OA17135A - Substituted nucleosides, nucleotides and analogs thereof - Google Patents

Substituted nucleosides, nucleotides and analogs thereof Download PDF

Info

Publication number
OA17135A
OA17135A OA1201400433 OA17135A OA 17135 A OA17135 A OA 17135A OA 1201400433 OA1201400433 OA 1201400433 OA 17135 A OA17135 A OA 17135A
Authority
OA
OAPI
Prior art keywords
optionally substituted
alkyl
formula
hydrogen
group
Prior art date
Application number
OA1201400433
Inventor
Guangyi Wang
David Bernard Smith
Leonid Beigelman
Jerome Deval
Marija Prhavc
Original Assignee
Alios Biopharma, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Alios Biopharma, Inc. filed Critical Alios Biopharma, Inc.
Publication of OA17135A publication Critical patent/OA17135A/en

Links

Abstract

Disclosed herein are nucleosides, nucleotides and analogs thereof, pharmaceutical compositions that include one or more of nucleosides, nucleotides and analogs thereof, and methods of synthesizing the same. Also disclosed herein are methods of ameliorating and/or treating a disease and/or a condition, including an infection from a paramyxovirus and/or an orthomyxovirus, with a nucleoside, a nucleotide and an analog thereof.

Description

[0001] The présent application relates to the fields of chemistry, biochemistry and medicine. More particularly, disclosed herein are nucleoside, nucléotides and analogs thereof, pharmaceutical compositions that inciude one or more nucleosides, nucléotides and analogs thereof, and methods of synthesizing the same. Also disclosed herein are methods of ameliorating and/or treating a paramyxovirus and/or an orthomyxovirus viral infection with one or more nucleosides, nucléotides and analogs thereof.
Description [0002] Respiratory viral infections, including upper and lower respiratory tract viral infections, infects and is the leading cause of death of millions of people each year. Upper <
respiratory tract viral infections involve the nose, sinuses, pharynx and/or larynx. Lower respiratory tract viral infections involve the respiratory system below the vocal cords, including the trachea, primary bronchi and lungs.
[0003] Nucleoside analogs are a class of compounds that hâve been shown to exert antiviral activity both in vitro and in vivo, and thus, hâve been the subject of widespread research for the treatment of viral infections. Nucleoside analogs are usually therapeutically inactive compounds that are converted by host or viral enzymes to their respective active antimetabolites, which, in tum, may inhibit polymerases involved in viral or cell prolifération. The activation occurs by a variety of mechanisms, such as the addition of one or more phosphate groups and, or in combination with, other metabolic processes.
SUM MARY [0004] Some embodiments disclosed herein relate to a compound of Formula (I), Formula (II) and/or Formula (III), or a pharmaceutically acceptable sait of the foregoing.
[0005] Some embodiments disclosed herein relate to methods of ameliorating and/or treating a paramyxovirus viral infection that can inciude administering to a subject suffering from the paramyxovirus viral infection an effective amount of one or more compounds of Formula (I), Formula (II) and/or Formula (ΙΠ), or a pharmaceutically acceptable sait of the foregoing, or a pharmaceutical composition that includes one or more compounds of Formula (I), Formula (II) and/or Formula (III), or a pharmaceutically acceptable sait of the foregoing.
Other embodiments described herein relate to using one or more compounds of Formula (I),
Formula (II) and/or Formula OII), or a pharmaceutically acceptable sait of the foregoing, in the manufacture of a médicament for améliora tin g and/or treating a paramyxovirus viral infection. Still other embodiments described herein relate to compounds of Formula (I), Formula 01) and/or Formula (III), or a pharmaceutically acceptable sait of the foregoing, that can be used for ameliorating and/or treating a paramyxovirus viral infection. Yet still other embodiments disclosed herein relate to methods of ameliorating and/or treating a paramyxovirus viral infection that can include contacting a cell infected with the paramyxovirus with an effective amount of one or more compounds of Formula 0), Formula 01) and/or Formula 0Π), or a pharmaceutically acceptable sait of the foregoing, or a pharmaceutical composition that includes one or more compounds of Formula (I), Formula (II) and/or Formula (ΙΠ), or a pharmaceutically acceptable sait of the foregoing. Some embodiments disclosed herein relate to methods of inhibiting the réplication of a paramyxovirus that can include contacting a cell infection with the paramyxovirus with an effective amount of one or more compounds of Formula 0), Formula (II) and/or Formula OU), or a pharmaceutically acceptable sait of the foregoing, or a pharmaceutical composition that includes one or more compounds of Formula 0), Formula 01) and/or Formula (III), or a pharmaceutically acceptable sait of the foregoing. For example, the paramyxovirus viral infection can be causcd by a henipavirus, a morbillivirus, a respirovirus, a rubulavirus, a pneumovirus (including a respiratory syncytial viral infection), a metapneumovirus, hendravirus, nipahvirus, measles, sendai virus, mumps, a human parainfluenza virus (HPIV-1, HPIV-2, HPIV-3 and HPIV-4) and/or a metapneumovirus.
[0006] Some embodiments disclosed herein relate to methods of ameliorating and/or treating an orthomyxovirus viral infection that can include administering to a subject suffering from the orthomyxovirus viral infection an effective amount of one or more compounds of Formula (I), Formula 01) and/or Formula OH), or a pharmaceutically acceptable sait of the foregoing, or a pharmaceutical composition that includes one or more compounds of Formula (I), Formula 01) and/or Formula (III), or a pharmaceutically acceptable sait of the foregoing. Other embodiments described herein relate to using one or more compounds of Formula (I), Formula 01) and/or Formula (III), or a pharmaceutically acceptable sait of the foregoing, in the manufacture of a médicament for ameliorating and/or treating an orthomyxovirus viral infection. Still other embodiments described herein relate to compounds of Formula (I), Formula 00 and/or Formula (III), or a pharmaceutically acceptable sait of the foregoing, that can be used for ameliorating and/or treating an orthomyxovirus viral infection. Yet still other embodiments disclosed herein relate to methods of ameliorating and/or treating an orthomyxovirus viral infection that can indude contacting a cell infected with the orthomyxovirus with an effective amount of one or more compounds of Formula (1), Formula (II) and/or Formula (III), or a pharmaceutically acceptable sait of the foregoing, or a pharmaceutical composition that includes one or more compounds of Formula (I), Formula (II) and/or Formula GH), or a pharmaceutically acceptable sait of the foregoing. Some embodiments disclosed herein relate to methods of inhibiting the réplication of an orthomyxovirus that can include contacting a cell infection with the orthomyxovirus with an effective amount of one or more compounds of Formula (I), Formula (II) and/or Formula (III), or a pharmaceutically acceptable sait of the foregoing, or a pharmaceutical composition that indudes one or more compounds of Formula (I), Formula (II) and/or Formula (III), or a pharmaceutically acceptable sait of the foregoing. For exampie, the orthomyxovirus viral infection can be an influenza viral infection (such as influenza A, B and/or C).
[0007] Some embodiments disclosed herein relate to methods of ametiorating and/or treating a paramyxovirus viral infection and/or an orthomyxovirus viral infection that can indude administering to a subject suffering from the viral infection an effective amount of a compound described herein or a pharmaceutically acceptable sait thereof (for exampie, one or more compounds of Formulae (I), (Π) and/or QU), or a pharmaceutically acceptable sait of the . foregoing), or a pharmaceutical composition that indudes one or more compounds described herein, in combination with one or more agents described herein. Some embodiments disclosed herein relate to methods of ameliorating and/or treating a paramyxovirus viral infection and/or an orthomyxovirus viral infection that can indude contacting a cell infected with the virus with an effective amount of a compound described herein or a pharmaceutically acceptable sait thereof (for example, one or more compounds of Formulae (J), 01) and/or (ΙΠ), or a pharmaceutically acceptable sait of the foregoing), or a pharmaceutical composition that indudes one or more compounds described herein, in combination with one or more agents described herein.
BRIEF DESCRIPTION OFTHE DRAWINGS [0008] Figure 1 shows example RSV agents.
DETAILED DESCRIPTION [0009] Paramyxoviridae family is a family of single stranded RNA viruses. Several généra of the paramyxoviridae family include henipavirus, morbitlivirus, respirovirus, rubulavirus, pneumovirus and metapneumovirus. These viruses can be transmitted person to person via direct or close contact with contaminated respiratory droplets or fomites. Species of henipavirus include hendravirus and nipahvirus. A species of morbillivirus is me as les. Species of respirovirus include sendai virus and human parainfluenza viruses 1 and 3; and species of rubulavirus include mumps virus and human parainfluenza viruses 2 and 4. A species of metapneumovirus is human metapneumovirus.
[0010] Human Respiratory Syncytial Virus (RSV), a species of pneumovirus, can cause respiratory infections, and can be associated with bronchiolitis and pneumonie. Symptoms of an RSV infection include coughing, sneezing, runny nose, fever, decrease in appetite, and wheezing. RSV is the most common cause of bronchiolitis and pneumonia in children under one year of âge in the worid, and can bc the cause of tracheobronchitis in older children and adults. In the United States, between 75,000 and 125,000 infants are hospitalized each year with RSV. Among adults older than 65 ycars of âge, an estimated 14,000 deaths and 177,000 hospîtalizations hâve been attributed to RSV.
[0011] Treatment options for people infected with RSV are currcntly limited. Antibiotics, usually prescribed to treat bacterial infections, and over-the-counter médication are not effective in treating RSV and may help only to relicve some of the symptoms. In severe cases, a nebulized bronchodilator, such as albuterol, may be prescribed to relieve some of the symptoms, such as wheezing. RespiGram® (RSV-IGIV, Medlmmune, approved for high risk children younger than 24 months of âge), Synagis® (palivizumab, Medlmmune, approved for hïgh risk children younger than 24 months of âge), and Virzole® (ribavirin by aérosol, ICN pharmaceuticals) hâve been approved for treatment of RSV.
[0012] Symptoms of the mcasles include fever, cough, runny nose, red eyes and a gencralized rash. Some individuals with measles can develop pneumonia, ear infections and bronchitis. Mumps Ieads to swelling of the salivary glands. Symptoms of mumps include fever, loss of appetite and fatigue. Individuals are often immunized against measles and mumps via a threc-part MMR vaccine (measles, mumps, and rubella). Human parainfluenza virus indudes four sérotypes types, and can cause upper and lower respiratory tract infections. Human parainfluenza virus 1 (HPIV-1) can be associated with croup; human parainfluenza virus 3 (HPIV-3) can be associated with bronchiolitis and pneumonia. According to the Ccnters of Disease Control and Prévention (CDC), there are no vaccines against human parainfluenza virus.
[0013] Influenza is a single strandcd RNA virus and a member of the Orthomyxoviridae family. There are currently three species of influenza; influenza A, influenza B and influenza C. Influenza A has been further classified based on the viral surface proteins
into hemagglutinin (H or HA) and neuramididase (N). There are approximately 16 H antigens (H1 to H16) and 9 N antigens (NI to N9). Influenza A includes several subtype, including H1N1, H1N2, H2N2, H3N1, H3N2, H3N8, H5N1, H5N2, H5N3, H5N8, H5N9, H7NI, H7N2, H7N3, H7N4, H7N7, H9N2, H ION 7. As with RSV, influenza viruses can be transmitted from person to person via direct contact with infected sécrétions and/or contaminated surfaces or objections. Complications from an influenza viral infection include pneumonia, bronchitis, déhydration, and sinus and ear infections. Médications currently approved by the FDA against an influenza infection include amantadine, rimantadine, Relenza® (zanamivir, GlaxoSmithKline) and Tamiflu® (oseltamivir, Genentech).
Définitions [0014] Unless defined otherwise, ail technical and scientifîc terms used herein hâve the same meaning as is commonly understood by one of ordinary skill in the art. Ail patents, applications, published applications and other publications referenced herein are încorporated by reference in their entirety unless stated otherwise. In the event that there are a plurality of définitions for a term herein, those in this section prevail unless stated otherwise.
[0015] As used herein, any R group(s) such as, without limitation, R1A, R2^ R3A, d4A „SA p6A p7A p8A p9A nlOA pllA Γ) I2A Dt3A pUA p ISA pt6A p 17A pt8A p ISA p20A ix y IX y IX f Ix y tx y Ix y Γχ. f IX f Γχ. | Γχ. f Γχ g Γχ. * IX y Γχ. y IX y IX g Γχ. y n2lA n22A n23A p24A n25A p2ÛA n27A p28A p29A n30A q31A q32A q33A q34A q35A q36A tx. y tx y tx ÿ tx * tx | tx y tx | tx | tx y tx | tx y tx y tx g tx f tx y tx g ρ37Α η38Α rjlB n2B ρ^Β p4B p3B p6B p7B ρ8θ P®® p 1®® P^® pl2B P^® P^® Pæ p2C tx. y tx y tx y tx | tx y tx | tx | tx y tx j tx | tx p tx | tx * tx y tx j tx y tx f tx y p3C p4C pîC p6C p7C p8C D9C plOC DI1C plîC p 13C pl4C plSC p16C pI7C DI8C pl9C p20C tx j tx | tx y tx a tx | tx y tx f tx | tx y tx. y tx. y tx y tx ( tx. | tx. y tx | tx y tx. f R21C, R220 and R23C represent substituents that can be attached to the indicated atom. An R group may be substituted or unsubstituted. If two R groups are described as being taken together the R groups and the atoms they are attached to can form a cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl or heterocycle. For example, without limitation, if R* and Rb of an NR* Rb group are indicated to be taken together, it means that they are covalently bonded to one another to form a ring:
,Ra
^Rb
In addition, if two “R” groups are described as being “taken together” with the atom(s) to which they are attached to form a ring as an alternative, the R groups are not limited to the variables or substituents defined previously.
[0016[ Whenever a group is described as being “optionally substituted” that group may be unsubstituted or substituted with one or more of the indicated substituents. Likewise, β
when a group is described as being “unsubstituted or substituted” if substituted, the substituent(s) may be selected from one or more the indicated substituents. If no substituents are indicated, it is meant that the indicated “optionally substituted” or “substituted” group may be substituted with one or more group(s) individually and independently selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxy, protected hydroxyl, alkoxy, aryloxy, acyl, mercapto, alkylthio, arylthio, cyano, halogen, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, protected C-carboxy, O-carboxy, isocyanate, thiocyanato, isothîocyanato, nitro, silyl, sulfenyl, sulfînyl, sulfonyl, haloalkyl, haloalkoxy, trihalomethanesulfonyl, trihalomethanesulfonamido, an amino, a mono-substituted amino group and a di-substituted amino group, and protected dérivatives thereof.
[0017] As used herein, “Câ to Ct>” in which “a” and “b” are integers refer to the number of carbon atoms in an alkyl, alkenyl or alkynyl group, or the number of carbon atoms in the ring of a cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl or heteroalicyclyl group. That is, the alkyl, alkenyl, alkynyl, ring of the cycloalkyl, ring of the cycloalkenyl, ring of the cycloalkynyl, ring of the aryl, ring of the heteroaryl or ring of the heteroalicyclyl can contain from “a” to “b”, inclusive, carbon atoms. Thus, for example, a “Ci to C< alkyl” group refera to ail alkyl groups having from 1 to 4 carbons, that is, CHj-, CH3CH2-, CH3CH2CH2-, (CFDjCH-, CH3CH2CH2CH2-, CH3CH2CH(CH3)- and (CHs^C-, If no “a” and “b” are designated with regard to an alkyl, alkenyl, alkynyl, cycloalkyl cycloalkenyl, cycloalkynyl, aryl, heteroaryl or heteroalicyclyl group, the broadest range described in these définitions is to be assumed.
[0018] As used herein, “alkyl” refers to a straight or branched hydrocarbon chain that comprises a fully saturated (no double or triple bonds) hydrocarbon group. The alkyl group may hâve l to 20 carbon atoms (whenever it appears herein, a numerical range such as “I to 20” refera to each integer in the given range; e.g., “1 to 20 carbon atoms” means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 20 carbon atoms, although the présent définition also covers the occurrence of the term “alkyl” where no numerical range is designated). The alkyl group may also be a medium size alkyl having 1 to 10 carbon atoms. The alkyl group could also be a lower alkyl having 1 to 6 carbon atoms. The alkyl group of the compounds may be designated as “C1-C4 alkyl” or similar désignations. By way of example only, “C1-C4 alkyl” indicates that there are one to four carbon atoms în the alkyl chain, i.e., the alkyl chain is selected from methyl, ethyl, propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, and t-butyl. Typical alkyl groups include, but are în no way limited
to, methyi, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl and hexyl. The alkyl group may be substituted or unsubstîtuted.
[0019] As used herein, “aikenyl” refers to an alkyl group that contains in the straight or branched hydrocarbon chain one or more double bonds. An aikenyl group may be unsubstîtuted or substituted.
[0020] As used herein, “alkynyl” refers to an alkyl group that contains in the straight or branched hydrocarbon chain one or more triple bonds. An alkynyl group may be unsubstîtuted or substituted.
[0021] As used herein, “cycloalkyl” refera to a completely saturated (no double or triple bonds) mono- or multi- cyclic hydrocarbon ring System. When composed of two or more rings, the rings may be joined together in a fused fashîon. Cycloalkyl groups can contain 3 to 10 atoms in the ring(s) or 3 to 8 atoms in the ring(s). A cycloalkyl group may be unsubstîtuted or substituted. Typical cycloalkyl groups include, but are in no way limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl.
[0022] As used herein, “cycloalkenyl” refera to a mono- or multi- cyclic hydrocarbon ring system that contains one or more double bonds in at least one ring; although, if there is more than one, the double bonds cannot form a fiilly delocalized pi-electron System throughout all the rings (otherwise the group would be “aryl,” as defined herein). When composed of two or more rings, the rings may be connected together in a fused fashion. A cycloalkenyl group may be unsubstîtuted or substituted.
[0023] As used herein, “cycloalkynyl” refera to a mono- or multi- cyclic hydrocarbon ring system that contains one or more triple bonds in at least one ring. If there is more than one triple bond, the triple bonds cannot form a fiilly delocalized pi-electron System throughout all the rings. When composed of two or more rings, the rings may be joined together in a fused fashion. A cycloalkynyl group may be unsubstîtuted or substituted.
[0024] As used herein, “aryl” refera to a carbocyclic (all carbon) monocyclic or multicyclic aromatic ring System (including fused ring Systems where two carbocyclic rings share a chemical bond) that has a fully delocalized pi-electron system throughout all the rings. The number of carbon atoms in an aryl group can vary. For example, the aryl group can be a C6-Ci4 aryl group, a Ce-Cio aryl group, or a Ce aryl group. Examples of aryl groups include, but are not limited to, benzene, naphthalene and azulene. An aryl group may be substituted or unsubstîtuted.
[0025] As used herein, “heteroaryl” refera to a monocyclic or multicyclic aromatic ring system (a ring system with fiilly delocalized pi-electron system) that contain(s) one or more hetcroatoms, that is, an element other than carbon, including but not limited to, nitrogen, oxygen and sulfur. The number of atoms in the ring(s) of a heteroaryl group can vary. For example, the heteroaryl group can contain 4 to 14 atoms in the ring(s), 5 to 10 atoms in the ring(s) or 5 to 6 atoms in the ring(s). Furthermore, the term “heteroaryr includes fused ring Systems where two rings, such as at least one aryl ring and at least one heteroaryl ring, or at least two heteroaryl rings, share at least one chemical bond. Examples of heteroaryl rings include, but arc not limited to, furan, furazan, thiophene, benzothiophene, phthalazine, pyrrole, oxazole, benzoxazole, 1,2,3-oxadiazole, 1,2,4-oxadiazole, thïazole, 1,23-thiadiazote, 1,2,4-thiadiazole, benzothiazole, imidazole, benzimidazole, indole, indazole, pyrazole, benzopyrazole, isoxazole, 10 benzoisoxazole, isothiazole, triazole, benzotriazole, thiadiazole, tetrazole, pyridine, pyridazine, pyrimidine, pyrazine, purine, pteridine, quînoline, isoquinoline, quinazoline, quînoxaline, cinnoline, and triazine. A heteroaryl group may be substituted or unsubstituted.
[0026] As used herein, “heterocyclyl” or “heteroalicyclyl” refera to three-, four-, fîve-, six-, seven-, eight-, nine-, ten-, up to 18-membered monocyclic, bicyclic, and tricyclic ring 15 system wherein carbon atoms together with from 1 to 5 hetcroatoms constitute said ring system.
A heterocycle may optionally contain one or more unsaturated bonds situated in such a way, however, that a fully delocalized pi-electron system does not occur throughout ail the rings. The heteroatom(s) is an element other than carbon including, but not limited to, oxygen, sulfur, and nitrogen. A heterocycle may further contain one or more carbonyl or thiocarbony! M functionalities, so as to make the définition include oxo-systems and thio-systems such as lactams, lactones, cyclic imides, cyclic thioimides and cyclic caibamates. When composed of two or more rings, the rings may be joincd together in a fused fashion. Additionally, any nitrogens in a heteroalicyclic may be quatemized. Heterocyclyl or heteroalicyclic groups may be unsubstituted or substituted. Examples of such “heterocyclyl” or “heteroalicyclyl” groups 25 include but are not limited to, 1,3-dioxin, 13-dioxane, 1,4-dioxane, 1,2-dioxotane, 1,3dioxotane, 1,4-dioxolanc, 1,3-oxathîane, 1,4-oxathiin, 1,3-oxathiolane, 1,3-dithiole, 1,3dithiolane, 1,4-oxathiane, tetrahydro-l,4-thiazine, 2H-l,2-oxazine, maleimide, succinimide, barbituric acid, thiobarbituric acid, dioxopiperazine, hydantoin, dihydrouracïl, trioxane, hexahydro-l,3,5-triazine, imidazoline, imidazolidine, isoxazoline, isoxazolidine, oxazoline, 30 oxazolidine, oxazolidinone, thiazoline, thiazolidine, morphotine, oxirane, piperidine A/’-Oxîde, piperidine, piperazine, pyrrolidine, pyrrolidone, pyrrotidione, 4-pîperidone, pyrazoline, pyrazolidine, 2-oxopyrrolidine, tetrahydropyran, 4H-pyran, tetrahydrothiopyran, thiamorpholine, thiamorpholine sulfoxide, thiamorpholine sulfone, and their benzo-fused analogs (e.g., benzîmidazolidinone, tetrahydroquinolinc, and 3,4-methylenedioxyphenyl).
[00271 As used herein, “aralkyl” and “aiyl(alkyl)” refer to an aryl group connected, as a substituent, via a lower alkylene group. The lower alkylene and aryl group of an aralkyl may be substituted or unsubstituted. Examples include but are not limited to benzyl, 2phenylalkyl, 3-phenylalkyl, and naphthylalkyl.
[0028] As used herein, “heteroaralkyl” and “heteroaryl(alkyl)” refer to a heteroaryl group connected, as a substituent, via a lower alkylene group. The lower alkylene and heteroaryl group of heteroaralkyl may be substituted or unsubstituted. Examples include but are not limited to 2-thîenylalkyl, 3-thienylalkyl, fiirylalkyl, thienylalkyl, pyrrolylalkyl, pyridylalkyl, isoxazolylalkyl, imidazolylalkyl, and their benzo-fused analogs. .
[0029] A “(heteroalicyclyl)alkyl” and “(heterocyclyl)alkyl” refer to a heterocyclic or a heteroalicyclylic group connected, as a substituent, via a lower alkylene group. The lower alkylene and heterocyclyi of a (heteroalicyclyl)alkyl may be substituted or unsubstituted. Examples include but are not limited tetrahydro-2H-pyran-4-yl)methyl, (piperidin-4-yl)ethyl, (piperidin-4-yl)propyl, (tetrahydro-2H-thiopyran-4-y!)methyl, and (l,3-thiazinan-4-yl)methyl.
[0030] “Lower alkylene groups” are straight-chained -CH2- tethering groups, forming bonds to connect molecular fragments via their terminal carbon atoms. Examples include but are not limited to methylene (-CH2-), ethylene (-CH2CH2-), propylene (CH2CH2CH2-), and butylène (-CH2CH2CH2CH2-). A lower alkylene group can be substituted by replacing one or more hydrogen of the lower alkylene group with a substituent(s) listed under the définition of “substituted.” [0031] As used herein, “alkoxy” refera to the formula -OR wherein R is an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, a cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, (heteroaryl)alkyl or (heteroalicyclyl)alkyl is defined herein. A nonlimiting list of alkoxys are methoxy, ethoxy, n-propoxy, 1-methylethoxy (isopropoxy), nbutoxy, îso-butoxy, sec-butoxy, tert-butoxy, phenoxy and benzoxy. An alkoxy may be Î5 substituted or unsubstituted.
[0032] As used herein, “acyl” refera to a hydrogen, alkyl, alkenyl, alkynyl, or aryl connected, as substituents, via a carbonyl group. Examples include formyl, acetyl, propanoyl, benzoyl, and acryl. An acyl may be substituted or unsubstituted.
[0033] As used herein, “hydroxyalkyl” refera to an alkyl group in which one or more of the hydrogen atoms are replaced by a hydroxy group. Exemplary hydroxyalkyl groups include but are not limited to. 2-hydroxyethyl, 3-hydroxypropyl, 2-hydroxypropyl, and 2,2dihydroxyethyl. A hydroxyalkyl may be substituted or unsubstituted.
[0034] As used herein, “haloalkyl” refera to an alkyl group in which one or more of the hydrogen atoms are replaced by a halogen (e.g., mono-haloalkyl, di-haloalkyl and trihaloalkyl). Such groups include but are not limited to, chloromethyl, fluoromethyl, difluoromethyl, trifluoromethyl, l-chloro-2-fluoromethyl and 2-fluoroisobutyl. A haloalkyl may 5 be substituted or unsubstituted.
[0035] As used herein, “haloalkoxy” refera to an alkoxy group in which one or more of the hydrogen atoms are replaced by a halogen (e.g., mono-haloalkoxy, di- haloalkoxy and trihaloalkoxy). Such groups include but are not limited to, chloromethoxy, fluoromethoxy, difluoromethoxy, trifluoromethoxy, l-chIoro-2-fluoromethoxy and 2-fluoroisobutoxy. A 1 o haloalkoxy may be substituted or unsubstituted.
[0036] As used herein, “arylthio” refera to RS-, in which R is an aryl, such as, but not limited to, phenyl. An arylthio may be substituted or unsubstituted.
[0037] A “sulfenyl group refera to an “-SR” group in which R can be hydrogen, alkyl, alkenyl, alkynyl, cycloalkyi, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, hcteroalicyclyl, 15 aralkyl, (heteroaryl)alkyl or (heteroalicyclyl)alkyl. A sulfenyl may be substituted or unsubstituted.
