Synthesis of compound (S$,7¾-3Sa-lb, Refer to Scheme 35a. To a solution of compound (5S,7S)-34-3b (60 mg, 0.1 1 mmol), DMAP ( 10 mg) and EtjN (0.5 mL) in DCM (2 mL) at 0 °C was added dihydrofuran-2,5-dione ( 108 mg, 1.1 mmol). After stirring at rt for 2 hr, the reaction mixture was concentrated and the residue was purified by preparative HPLC to give compound (5S,7.S)-35a-lb (40 mg, 56.5% yield) as a white solid. LC-MS (ESI): m/z 655.1 [M + Hf; Ή NMR (500 MHz, cADMSO): δ 12.29 (br , I H), 8,51 (q, J = 5.5 Hz, I H), 7.92 (t, J - 4.5 Hz, 2H), 7.84 (s, 1 H), 7.57 (s, 1 H), 7.30 (t, J = 8.5 Hz, 2H), 7.20 - 7.17 (m ,2H), 7.12 (d, J - 8.0 Hz, 2H), 4.90 (q, J = 6.5 Hz, 1 H), 4.12 4.15 (m, 2H), 4.04 (s, 2H), 3.39 (s, 3H), 2.90 (t, J = 4.5 Hz, I H), 2.84 (d , J - 4.0 Hz, 3H), 2.46 - 2.50 (m, 4 H), 1.63 (d, J « 6.5 Hz, 3H) ppm.
|0210| Synthesis of compound (5$, 7$) -35a- la. Following the same procedure as that used to prepare compound (5S,7S)-35a-lb and replacing compound (5S,7S)-34-3b with (5S,7S)-34-3a, compound (55,75>35a-la was obtained as a white solid. LC-MS (ESI): m/z 563 1 [M + Hf; Ή NMR (500 MHz, tADMSO): δ 8.55 (q, J = 5.0 Hz, I H), 7.97 (dd, J, - 9.0 Hz, J> = 5 5 Hz, 2H), 7.86 (s, I H), 7.59 (s, I H), 7.41 (t, J= 9.0 Hz, 2H), 4.90 (q, J - 6.5 Hz, I H), 4.13 4.16 (m, 2H), 4.40 (m, 2H), 3.39 (s, 3H), 2.89 - 2.94 (m, 1 H), 2.84 (d, J - 4 5 Hz, 3H), 2.43 2.48 (m, 4H), 1.63 (d, J = 6.0 Hz) ppm.
Figure imgf000110_0002
(SS,7S 38b-2 R = 4-F-PhO- im |021 11 Synthesis of compound (5_S,7.S)-35b-2a. Refer to Scheme 35b. To a stirred solution of compound (5S,7S)-34-3a (60 mg, 0.13 mmol) in THF (5 mL) was added Ι ,Γ- carbonyldiimidazole (CDl) (63 mg, 0.39 mmol) at rt. After stirring at rt overnight, compound (5S,75 -34-3a was completely converted to compound (5S,7-V)-35b-la determined by LC-MS. Subsequently, l ,4'-bipiperidine (327 mg, 1.95 mmol) was added into the reaction mixture. After stirring at rt for 10 hrs, the reaction mixture was concentrated and the residue was dissolved in DCM (25 mL). The mixture was washed with brine and dried with anhydrous Na2S0 . The solvent was removed and the residue was purified by preparative HPLC to give compound (5S,7S)-3Sb~2a (50 mg, 59% yield) as a white solid. LC-MS (ESI): m/z 657.1 [M + Hj+; ! H NMR (500 MHz, ( -DMSO): δ 8.52 (q, J = 4.0 Hz, 1 H), 7.95 - 7.98 (m, 2H), 7.84 (s, 1 H), 7.85 (s, 1 H), 7.40 (t, J - 8.5 Hz, 2H), 4.90 (q. J = 6.5 Hz, 1 H), 3.95 - 4.14 (m, 6H), 3.38 (s, 3H), 2.91 (br, 1 H), 2.84 (d , 7 = 5.0 Hz, 3H), 2.80 (br, 2H), 2.37 (br, 5H), 1.62 1 68 (m, 5H), 1.27 - 1.46 (m, 8H) ppm.
Figure imgf000111_0001
(5S7S)-3 -3a R = F <SS.7S>-36a-1a R - F
(5S.7S)-34-3b R » 4-F PhO (SS,7S»-3«a-1b R = 4-F-PhO-
|02I2| Synthesis of compound (&S,7S)-36a-lb Refer to Scheme 36a. To a solution of compound (5S,7S)-34-3b (40 mg, 0.0724 mmol), and NaOH (0.5 mL, 40% (wAv) in H20) in acetone (3 mL) was added dimethyl sulfate (0.6 mL) at rt. After stirring at rt overnight, the reaction mixture was concentrated and the residue was diluted with DCM (50 mL). The organic layer was washed with brine and dried over anhydrous Na2SCV The solvent was removed that the residue was purified by preparative HPLC to give compound (5S,75)-36a-lb (20 mg, 49% yield) as a white solid. LC MS (ESI): m z 569.2 [M + Hf ; ! H NMR (500 MHz, i -DMSO): δ 8.50 (q, J 4.5 Hz, 1 H), 7 92 (d, J ÷ 15 0 Hz, 2H), 7 82 (s, I H), 7 56 (s, I H), 7.29 (t, J= 17.5 Hz, 2H), 7.17 7.20 (m, 2H), 7.12 (d, J 17.5 Hz, 2H), 4.89 (q, J - 6.5 Hz, I H), 4.13 (d, J = 10.5 Hz, I H), 4.07 (t, J - 4.5 Hz, I H), 3.37 3.40 (m ,1 H), 3.37 (s, 3H), 3.27 - 3.30 (m, I H), 3.25 (s, 3H), 2.84 (d, J 4.5 Hz, 3H), 1.62 (d, J 6.5 Hz, 3H) ppm
|0213| Synthesis of compound (5S,7S)-36a-la. Following the same procedure as that used to prepare compound (5S,7S)-36a-lb and replacing compound (55,7S)-34~3b with (55,75)-34-3a, compound (5S,7S)-36a-la was obtained as a white solid. LC-MS (ESI): m/.r 477.1 [M + Hf; Ή NMR (500 MHz, </-D SO): δ 8,52 (q,J~ 3.5 Hz, S H), 7.97 (dd, Ji■■■■ 8.8 Hz, J2 ==· 5,5 Hz, 2H), 7.84 (s, IH), 7.59 (s, IH), 7.40 (t, J = 8.0 Hz, 2H), 4.90 (q, J :: 6.0 Hz, I H), 4,07 4,16 (m,2H), 3.39 (s, 3H), 3.27 - 338 (m, 2H), 3.27 (s, 3H), 2.84 - 2.88 (m, I H), 2.84 (d, J - 4.5 Hz, 3H), 1.62 (d,J= 6.5 Hz, 3H) ppm.
Figure imgf000112_0001
|0214| Following either Approach A or B described in Scheme 36b, the following analogs were
Figure imgf000112_0002
in
Figure imgf000113_0001
Figure imgf000114_0001
Figure imgf000115_0001
Π4
Figure imgf000116_0001
Figure imgf000117_0001
IH), 4.06 (s,2H),3.58~
3.43 (m,2H),3.37(s,3H),
3.30-3.27(m, iH), 300- 2.92(oi, 4H), 2,84 (d,J; 4.5 Hz, 3H), 3.78 (s, 2H), 1.62 (d,J « 60 Hz, 3H)
B 546.2 8.52 (q, 7 = 4.5 Hz, iH), prepared in u
7.97 (dd, J! - 6.5 Hz, J2 =
9.0 Hz, 2H), 7.83 (s, IH), obiained b ireaiing ihe
7.59 (s, IH), 7,40 (t, 7 = 9.0 compound shown in Enir 1
wish aq. HCHO in ihe Hz,2H), 4.89 (q,J = 6.5 Hz, presence of NaBHjCN and IH), 4.13(d,J = 13.5 Hz,
AcOH in eOH IH), 4.04 - 4.02 (m, IH),
3.99-3.94(01, IH), 340
3 Ί*j "IQ (m ill, \ i H ΓΪ¾ j 1.j Πf icS, U\
3.32 ~ 3.29 (m,2H), 2.90-
2.88 (m, IH), 2.84 (d,J- 5
Hz, 3H), 2.57 ~2.52 (m,
IH), 2.43 2.41 (m, IH),
2.30- 2.26 (m, IH), 2.20 {s,
3H), 2.00- 1.93 (m, IH), i.62 {d,J 65 Hz,3H)
B 546.2 8.52- 8.50 (m, IH), 7.97
(dd,Ji -6,0 Hz,J2 85 ob!ained by ireaiing ihe Hz,2H), 7.83 (s, iH), 7.59 compound shown in Eniry 13 (s, IH), 7.40 (t,J 8,5 Hz, wiJh eOC(0)CI in ihe 2H), 4.89 {q, J =* 60 Hz, presence of Ei3N in DCIVI
iH), 4.i3{d,J-9.5 Hz, !H), 4.06 (s, IH), 3.57 (s,
Figure imgf000119_0001
Scheme 33c
Figure imgf000119_0002
|021S| Step 1. Refer to Scheme 36c. To a stirred solution of compound (5S,75)-36b-3a (50 mg, 0.08 mmol) in dry DMF (2 mL) was added Cs2C03 (26 mg, 0, 16 mmol) and 2 -methyl- 1 H~ imidazole ( 13 mg, 0.16 mmol). After stirring at 50 °C for 12 hrs, the reaction mixture was concentrated. The residue was diluted with water (25 mL) and the resulting suspension was extracted with EtOAc (20 mL x 3). The combined organic extracts were washed with brine (20 mL), dried over anhydrous NajS0 , and concentrated. The residue was dried in vacuo to give crude compound (55,7 ?)-36c-l (50 mg) as light yellow solid, which was used directly for the next step without further purification LC MS (ESI) m z 542 2 [M + Hf.
|0216| Step 2. Following the same procedure as that used for converting compound 1-14 to 1-15 shown in Scheme 1 (WO2012/058125) and replacing compound 1-14 with (5S,7/?)-36c-l, compound (55,7 ?)-36c-2 was obtained as a light yellow solid. LC-MS (ESI): m/z 514.1 [M + Hf . |0217| Step 3. Following the same procedure as that used for converting compound 1-15 to 1-16 shown in Scheme 1 and replacing compound 1-15 with (5i',7/?)-36c-2, compound (5S,7/?)-36c-3 was obtained as a white solid. LC-MS (ESI): m/z 527.2 [M + Hf; Ή NMR (500 MHz, tt- DMSO); S 8.53 (q, J = 4.5 Hz, 1 H), 7.96 (t, J - 4 5 Hz, 2H), 7.84 (s, 1 H), 7.56 (s, I H), 7.40 (t, J 8.5 Hz, 2H), 7.07 (s, 1 H), 6.74 (s, 1 H), 4 86 (q, J■« 6.5 Hz, 1 H), 4.23 - 4.16 (m, 2H), 4.08 (d, J - 18.0 Hz, 1 H), 3.90 (dd, J, 15.0 Hz, J? 7 0 Hz, I H), 3.51 (s, 3H), 2.84 - 2.79 (m, I H), 2.79 (s,
(d, J - 7.0 Hz, 3H) ppm
Figure imgf000120_0001
(5S,7 38s-4
|0218| Synthesis of compound (5£,7i?)-36c-4. Following the same procedure as that used for preparing compound (5S,7/?)-36c-3 shown in Scheme 36c and replacing 2-methyl-lH-imidazole with I H-pyrazole, compound (5S,7,R)-36c-4 was obtained as a white solid. LC-MS (ESI): m/z 513.2 [M + Hf; Ή NMR (500 MHz, i/-DMSO): 6 8.51 (q, J - 5.0 Hz, I H), 7.98 - 7.95 (m, 2H), 7.84 (s, I H), 7.66 (s, I H), 7.57 (s, IH), 7.45 (s, I H), 7.40 (t, J 8.5 Hz, 2H), 6.23 (s, I H), 4.86 (q, J = 6.0 Hz, 1 H), 4.26 - 4.12 (m, 4H), 3.36 (s, 3H), 2.84 (d, J - 5.0 Hz, 3H), 1 .54 (d, 7 - 6.5 Hz, 3H) ppm.
Figure imgf000120_0002
|0219| Synthesis of compound (55,7i?)-36c-5. Following the same procedure as that used for preparing compound (55,7 ?)-36c-3 shown in Scheme 36c and replacing 2-methyl-S //-imidazole with pyrrolidin-2-one, compound (5S,7/?)-36c-5 was obtained as a white solid. LC-MS (ESI): /z 530.2 [M + Hf; Ή NMR (500 MHz, i -DMSO): δ 8.52 (q, J 4.5 Hz, 1 H), 7.97 (dd, J, 5.5 Hz, J2 = 8.5 Hz. 2H), 7.85 (s, I H), 7.59 (s, IH), 7.40 (t, J 8.5 Hz, 2H), 4.88 (q, J 6.5 Hz, I H), 4.08 4 06 (m, 2H), 3 52 - 3.47 (m, 2H), 3.38 (s, 3H), 3.10 - 3.06 (m, 2H), 2.84 (d, J = 4.5 Hz, 3H),
2 1 (m, 2H), 1.94 - 1 .89 (m, 2H), 1.62 (d, J = 6.0 Hz, 3H) ppm.
Figure imgf000121_0001
|0220| Synthesis of compound (5S,7/f)-36c-6. Following the same procedure as that used for preparing compound (5S R) 36c-3 shown in Scheme 36c and replacing 2-methyl- 1 H-imidazole with oxazo!idin-2-one, compound (55,7/?)-36c-6 was obtained as a white solid. LC-MS (ESI): mlz 532.1 [M + H] ; Ή NMR (500 MHz, -DMSO): δ 8.5 1 (q, J - 4.5 Hz, 1 H), 7.97 (dd, J, = 4.5 Hz, j2 = 7.5 Hz, 2H), 7 85 (s, I H), 7 59 (s, 1 H), 7.40 (t, J, = 8.5Hz, 2H), 4.90 (q, J - 6.5 Hz, 1 H), 4.28 ~ 4.24 (m, 2H), 4.13 - 4.1 1 (d, J « 9.5 Hz, 2H), 3.70 - 3.68 (m, 2H), 3.57 - 3.55 (m, 2H), 3.39 (s,
2.84 (d, J = 4.5 Hz, 3H), 1 .63 (d, J - 6.0 Hz, 3H) ppm.
Figure imgf000121_0002
|0221 | Synthesis of compound (&S,7/f)-36c-7, Following the same procedure as that used for preparing compound (5S,7 ?)-36c-3 shown in Scheme 36c and replacing 2-methyl- 1 H-imidazole with 1 ,3-oxazinam 2 one, compound (55,7/?)-36c-7 was obtained as a white solid. LC-MS (ESI): mlz 546.2 [M + Η] ; Ή NMR (500 MHz, J3 DMSO): δ 8.52 (q, J = 4.5 Hz, I H), 7.97 (dd, J, - 5.5 Hz, J2 - 8.5 Hz, 2H), 7.84 (s, 1 H), 7.59 (s, 1 H), 7.39 (t, J = 8.5 Hz, 2H), 4.90 (q, J - 6.5 Hz, 1 H), 4.21 - 4.05 (m, 4H), 3.48 - 3.40 (m, 4H), 3.38 (s, 3H), 3 08 - 3.01 (br, I H), 2.84 (d, J ~ 4 5
(d, J 6.0 Hz, 3H) ppm.
Figure imgf000121_0003
|0222| Synthesis of compound (5S,7iV)-36c-8. Following the same procedure as that used for preparing compound (5_?,7i?)-36c-3 shown in Scheme 36c and replacing 2-methyl- 1 H-imidazole with morpholin 3-one, compound (55,7 ?)-36c-8 was obtained as a white solid. LC-MS (ESI): m! 546,2 [M + H]+; Ή NMR (500 MHz, tt DMSO): δ 8.53 (q, J 4.5 Hz, I H), 7.97 (dd, Jj - 5.5 Hz, J2 - 8.5 Hz, 2H), 7.87 (s, I H), 7.60 (s, 1 H), 7.40 (t, J 8.5 Hz, 2H), 4.90 (q, J - 6.5 Hz, I H), 4.18 4.01 (m, 4H), 3.81 ~ 3.78 (t, J- 45 Hz, 2H), 3.61 356 (m, 1H), 3.52 - 3.48 (m, 2H), , 1 H), 284 (d, J - 45 Hz, 3H), 1.62 (d, J = 6.0 Hz, 3H) ppm.
Figure imgf000122_0001
| 2231 Synthesis of compound (5S,7i?)-36c-9. Following the same procedure as that used for preparing compound (55,7?) 36c-3 shown in Scheme 36c and replacing 2-methyl- 1 H-imidazole with piperidin-2-one, compound (5S,7/?)-36c-9 was obtained as a white solid. LC-MS (ESI) miz
5442 [M + Hf ; Ή NMR (500 MHz, (t DMSO): δ 8.52 (q, J·»■ 4.5 Hz, 1H), 7.97 (dd, J\ - 5.5 Hz, Ji - 8.5 Hz, 2H), 7.85 (s, IH), 7.58 (s, IH), 7.39 (t,J=8.5 Hz, 2H), 4.86 (q, J -6.5 Hz, 1H),413 4.05 (m, 2H), 361 355 (m, IH), 3.44 3.41 (m, 2H), 3.36 (s, 3H), 3.02-2.90 (m, IH), 2.84 - 4.5 Hz, 3H), 224 - 2.21 (m, 2H), 1.74- I 61 (m, 4H), 1.60 (d, J - 6.0 Hz, 3H) ppm.
Figure imgf000122_0002
|0224| Synthesis of compound (5S,7/r 36c-10. Following the same procedure as that used for preparing compound (5S,7?)-36e-3 shown in Scheme 36c and replacing 2-methyl- 1 H-imidazole with 1 , 1 -dioxo-isothiazolidme, compound (5S,7/?)-36c-10 was obtained as a white solid LC MS (ESI): miz = 566.1 [M + Hf ; lH NMR (500 MHz, ./-DMSO): δ 8.51 (q,J 45 Hz, IH), 7.97 (dd, Ji = 5.5 Hz, J2 - 8.5 Hz, 2H), 7.85 (s, 1 H), 7.59 (s, 1 H), 7.40 (t, J = 90 Hz, 2H), 4.90 (q, J « 6.5 Hz, IH), 4.20 -4.10 (m, 2H), 3.38 (s, 3H),3.21 -3.11 (m, 4H), 3.10 3.02 (m, 1H),2.89
- 4.5 Hz, 3H), 2.25 -2.18 (m, 2H), 1.65 (d, J - 6.0 Hz, 3H) ppm.
Figure imgf000122_0003
|0225| Synthesis of compound (5S R)-36c-\l. Following the same procedure as that used for preparing compound (55,7/?)-36c-3 shown in Scheme 36c and replacing 2-methyl- 1 H imidazole with 1 , l-dioxo-tetrahydro-2H- 1 ,2-thiazine, compound (55,7?)-36c-l 1 was obtained as a white solid. LC-MS (ESI): miz = 580.2 [M + Hj+;Ή NMR (500 MHz, i^-DMSO): δ 8.51 (q, J 45 Hz, IH), 7.97 (dd, J, - 5.5 Hz, J2 - 8.5 Hz, 2H), 7.85 (s, 1 H), 7.59 (s, IH), 7.40 (t, J - 90 Hz, 2H), 4.90 (q, J 6,5 Hz, 1 H), 4.14 ~ 4 07 (m, 2H), 3.43 - 3 40 (m, 2H), 3.37 (s, 3H), 3.16 - 3 06 (m, 4H), 2.85 (d, J 4,5 Hz, 3H), 2 84 (m, S H), 2 0 i (m, 2H), 1.64 (d, J - 6.0 Hz, 3H), 1 54 {m, 2H) ppm.
Figure imgf000123_0001
|0226| Step 1. Refer to Scheme 36d. To a stirred solution of compound (5S,7S)-36b-3b ( 1 10 mg, 0.18 mmoi) in DMF (3 mL) at 65 °C was added NaNj ( 140 mg, 2.15 mmol) in portions After stirring at 65 °C for 4 hrs, the reaction mixture was cooled to rt and diluted with water (20 mL) and EtOAc (60 mL). The organic layer was washed with saturated aq. NaHCOj (20 mL), dried over anhydrous Na2SCXi, and concentrated. The residue was purified by silica gel column chromatography to give compound (5S,7 ?)-36d-l (71 mg, 81 % yield) as a white solid. LC-MS (ESI): mlz 525.1 [M + Na]+.
|0227| Step 2. To a solution of trimethylsilylacetylene (85 μί, 0.597 mmol) and compound (5S,7/?)-36d-I ( 100 mg, 0.2 mmol) in DMF (5 mLVH20 (2 mL) was added Oil ( 1 14 mg, 0.60 mmol) and Et¾N (61 mg, 0.60 mmol). After stirring at rt for 24 hrs, the reaction mixture was diluted with water (30 mL). The resulting mixture was extracted with EtOAc (50 mL x 3). The combined organic extracts were washed with water (50 mL) and brine (50 mL), dried over anhydrous NaiSttj, and concentrated. The residue was dried in vacuo to give crude compound (55,7/?)-36d-2 (91 mg, 76% yield) as a white solid, which was used directly for the next step without further purification. LC-MS (ESI): mlz 601.2 [M + Hf ,
|0228| Step 3. Following the same procedure as that used for converting compound 1-14 to 1-15 shown in Scheme 1 (WO2012/058125) and replacing compound 1-14 with (5S,7/?)-36d-2, compound (5S,7 ?)-36d-3 (66 mg, 70%) was obtained as a white solid. LC-MS (ESI): miz 573.2 [M + Hf .
|0229| Step 4. Following the same procedure as that used for converting compound 1-15 to 1-16 shown in Scheme 1 (WO2012/058125) and replacing compound 1-15 with (5S,7/?)-36d-3, compound (55,7 ?)-36d-4 (56 mg, 83% yield) was obtained as a white solid. LC-MS (ESI): miz 586.2 [M + Hf.
|0230| Step 5. A solution of compound (5S,7/?)-36d-4 (56 mg, 0.1 mmo!) in 4 N HC1 in 1 ,4- dioxane (5 mL) was stirred at 60 °C for 4 hrs. Subsequently, the reaction mixture was concentrated and the residue was purified by preparative HPLC to give compound (5-?,7 ?)-36d-5 (5.5 mg, 1 1% yield) as a white solid. LC-MS (ESI): miz 514.1 [M + H]+; Ή NMR (500 MHz, ct- DMSO): 6 8.51 (q, J = 4.5 Hz, 1 H), 8.05 (s, 1 H), 7.96 (dd, J, - 5.5 Hz, J2 = 9.0 Hz, 2H), 7.85 (s, 1 H), 7.73 (s, I H), 7.57 (s, 1 H), 7.40 (t, J - 9.0 Hz, 1 H), 4.87 (q, J - 6.5 Hz, I H), 4.62 (d, J - 1 1.0 Hz, 1 H), 4.39 - 4.23 (m, 3H), 3.38 (s, 3H), 2.88 - 2.85 (m, 1 H), 2.83 (d, J « 4,5 Hz, 3H), 1.52 (d, J - 6.0 Hz, 3H) ppm.
Figure imgf000124_0001
102311 Step 1. Refer to Scheme 36e. To a stirred solution of compound (5S,7S)-34-3a (2.0 g, 4.3 mmol), DMAP (52 g, 0.43 mmol) and Et3N ( 1 mL, 8.3 mmol) in DCM (40 mL) at 0 °C was added a solution of TsCI ( 1.24 g, 6.5 mmol) in DCM (20 mL). After stirring at rt overnight, the reaction mixture was quenched by adding ice-water (50 mL). The organic layer was separated and washed with saturated aq. NaHCOi solution (20 mL x 3) and water (20 mL x 3), dried over anhydrous Na2SC>4, and concentrated. The residue was purified by silica gel column chromatography
(Petroleum ether/EtOAc = 2/1 (v/v)) to give compound (55,75)-36e-l (1 .8 g, 68% yield) as a light yellow solid. LC-MS (ESI): m/z 617.1 [M + Hf.
102321 Step 2. To a stirred solution of compound (5S,7S)-36e-l (1 .8 g, 3.0 mmol) in DMF (40 mL) at 65 °C was added NaNj (0.6 g, 9.0 mmol) portion-wise. After stirring at 65 UC for 2 hrs, the reaction mixture was cooled to rt and added ice- water (50 mL). The mixture was extracted with EtOAc (50 mL * 3). The combined organic extracts were washed with saturated aq. NaHCOj solution ( 10 mL x 3) and water ( 10 mL x 3), dried over anhydrous Na2S04, and concentrated. The residue was dried in vacuo to give crude compound (55,7/?)-36e-2 ( 1 .42 g, quantitative yield) as a white solid, which was used directly for the next step without further purification. LC-MS (ESI): m/z- 488.1 [M + Hf.
102331 Step 3. To a stirred solution of compound (5S,7/?)-36e-2 ( 1.42 g, 3.0 mmol) in THF (40 mL) and H20 (2 mL) was added PPhj ( 1.57 g, 6 mmol) at rt under an atmosphere of N2, After refluxing for 4 hrs, the reaction mixture was cooled to rt and added 1 .0 N aq. HCl solution (6 mL). The resulting mixture was stirred at rt for 1 hr and concentrated. The residue was diluted with water (20 mL) and extracted with hexane (20 mL x 3). Subsequently, the aq. phase was treated with 1 .0 N aq. NaOH solution to adjust the pH value > 9.0. The basic suspension was extracted with EtOAc (25 mL x 3). The combined organic extracts were dried with anhydrous Na2S04 and concentrated. The residue was dried in vacuo to give compound (55,7/?)-36e-3 (810 mg, 60% yield) as a white solid. LC-MS (ESI): m/z 462.1 [M + Hf.
102341 Step 4. To a stirred solution of 4-chlorobutanoic acid (74 mg, 0.6 mmol) in DMF (2 mL) was added HATU (456 mg, 1 .2 mmol). After stirring at rt for 30 min, the mixture was added DIPEA ( 193 mg, 1 .5 mmol) and compound (55,7/?)-36e-3 ( 192 mg, 0.4 mmol). The resulting reaction mixture was stirred at rt for 3 hrs and then poured into water (20 mL). The suspension was filtered; the solid was washed with water ( 10 mL x 2) and dried in vacuo to give crude compound (5S,7/?)-36e-4 (180 mg, 63% purity based on LC-MS) as a white solid, which was used directly for the next step without further purification. LC-MS (ESI): mlz 566.1 [M + H] . |0235| Step 5. To a stirred solution of compound (5S,7/?)-36e-4 ( 180 mg, 0.31 mmol) in dry THF (50 mL) was added 2CO1 (483 mg, 3.5 mmol). After re fluxing for 6 hrs, the reaction mixture was cooled to rt and filtered. The filtrate was concentrated and the residue was purified by preparative HPLC to give (5£,7i?)-36e-5 (28 mg, 17 yield) as a white solid. LC-MS (ESI): mlz 530.2 [M + Hf.
|0236| Synthesis of compound (&S,7 ?)-36e-6. To a stirred solution ofCDI ( 162 mg, 1.0 mmol) in DMF (2 mL) was added compound (55,7Λ)-36β-3 (92 mg, 0.2 mmol) portion-wise at rt. After stirring at 60 UC for 3 hrs, the reaction mixture was added 3-ch!oro- 1 -propylamine HC1 salt (260 mg, 2.0 mmol). The reaction mixture was stirred at 60 °C overnight and then concentrated. The residue was diluted with water (20 mL) and EtOAc (20 mL) and the aqueous layer was extracted with EtOAc (20 mL x 3). The combined organic extracts were washed with brine (25 mL), dried over anhydrous
Figure imgf000126_0001
and concentrated. The residue was dried in vacuo to give crude compound (5S,7/?)-36e-6 (99 mg, 86% yield) as a white solid, which was used directly for the next step without further purification. LC-MS (ESI): mlz 581.2 [M + Hf.
|0237| Synthesis of compound (&_»,7i?)-36e-7. To a solution of compound (5S,7/?)-36e-6 (99 mg, 0.17 mmol) in DMF (6 mL) was added 2C03 (71 mg, 0.51 mmol). After stirring at 80 UC overnight, the reaction mixture was cooled to rt, diluted with EtOAc (20 mL), and filtered through C elite* 545. The filtered cake was washed with EtOAc (20 mL x 2). Subsequently, the filtrate was washed with H2O (20 mL x 3) and brine (20 mL), dried over anhydrous Na2S04, and concentrated. The residue was purified by preparative HPLC to give compound (5S,7/?)-36e-7 (35 mg, 38% yield) as a white solid. LC-MS (ESI): mlz 545.2 [M + Hf; Ή NMR (400 MHz, c ~ DMSO): δ 8.52 (q, J = 5.5 Hz, 1 H), 7.96 (dd, J, =1 1.0 Hz, J2 = 7.0 Hz, 2H), 7.84 (s, 1 H), 7.58 (s, 1 H), 7.40 (t, J = 1 1.0 Hz, 2H), 4.87 (q, J = 7.5 Hz, 1 H), 4.28 - 3.98 (m, 4H), 3.36 (s, 3H), 3 35 (s, 1 H), 3.23 (s, 2H), 3.05 (d, J - 17.0 Hz, 2H), 2.84 (d, J = 5.5 Hz, 3H), 2.81 - 2.76 (m, 1 H), 1 86 (br, 2H), 1.63 (d, J = 9.0 Hz, 3H) ppm.
Figure imgf000126_0002
|0238| Step 1. Refer to Scheme 36f. A solution of compound (5S,7S)-36e-l (62 mg, 0.1 mmol) in 70% EtNH2 in water (5 mL) was stirred at 80 °C in a microwave reactor for 2 hrs. The reaction mixture was concentrated and the residue was diluted with water (30 mL). Subsequently, the resulting mixture was extracted with EtOAc (20 mL x 3); the combined organic extracts were dried over anhydrous NaiSO* and concentrated. The residue was dried in vacuo to give crude compound (5S,7/?)-36f-l (35 mg, 72% yield) as a white solid. LC-MS (ESI): mlz 490.2 [M + Hf. |0239| Step 2. To a solution of compound (5S,7/?)-36f-l (35 mg, 0.07 mmol) in DCM (5 mL) and Eti ( i mL) was added methyl chloroformate (6.7 mg, 0.07 mmol) at 0 °C. After stirring at rt overnight, the reaction mixture was added several drops of saturated aq. NaHCOj solution.
Subsequently, the mixture was concentrated and the residue was added water ( 15 mL). The resulting mixture was extracted with EtOAc ( 15 mL x 3); the combined organic extracts were dried over anhydrous Na2S0 and concentrated. The residue was purified by preparative HPLC to give compound (5S,7/?)-36f-2 (7 mg, 18% yield) as a white solid. LC-MS (ESI); mlz 548.2 [M + Hf; Ή NMR (500 MHz, c/-DMSO): 8 8.52 (q, J= 4.0 Hz, 1 H), 7.97 (dd, J, = 8.0 Hz, J2 » 8.5 Hz, 2H), 7.85 (s, 1 H), 7.59 (s, 1 H), 7.41 (t, J= 9.0 Hz, 2H), 4.89 (q, J = 6.5 Hz, I H), 4.10 - 4.06 (m, 2H), 3.60 (s, 3H), 3.45 - 3.38 {m, 4H), 3 12 3,09 (br, I H), 2 85 (d, J 5.5 Hz, 3H), 2 85 2.79 (m, 1 H), 1.62 (d, J- 6.5 Hz, 3H), 1.06 (t, J 7.0 Hz, 3H) ppm.
Figure imgf000127_0001
|0240| Synthesis of compound 36g-2 Refer to Scheme 36g To a stirred solution of CDI (300 mg, 1 .85 mmol) in dry DMF (5 mL) was added (/?)-tetrahydrofuran-3-ol ( 163mg, 1.85 mmol) at rt. After stirring at rt for 4 hrs, the reaction mixture was added compound (5-?,7 ?)-36e-3 (47 mg,
0.1 mmol) and TEA ( 1 mL, 6.6 mmol). The resulting reaction mixture was stirred at 60 °C overnight and then concentrated. The residue was diluted with water (20 mL) and extracted with
EtOAc (20 mL x 3). The combined organic extracts were washed with water (25 mL) and brine
(25 mL), dried over anhydrous Na2S04, and concentrated The residue was purified by
preparative HPLC to give compound 36g~2 (12 mg, 21%) as a white solid. LC MS (ESI): mlz
576.2 [M + Hf; f H NMR (500 MHz, DM SO): 8 8 52 (q,J 5.0 Hz, 1 H), 7 96 (dd, J, »= 5 5 Hz, J2 = 9.0 Hz, 2H), 7.84 (s, I H), 7.59 (s, 1 H), 7 40 (t, J - 9 0 Hz, 2H), 7.28(t, J - 4.5 Hz, I H), 5.10 (br, 1 H), 4.86(q, J - 5.0 Hz, 1 H), 4.09 (d, J 17 0 Hz , I H), 3 95 - 3.90 (m, 1 H), 3.74 - 3.65 (m, 4H), 3.10 - 2.95 (m, 2H), 2.94 (s, 3H), 2.84 (d, J 4 5 Hz, 3H), 2.65 - 2.60 (m, 1 H), 2.09 - 2.07
-1.85 (m, I H), 1 .60 (d, J 6.5 Hz, 3H) ppm.
Figure imgf000128_0001
|0241 | Synthesis of compound 36g-3. Following the same procedure as that used for preparing compound 36g-2 shown in Scheme 36g and replacing (/?) tetrahydrofuran-3-ol with (S)- tetrahydrofuran-3-ol, compound 36g-3 was obtained as a white solid. LC-MS (ESI): mlz 576.2 [M
+ Hf ; Ή NMR (500 MHz, i -DMSO): δ 8 51 (q, J - 5.0 Hz, 1 H), 7.98 (dd, J, = 5.5 Hz, J2 = 9.0 Hz, 2H), 7.84 (s, I H), 7.58 (s, I H), 7.40 (t, J 9.0 Hz , 2H), 7.27 (s, I H), 5.1 1 (t, J - 6.0 Hz, I H), 4.87(d, J = 5.0 Hz, 1 H), 4.13 (d, J = 17.0 Hz, I H), 3.98 - 3.92 (m, 1 H), 3.77 - 3.65 (m, 4H), 3.36 (s, 3H), 3.09 - 2.97 (m, 2H), 2.84 (d, J = 4.5 Hz, 3H), 2.80 2.73 (m, I H), 2. 15 - 2.05 (m, 1 H), - 1.82 (m, I H), 1.63 (d, J - 6.5 Hz, 3H) ppm.
Figure imgf000128_0002
|0242| Synthesis of compound 36g-4. Following the same procedure as that used for preparing compound 36g-2 shown in Scheme 36g and replacing ( ?)-tetrahydrofuran-3-ol with tetrahydro- 2H-pyran-4-ol, compound 36g-4 was obtained as a white solid LC-MS (ESI): mh 590.2 [M + H]+; 1 H NMR (500 MHz, </-DMSO): δ 8,5 1 (q, J - 5 0 Hz, I H), 7.97 (dd, J, = 5.5 Hz, J2 » 9.0 Hz, 2H), 7.84 (s, 1 H), 7.59 (s, 1 H), 7.40 (t, J - 9 0 Hz , 2H), 7 20 (s, 1 H), 4.87 (q, J 6.0 Hz, 1 H), 4.71 - 4.67 (m, 1 H), 4.12 (d, J ÷ 15.5 Hz, 1 H), 3.96 (m, I H), 3.81 - 3.79 (m, 2H), 3.46 -3 39 (m, 2H), 3.35 (s, 3H), 3.03 - 3.00 (m, 2H), 2.84 (d, J 4 5 Hz, 3H), 2.80 - 2.70 (m, I H), 1 .85™ 1 .82 (m, 2H), 1.62 (d, J - 6.5 Hz, 3H), 1.60 1 .40 (m, 2H) ppm
Figure imgf000128_0003
102431 104321 Step 1. Refer to Scheme 36h. To a stirred suspension of Cs2C03 (63 mg, 0.19 mmol) in dry DMF (2 mL) was added /<?r/-butyl thiazol-2-ylcarbamate (30 mg, 0.14 mmol) at rt under an atmosphere of Ar. The mixture was stirred at rt for 30 min and then (5S,7S)-36e-I (60mg, 0.1 mmol) was added. After stirring at 60 °C overnight, the reaction mixture was concentrated. The residue was diluted with water ( 15 mL) and EtOAc ( 15 mL). The aqueous phase was extracted with EtOAc ( 15 mL x 3). Subsequently, the combined organic extracts were washed with saturated aq. NaHC{¾ solution (20 mL x 2) and brine (20 mL), dried over anhydrous and concentrated. The residue was dried in vacuo to give crude compound (5S,7/?)-36h- 1 , which was used for the next step without further purification. LC-MS (ESI): mlz 645.2 [M +
Hf.
1024 1 Step 2. A mixture of compound (5S,7fl)-36h-l (50 mg, 0.08 mmol) in saturated HC1 in EtOAc (5 mL) was stirred at rt for 5 hrs. The mixture was concentrated and the residue was purified by preparative HPLC to give compound (55,7 ?)-36h-2 (13 mg, 31 % yield) as a white solid. LC-MS (ESI): mlz 545.1 [M + Hf; Ή NMR (500 MHz,
Figure imgf000129_0001
δ 8.5 i (q, J - 4.5 Hz. 1 H), 7.98 - 7.95 (m, 2H), 7.84 (s, 1 H), 7.59 (s, 1 H), 7.60 - 7.57 (m, 1 H), 7.40 (t, J - 9.0 Hz, 2H), 7.00 (s, I H), 6.60 (s, 1 H), 4.90 (q, J 6.5 Hz, 1 H), 4 18 {d, J* 10.5 Hz, S H), 4.12 4J 0 {m, 1 H), 3.39 ~ 3.31 (m, 2H), 3,35 (s, 3H), 2.84 (d, J 4 5 Hz, 3H), 2.89 - 2.84 (m, S H), 1.65 (d, J « 6 5 Hz, 3H) ppm.
Figure imgf000129_0002
102451 Synthesis of compound (§£,7$)-36h-3. Following the same procedure as that used for preparing compound (5S,7/?)-36h-2 shown in Scheme 36h and replacing te/7-butyl thiazol-2- y I carbamate with ten-butyl benzo[i/]thiazol-2-ylcarbamate, compound (5S,7 ?)-36h-3 was obtained as a white solid. LC-MS (ESI): mlz 595.1 [M + Hf, Ή NMR (500 MHz, tA DMSO) δ 8.51 (q, J - 4.5 Hz, 1 H), 8. 10 (t, J 5.0 Hz, 1 H), 7 96 (dd, J, 9.0 Hz, J2 5.0 Hz, 2H), 7.85 (s, 1 H), 7.66 (d, J - 7.5 Hz, 1 H), 7.60 (s, 1 H), 7.42™ 7.37 (m, 3H), 7.22 (t, J *» 7.0 Hz, 1 H), 7 02 (t, J = 7.5 Hz, 1 H), 4.92 (q, J = 6.5 Hz, 1 H), 4.25 4.18 (m, 2H), 3.53 - 3.39 (m, 2H), 3.41 (s, 3H), 2.90 (m, 1 H), 2.84 (d, J = 5.0 Hz, 3H), 1.66 (d, J - 6.0 Hz, 3H) ppm.
Figure imgf000130_0001
|0246| Synthesis of compound (&S,7J?)-36fa-4, Following the same procedure as that used for preparing compound (5S,7 ?)-36h-2 shown in Scheme 36h and replacing terf-butyl thiazol-2- ylcarbamate with ierf-butyl 2-(terf-butoxycarbonylamino)- 1 //-imidazole- 1 -carboxylate, compound (5S,7/?)-36h-4 was obtained as a white solid. LC-MS (ESI): m/z 528.2 [M + Hf; Ή NMR (500 MHz, -DMSO): 5 8.51 (q, J = 5.0 Hz, 1 H), 8.30 (s, 1 H), 7.97 (dd, J, - 9.0 Hz, J2 = 5.0 Hz, 2H), 7.82 (s, I H), 7.59 (s, I H), 7.40 (t, J - 9.0 Hz, 2H), 6.48 (s, 2H), 5.25 (m, 1 H), 4.91 (q, J = 6.5 Hz, 1 H), 4.1 6 (m, 1 H), 4.06 (m, 1 H), 3.35 (s, 3H), 3.21 - 3.12 (m, 2H), 2.88 - 2.84 (m,
= 5.0 Hz, 3H), 1.66 (d, J = 6.0 Hz, 3H) ppm.
Figure imgf000130_0002
|0247| Synthesis of compound (&S,7/?)-36h-5. Following the same procedure as that used for preparing compound (5S,7 ?)-36h-2 shown in Scheme 36h and replacing terf-butyl thiazol-2- ylcarbamate with terf-butyl 1 -methyl- 1 H.imidazol-2-ylcarboxylate, compound (SS,7/?)-36h-5 was obtained as a white solid. LC MS (ESI): m/z 542 2 [M + H]+; Ή NMR (500 MHz, t- DMSO): 6 8.51 (q, J = 5.0 Hz, I H), 8.30 (s, 1 H), 7.97 (dd, J\ - 9.0 Hz, J2 - 5.0 Hz, 2H), 7.83 (s, I H), 7.59 (s, 1 H), 7.40 (t, J 9.0 Hz, 2H), 6 61 (s, 1 H), 6.43 (s, 1 H), 5.25 (m, 1 H), 4.90 (q, J 7.0 Hz, l H), 4.20 (m, I H), 4. I O (m, I H), 3.34 (s, 3H), 3 23 3 19 (m, 2H), 2.88 - 2.84 (m, I H),
= 4.0 Hz, 3H), 1.63 (d, J 7.0 Hz, 3H) ppm.
Figure imgf000130_0003
|0248| Synthesis of compound (5S,7/?)-36h-6 Following the same procedure as that used for preparing compound (55,7 ?)-36h-2 shown in Scheme 36h and replacing terf-butyl thiazol-2- ylcarbamate with terf-butyl 2-(½rf-butoxycarbonylamino)- 1 //-benzo[<7) imidazole- 1 -carboxylate, compound (55,7 ?)-36h-6 was obtained as a white solid. LC MS (ESI): m/z 578.2 [M + Hf ; Ή NMR (500 MHz, -DMSO): 5 8.51 (q, J - 5.0 Hz, 1 H), 8.1 7 (s, 1 H), 7.96 (dd, J, = 9.0 Hz, J2 ¾ 5.0 Hz, 2H), 7.84 (s, 1 H), 7 60 (s, 1 H), 7. 13 (t, J 4.0 Hz, 2H), 6.86 (m, 2H), 6.52 (m, 1 H), 4.93 (q, J™ 7 0 Hz, I H), 4 23 (m, 1 H), 4 15 (m, 1 H), 3.43 - 3.39 (m, 2H), 3.35 (s, 3H), 2.92 - 2.89 fm,
, 1.63 (d, J 6.5 Hz, 3H) ppm
Figure imgf000131_0001
|0249| Synthesis of compound (&S,7 i)-36h-7 Following the same procedure as that used for preparing compound (5S,7 ?)-36h-2 shown in Scheme 36h and replacing rerr-butyl thiazol-2- ylcarbamate with tert butyl pyrimidin-2-ylcarbamate, compound (5S,7/?)-36h-7 was obtained as a white solid. LC-MS (ESI): miz 540.2 [M + H] ', Ή NMR (500 MHz, (/-DMSO): S 8.51 (q, J ~ 4.0 Hz, 1 H), 8.28 (d, J 5.0 Hz, 2H), 7 95 (dd, J/ 9.5 Hz, J2 5.0 Hz, 2H), 7.83 (s, 1 H), 7.59 (s, 1 H), 7.40 (t, J 9.0 Hz, 2H), 7 08 (t, J - 7 0 Hz, 1 H), 6 59 (t, J, - 4.5 Hz, 1 H), 4.90 (q, J = 7.0 Hz, I H), 4.10 - 4.05 (m, 2H), 3.40 (s, 3H), 3.45 - 3.35 (m, 2H), 2.92 - 2.85 (m 1 H), 2.84 (d, J =
- 7.0 Hz, 3H) ppm.
Figure imgf000131_0002
|0250| Synthesis of compound (&S",7J?)-36h-8. Following the same procedure as that used for preparing compound (5S,7 ?)-36h-2 shown in Scheme 36h and replacing reri-butyl thiazol-2- ylcarbamate with ieri-butyl 5-fluoropyridin-2-ylcarbamate, compound (5S,7 ?)-36h-8 was obtained as a white solid. LC-MS (ESS): miz 557.2 [M + H] ; Ή NMR (500 MHz, /"-DMSO): δ 8.51 (q, J = 4.5 Hz, I H), 7.98 ~ 7.92 (m, 3H), 7.83 (s, 1 H), 7 59 (s, I H), 7.41 - 7.32 (m, 3H), 6.58 - 6.52 (m, 2H), 4.90 ( 1 , J = 7.0 Hz, 1 H), 4.15 - 4.05 (m, 2H), 3 35 (s, 3H), 3.36 - 3.30 (m, 2H), - 2.85 (m I H), 2.84 (d, J = 4.5 Hz, 3H), 1.64 (d, J 7.0 Hz, 3H) ppm.
Figure imgf000131_0003
|02511 Synthesis of compound (&S,7i?)-36h-9. Following the same procedure as that used for preparing compound (5S,7/?)-36h-2 shown in Scheme 36h and replacing r/-butyl thiazol-2- ylcarbamate with /eri-butyl 5-chloropyridin-2-ylcarbamate, compound (5S,7 ?)-36h-9 was obtained as a white solid. LC MS (ESS); mh 573 1 [M + H] ; Ή NMR (500 MHz, (ADMSO): S 8.51 (q, J = 4.0 Hz, 1 H), 7.96 (dd, J, - 8 5 Hz, J2 6 0 Hz, 3H), 7.83 (s, 1 H), 7.59 (s, 1 H), 7.43 ~ 7.38 (m, 3H). 6.80 (t, J 5.0 Hz, I H), 6 58 (d, J 9,0 Hz, I H), 4.90 (q, J 6.5 Hz, 1 H), 4.10 4.05 (m, 2H), 3.34 (s, 3H), 3.35 - 3 30 (m, 2H), 2 92 2 85 (m 1 H), 2.84 (d, J 4.5 Hz, 3H),