[0038] A “sulfinyl” group refera to an “-S(=O)-R” group in which R can be the same as defined with respect to sulfenyl. A sulfinyl may be substituted or unsubstituted.
[0039] A sulfonyl” group refera to an “SO2R group in which R can be the same as 20 defined with respect to sulfenyl. A sulfonyl may be substituted or unsubstituted.
[0040] An O-carboxy” group refera to a “RC(=O)O-” group in which R can be hydrogen, alkyl, alkenyl, alkynyl, cycloalkyi, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, (heteroaryl)alkyl or (heteroalicyclyl)alkyl, as defined herein. An Ocarboxy may be substituted or unsubstituted.
[0041] The terms “ester and “C-carboxy” refer to a “-C(=O)OR” group in which R can be the same as defined with respect to O-carboxy. An ester and C-carboxy may be substituted or unsubstituted.
[0042] A “thiocarbonyl” group refera to a “-C(=S)R” group in which R can be the same as defined with respect to O-carboxy. A thiocarbonyl may be substituted or unsubstituted.
[0043] A “trihalomethanesulfonyl” group refera to an X3CSO2-” group wherein each X is a halogen.
[0044] A “trihalomethanesulfonamido” group refera to an *‘X3CS(O)2N(Ra)-” group wherein each X is a halogen, and Ra hydrogen, alkyl, alkenyl, alkynyl, cycloalkyi, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, (heteroaryl)alkyl or (heteroaticyclyl)alkyl.
[0045] The term “amino” as used herein refera to a -NH2 group.
[0046] As used herein, the term “hydroxy” refera to a -OH group.
[0047] A “cyano” group refera to a “-CN” group.
[0048] The term “azido” as used herein refera to a -Nj group.
[0049] An “isocyanato” group refera to a “-NCO” group.
[0050] A “thiocyanato” group refera to a “-CNS group.
[0051] An “isothiocyanato” group refera to an “ -NCS” group.
[0052] A “mercapto” group refers to an “-SH group.
[0053] A “carbonyl group refera to a C=O group.
[0054] An “S-sulfonamido” group refera to a “-SO2N(RaRb)” group in which Ra and Rb can be independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, (heteroaryl)alkyl or (heteroalicyclyl)alkyl. An S-sulfonamido may be substituted or unsubstituted.
[0055] An “N-sulfonamido” group refera to a “RSO2N(Ra)-” group in which R and Ra can be independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, (heteroaryl)alkyl or (heteroalicyclyl)alkyl. An N-sulfonamido may be substituted or unsubstituted.
[0056] An “O-carbamyl” group refera to a “-OC(=O)N(RaRb) group in which Ra and Rb can be independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, (heteroaryl)alkyl or (heteroalicyclyl)alkyl. An O-carbamyl may be substituted or unsubstituted.
[0057] An “N-carbamyl” group refera to an “ROC(=O)N(Ra)-” group in which R and Ra can be independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, (heteroaryl)alkyl or (heteroalicyclyl)alkyl. An N-carbamyl may be suhstituted or unsubstituted.
[0058] An “O-thiocarbamyl” group refera to a *‘-OC(=S)-N(RaRb)” group in which Ra and Rb can be independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, (heteroaryl)alkyl or (heteroalicyclyl)alkyl. An O-thiocarbamyl may be substituted or unsubstituted.
[0059] An “N-thiocarbamyl” group refera to an “ROC(=S)N(Ra)- group in which R and Ra can be independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyt, aryl, heteroaryl, heteroalicyclyl, aralkyl, (heteroarylalkyl or (heteroaticyclyl)alkyt. An N-thiocarbamyt may be substituted or unsubstituted.
[0060] A “C-amido” group refera to a **-C(=O)N(RaRb)” group in which Ra and Rb can be independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cydoalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, (heteroaryl)alkyl or (heteroalicyclyl)alkyl. A C-amido may be substituted or unsubstituted.
[0061] An “N-amido” group refera to a ‘*RC(=O)N(Ra)-” group in which R and Ra can be independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cydoalkenyl, cycloalkynyt, aryl, heteroaryl, heteroalicyclyl, aralkyl, (heteroarylalkyl or (heteroalicydyl)alkyl. An 1 ° N-amido may be substituted or unsubstituted.
[0062] The term “halogen atom” or “halogen” as used herein, means any one of the radio-stable atoms of column 7 of the Periodic Table of the Eléments, such as, fluorine, chlorine, bromine and iodine.
[0063] Where the numbera of substituents is not specifîed (e.g. haloalkyl), there may 15 be one or more substituents présent. For example haloalkyl” may include one or more of the same or different halogens. As another exampie, “C1-C3 alkoxyphenyl” may include one or more of the same or different alkoxy groups containing one, two or three atoms.
[0064] As used herein, the abbreviations for any protective groups, amino acids and other compounds, are, unless indicated otherwise, in accord with their common usage, 20 recognized abbreviations, or the IUPAC-RJB Commission on Biochemical Nomenclature (See,
Biochem. 11:942-944 (1972)).
[0065] The term “nucleoside” is used herein in its ordinary sense as understood by those skilled in the art, and refera to a compound composed of an optionally substituted pentose moiety or modified pentose moiety attached to a heterocyciic base or tautomer thereof via a N25 gtycosidic bond, such as attached via the 9-posîtion of a purine-base or the 1-position of a pyrimidine-base. Examples include, but are not limited to, a ribonucleoside comprising a ribose moîety and a deoxyribonucteoside comprising a deoxyribose moiety. A modified pentose moiety is a pentose moiety in which an oxygen atom has been replaced with a carbon and/or a carbon has been replaced with a sulfur or an oxygen atom. A “nucleoside” is a monomer that
3Q can hâve a substituted base and/or sugar moiety. Additionally, a nucleoside can be incorporated into larger DNA and/or RNA polymère and otigomers. In some instances, the nucleoside can be a nucleoside analog drug.
[0066] The term nucléotide” is used herein in its ordinary sense as understood by those skilled in the art, and refers to a nucleoside having a phosphate ester bound to the pentose moiety, for example, at the 5*-position.
[0067] As used herein, the term “heterocyclic base” refera to an optionally substituted nitro gen-containing heterocyclyl that can be attached to an optionally substituted pentose moiety or modified pentose moiety. In some embodiments, the heterocyclic base can be selected from an optionally substituted purine-base, an optionally substituted pyrimidine-base and an optionally substituted triazole-base (for example, a 1,2,4-triazole). The term purinebase” is used herein in its ordinary sense as understood by those skilled in the art, and includes its tautomera. Sîmîlarly, the term pyrimidine-base” is used herein in its ordinary sense as understood by those skilled in the art, and includes its tautomera. A non-lîmiting list of optionally substituted purine-bases includes purine, adenine, guanine, hypoxanthine, xanthine, alloxanthine, 7-alkylguanine (e.g. 7-methylguanine), theobromine, caffeine, une acid and isoguanine. Examples of pyrimidine-bases include, but are not limited to, cytosine, thymine, uracil, 5,6-dihydrouracit and 5-alkylcytosine (e.g., 5-methylcytosine). An example of an optionally substituted triazole-base is l,2,4-triazole-3-carboxamide. Other non-iimiting examptes of heterocyclic bases include diamînopurine, e-oxo-N^-alkyladenine (e.g., β-οχο-Ν^methyladenine), 7-deazaxanthîne, 7-deazaguanine, 7-deazaadenine, N4,N4-cthanocytosin, Ν^,Ν6ethano-2,6-diaminopurine, 5-haIouracil (e.g., 5-fluorouracil and 5-bromouracil), pseudoisocytosine, isocytosine, isoguanine, and other heterocyclic bases described in U.S. Patent Nos. 5,432,272 and 7,125,855, which are incorporated herein by reference for the limited purpose of disclosing additional heterocyclic bases. In some embodiments, a heterocyclic base can be optionally substituted with an amine or an enol protecting group(s).
[0068] The term -N-linked amino acid” refera to an amino acid that is attached to the indicated moiety via a main-chain amino or mono-substituted amino group. When the amino acid is attached in an -N-linked amino acid, one of the hydrogens that is part of the main-chain amino or mono-substituted amino group is not présent and the amino acid is attached via the nitrogen. N-linked amino acids can be substituted or unsubstituted.
[0069] The term “-N-linked amino acid ester dérivative” refers to an amino acid in which a main-chain carboxylic acid group has been converted to an ester group. In some embodiments, the ester group has a formula selected from alkyI-O-C(=O)-, cycloalkyl-O-C(=O), aryt-O-C(=O)- and aryl(alkyl)-O-C(=O)-. A non-lîmiting list of ester groups include substituted and unsubstituted versions of the foliowing: methyl-O-C(=O)-, ethyl-O-C(=O)-, npropy 1-O-C(=O)-, isopropyl-O-C(=O)-, n-butyl-O-C(=O)-, isobutyI-O-C(=O)-, tert-butyl-O14
C(=O)-, neopentyl-O-C(=O)-, cyclopropyI-O-C(=O)-, cyclobutyl-O-C(=O)-, cyclopentyl-OC(=O)-, cyclohexyl-O-C(=O)-, phenyl-O-C(=O)-, benzyl-O-C(=O)-, and naphthyl-O-C(=O)-. N-linked amino acid ester dérivatives can be substituted or unsubstituted.
[0070] The term “-O-tinked amino acid” refera to an amino acid that is attached to the indicated moiety via the hydroxy from its main-chain carboxylic acid group. When the amino acid is attached in an -O-linked amino acid, the hydrogen that is part of the hydroxy from its main-chain carboxylic acid group is not présent and the amino acid is attached via the oxygen. O-linked amino acids can be substituted or unsubstituted.
[0071 [ As used herein, the term “amino acid” refera to any amino acid (both standard and non-standard amino acids), including, but not limited to, α-amino acids, β-amino acids, γamino acids and δ-amino acids. Examples of suitable amino acids include, but are not limited to, alanine, asparagine, aspartate, cysteine, glutamate, glutamine, glycine, proline, serine, tyrosine, arginine, histidine, isoleucine, leucine, lysine, méthionine, phenytalanine, threonine, tryptophan and valine. Additional examples of suitable amino acids include, but are not limited to, omithine, hypusine, 2-aminoisobutyric acid, dehydroalanine, gamma-aminobutyric acid, citrulline, beta-alanine, alpha-ethyl-glycine, alpha-propyl-glycine and norleucine.
[0072] The terms “phosphorothioate” and “phosphothioate” refer to a compound of
the general formula 0'
OH
0' ’ its protonated forms (for example, 0' and
OH ) and its tautomers (such as ÔH ).
[0073] As used herein, the term “phosphate” is used in its ordinary sense as underatood by those skilled in the art, and includes its protonated forms (for example,
OH
OH
0' and OH ). As used herein, the terms “monophosphate,” “diphosphate,” and “triphosphate” are used in their ordinary sense as understood by those skilled in the art, and include protonated forms.
[0074] The terms “protecting group” and “protecting groups” as used herein refer to any atom or group of atoms that is added to a molécule in order to prevent existîng groups in the molécule from undergoing unwanted chemical reactions. Examples of protecting group moieties are described in T. W. Greene and P. G. M. Wuts, Protective Groups in Organic Synthesis, 3.
Ed. John Wiley & Sons, 1999, and in J.F,W. McOmie, Protective Groups in Organic Chemistry
Plénum Press, 1973, both of which are hereby incorporated by reference for the limited puipose of disclosing suitable protecting groups. The protecting group moiety may be chosen in such a way, that they are stable to certain reaction conditions and readily removed at a convenient stage using methodology known from the art. A non-limiting list of protecting groups inciude benzyl;
substituted benzyl; alkylcarbonyls and alkoxycarbonyls (e.g., t-butoxycarbonyl (BOC), acetyl, or isobutyiyl); arylalkylcarbonyls and arylalkoxycaibonyls (e.g., benzyloxycarbonyl); substituted methyl ether (e.g. mcthoxymcthyl ether); substituted ethyl ether, a substituted benzyl ether, tctrahydropyranyl ether, silyls (e.g., trimcthylsilyl, tricthylsilyl, triisopropylsilyl, t10 butyldimcthylsilyl, tri-iso-propylsilyloxymethyl, [2-(trimethylsilyl)ethoxy]mcthyl or tbutyldiphenylsilyl); esters (e.g. benzoate ester); carbonates (e.g. methoxymethylcarbonate); sulfonates (e.g. tosylate or mesylate); acyclic ketal (e.g. dimethyl acetal); cyclic ketals (e.g., 1,3dioxane, 1,3-dioxolanes, and those described herein); acyclic acetal; cyclic acetal (e.g., those described herein); acyclic hemiacetal; cyclic hemiacetal; cyclic dithioketals (e.g., 1,3-dithiane or 15 1,3-dithiolane); orthoesters (e.g., those described herein) and triarylmethyl groups (e.g., trityl;
monomethoxytrityl (MMTr); 4,4'-dimethoxytrityl (DMTr); 4,4',4'*-trimethoxytrityl (TMTr); and those described herein).
[0075] The term “pharmaceutically acceptable sait” refers to a sait of a compound that does not cause significant irritation to an organism to which it is administered and does not 20 abrogate the biological activity and properties of the compound. In some embodiments, the sait is an acid addition sait of the compound. Pharmaceutical salts can be obtained by reacting a compound with inorganic acids such as hydrohalic acid (e.g., hydrochloric acid or hydrobromic acid), sulfurie acid, nitric acid and phosphoric acid. Pharmaceutical salts can also be obtained by reacting a compound with an organic acid such as aliphatic or aromatic carboxylic or sulfonic 25 acids, for example formic, acetic, succinic, lactic, malic, tartane, citric, ascorbic, nicotinic, methanesulfonic, ethanesulfonic, p-tolucnsulfonic, salicylic or naphthalenesulfonic acid. Pharmaceutical salts can also be obtained by reacting a compound with a base to form a sait such as an ammonium salL an alkali métal sait, such as a sodium or a potassium sait, an alkalîne earth métal sait, such as a calcium or a magnésium sait, a sait of organic bases such as 30 dicyclohexylamine, N-methyl-D-glucamine, tris(hydroxymethyl)methylamine, C1-C7 alkylamine, cyclohexylamine, triethanolamîne, ethylenediamine, and salts with amino acids such as arginine and lysine.
[0076] Ternis and phrases used in this application, and variations thereof, especially in the appended daims, unless otherwise expressly stated, should be construed as open ended as opposed to limiting. As examples of the foregoing, the term ‘including’ should be read to mean ‘including, without limitation,’ ‘including but not limited to,’ or the like; the term ‘comprising’ as used herein is synonymous with ‘including,* ‘containing,’ or ‘characterized by,’ and is inclusive or open-ended and does not exclude additional, unrecited éléments or method steps; the tenu ‘having* should be interpreted as ‘having at least;* the term ‘includes* should be interpreted as ‘includes but is not limited to;’ the term ‘exampie’ is used to provide exemplary instances of the item in discussion, not an exhaustive or limiting list thereof; and use of terms like ‘preferably,’ ‘preferred,* ‘desired,’ or ‘désirable,’ and words of similar meaning should not be understood as implying that certain features are critical, essential, or even important to the structure or function of the invention, but instead as merely intended to highlight alternative or additional features that may or may not be utilized in a particular embodiment. In addition, the term comprising” is to be interpreted synonymously with the phrases having at least or including at least. When used in the context of a process, the term comprising means that the process includes at least the recited steps, but may include additional steps. When used in the context of a compound, composition or device, the term comprising means that the compound, composition or device includes at least the recited features or components, but may also include additional features or components. Likewise, a group of items linked with the conjunction ‘and’ should not be read as requiring that each and every one of those items be présent in the grouping, but rather should be read as ‘and/or* unless expressly stated otherwise. Similarly, a group of items linked with the conjunction ‘or’ should not be read as requiring mutual exclusivity among that group, but rather should be read as ‘and/or’ unless expressly stated otherwise.
[0077) With respect to the use of substantiaily any plural and/or singular terms herein, those having skill in the art can translate from the plural to the singular and/or from the singular to the plural as is appropriate to the context and/or application. The various singular/plural permutations may be expressly set forth herein for sake of clarity. The indefînite article “a” or “an” does not exclude a plurality. A single processor or other unit may fulfill the fonctions of several items recited in the claims. The mere fact that certain measures are recited in mutually different dépendent claims does not indicate that a combination of these measures cannot be used to advantage. Any reference signe in the claims should not be construed as limiting the scope.
[0078] It is understood that, in any compound described herein having one or more chiral centers, if an absolute stereochemîstry is not expressly indicated, then each center may independently be of R-configuration or S-configuration or a mixture thereof. Thus, the compounds provided herein may be enantiomerically pure, enantiomerically enriched, racemic mixture, diastereomerically pure, diastereomerically enriched, or a stereoisomeric mixture. In addition it is understood that, in any compound described herein having one or more double bond(s) generating geometrical isomers that can be defined as E or Z, each double bond may independently be E or Z a mixture thereof.
[0079] Likewise, it is understood that, in any compound described, all tautomeric forms are also intended to be included. For example all tautomers of a phosphate and a phosphorothioate groups are intended to be included. Examples of tautomers of a phosphorothioate include the following:
Furthermore, all tautomers of heterocyclic bases known in the art are intended to be included, including tautomers of naturel and non-natural purine-bases and pyrimidine bases.
[0080] It is to be understood that where compounds disclosed herein hâve unfilted valencies, then the valencies are to be fitled with hydrogens or isotopes thereof, e.g., hydrogen-1 (protium) and hydrogen-2 (deuterium).
[0081] It is understood that the compounds described herein can be labeied isotopically. Substitution with isotopes such as deuterium may afford certain therapeutic advantages resulting from greater metabolic stability, such as, for example, increased in vivo half-life or reduced dosage requirements. Each chemical element as represented in a compound structure may inctude any isotope of said element For example, in a compound structure a hydrogen atom may be explicitly disclosed or understood to be présent in the compound. At any position ofthe compound that a hydrogen atom may be présent, the hydrogen atom can be any isotope of hydrogen, including but not limited to hydrogen-1 (protium) and hydrogen-2 (deuterium). Thus, reference herein to a compound encompasses all potential isotopic forms unless the context ctearly dictâtes otherwise.
[0082] It is understood that the methods and combinations described herein include crystalline forms (also known as polymorphs, which include the different crystal packing arrangements of the same elemental composition of a compound), amorphous phases, salts, solvatés, and hydrates. In some embodiments, the compounds described herein exist in solvated forms with pharmaceutically acceptable solvents such as water, éthanol, or the like. In other embodiments, the compounds described herein exist in unsolvated form. Solvatés contain either stoichiometric or non-stoichiometric amounts of a solvenL and may be formed during the process of crystallization with pharmaceutically acceptable solvents such as water, éthanol, or the like. Hydrates are formed when the solvent is water, or alcoholates are formed when the solvent is alcohol. In addition, the compounds provided herein can exist in unsolvated as well as solvated forms. In general, the solvated forms are considered équivalent to the unsolvated forms for the purposes ofthe compounds and methods provided herein.
[0083] Where a range of values is provided, it is understood that the upper and lower
Ιίπΰζ and each intervening value between the upper and lower limit of the range is encompassed 1 θ within the embodiments.
Compounds [0084] Some embodiments disclosed herein relate to a compound selected from Formula (I), Formula (II) and Formula (III), or a pharmaceutically acceptable sait of the foregoing:
wherein: B1A, B1b and B1C can be independently an optionally substituted heterocyclic base or an optionally substituted heterocyclic base with a protected amino group; R1A can be selected from hydrogen, an optionally substituted acyl, an optionally substituted O-linked amino acid,
a single bond, Rm can be CH2, and R3A can be O (oxygen); when the dashed line (----) of
Formula (I) is absent, can be selected from an optionally substituted C]_6 alkyl, an optionally
Τ3Λ r10aJ_^
R11a ; when the dashed line (----) of Formula (I) is
substituted C2-6 alkenyl, an optionally substituted C2.6 alkynyl, an optionally substituted C» cycloalkyl, an optionally substituted -O-Ci-β alkyl, an optionally substituted O-C3-6 alkenyl, an optionally substituted O-Cj^ alkynyl and cyano, and R3A can be selected from OH, -OC(=O)R A and an optionally substituted O-linked amino acid; RIB can be selected from O\ OH,
optionally substituted N-linked amino acid and an optionally substituted N-linked amino acid ester dérivative; R,c and R2C can be independently selected from O, OH, an optionally
, an optionally substituted N-linked amino acid and an optionally substituted N-linked amino acid ester dérivative; or R,c can be
R2C can be 0' or OH; R20 and R3C can be independently selected from an optionally substituted
C145 alkyl, an optionally substituted C245 alkenyl, an optionally substituted C2-6 alkynyl, an optionally substituted -O-Cw alkyl, an optionally substituted -O-C3-6 alkenyl, an optionally substituted-O-C^ alkynyl, an optionally substituted C3.6 cycloalkyl and cyano; R4C can be selected from OH, •OC(i:O)Rc and an optionally substituted O-linked amino acid; R4A, R3B and R3C can be independently a halogen; RSA, R4B and R60 can be independently hydrogen or halogen; R6A, R7A and R^ can bc independently selected from absent, hydrogen, an optionally substituted Cr24 alkyl, an optionally substituted C2-24 alkenyl, an optionally substituted C2.24 alkynyl, an optionally substituted C3.6 cycloalkyl, an optionally substituted C345 cycloalkenyl, an optionally substituted aryl, an optionally substituted heteroaryl, an optionally substituted aryl(Ci-6 alkyl), an optionally substituted *-(CRlsAR16A)p-O-Ci.24 alkyl, an optionally
substituted *-<CR17AR,8A)q-O-Ci.24 alkenyl,
28A and
R29A ; or ReA can r12Aq_p--OR13A m and R7A can be absent or hydrogen; or R6A and R7A can be
aken together to form a moiety selected from an optionally substituted and and an i jptîonally
phosphorus and the moiety form a six-membered to ten-membered ring system; R9A can be ndependently selected from an optionally substituted C1.24 alkyl, an optionally substituted C2-24 ilkenyl, an optionally substituted C2-24 alkynyl, an optionally substituted C34 cycloalkyl, an jptionally substituted CM cycloalkenyl, NR3OAR31A, an optionally substituted N-linked amino icid and an optionally substituted N-linked amino acid ester dérivative; R10A and RllA can be independently an optionally substituted N-linked amino acid or an optionally substituted Ninked amino acid ester dérivative; R12A, R13A and R14A can be independently absent or îydrogen; each R1ÎA, each R16A, each R17A and each R18A can be independently hydrogen, an jptionally substituted C,^ alkyl or alkoxy, R19A, R20A, R2^, R23A, R3B, R6B, R8B, R9B, R90, R10C, R?2C and R13C can be independently selected from hydrogen, an optionally substituted C1.24 alkyl and an optionally substituted aryl; R21A, R24A, R™, RIOB, R1IC and R14c can be independently selected from hydrogen, an optionally substituted C1.24 alkyl, an optionally substituted aryl, an optionally substituted -O-C].24 alkyl and an optionally substituted -O-aryl; R23A, R29A, RllB and R15C can be independently selected from hydrogen, an optionally substituted Ci.24 alkyl and an optionally substituted aryl; R16C, Rl7C and Rl8C can be independently absent or hydrogen; R26A and R27A can be independently -ON or an optionally substituted substituent selected from C2-8 organylcarbonyl, C2-8 alkoxycarbonyl and C2-8organylaminocarbonyl; R28A can be selected from hydrogen, an optionally substituted Ci.24-alkyl, an optionally substituted C2.24 alkenyl, an optionally substituted C2.24 alkynyl, an optionally substituted C34 cycloalkyl and an optionally substituted Cj-β cycloalkenyl; R30A and R3IA can be independently selected from hydrogen, an optionally substituted Ci-24-alkyl, an optionally substituted C2-24 alkenyl, an optionally substituted C2-24 alkynyl, an optionally substituted C345 cycloalkyl and an optionally substituted -3-6 cycloalkenyl; for Formula (III),-----can be a single bond or a double bond; when-----is single bond, each R70 and each R8C can be independently hydrogen or halogen; and when--- is a double bond, each R7C is absent and each R8C can be independently hydrogen or halogen; < A and R° can be independently an optionally substituted Ci-24-alkyl, m and n can be ndependently 0 or 1; p and q can be independently selected from 1,2 and 3; r can be 1 or 2; ζ, Z1*, Z3A, Z4*, ZIB, Z2B and Z1C can be independently O or S; and provided that when the 72A
R^O— lashed line (----) of Formula (I) is absent; R1A is R9A wherein R8A is an insubstituted Cm alkyl or phenyl optionally para-substituted with a halogen or methyl and R9A s methyl ester, ethyl ester, isopropyl ester, n-butyl ester, benzyl ester or phenyl ester of an imino acid selected from glycine, alanine, valine, leucine, phenylalanine, tryptophan, nethionine and proline; R3A is OH; R4A is fluoro; RÎA is fluoro or hydrogen; and B1A is an insubstituted uracil; then R2* cannot be -OCH3; provided that when the dashed line (----) of formula (I) is absent; R1A is H; R3A is OH; R4A is fluoro; R3A is fluoro; and BIA is an 1 insubstituted cytosine; then R2A cannot be allenyl; provided that when the dashed line (----) of formula (I) is absent; RIA is H; R3A is OH; R4A is fluoro; RîA is hydrogen; and BÎA is an 1 insubstituted thymine; then R2* cannot be Ci alkyl substituted with an optionally substituted Namido (for example, -NC(=O)CF3); and provided that when the dashed line (----) of Formula
11) is absent; R1A is H; R3A is OH; R4A is fluoro; RÎA is fluoro; and B1A is an unsubstituted 1 :ytosine; then R2* cannot be ethynyl.