- 6.5 Hz, 3H) ppm.
Figure imgf000132_0001
102521 Synthesis of compound (&S,7J?)-36h-10, Following the same procedure as that used for preparing compound (5S,7 ?)-36h-2 shown in Scheme 36h and replacing terf-butyl thiazol-2- ylcarbamate with terf-butyl 5 trifluoromethylpyridin 2 ylcarbamate, compound (5S,7 ?)-36h-10 was obtained as a white solid. LC MS (ESI): mlz 607.2 [M + Hf; 1 H NMR (500 MHz, if
DMSO): δ 8.51 (q, J ~ 4.0 Hz, 1 H), 8.30 (s, 1 H), 7.96 (dd, J, = 8.5 Hz, = 5.5 Hz. 3H), 7.84 (s, 1 H), 7.63 (m, 1 H), 7.59 (s, I H), 7 42 - 7.36 (m, 3H). 6 66 (d, J = 9.0 Hz, 1 H), 4.90 (q, J ~ 6.5 Hz, I H), 4.18— 4.10 (m, 2H), 3.43 3,38 (m, 2H), 3.35 (s, 3H), 2.92 - 2.85 (m 1 H), 2.84 (d, J - 4.5
3H) ppm
Figure imgf000132_0002
|0253| Synthesis of compound (5S,7i?)-36h-U. Following the same procedure as that used for preparing compound (5S,7 ?) 36h-2 shown in Scheme 36h and replacing fer/ butyl thiazoL2 ylcarbamate with ten butyl 4- fluoropyridtn 2 -ylcarbamate, compound (5S.7 ?) 36h-U was obtained as a white solid. LC MS (ESI): mlz 557.2 [M + Hf , Ή NMR (500 MHz, t/ DMSO): δ 8.51 (q, J ~ 4.0 Hz, 1 H), 8 00 «· 7 95 (m, 3H), 7,84 (s, I H), 7.59 (s, I H), 7.40 (t, J 8 5 Hz, 2H), 6.80 (t, J - 6.0 Hz, 1 H), 6.39 (m, 1 H). 6.31 (d. J s 1 .5 Hz, I H), 4.90 (q, J * 6 5 Hz, 1 H), 4 17 (m, I H), 4.08 (m, I H), 3.38 3.28 (m, 2H), 3.35 (s5 3H), 2.92 - 2 85 (m I H), 2.84 (d, J 4,5 Hz, 3H),
- 6.5 Hz, 3H) ppm.
Figure imgf000132_0003
10254| Synthesis of compound (5$,7Λ)-36Ιι-1 . Following the same procedure as that used for preparing compound (5S,7?)-36h-2 shown in Scheme 36h and replacing tert butyl thiazol 2 ylcarbamate with ierf-but l 4-c hloropyridin-2 -ylcarbamate, compound (SS R) 36h-l was obtained as a white solid. LC-MS (ESI): mlz 573.1 [M + H] ; Ή NMR (500 MHz, </~DMSO): δ 8.51 (q,J~ 4.0 Hz, I H), 7.97 - 7.92 (m, 3H), 7.84 (s, 1 H), 7.59 (s, 1 H), 740 (t, J - 9.0 Hz, 2H), 6.83 (m, 1 H).6.62 (d,J= 1.5 Hz, 1 H), 6.55 (dd, J, 55 Hz, J2 1.5 Hz, 1 H), 490 (q, J 6.0 Hz, 1H), 4.15 (m, 1H), 4.07 (m, IH), 3.40-3.27 (m, 2H), 335 (s, 3H), 2.89 2.85 (m 1H), 2.84 (d, J = - 6.5 Hz, 3H)ppm.
Figure imgf000133_0001
|0255| Synthesis of compound (5£,7i?)-36h-13. Following the same procedure as that used for preparing compound (5S,7?)-36h-2 shown in Scheme 36h and replacing teri-butyl thiazol-2- ylcarbamate with teri-butyl 5-trifluoromethylpyridin-2-ylcarbamate, compound (5S,7i?)-36h-13 was obtained as a white solid. LC-MS (ESI): mlz 6072 [M + Hf; Ή NMR (500 MHz, <*- DMSO): 68.51 (q,J = 4.0 Hz, 1H), 8.20 (d, J - 5.0 Hz, I H), 7.95 (dd, J, = 9.0 Hz, J2 5.5 Hz, 2H), 7.84 (s, I H), 7.60 (s, 1 H), 7.40 (t, J - 8.5 Hz, 2H), 711 (t, J = 6.0 Hz, I H), 6.87 (s, 1 H), 6.72 (d,J=5.5 Hz, 1H), 4.90 (q,J=6.0 Hz, IH), 4.15 (m, IH), 4 i0(m, 1 H), 3.44 - 3.41 (m,
- 2.85 (m IH), 2.84 (d,J 5.0 Hz, 3H), 1.64 (d, J = 6.0 Hz, 3H) ppm.
Figure imgf000133_0002
102561 Synthesis of compound (5-?,7Λ)-36η-1 . Following the same procedure as that used for preparing compound (55,7/?)-36h-2 shown in Scheme 36h and replacing rerf-butyl thiazol-2- ylcarbamate with ieri-butyi pyridazin-3-ylcarbamate, compound (5S,7?)-36h-l4 was obtained as a white solid. LC-MS (ESI); mlz 540.2 [M + Hf ; Ή NMR (500 MHz, -DMSO): 58.51 (q,J 4.0 Hz, 1 H), 8.42 (d, J - 4.5 Hz, IH), 7.96 (dd, J\ 9.0 Hz, J2 5.5 Hz, 2H), 7.84 (s, 1 H), 7.60 (s, IH), 7.40 (t,J=9.0 Hz, 2H), 7.21 (dd, J, * 9.0 Hz,J2 4.5 Hz, IH), 6,86 (d, J - 8,5 HZ, IH), 6.82 (m, IH), 4.91 (q,J=6.0 Hz, IH), 4.20-4.15 (m, 2H), 345- 3.42 (m, 2H), 3.35 (s, 3H), 2.89 - 2.85 (m 1 H), 2.84 (d, J - 4.5 Hz, 3H), 1.64 (d,J 6.0 Hz, 3H) ppm.
Figure imgf000134_0001
|0257| Synthesis of compound (&S,7i?)-36h-15. Following the same procedure as that used for preparing compound (5S,7/?)-36h-2 shown in Scheme 36h and replacing terf- butyl thiazol-2- ylcarbamate with feri-butyS S-chloropyrazin-2-ylcarbamate, compound (SS,7/?)-36h-15 was obtained as a white solid. LC-MS (ESI): mlz 574.1 [M + Hf; Ή NMR (500 MHz, ίΛ-DMSO): 5 8.51 (q, J = 4.0 Hz, I H), 8.05 (s, I H), 7.96 (dd. J, = 9.0 Hz, J2 = 5.5 Hz, 2H), 7.84 (s, 1 H), 7.82 (s, 1 H), 7.40 (t, J = 9.0 Hz, 2H), 7.33 (t, J = 5.0 Hz, I H), 4.89 (q, J = 6.0 Hz, I H), 4.17 - 4.1 1 (m, 2H), 3.56 (s, 3H), 3.56 - 3.27 (m, 2H), 2.89 - 2.85 (m I H), 2.84 (d, J = 4.5 Hz, 3H), 1.64 (d, J 6.5 Hz, 3H) ppm.
Figure imgf000134_0002
|0258| Synthesis of (5S,7S)-36i-l Refer to Scheme 361, To a stirred suspension of Cs3C03 (3 mg, 0 1 mmol) in dry DMF ( 1 mL) was added 4-fluorophenol (6 mg, 0.05 mmol) at rt under an atmosphere of Ar. After stirring at rt for 30 min, compound (55,75 -36e-l (30 mg, 0.05 mmol) was added. The resulting reaction mixture was stirred at 60 "C overnight and then diluted with water ( 10 mL). The mixture was adjusted pH value to 3-4 by adding 2 N aq. HCl solution The resulting suspention was filtered and the solid was collected and further purified by preparative HPLC to give compound (5S,7S>36.-1 ( 13 mg, 48% yield) as a white solid. LC MS (ESI): mlz 557.1 [M + Hf; Ή NMR (500 MHz, </ DMSO): δ 8 52 (q, J 4.5 Hz, I H), 7.97 (dd, J, - 8 0 Hz, J - 5 0 Hz, 2H), 7.86 (s, I H), 7 61 (s, I H), 7 40 (t, J 8.5 Hz, 2H), 7. l0 (t, J 9.0 Hz, 2H), 6.97 6 94 (m, 2H), 4.96 (q, J 6.0 Hz, 1 H), 4.27 {d, J ~ 1 1.0 Hz, 2H), 4.01 - 3.97 (m, 2H), 3 40 (s, 3H), 3 03 - 2.99 (mf 1 H), 2.84 (d, J 5.0 Hz, 3H), 1.65 (d, J 5.0 Hz, 3H) ppm.
Figure imgf000134_0003
102591 Synthesis of compound (5S,75)-36i-2. Following the same procedure as that used for preparing compound (5S,7S)-36i-l shown in Scheme 36k and replacing 4-fluorophenol with 3- fluorophenol, compound (55,75)-36i-2 was obtained as a white solid. LC-MS (ESI): mlz 557.1 [M + H]+; Ή NMR (500 MHz, ( "-DMSO): δ 8.52 (q, J 5 0 Hz, 1 H), 7.97 (dd, Js =5.5 Hz. J2 = 9.0 Hz, 2H), 7.87 (s, I H), 7.61 (s, l H), 7.40 (t, J 8.5 Hz, 2H), 7.30 (q, 1 H), 6.84 - 6.74 (m, 3H), 4.97 (q, J = 6.5 Hz, I H), 4.29 - 4.25 (m, 2H), 4.07 4 01(m, 2H), 3.41 (s, 3H), 3.05 - 2.99 (m,
= 4.0 Hz, 3H), 1.65 (d, J- 6.0 Hz, 3H) ppm.
Figure imgf000135_0001
102601 Synthesis of compound (5&,7£)-36i-3. Following the same procedure as that used for preparing compound (55,75)-36i-l shown in Scheme 36i and replacing 4-fluorophenol with 3,4- difluorophenol, compound (55,75)-36I-3 was obtained as a white solid. LC-MS (ESI): mlz 575.1 [M + Hf; 1 H NMR (500 MHz, d6-DMSO): δ 8.52 (q, J 5.0 Hz, I H), 7.98 (dd, Jt -5.5 Hz, - 9.0 Hz, 2H), 7.87 (s, I H), 7.61 (s, 1 H), 7.40 (t, J- 8 5 Hz, 2H), 7.33 (q, J = 10.0 Hz, 1 H), 7 1 1— 7.06 (m, l H), 6.78 ~ 6.77(d, J 9,0 Hz, I H), 4 96 (q, J« 6 5 Hz, 1 H), 4.25 4.20 (m, 2H), 4.03 4.01(m, 2H), 3.40 (s. 3H), 3.05 - 2.99 (m, I H), 2.85 (d, J * 4.0 Hz, 3H), 1.64 (d, J - 6.0 Hz. 3H) ppm.
Figure imgf000135_0002
102611 Synthesis of compound (55,75 -36i-4. Following the same procedure as that used for preparing compound (55,75 36Ι~1 shown in Scheme 36i and replacing 4-fluorophenol with 2,4 difluorophenol, compound (55,75) -361-4 was obtained as a white solid. LC-MS (ESI): mlz 575.1 [M + Hf; Ή NMR (500 MHz, <f -DMSO): 6 8 52 (q, J 5.0 Hz, 1 H), 7.98 (dd, Ji ' 5.5 Hz, J2 » 9.0 Hz, 2H), 7.87 (s, 1 H), 7 61 (s, I H), 7 40 (t, J - 9.0 Hz, 2H), 7.29 - 7.20 (m, 2H), 7 01 (t, J 9.0 Hz, I H), 4.96 (q, J 6 5 Hz, 1 H), 4.30 4 20 (m, 2H), 4.09 (d, J 4.5 Hz, 2H), 3.40 (s, 3H), 3.05 - 2.99 (m, I H), 2.85 (d, J 4.0 Hz, 3H), 1 64 (d, J - 6 5 Hz, 3H) ppm.
Figure imgf000136_0001
|02621 Synthesis of compounds (5S,7S)-36j-la and (5S,7#)-36j-ib. Refer to Scheme 36j. To a stirred suspension of NaH ( 10 mg, 0.4 mmol) in dry DMF ( I mL) was added 5-chloropyridin-2-ol (31 mg, 0.24 mmol) at rt under an atmosphere of Ar. After stirring at rt for 30 min, the reaction mixture was added compound (55,75)-36e-i (50 mg, 0.08 mmol). The resulting mixture was stirred at 60 °C for 4 hrs and added several drops of saturated aq, NH4CI solution to quench the reaction. Subsequently, the mixture was added water ( 10 mL). The resulting suspension was filtered and the solid was collected and further purified by preparative HPLC to give compounds (55,75)-36j-la ( 12 mg, 26% yield) and (5S,7/?)-36j-Ib (2 mg, 4% yield) as white solid. (55,75)- 36]- ί a: LC-MS (ESI): mlz 574.1 [M + Hf; Ή NMR (500 MHz, tADMSO): 6 8.52 (q, J= 4.0 Hz, 1 H), 8.21 (s, 1 H), 7.97 (t, J - 7.0 Hz, 2H), 7.86 (s, 1 H), 7.80 (d, J = 9.0 Hz, 1 H), 7.61 (s, 1 H). 7.40 (t, J = 9.0 Hz, 2H), 6.89 (d, J = 9.0 Hz , I H), 4.94 (q, J = 6.5 Hz, 1 H), 4.30 - 4.23 (m, 4H), 3.39 (s, 3H), 3.03 - 2.99 (m, 1 H), 2.84 (d, J = 4.0 Hz, 3H), 1.64 (d, J = 6.0 Hz, 3H) ppm. (55,7/?)- 36j-lb: LC-MS (ESI): mlz 574.1 [M + Hf; 1 H NMR (500 MHz, cZ-DMSO): 6 8 50 (q, J - 4 5 Hz, 1 H), 7.97 (dd, J, = 5.0 Hz, J2 = 9.0 Hz. 2H), 7.86 (s, I H), 7,74 (s, 1 H), 7.58 (s, 1 H), 7 51 (dd, J, = 3.0 Hz, Jj = 10.0 Hz, 2H), 7.40 (t, J = 9.0 Hz, 2H), 6.45 (d, J = 10.0 Hz ,1 H), 4.83 (q, J « 6.5 Hz, I H), 4.25 - 4.10 (m, 3H), 3.64 - 3.63 (m, 1 H), 3.38 (s, 3H), 2 84 (d, J - 5.0 Hz, 3H), 1 .52 (d, J - 6.5 Hz, 3H) ppm.
Figure imgf000136_0002
1026 1 Synthesis of compounds (55,75 -36j-2a and (55,7 ?)-36j-2b. Following the same procedure as that used for preparing compounds (55,75)-36j-la and (55,7 ?)-36j-ib shown in Scheme 36j and replacing 5-chloropyridin-2-ol with 5 trifiuoromethylpyridin-2-ol, compounds (55,75)-36j-2a and (55,7/?)-36j-2b were obtained as white solid. (55,75)-36j-2a. LC MS (ESI): mlz 608.1 [M + Hf; 1 H NMR (500 MHz, c/*-DMSO): δ 8 58 (s, 1 H), 8.52 (q, J - 4 0 Hz, 1 H), 8.07 (dd, J, = 9.0 Hz, J2 = 2.5 Hz, I H), 7.97 (dd, J, - 9 0 Hz, J2 5.0 Hz, 2H), 7 87 (s, 1 H), 7 61 (s, 1 H), 7 42 0, J 9 0 Hz, 2H), 7.03 (d, J =« 9 0 Hz, I H), 4.95 (q, J 6.5 Hz, 1 H), 4.39 4 24 (ra, 4H), 3 39 (s, 3H). 3 09 3 03 (m, 1 H), 2.84 (d, J 4.0 Hz, 3H), 1.64 (d, J 6.5 Hz, 3H) ppm, (55,7/?) 36j-2b LC MS (ESI): miz 608 I [M + Hf; Ή NMR (500 MHz, cZ-DMSO): δ 8.49 (q, J 4.0 Hz, 1H), 7 96 (dd, J\ - 9.0 Hz, J2 5.5 Hz, 2H), 7.86 (s, 1 H), 7.78 (d, J ** 7.0 Hz, i H), 7.56 (s, 1 H), 7.40 (i, J 9 0 Hz, 2H), 6.80 (s, 1 H), 6.47 (dd, J, - 9.0 Hz, J2 2.5 Hz, I H), 4.83 (q, J - 6.5 Hz, 1 H), 4.28 4.21 (m, 3H), 3 76 3 71 (m, 1 H), 3 37 (s, 3H), 2.84 (d, J 4.5 Hz, 3H), 1.49 (d, J 6 5 Hz, 3H) ppm.
Figure imgf000137_0001
|0264| Synthesis of compounds (5_V,7S)-36j-3a and {5S,7i?)-36j-3b. Following the same procedure as that used for preparing compounds (5S,7S)-36j-la and (5S,7 ?)-36j-lb shown in
Scheme 36j and replacing 5-chloropyridin-2 ol with 4-tri fluoromethy lpyr idin 2-ol , compounds
(5S.7S) 36j-3a and (5S,7 ?)-36j-3b were obtained as white solid. (5S,7S)-36j-3a: LC-MS (ESI): miz 608.1 [M + H]+; 1 H NMR (500 MHz, </-DMSO): 6 8.51 (q, J - 4.0 Hz, I H), 8.43 (d, j - 5.0
Hz, I H), 7.97 (dd, J, = 9.0 Hz, Ji = 5.0 Hz, 2H), 7.87 (s, I H), 7 61 (s, I H), 7.40 <t, J 9.0 Hz,
2H), 7.34 (d, i = 5.0 Hz, 1 H), 7.23 (s, 1 H), 4.96 (q, J = 6.5 Hz, 1 H), 4.41 - 4.26 (m, 4H), 3.40 (s,
3H), 3.09 - 3.03 (m, 1 H), 2.84 (d, J = 4.0 Hz, 3H), 1.64 (d, J - 6 5 Hz, 3H) ppm. (5S R) -36j~3b'
LC-MS (ESI), miz 608.1 [M + Hj+; Ή NMR (500 MHz, (f DMSO): δ 8 50 (q, J - 4.0 Hz, I H),
8.10 (s, 1 H), 7.96 (dd, J, = 9.0 Hz, J2 = 5.5 Hz, 2H), 7.87 (s, 1 H), 7.70 (dd, A = 9 5 Hz, J2 - 2 5
Hz, 1 H), 7.57 (d, J = 5.0 Hz, 1 H), 7.40 (t, J = 9.0 Hz, 2H), 6.57 (d, J 9 5 Hz, 1 H), 4 81 (q, J
5.5 Hz, I H), 4.38 (dd, J\ = 13.5 Hz, j2 = 3.0 Hz, I H), 4 27 - 4. l6 (m, 2H), 3.65 (m, I H), 3.38 (s,
3H), 2.87 (d, J~ 4.5 Hz. 3H), 1.46 (d, J = 6.5 Hz, 3H) ppm.
Scheme 36k
Figure imgf000137_0002
10265| Synthesis of compound {5»S,7/f)-36k-l. Refer to Scheme 36k. A mixture of compound (5S,7/?)-36e-3 (30 mg, 0.065 mmol), 2-bromopyridine (20 mg, 0 13 mmol), Pd2dba3 (6.3 mg, 0.0065 mmol), Xantphos ( 1 1 mg, 0.13 mmol) and f BuONa ( 18 mg, 0.195 mmol) in dry DMF (8 ml) was stirred at 100 "C for 5 hrs under an atmosphere of N2. The mixture was subsequently concentrated and the residue was diluted with water (20 mL) and EtOAc (20 mL). The aqueous phase was extracted with EtOAc (20 mL x 3) and the combined organic extracts were washed with brine and dried over anhydrous NajSO . The solvent was removed and the residue was purified by preparative HPLC to give compound (5S,7 ?)-36k-l ( 13 mg, 24% yield) as a white solid. LC-MS (ESI): mlz 539.2 [M + Hf; Ή NMR (500 MHz, -DMSO): 5 8.51 (q, J = 5.0 Hz, 1 H), 7.98 - 7.95 (m, 3H), 7.83 (s, I H), 7.59 (s, 1 H), 7.37 (t, J - 6.0 Hz, 3H), 6.53(d, J = 8.5 Hz, 1 H), 6.47 (t, J = 5.5 Hz, 2H), 4.89 (q, J » 6.5 Hz, 1 H), 4.10 4.07 (m, 2H), 3.40 (s, 3H), 3.35 - 3.30 (m, 2H), 2.85 - 2.80 (m, 1 H), 2.84 (d, J- .5 Hz, 3H), 1.64 (d, J = 6.0 Hz, 3H) ppm.
|0266| Synthesis of compound (f¾7j?)-36h~8. Following the same procedure as that used for preparing compound ( 55,7/?) -36k- 1 shown in Scheme 36k and replacing 2-bromopyridine with 2- bromo-5-fluoropyridine, compound (55,7/?)-36h-8 was obtained as a white solid. LC-MS (ESI): mlz 557.2 [M + Hf .
|0267| Synthesis of compound (&S,7 ?)-36h-9. Following the same procedure as that used for preparing compound (55,7/?) 36k- 1 shown in Scheme 36k and replacing 2-bromopyridine with 2 bromo-5-chloropyridine, compound (55,7/?) 36h-9 was obtained as a white solid. LC-MS (ESI); mlz 557.2 [M + H]T
Scheme 36.
Figure imgf000138_0001
|0268| Synthesis of compound (55,7fl)-36l-l. Refer to Scheme 361. To a stirred solution of compound (55,7/?) 36e-3 (45 mg, 0 1 mmol) in DMF (2 mL) was added 2-chloro-5- fluoropyrimidine (39 mg, 0 29 mmol) and DIPEA (51 mg, 0 39 mmol). After stirring at 120 "C for 3 hrs, the reaction mixture was concentrated and the residue was purified by preparative HPLC to give compound (55,7/?) 36Ϊ-1 (5 mg, 9% yield) as a white solid. LC-MS (ESI): mlz 558.2 [M + H] ; Ή NMR (500 MHz, tf DMSO): 6 8 51 (q, J - 4.5 Hz, 1 H), 8.37 (s, 1 H), 7.96 (dd, J I - 8.8 Hz, Ji 5.5 Hz, 2H), 7 84 (s, 1 H), 7 59 (s, 1 H), 7.40 (t, J = 8.5 Hz, 2H), 7.21 (t, J 5.5 Hz, 1 H), 4.89 (q, J - 6,0 Hz, I H), 4.18 (d, J - 13.5 Hz, 1 H), 4.10 (br, 1 H), 3.34 (s, 3H), 3.26 3.18 (m, 2H), 2.84 (d, J = 4.0 Hz, 3H), 2.81 - 2.77 (m, 1 H), 1.62 <d, J 6.0 Hz, 3H) ppm.
|0269| Synthesis of compound (5S,7/?)-36h-7. Following the same procedure as that used for preparing compound (55,7Λ)-36Ι-Ι shown in Scheme 361 and replacing 2-chloro-5- fluoropyrimidine with 2-chloropyrimidine, compound (5S,7/?)-36h-7 was obtained as a white -MS (ESI): mlz 540.2 [M + Hf.
Figure imgf000139_0001
|0270| Synthesis of compound (5S,7J?)-36I-2. Following the same procedure as that used for preparing compound (5S,7 ?)- 361-1 shown in Scheme 361 and replacing 2-chloro-5- fluoropyrimidine with 2,5-dichloropyrimidine, compound (5-?,7/?)-36Ι-2 was obtained as a white solid. LC-MS (ESI): mlz 574.2 [M + Hf; Ή NMR (500 MHz, iZ-DMSO): S 8.50 (q, J- 5.5 Hz, 1 H), 8.35 (s, 2H), 7.97 (dd, J, = I I .O Hz, J2 = 7.0 Hz, 2H), 7.84 (s, 1 H), 7.59 (s, 1 H), 7.46 (t, J = 7.5 Hz, 1 H), 7.40 (t, J = 1 1.0 Hz, 2H), 4.89 (q, J ~ 8.0 Hz, 1 H), 4.19 - 4.10 (m, 2H), 3.38 3.37 (m, 1 H), 3.35 (s, 3H), 3.28 - 3 21 (m, 1 H), 2.84 (d, J 5 5 Hz, 3H), 2.79 (m, 1 H), 1.62 (d, J - 8.0 Hz, 3H) ppm.
Figure imgf000139_0002
|02711 Synthesis of compound (&S",7 f)-36l-3 Following the same procedure as that used for preparing compound (55,7 ?) 361-1 shown in Scheme 361 and replacing 2-chloro-5- fluoropynmidine with 2-chloro-5 trifluoromethylpyrimidine, compound (5S,7/?)-361-3 was obtained as a white solid LC MS (ESI): mlz 608 2 [M + Hf, Ή NMR (500 MHz, ( -DMSO): δ 8.66 (s, 1 H), 8 63 (s. 1 H), 8 50 (q, J 5.0 Hz, I H), 8 09 (t, J 5.5 Hz, 1 H), 7.97 (dd, J\ 9 0 Hz, J2 - 5.5 Hz, 2H), 7.95 (s, 1 H), 7.59 (s. 1 H), 7.40 (t, J - 9.0 Hz, 2H), 4.90 (q, J 7.0 Hz, I H), 4.20 4.14 (m, 2H), 3.45 3 40 (m, I H), 3 32 (s, 3H), 2 85 ~ 2.80 (m, 1 H), 2.84 (d, J 4.5 Hz, 3H), 1.62 (d, J 6.5 Hz, 3H) ppm.
Figure imgf000140_0001
|0272| Synthesis of compound (&S,7i?)-361-4. Following the same procedure as that used for preparing compound (5-?,7/?)-361-l shown in Scheme 361 and replacing 2-chloro-5- fluoropyrimidine with 3,6-dichloropyridazine, compound (5-?,7Λ)-361-4 was obtained as a white solid. LC-MS (ESI): mlz 574.1 [M + Η]τ; Ή NMR (500 MHz, </-DMSO): δ 8.52 (q, J 5.0 Hz, 1 H), 7.97 (dd, J, = 9.0 Hz, J2 = 5.5 Hz, 2H), 7.85 (s, 1 H), 7.60 (s, 1 H), 7.43 - 7.36 (m, 3H), 7.15 (t, J= 5.0 Hz, 1 H), 7.00 (d, J- .0 Hz, I H), 4.92 (q, J= 6.5 Hz, 1 H), 4.22 - 4.10 (m, 2H), 3.50 ~
- 2.85 (m, 2H), 2.84 (d, J= 4.5 Hz, 3H), 1.65 (d, J= 6.0 Hz, 3H) ppm.
Figure imgf000140_0002
102731 Synthesis of compound (5£,7Ι?)-36Ι-5. Following the same procedure as that used for preparing compound (55,7 ?)-361-l shown in Scheme 361 and replacing 2-chloro-5- fluoropyrimidine with 3 -chloro-6-tri l uorome thylpyridazine, compound (55",7Λ) 361-5 was obtained as a white solid LC-MS (ES ): mlz 608.2 [M + Hf , Ή NMR (500 MHz, c DMSO): δ 8.52 (q, J= 5.0 Hz, 1 H), 7.97 (dd, J/ - 9,0 Hz, J2 - 5.5 Hz, 2H), 7.85 (s, 1 H), 7.74 (m, 1 H), 7 70 (d, J - 9.0 Hz, 1 H), 7.60 (s, I H), 7 40 (t, J 8.5 Hz, I H), 7.08 (d, J 9.0 Hz, 1 H), 4 92 (q, J 6.5 Hz, I H), 4.22 - 4.10 (m, 2H), 3.50 {m, 2H), 3.37 (d, J · 5.5 Hz, 3H), 2.90 (m, I H), 2.84 (d, J- - 6.5 Hz, 3H) ppm.
Figure imgf000140_0003
102741 Synthesis of compound (5_S,7J?)-36Ms Following the same procedure as that used for preparing compound (55",7Λ)-361-1 shown in Scheme 361 and replacing 2-chloro-5- fluoropyrimidine with 4 chloroquinazoline, compound (55, 7Λ) 361-6 was obtained as a white solid. LC-MS (ESI): mlz 590.2 [M + Hf ; Ή NMR (500 MHz, i -DMSO): δ 8.52 (q, J 5 0 Hz, 1 H), 8.38 (s, I H), 8.34 (t, J - 15 Hz, I H), 8.29 (d, J - 8.5 Hz, 1 H), 7.97 (dd, J, » 9.0 Hz, J2 - 5.5 Hz, 2H), 7.85 (s, I H), 7.78 (t, J 8.5 Hz, I H), 7 70 (d, J 8.0 Hz, I H), 7.60 (s, I H), 7 53 (t, J 7.0 Hz, 1H), 740 (t, J 85 Hz, 1H),493 (q, J* 6.5 Hz, 1H), 4.31 (m, 1H), 4.23 (m, IH), 3.66- (m, IH), 2.84 (dtJ 40 Hz, 3H), 1.64 (d,J= 6.5 Hz, 3H) ppm.
Figure imgf000141_0001
|02751 Synthesis of compound (55,7Λ)-361-7. Following the same procedure as that used for preparing compound ( 55", 7Λ) 361-1 shown in Scheme 361 and replacing 2-chIoro-5- fluoropyrimidine with 2-chloroquinazoline, compound (55,7/?) 361-7 was obtained as a white solid. LC MS (ESI): m/z 5902 [M + Hf, Ή NMR (500 MHz, -DMSO): δ 9.16 (s, 1 H), 8.50 (q, J- 5.5 Hz, 1H),7.97 (dd,J,= 9.0 Hz, J2 - 5.5 Hz, 2H), 7.84 (s, IH), 7.81 (d,J=6.5 Hz, 1H), 7.70 (m, 1 H), 760 (s, I H), 747 (d, J - 8.0 Hz, 1 H), 7.40 (t, J - 8.5 Hz, 1 H), 7.34 (m, 1 H), 7.24 (t, J = 8.0 Hz, IH), 4.92 (q, J 60 Hz, IH), 4.28 - 4.22 (m, 2H), 3.48 - 3.38 (m, 2H), 3.35 (s, 3H),
=4.0 Hz, 3H), l.64(d,J-6.0 Hz, 3H) ppm.
Figure imgf000141_0002
|0276| Synthesis of compound (5_ϊ,71?)-361-8. Following the same procedure as that used for preparing compound (55",7/?)-361-l shown in Scheme 361 and replacing 2-chloro-5
fluoropyrimidine with 4-chlorothieno[3,2-i]pyrimidine, compound (55,7Λ)-361-8 was obtained as a white solid. LC-MS (ESI): m/z 596.1 [M + Hf ; Ή NMR (500 MHz,
Figure imgf000141_0003
δ 8.50 (q, J* 5.5 Hz, IH), 8.45 (s, IH), 8.10 (d, J =5.5 Hz, IH), 7.98-7.94 (m, 3H),7.84 (s, IH), 759 (s, IH), 7.42 - 7.28 (m, 3H), 4.92 (q, J= 6.5 Hz, 1 H), 4.24 - 4.20 (m, 2H), 3.62 - 3.48 (m, 2H), 334 (s, 3H), 2.90- =4.5 Hz, 3H), 1.62 (d,J= 6.5 Hz, 3H) ppm.
4-F-PnO-, R
Figure imgf000141_0004
[02771 Synthesis of compound (&S,7-S)-37-3a-l. Refer to Scheme 37. To a stirred solution of compound (55,75)-37-1β (60 mg, 0. 1 15 mmol) in DCE (3 mL) was added PC15 (72 mg, 0.344 mmol) at rt. After refluxing for 3 hrs, the reaction mixture was cooled to rt. Subsequently,
NHjOH HC1 (23 mg, 0.344 mmol) and DIPEA (369 mg, 2.86 mmol) were added and the resulting mixture was re fluxed for 3 hrs. The mixture was concentrated and the residue was purified by preparative HPLC to give compound (5-?,75 37-3a-l (40 mg, 65% yield) as a white solid. LC-MS (ESI); m/z 540.1 [M + Hf; Ή NMR (500 MHz, i -DMSO): 6 9.81 (s, 1 H), 8.40 (s, 1 H), 7.94 - 7.97 (m, 2H), 7.90 (s, I H), 7.46 (s, 1 H), 7.37 - 7 44 (t, 2H), 6.25 (q, J = 5.0 Hz, 1 H), 4.99 (q, J = 6.5 Hz, 1 H), 4.40 (t, 7 = 6.5 Hz, 1 H), 4.20 4 22 (m, 1 H), 3.40 (s, 3H), 3.23 (br, 1 H), 3.04 (s, 3H), 2.92 - 2.97 (br, 1 H), 2.48 (d, J = 5.5 Hz, 3H), 1 64 (d, J - 6.5 Hz, 3H) ppm.
|0278| Synthesis of compound (55,75 7-3a-2. Following the same procedure as that used for the preparation of compound (55,7S)-37-3a-l and replacing NH2OH HCl with NH2OMe HCl, compound (5S,7S)-37«3a-2 (45 mg, 71 % yield) was obtained as a white solid. LC-MS (ESI): m/z 554.1 [M + Hf; Ή NMR (500 MHz,
Figure imgf000142_0001
δ 7.94 (m, 2H), 7.89 (s, 1 H), 7.47 (s, 1 H), 7.43 (m, 2H), 6.48 (q, J = 4.5 Hz, 1 H), 4.98 (m, 1 H), 4.39 (m, I H), 4.20 (m, 1 H), 3.72 (s, 3H), 3.39 (s, 3H), 3.23 3.30 m, 2H), 3.04 (s, 3H), 2.93 (m, 1 H), 2.45 (d, J = 6.5 Hz, 3H), 1.64 (d, J = 6.5 Hz, 3H) ppm.
Scheme 38
Figure imgf000143_0001
(0279] Synthesis of compounds 38-14a/b, 38-15a b and related derivatives. Refer to Scheme 38. Following the procedures described in a reported protocol (Aust, J. Chem. 1981, 34, 927) with some modification, compound 38-4 is obtained using commercially available compound 38-1 as the starting material. De-methylation of compound 38-4, followed by reduction of the -NO2 moiety and acylation of the resulting product gives compound 38-6. Sonogashira coupling of compound 38-6 with 4-fluorophenyl acetylene or 4-fluorophenoxyl-4-phenylacetylene, followed by soapnification, cyclization, and mesylation gives compound 38-9a or 38-9b, which is converted to compound 38- 10a or 38- 10b by a Pd-mediated vinylation. A-alkylation of compound 38- 10a or 38-10b with (/?)-(2,2-dimethyl-l,3-dioxolan-4-yl)methyl 4- methylbenzenesulfonate, followed by TsOH-mediated one-pot deprotection and cyclization gives compound 38-12a or 38-12b, which is converted to compound 38-13a or 38-13b by selective iodi nation, followed by a Pd-mediated carbonylation. Finally, following the procedures described for the preparation of those functionalized benzofuran derivatives described in Schemes 34a, 35, 35a, 35b, 36a, 36b, and 37 and using 38-13a or 38- 13b as the substrate, compounds 38-14a/b, 38- 15a b, O-alkylated or -acylated derivatives of 38-14a b, or W-hydroxy(or alkoxyl)-W- methylacetimidamide derivatives of 38-14a b or 38-15a/b are obtained.
Scheme 39
Figure imgf000144_0001
3S-12B-1 R = F. R" = -Me 3S-13S-1 R = F. " = -Me
39-12*-2 = F. R- = -OMe 39.13a.2 R = F. R- = -OMe
39-12b-1 R = 4-F-PhO-. R' 38-136-1 R ^F-PhO-. R"
39-12b-2 = 4-F-PhO-. R' 39.13b-2 R s4-F-PhO-. R-
[0280] Synthesis of compounds 39-12a b, 39-13a b and related derivatives. Refer to Scheme 39. Selective formylation of commercially available 39-1, followed by Baeyer-Villiger reaction and soapnification gives compound 39-3. De-methylation of compound 39-3 and acylation of the resulting phenol derivative give compound 39-4, which undergoes a Sonogashira coupling with 4- fluorophenylacetylene or 4-fluorophenoxyl-4-phenylacetylene to give compound 39-5a or 39-5b. One-pot benzofuran formation and carbonylation of compound 39-5a or 39-5b gives compound 39-6a or 39-6b, which is nitrated, followed by triflation and a Pd-mediated vinylation to give compound 39-8a or 39-8b. Finally, following the procedures described for the preparation of those functionalized benzofuran derivatives described in Schemes 29, 34a, 35, 35a, 35b, 36a, 36b, and 37 and using 39-8a or39-8b as the substrate, compounds 39-12a/b, 39-13a b, -alkylated or -acylated derivatives of 39-12a/b, orJV-hydroxy(or alkoxy.)-N-methylacetim idamide derivatives of 39-12a b or 39-13a/b are obtained
Figure imgf000145_0001
[0281] Step 1. Refer to Scheme 39a. To a stirred solution of compound 39a- 1 (5.1 g, 17 mmol) (prepared by following the procedure descried in WO200759421 with some modifications) in CH3CN (300 mL) was added Selectfluor® (7.25 g, 20.4 mmol) portion-wise at rt. After stirring at rt overnight, the reaction mixture was concentrated. The residue was added water (50 mL) and the resulting mixture was extracted with EtOAc (100 mL x 3). The combined organic extracts were washed with brine, dried over anhydrous Na2SC>4 and concentrated. The residue was purified by silica gel column chromatography (Petroleum ether EtOAc = 9/1 (v V)) to give the desired regio- isomer compound 39a-2 (1.35 g, 25% yield) as a yellow solid. LC-MS (ESI): m/z 319.1 [M + H]+.
[0282] Step 2. To a stirred solution of H 03 (5 mL) in DCM (15 mL) at 0°C was added a solution of compound 39a-2 (954 mg, 3 mmol) in DCM (15 mL) dropwise (Caution: the reaction mixture must be kept around 0 °C). After stirring at 0 °C for 1 hr, the reaction mixture was slowly warmed to rt and diluted with ice-water (15 mL). The organic phase was washed with ¾0 (25 mL x 3), dried over anhydrous a2S04, and concentrated. The residue was dried in vacuo to give crude compound 39a-3 (850 mg) as a dark yellow solid, which was used for the next step without further purification. LC-MS (ESI): m/z 364.1 [M + H .
[0283] Step 3. To a stirred solution of crude compound 39a-3 ( 1.24 g, about 3.4 mmol) in DCM (50 mL) was added DIPEA (1.7mL, 10.2 mmol) and DMAP (5 mg, 0.34 mmol), followed by Tf20 (0.86 mL, 5.1 mmol) at 0 °C. After stirring at rt for 5 hrs, the reaction mixture was diluted with water (50 mL) and DCM (50 mL). The organic phase was washed with brine (50 mL), dried over anhydrous Na2S04. and concentrated. The residue was purified by silica gel column chromatography (Petroleum ether/EtOAc = 15/1 (v/v)) to give compound 39a-4 (850 mg, 51% yield) as a yellow solid. LC-MS (ESI): m/z 496.0 [M + H]+.
[0284] Step 4. To a solution of the compound 39a-4 (1.5 g, 3.0 mmol) in DME (50 mL) and 2 M Na2C03 solution (4 mL) was added Pd(PPh3)2Cl2 (210 mg, 0.3 mmol), the resulting mixture was saturated with N2. Next, vinylboronic acid pinacol ester (0.6 mL, 4.5 mmol) was added and the resulting mixture was stirred at 80 °C for 4 hrs under an atmosphere of N2. Subsequently, the mixture was concentrated and the residue was added water (50 mL) and EtOAc (50 mL). The aqueous phase was extracted with EtOAc (25 mL x 3). The combined organic extracts were washed with brine (50 mL), dried over anhydrous Na2S04, and concentrated. The residue was purified by silica gel column chromatography (Petroleum ether/EtOAc = 15/1 (v/v)) to give compound 39a-5 (1.0 g, 89% yield) as a yellow solid. LC-MS (ESI): m/z 374.1 [M + H]+.
10285] Step 5. To a stirred solution of the compound 39a-5 (1.0 g, 2.7 mmol) in EtOAc (30 mL) was added SnC 2-2H20 ( 1.22 g, 5.4 mmol) portion-wise. After refluxing for 1 hr under an atmosphere of N2, the reaction mixture was cooled to rt and slowly added saturated aq. NaHC03 solution (20 mL). The mixture was filtered through Celite® 545 and the filtered cake was washed with EtOAc (25 mL x 3). The organic phase of the filtrate was dried over anhydrous Na2S04 and concentrated. The residue was dried in vacuo to give the amine as a light yellow solid, which was used for the next step without further purification. LC-MS (ESI): m/z 344.1 [M + H]+. Next, to a stirred solution of the crude amine in DCM (30 mL) was added TEA ( 1.8 mL, 13.5 mmol) and DMAP (50 mg), followed by MsCl (0.8 mL, 10 mmol) at 0 °C. After stirring at rt overnight, the reaction mixture was added saturated aq. NaHC03 solution (5 mL), water (25 mL), and DCM (50 mL). The organic phase was washed with brine (25 mL), dried over anhydrous NaiSG*, and concentrated. The residue was purified by silica gel column (Petroleum ether EtOAc = 2 1 (v/v)) to give compound 39a-6 (700 mg, 62% yield for two steps). LC-MS (ESI): mk 422.1 [M + H]+. (0286] Step 6. Following the same procedure as that used for preparing compound 34a-2 show in Scheme 34a and replacing compound 29-3 with 39a-6, compound 39a-7 (66 mg, 26% yield) was obtained as a yellow oil. LC-MS (ESI): mlz 536.1 [M + Hf.
[0287] Step 7. Following the same procedure as that used for preparing compound (55,75)-34-2a shown in Scheme 34a and replacing compound 34a-2 with 39a-7, compound 39a-8 (35 mg, 25% yield) was obtained as a white solid. LC-MS (ESI): mlz 518.1 [M + Na]+; Ή NMR (500 MHz, CDC13): δ 7.93 (dd, J,= 9.0 Hz, J2 = 5.5 Hz, 2H), 7.32 (s, 1H), 7.17 (t, J= 9.0 Hz, 2H), 5.80 (q, J = 7.5 Hz, 1H), 4.40 (q, J= 7.0 Hz, 1H), 4.04 (d, J= 14.0 Hz, 1H), 3.76 - 3.61 (m, 2H), 3.25 (s, 3H), 3.30 - 3.20 (m, 1 H), 2.05 - 1.90 (m, 1H), 1.75 (d, J= 7.0 Hz, 3H), 1.36 (d, J = 7.5 Hz, 3H) ppm.
[0288] Step 8. Following the same procedure as that used for preparing compound 1-15 shown in Scheme 1 (WO2012/058125) and replacing compound 1-14 with 39a-8, compound 39a-9 was obtained as a light solid. LC-MS (ESI): mlz 490.0 [M + Na]+.
|0289] Step 9. Following the same procedure as that used for preparing compound 1-16 shown in Scheme 1 (WO2012/058125) and replacing compound 1-15 with 39a-9, compound 39-12a-l was obtained as a white solid. LC-MS (ESI): mlz =481.1 [M + H]+; Ή NMR (500 MHz, c -DMSO): δ 8.71 (q, J= 4.0 Hz, 1 H), 7.89 (dd, J, = 9.0 Hz, J2 = 6.0 Hz, 2H), 7.73 (s, 1H), 7.42 (t, J= 9.0 Hz, 2H), 5.57 (q, J= 7.0 Hz, 1H), 4.83 (q, J= 5.5 Hz, 1H), 4.17 (d, J= 14.0 Hz, 1H), 3.48 - 3.43 (m, l H), 3.45 (s, 3H), 3.40 - 3.30 (m, 1 H), 2.93 - 2.92 (m, 1 H), 2.81 (d, J= 4.5 Hz, 3H), 1.61 (d,J= 7.0 Hz, 3H) ppm; !9F NMR (376 MHz, c -DMSO): δ -110.21 , -123.87 ppm.
Scheme 40
Figure imgf000148_0001
[0290 [ Step 1. Refer to Scheme 40. To a stirred solution of 6-amino-pyridin-2-ol (5g, 45.5 mmol) in acetic acid (50 mL) was added a solution of N-iodosuccinimide (20.45 g, 90.9 mmol) in acetic acid (90 mL). After stirring at rt for lhr, the resulting precipitate was collected by filtration, washed with acetic acid, and dried in vacuo to give compound 40-2 (12.5g, 84% yield) as yellow solid. LC-MS (ESI): m/z 462.7 [M + Hf.
[0291] Step 2. A solution of compound 40-2 (10 g, 27.6 mmol) in Ac20 (50 mL) was stirred at 90 °C for 1 hr and then concentrated. The residue was carefully added saturated aq. NaHCOj solution (100 mL) at 0 °C and the resulting mixture was extracted with DCM (150 mL x 3). The organic extracts were combined, dried over anhydrous Na2S04, and concentrated. The residue was dried in vacuo to give compound 40-3 ( 15 g, 68 % yield) as a light yellow solid. LC-MS (ESI): m/z 510.5 [M + H]+. (0292] Step 3. To a stirred solution of compound 40-3 (10 g, 20.5 mmol) and EtjN (6 mL, 41 mmol) in THF (100 ml) were added Cul (0.18 g, 0.943 mmol) and Pd(PPh3)2Cl2 (575 mg, 0.82 mmol) at 0 °C under an atmosphere of N2. Subsequently, a solution of 4-fluorophenylacetylene (2.5 g, 20.5 mmol) in THF (80 mL) was added dropwise over a period of 2 hrs. After stirring at 0