[0085] In some embodiments, the compound can be a compound of Formula (I), or a pharmaceutically acceptable sait thereof, wherein: B1A can be an optionally substituted lieterocyclic base or an optionally substituted heterocyclic base with a protected amino group;
-»1A ,2A T-3A
Z' r6AO— R8^— R1oa—P—
I <lA can be selected from hydrogen, OR7A, R9A and R11 A ; when the dashed line (----) of Formula (I) is a single bond, R2* is CH2, and R3A is O (oxygen); when the dashed line ( ) of Formula (I) is absent, RM can be selected from an optionally substituted Cm alkyl, an optionally substituted C2-6 alkenyl, an optionally substituted C2-6 alkynyl, an optionally substituted -Ο-Cm alkyl, an optionally substituted -O-C345 alkenyl, an optionally
Îubstituted -O-Ca-β alkynyl and cyano, and R3A is OH; R4A can be a halogen; RÎA can be îydrogen or halogen; R6A, R7A and R8A can be independently selected from absent, hydrogen, an jptionally substituted C1.24 alkyl, an optionally substituted C2.24 alkenyl, an optionally substituted C2-24 alkynyl, an optionally substituted C345 cycloalkyl, an optionally substituted C3-6 :ycloalkenyl, an optionally substituted aryl, an optionally substituted heteroaryl, an optionally substituted aryt(Ci^ alkyl), an optionally substituted *-(CRlîAR16A)p-O-Ci.24 alkyl, an
^12Aq—P
can be absent or hydrogen; or R6A and R7A can be *
taken together to form a moiety selected from an optionally substituted
and an
ôptionally substituted , wherein the oxygens connected to R6A and R7A, the jjhosphonis and the moiety form a six-membered to ten-membered ring system;. R9A can be independently selected from an optionally substituted C1-24 alkyl, an optionally substituted C2.24 independently an optionally substituted N-linked amino acid or an optionally substituted Nlinked amino acid ester dérivative; R12A, R13A and R14A can be independently absent or alkenyl, an optionally substituted C2-24 alkynyl, an optionally substituted C345 cycloalkyl, an optionally substituted cydoalkenyl, NR30AR3IA, an optionally substituted N-linked amino acid and an optionally substituted N-linked amino acid ester dérivative; R10A and R1IA can be
lydrogen; each Rl3A, each R16A, each Rl7A and each R18A can be independently hydrogen, an jptionally substituted C1.24 alkyl or alkoxy, R19A, R20A, R22A and R23* can be independently selected from hydrogen, an optionally substituted C1.24 alkyl and an optionally substituted aryl; <21A and R24A can be independently selected from hydrogen, an optionally substituted C1.24 îlkyl, an optionally substituted aryl, an optionally substituted -O-C1-24 alkyl and an optionally substituted -O-aryl; R2îA and R29A can be independently selected from hydrogen, an optionally substituted C1.24 alkyl and an optionally substituted aryl; R26A and R27A can be independently 10
3=N or an optionally substituted substituent selected from C2.8 organylcarbonyl, C2-8 ilkoxycarbonyl and C2.8 organylaminocarbonyt; R28A can be selected from hydrogen, an optionally substituted Ci.24*alkyl, an optionally substituted C2-24 alkenyl, an optionally substituted C2.24 alkynyl, an optionally substituted cycloalkyi and an optionally substituted -3^ cycloalkenyl; R30A and R31A can be independently selected from hydrogen, an optionally substituted Ci.24-alkyl, an optionally substituted C2-24 alkenyl, an optionally substituted C2-24 ilkynyl, an optionally substituted C3-6 cycloalkyi and an optionally substituted ¢3.6 cycloalkenyl; n can be 0 or t ; p and q can be independently selected from 1,2 and 3; r can be 1 or 2; Z1A, Z2*, Z3A and Z4A can be independently O or S. In some embodiments, a compound of Formula (I) :an hâve a structure shown herein, provided that when the dashed line (----) of Formula (I) is
ibsent; R1A is R9A wherein R8A is an unsubstituted Cm alkyl or phenyl optionally )ara-substituted with a halogen or methyl and R9A is methyl ester, ethyl ester, isopropyl ester, njutyl ester, benzyt ester or phenyl ester of an amino acid selected from glycine, alanine, valine, eucine, phenylalanine, tryptophan, méthionine and proline; R3A is OH; Ru is fluoro; R5A is luoro or hydrogen; and B1A is an unsubstituted uracil; then Ru cannot be -OCH3; provided that vhen the dashed line (----) of Formula (I) is absent; R1A is H; R3A is OH; Ru is fluoro; R3A is luoro; and B is an unsubstituted cytosine; then R cannot be allenyl; provided that when the lashed line (----) of Formula (I) is absent; R1A is H; R3A is OH; R4A is fluoro; R3A is hydrogen;
ind B1A is an unsubstituted thymine; then cannot be Ci alkyl substituted with an N-amido; ind provided that when the dashed line (----) of Formula (I) is absent; R1A is H; R3A is OH;
<4A is fluoro; R3A is fluoro; and BlA is an unsubstituted cytosine; then cannot be ethynyl.
r6ao— [0086] ln some embodiments, R1A can be OR7A. In some embodiments, <6A and R7A can be both hydrogen. In other embodiments, R6A and R7A can be both absent. In
! till other embodiments, at least one R6A and R7A can be absent. In yet still other embodiments, i it least one R6A and R7A can be hydrogen. Those skilled in the art understand that when R6A i ind/or R7Aare absent, the associated oxygen(s) will hâve a négative charge. For example, when R6A is absent, the oxygen associated with R6A will hâve a négative charge. In some embodiments, Z1A can be O (oxygen). In other embodiments, Z1A can be S (sulfur). In some mbodiments, R,A can be a monophosphate. In other embodiments, R1A can be a nonothiop hosphate.
>1A
[0087] In some embodiments, when R1A is OR7A, one of R6A and R7A can be hydrogen, and the other of R6A and R7A ts selected from an optionally substituted C].34 alkyl, t n optionally substituted C2.24 alkenyl, an optionally substituted C2.24 alkynyl, an optionally i ubstituted C3^ cycloalkyl, an optionally substituted C« cycloalkenyl, an optionally substituted rryl, an optionally substituted heteroaryl and an optionally substituted aryl(Ci_6 alkyl). In some embodiments, one of R6A and R7A can be hydrogen, and the other of RfiA and R7A can be an optionally substituted Ci,24 alkyl. In other embodiments, both R6A and R7A can be independently selected from an optionally substituted Ct-24 alkyl, an optionally substituted C2-24 alkenyl, an tiptionally substituted C2-24 alkynyl, an optionally substituted C3_6 cycloalkyl, an optionally s ubstituted C3-e cycloalkenyl, an optionally substituted aryl, an optionally substituted heteroaryl md an optionally substituted aryl(Ci^ alkyl). In some embodiments, both R6A and R7A can be an optionally substituted C1.24 alkyl. In other embodiments, both R6A and R7A can be an < iptionally substituted C2_24 alkenyl. In some embodiments, R6A and R7A can be independently an optionally substituted version of the following: myristolcyl, myristyl, palmitolcyl, palmityl, sapienyl, olcyl, elaidyl, vaccenyl, linoleyl, α-linotenyl, arachidonyl, eicosapentaenyl, erucyl, t ocosahexaenyl, caprylyl, capryl, lauiyl, stearyl, arachidyl, behenyl, lignoceryl, and cerotyl.
[0088] In some embodiments, at least one of R6A and R7A can be *-(CR15AR16A)p-OÎi_24 alkyl. In other embodiments, R6A and R7A can be both *-{CRlîAR16A)p-O-Ci.24 alkyl. In >me embodiments, each Rt3A and each R16Aare hydrogen. In other embodiments, at least one if R1îa and RlfiA is an optionally substituted C1.24 alkyl. In other embodiments, at least one of .1ÎA and R16A is an alkoxy (for example, benzoxy). In some embodiments, p can be 1. In other ^mbodiments, p can be 2. In still other embodiments, p can be 3.
Σ2-24 alkenyl. In other embodiments, R6A and R7A can be both *-{CR,7AR,8A)q-O-C2-24 alkenyl. l some embodiments, each Rl7A and each R18A are hydrogen. In other embodiments, at least
one of Rl7A and RI8A is an optionally substituted C1.24 alkyl. In some embodiments, q can be 1.
n other embodiments, q can be 2. In still other embodiments, q can be 3. When at least one of lt6A and R7A is *4CR,SAR16A)p-O-Ci-24 alkyl or *4CR17AR18A)q-O-C2-24 alkenyl, the Ci-24 alkyl can be selected from caprylyl, capryl, lauryl, myristyl, palmityl, steaiyl, arachidyl, behenyl,
Iignoceryl, and cerotyl, and the C2-24 alkenyl can be selected from myristoleyl, palmitoleyl, ! apîenyl, oleyl, elaidyl, vaccenyl, linoleyl, α-linolenyl, arachidonyl, eicosapentaenyl, erucyl and docosahexaenyl.
I0090J
In some embodiments, when R
selected
P
OR7A, at least one of R6A and
; and the other of RSA and R7A can be selected from absent, îydrogen, an optionally substituted C1.24 alkyl, an optionally substituted C2-24 alkenyl, an jptionally substituted C2-24 alkynyl, an optionally substituted C3^ cycloalkyl, an optionally lubstituted C3-6 cycloalkenyl, an optionally substituted aryl, an optionally substituted heteroaryl ind an optionally substituted aryl(Ci-e alkyl).
. In some embodiments, both R
20A can be independently [0091] In some embodiments, at least one of R6A and R7A can be
Vhen one or both of R6A and R7A selected from hydrogen, an optionally substituted Ci-24 alkyl and an optionally substituted aryl; und R21a can be selected from hydrogen, an optionally substituted C1.24 alkyl, an optionally substituted aryl, an optionally substituted -O-Ci-24 alkyl and an optionally substituted -O-aryl.
[n some embodiments, R19A and R20A can be hydrogen. In other embodiments, at least one of
V9A and R20A can be an optionally substituted C1.24 alkyl or an optionally substituted aryl. In some embodiments, R21A can be an optionally substituted C1.24 alkyl. In other embodiments, <21A can be an optionally substituted aryl. In still other embodiments, R21Acan be an optionally substituted -O-C1.24 alkyl or an optionally substituted -O-aryl.
[0092] In some embodiments, both R6A and R7A can be
R^*R2“ fl
When one or both of R6A and R7A are , R22A and R23* can be
ndependently selected from hydrogen, an optionally substituted C1.24 alkyl and an optionally substituted aryl; R24A can be independently selected from hydrogen, an optionally substituted Ci. : >4 alkyl, an optionally substituted aryl, an optionally substituted -O-C1.24 alkyl and an optionally iubstituted -O-aryl; and Z4A can be independently O (oxygen) or S (sulfur). In some 1 îmbodiments, R22A and R23A can be hydrogen. In other embodiments, at least one of R22A and <23A can be an optionally substituted C1.24 alkyl or an optionally substituted aryl. In some embodiments, R24A can be an optionally substituted C1.24 alkyl. In other embodiments, R24Acan >e an optionally substituted aryl. In still other embodiments, R24A can be an optionally j iubstituted -O-C1.24 alkyl or an optionally substituted -O-aryl. In some embodiments, Z4A can 1 >e O (oxygen). In other embodiments, Z4A can be or S (sulfur). In some embodiments, one or 1 )oth of R and R can be isopropylcarbonyloxymethyl. In some embodiments, one or both of : Ie A and R7A can be pivaloyloxymethyl.
independently -ON or an optionally substituted substituent selected from C2.8 organylcarbonyl, <^2-8 alkoxycarbonyl and C2-8 organylaminocarbonyl; R2aA can be selected from hydrogen, an optionally substituted Ci-24-alkyl, an optionally substituted C2.24 alkenyl, an optionally substituted C2.24 alkynyl, an optionally substituted C3^ cycloalkyl and an optionally substituted
Cj-β cycloalkenyl; and r can be 1 or 2. In some embodiments, R26A can be -CsN and R27A can be
m optionally substituted Cm alkoxycarbonyl, such as -C(=O)OCH3. In other embodiments, <26A can be -C=N and R27A can be an optionally substituted organylaminocarbonyt, for example, -C(=O)NHCH2CH3 and -C(=O)NHCH2CH2phenyI. In some embodiments, both R26A md R27A can be an optionally substituted C2_8 organylcarbonyl, such as -C(=O)CH3. In some embodiments, both R26A and R27A can be an optionally substituted Cm alkoxycarbonyl, for example, -C(=O)OCH2CH3 and -C(=O)OCHj. In some embodiments, including those lescribcd in this paragraph, R28A can be an optionally substituted Cm-alkyt. In some embodiment, R28A can be methyl or tert-butyl. In some embodiments, r can be 1. In other embodiments, r can be 2.
[0094] ollowing:
include, but are not limited to the
o
[0095] In some embodiments, R6A and R7A can be both an optionally substituted aryl.
In some embodiments, at least one of R6A and R7A can be an optionally substituted aryl. For example, both R6A and R7A can be an optionally substituted phenyl or an optionally substituted naphthyl. When substituted, the substituted aryl can be substituted with l, 2, 3 or more than 3 substituents. When more the two substituents are présent, the substituents can be the same or different. In some embodiments, when at least one of R6A and R7A is a substituted phenyl, the substituted phenyl can be a para-, ortho- or meta-substituted phenyl.
[0096] In some embodiments, R6A and R7A can be both an optionally substituted aryl (Cm alkyl). In some embodiments, at least one of R6A and R7A can be an optionally substituted aryl(CM alkyl). For example, both R6A and R7A can be an optionally substituted benzyl. When substituted, the substituted benzyl group can be substituted with 1, 2, 3 or more than 3 substituents. When more the two substituents are présent, the substituents can be the same or different. In some embodiments, the aryl group of the aryl(CM alkyl) can be a para-, artho- or meta-substituted phenyl.
[0097]
In some embodiments, Rîa and R7A can be both
some embodiments, at least one of RSA and R7A can be r ü R25A, In some embodiments, R23A can be hydrogen. In other embodiments, R23A can be an optionally substituted C124 alkyl. In still other embodiments, R2ÎA can be an optionally substituted aryl. In some embodiments, R23A can be a Cm alkyl, for example, methyl, ethyl, n-propyl, isopropyl, njutyl, isobutyl, tert-butyl, pentyl (branched and straight-chained), and hexyl (branched and straight-chained).
[0098]
In some embodiments, R6A and R7A can be both
In some embodiments, atleastoneofR6A andR7Acanbe R29*. Insomeembodiments, R29* can be hydrogen. In other embodiments, R29A can be an optionally substituted C144 alkyl. In i orne embodiments, R29* can be a Cm alkyl, such as methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl and t-butyl. In still other embodiments, R29A can be an optionally substituted aryl, such as an optionally substituted phenyl or an optionally substituted naphthyl.
7IA [0099] In some embodiments, , R1A can be
O
R12AO—P— or13A rBAq—
ÔR7A; R6A can be
L -Jm; R7A can be absent or hydrogen; Rl2A, R13A and Rl4A can be ndependently absent or hydrogen; and m can be 0 or 1. In some embodiments, m can be 0, and <7A, R12A and R,3A can be independently absent or hydrogen. In other embodiments, m can be I, and R7A, RI2A, R,3A and RHA can be independently absent or hydrogen. Those skilled in the irt understand that when m is 0, R6A can be diphosphate, when ZlA is oxygen, or an alphahiodiphosphate, when Z>A is sulfur. Likewise, those skilled in the art understand that when m is 1, R6A can be triphosphate, when Z,A is oxygen, or an alpha-thiotriphosphate, when Z1A is uulfur.
[0100]
In some embodiments, R6A and R7A can be taken together to form an *
optionally substituted
. For example, RlA can be an optionally substituted
. When substituted, the ring can be substituted 1,2, 3 or 3 or more times. When Substituted with multiple substituents, the substituents can be the same or different. In some
:mbodiments, when R , the ring can be substituted with an optionally lubstituted aryl group and/or an optionally substituted heteroaryl. An example of a suitable îeteroaryl is pyridinyl. In some embodiments, R6A and R7A can be taken together to form an
optionally substituted
such as
can be an optionally substituted aryl, an optionally substituted heteroaryl or an optionally substituted heterocyclyl.
[0101] In some embodiments, R6A and R7A can be taken together to form an optionally substituted
, wherein the oxygens connected to R6A and R7A, the ïhosphorus and the moiety form a six-membered to ten-membered ring system. Example of an
[0102] In some embodiments, R6A and R7A can be the same. In some embodiments, 1 l6A and R7A can be the different [0103] In some embodiments, Z1A can be oxygen. In other embodiments, Z1A can be i ulfur.
[0104]
In some embodiments, R1A can be
In some embodiments, R8A can be selected from absent, hydrogen, an optionally substituted C1.24 alkyl, an optionally j ubstituted C2-24 alkenyl, an optionally substituted C2.24 alkynyl, an optionally substituted C3-6 Cycloalkyl and an optionally substituted cycloalkenyl; and R9A can be independently selected from an optionally substituted C1.24 alkyl, an optionally substituted C2-24 alkenyl, an optionally substituted C2.24 alkynyl, an optionally substituted Cj_e cycloalkyl and an optionally substituted cycloalkenyl.
[0105] In some embodiments, R8A can be hydrogen, and R9A can be an optionally substituted Cm alkyl. Examples of suitable Cm alkyls include methyl, ethyl, n-propyl, hopropyl, n-butyl, isobutyl, tert-butyl, pentyl (branched and straight-chained), and hexyl ^branched and straight-chained). In other embodiments, R8A can be hydrogen, and R9A can be
NR30AR31A, wherein R30 and R31 can be independently selected from hydrogen, an optionally iubstituted C1.24 alkyl, an optionally substituted C2-24 alkenyl, an optionally substituted C2.24 ilkynyl, an optionally substituted C34 cycloalkyl and an optionally substituted C34 cycloalkenyl.
[0106] In some embodiments, R8A can be absent or hydrogen; and R9A can be an optionally substituted N-linked amino acid or an optionally substituted N-linked amino acid ;ster dérivative. In other embodiments, R8A can be an optionally substituted aryl; and R9A can be 1 in optionally substituted N-linked amino acid or an optionally substituted N-linked amino acid < :ster dérivative. In still other embodiments, R8A can be an optionally substituted heteroaryl; and <9A can be an optionally substituted N-linked amino acid or an optionally substituted N-linked iimino acid ester dérivative. In some embodiments, R9A can be selected from alanine, î isparagine, aspartate, cysteine, glutamate, glutamine, glycine, proline, serine, tyrosine, arginine, îistidine, isoleucine, leucine, lysine, méthionine, phenylalanine, threonine, tryptophan, valine Înd ester dérivatives thereof. Examples of an optionally substituted N-linked amino acid ester erivatives include optionally substituted versions of the following: alanine isopropyl ester,
Janine cyclohexyl ester, alanine neopentyl ester, valine isopropyl ester and leucine isopropyl ester. In some embodiments, R9A can hâve the structure
wherein R33A can be i elected from hydrogen, an optionally substituted Cw-alkyl, an optionally substituted C34 cycloalkyl, an optionally substituted aryl, an optionally substituted aryl(Ci-e alkyl) and an I ptionally substituted haloalkyl; R34A can be selected from hydrogen, an optionally substituted alkyl, an optionally substituted Ci4 haloalkyl, an optionally substituted C34 cycloalkyl, an ptionally substituted Ce aryl, an optionally substituted Cio aryl and an optionally substituted ryl(Ci4 alkyl); and R33A can be hydrogen or an optionally substituted Ci^-alkyl; or R34A and can be taken together to form an optionally substituted C34 cycloalkyl.
[0107] When R34A is substituted, R34A can be substituted with one or more substituents selected from N-amido, mercapto, alkylthio, an optionally substituted aryl, hydroxy, an optionally substituted heteroaryl, O-carboxy, and amino. In some embodiments, R34A can be In unsubstituted Ci4-alkyl, such as those described herein. In some embodiments, R34A can be ydrogen. In other embodiments, R34A can be methyl. In some embodiments, R33A can be an ptionally substituted C14 alkyl. Examples of optionally substituted Ci-e-alkyls include ptionally substituted variants of the following: methyl, ethyi, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, pentyl (branched and straight-chained), and hexyl (branched and straight32
chained). In some embodiments, R33A can be methyl or isopropyl. In some embodiments, R33A can be ethyl or neopentyl. In other embodiments, R33A can be an optionally substituted Cm cycloalkyl. Examples of optionally substituted Cm cycloalkyl include optionally substituted variants of the following: cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. In an embodiment, R33a can be an optionally substituted cyclohexyl. In still other embodiments, R33A can be an optionally substituted aryl, such as phenyl and naphthyl. In yet still other embodiments, R33A can be an optionally substituted aryl(Ci^ alkyl). In some embodiments, R33A can be an optionally substituted benzyl. In some embodiments, R33A can be an optionally substituted Cm haloalkyl, for example, CF3. In some embodiments, R3ÎA can be hydrogen. In Dther embodiments, R3ÎA can be an optionally substituted Ci^-alkyl, such as methyl, ethyl, npropyl, isopropyl, n-butyl, isobutyl and tert-butyl. In an embodiment, R3ÎA can be methyl. In some embodiments, R34A and R3ÎA can be taken together to form an optionally substituted C» cycloalkyl. Examples of optionally substituted Cm cycloalkyl include optionally substituted variants of the following: cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. Depending on the groups that are selected for R34A and R33A, the carbon to which R34A and R33A are attached may be a chiral center. In some embodiment, the carbon to which R34A and R33A are attached may be a (R)-chiral center. In other embodiments, the carbon to which R34A and R33A are attached may be a (S)-chiral center.
[0108] In some embodiments, when R1A is R9A , can be O (oxygen).
In other embodiments, when RlA îs R9A , can be S (sulfur).
Z3A
R1QA-J3— [0109] In some embodiments, R1A can be R11A . In some embodiments, R10A and Rua can be both an optionally substituted N-linked amino acid or an optionally substituted N-linked amino acid ester dérivative. In some embodiments, R10A and RUA can be independently selected from alanine, asparagine, aspartate, cysteine, glutamate, glutamine, glycine, praline, serine, tyrosine, arginine, histidine, isoleucine, leucine, lysine, méthionine, phenylalanine, threonine, tryptophan, valine and ester dérivatives thereof. In some embodiments, R10A and RllA can be an optionally substituted version of the following: alanine isopropyl ester, alanine cyclohexyl ester, alanine neopentyl ester, valine isopropyl ester and
ïeucine isopropyl ester. In some embodiments, R10A and RnA can independently hâve the structure
wherein R36A can be selected from hydrogen, an optionally substituted Ci^-alkyl, an optionally substituted cycloalkyl, an optionally substituted aryl, an ciptîonally substituted aryl(Cw alkyl) and an optionally substituted haloalkyl; R37A can be selected from hydrogen, an optionally substituted Ct^ alkyl, an optionally substituted Cu Îaloalkyl, an optionally substituted C34 cycloalkyl, an optionally substituted Ce aryl, an ptionally substituted Cio aryl and an optionally substituted aryl(Ci_6 alkyl); and R3SA can be ydrogen or an optionally substituted Cu-alkyl; or R37A and R38A can be taken together to form ώι optionally substituted C« cycloalkyl.
[0110] When R37A is substituted, R37A can be substituted with one or more substituents selected from N-amido, mercapto, alkylthio, an optionally substituted aryl, hydroxy, an optionally substituted heteroaryl, O-carboxy, andamino. In some embodiments, R37A can be an unsubstituted Cj^-alkyl, such as those described herein. In some embodiments, R37A can be 1 ydrogen. In other embodiments, R37A can be methyl. In some embodiments, R36A can be an optionally substituted Ci_6 alkyl. Examples of optionally substituted Ci^-alkyls include c ptionally substituted variants of the foliowing: methyl, ethyl, n-propyl, isopropyl, n-butyl, i «butyl, tert-butyl, pentyl (branched and straight-chained), and hexy! (branched and straightenained). In some embodiments, R36A can be methyl or isopropyl. In some embodiments, R3<5A Jan be ethyl or ncopentyl. In other embodiments, R36A can be an optionally substituted C« jycloalkyl. Examples of optionally substituted C34 cycloalkyl include optionally substituted ariants of the foliowing: cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. In an embodiment, R36A can be an optionally substituted cyclohexyl. In still other embodiments, R36* Jan be an optionally substituted aryl, such as phenyl and naphthyl. In yet still other Jmbodiments, R36* can be an optionally substituted aryl(Ci_6 alkyl). In some embodiments, R3®* can be an optionally substituted benzyl. In some embodiments, R36A can be an optionally Jubstituted Ci_6 haloalkyl, for example, CF3. In some embodiments, R38A can be hydrogen. In e ther embodiments, R38A can be an optionally substituted Cm-alkyl, such as methyl, ethyl, nj ropyl, isopropyl, n-butyl, isobutyl and tert-butyl. In an embodiment, R38A can be methyl. In some embodiments, R37A and R38A can be taken together to form an optionally substituted C« cycloalkyl. Examples of optionally substituted C3-6 cycloalkyl include optionally substituted variants of the foliowing: cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. Depending on t îe groups that arc selected for R37A and R38A, the carbon to which R37A and R38A are attached may be a chiral center. In some embodiment, the carbon to which R37A and R38A are attached may be a (R)-chiral center. In other embodiments, the carbon to which R37A and R38A are attached may be a (S)-chiral center.
[OUI] Examples of suitable
groups p34A R3®*
include the following:
[0112] In some embodiments, R10A and R1IA can be the same. In some embodiments, R,0A and R1,A can be the different.
[0113] In some embodiments, Z3A can be O (oxygen). In other embodiments, Z3A :an be S (sulfur).
[0114] In some embodiments, R1A can be hydrogen. In some embodiments, R1A can >e an optionally substituted acyl. In other embodiments, RlA can be -C(=O)R39A, wherein R39A ;an be selected from an optionally substituted C1-12 alkyl, an optionally substituted C2-12 alkenyl, m optionally substituted C2.12 alkynyl, an optionally substituted C3.8 cycloalkyl, an optionally substituted C34 cydoalkenyl, an optionally substituted Ce-io aryl, an optionally substituted leteroaryl, an optionally substituted heterocyclyl, an optionally substituted aryl(Ci-e alkyl), an jptionally substituted heteroaryl(Ci-6 alkyl) and an optionally substituted heterocyclyl(Ci_6 ilkyl). In some embodiments, R39A can be a substituted Cu2 alkyl. In other embodiments, R39A :an be an unsubstituted C1.12 alkyl.
[0115] In still other embodiments, R,A can be an optionally substituted O-linked imino acid. Examples of suitable O-lînked amino acids include alanine, asparagine, aspartate, ;ysteine, glutamate, glutamine, glycine, praline, serine, tyrosine, arginine, hîstidine, isoleucine, eucine, lysine, méthionine, phenylalanine, threonine, tryptophan and valine. Additional ïxamples of suitable amino acids include, but are not limited to, omithine, hypusine, 2iminoisobutyric acid, dehydroalanine, gamma-aminobutyric acid, citrulline, beta-alanine, alpha:thyl-glycine, alpha-propyl-glycine and norleucine. In some embodiments, the O-linked amino icid can hâve the structure
40A can be selected from hydrogen, an jptionally substituted Ci_c alkyl, an optionally substituted Ci-β haloalkyl, an optionally iubstituted C3-6 cycloalkyl, an optionally substituted Ce aryl, an optionally substituted Cio aryl md an optionally substituted aryl(Ci^ alkyl); and R4lA can be hydrogen or an optionally lubstituted Cu-alkyl; or R40A and R4lA can be taken together to form an optionally substituted
-3-6 cycloalkyl. Those skilled in the art understand that when RIA is an optionally substituted 0inked amino acid, the oxygen of R1AO- of Formula (I) is part of the optionally substituted 0-
inked amino acid. For example, when R1A is i s the oxygen of R1AO- of Formula (I).
[0116] When R40A is substituted, R40A can f the oxygen indicated with **♦’* be substituted with one or more ^ubstituents selected from N-amido, mercapto, alkylthio, an optionally substituted aryl, hydroxy, i n optionally substituted heteroaryl, O-carboxy, and amino. In some embodiments, R40A can be un unsubstituted Ci^-alkyl, such as those described herein. In some embodiments, R40A can be 1 lydrogen. In other embodiments, R40A can be methyl. In some embodiments, R41A can be hydrogen. In other embodiments, R41A can be an optionally substituted Ci-i-alkyl, such as methyl, ethyi, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl. In an embodiment, R41A can be methyl. Depending on the groups that are selected for R40A and R41A, the carbon to which r40A and R41A are attached may be a chiral center. In some embodiment, the carbon to which R40A and R4lA are attached may be a (R)-chîral center. In other embodiments, the carbon to t /hich R40A and R4lA are attached may be a (S)-chiral center.
[0117] Examples of suitable
J«A rU1A
include the following:
Ha
NH2
O
O *
NH2 f O NH2| O NH2,
[0118]
NHz and O NHî .
In some embodiments, the dashed line (----) can be a single bond, RM can t e CH2, and R3A can bc O (oxygen). When the dashed line (----) is a single bond, RM is CH2, and R3A is O (oxygen), a 4-membered ring is formed that includes the 4’-carbon and 3’-carbon c f the pentose ring. In other embodiments, the dashed line (----) can be absent, RM can be selected from an optionally substituted Ci^ alkyl, an optionally substituted C2^ alkenyl, an optionally substituted Cm alkynyl, an optionally substituted -Ο-Cm alkyl, an optionally substituted -O-C3-6 alkenyl, an optionally substituted -O-Cj^s alkynyl and cyano, and R3A can >e selected from OH, -OC(=O)R A and an optionally substituted O-linked amino acid.
[0119] Various groups can be attached to the 4*-position of the pentose ring. In some embodiments, R2* can be an optionally substituted Cm alkyl. Examples of suitable Cm i .lkyls include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, pentyl (branched nnd straight-chained), and hexyl (branched and straight-chalned). In some embodiments, Ru can be an unsubstituted Cm alkyl. In other embodiments, Ru can be a substituted Cm alkyl. l:or example, Ru can be a halogen substituted Cm alkyl, a hydroxy substituted Cm alkyl, an clkoxy substituted Cm alkyl or a sulfenyl substituted Cm alkyl (for example, -Cm alkyl-S-CM i lkyt). In other embodiments, R2A can be a Cm haloalkyl. In other embodiments, Ru can be an optionally substituted Cm alkenyl. In some embodiments, R2* can be a substituted Cm alkenyl. ] n other embodiments, R2* can be an unsubstituted Cm alkenyl. For example, Ru can be cthenyl, propenyl or atlenyl. In still other embodiments, R2* can be an optionally substituted C2. « alkynyl. In some embodiments, R2* can be a substituted C24s alkynyl. In other embodiments, R2* can be an unsubstituted Cm alkynyl. Suitable Cm alkynyls include ethynyl and propynyl. ] n yet still other embodiments, R1* can be an optionally substituted Cm cycloalkyl. In some «mbodimcnts, Ru can be a substituted Cm cycloalkyl. In other embodiments, Ru can be an unsubstituted C3-6 cycloalkyl. A non-limiting list of Cj^ cyctoalkyls include cyclopropyl, dyclobutyl, cyclopcntyl and cyclohexyl. In some embodiments, R2* can be an optionally î ubstituted -Ο-Cm alkyl. In some embodiments, R1* can be a substituted -Ο-Cm alkyl. In c ther embodiments, Ru can be an unsubstituted -Ο-Cm alkyl. Exemptes of suitable O-Cm t lkyl groups include methoxy, ethoxy, n-propoxy, iso-propoxy, n-butoxy, isobutoxy, tertbutoxy, pentoxy (branched and straight-chained), and hexoxy (branched and straight-chained). In other embodiments, Ru can be an optionally substituted -Ο-Cm alkenyl. In some embodiments, Ru can be a substituted -O-C3-6 alkenyl. In other embodiments, R2* can be an unsubstituted -O-Cî-e alkenyl. In still other embodiments, R2* can be an optionally substituted -O-C3-6 alkynyl. In some embodiments, R2* can be a substituted -Ο-Cm alkynyl. In other ¼bodiments, Ru can be an unsubstituted -Ο-Cm alkynyl. In yet still other embodiments, R2* can be cyano.
[0120] The groups attached to the 3*-position of the pentose ring can vary. In some embodiments, including those of paragraph [0119], R3A can be OH. In other embodiments, including those of paragraph [0119], R3A can be an optionally substituted O-linked amino acid.
Exemptes of suitable O-tinked amino acids include alanine, asparagine, aspartate, cysteine,
I ;lutamate, glutamine, glycine, proline, serine, tyrosine, arginine, hïstidine, isoleucine, teucine, lysine, méthionine, phenylatanine, threonine, tryptophan and valine. Additional examples of suitable amino acids inciude, but are not limited to, omithine, hypusine, 2-aminoisobutyric acid, (Lhydroatanine, gamma-aminobutyric acid, citrulline, beta-alanine, alpha-ethyl-glycine, alphaJiropyl-glycine and norteucine. In some embodiments, the O-linked amino acid can hâve the
structure O NH2 f wherein R42A can be selected from hydrogen, an optionally substituted Cm alkyl, an optionally substituted Ci_e haloalkyl, an optionally substituted cycloalkyl, an optionally substituted Ce aryl, an optionally substituted Cio aryl and an optionally s ubstituted aryl(Ci^ alkyl); and R43A can be hydrogen or an optionally substituted Cu-alkyl; or R42A and R43A can be taken together to form an optionally substituted Cj-β cycloalkyl.
[0121] When R42A is substituted, R4IA can be substituted with one or more substituents selected fromN-amido, mercapto, alkylthio, an optionally substituted aryl, hydroxy, rn optionally substituted heteroaryl, O-carboxy, and amino. In some embodiments, R42A can be rn unsubstituted Ci_6-alkyl, such as those described herein. In some embodiments, R42A can be hydrogen. In other embodiments, R42A can be methyl. In some embodiments, R43A can be tydrogen. In other embodiments, R43A can be an optionally substituted Cm-alkyl, such as riethyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl. In an embodiment, R43A can t e methyl. Depending on the groups that arc selected for R42A and R4JA, the carbon to which R42A and R43A are attached may be a chiral center. In some embodiment, the carbon to which Il42A and R4JA are attached may be a (R)-chiral center. In other embodiments, the carbon to which R42A and R43A arc attached may be a (S)-chiral center.
[0122]
Examples of suitable inciude the following:
O
NHa f O
NH2 and 0 [0123] In still other embodiments, including those of paragraph [0119], R3A can be OC(=O)R A, wherein R A can be an optionally substituted C1.24 alkyl. In some embodiments, < A can be a substituted Cm alkyl. In other embodiments, ra can be an unsubstituted Cm ilkyl. In still other embodiments, including those of paragraph [0119], R3A can be an optionally substituted -O-acyl, In yet still other embodiments, including those of paragraph [0119], R3A :an be -OC(=0)R44A, wherein R44A can be selected from an optionally substituted Ci.12 alkyl, an jptionally substituted C2.12 alkenyl, an optionally substituted C2-12 alkynyl, an optionally substituted C3.8 cycloalkyl, an optionally substituted Cm cycloalkenyl, an optionally substituted -6-10 aryl, an optionally substituted heteroaryl, an optionally substituted heterocyclyl, an iptionally substituted aryl(C|-e alkyl), an optionally substituted heteroaryl(CM alkyl) and an < pptionally substituted heterocyclyl(CM alkyl). In some embodiments, R44A can be a substituted 21.12 alkyl. In other embodiments, R44A can be an unsubstituted C1.12 alkyl.
[0124] Various substituents can be présentât the 2’-position ofthe pentose ring. In some embodiments, R3A can be hydrogen. In other embodiments, R3A can be halogen, for exemple, fluoro. In some embodiments, R4A can be halogen, such as fluoro. In some embodiments, R3A can be hydrogen and R4A can be halogen. In other embodiments, R4A and <îA can both be halogen.
[0125] In some embodiments, — can be a single bond, R4* can be fluoro, R3A can 1 >e hydrogen and R2A can be a Cm haloalkyl. In some embodiments, — can be a single bond, 1 <4A can be fluoro, R3A can be hydrogen, R2A can be a Cm haloalkyl and B1A can be cytosine.
[0126] In some embodiments, R2A cannot be methoxy. In some embodiments, R2A not be methoxy when B1A is substituted or unsubstituted uracil. In some embodiments, B1A substituted or unsubstituted cytosine. In other embodiments, BIA is substituted or ubstituted thymine. In still other embodiments, B1A cannot be an unsubstituted uracil. In
some embodiments, R2A cannot be methoxy when ZlA is
R9A , wherein R8A is an unsubstituted Cm alkyl or a para-substituted phenyl; and R9A is an optionally substituted Nhnked amino acid or an optionally substituted N-linked amino acid ester dérivative. In some ,2A « mbodiments, R2A cannot be methoxy when Z1A is
R9A . In some embodiments, R2A
:annot be an alkoxy (for example, when Z,A is R9A ). In some embodiments, B1A eannot be cytosine when R2* is an unsubstituted alkenyl or an unsubstiuted alkynyl. In some embodiments, B,A cannot be thymine when R2A is an optionally substituted alkyl. In some embodiments, R2A cannot be an unsubstituted alkoxy (such as methoxy), an optionally ; iubstituted alkenyl (such as allenyl), an unsubstituted alkynyl (such as ethynyl) or a Ci alkyl substituted with a non-halogen substituent. In some embodiments, R2A cannot be an ' insubstituted alkoxy (such as methoxy), an optionally substituted alkenyl (such as allenyl), an i eptionallys substituted substituted alkynyl (such as ethynyl) or a Cm alkyl substituted with a i ion-halogen substituent In some embodiments RlA cannot be H. In some embodiments R,A * :annot be H when BlA is an optionally substituted cytosine or an optionally substituted thymine.
[0127] Various optionally substituted heterocyclic bases can be attached to the ] >entose ring. In some embodiments, one or more of the amine and/or amino groups may be ] irotected with a suitable protecting group. For example, an amino group may be protected by i ransforming the amine and/or amino group to an amide or a carbamate. In some embodiments, an optionally substituted heterocyclic base or an optionally substituted heterocyclic base with one or more protected amino groups can hâve one of the following structures:
wherein: R^ can be selected from hydrogen, halogen and NHRJ2, wherein R12 can be selected from hydrogen, -CÎ^OjR1^2 and -C(=O)ORL2; RB2 can be halogen or NHRW2, wherein R1^2 can be selected from hydrogen, an optionally substituted Cm alkyl, an optionally substituted C2-e ί lkenyl, an optionally substituted C3-8 cycloalkyl, -C(=O)RM2 and -C(=O)ORN2; R02 can be l ydrogen or NHR02, wherein R02 can be selected from hydrogen, -C(=O)RP2 and -C(=O)ORQ2;
IL02 can be selected from hydrogen, halogen, an optionally substituted Cm alkyl, an optionally
substituted C2_6 alkenyl and an optionally substituted C2-6 alkynyl; RQ can be selected from lydrogen, hydroxy, an optionally substituted Cm alkyl, an optionally substituted Cm cycloalkyl, C(=O)RR2 and -C(=O)ORS2; Rn can be selected from hydrogen, halogen, an optionally substituted Cm alkyl, an optionally substituted C2-6 alkenyl and an optionally substituted C2-6 ilkynyl; Y2 and Y3 can be independently N (nitrogen) or CRn, wherein R12 can be selected from lydrogen, halogen, an optionally substituted CM-alkyl, an optionally substituted C2-6-alkenyl md an optionally substituted Cî-i-alkynyl; R02 can be an optionally substituted Cm alkyl; R112 :an be hydrogen or NHR72, wherein R72 can be independently selected from hydrogen, C(=O)RU2 and -C(=O)ORV2; and R*2, Ru, R342, RN2, Rn, R52, RM, RS2, Rm and R72 can be ndependently selected from Cm alkyl, C2-« alkenyl, Cm alkynyl, C» cycloalkyl, Cm cycloalkenyl, Ce-io aryl, heteroaryl, heteroalicyclyl, aryl(CM alkyl), heteroaryl(CM alkyl) and 1 ieteroalicyclyl(CM alkyl). In some embodiments, the structures shown above can be modified Dy replacing one or more hydrogens with substituents selected from the list of substituents provided for the définition of “substituted.”
[0128] In some embodiments, B1A can be wLa . In other
O II
0 embodiments, B1A can be . In still other embodiments, B1A can be
:an bc hydrogen. In other embodiments, B1A can be . In some embodiments, <B2 can be NH2. In other embodiments, RB2 can be NHR^, wherein R^ can be -C(=O)Rnc or
-C(=O)ORN2. In still other embodiments, B1A
In some
[0129] In some embodiments, a compound of Formula (I) can hâve a structure j elected from one of the following:
CO
, or a pharmaceutically acceptable sait of
1A the foregoing. In some embodiments of this paragraph, B1A can be an optionally substituted Eurine base. In other embodiments of this paragraph, B1A can be an optionally substituted yrimidine base. In some embodiments of this paragraph, B1A can be guanine. In other embodiments of this paragraph, B1A can be thymine. In still other embodiments of this jaragraph, B1A can be cytosine. In yet still other embodiments of this paragraph, B1A can be iracil. In some embodiments of this paragraph, B1A can be adenine. In some embodiments of his paragraph, R1A can be hydrogen. In other embodiments of this paragraph, R1A can be an optionally substituted acyl. In still other embodiments of this paragraph, R1A can be mono-, dior tri-phosphate. In yet other embodiments of this paragraph, R1A can be phosphoroamidate. In tome embodiments of this paragraph, R1 A can be an acyloxyalkyl ester phosphate prodrug.
[0130] In some embodiments, the compound can be a compound of Formula (Π), or a pharmaceutically acceptable sait thereof, wherein: Bia can be an optionally substituted îeterocyclic base or an optionally substituted heterocyclic base with a protected amino group;
an optionally substituted N-linked amino acid and an optionally substituted N-linked amino acid ester dérivative; R13 can be selected from an optionally substituted Cm alkyl, an optionally substituted C2^ alkenyl, an optionally substituted C2^ alkynyl, an optionally substituted -O-Ci_6 alkyl, an optionally substituted -O-C3-6 alkenyl, an < ptionally substituted -O-C3^ alkynyl and cyano; R3B can be a halogen; R4B can be hydrogen or halogen; RÎB, R68, R8B and R9B can be independently selected from hydrogen, an optionally substituted C1.24 alkyl and an optionally substituted aryl; R70 and R10B can be independently selected from hydrogen, an optionally substituted C1.24 alkyl, an optionally substituted aryl, an t ptionally substituted -O-C]_24 alkyl and an optionally substituted -O-aryl; R11B can be selected from hydrogen, an optionally substituted Ci.24 alkyl and an optionally substituted aryl; Z1B and
Z2D can be independently O or S.
[0131] In some embodiments, R1B can be O'. In other embodiments, R1B can be OH.
>68 [0132] In some embodiments, RlB can bc θ wherein RSB and R60 :an be independently selected from hydrogen, an optionally substituted C]_24 alkyl and an ' jptionally substituted aryl; and R™ can be selected from hydrogen, an optionally substituted Ci. : 4 alkyl, an optionally substituted aryl, an optionally substituted -O-Ci. 24 alkyl and an optionally substituted -O-aryl. In some embodiments, RJB and R60 can be hydrogen. In other Îmbodiments, at least one of RJB and R60 can be an optionally substituted C1.24 alkyl or an ptionally substituted aryl. In some embodiments, R7B can be an optionally substituted C1.24 nlkyl. In other embodiments, R70 can be an optionally substituted aryl. In still other < îmbodiments, R™ can be an optionally substituted -O-C1.24 alkyl or an optionally substituted aryl.
[0133] In some embodiments, RlB can be 0'
î nd R90 can be independently selected from hydrogen, an optionally substituted C1.24 alkyl and î n optionally substituted aryl; Rl0B can be independently selected from hydrogen, an optionally substituted C1.24 alkyl, an optionally substituted aryl, an optionally substituted -O-C1.24 alkyl î nd an optionally substituted -O-aryl; and Z20 can be independently O (oxygen) or S (sulfiir). In some embodiments, R80 and R90 can be hydrogen. In other embodiments, at least one of R80 and R90 can be an optionally substituted C1.24 alkyl or an optionally substituted aryl. In some embodiments, R100 can be an optionally substituted C1.24 alkyl. In other embodiments, R100 can be an optionally substituted aryl. In still other embodiments, R100 can be an optionally substituted -O-C1.24 alkyl or an optionally substituted -O-aryl. In some embodiments, Z20 can l e O (oxygen). In other embodiments, Z20 can be or S (sulfur). In some embodiments, R10 can be isopropylcarbonyloxymethyloxy. In some embodiments, R10 can be pivaloyloxymethyloxy.
[0134] In some embodiments, R10 can be . In some embodiments, R110 can be hydrogen. In other embodiments, R110 can be an optionally substituted Cj.24 alkyl. In still other embodiments, R some embodiments, Rl,B t s raight-chained).
. 0 can be an optionally substituted aryl. In can be a C|_e alkyl, for example, methyl, ethyl, n-propyl, isopropyl, nutyl, isobutyl, tert-butyl, pentyl (branched and straight-chained), and hexyl (branched and tv
[0135] In some embodiments, R18 can be an optionally substituted N-linked amino acid or an optionally substituted N-linked amino acid ester dérivative. For example, R1B can be optionally substituted version of the following: alanine, asparagine, aspartate, cysteine, glutamate, glutamine, glycine, proline, serine, tyrosine, arginine, histidine, isoleucine, leucine, lysine, méthionine, phenylalanine, threonîne, tryptophan, valine and ester dérivatives thereof. In some embodiments, RlB can be selected from alanine isopropyl ester, alanine cyclohexyl ester, alanine neopenty! ester, valine isopropyl ester and leucine isopropyl ester. In some
Îmbodiments, RlB can hâve the structure
can be selected from îydrogen, an optionally substituted Cu-alkyl, an optionally substituted Cm cycloalkyl, an jptionally substituted aryl, an optionally substituted aryt(Ci_6 alkyl) and an optionally ïubstituted haloalkyl; R138 can be selected from hydrogen, an optionally substituted Cm alkyl, in optionally substituted Cm haloalkyl, an optionally substituted Cm cycloalkyl, an optionally ; substituted C6 aryl, an optionally substituted Ci0 aryl and an optionally substituted aryl(CM alkyl); and R148 can be hydrogen or an optionally substituted Cu-alkyl; or R13B and R148 can be ' aken together to form an optionally substituted Cm cycloalkyl.
[0136] When R13B is substituted, R13B can be substituted with one or more ; ubstituents selected from N-amîdo, mercapto, alkylthio, an optionally substituted aryl, hydroxy, m optionally substituted heteroaryl, O-carboxy, and amino. In some embodiments, R13B can be î Ln unsubstituted Ci_e*alkyl, such as those described herein. In some embodiments, R13B can be hydrogen. In other embodiments, R138 can be methyl. In some embodiments, R12B can be an optionally substituted Cm alkyl. Examples of optionally substituted Ci-6-aIkyls include optionally substituted variants of the following: methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, pentyl (branched and straight-chained), and hexyl (branched and straightî hained). In some embodiments, R12B can be methyl or isopropyl. In some embodiments, R12B c an be ethyl or neopentyl. In other embodiments, R128 can be an optionally substituted Cm cycloalkyl. Examples of optionally substituted Cm cycloalkyl include optionally substituted variants of the following: cyclopropyt, cyclobutyl, cyclopentyl, and cyclohexyl. In an embodiment, R128 can be an optionally substituted cyclohexyl. In still other embodiments, R12B can be an optionally substituted aryl, such as phenyl and naphthyl. In yet still other embodiments, R128 can be an optionally substituted ary[(C]_6 alkyl). In some embodiments, R12B can be an optionally substituted benzyl. In some embodiments, R12B can be an optionally s ibstituted Cm haloalkyl, for example, CF3. In some embodiments, R148 can be hydrogen. In
other embodiments, R14B can be an optionally substituted Cm-alkyl, such as methyi, ethyl, n] sropyl, isopropyl, n-butyl, isobutyl and tert-butyl. In an embodiment, R14B can be methyi. In iiome embodiments, R13B and RI4B can be taken together to form an optionally substituted C« cycloalkyl. Examples of optionally substituted Cj-β cycloalkyl include optionally substituted variants of the following: cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. Depending on Îe groups that are selected for R13B and R14B, the carbon to which Rl3B and R14B are attached ay be a chiral center. In some embodimenL the carbon to which R13B and R14B are attached may be a (R)-chiral center. In other embodiments, the carbon to which R13B and R14B are î ttached may be a (S)-chiral center.
O
In some embodiments, R2B can be an optionally substituted Cm alkyl. Examples ofsuitable Cm ilkyls include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, pentyl (branched ind straight-chained), and hexyl (branched and straight-chained). In some embodiments, R2B :an be an unsubstituted Cm alkyl. In other embodiments, R2B can be a substituted Cm alkyl. Tor example, R23 can be a halogen substituted Cm alkyl, a hydroxy substituted Cm alkyl, an itkoxy substituted Cm alkyl or a sulfenyt substituted Cm alkyl (for example, -Cm alkyl-S-CM ilkyl). In other embodiments, R2B can be a Cm haloalkyl. . In other embodiments, R2B can be in optionally substituted Cm alkenyl. In some embodiments, R2B can be a substituted C« ; ilkenyl. In other embodiments, R2B can be an unsubstituted Cm alkenyl. For example, R2B can 1 >e ethenyl, propenyl or allenyl. In still other embodiments, R2B can be an optionally substituted Cm alkynyl. In some embodiments, R2B can be a substituted Cm alkynyl. In other embodiments, R2B can be an unsubstituted Cm alkynyl. Suitable Cm alkynyls include ethynyl end propynyl. In yet still other embodiments, R2B can be an optionally substituted Cm cycloalkyl. In some embodiments, R2B can be a substituted Cm cycloalkyl. ln other embodiments, R2B can be an unsubstituted Cm cycloalkyl. A non-limiting list of Cm (ycloalkyts include cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl. In some embodiments, R2B can be an optionally substituted -Ο-Cm alkyl. In some embodiments, R2B can be a substituted -Ο-Cm alkyl. In other embodiments, R2B can be an unsubstituted -O-Cm alkyl. Examples of suitable Ο-Cm alkyl groups include methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, tert-butoxy, pentoxy (branched and straight-chained), and hexoxy (branched and straight-chained). In other embodiments, R20 can be an optionally substituted Ô-Cm alkenyl. In some embodiments, R2B can be a substituted - O-C3^ alkenyl. In other < mbodiments, R2B can be an unsubstituted -Ο-Cm alkenyl. In still other embodiments, R2B can t e an optionally substituted -Ο-Cm alkynyl. ln some embodiments, R2B can be a substituted 20
O—Cm alkynyl. In other embodiments, R2B can be an unsubstituted -0~Cm alkynyl. In yet still ather embodiments, R2B can be cyano.
[0139] Variety of substituents can be présent at the 2*-position of the pentose ring, h some embodiments, R43 can be hydrogen. In other embodiments, R4B can be halogen, such as luoro. In some embodiments, R3B can be halogen, such as fluoro. In some embodiments, R4D ;an be hydrogen and R3B can be halogen. In other embodiments, R3B and R4B can be both talogen. For example, R3B and R4B can be both fluoro.
[0140] In some embodiments, Z1B can be O (oxygen). In other embodiments, ZlB :an be S (sulfur).
[0141] Various optionally substituted heterocyclic bases can be attached to the jentose ring. In some embodiments, one or more of the amine and/or amino groups may be ] >rotected with a suitable protecting group. For example, an amino group may be protected by Iransforming the amine and/or amino group to an amide or a carbamate. In some embodiments, tin optionally substituted heterocyclic base or an optionally substituted heterocyclic base with qne or more protected amino groups can hâve one of the following structures:
nhrEB2
» «λλλλ
ΛΛΛ/»
AB2 s
wherein R1 »
v'herein: R*32 can be selected from hydrogen, halogen and NHRra2, wherein Rro2 can be lïlected from hydrogen, -C(=O)RKB2 and -C^OJOR1*82; RBB2 can be halogen or NHRWB2,
-—L _^2 can be selected from hydrogen, an optionally substituted Cm alkyl, an optionally ubstituted C2-e alkenyl, an optionally substituted Cm cycloalkyl, -C(=O)RMBI and - - - NB2. rCbî can hydf-Qgen or nhrobi, wherein R032 can be selected from hydrogen, (=O)RPB2 and -C(=O)ORQB2; RDBI can be selected from hydrogen, halogen, an optionally s
C(=O)OR’
Ci s ibstituted Cm alkyl, an optionally substituted C2_6 alkenyl and an optionally substituted C2-e a kynyl; RED2 can be selected from hydrogen, hydroxy, an optionally substituted Cm alkyl, an i Jtionally substituted C3-8 cycloalkyl, -C^OJR1132 and -C(=O)ORSB2; Rra2 can be selected from
Ο hydrogen, halogen, an optionally substituted Cm alkyl, an optionally substituted C2^s alkenyl and an optionally substituted C2-6 alkynyl; Y2B and Y3B can be independently N (nitrogen) or CR®2, wherein R®2 can be selected from hydrogen, halogen, an optionally substituted Cw-alkyl, an optionally substituted Cm-alkenyl and an optionally substituted C2alkynyl; RGB2 can be an optionally substituted Cm alkyl; R1®2 can be hydrogen or NHR™, wherein R102 can be ndependently selected from hydrogen, -C(=O)RU02 and -C^OJOR702; and R1®2, RLB2, R3402, V®2, RPB2, RQB2, R1®2, RSB2, R1®2 and R3®2 can be independently selected from Cm alkyl, C2-6 ilkenyl, C2-e alkynyl, C3^ cycloalkyl, Cm cycloalkenyl, Cmo aryl, heteroaryl, heteroalicyclyl, ;iryl(CM alkyl), heteroaryl(CM alkyl) and hetcroalicyclyl(CM alkyl). In some embodiments, the structures shown above can be modified by replacing one or more hydrogens with substituents selected from the list of substituents provided for the définition of “substituted.”
embodiments, B1b [0142] In some embodiments, B
nhrEB2 , for example, -vlv can be hydrogen. In other embodiments, B
In still other embodiments, B1b can be yet still other embodiments, B1b can , In some embodiments, R002 . In some embodiments,
RBB2 can be NH2. In other embodiments, RBB2 can be NHR^2, wherein R^2 can be C(=O)Rmd2 or -C^OJOR™2.