°C for 5 hrs and then at rt overnight, the reaction mixture was filtered and the filtrate was concentrated. The residue was dried in vacuo to give crude compound 40-4a as a brown solid, which was used directly for the next step without further purification, LC-MS (ESI): mlz 502.6 [M + Hf.
[0293 [ Step 4. To a stirred solution of compound 40-4a (1.5 g, 3.1 mol) in MeOH (10 mL) was added K2C03 ( 1.7 g, 12.4 mmol) at rt. After stirring at rt for 16 hrs, the reaction mixture was concentrated and the residue was diluted with THF (25 mL). The mixture was filtered through a Celite®545 plug and the filtered cake was washed with THF (25 mL x 2). The filtrate was concentrated and the residue was dried in vacuo to give crude compound 40-5a (700 mg, 63% yield) as a yellow solid. LC-MS (ESI): mlz 354.8 [M + Hf.
10294] Step 5. To a stirred solution of compound 40-5a (5 g, 14.1 mol) in anhydrous THF (50 mL) was added 1 M TBAF in THF (14.1 mL). After refluxing for 16 hrs, the reaction mixture was concentrated. The residue was purified by silica gel column chromatography
(DCM Petroleum ether = 1/1 (vV)) to give compound 40-6a (3.5 g, 70% yield) as a gray solid. LC-MS (ESI): m/z 354.7 [M + H]+.
[0295] Step 6. To a stirred solution of compound 40-6a (5.0 g, 14 mmol) and anhydrous Et3N ( 11.6 mL, 85 mmol) in dry DCM (200 mL) at 0 °C was added MsCl (4.3 mL, 56 mmol) dropwise. After stirring at rt for 16 hrs, the reaction mixture was concentrated and the residue was diluted with water (100 mL). The mixture was extracted with EtOAc (100 mL x3). The organic extracts were combined, dried with anhydrous Na2SC>4, and concentrated. The residue was dried in vacuo and then dissolved in MeOH (150 mL), followed by adding LiOH ( 1 M aq. solution, 25 mL). After stirring at rt for 1 hr, the reaction mixture was concentrated. The residue was added water ( 100 mL) and the mixture was extracted with ether (50 mL x 3). The aq. layer was adjusted to pH 6 with saturated aq. citric acid. The precipitate was collected by filtration, washed with water, and dried in a lyophilizerto give compound 40-7a (400 mg, 65% yield) as a brown solid. LC-MS
m (ESI): mlz 510.5 [M + H]+; Ή NMR (500 MHz, i/'-DMSO): δ 9.90 (br, 1H), 8.59 (s, 1 H), 8.00 (t, J= 5.5 Hz, 2H), 7.39 - 7.35 (m, 3H), 3.40 (s, 3H) ppm.
[0296] Step 7. To a stirred solution of the compound 40-7a (2.5 g, 5.8 mmol) in DME (30 mL) and 2 M aq. Na2C03 solution (7.5 mL) was added Pd(PPh3)2Cl2 (407 mg, 0.58 mmol), followed by vinylboronic acid pinacol ester (1.13 g, 8.7 mmol) under an atmosphere of N2. After stirring at 80 °C for 4 hrs, the reaction mixture was concentrated and the residue was poured into water ( 100 mL). The mixture was extracted with EtOAc (50 mL x 3). The extracts were combined, washed with brine, dried with anhydrous Na2S04, and concentrated. The residue was purified by silica gel column chromatography (DCM) to give compound 40-8a (900 mg, 47% yield) as a yellow solid. LC-MS (ESI): mJz 333.1 [M + Hf.
10297] Step 8. To a stirred solution of compound 40-8a (900 mg, 2.7 mmol) in dry DMF (5.4 mL) was added Cs2C(¼ (3.52 g, 10.8 mmol). The mixture was stirred at rt for 30 min and then ( ?)-(2,2-dimethyl-l,3-dioxolan-4-yl)methyl 4-methylbenzenesulfonate (3.1 1 g, 10.8 mmol) was added. After stirring at 1 10 °C for 48 hrs, the reaction mixture was concentrated. The residue was diluted with water (100 mL) and the resulting mixture was extracted with EtOAc (50 mL x 3). The extracts were combined, washed with brine, dried with anhydrous Na2S04, and concentrated. The residue was purified by silica gel column chromatography (Petroleum ether DCM = 2/1
(v/v)) to give compound 40-9a (600 mg, 50% yield) as a yellow solid. LC-MS (ESI): mlz 447.1
Figure imgf000150_0001
[0298 [ Step 9. To a stirred solution of compound 40-9a (800 mg, 1.76 mmol) in DCE ( 100 mL) at 90 °C was added TsOH (600 mg, 3.5 mmol). After re fluxing overnight, the reaction mixture was concentrated. The residue was dissolved in DCM (100 mL) and the resulting mixture was washed with water (25 mL x 2), dried with anhydrous Na2SC>4, and concentrated. The residue was purified by silica gel column chromatography (Petroleum ether/EtOAc = 2/1 (v/v) then
DCM acetone = 20/1 (v/v)) to give compound 40-10a (400 mg, 52%, yield; (5S, 7S)/(57?, 75) ~ 1/1) as a white solid. The desired diastereomer was isolated by chiral HPLC. LC-MS (ESI): mlz 407.1 [M + Hf; Ή NMR (500 MHz, cZ-DMSO): 5 8.19 (s, 1H), 8.03 (dd, J/ = 5.5 Hz, J2 = 8.5 Hz, 2H), 7.51 (s, 1H), 7.39 (t, J = 8.5 Hz, 2H), 4.86 (t, J= 6.0 Hz, 1H), 4.78 (q,J = 6.5 Hz, 1H), 4.19 (d, J= 13.0 Hz, 1H), 3.83 (t, J= 5.5 Hz, I H), 3.53 (s, 3H), 3.51- 3.49 (m, 1H), 3.30 - 3.26 (m, 1H), 2.85 - 2.81 (m, I H), 1.64 (d, J= 6.5 Hz, 3H) ppm. 10299] Step 10. To a solution of compound 40- 10a (200 mg, 0.5 mmol) and TFA (1 mL) in CH2Cb (2 mL) was added NIS (160 mg, 0.60 mmol) portionwise at rt. After stirring at rt for 3 hrs, the reaction mixture was concentrated and the residue was purified by silica gel column chromatography (Petroleum ether/EtOAc = 4/1 (v/v)) to give compound 40-1 la (200 mg, 76% yield) as a yellow solid. LC-MS (ESI): mlz 533.0 [M + H]+.
[0300] Step 11. To a solution of compound 40- 11 a ( 100 mg, 0.19 mmol) and Et3N (0.13 mL, 0.94 mmol) in DMF (3 mL) and MeOH (3 mL) was added
Figure imgf000151_0001
( 13 mg, 0.019 mmol) under an atmosphere of N2. Subsequently, the reaction mixture was saturated with CO and stirred at 60 °C for 24 hrs under an atmosphere of CO. The reaction mixture was concentrated and the residue was diluted with water (25 mL). The mixture was extracted with EtOAc (25 mL x 4). The organic extracts were combined, washed with brine, dried with anhydrous a2S04, and concentrated. The residue was purified by silica gel column chromatography (Petroleum ether EtOAc = 8/1 (v v)) to give compound 40-12a (70 mg, 79% yield) as a yellow solid. LC-MS (ESI): mlz 465 Λ [M + H]+.
[0301] Synthesis of compound 40-14a. Following the same experimental procedures as described for the conversion of compound 1-14 to 1-16 in Scheme 1 (WO2012/058125) and replacing compound 1-14 with 40-12a, compound 40-14a was obtained as a white solid. LC-MS (ESI): mlz 464.1 [M + H]+; Ή NMR (500 MHz, i/-DMSO): δ 8.56 (q, J=5.0 Hz, 1 H), 8.09 (s, 1H), 8.03 (dd, J, = 5,0 Hz, J2 = 9.0 Hz, 2H), 7.40 (t, J= 9.0 Hz, 2H), 4.87 (t, J= 7.0 Hz, 1H), 4.81 (q, J= 6.5 Hz, 1H), 4.21 (d, J= 13.5 Hz, 1 H), 3.84 (t, J= 7.0 Hz, 1H), 3.51 (s, 3H), 3.51- 3.49 (m, 1 H), 3.30 - 3.26 (m, 1 H), 2.85 (s, 3H), 2.85 -2.81 (m, 1 H), 1.65 (d, J = 6.5 Hz, 3H) ppm.
[0302] Synthesis of compound 40- 14b. Following the same experimental procedures as described for the conversion of compound 1-14 to 1-16 in Scheme 1 (WO2012/058125) and replacing compound 1-14 with 40-12a, and MeNH2 HCl with MeONH2 HCl, compound 40-14b was obtained as a white solid. LC-MS (ESI): mlz 479.4 [M + Hf. Ή NMR (500 MHz, ct- DMSO): δ 8.10 (s, 1H), 8.04 (dd, Jx = 5.0 Hz, J2 = 8.5 Hz, 2H), 7.43 (t, J= 8.5 Hz, 2H), 4.87 (t, J = 7.0 Hz, 1H), 4.81 (q, J= 6.0 Hz, 1H), 4.21 (<L J = 13.5 Hz, 1 H), 3.85-3.83 (m, 1H), 3.78 (s, 3H), 3.54 (s, 3H), 3.50 - 3.49 (m, 1H), 2.86 - 2.81 (m, 1H), 1.65 (d, J= 6.5 Hz, 3H) ppm. [0303 J Synthesis of compound 40-15a. Following the same experimental procedures as described for the conversion of compound 28-1 lb to 28-13b in Scheme 28 and replacing compound 28-1 lb with 40-14a, compound 40-15a was obtained as a white solid. LC-MS (ESI): mlz 526.1 [M + H]+; Ή NMR (500 MHz, cZ-DMSO): δ 8.56 (q, J= 4.5 Hz, 1 H), 8.1 1 (s, 1H), 8.04 (dd, J i = 6.0 Hz, J2 = 9.0 Hz, 2H), 7.41 (t, J = 9.0 Hz, 2H), 4.89 (q,J= 6.0 Hz, 1 H), 4.29 (t, J= 7.0 Hz, 1H), 4.19 (d, 7=14.0 Hz, 1H), 3.53 (s, 3H), 3.41- 3.36 (m, 2H), 3.05 (s, 3H), 2.97 (t, J= 12.5 Hz, 1H), 2.85 (d, J= 5.0 Hz, 3H), 1.69 (d, J = 6.5 Hz, 3H) ppm.
[0304] Synthesis of compound 40-15b. Following the same experimental procedures as described for the conversion of compound 28-1 lb to 28-13b in Scheme 28 and replacing compound 28-1 lb with 40-14b, compound 40-15b was obtained as a white solid. LC-MS (ESI): mlz 542.1 [M + H]+. Ή NMR (500 MHz, iZ-DMSO): δ 8.36 (s, 1H), 8.13 (s, 1H), 8.05 (m, 2H), 7.43 (t, 7 = 8.0 Hz, 2H), 4.90 (q, J= 6.5 Hz, 1 H), 4.30 (t, J = 6.0 Hz, 1H), 4.19 (d, J= 14.0 Hz, 1H), 3.78 (s, 3H), 3.75-3.73 (m, 1H), 3.53 (s, 3H), 3.54- 3.52 (m, 1 H), 3.05 (s, 3H), 2.97 (t, J=
= 6.0 Hz, 3H) ppm.
Figure imgf000152_0001
[0305] Step 1. Refer to Scheme 42a. To a stirred solution of sodium propane-2-thiolate (/-PrSNa) (120 mg, 1.22 mmol) in anhydrous DMF (8 mL) was added I (267 mg, 1.62 mmol) at rt under an atmosphere of N2. After stirring at rt for 30 min, compound 36e-l (500 mg, 0.81 mmol) was added in small portions. The resulting mixture was stirred at rt overnight and then diluted with ice water. The suspension was filtered and the solid was washed with water and dried in vacuo to give crude compound 42a-l (450 mg, 67% purity based on LC-MS), which was used for the next step without further purification. LC-MS (ESI): mlz 521.2 [M + Hf.
[0306] Step 2. To a stirred solution of compound 42a-l (450 mg, 0.81 mmol) in acetone (50 mL) was added hydrogen peroxide (20 mL, 30 wt. % in H2Q at rt. After stirring at 60 °C overnight, the reaction mixture was cooled to rt and quenched with saturated aq. Na2S03 solution (20 mL). The mixture was concentrated to remove acetone and the resulting suspension was filtered. The solid was washed with water and purified by preparative HPLC to give compound 42a-2 (110 mg, 25% yield) as a white solid. LC-MS (ESI): mlz 553.1 [M + Hf; Ή NMR (500 MHz, t/-DMSO): δ 8.53 (d, J= 4.5 Hz, 1H), 7.98 (dd, Ji = 5.5 Hz, J2= 8.5 Hz, 2H), 7.88 (s, 1H), 7.60 (s, 1H), 7.41 (t, J = 8.5 Hz, 2H), 4.97 (q, J= 6.5 Hz, 1H), 4.39 (t, J = 10.5 Hz, 1H), 4.21 (t, J = 14.5 Hz, 1H), 3.41 (s, 3 H), 3.41- 3.87 (m, 2H), 3.18 (dd. J, = 1.5 Hz, J2 = 15.0 Hz, 1H), 2.94 (br, 1H), 2.85 (d, J = J = 6.0 Hz, 3H), 1.26 (d, J= 7.0 Hz, 3H), 1.23 (d, J = 7.0 Hz, 3H) ppm.
Figure imgf000153_0001
103071 Synthesis of compound 42a-3. Following the same procedure as that used for preparing compound 42a-2 shown in Scheme 42a and replacing i-PrSNa with EtSNa, compound 42a-3 was obtained as a white solid. LC-MS (ESI): mlz 539.1 [M + Hf.
Figure imgf000153_0002
103081 Synthesis of compound 42a-4. Following the same procedure as that used for preparing compound 42a-2 shown in Scheme 42a and replacing i-PrSNa with /-BuSNa, compound 42a-4 was obtained as a white solid. LC-MS (ESI): mlz 567.2 [M + Hf.
Figure imgf000153_0003
10309] Synthesis of compound 42a-5. Following the same procedure as that used for preparing compound 42a-2 shown in Scheme 42a and replacing /-PrSNa with 4-F-PhSNa, compound 42a-5 was obtained as a white solid. LC-MS (ESI): mlz 605.1 [M + Hf.
Figure imgf000153_0004
(03101 Synthesis of compound 42a-6. Following the same procedure as that used for preparing compound 42a-2 shown in Scheme 42a and replacing /-PrSNa with 4-F-BnSNa, compound 42a-6 was obtained as a white solid. LC-MS (ESI): mlz 619.1 [M + Hf.
Figure imgf000154_0001
1031 II Synthesis of compound 42a-7. Following the same procedure as that used for preparing compound 42a-2 shown in Scheme 42a and replacing i-PrSNa with HOCH2CH2SH under the condition of ^CCVanhydrous DMF/80 °C/overnight, the anticipated hydroxyethyl thiol ether was obtained. Subsequently, acylation of the hydroxyethylthiol ether under the condition of
Ac20/Et3N/DCM/rt/ovemight, followed by oxidation of the thioether moiety under the condition of H2<I)2/acetone/60 0C/ovemight and soapnification of the acetate residue under the condition of 2C03 MeOH H20/rt 4 hr gave compound 42a-7 as a white solid. LC-MS (ESI): miz 555.1 [M + +.
Figure imgf000154_0002
]0312] Synthesis of compound 42a-8. Following the same procedure as that used for preparing compound 42a-7 described above and replacing HOCH2CH2SH with HOCH2CH2CH2SH, compound 42a-8 as obtained a white solid. LC-MS (ESI): miz 569.1 [M + Hf.
Scheme 42b
Figure imgf000154_0003
J0313J Step 1. Refer to Scheme 42b. To a stirred solution of compound 36e-l (1.0 g, 1.62 mmol) in anhydrous DMF ( 10 mL) was added potassium ethanethioate (KSAc) (277 mg, 2.43 mmol) at rt under an atmosphere of Ar. After stirring at 90 °C for 3 hr, the reaction mixture was cooled to rt and added ice water (60 mL). The suspension was filtered and the solid was washed with water and dried in vacuo to give crude compound 42b-l (700 mg, 83% purity based on LC-MS) as a pale yellow solid, which was used for the next step without further purification. LC-MS (ESI): mlz 521.1 [M + H]\
I0314I Step 2. To a stirred solution of compound 42b-2 (100 mg, 0.192 mmol) and 4- chlorotetrahydro-2H-pyran (46 mg, 0.384 mmol) in anhydrous DMF/EtOH (2 rnL/l mL) was added NaOH (15 mg, 0.38 mmol) at 0 °C under an atmosphere of Ar. After stirring at 50 °C for 16 hr, the reaction mixture was cooled to rt and added ice water (30 mL). The mixture was extracted with EtOAc (30 mL x 3) and the combined organic extracts were washed with brine and dried over anhydrous Na2S04. The solvent was removed and the residue was dried in vacuo to give crude compound 42b-2 (80 mg, 87% purity based on LC-MS) as a white solid, which was used for the next step without further purification. LC-MS (ESI): mlz 563.2 [M + H|\
103151 Step 3. To a stirred solution of compound 42b-2 (50 mg, 0.088 mmol) in AcOH (3 mL) was added H202 (3 mL, 30 wt.% in H20). After stirring at 50 °C for 3 hr, the reaction mixture was cooled to rt and added ice water (20 mL). The suspension was filtered and the solid was washed with water and purified by preparative HPLC to give compound 42b-3 (15 mg, 29% yield) as a white solid. LC-MS (ESI): mlz 595.2 [M + Hf; Ή NMR (500 MHz,
Figure imgf000155_0001
5 8.53 (q, J= 4.0 Hz, 1H), 7.97 (dd, J, = 8.5 Hz, J? = 5.5 Hz, 2H), 7.88 (s, 1 H), 7.61 (s, 1H), 7.41 (t, J= 8.5 Hz, 2H), 4.99 (q, J= 6.0 Hz, 1H), 4.41 (t, J = 10.5 Hz, 1H), 4.22 (d, J= 10.0 Hz, 1H), 4.04 - 3.93 (m, 2H), 3.52 (t, J = 11.5 Hz, 1H), 3.39 (s, 5H), 3.29 - 3.19 (m, 2H), 2.91 - 2.84 (m, 1 H), 2.84 (t, J = 4.5 Hz, I H), 1.96 - 1.90 (m, 1 H), 1.67 (m, J = 6.0 Hz, 1 H), 1.56 - 1.68 (m, 2H) ppm.
Figure imgf000155_0002
10316] Synthesis of compound 42b-4. Following the same procedure as that used for preparing compound 42b-3 shown in Scheme 42b and replacing 4-chlorotetrahydro-2/ -pyrane with bromocyclobutane, compound 42b-4 was obtained as a white solid. LC-MS (ESI): mlz 565.1 [M
+ Hf.
Figure imgf000156_0001
10317] Synthesis of compound 42b-5. Following the same procedure as that used for preparing compound 42b-3 shown in Scheme 42b and replacing 4-chlorotetrahydro-2H-pyrane with 3- bromooxetane, compound 42b-5 was obtained as a white solid. LC-MS (ESI): mlz 567.1 [M + +.
Figure imgf000156_0002
10318] Synthesis of compound 42b-6. Following the same procedure as that used for preparing compound 42b-3 shown in Scheme 42b and replacing 4-chlorotetrahydro-2H-pyrane with tert- butyl 3-bromoazetidine- 1 -carboxy late, compound 42b-6 was obtained as a white solid. LC-MS
+.
Figure imgf000156_0003