In still other embodiments, B,b can be
rame embodiments, Bib can be
[0143] In some embodiments, a compound of Formula (II) can hâve the following
embodiments of this paragraph, B1b can be an optionally substituted purine base. In other embodiments of this paragraph, Bib can be an optionally substituted pyrimidine base. In some embodiments of this paragraph, B,b can be guanine. In other embodiments of this paragraph, î,B can be thymine. In still other embodiments of this paragraph, B1b can be cytosine. In yct still other embodiments of this paragraph, B,b can be uracil. In some embodiments of this jaragraph, B1b can be adénine. In some embodiments of this paragraph, ZIB can be oxygen. In some embodiments of this paragraph, Z,B can be sulfur. In still other embodiments of this jaragraph, R1B can be alkylcarbonyloxyalkoxy.
[0144] In some embodiments, the compound can be a compound of Formula (III), or a pharmaceutically acceptable sait thereof, wherein: B1C can be an optionally substituted îeterocyclic base or an optionally substituted heterocyclic base with a protected amino group; <1C and R2C can be independentlyselected from O, OH, an optionally substituted Cm alkoxy,
optionally substituted N-linked amino acid and an optionally substituted N-linked amino acid Aster dérivative; R3C can be selected from an optionally substituted Cm alkyl, an optionally substituted C2_6 alkenyl, an optionally substituted C2_6 alkynyl, an optionally substituted -O-Cm alkyl, an optionally substituted -O-C3-6 alkenyl, an optionally substituted -Ο-Cm alkynyl, an optionally substituted C3-6 cycloalkyl and cyano; R4C can be selected from OH, -OC(=O)Rc and m optionally substituted O-linked amino acid; R3C can be a halogen; R6C can be hydrogen or lalogen; R90, Rloc, Rl2C and Rl3C can be independently selected from hydrogen, an optionally substituted Ci-24 alkyl and an optionally substituted aryl; R1IC and Rl4C can be independently selected from hydrogen, an optionally substituted C1.24 alkyl, an optionally substituted aryl, an jptionally substituted -O-C1.24 alkyl and an optionally substituted -O-aryl; RIîC can be selected rom hydrogen, an optionally substituted Ομ24 alkyl and an optionally substituted aryl;----:an be a single bond or a double bond; when-----is a single bond, each R7C and each R80 can )e independently hydrogen or halogen; and when-----is a double bond, each R7C is absent and :ach R8C can be independently hydrogen or halogen; Z1C can be O (oxygen) or S (sulfur); and ] lc can be an optionally substituted Ci-24-alkyl.
[0145] In some embodiments,-----can be a single bond such that Formula (ΙΠ) has
the structure r5C , wherein each R7C and each R80 can be independently
I ydrogen or halogen. In some embodiments, the R7C and the R8C groups can ail be hydrogen. In t ther embodiments, one R7C can be halogen, one R7C can bc hydrogen and both Rsc groups can 8 11 be hydrogen. In still other embodiments, one R7C can be halogen, one R7C can be hydrogen, cne R8C can be halogen and one Rsc can be hydrogen. In some embodiments, the carbon adjacent to the phosphorus and the 5’-carbon can each be independently a (S)-chiral center. In same embodiments, the carbon adjacent to the phosphorus and the 5’-carbon can each be independently a (R)-chiral center.
[0146] In some embodiments,-----can be a double bond such that Formula (III)
r5C , wherein each R7C is absent and each R8C can be has the structure independently hydrogen or halogen. In some embodiments, both R80 groups can be hydrogen. In other embodiments, one R8C can be halogen and the other R8C can be hydrogen. In some embodiments, both R8C groups can be halogen. In some embodiments, the double bond has a ^-configuration, In some embodiments, the double bond has a (E)-configuration.
[0147] In some embodiments, Rlc and/or R2C can be O. In other embodiments, RIC md/or R2C can be OH. In some embodiments, R1C and R2C can be both OH.
□9C [0148] In some embodiments, R1C and/or R2C can be O wherein
I90 and Rloc can be independently selected from hydrogen, an optionally substituted C].24 alkyl ind an optionally substituted aryl; and Rllc can be selected from hydrogen, an optionally substituted Ci.24 alkyl, an optionally substituted aryl, an optionally substituted -O-Ci.24 alkyl md an optionally substituted -O-aryl. In some embodiments, R9C and Rloccan be hydrogen. In i jther embodiments, at least one of R90 and Rloc can be an optionally substituted C],24 alkyl or an optionally substituted aryl. In some embodiments, Rllc can be an optionally substituted C].24 alkyl. In other embodiments, R11C can be an optionally substituted aryl. In still other i :mbodiments, Rnccan be an optionally substituted -O-C1.24 alkyl or an optionally substituted R9C d10C Ao>Y,,c <)-aryl. In some embodiments, RIC and R2C can be both O .
[0149]
In some embodiments, RIC and/or R2C can be
Îherein R12C and R13C can be independently selected from hydrogen, an optionally substituted
1.24 alkyl and an optionally substituted aryl; RHC can be independently selected from hydrogen, optionally substituted C1.24 alkyl, an optionally substituted aryl, an optionally substituted-Oi?i_24 alkyl and an optionally substituted -O-aryl; and Z1C can be independently O (oxygen) or S sulfur). In some embodiments, R12C and Rnc can be hydrogen. In other embodiments, at least one of Rl2C and Rnc can be an optionally substituted Ci-24 alkyl or an optionally substituted iryl. In some embodiments, R14C can be an optionally substituted C].24 alkyl. In other embodiments, RI4C can be an optionally substituted aryl. In still other embodiments, RUC can be an optionally substituted -O-Ci.24 alkyl or an optionally substituted -O-aryl. In some ( mbodiments, Z1C can be O (oxygen). In other embodiments, ZIC can be or S (sulfur). In some embodiments, R1C and/or R2C can be isopropylcarbonyloxymethoxy. In some embodiments, Rlc and/or R2C can be pivaloyloxymethoxy. In some embodiments, R,c and R2C can be both r2C r2C
R14C.
i sopropylcarbonyloxymethoxy. ] ) i valoyloxymethoxy.
[0150] In some
O
In some embodiments,
In other embodiments, embodiments, R1C
R1C and
R,c and and/or R2C
S risc . In some embodiments, R13C can be can be can be can hydrogen. In both both be other c mbodiments, R,ÎC can be an optionally substituted C144 alkyl. In still other embodiments, R1ÎC can be an optionally substituted aryl. In some embodiments, R1ÎC can be a Ci-e alkyl, for cxamplc, methyi, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, pentyl (branched and straight-chained), and hexyl (branched and straight-chained). In some embodiments, Rlc and
F .2C can be both [0151] In some embodiments, R,c and/or R2C can be an optionally substituted Nnked amino acid or an optionally substituted N-linked amino acid ester dérivative. For ticample, R,c and/or R2C can be optionally substituted version of the following: alanine, e
a iparagine, aspartate, cysteine, glutamate, glutamine, glycine, proline, serine, tyrosine, arginine, histidine, isoleucine, leucine, lysine, méthionine, phenylalanine, threonine, tryptophan, valine a a
aid leucine isopropyl ester. In some embodiments, R1C and/or R' . id ester dérivatives thereof. In some embodiments, R1C and/or R2C can be selected from . anine isopropyl ester, alanine cyclohexyl ester, alanine neopentyl ester, valine isopropyl ester 2C can hâve the structure
, wherein R19C can be selected from hydrogen, an optionally substituted C^kyl, an optionally substituted C34 cycloalkyl, an optionally substituted aryl, an optionally substituted aryI(C]^ alkyl) and an optionally substituted haloalkyl; R20C can be selected from hydrogen, an optionally substituted Ci_6 alkyl, an optionally substituted Cu haloalkyl, an optionally substituted C3-6 cycloalkyl, an optionally substituted Ce aryl, an optionally substituted Cio aryl and an optionally substituted aryl(Ci_e alkyl); and R2IC can be hydrogen or an optionally sx.bstituted Ci-4-alkyl; or R200 and R2IC can be taken together to form an optionally substituted C.w cycloalkyl.
[0152] When R20C is substituted, R20C can be substituted with one or more substituents selected from N-amido, mcrcapto, alkylthio, an optionally substituted aryl, hydroxy, i in optionally substituted heteroaryl, O-carboxy, and amino. In some embodiments, R20C can be i in unsubstituted Cj^-alkyl, such as those described herein. In some embodiments, R200 can be hydrogen. In other embodiments, R20C can be methyl. In some embodiments, R19C can be an optionally substituted Ci-β alkyl. Examples of optionally substituted Ci4-alkyls include optionally substituted variants of the following: methyl, ethyi, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, pentyl (branched and straight-chained), and hexyl (branched and straightchained). In some embodiments, R19C can be methyl or isopropyl. In some embodiments, R19C can be ethyi or neopentyl. In other embodiments, R19C can be an optionally substituted C34 rycloalkyl. Examples of optionally substituted C34 cycloalkyl include optionally substituted variants of the following: cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. In an embodiment, Rl9C can be an optionally substituted cyclohexyl. In stillother embodiments, R19C can be an optionally substituted aryl, such as phenyl and naphthyl. In yet still other embodiments, Rl9C can be an optionally substituted aryl(C]4 alkyl). In some embodiments, R19C Jan be an optionally substituted benzyl. In some embodiments, R19C can be an optionally substituted C14 haloalkyl, for example, CF3. In some embodiments, R21C can be hydrogen. In ether embodiments, R21C can be an optionally substituted Ci^-alkyl, such as methyl, ethyi, npropyl, isopropyl, n-butyl, isobutyl and tert-butyl. In an embodiment, R2lc can be methyl. In same embodiments, R200 and R2lc can be taken together to form an optionally substituted C34 Îcloalkyl. Examples of optionally substituted C34 cycloalkyl include optionally substituted riants of the following: cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. Depending on tl 1e groups that are selected for R20C and R2IC, the carbon to which R20C and R21C are attached may be a chiral center. In some embodiment, the carbon to which R20C and R21C are attached may be a (R)-chiral center. In other embodiments, the carbon to which R20C and R21C are a lached may be a (S)-chiral center.
suitable [0153] Examples of
groups include the following:
[0154] In some embodiments, R1C and R2C can be the same. In other embodiments, IL1C and R2C can be different.
RieCQ_| ôr17C
LJ-o-L
I ... I ... 4
ORiac Jn and R2C can will be a diphosphate. In other embodiments, when n is 1, the compound of Formula (ΙΠ) will be a triphosphate.
[0155] In some embodiments, R1C can be l e O or OH, wherein R16C, R17C and R1SC can be absent or hydrogen; and n can be 0 or 1. Those skilled in the art understand that when R16C, R17C and/or RI8C are absent, the associated oxygen will be negatively charge. In some embodiments, when n is 0, the compound of Formula (III) [0156] A variety of substituents can be présent at the 4*-position of the pentose ring. 1 n some embodiments, R3C can be an optionally substituted Cm alkyl. Examples of suitable Cm i lkyls include methyl, ethyi, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, pentyl (branched tnd straight-chained), and hexyl (branched and straight-chained). In some embodiments, R3C Îan be an unsubstituted Cm alkyl. In other embodiments, R3C can be a substituted Cm alkyl. or example, R3C can be a halogen substituted Cm alkyl. In other embodiments, R3C can be an t ptionally substituted C2-6 alkenyl. In some embodiments, R3C can be a substituted Cm alkenyl. In other embodiments, R3C can be an unsubstituted Cm alkenyl. For example, R3C can be ethenyl, propenyl or allenyl. In still other embodiments, R3C can be an optionally substituted C2. alkynyl. In some embodiments, R3C can be a substituted Cm alkynyl. In other embodiments, can be an unsubstituted C2-« alkynyl. Suitable Cm alkynyls include ethynyl and propynyl.
’ ‘ “ _.3C can be an optionally substituted Cm cycloalkyl. In some can be an
R3C yet still other embodiments, R embodiments, R3C can be a substituted Cm cycloalkyl. In other embodiments, R30___________ unsubstituted Cm cycloalkyl. A non-limiting list of Cm cycloalkyls include cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl. In some embodiments, R3C can be an optionally s ibstituted -O-Cm alkyl. In some embodiments, R3C can be a substituted -Ο-Cm alkyl. In other embodiments, R3C can be an unsubstituted -O—Cm alkyl. Examples of suitable O-Cm akyl groups include methoxy, ethoxy, n-propoxy, iso-propoxy, n-butoxy, isobutoxy, tertb itoxy, pentoxy (branched and straight-chained), and hexoxy (branched and straight-chained). In other embodiments, R3C can be an optionally substituted -Ο-Cm alkenyl. In some enbodiments, R3C can be a substituted -O~C3_e alkenyl. In other embodiments, R3C can be an unsubstituted -O-C3-6 alkenyl. In still other embodiments, R3C can be an optionally substituted - 2>-Cm alkynyl. In some embodiments, R3C can be a substituted -O—Cm alkynyl. In other e nbodiments, R3C can be an unsubstituted -O-C3-6 alkynyl. In yet still other embodiments, R3C can be cyano.
[0157] The substituents that can be présent on the 3‘-position of the pentose ring can viry. In some embodiments, R4C can be OH. In other embodiments, R4C can be an optionally substituted O-linked amino acid. Examples of suitable O-linked amino acids include alanine, asparagine, aspartate, cysteine, glutamate, glutamine, glycine, proline, serine, tyrosine, arginine, histidine, isoleucine, leucine, lysine, méthionine, phenylalanine, threonine, tryptophan and v iline. Additional examples of suitable amino acids include, but are not limited to, omîthine, hjrpusine, 2-aminoisobutyric acid, dehydroalanine, gamma-aminobutyric acid, citrulline, betaalanine, alpha-ethyl-glycine, alpha-propyl-glycine and norleucine. In some embodiments, the , wherein R22C
O-linked amino acid can hâve the structure
can be selected : rom hydrogen, an optionally substituted Cm alkyl, an optionally substituted Cm haloalkyl, an Aptionally substituted Cm cycloalkyl, an optionally substituted C6 aryl, an optionally substituted < 2io aryl and an optionally substituted aryl(Ci_6 alkyl); and R231 can be hydrogen or an optionally ! ubstituted Ci-4-alkyl; or R220 and R23C can be taken together to form an optionally substituted * -3-6 cycloalkyl.
[0158] When R22C is substituted, R22C can be substituted with one or more substituents selected fromN-amido, mercapto, alkylthio, an optionally substituted aryl, hydroxy, un optionally substituted heteroaryl, O-carboxy, and amino. In some embodiments, R22C can be un unsubstituted CM-alkyl, such as those described herein. In some embodiments, R22C can be îydrogen. In other embodiments, R220 can be methyl. In some embodiments, R23C can be îydrogen. In other embodiments, R23C can be an optionally substituted Cu-alkyl, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl. In an embodiment, R23C can je methyl. Depending on the groups that are selected for R22C and R23C, the carbon to which <22C and R23C are attached may be a chiral center. In some embodiment, the carbon to which : <22C and R230 are attached may be a (R)-chiral center. In other embodiments, the carbon to 1 vhich R22C and R230 are attached may be a (S)-chiral center.
[0159]
Examples of suitable include the following:
[0160] In still other embodiments, R4C can be -OC^OjR0, wherein R° can be an i jptionally substituted C1.24 alkyl. In some embodiments, RC can be a substituted Ci. 12 alkyl. In jther embodiments, R”0 can be an unsubstituted Cm 2 alkyl. In still other embodiments, R0 can >e a substituted Cm alkyl. In yet still other embodiments, R° can be an unsubstituted Cm ilkyl. In some embodiments, R4C can be an optionally substituted acyl. In other embodiments,
R4C can be -OC(=O)Rc, wherein R”c can be selected from an optionally substituted C1.12 alkyl, an optionally substituted C2-12 alkenyl, an optionally substituted C2-12 alkynyl, an optionally iubstituted Cj-g cycloalkyl, an optionally substituted Cj_8 cycloalkenyl, an optionally substituted Çs-io aryl, an optionally substituted heteroaryl, an optionally substituted heterocyclyl, an optionally substituted aryl (Cm alkyl), an optionally substituted heteroaryl(CM alkyl) and an optionally substituted heterocyclyl(Ci_6 alkyl). In some embodiments, R c can be a substituted Ci-12 alkyl. In other embodiments, R0 can be an unsubstituted C1-12 alkyl.
]0161] A variety of substituents can also be présent at the 2’-position of the pentose nng. In some embodiments, R6C can be hydrogen. In other embodiments, R6C can be halogen, such as fluoro. In some embodiments, Rsc can be halogen, such as fluoro. In some embodiments, R60 can be hydrogen and RÎC can be halogen. In other embodiments, RÎC and R6C can be both halogen. For example, RÎC and R6C can be both fluoro.
[0162] Various optionally substituted heterocyclic bases can be attached to the pentose ring. In some embodiments, one or more of the amine and/or amino groups may be protected with a suitable protecting group. For example, an amino group may be protected by t •ansforming the amine and/or amino group to an amide or a carbamate. In some embodiments, an optionally substituted heterocyclic base or an optionally substituted heterocyclic base with one or more protected amino groups can hâve one of the foliowing structures:
NHREC2
I substituted Cm alkyl, an optionally substituted C2-6 alkenyl and an optionally substituted C2-6 wherein: RAC2 can be selected from hydrogen, halogen and NHRJC2, wherein RJC2 can be sïlectcd from hydrogen, -C(=O)RKC2 and -C(=O)ORLC2; RDC2 can be halogen or NHRWC2, wherein RWC2 can be selected from hydrogen, an optionally substituted Cm alkyl, an optionally s ibstituted C2-6 alkenyl, an optionally substituted Cm cycloalkyl, -C(=O)RMC2 and C (=O)ORNC2; RCC2 can be hydrogen or NHR002, wherein R002 can be selected from hydrogen, Ç(=O)RPC2 and -QOjOR902; R002 can be selected from hydrogen, halogen, an optionally ; tlkynyl; Ræ2 can be selected from hydrogen, hydroxy, an optionally substituted Cm alkyl, an optionally substituted C3-8 cycloalkyl, -C(=O)RRC2 and -C(=0)ORSC2; RFC2 can be selected from
îydrogen, halogen, an optionally substituted Cm alkyl, an optionally substituted C2-6 alkenyl and 1 in optionally substituted C2-6 alkynyl; Y2C and Y3C can be independently N (nitrogen) or CRIC2, vhereîn R1C2 can be selected from hydrogen, halogen, an optionally substituted CM-alkyl, an ?ptionally substituted C2^-aIkenyl and an optionally substituted C2^-alkynyl; R002 can be an ptionally substituted Cm alkyl; RI1C2 can be hydrogen or NHR702, wherein R702 can be mdependently selected from hydrogen, -C(=O)RUC2 and -C(=O)ORVC2; and RKC2, RLC2, RMC2, RNC2, RPC2, R*202, RRC2, RSC2, RUC2 and RVC2 can be independently selected from Cm alkyl, Cm hlkenyl, C2-e alkynyl, Cm cycloalkyl, C3-« cycloalkenyl, Ce-ιο aryl, heteroaryl, heteroalicyclyl, ârylfCM alkyl), heteroaryl(CM alkyl) and heteroalicyclyl(CM alkyl). In some embodiments, the structures shown above can be modified by replacing one or more hydrogens with substituents s elected from the list of substituents provided for the définition of “substituted.*’
embodiments, B1C
In still other [0163] In some embodiments, B1C
embodiments, B1C can be
NHREC2 , for example, . In some embodiments, R0^
In yet still other embodiments, B1C can
can be hydrogen. In other embodiments, B1C can be . In some embodiments,
I DC2 can be NH2. In other embodiments, RBC2 can be NHRWC2, wherein RWC2 can be C(=O)RMC2 or-C(=O)ORNC2. In still other embodiments, B1C
some embodiments, B1C can be
[0164]
In some embodiments, the compound of Formula (III) can hâve one of the following
R/ structures:
R2C
F
R** R50
R2C
LC
t
R5C . In s jme embodiments of this paragraph, B1C can be an optionally substituted purine base. In other embodiments of this paragraph, Blc can be an optionally substituted pyrimidine base. In some embodiments of this paragraph, B1C can be guanine. In other embodiments of this paragraph, E 1C can be thymine. In still other embodiments of this paragraph, B1C can be cytosine. In yet s :ill other embodiments of this paragraph, B1C can be uracil. In some embodiments of this paragraph, Blc can be adenine. In some embodiments of this paragraph, Rlc and R2C can each be an optionally substituted ϋμ alkyl. In other embodiments of this paragraph, RlA can be an φ : optionally substituted acyl. In stitl other embodiments of this paragraph, R1C and R2C can form a nono-, di- or tri-phosphate. In yet other embodiments of this paragraph, R1C and R2C can each je an alkytcarbonytoxyalkoxy. In some embodiments of this paragraph, R4C can be OH. In î tome embodiments of this paragraph, RÎC can be F and R6C can be hydrogen.
[0165] Examples of suitable compounds of Formula (I) include, but are not limited to he following:
'oregoing.
[0166] Additional examples of a compound of Formula (I) include the following:
ββ
hJLJLJAh Ah Ah
[0t67] Further examples of a compound of Formula (I) include, but are not limited to î he following:
es
sait of the foregoing.
[0168] Examples of a compound of Formula (II) include, but are not limited to, the following:
[0169] Exemples of a compound of Formula (III) inciude, but are not limited to, the following:
F
pharmaceutically acceptable sait of the foregoing.
[0170] Further examples of a compound of Formula (ΙΠ) include, but are not limited t), the following:
pharmaceutically acceptable sait of the foregoing.
Synthesis [0171] Compounds of Formula (I) Formula (II) and Formula (III), and those described herein may be prepared in various ways. Some compounds of Formulae (I), (II) and ( II) can be obtained commercially and/or prepared utilizing known synthetic procedures. General synthetic routes to the compounds of Formulae (I), (Π) and (ΠΙ), and some examplcs of s arting materials used to synthesize the compounds of Formulae (I), (II) and (ΠΙ) arc shown and described herein. The routes shown and described herein are illustrative only and arc not intended, nor are they to be construed, to limit the scope of the claims in any manner whatsoever. Those skitled in the art will be able to recognize modifications of the disclosed s;mtheses and to devise altemate routes based on the disclosures herein; ail such modifications a id altemate routes arc within the scope of the claims.
; îchemc 1
(A)
(B)
[0172] As shown in Scheme 1, compounds of Formula (I) can be prepared from a nucleoside, for exampie, a nucleoside of Formula (A). In Scheme 1, R3, R41, R3*, and B11 can t protecting group. A hydroxyalkyt group can be formed at the 4’-position of the pentose ring using suitable conditions known to those skilled in the art. Examples of suitable conditions for forming a hydroxyalkyt include the use of 2-iodoxybenzoic acid (IBX) aqueous formaldéhyde aid sodium borohydridc. A compound of Formula (B) can be oxidized to an aldéhyde using a si litable oxidizing agent(s) to form a compound of Formula (C). An example of suitable o e the same as R3A, R4A, RSA, and BlA as described herein for Formula (I), and PG* is a suitable xïdizing agent is Dess-Martin periodinane. An optionally substituted C2^ alkenyl or an • stionally substituted C2-6 alkynyl can be formed at the 4*-position using methods known to efination reaction, and Corey Fuchs reaction. An optionally substituted Cm alkyl can be o
those skilled in the art, for exampie, Wittig reagent and n-BuLi, Wittig-type reactions, Peterson o obtained by hydrogenating the unsaturatcd group attached to the 4*-position, for exampie, using h; ïdrogen over palladium on carbon.
[0173] Attematively, a compound of Formula (B) can be transformed to a haloalkyl uiing a suitable agent(s), for exampie, to an iodide using imidazote, triphenylphosphine and iedine; to a fluoro using diethytaminosulfur trifluoride (DAST); or to a chtoro using tr phenytphosphine and carbontetrachloride in dichloroethylene (DCE). An iodoalkyl can be tr insformed to an unsubstituted Cm alkyl group using methods known to those skilled in the art, fc r exampie, hydrogen over palladium on carbon. A compound of Formula (C) can be reacted w ith hydroxylamine to form an oxime. The oxime can be transformed to a cyano group using msthods known to those skilled in the art, for example, using mcthanesulfonyl chloride.
7S
Scheme 2
SU
Fo [0174] As shown in Scheme 2, compounds of Formula (I), where R2A is an t ptionally substituted -Ό-Ci^ alkyl, an optionally substituted -O-Cj-e alkenyl or an optionally ^ibstitutcd --O-C34S alkynyl, can be prepared from a nucleoside, for exemple, a nucleoside of 'ormuta (A). In Scheme 2, R2*, R3*, R4*, R5* and B1* can be the same as R2*, R3A, R4A, R3A and B1A as described herein for Formula (I), and PG2 can be a suitable protecting group. The nucleoside can undergo élimination and form an otefin having the general formula of Formula D). A compound of Formula (D) can be treated with an iodinating reagent in the presence of :ad carbonate and an alkoxy source to form a compound of Formula (E). A compound of ormula (E) can then be transformed to a compound of Formula (I) through disptacemcnt of the l!
F<
iodide with an oxygen nucteophile.
-7677
Scheme 3
(F)
Scheme 4
[0175] Compounds of Formula (I) having a phosphorus containing group attached to position of the pentose ring can be prepared using various methods known to those skilled tlie 5*-] in the art. Examples of methods are shown in Schemes 3 and 4. A phosphorus containing precursor can be coupied to the nucleoside, for example, a compound of Formula (F) or a c jmpound of Formula (G). As shown in Scheme 3, following the coupling of the phosphorus containing precursor, any leaving groups can be cleaved under suitable conditions, such as hVdrolysis. Further phosphorus containing groups can be added using methods known to those skilled in the art, for example using a pyrophosphate.