10319] Synthesis of compound 42b-7. Following the same procedure as that used for preparing compound 42b-3 shown in Scheme 42b and replacing 4-chlorotetrahydro-2H-pyrane with chlorocyclopentane, compound 42b-6 was obtained as a white solid. LC-MS (ESI): mlz 579.2 [M
Figure imgf000156_0004
|0320| Synthesis of compound 42b-8. Following the same procedure as that used for preparing compound 42b-3 shown in Scheme 42b and replacing 4-bromtetrahydro-2H-pyrane with tert- butyl 4-bromopiperidine- 1-carboxylate, compound 42b-8 was obtained as a white solid. LC-MS
(ESI): mlz 694.3 [M + Hf.
Figure imgf000157_0001
103211 Synthesis of compound 42b-9. Treatment of compound 42b-8 with 4.0 N HC1 in dioxane, followed by preparative HPLC purification gave compound 42b-9 as a white solid. LC-MS (ESI):
+.
Figure imgf000157_0002
103221 Synthesis of compound 42b-10. Treatment of compound 42b-9 with paraformalde in the presence of AcOH and NaBH3CN in EtOH, followed by preparative HPLC purification gave
. LC-MS (ESI): mlz 608.2 [M + Hf.
Figure imgf000157_0003
I0323| Synthesis of compound 42b-l l. Treatment of compound 42b-9 with 2,2,2-trifluoroethyl trifluoromethanesulfonate in the presence of Et3N in DCM at reflux, followed by preparative
42b- 11 as a white solid. LC-MS (ESI): mlz 676.2 [M + H]+.
Figure imgf000157_0004
103241 Synthesis of compound 42b-12. Treatment of compound 42b-9 with methyl
chloroformate in the presence of Et3N in DCM at rt, followed by preparative HPLC purification gave compound 42b-12 as a white solid. LC-MS (ESI): mlz 652.2 [M + H]+.
Figure imgf000158_0001
J0325I Step 1. Refer to Scheme 42c. To a stirred solution of compound 36b-3b (3 g, 4.75 mmol) in anhydrous DMF (30 mL) was added KSAc (650 mg, 5.7 mmol) at rt under an atmosphere of AT. After stirring at 90 °C for 3 hr, the reaction mixture was cooled to rt and added ice water (100 mL), The resulting suspension was filtered and the solid was washed with water and dried in vacuo to give crude compound 42c-l (2.2 g, 86% purity based on LC-MS) as a white solid, which was used for the next step without further purification. LC-MS (ESI): mlz 558.1 [M + Naf.
103261 Step 2. To a stirred solution of hydrochloric acid (2 M aq. solution, 1 mL) in acetonitrile (20 mL) was added NCS (994 mg, 7.47 mmol), followed by a solution of compound 43c-l (1.0 g, 1.86 mmol) in acetonitrile (2 mL) at 0 °C. After stirring at 0 °C for 3 hr, the reaction mixture was diluted with water (20 mL) and concentrated. The residue was extracted with EtOAc (50 mL x 3) and the combined organic extracts were washed with brine and dried over anhydrous Na2S04. The solvent was removed and the residue was dried in vacuo to give crude compound 42e-2 (600 mg, 87% purity based on LC-MS) as a white solid, which was used for the next step without further purification. LC-MS (ESI): mlz 582.1 [M + Naf.
I0327I Step 3. To a stirred solution of compound 42c-2 (150 mg, 0.26 mmol) in DCM (5 mL) was added methanamine hydrochloride (54 mg, 0.81 mmol), followed by an aqueous solution of 2CO3 (2 M, 0.5 mL) at 0 °C. After stirring at rt for 3 hr, the reaction mixture was diluted with water (5 mL). The mixture was extracted with DCM (20 mL x 3) and the combined organic extracts were washed with brine and dried over anhydrous Na2S04. The solvent was removed and the residue was dried in vacuo to give crude compound 42c-3 (120 mg, 85% purity based on LC- MS) as a white solid, which was used for the next step without further purification. LC-MS (ESI): mlz 555.1 [M + H]+.
10328| Step 4. Following the same procedure used for preparing compound 4-9 shown in Scheme 4 and replacing compound 4-7 with 42c-3, compound 42c-5 was obtained as a white solid. LC- MS (ESI): mlz 540.1 [M + Hf; Ή NMR (500 MHz,
Figure imgf000159_0001
δ 8.53 (q, J= 4.5 Hz, I H), 7.95 - 7.98 (m, 2H), 7.87 (s, IH), 7.59 (s, IH), 7.40 (t, J= 9.0 Hz, 2H), 6.97 (q, J= 4.5 Hz, IH), 4.95 (q, J= 6.0 Hz, IH), 4.35 (m, IH), 4.18 (d, J = 18.0 Hz, IH), 3.39 (s, 3H), 3.09 - 3.20 (m, 2H), 2.89
= 4.5 Hz, 3H), 2.59 (d, J= 5.0 Hz, 3H), 1.64 (d, J = 6.5 Hz, 3H) ppm.
Figure imgf000159_0002
|0329] Synthesis of compound 42c-6. Following the same procedure as that used for preparing compound 42c-5 shown in Scheme 42c and replacing methylamine with ammonia, compound -6 was obtained as a white solid. LC-MS (ESI): mlz 526.6 [M + Hf .
Figure imgf000159_0003
103301 Synthesis of compound 42c-7. Following the same procedure as that used for preparing compound 42c-5 shown in Scheme 42c and replacing methylamine with dimethylamine, compound 42c-7 was obtained as a white solid. LC-MS (ESI): mlz 554.1 [M + Hf,
Figure imgf000159_0004
103311 Synthesis of compound 42c-8. Following the same procedure as that used for preparing compound 42c-5 shown in Scheme 42c and replacing methylamine with morpholine, compound 42c-8 was obtained as a white solid. LC-MS (ESI): mlz 596.1 [M + Hf.
Figure imgf000160_0001
103321 Step 1. Refer to Scheme 42d. To a stirred solution of compound 42b-l (300 mg, 0.58 mmol) in acetonitrile (10 mL) was added N-chlorosuccinimide (1 15 mg, 0.86 mmol) and hydrochloric acid aqueous solution (1 M, 2 mL) at 0 °C. After stirring at 0 °C for 3 hr, the reaction mixture was diluted with ice water (40 mL). The suspension was filtered and the solid was washed with water and dried in vacuo to give crude compound 42d~l (300 mg, 90% purity based on LC-MS) as a white solid, which was used for the next step without further purification. LC- MS (ESI): mlz 545.1 [M + H]+.
103331 Step 2. To a stirred solution of piperidine (19 mg, 0.2 mmol) in anhydrous DMF (2 mL) was added K2C03 (28 mg, 0.2 mmol), followed by compound 42d-l (60 mg, 0.11 mmol) at rt. After stirring at rt for 2 hr, the reaction mixture was diluted with ice water (10 mL). The suspension was filtered and the solid was washed with water and purified by preparative HPLC to give compound 42d-2 (12 mg, 18.5% yield) as a white solid. LC-MS (ESI): mlz 594.2 [M + H ; Ή NMR (500 MHz, iZ-DMSO): δ 8.55 (q, J =5.0 Hz, 1H), 7.97 (dd, Ji = 5.5 Hz, J2= 9.0 Hz, 2H), 7.86 (s, 1H), 7.60 (s, 1H), 7.41 (t, J= 9.0 Hz, 2H), 4.95 (q, J= 5.0 Hz, IH), 4.39 (t, J = 10.5 Hz, IH), 4.21 (t, J= 14.5 Hz, lH), 3.39 (s, 3H), 3.23 - 3.20 (m, IH), 3.20 - 3.10 (m, 5H), 2.89 - 2.84 (m, IH), 2.84 (<L J= 4.0 Hz, 3H), 1.65 (d, J= 6.5 Hz, 3H), 1.56 (m, 3H), 1.23 (d,J= 4.5 Hz, 3H) ppm.
Figure imgf000160_0002
10334] Synthesis of compound 42d-3. Following the same procedure as that used for preparing compound 42d-2 shown in Scheme 42d and repiacing piperidine with pyrrolidine, compound 42d-3 was obtained as a white solid. LC-MS (ESI): mlz 580.2 [M + H]+.
Figure imgf000161_0001
] 03351 Synthesis of compound 42d-4. Following the same procedure as that used for preparing compound 42d-2 shown in Scheme 42d and replacing piperidine with iV-methylpiperizine, s a white solid. LC-MS (ESI): mlz 609.2 [M + H]+.
Figure imgf000161_0002
10336] Synthesis of compound 42d-5. Following the same procedure as that used for preparing compound 42d-2 shown in Scheme 42d and replacing piperidine with thiomorphol ine-dioxane, compound 42d-5 was obtained as a white solid. LC-MS (ESI): mlz 644.1 [M + Hf .
Figure imgf000161_0003
10337 J Synthesis of compound 42d-4. Following the same procedure as that used for preparing compound 42d-2 shown in Scheme 42d and replacing piperidine with N-methy lpiperi zine, as a white solid. LC-MS (ESI): mlz 609.2 [M + Hf.
Figure imgf000161_0004
103381 Synthesis of compound 42d-5. Following the same procedure as that used for preparing compound 42d-2 shown in Scheme 42d and replacing piperidine with thiomorpholine-dioxane, compound 42d-5 was obtained as a white solid. LC-MS (ESI): mlz 644.1 [M + Hf. Scheme 42β
Figure imgf000162_0001
|0337J Synthesis of compound 42e-l. Amide coupling of compound 42c-6 with cyclopropyl carboxylic acid in the presence of EDCI, DMAP and EtjN in DCM gave compound 42e-l as a whilte solid. LC-MS (ESI): mlz 594.1 [M + H]+.
{03381 Synthesis of compound 42e-2. Amide coupling of compound 42c-6 with /-butyl carboxylic acid in the presence of EDCI, DMAP and Et3N in DCM gave compound 42e-2 as a whilte solid. LC-MS (ESI): mlz 610.2 [M + H]+.
10338| icai activity of the compounds of the invention was determined using an HCV replicon assay. The lb_Huh-Luc/Neo-ET cell line persistently expressing a bicistronic genotype lb replicon in Huh 7 cells was obtained from ReBLikon GMBH. This cell line was used to test compound inhibition using luciferase enzyme activity readout as a measurement of compound inhibition of replicon levels.
III. Biological A ci uvin
103391 Biological activity of the compounds of the invention was determined using an HCV replicon assay. The 1 b_Huh-Luc Neo-ET cell line persistently expressing a bicistronic genotype lb replicon in Huh 7 cells was obtained from ReBLikon GMBH. This cell line was used to test compound inhibition using luciferase enzyme activity readout as a measurement of compound inhibition of replicon levels.
10340 [ On Day 1 (the day after plating), each compound was added in triplicate to the cells. Plates were incubated for 72 hours prior to running the luciferase assay. Enzyme activity was measured using a Bright-Glo Kit (cat. number E2620) manufactured by Promega Corporation. The following equation was used to generate a percent control value for each compound.
% Control = (Average Compound Value/Average Control)* 100
10341] The EC» value was determined using GraphPad Prism and the following equation: Y « Bottom + (Top-Bottom)/ (1+10Λ ((LogIC50-X)*HillSlope))
|0342] EC50 values of compounds were repeated several times in the replicon assay.
|0343] Compounds of the invention can inhibit multiple genotypes of HCV selected but not limited to la, l b, 2a, 3a, and 4a. The ECs0s are measured in the corresponding replicon assays that are similar to HCV lb replicon assay as described above.
(03441 Exemplary compounds of the disclosed invention are illustrated in the Tables attached as Appendix A and Appendix B. Apendix A shows inhibitory activity of the compound against HCV lb. The biological activity against HCV lb is indicated as being *, **, ***, or ***, which corresponds to EC*) ranges of EC50 > 1000 nM, 100 nM < EC50≤ 1000 nM, 10 nM < ECso≤ 100 nM, or EC50≤ 10 nM, respectively.
10345J Appendix A shows structures of compounds of the invention identified by ID NOD B243- B577, and ECJO values determined for the compounds. Of these, all of compounds B243-B282, B285-B289, B292, B294-B426, B429-B438, B440-B443, B445-B469, B471-B540, B542-B549, B551 -B552, B554-B556, B558-B559, B561 -B575 had a measured activity of either 10 nM < EC50≤ 100 nM, or EC50≤ 10 nM.
IV. Pharmaceutical Compositions
103461 In another aspect, the invention includes a pharmaceutical composition comprising one or more of the compounds named above, together with one or more pharmaceutically acceptable excipients or vehicles, and optionally other therapeutic and/or prophylactic ingredients. The composition may further include one, two or three anti-HCV agent(s) selected from the group consisting of interferon-alpha, ribavirin, cyclosporine derivatives, NS3 protease inhibitors, NS4B inhibitors, NS5A inhibitors, NS5B polymerase inhibitors, and p7 inhibitors.
10347] The compounds described herein may be formulated in a pharmaceutical composition comprising one or more compounds, optionally together with one or more pharmaceutically acceptable excipients or vehicles, and optionally other therapeutic and/or prophylactic ingredients. Such excipients are known to those of skill in the art. Pharmaceutically acceptable salts can be used in the compositions of the present invention and include, for example, mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates and the like; and the salts of organic acids such as acetates, propionates, malonates, benzoates and the like. A thorough discussion of pharmaceutically acceptable excipients and salts is available in Remington's Pharmaceutical Sciences, 18th Edition (Easton, Pennsylvania: Mack Publishing Company, 1990), and in
Handbook of Pharmaceutical Excipients, 6lb Edition, Ed. R. C. Rowe, P. J. Sheskey, and M. E. Quinn (American Pharmacists Association, 2009).
{03481 Depending on the intended mode of administration, the pharmaceutical compositions may be in the form of solid, semi-solid or liquid dosage forms, such as, for example, tablets, suppositories, pills, capsules, powders, liquids, suspensions, creams, ointments, lotions or the like, preferably in unit dosage form suitable for single administration of a precise dosage. The compositions will include an effective amount of the selected drug in combination with a pharmaceutically acceptable carrier and, in addition, may include other pharmaceutical agents, adjuvants, diluents, buffers, etc.
{0349| The compound comprised within the pharmaceutical composition includes isomers, racemic or non-racemic mixtures of isomers, or pharmaceutically acceptable salts or solvates thereof, together with one or more pharmaceutically acceptable carriers and optionally other therapeutic and/or prophylactic ingredients.
(03501 For solid compositions, conventional nontoxic solid carriers include, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talc, cellulose, glucose, sucrose, magnesium carbonate and the like.
103511 For oral administration, the composition will generally take the form of a tablet, capsule, a softgel capsule nonaqueous solution, suspension or syrup. Tablets and capsules are preferred oral administration forms. Tablets and capsules for oral use will generally include one or more commonly used carriers such as lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. When liquid suspensions are used, the active agent may be combined with emulsifying and suspending agents. If desired, flavoring, coloring and/or sweetening agents may be added as well. Other optional components for incorporation into an oral formulation herein include, but are not limited to, preservatives, suspending agents, thickening agents and the like.
{03521 In yet another aspect, provided herein is the use of the compounds of the invention in the manufacture of a medicament, e.g., for the treatment of hepatitis C.
m V. Method of Treating HCV
103531 Also provided is a method of treating hepatitis C. The method comprises administering to a subject in need thereof, a therapeutically effective amount of a compound of the invention, optionally contained in a pharmaceutical composition. A pharmaceutically or therapeutically effective amount of the composition will be delivered to the subject. The precise effective amount will vary from subject to subject and will depend upon the species, age, the subject's size and health, the nature and extent of the condition being treated, recommendations of the treating physician, and the therapeutics or combination of therapeutics selected for administration. Thus, the effective amount for a given situation can be determined and optimized by routine experimentation. The subject may be administered as many doses as is required to reduce and/or alleviate the signs, symptoms or causes of the disorder in question, or bring about any other desired alteration of a biological system. One of ordinary skill in the art of treating such diseases will be able, without undue experimentation and in reliance upon personal knowledge and the disclosure of this application, to ascertain a therapeutically effective amount of the compounds of this invention for a given disease.