[0176] In some embodiments, an alkoxide can be generated from a compound of F irmula (G) using an organometallic reagent, such as a Grignard reagent. The alkoxide can be coupied to the phosphorus containing precursor. Suitable Grignard reagents arc known to those skilled in the art and include, but are not limited to, alkylmagnesium chtorides and alkylmagnesium bromides. In some embodiments, an appropriate base can be used. Exemples suitable bases include, but are not limited to, an amine base, such as an alkyl amine (including llcyl or st mMio-, di- and tri-alkylamines (e.g., triethylamîne)), optionally substituted pyridines (e.g. col ph pb llidine) and optionally substituted imidazolcs (e.g., N-methytimidazole)). Altematively, a osphorus containing precursor can be added to the nucleoside and form a phosphite. The osphite can be oxidized to a phosphate using conditions known to those skilled in the art.
Suitable conditions include, but are not limited to, meta-chloropcroxybenzoic acid (MCPBA) ind iodine as the oxidizing agent and water as the oxygen donor.
[0177] When compounds of Formula (I) hâve ZlA, Z2* or Z3A being sulfur, the sulfur can be added in various manners known to those skilled in the art. In some embodiments, the ReAQ_ C|orOH i ulfur can be part of the phosphorus containing precursor, for example, R7AO or n^o—|—a
R9A . Altematively, the sulfur can be added using a sulfurization reagent. Suitable sulfurization agents are known to those skilled in the art, and include, but are not limited to, elemental sulfur, Lawesson's reagent, cyclooctasulfur, 3H-!^-Bcnzodithiole-3-one-l,l-dioxide (3eaucage’s reagent), 3-((N,N-dimethylaniinomethyIidene)amino)-3H-l,2,4-dithiazole-5-thione ( DDTT) and bis(3-triethoxysilyl)propyl-tetrasulfide (TEST).
[0178] Suitable phosphonis containing precursors can be commercially obtained or > repared by synthetic methods known to those skilled in the art. Examples of general structures i F phosphorus containing precursors are shown in Schemes 3 and 4.
:heme 5:
oxidation reagent (Z1B “ O) sulfurization reagent (Z1B “ S)
[0179] A method for forming a compound of Formula (II) is shown in Schemc 5. In Scheme 5, Rlb, R2b, R3b, R4b and Blb can be the same as R1b, R2B, R3b, R4B and B,b as described herein for Formula (Π), each L1 can be a halogen, a sulfonate ester or an amine (mono- or di substituted), and X can be oxygen or sulfur. As shown in Scheme 5, a compound having a hydroxy group attached to the 3’-carbon and a hydroxy group attached to the 5’-carbon can be rei icted with a compound having the formula, (R,b)P(L’)2, in the presence of a base, to produce a
phosphite compound. Suitable bases are known to those skitled in the art and described herein. The phosphores can then be oxidized to phosphorus(V) using a suitable oxidizing agent, to produce a compound where X is O (oxygen). Altematively, the phosphite compound can be reacted with a sulfurization reagent to produce a compound where X is S (sulfur). Suitable oxidizing and sulfurization agents are known to those skilled in the art. For example, the jxidation can be carried out using iodine as the oxidizing agent and water as the oxygen donor. Suitable sulfurization agents are described herein.
Scheme 6
[0180| A method for forming a compound of Formula (III) is shown in Scheme 6. In Scheme 6, Rlc, R2c, R3c, R4', R3', R6® and Blc can be the same as RIC, RÎC, RÎC, R4C, Rsc, R60 and B,c as described herein for Formula (ΠΙ), and R70 and R8C are not shown. The oxygen attached ti j the 5’-carbon of the compound of Formula (H) can be oxidized to a ketone using methods and reagents known to those skilled in the art. For example, an oxidizing agent, such as Dess-Martin periodinane, can be utilized. A phosphorus-containing reagent can then be added to a compound f Formula (J) in the presence of a strong base (e.g., sodium hydride). The double bond can be li ydrogenated, for example using hydrogen gas or Pd/C, to a single bond. Additional phosphates cm be added via phosphorylation to form a di- or tri-phosphate using suitable reagents, such as a, pyrophosphate (e.g., tetrabutylammonium pyrophosphate).
[0181] An acyl group can be added to the 5'-position and/or the 3'-position of a compound of Formula (I) or (ΙΠ) using methods known to those skilled in the art. One suitable r lethod is using an anhydride in pyridine.
[0182] During the synthesis of any of the compounds described herein, if desired, s ny hydroxy groups attached to the pentose ring, and any -NH and/or NH2 groups présent on the Bla, Blb and B!c can be protected with one or more suitable protecting groups. Suitable p rotectïng groups are described herein. For example, when R3* and/or R4c is a hydroxy group, R3* and/or R4® can be protected with a triarylmethyl group or a silyl group. Likewise, any -NH and/or NH2 groups présent on the B1*, BIb and B!e can be protected, such as with a triarylmethyl and a silyl group(s). Examples of triarylmethyl groups include but are not limited to, trityl, monomethoxytrityl (MMTr), 4,4'-dimethoxytrityl (DMTr), 4,4',4-trimethoxytrityl (TMTr),. 4,4',4-tris- (benzoyloxy) trityl (TBTr), 4,4',4-tris (4,5-dichlorophthalimido) trityl (CPTr), ^'/P-tris (levulinyloxy) trityl (TLTr), p-anîsyl-1- naphthylphenylmethyl, di-o-anisyl-1naphthylmethyl, p-tolyldipheylmethyl, 3-(imidazolylmethyl)-4,4'-dimethoxytrityl, 9Îenylxanthen-9-yl (Pixyl), 9-(p-methoxyphenyl) xanthen-9-yl (Mox), 4-decyloxytrityl, 4xadecyloxytrityl, 4,4'-dioctadecyltrityl, 9-(4- octadecyloxyphenyl) xanthen-9-yl, l,l'-bis-(4rlethoxyphenyl)-l’-pyrenylmethyl, 4,4',4-tris- (tert-butylphenyl) methyl (TTTr) and 4,4'-di-3, 5 -hexadienoxytrityl. Examples of silyl groups include, but are not limited to, trimethylsilyl C rMS), tert-butyldimethylsilyl (TBDMS), triisopropylsilyl (TIPS), tert-butyldiphenylsilyl f rBDPS), tri-£so-propylsilyloxymethyl and [2-(trimethylsilyl)ethoxy]methyl. Altematively, R3* a îd R4* and/or R4® and Rîe can be protected by a single achiral or chiral protecting group, for e: cample, by forming an orthoester, a cyclic acetal or a cyclic ketal. Suitable orthoesters include lethoxymethylene acetal, ethoxymethylene acetal, 2-oxacyclopentylidene orthoester, methoxymethylene orthoester, 1-methoxyethylidene orthoester, 1-ethoxyethylidene orthoester, π d nethylidene orthoester, phthalide orthoester 1,2-dimethoxyethylidene orthoester, and alpharr ethoxybenzylidene orthoester; suitable cyclic acetals include methylene acetal, ethylidene acetal, t-butylmethylidene acetal, 3-(benzyloxy)propyl acetal, benzylidene acetal, 3,4d methoxybenzylidene acetal and p-acetoxybenzylidene acetal; and suitable cyclic ketals include t-butylethylidene ketal, l-phenylethylidene ketal, isopropylidene ketal, cyclopentylidene ketal, cyclohexylidene ketal, cycloheptylidene ketal and l-(4-methoxyphenyl)ethylidene ketal. Those skilled in the art will appreciate that groups attached to the pentose ring and any -NH and/or
NH2 groups présent on the BIe, Blb and Blc can be protected with various protecting groups, and any protecting groups présent can be exchanged for other protecting groups. The sélection and exchange of the protecting groups is within the skill of those of ordinary skill in the art. Any protecting group(s) can be removed by methods known in the art, for example, with an acid (e.g., a minerai or an organic acid), a base or a fluoride source.
Pharmaceutical Compositions [0183] Some embodiments described herein relates to a pharmaceutical composition,
Iiarmaceutically acceptable sait of the foregoing) and a pharmaceutically acceptable carrier, t iat can include an effective amount of one or more compounds described herein (e.g., a campound of Formula (I), a compound of Formula (Π) and/or a compound of Formula (III), or a P diluent, excipient or combination thereof.
[0184] The term “pharmaceuticat composition” refera to a mixture of one or more ci impounds disclosed herein with other chemical components, such as diluents or carriers. The p îarmaceutical composition facilitâtes administration of the compound to an organisai. P larmaceutical compositions can also be obtained by reacting compounds with inorganic or o ’ganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric aAid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, and salicylic acid. P larmaceutical compositions will generally be tailored to the spécifie intended route of administration.
[0185] The term “physiologically acceptable” defines a carrier, diluent or excipient th at does not abrogate the biological activity and properties of the compound.
[0186] As used herein, a carrier” refera to a compound that facilitâtes the incorporation of a compound into cells or tissues. For example, without limitation, dimethyl su lfoxïde (DMSO) is a commonly utilized carrier that facilitâtes the uptake of many organic cc mpounds into cells or tissues of a subject.
[0187] As used herein, a diluent” refera to an ingrédient in a pharmaceutical mposition that lacks pharmacological activity but may be pharmaceutically necessary or sirable. For exemple, a diluent may be used to increase the bulk of a potent drug whose mass too small for manufacture and/or administration. It may also be a liquid for the dissolution of a drug to be administered by injection, ingestion or inhalation. A common form of diluent in the ar : is a buffered aqueous solution such as, without limitation, phosphate buffered saline that mies the composition of human blood.
[0188] As used herein, an excipient” refera to an inert substance that is added to a pharmaceutical composition to provide, without limitation, bulk, consistency, stability, binding ab ility, lubrication, disintegrating ability etc., to the composition. A “diluent” is a type of ex ripient.
CCI de is mi [0189] The pharmaceutical compositions described herein can be administered to a îuman patient per se, or in pharmaceutical compositions where they are mixed wîth other active : ngredients, as in combination therapy, or carriers, diluents, excipients or combinations thereof.
’roper formulation is dépendent upon the route of administration chosen. Techniques for i brmulation and administration of the compounds described herein are known to those skilled in
the art.
[0190] The pharmaceutical compositions disclosed herein may be manufactured in a manner that is itself known, e.g., by means of conventional mixing, dissolving, granulating, ( ragee-making, levigating, emulsifying, encapsulating, entrapping or tableting processes. Additionally, the active ingrédients are contained in an amount effective to achieve its intended I urpose. Many of the compounds used in the pharmaceutical combinations disclosed herein may te provided as salts with pharmaceutically compatible counterions.
[0191] Multiple techniques of administering a compound exist in the art including, tut not limited to, oral, rectal, topical, aérosol, injection and parenterai delivery, including iitramuscular, subcutaneous, intravenous, intramedullary injections, intrathecal, direct iitraventricular, intraperitoneal, intranasal and intraocular injections.
[0192] One may also administer the compound in a locat rather than systemic manner, for example, via injection of the compound directly into the infected area, often în a depot or sustained release formulation. Furthermore, one may administer the compound in a targeted drug delivery system, for example, in a liposome coated with a tissue-specifïc antibody.
The liposomes will be targeted to and taken up selectively by the organ.
[0193] The compositions may, if desired, be presented in a pack or dispenser device v hich may contain one or more unit dosage forms containing the active ingrédient. The pack may for example comprise meta] or plastic foil, such as a blister pack The pack or dispenser device may be accompanied by instructions for administration. The pack or dispenser may also be accompanied with a notice associated with the container in form prescribed by a govcmmental agency regulatîng the manufacture, use, or sale of pharmaceuticals, which notice ii reflective of approvat by the agency of the form of the drug for human or veterinary administration. Such notice, for example, may be the labeling approved by the U.S. Food and drug Administration for prescription drugs, or the approved product insert. Compositions that cnn include a compound described herein formulated in a compatible pharmaceutical carrier ay also be prepared, placed in an appropriate container, and labeled for treatment of an ir dicated condition.
vfethods of Use:
[0194] Some embodiments described herein relate to a method of ameliorating, reating and/or preventing a viral infection selected from a paramyxovirus viral infection and an orthomyxovirus viral infection, which can include administering to a subject an effective ; mount of one or more compounds described herein, or a pharmaceutical composition that inciudes one or more compounds described herein (e.g., a compound of Formula (I), a compound of Formula (Π) and/or a compound of Formula (III), or a pharmaceutically acceptable f ait of the foregoing). In some embodiments, the subject is identified as suffering from the viral infection (for example, a paramyxovirus viral infection or an orthomyxovirus viral infection).
[0195] Other embodiments described herein relate to a method of inhiblting viral réplication of a virus selected from a paramyxovirus and an orthomyxovirus, which can include contacting a cell infected with the virus wîth an effective amount of a compound of Formula (I), or a pharmaceutically acceptable sait thereof, an effective amount of a compound of Formula (Π), or a pharmaceutically acceptable sait thereof, an effective amount of a compound of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a pharmaceutical composition that inciudes one or more compounds described herein (e.g., a compound of Formula (I), a compound of Formula (II) and/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing).
[0196] Other embodiments described herein relate to the use of an effective amount or a compound selected from Formula (I), Formula (Π), and Formula (IlI), or a pharmaceutically acceptable sait of the foregoing, for amelioring or treating a viral infection caused by a virus sllected from a henipavirus, a morbillivirus, a respirovirus, a rubulavirus and a metapneumovirus. Still other embodiments described herein relate to a method for ameliorating o ‘ treating a viral infection caused by a virus that can include administering to or contacting a c :11 in a subject with an effective amount of a compound selected from Formula (I), Formula (II), and Formula (III), or a pharmaceutically acceptable sait of the foregoing, wherein the virus c;m be selected from a henipavirus, a morbillivirus, a respirovirus, a rubulavirus and a n etapneumovirus. Yet still other embodiments described herein relate to using an effective amount of a compound selected from Formula (I), Formula (Π), and Formula (ΙΠ), or a pharmaceutically acceptable sait of the foregoing, for inhiblting réplication of a virus selected from a henipavirus, a morbillivirus, a respirovirus, a rubulavirus and a metapneumovirus (for ej ample by inhiblting the virus’ polymerase complex).
[0197] In some embodiments, an effective amount of one or more compounds of
Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula )0
Π), or a pharmaceutically acceptable sait thereof, an effective amount of one or more :ompounds of Formula (ΙΠ), or a pharmaceutically acceptable sait thereof, and/or a îharmaceutical composition that indudes one or more compounds described herein (e.g., a :ompound of Formula (I), a compound of Formula (H) and/or a compound of Formula (III), or a iharmaceutically acceptable sait of the foregoing) can be used to treat and/or ameliorate a i espiratory syncytial viral (RSV) infection. In some embodiments, an effective amount of one or more compounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula (II), or a pharmaceutically acceptable sait thereof, an effective amount of one or more compounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a pharmaceutical composition that indudes one or more compounds described herein (e.g., a compound of Formula (I) a compound of Formula (II) and/or a compound of Formula (ΠΙ), or a pharmaceutically acceptable sait of the foregoing) can be used to prevent a respiratory syncytial 5irai infection. In some embodiments, an effective amount of one or more compounds of
Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula ΠΙ), or a pharmaceutically acceptable sait thereof, an effective amount of one or more compounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a pharmaceutical composition that indudes one or more compounds described herein (e.g., a c ompound of Formula (I), a compound of Formula (II) and/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing) can be used to inhibit the réplication of a respiratory syncytial virus. In some embodiments, an effective amount of one or more compounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or more
Împounds of Formula (II), or a pharmaceutically acceptable sait thereof, an effective amount of e or more compounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a . armaceutical composition that indudes one or more compounds described herein (e.g., a compound of Formula (I), a compound of Formula (Π) and/or a compoundof Formula (III), or a p larmaceutically acceptable sait of the foregoing) can be used to inhibit the RSV polymerase
[0198] In other embodiments, an effective amount of one or more compounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula (II), or a pharmaceutically acceptable sait thereof, an effective amount of one or more c împounds of Formula (ΙΠ), or a pharmaceutically acceptable sait thereof, and/or a p îarmaceutical composition that indudes one or more compounds described herein (e.g., a compound of Formula (I) a compound of Formula (II) and/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing) can be used to treat and/or ameliorate an nfluenza viral infection. In other embodiments, an effective amount of one or more compounds i >f Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of : :ormula (Π), or a pharmaceutically acceptable sait thereof, an effective amount of one or more compounds of Formula (ΙΠ), or a pharmaceutically acceptable sait thereof, and/or a pharmaceutical composition that includes one or more compounds described herein (e.g., a compound of Formula (1), a compound of Formula (II) and/or a compound of Formula 011), or a I harmaccutically acceptable sait of the foregoing) can be used to prevent an influenza viral i ifection. In some embodiments, an effective amount of one or more compounds of Formula (1), cr a pharmaceutically acceptable sait thereof, one or more compounds of Formula 01), or a harmaccutically acceptable sait thereof, an effective amount of one or more compounds of
F
Formula (ΠΙ), or a pharmaceutically acceptable sait thereof, and/or a pharmaceutical c imposition that includes one or more compounds described herein (e.g., a compound of Formula 0), a compound of Formula (II) and/or a compound of Formula 011), or a p larmaccutically acceptable sait of the foregoing) can be used to inhibit the réplication of an influenza virus. In some embodiments, an effective amount of one or more compounds of F annula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula (Π), or a pharmaceutically acceptable sait thereof, an effective amount of one or more compounds of Formula 0Π), or a pharmaceutically acceptable sait thereof, and/or a P côi ph îarmaccutical composition that includes one or more compounds described herein (e.g., a mpound of Formula (I), a compound of Formula 01) and/or a compound of Formula 011), or a larmaccutically acceptable sait of the foregoing) can be used to inhibit the influenza pôlymerase complex.
[0199] In some embodiments, an effective amount of one or more compounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula 0), or a pharmaceutically acceptable sait thereof, an effective amount of one or more cempounds of Formula 0Π), or a pharmaceutically acceptable sait thereof, and/or a pharmaceutical composition that includes one or more compounds described herein (e.g., a compound of Formula 0), a compound of Formula (II) and/or a compound of Formula 011), or a pharmaccutically acceptable sait of the foregoing) can be used to treat and/or ameliorate a henipaviral infection. In some embodiments, an effective amount of one or more compounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula 01), or a pharmaceutically acceptable sait thereof, an effective amount of one or more co npounds of Formula (ΙΠ), or a pharmaceutically acceptable sait thereof, and/or a ph irmaceutical composition that includes one or more compounds described herein (e.g., a !0 :ompound of Formula (I) a compound of Formula (II) and/or a compound of Formula (III), or a jhannaceutically acceptable sait of the foregoing) can be used to prevent a henipavira! infection md/or nipahviral infection. In some embodiments, an effective amount of one or more compounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula (II), or a pharmaceutically acceptable sait thereof, an effective amount of one or more compounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a Îharmaceutical composition that inciudes one or more compounds described herein (e.g., a ompound of Formula (I), a compound of Formula (Π) and/or a compound of Formula (III), or a harmaceutically acceptable sait of the foregoing) can be used to inhibit the réplication of a 1 enipavirus. In some embodiments, an effective amount of one or more compounds of Formula ([), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula (II), or a pharmaceutically acceptable sait thereof, an effective amount of one or more compounds of Formula (ΠΙ), or a pharmaceutically acceptable sait thereof, and/or a pharmaceutical composition that inciudes one or more compounds described herein (e.g., a compound of Formula (I), a compound of Formula (II) and/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing) can be used to inhibit the henipavirus polymerase complex and/or nipahvirus polymerase complex.
[0200] In some embodiments, an effective amount of one or more compounds of ormula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula I), or a pharmaceutically acceptable sait thereof, an effective amount of one or more compounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a p îarmaceutical composition that inciudes one or more compounds described herein (e.g., a compound of Formula (I), a compound of Formula (Π) and/or a compound of Formula (III). or a pharmaceutically acceptable sait of the foregoing) can be used to treat and/or ameliorate a hendraviral infection and/or nipahviral infection. In some embodiments, an effective amount of one or more compounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula (II), or a pharmaceutically acceptable sait thereof, an effective amount of one or more compounds of Formula (ΙΠ), or a pharmaceutically acceptable sait thereof, and/or a pharmaceutical composition that inciudes one or more compounds described hèrein (e.g., a compound of Formula (I) a compound of Formula (II) and/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing) can be used to prevent a hendraviral infection and/or nipahviral infection. In some embodiments, an effective amount of or e or more compounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or m >re compounds of Formula (II), or a pharmaceutically acceptable sait thereof, an effective
JO amount of one or more compounds of Formula (ΙΠ), or a pharmaceutically acceptable sait hereof, and/or a pharmaceutical composition that includes one or more compounds described I îerein (e.g., a compound of Formula (I), a compound of Formula 01) and/or a compound of Î'ormula (III), or a pharmaceutically acceptable sait of the foregoing) can be used to inhibit the eplication of a hendravirus and/or nipahvirus. In some embodiments, an effective amount of ( ne or more compounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula (Π), or a pharmaceutically acceptable sait thereof, an effective î mount of one or more compounds of Formula (ΊΙΙ), or a pharmaceutically acceptable sait t îereof, and/or a pharmaceutical composition that includes one or more compounds described 1 erein (e.g., a compound of Formula (I), a compound of Formula (II) and/or a compound of Formula 011), or a pharmaceutically acceptable sait of the foregoing) can be used to inhibit the 1 endravirus polymerase complex and/or nipahvirus polymerase complex.
[0201] In some embodiments, an effective amount of one or more compounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula I), or a pharmaceutically acceptable sait thereof, an effective amount of one or more cbmpounds of Formula 0Π), or a pharmaceutically acceptable sait thereof, and/or a pharmaceutical composition that includes one or more compounds described herein (e.g., a ompound of Formula (I), a compound of Formula (II) and/or a compound of Formula 011), or a pharmaceutically acceptable sait of the foregoing) can be used to treat and/or ameliorate a orbilliviral infection. In some embodiments, an effective amount of one or more compounds of Formula 0), or a pharmaceutically acceptable sait thereof, one or more compounds of
Î
Formula (II), or a pharmaceutically acceptable sait thereof, an effective amount of one or more ompounds of Formula (ΙΠ), or a pharmaceutically acceptable sait thereof, and/or a p îarmaceutical composition that includes one or more compounds described herein (e.g., a compound of Formula (I) a compound of Formula (II) and/or a compound of Formula (III), or a p îarmaceutically acceptable sait of the foregoing) can be used to prevent morbilliviral infection. In some embodiments, an effective amount of one or more compounds of Formula (I), or a P pliarmaceutically acceptable sait thereof, an effective amount of one or more compounds of
Formula (III), or a pharmaceutically acceptable sait thereof, and/or a pharmaceutical composition that includes one or more compounds described herein (e.g., a compound of
Formula (I), a compound of Formula (II) and/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing) can be used to inhibit the réplication of a morbillivirus. In some embodiments, an effective amount of one or more compounds of îarmaceutically acceptable sait thereof, one or more compounds of Formula (Π), or a larmaceutically acceptable sait thereof, an effective amount of one or more compounds of
Pormula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula (Π), or a pharmaceutically acceptable sait thereof, an effective amount of one or more compounds of Formula (ΙΠ), or a pharmaceutically acceptable sait thereof, and/or a pharmaceutical composition that includes one or more compounds described herein (e.g., a c ompound of Formula (I), a compound of Formula (II) and/or a compound of Formula (III), or a I harmaceuticatly acceptable sait of the foregoing) can be used to inhibit the morbillivirus f olymerase complex.
[0202] In some embodiments, an effective amount of one or more compounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula I), or a pharmaceutically acceptable sait thereof, an effective amount of one or more compounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a pharmaceutical composition that includes one or more compounds described herein (e.g., a compound of Formula (I), a compound of Formula (II) and/or a compound of Formula (ΠΙ), or a pharmaceutically acceptable sait of the foregoing) can be used to treat and/or ameliorate measles. In some embodiments, an effective amount of one or more compounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula (II), or a P
Formula (III), or a pharmaceutically acceptable sait thereof, and/or a pharmaceutical composition that includes one or more compounds described herein (e.g., a compound of Formula (I) a compound of Formula (Π) and/or a compound of Formula (III), p larmaceutically acceptable sait of the foregoing) can be used to prevent measles. In embodiments, an effective amount of one or more compounds of Formula (I), pi P
Formula (ΠΙ), or a pharmaceutically acceptable sait thereof, and/or a pharmaceutical a imposition that includes one or more compounds described herein (e.g., a compound of Formula (I), a compound of Formula (II) and/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing) can be used to inhibit the réplication of a msasles virus. In some embodiments, an effective amount of one or more compounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula ), or a pharmaceutically acceptable sait thereof, an effective amount of one or more ccmpounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a pl armaceutical composition that includes one or more compounds described herein (e.g., a compound of Formula (I), a compound of Formula (II) and/or a compound of Formula (III), or a or a some or a larmaceutically acceptable sait thereof, one or more compounds of Formula (Π), îarmaceutically acceptable sait thereof, an effective amount of one or more compounds of or a pharmaceutically acceptable sait of the foregoing) can be used to inhibit the measles polymerase c omptex.
[0203] In some embodiments, an effective amount of one or more compounds of formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula , II), or a pharmaceutically acceptable sait thereof, an effective amount of one or more (I compounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a P c ompound of Formula (I), a compound of Formula (II) and/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing) can be used to treat and/or ameliorate a r ibutaviral infection. In some embodiments, an effective amount of one or more compounds of F ormula (I), or a pharmaceuticany acceptable sait thereof, one or more compounds of Formula I), or a pharmaceutically acceptable sait thereof, an effective amount of one or more harmaceutical composition that includes one or more compounds described herein (e.g., a
C compounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a p larmaceutical composition that includes one or more compounds described herein (e.g., a ci jmpound of Formula (I) a compound of Formula (II) and/or a compound of Formula (ΠΙ), or a p larmaceutically acceptable sait ofthe foregoing) can be used to prevent a rubulaviral infection. In some embodiments, an effective amount of one or more compounds of Formula (I), or a p îarmaceuticalty acceptable sait thereof, one or more compounds of Formula (Π), or a p îarmaceutically acceptable sait thereof, an effective amount of one or more compounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a pharmaceutical composition that includes one or more compounds described herein (e.g., a compound of Formula (I), a compound of Formula (II) and/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing) can be used to inhibit the réplication of a nbulavirus. In some embodiments, an effective amount of one or more compounds of Formula (l|, or a pharmaceutically acceptable sait thereof, one or more compounds of Formula (II), or a pharmaceutically acceptable sait thereof, an effective amount of one or more compounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a pharmaceutical composition that includes one or more compounds described herein (e.g., a compound of Formula (I), a compound of Formula (II) and/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing) can be used to inhibit the rubutavirus pclymerase complex.
[0204] In some embodiments, an effective amount of one or more compounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula (II), or a pharmaceutically acceptable sait thereof, an effective amount of one or more or a some or a or a larmaceutically acceptable sait of the foregoing) can be used to inhibit the réplication of a In some embodiments, an effective amount of one or more compounds of ormula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula I), or a pharmaceutically acceptable sait thereof, an effective amount of one or more :ompounds of Formula (ΙΠ), or a pharmaceutically acceptable sait thereof, and/or a jharmaceutical composition that includes one or more compounds described herein (e.g., a compound of Formula (I), a compound of Formula (Π) and/or a compound of Formula (III), or a ' iharmaceutically acceptable sait of the foregoing) can be used to treat and/or ameliorate mumps. 1 n some embodiments, an effective amount of one or more compounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula (II), or a pharmaceutically acceptable sait thereof, an effective amount of one or more compounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a pharmaceutical composition that includes one or more compounds described herein (e.g., a compound of Formula (I) a compound of Formula (II) and/or a compound of Formula (ΠΙ), pharmaceutically acceptable sait of the foregoing) can be used to prevent mumps. In embodiments, an effective amount of one or more compounds of Formula (I), pharmaceutically acceptable sait thereof, one or more compounds of Formula (Π), pharmaceutically acceptable sait thereof, an effective amount of one or more compounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a pharmaceutical camposition that includes one or more compounds described herein (e.g., a compound of Formula (I), a compound of Formula (II) and/or a compound of Formula (III), or a P mumps virus. F C compounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a p îarmaceutical composition that includes one or more compounds described herein (e.g., a compound of Formula (I), a compound of Formula (II) and/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing) can be used to inhibit the mumps polymerase complex.
[0205] In some embodiments, an effective amount of one or more compounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula (II), or a pharmaceutically acceptable sait thereof, an effective amount of one or more cjmpounds of Formula (ΙΠ), or a pharmaceutically acceptable sait thereof, and/or a pharmaceutical composition that includes one or more compounds described herein (e.g., a cc mpound of Formula (I), a compound of Formula (II) and/or a compound of Formula (III), or a pl armaceutically acceptable sait of the foregoing) can be used to treat and/or ameliorate a setidai viral infection. In some embodiments, an effective amount of one or more compounds of : formula (1), or a pharmaceuticalty acceptable sait thereof, one or more compounds of Formula ΪΠ), or a pharmaceutically acceptable sait thereof, an effective amount of one or more ompounds of Formula (ΙΠ), or a pharmaceutically acceptable sait thereof, and/or a harmaceutical composition that includes one or more compounds described herein (e.g., a compound of Formula (1) a compound of Formula (II) and/or a compound of Formula (ΠΙ), or a pharmaceutically acceptable sait of the foregoing) can be used to prevent a sendai viral i ifectïon. In some embodiments, an effective amount of one or more compounds of Formula (1), c r a pharmaceutically acceptable sait thereof, one or more compounds of Formula (II), or a p harmaceutically acceptable sait thereof, an effective amount of one or more compounds of Formula (ΠΙ), or a pharmaceutically acceptable sait thereof, and/or a pharmaceuticai composition that includes one or more compounds described herein (e.g., a compound of Formula (1), a compound of Formula (II) and/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing) can be used to inhibit the réplication of a s :ndai virus. In some embodiments, an effective amount of one or more compounds of Formula ( ), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula (II), or a P Formula (ΙΠ), or a pharmaceutically acceptable sait thereof, and/or a pharmaceuticai composition that includes one or more compounds described herein (e.g., a compound of F îarmaceuticalty acceptable sait thereof, an effective amount of one or more compounds of armula (1), a compound of Formula (II) and/or a compound of Formula (III), or a îarmaceutically acceptable sait of the foregoing) can be used to inhibit the sendai virus nlymerase complex.
[0206] In some embodiments, an effective amount of one or more compounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula (Π), or a pharmaceutically acceptable sait thereof, an effective amount of one or more compounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a p îarmaceutical composition that includes one or more compounds described herein (e.g., a compound of Formula (1), a compound of Formula (II) and/or a compound of Formula (III), or a plu re armaceutically acceptable sait of the foregoing) can be used to treat and/or ameliorate a spiroviral infection. In some embodiments, an effective amount of one or more compounds of
F >rmula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula ), or a pharmaceutically acceptable sait thereof, an effective amount of one or more compounds of Formula (ΙΠ), or a pharmaceutically acceptable sait thereof, and/or a pharmaceuticai composition that includes one or more compounds described herein (e.g., a ccmpound of Formula (I) a compound of Formula (II) and/or a compound of Formula (III), or a <0 pharmaceutically acceptable sait of the foregoing) can be used to prevent a respiroviral nfection. In some embodiments, an effective amount of one or more compounds of Formula (I), * )r a pharmaceutically acceptable sait thereof, one or more compounds of Formula (II), or a ] îharmaceutically acceptable sait thereof, an effective amount of one or more compounds of 1 formula (III), or a pharmaceutically acceptable sait thereoC and/or a pharmaceutical composition that includes one or more compounds described herein (e.g., a compound of Formula (I), a compound of Formula (II) and/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing) can be used to inhibit the réplication of a ijespirovirus. In some embodiments, an effective amount of one or more compounds of Formula Π), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula (II), or a j harmaceutically acceptable sait thereof, an effective amount of one or more compounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a pharmaceutical composition that includes one or more compounds described herein (e.g., a compound of formula (I), a compound of Formula (II) and/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing) can be used to inhibit the respirovirus polymerase complex.
[0207] In some embodiments, an effective amount of one or more compounds of
Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula (1 compounds of Formula (ΙΠ), or a pharmaceutically acceptable sait thereof, and/or a P c jmpound of Formula (I), a compound of Formula (Π) and/or a compound of Formula (III), or a P
I), or a pharmaceutically acceptable sait thereof, an effective amount of one or more larmaceutical composition that includes one or more compounds described herein (e.g., a îarmaceutically acceptable sait of the foregoing) can be used to treat and/or ameliorate a E PIV-I infection and/or HPIV-3 infection. In some embodiments, an effective amount of one o:1 more compounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula (II), or a pharmaceutically acceptable sait thereof, an effective amount of oi le or more compounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a pnarmaceutical composition that includes one or more compounds described herein (e.g., a compound of Formula (I) a compound of Formula (II) and/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing) can be used to prevent a HPIV-I infection ar d/or HPIV-3 infection. In some embodiments, an effective amount of one or more compounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula (II), or a pharmaceutically acceptable sait thereof, an effective amount of one or more compounds of Formula (ΙΠ), or a pharmaceutically acceptable sait thereof, and/or a
pharmaceutical composition that includes one or more compounds described herein (e.g., a compound of Formula (I), a compound of Formula (II) and/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing) can be used to inhibit the réplication of 1PIV-1 and/or HPIV-3. In some embodiments, an effective amount of one or more compounds )f Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of 'ormula (II), or a pharmaceutically acceptable sait thereof, an effective amount of one or more compounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a pharmaceutical composition that includes one or more compounds described herein (e.g., a < ompound of Formula (I), a compound of Formula (Π) and/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing) can be used to inhibit the HPIV-1 polymerase complex and/or HPIV-3 polymerase complex.
[0208] In some embodiments, an effective amount of one or more compounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula ΠΙ), or a pharmaceutically acceptable sait thereof, an effective amount of one or more compounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a Jharmaceutical composition that includes one or more compounds described herein (e.g., a compound of Formula (I), a compound of Formula (II) and/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing) can be used to treat and/or ameliorate a F PIV-2 infection and/or HPIV-4 infection. In some embodiments, an effective amount of one or more compounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula (II), or a pharmaceutically acceptable sait thereof, an effective amount of o le or more compounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a p larmaceutica! composition that includes one or more compounds described herein (e.g., a compound of Formula (I) a compound of Formula (II) and/or a compound of Formula (ΠΙ), or a p harmaceutically acceptable sait of the foregoing) can be used to prevent a HPIV-2 infection and/or HPIV-4 infection. In some embodiments, an effective amount of one or more compounds o ' Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula (Π), or a pharmaceutically acceptable sait thereof, an effective amount of one or more compounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a pharmaceutical composition that includes one or more compounds described herein (e.g., a compound of Formula (I), a compound of Formula (II) and/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing) can be used to inhibit the réplication of HPIV-2 and/or HPIV-4. In some embodiments, an effective amount of one or more compounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of ’ormula (Π), or a pharmaceutically acceptable sait thereof, an effective amount of one or more :ompounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a pharmaceutical composition that includes one or more compounds described herein (e.g., a compound of Formula (I), a compound of Formula (Π) and/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing) can be used to inhibit the HPIV-2 polymerase complex and/or HPIV-4 polymerase complex, [0209] In some embodiments, an effective amount of one or more compounds of ] formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula Π), or a pharmaceutically acceptable sait thereof, an effective amount of one or more compounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a pharmaceutical composition that includes one or more compounds described herein (e.g., a tSompound of Formula (I), a compound of Formula (II) and/or a compound of Formula (III), or a harmaceutically acceptable sait of the foregoing) can be used to treat and/or ameliorate a îtapncumoviral infection. In some embodiments, an effective amount of one or more mpounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or more mpounds of Formula (II), or a pharmaceutically acceptable sait thereof, an effective amount of c ne or more compounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a harmaceutical composition that includes one or more compounds described herein (e.g., a
RI compound of Formula (I) a compound of Formula (II) and/or a compound of Formula (III), or a F infection. In some embodiments, an effective amount of one or more compounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula (II), or a pharmaceutically acceptable sait thereof, an effective amount of one or more compounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a pharmaceutical composition that includes one or more compounds described herein (e.g., a compound of Formula (I), a compound of Formula (II) and/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing) can be used to inhibit the réplication of a metapneumovirus. In some embodiments, an effective amount of one or more compounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula (II), or a pharmaceutically acceptable sait thereof, an effective amount of one or more compounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a p larmaceutical composition that includes one or more compounds described herein (e.g., a compound of Formula (I), a compound of Formula (II) and/or a compound of Formula (III), or a P harmaceutically acceptable sait of the foregoing) can be used to prevent a metapneumoviral larmaceutically acceptable sait of the foregoing) can be used to inhibit the metapneumovirus polymerase complex. In some embodiments, including those of this paragraphe, the metapneumovirus can be a human metapneumovirus.
[0210] In some embodiments, an effective amount of one or more compounds of Ïormula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula I), or a pharmaceutically acceptable sait thereof, an effective amount of one or more compounds of Formula (ΙΠ), or a pharmaceutically acceptable sait thereof, and/or a pharmaceutical composition that includes one or more compounds described herein (e.g., a compound of Formula (I), a compound of Formula (II) and/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing) can be used treat and/or ameliorate an upper respiratory viral infection caused by a virus selected from a henipavirus, a morbillivirus, a r espirovirus, a rubulavirus, a pneumovirus, a metapneumovirus and influenza virus. In some embodiments, an effective amount of one or more compounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula (Π), or a pharmaceutically acceptable sait thereof, an effective amount of one or more compounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a pharmaceutical composition that includes one or more compounds described herein (e.g., a compound of Formula (I), a compound of Formula (II) and/or a compound of Formula (III), or a p larmaccutically acceptable sait of the foregoing) can be used treat and/or ameliorate a lower respiratory viral infection caused by a virus selected from a henipavirus, a morbillivirus, a respirovirus, a rubulavirus, a pneumovirus, a metapneumovirus and influenza virus. In some cnbodiments, an effective amount of one or more compounds of Formula (I), or a p larmaccutically acceptable sait thereof, one or more compounds of Formula (Π), or a p îarmaceutically acceptable sait thereof, an effective amount of one or more compounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a pharmaceutical composition that includes one or more compounds described herein (e.g., a compound of F p îarmaceutically acceptable sait of the foregoing) can be used treat and/or ameliorate one or rr irmula (I) a compound of Formula (II) and/or a compound of Formula (ΠΙ), or a ore symptoms of an infection caused by a virus selected from a henipavirus, a morbillivirus, a spirovirus, a rubulavirus, a pneumovirus, a metapneumovirus and influenza virus (such as ose described herein).
[0211] In some embodiments, an effective amount of one or more compounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula or a pharmaceutically acceptable sait thereof, an effective amount of one or more et mpounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a re th pharmaceutical composition that includes one or more compounds described herein (e.g., a i :ompound of Formula (I), a compound of Formula (Π) and/or a compound of Formula (III), or a ] (harmaceutically acceptable sait of the foregoing) can be used treat and/or ameliorate an upper respiratory viral infection caused by RSV infection, meastes, mumps, parainfluenza infection, i netapneumovirus and/or influenza infection. In some embodiments, an effective amount of one < r more compounds of Formula (I), or a pharmaceuticatty acceptable sait thereof, one or more (ompounds of Formula (II), or a pharmaceutically acceptable sait thereof, an effective amount of c ne or more compounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a I harmaceutical composition that includes one or more compounds described herein (e.g., a compound of Formula (I), a compound of Formula (II) and/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing) can be used treat and/or ameliorate a lower r ïspiratory viral infection caused by RSV infection, measles, mumps, parainfluenza infection, n letapneumovirus and/or influenza infection. In some embodiments, an effective amount of one or more compounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or more c ampounds of Formula (II), or a pharmaceutically acceptable sait thereof, an effective amount of o ne or more compounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a p iarmaceuticat composition that includes one or more compounds described herein (e.g., a c ampound of Formula (I) a compound of Formula (II) and/or a compound of Formula (ΠΙ), or a p larmaccuticalty acceptable sait of the foregoing) can be used treat and/or ameliorate one or nore symptoms of an infection caused by RSV infection, measles, mumps, parainfluenza infection, metapneumovirus and/or influenza infection (such as those described herein).
[0212] In some embodiments, an effective amount of one or more compounds of F jrmula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula Î), or a pharmaceutically acceptable sait thereof, an effective amount of one or more mpounds of Formula (ΙΠ), or a pharmaceutically acceptable sait thereof, and/or a piarmaceutical composition that includes one or more compounds described herein (e.g., a compound of Formula (I), a compound of Formula (II) and/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing) can be used treat and/or ameliorate bronchiolitis and/or tracheobronchitîs due to a RSV infection, influenza infection and/or human parainfluenza virus 3 (HPIV-3) infection. In some embodiments, an effective amount of one or more compounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or more cc mpounds of Formula (II), or a pharmaceutically acceptable sait thereof, an effective amount of oie or more compounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a piarmaceutical composition that includes one or more compounds described herein (e.g., a ίο;
h îompound of Formula (I), a compound of Formula (Π) and/or a compound of Formula (III), or a jharmaceutically acceptable sait of the foregoing) can be used treat and/or ameliorate mcumonia due to a RSV infection, influenza infection and/or human parainfluenza virus 3 ΉΡΓν-3) infection. In some embodiments, an effective amount of one or more compounds of :ormula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula Π), or a pharmaceutically acceptable sait thereof, an effective amount of one or more Îompounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a harmaceutical composition that includes one or more compounds described herein (e.g., a compound of Formula (I), a compound of Formula (II) and/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing) can be used treat and/or ameliorate croup due to a RSV infection, influenza infection and/or human parainfluenza virus 1 (HPIV-I) infection.
[0213] In some embodiments, an effective amount of one or more compounds of Îormula (1), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula
d), or a pharmaceutically acceptable sait thereof, an effective amount of one or more ompounds of Formula (ΙΠ), or a pharmaceutically acceptable sait thereof, and/or a harmaceutical composition that includes one or more compounds described herein (e.g., a compound of Formula (I), a compound of Formula (Π) and/or a compound of Formula 011), or a pharmaceutically acceptable sait of the foregoing) can be used treat and/or ameliorate a fever, caugh, runny nose, red eyes, a generalized rash, pneumonia, an ear infection and/or bronchitis due to measles. In some embodiments, an effective amount of one or more compounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula ( I), or a pharmaceutically acceptable sait thereof, an effective amount of one or more cjmpounds of Formula (ΙΠ), or a pharmaceutically acceptable sait thereof, and/or a p larmaceutical composition that includes one or more compounds described herein (e.g., a compound of Formula (I), a compound of Formula (Π) and/or a compound of Formula (III), or a P o îarmaceutically acceptable sait of the foregoing) can be used treat and/or ameliorate swellîng 7the satîvary glands, fever, loss of appetite and/or fatigue due to mumps.
[0214] In some embodiments, an effective amount of one or more compounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula (u), or a pharmaceutically acceptable sait thereof, an effective amount of one or more compounds of Formula (III), or a pharmaceutically acceptable sait thereof, and/or a pharmaceutical composition that includes one or more compounds described herein (e.g., a compound of Formula (I), a compound of Formula (Π) and/or a compound of Formula (III), or a P· armaceutîcally acceptable sait of the foregoing) can be used to prevent an influenza viral nfection. In some embodiments, the influenza viral infection can be an influenza A viral nfection. In other embodiments, the influenza viral infection can be an influenza B viral infection. In still other embodiments, the influenza viral infection can be an influenza C viral i nfection. In some embodiments, one or more compounds of Formula (I), or a pharmaceutically î cceptable sait thereof, one or more compounds of Formula (II), or a pharmaceutically i cceptable sait thereof, and/or one or more compounds of Formula (III), or a pharmaceutically t cceptable sait thereof, can be used to treat and/or ameliorate one or more subtypes of influenza. For example, one or more compounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula (Π), or a pharmaceutically acceptable sait thereof.
Fi ( armaceutically acceptable sait of the foregoing) can be used to prevent a human parainfluenza lereof, one or more compounds of Formula (Π), or a pharmaceutically acceptable sait thereof, nd/or one or more compounds of Formula (III), or a pharmaceutically acceptable sait thereof, cian be used to treat H1N1 and/or H3N2.
[0215] In some embodiments, an effective amount of one or more compounds of ormula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula il), or a pharmaceutically acceptable sait thereof, an effective amount of one or more compounds of Formula (ΙΠ), or a pharmaceutically acceptable sait thereof, and/or a pharmaceutical composition that includes one or more compounds described herein (e.g., a compound of Formula (I), a compound of Formula (II) and/or a compound of Formula (III), or a ph;
viral infection. In some embodiments, the human parainfluenza viral infection can be a human P cm be a human parainfluenza virus 2 (HPIV-2).
P e nbodiments, the human parainfluenza viral infection can be a human parainfluenza virus 4 (HPIV-4). In some embodiments, one or more compounds of Formula (I), or a pharmaceutically acceptable sait thereof, one or more compounds of Formula (II), or a pharmaceutically acceptable sait thereof, and/or one or more compounds of Formula (ΠΙ), or a pharmaceutically acceptable sait thereof, can be used to treat and/or ameliorate one or more subtypes of human parainfluenza virus. For example, one or more compounds of Formula (I), or a pharmaceutically irainflucnza virus I (HPIV-1). In other embodiments, the human parainfluenza viral infection In other embodiments, the human arainflucnza viral infection can be a human parainfluenza virus 3 (HPIV-3). In other sa th acceptable sait thereof, one or more compounds of Formula (II), or a pharmaceutically acceptable sait thereof, and/or one or more compounds of Formula (III), or a pharmaceutically acceptable sait thereof, can be used to treat HPIV-1 and/or HPIV-3.
[0216] The one or more compounds of Formula (I) or a pharmaceutically acceptable lt thereof, one or more compounds of Formula (II), or a pharmaceutically acceptable sait ereof, and/or one or more compounds of Formula (ΙΠ), or a pharmaceutically acceptable sait
1UU thereof, that can be used to treat, ameliorate and/or prevent a paramyxovirus and/or or an irthomyxovirus vira! infection can be a compound of Formula (I), or pharmaceutically icceptable sait thereof, and/or a compound of Formula (Π), or a pharmaceutically acceptable sait hereof, and/or a compound of Formula (ΠΙ), or a pharmaceutically acceptable sait thereof, irovided in any of the embodiments described in paragraphs [0084]-[0l70J.
[0217] As used herein, the terms “prevent” and “preventing,” mean a subject does not develop an infection because the subject has an immunity against the infection, or if a iubject becomes infected, the severity of the disease is less compared to the severity of the disease if the subject has not been administered/received the compound. Examples of forms of Prévention include prophylactic administration to a subject who has been or may be exposed to m infectious agent, such as a paramyxovirus (e.g., RSV) and/or an orthomyxovirus (e.g., i ifluenza).
[0218] As used herein, the terms “treat,” “treating,” “treatment,” “therapeutic,” and therapy do not necessarily mean total cure or abolition of the disease or condition. Any lleviation of any undesired signs or symptoms of a disease or condition, to any extent can be considered treatment and/or therapy. Furthermore, treatment may include acts that may worsen îe subject’s overall feeling of well-being or appearance.
[0219] The terms “therapeutically effective amount” and “effective amount are used indicate an amount of an active compound, or pharmaceutical agent, that elicits the biologica! médicinal response indicated. For example, a therapeutically effective amount of compound c m be the amount needed to prevent, alleviate or ameliorate symptoms of disease or prolong the s irvival ofthe subject being treated This response may occur in a tissue, System, animal or himan and includes aliénation of the signs or symptoms of the disease being treated. E eterminatîon of an effective amount is well within the capability of those skilled in the art, in v .ew of the disciosure provided herein. The therapeutically effective amount of the compounds dsclosed herein required as a dose will dépend on the route of administration, the type of a limai, including human, being treated, and the physical characteristics of the spécifie animal u ider considération. The dose can be tailored to achieve a desired effect, but will dépend on such factors as weight, diet, concurrent médication and other factors which those skilled in the m edical arts will recognize.
[0220] Various indicators for determining the effectiveness of a method for treating a viral infection, such as a paramyxovirus and/or an orthomyxovirus infection, are known to those skilled in the art. Example of suitable indicators include, but are not limited to, a réduction in vira! load, a réduction in viral réplication, a réduction in time to séroconversion (virus
101 i indétectable in patient sérum), a réduction of morbidity or mortality in clinical outcomes, and/or other indicator of disease response.
[0221] In some embodiments, an effective amount of a compound of Formulae (I), < Π) and/or (III), or a pharmaceutically acceptable sait of the foregoing, is an amount that is < ffective to reduce viral titers to undetectable levels, for example, to about 1000 to about 5000, ta about 500 to about 1000, or to about 100 to about 500 genome copies/mL sérum. In some
3mbodiments, an effective amount of a compound of Formulae (I), (II) and/or (III), or a harmaceutically acceptable sait of the foregoing, is an amount that is effective to reduce viral load compared to the viral load before administration of the compound of Formulae (I) ,(II) and/or (III)» or a pharmaceutically acceptable sait of the foregoing. For example, wherein the viral load is measure before administration of the compound of Formulae (I), (Π) and/or (III), or pharmaceutically acceptable sait of the foregoing, and again after completion of the treatment régime with the compound of Formulae (I), (II) and/or (ΙΠ), or a pharmaceutically acceptable s ilt of the foregoing (for exampie, 1 week after completion). In some embodiments, an effective anount of a compound of Formulae (I), (II) and/or (ΙΠ), or a pharmaceutically acceptable sait of the foregoing, can be an amount that is effective to reduce viral load to lower than about 100 genome copies/mL sérum. In some embodiments, an effective amount of a compound of Formulae (I), (II) and/or (III), or a pharmaceutically acceptable sait of the foregoing, is an a; nount that is effective to achieve a réduction in viral titer in the sérum of the subject in the rtnge of about 1.5-log to about a 2.5-log réduction, about a 3-log to about a 4-log réduction, or a greater than about 5-log réduction compared to the viral load before administration of the compound of Formulae (I), (II) and/or (III), or a pharmaceutically acceptable sait of the foregoing. For example, wherein the viral load îs measure before administration of the compound of Formulae (I), (Π) and/or (110, or a pharmaceutically acceptable sait of the foregoing, and again after completion of the treatment régime with the compound of Formulae (I), (Π) and/or (III), or a pharmaceutically acceptable sait of the foregoing (for example, 1 week after completion).
[0222] In some embodiments, a compound of Formulae (I), (II) and/or (III), or a pharmaceutically acceptable sait of the foregoing, can resuit in at least a 1,2,3,4,5, 10, 15,20, 2l·, 50, 75, 100-fold or more réduction in the réplication of a paramyxovirus and/or an orthomyxovirus relative to pre-treatment levels in a subject, as determined after completion of the treatment régime (for example, I week after completion).
cc fo
In some embodiments, a mpound of Formulae (I), (II) and/or (III), or a pharmaceutically acceptable sait of the regoing, can resuit in a réduction of the réplication of a paramyxovirus and/or an
102
Ï5 orthomyxovirus relative to pre-treatment Ievels in the range of about 2 to about 5 fold, about 10 to about 20 fold, about 15 to about 40 fold, or about 50 to about 100 fold. In some embodiments, a compound of Formulae (I), (II) and/or (ΠΙ), or a pharmaceutically acceptable i ait of the foregoing, can resuit in a réduction of paramyxovirus réplication in the range of 1 to : .5 log, 1.5 log to 2 log, 2 log to 2.5 log, 2.5 to 3 log, 3 log to 3.5 log or 3.5 to 4 log more réduction of paramyxovînis réplication compared to the réduction of paramyxovirus réduction ε chieved by ribavirin (Virazole®), or may achieve the same réduction as that of ribavirin (Virazole®) therapy in a shorter period of time, for example, in one week, two weeks, one r lonth, two months, or three months, as compared to the réduction achieved after six months of ribavirin (Virazole®) therapy. In some embodiments, a compound of Formulae (I), (II) and/or
III), or a pharmaceutically acceptable sait of the foregoing, can resuit in a réduction of rthomyxovirus réplication in the range of 1 to 1.5 log, 1.5 log to 2 log, 2 log to 2.5 log, 2.5 to 3 (
ci bg, 3 log to 3.5 log or 3.5 to 4 log more réduction of orthomyxovirus réplication compared to tie réduction of orthomyxovirus réduction achieved by oseltamivir (Tamiflu®), or may achieve t îe same réduction as that of oseltamivir (Tamiflu®) therapy in a shorter period of time, for example, in one week, two weeks, one month, two months, or three months, as compared to the r :duction achieved after six months of oseltamivir (Tamiflu®) therapy.