[0354] A treatment regimen using one or more of the compounds provided herein, optionally in combination with one or more anti-HCV agents, will generally involve administering a therapeutically effective dose of the compound, which can readily be determined by one skilled in the art taken into account variables as previously described. A typical dose of a compound as provided herein will generally range from about 10 mg to 1000 mg a day. Representative dosage ranges include from about 10 mg to 900 mg a day, or from about 30 mg to about 700 mg a day, from about 50 mg to about 600 mg a day. A therapeutically effective dosage amount will typically range from 0.1 mg kg body weight to about 30 mg/kg body weight, or from about 0.1 mg kg body weight to about 15 mg kg body weight. The compound may be administered once or multiple times (two to three times) daily, once a week, twice a week, three times a week, etc., over a time course effective to achieve a positive virologic response. Representative courses of treatment include but are not limited to 10 weeks, 12 weeks, 24 weeks, 36 weeks, 48 weeks, and the like. A positive virologic response generally refers to undetectable HCV RNA in serum as measured by PCR and a biochemical response (normalization of aminotransferase levels).
Although one may gauge success of treatment by a histologic response (e.g., pathology of liver biopsy), post-treatment biopsies are generally less preferable to an assessment based upon a virologic response.
10103] The treatment compound may be one or more of the compounds of B243, B244, B245, B246, B248, B249, B252, B253, B255, B256, B257, B258, B259, B260, B261, B262,B263, B264, B265, B266, B267, B268, B269, B270, B271, B272, B273, B274, B275, B276, B277, B278, B279, B280, B281, B282, B283, B284, B285, B286, B287, B288, B289, B290, B291, B293, B294, B295, B296, B297, B298, B299, B300, B301, B302, B303, B304, B305, B306, B307, B308, B309, B310, B31 1, B312, B313, B314, B315, B316, B317, B318, B319, B320, B321, B322, B323, B326, B327, B328, B329, B330, B333, B334, B335, B336, B337, B338, B339, B340, B341. B342, B343, B344, B345, B346, B347, B348, B349, B350, B351, B352, B353, B354, B355, B356, B357, B358, B360, B361, B362, B363, B364, B365, B366, B367, B368, B369, B370, B371, B372, B373, B374, B375, B376, B377, B378, B379, B380, B381, B382, B383, B384, B385, B386, B387, B388, B389, B390, B391, B392, B393, B394, B395, B396, B397, B398, B399, B400, B401, B402, B403, B404, B405, B406, B407, B408, B409, B410, B41 1, B412, B413, B414, B415, B416, B417, B418, B419, B420, B421, B422, B423, B424, B425, B426, B427, B428, B429, B430, B431, B432, B433, B434, B435, B436, B437, B438, B439, B440, B441, B442, B443, B444, B446, B447, B448, B449, B450, B451, B452, B453, B454, B455, B456, B457, B458, B459, B460, B461 , B462, B463, B464, B465, B466, B467, B468, B469, B470, B471, B472, B473, B474, B475, B476, B477, B478, B479, B480, B481, B482, B483, B484, B485, B486, B487, B488, B489, B490, B491, B492, B493, B494, B495, B496, B497, B498, B499, B500, B501, B502, B503, B504, B505, B506, B507, B508, B509, B510, B511, B512, B513, B514, B515, B516, B517, B518, B519, B520, B521, B522, B523, B524, B525, B526, B527, B528, B529, B530, B531, B532, B533, B534, B535, B536, B537, B538, B539, B540, B541, B542, B543, B544, B545, B546, B547, B548, B549, B550, B551, B552, B553, B554, B555, B556, B557, B558, B561, B562, B563, B564, B565, B566, B567, B568, B569, B570, B571, B572, B573, B574, B575, B576, and B577. Specific compounds include those from Group 1, Group 2, or Group 3 above.
103551 The administering may be by oral administration. Also disclosed is a compound as named above for use in treating HCV infection in an infected subject.
m 103561 Generally, treatment by administering a compound as provided herein is effective to result in at least a 2 log reduction in HCV RNA levels over the course of treatment. In some cases HCV RNA will still remain positive (detectable) after a course of treatment, although ideally, treatment is effective to achieve undetectable HCV RNA after completing a round of therapy. Subjects may also be assessed following completion of therapy to confirm maintenance of a positive and sustained virologic response (undetectable HCV RNA levels). Follow-up assessments may be carried out over the course of months or even years following treatment.
VI. ComblnationTheraDV
J0357[ The compounds of the present invention and their isomeric forms and pharmaceutically acceptable salts thereof are useful in treating and preventing HCV infection alone or when used in combination with other compounds targeting viral or cellular elements or functions involved in the HCV life cycle. Classes of compounds useful in the invention may include, without limitation, all classes of HCV an ivirals, both direct-acting and indirect-acting ('cell-targeted' inhibitors of HCV replication). For combination therapies, mechanistic classes of agents that may be useful when combined with the compounds of the present invention include, for example, nucleoside and non-nucleoside inhibitors of the HCV protease inhibitors, helicase inhibitors, NS5A inhibitors, NS4B inhibitors and medicinal agents that functionally inhibit the internal ribosomal entry site (IRES), other NS5B inhibitors and other medicaments that inhibit HCV cell attachment or virus entry, HCV RNA translation, HCV RNA transcription, replication or HCV maturation, assembly or virus release.
103581 Specific compounds in these classes and useful in the invention include, but are not limited to, linear, macrocyclic, and heterocyclic HCV protease inhibitors such as telaprevir (VX- 950), boceprevir (SCH-503034), narlaprevir (SCH-900518), ITMN-191 (R-7227), TMC-435350 (a.k.a. TMC-435), M -7009, M -5172, Bl-201335, BMS-650032, ACH- 1625, ACH-2784, ACH-1095 (HCV NS4A protease co-factor inhibitor), AVL-181, AVL-192, VX-813, PHX-1766, PHX-2054, 1DX-136, IDX-316, IDX-320, GS-9256, GS-9265, GS-9451, ABT-450, EP-013420 (and congeners) and VBY-376; the nucleosidic HCV polymerase (replicase) inhibitors useful in the invention include, but are not limited to, ALS2200, R7128, PSI-7851, PS1-7977 (GS-7977), PSI-938, PSI-879, PSI-6130, IDX-184, IDX-102, ΓΝΧ-189, R1479, R1626, UNX-08189, and various other nucleoside and nucleotide analogs and HCV inhibitors including, but not limited to, those derived from 2'-C-methyl modified nucleos(t)ides, 4'-aza modified nucleos(t)ides, and T- deaza modified nucleos(t)ides. NS5A inhibitors useful in the invention, include, but are not limited to, ΡΡΙ-46 , PPI-668, BMS-790052, BMS-824393, GS-5885, EDP-239, ACH-2928, AZD-7295, IDX-719, IDX-380, ABT-267, GS -2336805, CF-102, A-831 and ΓΓΜΝ-9916. Non-nucleosidic HCV polymerase (replicase) inhibitors useful in the invention, include, but are not limited to, VCH-759, VCH-916, VCH-222 (VX-222), ANA-598, M -3281, ABT-333, ABT- 072, PF-00868554 (filibuvir), BI-207127, GS-9190, A-837093, GS -625433, J T-109, GL- 59728 and GL-60667. HCV P7 inhibitors useful in the present invention include BIT-225 and other P7 inhibitors, as well as HCV NS4B inhibitors including but not limited to histamine agents that antagonize HCV NS4B function.
J0359I In addition, NS5B inhibitors of the present invention may be used in combination with cyclophyllin and immunophyllin antagonists (eg, without limitation, DEBIO compounds, NM- 81 1, SCY-635, EP-CyP282, as well as cyclosporine and its derivatives), kinase inhibitors, inhibitors of heat shock proteins (e.g., HSP90 and HSP70), other immunomodulatory agents that may include, without limitation, interferons (-alpha, -beta, -omega, -gamma, -lambda or synthetic) such as Intron A™, Roferon-A™, Canferon-A300™, Advaferon™, Infergen™, Humoferon™, Sumiferon MP™, Alfaferone™, IFN-β™, Feron™ and the like; polyethylene glycol derivatized (pegylated) interferon compounds, such as PEG interferon-a-2a (Pegasys™), PEG interferon-o-2b (PEGIntron™), pegylated IFN-a-conl and the like; long acting formulations and derivatizations of interferon compounds such as the albumin-fused interferon, Albuferon™ , Locteron™ and the like; interferons with various types of controlled delivery systems (e.g.
ITCA-638, omega-interferon delivered by the DUROS™ subcutaneous delivery system);
compounds that stimulate the synthesis of interferon in cells, such as resiquimod and the like; interleukins; compounds that enhance the development of type 1 helper T cell response, such as SCV-07 and the like; TOLL-like receptor agonists such as CpG-10101 (actilon), isotorabine, ANA773, SD-101, IMO-2125, and the like; thymosin a -1; ANA-245 and ANA-246; histamine dihydrochloride; propagerrnanium; tetrach lorodecaoxide; ampligen; IMP-321; KRN-7000:
antibodies, such as civacir, XTL-6865 and the like and prophylactic and therapeutic vaccines such as GI-5005, TG-4040, InnoVac C, HCV E1 E2/MF59 and the like. In addition, any of the above- described methods involving administering an NS5B inhibitor, a Type I interferon receptor agonist (e.g., an IFN-a) and a Type II interferon receptor agonist (e.g., an IFN-γ) can be augmented by administration of an effective amount of a TNF- antagonist Exemplary, non- limiting TNF-a antagonists that are suitable for use in such combination therapies include ENBREL™, REMICADE™ and HUMI A™.
J0360J NS5B inhibitors of the present invention may also be used with alternative forms of interferons and pegylated interferons, ribavirin or its analogs (e.g., tarabavarin, levoviron), microRNA, small interfering RNA compounds (e.g., S1RPLEX-140-N and the like) and microR A agents (such as micro-RNA-122), nucleotide or nucleoside analogs, multifunction inhibitors such as nitazoxanide, immunoglobulins, hepatoprotectants, anti-inflammatory agents and other direct and indirect inhibitors of HCV replication. Inhibitors of other targets in the HCV life cycle include NS3 helicase inhibitors; NS4A co- factor inhibitors; and sense oligonucleotide inhibitors, such as ISIS- 14803, ISlS-11, AVI-4065 and the like; vector-encoded short hairpin RNA (shRNA); HCV specific ribozymes such as heptazyme, RPI-13919 and the like; entry inhibitors such as HepeX-C, HuMax-HepC and the like; alpha glucosidase inhibitors such as celgosivir, UT-231B and the like; KPE-02003002 and BIVN 401 and IMPDH inhibitors.
J 0361 J Other illustrative HCV inhibitor compounds include those disclosed in the following publications: U.S. Pat. No. 5,807,876; U.S. Pat. No. 6,498,178; U.S. Pat. No. 6,344,465; U.S. Pat. No. 6,054,472; U.S. Pat. No. 7,759,495; U.S. Pat. No. 7,704,992; U.S. Pat. No. 7,741, 347; WO 02/04425; WO 03/007945; WO 03/010141; WO 03/000254; WO 03/037895; WO 02/100851; WO 02/100846; EP 1256628; WO 02/18369; WO 05/073216; WO 05/073195; WO 08/021927; US 20080050336; US 20080044379; US 2009004716; US 20090043107; US 20090202478; US 20090068140; WO 10/096302; US 20100068176; WO 10/094977; WO 07/138242; WO
10/096462; US 2010091413; WO 10/075380; WO 10/062821 ; WO 10/09677; WO 10/065681 and WO 10/065668.
103621 Additionally, combinations of, for example, ribavirin a NS3 protease inhibitor, a replicase inhibitor and interferon, may be administered as multiple combination therapy with at least one of the compounds of the present invention. The present invention is not limited to the
aforementioned classes or compounds and contemplates known and new compounds and combinations of biologically active agents (see, for example, lebl et al. "Host cell targets in HCV therapy: novel strategy or proven practice, Antiviral Chemistry & Chemotherapy 16:69-90, which is incorporated by reference herein). It is intended that combination therapies of the present invention include any chemically compatible combination of a compound of this inventive group with other compounds of the inventive group or other compounds outside of the inventive group, as long as the combination does not eliminate the anti-viral activity of the compound of this inventive group or the anti-viral activity of the pharmaceutical composition itself
[0363 J Combination therapy can be sequential, that is treatment with one agent first and then a second agent (for example, where each treatment comprises a different compound of the invention or where one treatment comprises a compound of the invention and the other comprises one or more biologically active agents) or it can be treatment with both agents at the same time (concurrently). Sequential therapy can include a reasonable time after the completion of the first therapy before beginning the second therapy. Treatment with both agents at the same time can be in the same daily dose or in separate doses. The dosages for both concurrent and sequential combination therapy will depend on absorption, distribution, metabolism and excretion rates of the components of the combination therapy as well as other factors known to one of skill in the art. Dosage values will also vary with the severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens and schedules may be adjusted over time according to the individual's need.
[0364| All publications and patent applications cited in this specification are herein incorporated by reference as if each individual publication or patent application were specifically and individually indicated to be incorporated by reference.
10365] Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be readily apparent to one of ordinary skill in the art in light of the teachings of this invention that certain changes and modifications may be made thereto without departing from the spirit or scope of the invention as defined in the appended claims.
Figure imgf000171_0001
ApptmllxA Pagei
Figure imgf000172_0001
Appendix A Page 2
Figure imgf000173_0001
A endix A PI¾B 3 A endix A
Figure imgf000174_0001
Ap en ix A Fs§& 4 Appendix A
Figure imgf000175_0001
Appendix A Pap 5
Figure imgf000176_0001
Figure imgf000177_0001
ApfMfHjixA
Figure imgf000178_0001
Figure imgf000179_0001
Figure imgf000180_0001
i ill Appendix A
mt
- ....
S389 SS54 em
m& —
mm ****
**** ·**
Mi
OSS SS1S
Apfiemi«APag»li
Figure imgf000182_0001
Figure imgf000183_0001
Figure imgf000184_0001
ApptmflxAPaftM Appendix A
Figure imgf000185_0001
Ap endix A PafsIS
Figure imgf000186_0001
Appendix A Pagff 16 Appendix A
Figure imgf000187_0001
Appendix A Psg* 17
Figure imgf000188_0001
Figure imgf000189_0001
Figure imgf000190_0001
AppemfccA
Figure imgf000191_0001
Appendix A Pane 21
Figure imgf000192_0001
AppncttxAPagttt Appendix A
Figure imgf000193_0001
Apend A Pa 23
Figure imgf000194_0001
Figure imgf000195_0001
Figure imgf000196_0001
Figure imgf000197_0001
Figure imgf000198_0001
Figure imgf000199_0001
Figure imgf000200_0001
Figure imgf000201_0001
Figure imgf000202_0001
Figure imgf000203_0001
Figure imgf000204_0001
Figure imgf000205_0001
Figure imgf000206_0001
Łppwtdbtt
Figure imgf000207_0001
Figure imgf000208_0001
Figure imgf000209_0001
Figure imgf000210_0001
Appendix B
Figure imgf000211_0001
Figure imgf000212_0001
Figure imgf000213_0001

Claims

IT IS CLAIMED:
1. A com ound havin the structural formula:
Figure imgf000214_0001
Figure imgf000214_0002
21}
Figure imgf000215_0001
214
Figure imgf000216_0001
Figure imgf000217_0001
216
Figure imgf000218_0001
117
Figure imgf000219_0001
Figure imgf000220_0001
°c ¾ O' o TT¾
Figure imgf000220_0002
s and
X' is selected from =0, =N-OH, .23CH3, and R'3 is selected from -CH3 and -OCH3.
2. The compound of claim 1, wherein: R'| is para-Y, X' is =0, and R'3 is -CH3.
3. The compound of claim I, selected from the group consisting of B243, B244, B245, B246, B248, B249, B252, B253, B255, B256, B257, B258, B259, B260, B261, B262.B263, B264, B265, B266, B267, B268, B269, B270, B271, B272, B273, B274, B275, B276, B277, B278, B279, B280, B281, B282, B283, B284, B285, B286, B287, B288, B289, B290, B291, B293, B294, B295, B296, B297, B298, B299, B300, B301, B302, B303, B304, B305, B306, B307, B308, B309, B310, B31 1 , B312, B313, B314, B315, B316, B317, B3 18, B319, B320, B321 , B322, B323, B326, B327, B328, B329, B330, B333, B334, B335, B336, B337, B338, B339, B340, B341 , B342, B343, B344, B345, B346, B347, B348, B349, B350, B351 , B352, B353, B354, B355, B356, B357, B358, B360, B361, B362, B363, B364, B365, B366, B367, B368, B369, B370, B371, B372, B373, B374, B375, B376, B377, B378, B379, B380, B381 , B382, B383, B384, B385, B386, B387, B388, B389, B390, B391 , B392, B393, B394, B395, B396, B397, B398, B399, B400, B401, B402, B403, B404, B405, B406, B407, B408, B409, B410, B41 1, B412, B413, B414, B 15, B416, B417, B418, B419, B420, B421, B422, B423, B424, B425, B426, B427, B428, B429, B430, B431 , B432, B433, B434, B435, B436, B437, B438, B439, B440, B441, B442, B443, B444, B446, B447, B448, B449, B450, B451 , B452, B453, B454, B455, B456, B457, B458, B459, B460, B461, B462, B463, B464, B465, B466, B467, B468, B469, B470, B471, B472, B473, B474, B475, B476, B477, B478, B479, B480, B48 I , B482, B483, B484, B485, B486, B487, B488, B489, B490, B491 , B492, B493, B494, B495, B496, B497, B498, B499, B500, B501 , B502, B503, B504, B505, B506, B507, B508, B509, B510, B51 1 , B512, B513, B514, B515, B516, B517, B518, B519, B520, B521 , B522, B523, B524, B525, B526, B527, B528, B529, B530, B531 , B532, B533, B534, B535, B536, B537, B538, B539, B540, B541, B542, B543, B544, B545, B546, B547, B548, B549, B550, B551, B552, B553, B554, B555, B556, B557, B558, B561, B562, B563, B564, B565, B566, B567, B568, B569, B570, B571, B572, B573, B574, B575, B576, and B577.
4. The compound of claim 1,
Figure imgf000221_0001
5. The compound of claim 1, compound having the structural formula:
wherein:
R'i is selected from -H, -F, -C
Figure imgf000221_0002
I, -Br, -1, and
Figure imgf000221_0003
Figure imgf000222_0001
221
Figure imgf000223_0001
X' is selected from =0, =N-OH, =N-0(CH2CH20)o.gCH,, and =N-0(CH2)] 23CH3, and R'3 is selected from -CH3 and -OCH3.
6. The compound of claim 5, wherein: R'i is para-?, X' is =0, and R'3 is -CHj.
7. The compound of claim 5, selected from the group consisting of B243, B244, B245, B246, B248, B249, B252, B253, B255, B256, B257, B258, B259, B260, B261 , B262.B263, B264, B265, B266, B267, B268, B269, B270, B27I, B272, B273, B274, B275, B276, B277, B278, B279, B280, B281, B282, B285, B286, B289, B300, B302, B306, B307, B308, B309, B310, B31 1, B314, B316, B317, B318, B319, B320, B333, B335, B336, B337, B338, B343, B344, B345, B346, B347, B348, B349, B350, B351 , B352, B353, B354, B373, B378, B379, B381, B383, B384, B385, B386, B388, B389, B390, B391 , B392, B394, B395, B397, B399, B400, B401 , B402, B404, B405, and B409.
8. The compound of claim 1, compound having the structural formula:
Figure imgf000224_0001
wherein:
Figure imgf000224_0002
M io F_- > o X #,
Figure imgf000225_0001
234
Figure imgf000226_0001
H0 Δ 0¾ - j o¾ H0H0 I 0¾ <U o
Figure imgf000226_0002
X' is selected from -O, =N-OH,
Figure imgf000226_0003
and R'i is selected from -CH3 and -OCH3.
9. The compound of claim 8, wherein: 1 is para-F, X' is =0, and R'3 is -CH3.
10. The compound of claim 8, selected from the group consisting of B283, B284, B287, B288,
Figure imgf000226_0004
1 1. The compound of claim 1 , compound having the structural formula:
Figure imgf000227_0001
wherein:
selected from -H, -F, -CI, -Br, -I,
Figure imgf000227_0002
Figure imgf000227_0003
Figure imgf000228_0001
Figure imgf000229_0001
Χ' is selected from =0, =N-OH,
Figure imgf000229_0002
and R'3 is selected from -CH3 and -OCH3.
12. The compound of claim 1 1, wherein: R'i is para-F, X' is =0, and R'3 is -CH3.
13. The compound of claim 1 1, selected from the group consisting of B294, B295, B296, B297, B298, B299, B301 , B303, B304, B305, B312, B313, B315, B321, B322, B323, B326, B327, B328, B329, B330, B334, B339, B340, B341, B342, B355, B356, B357, B358, B360, B361, B362, B363, B364, B365, B366, B367, B368, B369, B370, B371, B372, B374, B375, B376, B377, B380, B382, B387, B393, B396, B398, B403, B406, B407, B408, B410, B41 1, B412, B413, B414, B415, B416, B417, B418, B419, B420, B421, B425, B427, B428, B429, B430, B431, B433, B434, B435, B436, B437, B438, B439, B440, B441, B442, B444, B448, B449, B450, B451, B452, B453, B454, B457, B458, B460, B461, B462, B465, B468, B471, B472, B473, B474, B475, B476, B477, B480, B481, B482, B483, B484, B485, B486, and B490.
14. A compound having the structural formula:
m 0'-¾'CH3
o
wherein:
R'i is selected from para-Έ
Figure imgf000230_0001
;
R'2 is selected from -OH and -SOjMe; and
R'i is selected from -CH3 and -OCH3, as exemplified by B324, B325, B331, and B332.
15. A compound having the structural formula:
wherein:
R'i is selected from para-F
Figure imgf000230_0002
R*2 is selected from -OH and -SOjMe; and
R'3 is selected from -CH3 and -OCH3, as exemplified by compound B359.
16. A pharmaceutical composition comprising a compound of any one of claims 1-15 or its pharmaceutically acceptable salts, together with one or more pharmaceutically acceptable excipients or vehicles, and optionally other therapeutic and/or prophylactic ingredients.
17. The composition of claim 16, with further comprises additional one, two or three anti-HCV agent(s) selected from the group consisting of interferon-alpha, ribavirin, cyclosporine derivatives, HCV NS3 protease inhibitors, HCV NS4B inhibitors, HCV NS5A inhibitors, HCV NS5B polymerase inhibitors, and p7 inhibitors.
18. A method of treating HCV infection in a subject comprising administering to the subject, a pharmaceutically acceptable dose of the compound of any one of claims 1-15, and continuing the administering until a selected reduction in the subject's HCV titer is achieved.
1 . The method of claim 18, wherein the compound is selected from the group consisting of B243, B244, B245, B246, B248, B249, B252, B253, B255, B256, B257, B258, B259, B260, B261, B262,B263, B264, B265, B266, B267, B268, B269, B270, B271, B272, B273, B274, B275, B276, B277, B278, B279, B280, B281, B282, B283, B284, B285, B286, B287, B288, B289, B290, B291, B293, B294, B295, B296, B297, B298, B299, B300, B301, B302, B303, B304, B305, B306, B307, B308, B309, B310, B31 1, B312, B313, B314, B315, B316, B317, B318, B319, B320, B321, B322, B323, B326, B327, B328, B329, B330, B333, B334, B335, B336, B337, B338, B339, B340, B341, B342, B343, B344, B345, B346, B347, B348, B349, B350, B351 , B352, B353, B354, B355, B356, B357, B358, B360, B361, B362, B363, B364, B365, B366, B367, B368, B369, B370, B371, B372, B373, B374, B375, B376, B377, B378, B379, B380, B381, B382, B383, B384, B385, B386, B387, B388, B389, B390, B391, B392, B393, B394, B395, B396, B397, B398, B399, B400, B401, B402, B403, B404, B405, B406, B407, B408, B409, B410, B41 1, B412, B413, B414, B415, B416, B417, B418, B419, B420, B421, B422, B423, B424, B425, B426, B427, B428, B429, B430, B431, B432, B433, B434, B435, B436, B437, B438, B439, B440, B441, B442, B443, B444, B446, B447, B448, B449, B450, B451, B452, B453, B454, B455, B456, B457, B458, B459, B460, B461, B462, B463, B464, B465, B466, B467, B468, B469, B470, B471, B472, B473, B474, B475, B476, B477, B478, B479, B480, B481, B482, B483, B484, B485, B486, B487, B488, B489, B490, B491, B492, B493, B494, B495, B496, B497, B498, B499, B500, B501, B502, B503, B504, B505, B506, B507, B508, B509, B510, B51 1, B512, B5 I 3, B514, B515, B516, B517, B518, B519, B520, B521 , B522, B523, B524, B525, B526, B527, B528, B529, B530, B531 , B532, B533, B534, B535, B536, B537, B538, B539, B540, B541, B542, B543, B544, B545, B546, B547, B548, B549, B550, B551, B552, B553, B554, B555, B556, B557, B558, B561, B562, B563, B564, B565, B566, B567, B568, B569, B570, B571 , B572, B573, B574, B575, B576, and B577.
2M
20. The method of claim 18, wherein the compound is selected from the group consisting of B243, B244, B245, B246, B248, B249, B252, B253, B255, B256, B257, B258, B259, B260, B261, B262.B263, B264, B265, B266, B267, B268, B269, B270, B271, B272, B273, B274, B275, B276, B277, B278, B279, B280, B281, B282, B285, B286, B289, B300, B302, B306, B307, B308, B309, B310, B31 1, B314, B316, B317, B318, B319, B320, B333, B335, B336, B337, B338, B343, B344, B345, B346, B347, B348, B349, B350, B351, B352, B353, B354, B373, B378, B379, B381, B383, B384, B385, B386, B388, B389, B390, B391, B392, B394, B395, B397, B399, B400, B401, B402, B404, B405, and B409.
21. The method of claim 18, wherein the compound is selected from the group consisting of B283, B284, B287, B288, B290, B291, B293, B422, B423, B424, B426, B432, B443, B446, B447, B455, B456, B459, B463, B464, B466, B467, B470, B478, B479, B487, B488, B489, B491, B492, B493, B494, B495, B496, B497, B498, B499, B500, B501, B502, B503, B504, B505, B506, B507, B508, B509, B510, BS 1 1, B512, B513, B514, B515, B516, B517, B518, B519, B520, B521, B522, B523, B524, B525, B526, B527, B528, B529, B530, B531, B532, B533, B534, B535, B536, B537, B538, B539, B540, B541, B542, B543, B544, B545, B546, B547, B548, B549, B550, B551, B552, B553, B554, B555, B556, B557, B558, B561, B562, B563, B564, B565, B566, B567, B568, B569, B570, B571, B572, B573, B574, B575, B576, and B577.
22. The method of claim 18, wherein the compound is selected from the group consisting of B294, B295, B296, B297, B298, B299, B301, B303, B304, B305, B312, B313, B315, B321, B322, B323, B326, B327, B328, B329, B330, B334, B339, B340, B341, B342, B355, B356, B357, B358, B360, B361, B362, B363, B364, B365, B366, B367, B368, B369, B370, B371, B372, B374, B375, B376, B377, B380, B382, B387, B393, B396, B398, B403, B406, B407, B408, B410, B41 1, B412, B413, B414, B415, B416, B417, B418, B419, B420, B421, B425, B427, B428, B429, B430, B431, B433, B434, B435, B436, B437, B438, B439, B440, B441 , B442, B444, B448, B449, B450, B451, B452, B453, B454, B457, B458, B460, B461, B462, B465, B468, B471, B472, B473, B474, B475, B476, B477, B480, B481, B482, B483, B484, B485, B486, and B490.
23i
23. The method of claim 18, wherein the administering is by oral administration.
24. A compound of any one of claims 1-15 for use in treating HCV infection in an infected subject.
PCT/US2013/038275 2012-04-25 2013-04-25 Inhibitors of hepatitis c virus WO2013163466A1 (en)

Applications Claiming Priority (10)

Application Number Priority Date Filing Date Title
US13/456,181 2012-04-25
US13/456,181 US9085587B2 (en) 2010-10-26 2012-04-25 Inhibitors of hepatitis C virus
US201261641769P 2012-05-02 2012-05-02
US61/641,769 2012-05-02
US13/544,261 US8614207B2 (en) 2010-10-26 2012-07-09 Inhibitors of hepatitis C virus
US13/544,261 2012-07-09
US201261722779P 2012-11-05 2012-11-05
US61/722,779 2012-11-05
US201261746548P 2012-12-27 2012-12-27
US61/746,548 2012-12-27

Publications (1)

Publication Number Publication Date
WO2013163466A1 true WO2013163466A1 (en) 2013-10-31

Family

ID=49483903

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/038275 WO2013163466A1 (en) 2012-04-25 2013-04-25 Inhibitors of hepatitis c virus

Country Status (1)

Country Link
WO (1) WO2013163466A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2014500861A (en) * 2010-10-26 2014-01-16 プレシディオ ファーマシューティカルズ インコーポレイテッド Hepatitis C virus inhibitor
CN105418620A (en) * 2014-09-23 2016-03-23 天津药明康德新药开发有限公司 Synthesis method for 4-(tert-butoxycarbonyl) octahydrofuro[3,2-b] pyridine-6-carboxylic acid
CN113173911A (en) * 2021-04-20 2021-07-27 梯尔希(南京)药物研发有限公司 Synthesis method of imidacloprid metabolite 5-hydroxy imidacloprid

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100063068A1 (en) * 2008-09-11 2010-03-11 Bristol-Myers Squibb Company Compounds for the Treatment of Hepatitis C
US20100093694A1 (en) * 2008-09-11 2010-04-15 Bristol-Myers Squibb Company Compounds for the Treatment of Hepatitis C
US20120015907A1 (en) * 2010-04-21 2012-01-19 Novartis Ag Furopyridine compounds and uses thereof
US20120022046A1 (en) * 2009-02-27 2012-01-26 Byrd Chelsea M Thienopyridine Derivatives for the Treatment and Prevention of Dengue Virus Infections
US20120251497A1 (en) * 2010-10-26 2012-10-04 Min Zhong Inhibitors of hepatitis c virus

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100063068A1 (en) * 2008-09-11 2010-03-11 Bristol-Myers Squibb Company Compounds for the Treatment of Hepatitis C
US20100093694A1 (en) * 2008-09-11 2010-04-15 Bristol-Myers Squibb Company Compounds for the Treatment of Hepatitis C
US20120022046A1 (en) * 2009-02-27 2012-01-26 Byrd Chelsea M Thienopyridine Derivatives for the Treatment and Prevention of Dengue Virus Infections
US20120015907A1 (en) * 2010-04-21 2012-01-19 Novartis Ag Furopyridine compounds and uses thereof
US20120251497A1 (en) * 2010-10-26 2012-10-04 Min Zhong Inhibitors of hepatitis c virus

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2014500861A (en) * 2010-10-26 2014-01-16 プレシディオ ファーマシューティカルズ インコーポレイテッド Hepatitis C virus inhibitor
US9085587B2 (en) 2010-10-26 2015-07-21 Presidio Pharmaceuticals, Inc. Inhibitors of hepatitis C virus
US9309260B2 (en) 2010-10-26 2016-04-12 Presidio Pharmaceuticals, Inc. Inhibitors of hepatitis C virus
CN105418620A (en) * 2014-09-23 2016-03-23 天津药明康德新药开发有限公司 Synthesis method for 4-(tert-butoxycarbonyl) octahydrofuro[3,2-b] pyridine-6-carboxylic acid
CN105418620B (en) * 2014-09-23 2018-05-04 天津药明康德新药开发有限公司 A kind of synthetic method of 4- (tertbutyloxycarbonyl) octahydros furans simultaneously [3,2-b] pyridine -6- carboxylic acids
CN113173911A (en) * 2021-04-20 2021-07-27 梯尔希(南京)药物研发有限公司 Synthesis method of imidacloprid metabolite 5-hydroxy imidacloprid

Similar Documents

Publication Publication Date Title
US8865756B2 (en) Inhibitors of HCV NS5A
US9150554B2 (en) Fused ring inhibitors of hepatitis C
EP2373172B1 (en) Inhibitors of hcv ns5a
AU2011320696B2 (en) Inhibitors of Hepatitis C Virus
US8877707B2 (en) Inhibitors of HCV NS5A
US8999967B2 (en) Tricyclic fused ring inhibitors of hepatitis C
EP2575475A1 (en) Inhibitors of hcv ns5a
US8822520B2 (en) Substituted bicyclic HCV inhibitors
WO2010111673A1 (en) Substituted bicyclic hcv inhibitors
WO2013163466A1 (en) Inhibitors of hepatitis c virus
AU2012360910A1 (en) Quinazolinone derivatives as HCV inhibitors

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 13781522

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13781522

Country of ref document: EP

Kind code of ref document: A1