[0223] In some embodiments, an effective amount of a compound of Formula (I), a c jmpound of Formula (II) and/or a compound of Formula (III), or a pharmaceutically acceptable s lit of the foregoing, is an amount that is effective to achieve a sustained viral response, for e «ample, non-detectable or substantially non-detectable paramyxovirus and/or orthomyxovirus P NA (e.g., less than about 500, less than about 400, less than about 200, or less than about 100 genome copies per milliliter sérum) is found in the subject’s sérum for a period of at least about oLe week, two weeks, one month, at least about two months, at least about three months, at least a xjut four months, at least about five months, or at least about six months following cessation of therapy.
[0224] After a period of time, infectious agents can develop résistance to one or n ore therapeutic agents. The term “résistance” as used herein refers to a viral strain dispiaying delayed, lessened and/or null response to a therapeutic agentfs). For example, after treatment vJith an antiviral agent, the viral load of a subject infected with a résistant virus may be reduced t< a lesser degree compared to the amount in viral load réduction exhibited by a subject infected vJith a non-resistant strain. In some embodiments, a compound of Formula (I), a compound of Formula (Π) and/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing, can be administered to a subject infected with RSV that is résistant to one or more
103 different anti-RSV agents (for example, ribavirin). In some embodiments, development of résistant RSV straîns can be detayed when subjects are treated with a compound of Formula (I), o compound of Formula (II) and/or a compound of Formula (III), or a phannaceutically acceptable sait of the foregoing, compared to the development of RSV straîns résistant to other RSV drugs. In some embodiments, a compound of Formula 0), a compound of Formula (II) and/or a compound of Formula (ΙΠ), or a pharmaceutically acceptable sait of the foregoing, can be administered to a subject infected with an influenza virus that is résistant to one or more ( ifferent anti-influenza agents (for exampte, amantadine and rimantadine). In some embodiments, development of résistant influenza straîns can be delayed when subjects are treated with a compound of Formula (I), a compound of Formula (Π) and/or a compound of Formula (ΠΙ), or a phannaceutically acceptable sait of the foregoing, compared to the t cvelopment of influenza straîns résistant to other influenza drugs.
[0225] In some embodiments, a compound of Formula (I), a compound of Formula il) and/or a compound of Formula (III), or a phannaceutically acceptable sait of the foregoing, can decrease the percentage of subjects that expérience complications from a RSV viral infection compared to the percentage of subjects that expérience complication being treated with r bavirin. In some embodiments, a compound of Formula (I), a compound of Formula (II) id/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing, can Ixrease the percentage of subjects that expérience complications from an influenza viral a
d infection compared to the percentage of subjects that expérience complication being treated with i
Jeltamivir. For example, the percentage of subjects being treated with a compound of Formula , a compound of Formula (II) and/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing, that expérience complications can be 10% , 25%, 40%, 50%, 6 )%, 70%, 80% and 90% less compared to subjects being treated with ribavirin or oseltamivir.
[0226] In some embodiments, a compound of Formula (I), a compound of Formula (Π) and/or a compound of Formula (ΙΠ), or a pharmaceutically acceptable sait of the foregoing, or a pharmaceutical composition that includes a compound described herein, can be used in combination with one or more additional agent(s). In some embodiments, a compound of Formula Q), a compound of Formula (II) and/or a compound of Formula (III), or a
Î0
h) pharmaceutically acceptable sait of the foregoing, can be used in combination with one or more agents currently used for treating RSV. For example, the additional agent can be ribavirin, pi .livizumab and RSV-IGIV. For the treatment of RSV, additional agents include but are not limited to ALN-RSVOI (Alnylam Pharmaceutlcals), BMS-433771 (l-cyclopropyl-3-[[l-(4droxybuty!)benzimidazol-2-yl]methy!]imidazo[4,5-c]pyridin-2-one), RFI-64 ! ((4,4-bis-{4,6104 )is-[3-(bis-carbamoylmethyl-sulfamoyl)-phenylamino]-(l ,3,5)triazin-2-ylamino) -btphenyl-2,2iisulfonic-acid)), RSV604 ((S)-l-(2-fluorophenyl)-3-(2-oxo-5-phenyl-2,3-dihydro-lH)enzo[e][l,4]di-azepin-3-yl)-urea), MDT-637 ((4Z)-2-methylsulfanyl-4-[(E)-3-thiophen-271prop-2-enylidene]-l,3-thiazol-5-one), BTA988I, TMC-353121 (Tibotec), MBX-300, YM‘ Î3403 (N-cyclopropyl-6-[4-[(2-phenylbenzoyl)amino]benzoyl]-4,5-dihydrothieno[3,2-
d][l]benzazepine-2-carboxamide), motavizumab (Medi-524, Medlmmune), Medi-559, Medih34, Medi-557, RV568 and a RSV-F Particle Vaccine (Novavax). In some embodiments, a compound of Formula 0), a compound of Formula 01) and/or a compound of Formula 011), or a pharmaceutically acceptable sait of the foregoing, can be used in combination with one or more agents currently used for treating influenza. For example, the additional agent can be amantadine, rimantadinc, zanamivir and oseltamivir. For the treatment of influenza, additional agents include but are not limited to peramivir ((IS,2S,3S,4R)-3-[(lS)-I-acetamido-2ethylbutyl]-4-(diaminomethylideneamino)-2-hydroxycyclopentane-l-carboxylic acid), l ininamivir ((4S,5R,6R)-5-acetamido-4-carbamimidamido-6-[( 1 R,2R)-3-hydroxy-2Îethoxypropyl]-5,6-dihydro-4H-pyran-2-carboxylic acid), favipiravir (T-705, 6-fluoro-3'droxy-2-pyrazinccarboxamide), fludasc (DAS181, NexBio), ADS-8902 (Adamas Pharmaccuticals), IFN-b (Synairgen), beraprost (4-[2-hydroxy-l-[(E)-3-hydroxy-4-methyloct-le and VGX-3400X 0novio).
i-6-ynyl]-2,3,3a,8b-tetrahydro-lH-cyclopenta[b][l]bcnzofiiran-5-yl]butanoic acid), Neugene® [0227] In some embodiments, a compound of Formula 0), a compound of Formula (II) and/or a compound of Formula (ΙΠ), or a pharmaceutically acceptable sait ofthe foregoing, can be administered with one or more additional agent(s) together in a single pharmaceutical imposition. In some embodiments, a compound of Formula (I), a compound of Formula (II) d/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing, can be administered with one or more additional agent(s) as two or more separate pharmaceutical compositions. For example, a compound of Formula (I), a compound of Formula (II) and/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing, can be at ministered in one pharmaceutical composition, and at least one of the additional agents can be ac ministered in a second pharmaceutical composition. If there are at least two additional agents, one or more of the additional agents can be in a first pharmaceutical composition that includes a cc mpound of Formula (I), a compound of Formula (II) and/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing, and at least one of the other additional agent(s) can be in a second pharmaceutical composition.
105 [0228] The order of administration of a compound of Formula (I), a compound of : 7ormula (Π) and/or a compound of Formula (ΠΙ), or a pharmaceutically acceptable sait of the : oregoing, with one or more additional agent(s) can vaiy. In some embodiments, a compound of Formula (I), a compound of Formula (II) and/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing, can be administered prior to all additional agents. In other embodiments, a compound of Formula (I), a compound ofFormula (II) and/or a compound of Formula (III), or a pharmaceutically acceptable sait of the foregoing, can be
i.dministered prior to at least one additional agent. In still other embodiments, a compound of Îôrmula (I), a compound of Formula (II) and/or a compound of Formula (III), or a harmaceutically acceptable sait of the foregoing, can be administered concomitantly with one < ir more additional agent(s). In yet still other embodiments, a compound of Formula 0), a < ompound of Formula (II) and/or a compound of Formula (III), or a pharmaceutically acceptable fait of the foregoing, can be administered subséquent to the administration of at least one î dditional agent. In some embodiments, a compound of Formula (I), a compound of Formula SU) and/or a compound of Formula 0Π), or a pharmaceutically acceptable sait of the foregoing, an be administered subséquent to the administration of all additional agents.
[0229] A potential advantage of utilizing a compound of Formula 0), a compound of Formula (H) and/or a compound of Formula (ΠΙ), or a pharmaceutically acceptable sait of the f oregoing, in combination with one or more additional agent(s) described in paragraph [0226], i ictuding pharmaceutically acceptable salts and prodrugs thereof, may be a réduction in the r:quired amount(s) of one or more compounds of paragraph [0226] (including pharmaceutically acceptable salts and prodrugs thereof) that is effective in treating a disease condition disclosed herein (for example, RSV and/or influenza), as compared to the amount required to achieve sime therapeutic resuit when one or more compounds described in paragraph [0226], including pharmaceutically acceptable salts and prodrugs thereof, are administered without a compound of Formula 0), a compound of Formula 01) and/or a compound of Formula 011), or a pharmaceutically acceptable sait the foregoing. For example, the amount of a compound Jcscribed in paragraph [0226], including a pharmaceutically acceptable sait and prodrug thereof, c m be less compared to the amount of the compound described in paragraph [0226], including a pharmaceutically acceptable sait and prodrug thereof, needed to achieve the same viral load réduction when administered c jmpound of Formula (I), a compound of Formula (II) and/or a compound of Formula 011), or a p larmaceutically acceptable sait of the foregoing, in combination with one or more additional a’ent(s) described in paragraph [0226], including pharmaceutically acceptable salts and as a monotherapy. Another potentiel advantage of utilizing a
106 prodrugs thereof, is that the use of two or more compounds having different mechanism of actions can create a higher barrier to the development of résistant viral strains compared to the b amer when a compound is administered as monotherapy.
[0230] Additional advantages of utilizing a compound of Formula (I), a compound of Formula (II) and/or a compound of Formula (III), or a pharmaceutically acceptable sait the foregoing, in combination with one or more additional agent(s) described in paragraph [0226], including pharmaceutically acceptable salts and prodrugs thereof, may include little to no cross résistance between a compound of Formula (I), a compound of Formula (II) and/or a compound < f Formula (ΙΙΓ), or a pharmaceutically acceptable sait the foregoing, and one or more additional tgent(s) described in paragraph [0226] (including pharmaceutically acceptable salts and prodrugs thereof); different routes for élimination of a compound of Formula (I), a compound of Formula (II) and/or a compound of Formula (III), or a pharmaceutically acceptable sait the foregoing, and one or more additional agent(s) described in paragraph [0226] (including pharmaceutically acceptable salts and prodrugs thereof); little to no overlapping toxicities Ïetwcen a compound of Formula (I), a compound of Formula (II) and/or a compound of ormula (III), or a pharmaceutically acceptable sait the foregoing, and one or more additional agents) described in paragraph [0226] (including pharmaceutically acceptable salts and prodrugs thereof); little to no significant effects on cytochrome P450; and/or little to no pharmacokinetic interactions between a compound of Formula (I), or a pharmaceutically Jcceptable sait thereof, and one or more additional agent(s) described in paragraph [0226] (including pharmaceutically acceptable salts and prodrugs thereof).
[0231] Some embodiments described herein relate to the use of an effective amount of a compound, or a pharmaceutically acceptable sait thereof, for amelioring or treating a viral infection caused by a virus selected from a henipavirus, a morbillivirus, a respirovirus, a ribulavirus and a metapneumovirus, wherein the compound can be selected from:
^yNH2
-10617135
ΊΙΠ
OH , - ï-°->v^o
OH OHN/VJ-OH h<5 . Other embodiments desecribed herein related to a for ameliorating or treating a viral infection caused by a virus that can include i nethod ; idministering to or contacting a cell in a subject with an effective amount of a compound, or a ] iharmaceutically acceptable sait of thereof, wherein the virus is selected from a henipavirus, a moibillivirus, a respirovtrus, a rubulavirus and a metapneumovirus, and wherein the compound
HO-P-O-P-O OH OH
Still other embodiments described herein relate to using f n effective amount of a compound, or a pharmaceutically acceptable sait thereof, for inhibiting tie réplication of a virus selected from a henipavirus, a morbillivirus, a respirovirus, a rubulavirus and a metapneumovirus (for example, by inhiting the polymerase complex of the selected
ΙΟ-ρ-Ο-Ρ-Ο-Ρ-Ο compound can be selected from:
virus), wherein the
HO' nh2
»
108
40-1 and nh2 g g g O (-o-f-o-f’-o -VAX
OH OH OH N3 °
HO' >
embodiments of this paragraph, the virus can be a henïpavirus (for example, a nipahvirus). In other embodiments of this paragraph, the virus can be a morbillivirus, such as mcasles virus. In itill other embodiments of this paragraph, the virus can be a respirovirus. In some embodiments, the respirovirus can be human paraînfluenza virus 1. In other embodiments, the respirovirus can be human paraînfluenza virus 3. In yet still other embodiments of this paragraph, the virus can be a rubulavirus. . In some embodiments, the rubulavirus can be human paraînfluenza virus 2. . In other embodiments, the rubulavirus can be human paraînfluenza virus 4. . In still other embodiments, the rubulavirus can be the mumps virus. In some embodiments of this paragraph, the virus can be a metapneumovirus, such as the human r letapneumovirus.
[0232] As wili be readily apparent to one skilled in the art, the useful in vivo dosage t ) be administered and the particular mode of administration wili vary depending upon the âge, weight, the severity of the affliction, and mammalian species treated, the particular compounds employed, and the spécifie use for which these compounds are employed. The détermination of effective dosage levels, that is the dosage levels necessary to achieve the desired resuit, can be accompilshed by one skilled in the art using routine methods, for exampie, human clinical trials and in vitro studies.
[0233] The dosage may range broadly, depending upon the desired effects and the tnerapcutic indication. Altematively dosages may be based and calculated upon the surface area of the patient, as understood by those of ski IL in the art. Although the exact dosage wili be determined on a drug-by-drug basis, in most cases, some generalizations regardîng the dosage c in be made. The daily dosage regimen for an adult human patient may be, for exampie, an oral dose of between 0.01 mg and 3000 mg of each active ingrédient preferably between 1 mg and 7pO mg, e.g. 5 to 200 mg. The dosage may be a single one or a sériés of two or more given in the course of one or more days, as is needed by the subject In some embodiments, the compounds wili be administered for a period of continuous therapy, for example for a week or n ore, or for months or years.
[0234] In instances where human dosages for compounds hâve been established for
In some ai least some condition, those same dosages may be used, or dosages that are between about 0
1% and 500%, more preferably between about 25% and 250% of the establishcd human
109 dosage. Where no human dosage is established, as will be the case for newly-discovered ] iharmaceutical compositions, a suitable human dosage can be inferred from ED» or IDjo values, or other appropriate values derived from in vitro or in vivo studies, as qualîfied by toxicity étudies and efficacy studies in animais.
[0235] In cases of administration of a pharmaceutically acceptable sait, dosages may be calculated as the free base. As will be understood by those of skill in the art, in certain s ituatïons it may be necessary to administer the compounds disclosed herein in amounts that < xceed, or even far exceed, the above-stated, preferred dosage range in order to effectively and i ggressivcly treat particulariy aggressive diseases or infections.
[0236] Dosage amount and interval may be adjusted îndividually to provide plasma έlevels of the active moiety which are sufficient to maintain the modulating effects, or minimal ective concentration (MEC). The MEC will vary for each compound but can be estimated m in vitro data. Dosages necessary to achieve the MEC will dépend on individual iracteristics and route of administration. However, HPLC assays or bioassays can be used to d etermine plasma concentrations. Dosage intervals can also be determined using MEC value. Compositions should be administered using a rcgimen which maintains plasma levels above the MEC for 10-90% of the time, preferably between 30-90% and most preferably between 50-90%.
i cases of local administration or sélective uptake, the effective local concentration ofthe drug n lay not be related to plasma concentration.
[0237] It should be noted that the attending physicien would know how to and when to terminate, interrupt, or adjust administration due to toxicity or organ dysfunctions. Convcrsely, the attending physîcian would also know to adjust treatment to higher levels if the c dose in the management of the dïsorder of interest will vary with the severity of the condition to be treated and to the route of administration. The severity of the condition may, for example, be elaluated, in part, by standard prognostic évaluation methods. Further, the dose and perhaps dose frequency, will also vary according to the âge, body weight, and response of the individual patient. A program comparable to that discussed above may be used in veterinary medicine.
[0238] Compounds disclosed herein can be evaluated for efficacy and toxicity using Îown methods. For example, the toxicology of a particular compound, or of a subset of the mpounds, sharing certain chemical moieties, may be established by determining in vitro cicity towards a cell line, such as a mammalian, and preferably human, cell line. The results ’ such studies are often prédictive of toxicity in animais, such as mammals, or more inical response were not adéquate (precluding toxicity). The magnitude of an administrated i
( o
specifically, humans. Altematively, the toxicity of particular compounds in an animal model,
110 such as mice, rats, rabbits, or monkeys, may be determined using known methods. The eflicacy of a particular compound may be establishcd using several recognized methods, such as in vitro methods, animal models, or human clinical trials. When selecting a model to détermine < fïïcacy, the skilled artisan can be guided by the state of the art to choose an appropriate model, c ose, route of administration and/or régime.
EXAMPLES [0239] Additional embodiments are disclosed in further detail in the following examples, which are not in any way intcnded to limit the scope of the claims.
EXAMPLE 1
Préparation of Compound (ta)
MMTrO
MMTrÔ
MMTrÔ
P1-4 [0240] Préparation of (Pt-2): To an ice cooled solution of Pl-1 (10.0 g, 40.8 nmol) in dry pyridine (100 mL) was added TBSC1 in pyridine (IM, 53 mL) dropwise at room température (R.T.). The reaction mixture was stirred at R.T. for 16 hours. The réaction mixture was then quenched with water, concentrated to give a residue. The residue was separated by ethyl acetate (EA) and saturated NaHCOj aq. solution. The organic phase was dried and concentrated. The residue was purified on a silica gel column (5% MeOH in DCM) to give a crude 5’-O-TBS protected intermediate as a white solid (13.4 g, 91%). The intermediate was dissolved in anhydrous DCM (100 mL) and sym-collidine (17.9 g, 149.2 mmol), AgNOj (25 g,
149.2 mmol) and MMTrCI (45 g, 149.2 mmol) were added. The mixture was stirred at R.T. for 16 hours. The mixture was quenched with water, and the organic layer was separated and cc ncentrated. The residue purified on a silica gel column (30% PE in EA) to give the crude product. The crude product was dissolved in IM TB AF (50 mL) in THF. The mixture was
111 s irred at R.T. for 2 hours. The solvent was removed, and the residue was purified on a silica gel c olumn (50% PE in EA) to give Pl-2 as a white solid (21.4 g, 66% for three steps).
[0241] Préparation of ÎP1-3): To a solution of pyridine (521 mg, 6.59 mmol) in aihydrous DMSO (5 mL) was added TFA (636 mg, 5.58 mmol) dropwise at 10°C under nitrogen. The reaction mixture was stirred until the solution became clear. The solution was then added into a mixture of Pl-2 (4.0 g, 5.07 mmol) and DCC (3.86 g, 18.76 mmol) in aihydrous DMSO (18 mL) at R.T. under nitrogen. The reaction mixture was stirred at 30°C ovemight. Water (80 mL) was added into the mixture, diluted with EtOAc (100 mL) and f Itered. The filtrate was extracted with DCM (100 mL x 6). The organic layer was washed v ith saturated aq. NaHCOj, dried over Na2SO4 and concentrated in vacuo. The residue was purified on a silica gel column eluted with 1% MeOH in DCM to give the intermediate (3.5 g, 87.7%) as a yeliow solid. The intermediate (3.5 g, 4.45 mmol) was dissolved in dioxane (25 mL) and aq. HCHO (668 mg, 22.25 mmol) was added at R.T. 2N NaOH (4.5 mL, 8.9 mmol) was tien added. The reaction mixture was stirred at 30°C ovemight. NaBFLj (593 mg, 15.6 mmol) v /as added in by portions at 5°C, and the mixture was stirred at R.T. for 15 min. The reaction v /as quenched with water, and the mixture was extracted with EtOAc (100 mL x 3). The organic t lyer was dried over NaîSO4 and concentrated in vacuo. The residue was purified on a silica gel column eluted wîth 1% MeOH in DCM to give Pl-3 as a yeliow solid (2.5 g, 67%). *H NMR (CDCIj, 400 MHz) J6.82-7.50 (m, 29H), 5.40 (d, J= 23.2 Hz, 1H), 4.99 (d, J = 7.6 Hz, 1H), < .46 (dd, Λ = 6.0 Hz, J2 = 54.4 Hz, IH), 3.94 (dd, Jt = 4.4 Hz, J2 « 12.4 Hz, IH), 3.78 (s, 6H), 2.42-3.69 (m, 2H), 2.71-3.05 (m, 2H), 2.45 (m, IH).
[0242] Préparation of ÎP1-4): To an ice cooled solution of Pl-3 (4.0 g, 4.9 mmol) i i dry pyridine (20 mL) was added dropwise TBSCt in pyridine (IM, 5.88 mL). The reaction riixture was stirred at R.T. for 16 hours. The reaction mixture was then quenched with water, concentrated to give a residue. The residue was separated in EA and saturated aq. NaHCOj. ' lie organic layer was separated and dried, and then concentrated. The residue was purified on a iilica gel column (1% MeOH in DCM) to give the intermediate as a yeliow solid (3.2 g, 70%). H NMR (CDCIj, 400 MHz) <57.53-6.83 (m, 29H), 5.51 (d, J= 21.2 Hz, 1H), 4.98 (d, J = 7.6 Hz, IH), 4.67 (dd, Jt = 5.6 Hz, J2 = 22.4 Hz, IH), 4.22 (dd, Jt = 5.6 Hz, J2 = 53.2 Hz, IH), 4,07 cm, IH), 3.89 (m, IH), 3.80 (s, 6H), 3.70-3.67 (m, IH), 3.03-2.98 (m, IH), 2.26 (m, IH), 0.93 (s,
9H), 0.10 (s, 6H).
[0243] The obtained intermediate was dissolved in anhydrous DCM (20 mL) and collidine (360 mg, 3 mmol), and AgNOj (500 mg, 3 mmol) and MMTrCl (606 mg, 2 mmol) were added. The mixture was stirred at R.T. for 16 hours. The reaction mixture was quenched
112
I vith water, and the organic layer was separated and concentrated. The residue was purified on a j îlica gel column (0.5% MeOH in DCM) to give the fully protected intermediate as a yellow î olid (3.3 g, 80%). The intermediate was dissoived în IM TBAF in THF (5 mL) and was stirred
II R.T. for 2 hours. The solution was concentrated, and the residue was purified on a silica gel tolumn (1% MeOH în DCM) to give a mixture of Pl-3 and Pl-4, which was separated by HPLC séparation (MeCN and 0.1% HCOOH in water) to give Pl-4 as a white solid (1.5 g, 75%).
[0244] Préparation of (Pl-5): Pl-4 (1.5 g, 1.22 mmol) was suspended in anhydrous DCM (50 mL), and Dess Martin periodînane (1.2 g, 2.73 mmol) was added at (TC. The reaction r lixture was stirred at R.T. for 3 hours. The reaction mixture was then quenched with saturated sq. NaîSîOj and NaîCOj. The organic layer was separated and dried, and then concentrated to £ ive the aldéhyde intermediate as a white solid.
[0245] A solution of ClCHîPPhjBr (2.19 g, 5.6 mmol) în anhydrous THF (40 mL) v <as cooled to -78°C. n-BuLi (2.5 M, 2.3 mL) was added în dropwise. After the addition, the r lixture was stirred at 0°C for 2 hours. A solution of the aldéhyde in anhydrous THF (10 mL) was then added. The mixture was stirred at R.T. for 16 hours. The reaction was quenched with saturated NH4CI aq. and extracted by EA. The organic layer was separated, dried and concentrated. The residue was purified on a silica gel column (1% MeOH in DCM) to give the intermediate as a yellow solid (1.1 g, 73%). To a solution of the intermediate (1.1 g, 0.98 mmol) in anhydrous THF (40 mL) was added n-BuLi (2.5M, 6 mL) -78°C dropwise. The mixture was snrred at -78°C for 5 hours and then quenched with a saturated NH4CI aq. solution. The mixture V'as extracted with EA. The organic layer was separated, dried and concentrated. The residue was purified on a silica gel column (2% MeOH in DCM) to give Pl-5 as a yellow solid (910 n îg, 86%).
[0246] Préparation of (la): Pl-5 (910 mg, 0.84 mmol) was suspended în 80% CHjCOOH (50 mL), and the reaction mixture was stirred at 40°C for 15 hours. The solvents were evaporated, and the residue was co-evaporated with toluene to remove traces of acid and water. The residue was purified by HPLC séparation (MeCN and 0.1% HCOOH in water) to give pure compound la as a white solid (101 mg, 45%). *H NMR (MeOD, 400 MHz) J7.90 (d, J = 7.2 Hz, IH), 6.04 (d, J= 19.6 Hz, !H), 5.87 (d, J= 7.6 Hz, IH), 5.00 (dd, Jt ~ 5.2 Hz, J2 =
53.6 Hz, IH), 4.47 (dd, Jt = 5.2 Hz, J2 = 22.8 Hz, IH), 3.86 (d, J= 12.4 Hz, IH), 3.73 (d, J = 1L·Hz, IH), 3.08 (s, IH); ESI-TOF-MS: m/z270.09 [M + H]+, 539.17 [2M + H]+.
113
EXAMPLE 2
Préparation of Compound (2a)
la 2a (0247] To a stirred solution of compound la (50 mg, 0.186 mmol) in anhydrous THF (3 mL) was added dropwise a solution of r-BuMgCl (0.37 mL, IM in THF) at -78°C. The mixture was then stirred at 0°C for 30 min and re-coolcd to -78°C. A solution of phenyl (isopropoxy-L-alaninyl) phosphorochloridate (104 mg, 0.4 mmol) in THF (0.5 mL) was added tropwise. After addition, the mixture was stirred at 25°C for 16 hours. The reaction was c uenched with HCOOH (80% aq.) at 0°C. The solvent was removed, and the residue was purified on silica gel (DCM:McOH = 50:1 to 10:1) to give compound 2a as a white solid (a mixture of two P isomers, 8.0 mg, 7.9 %). *H NMR (MeOD, 400 MHz) £7.71,7.68 (2d, J= 7.6 Hz, IH), 7.17-7.37 (m, 5H), 6,02, 6.00 (2d, J= 20,4 Hz, IH), 5.90, 5.86 (2d, J= 7.6 Hz, IH), f .03-5.18 (m, IH), 4.91-4.99 (m, IH), 4.45-4.55 (m, IH), 4.34-4.43 (m, IH), 4.26-4.33 (m, IH), 3.87-3.95 (m, IH), 3.25,3.22 (2s, IH), 1.29-1.34 (m, 3H), 1.20-1.22 (m, 6H). 3,PNMR (MeOD, 162 MHz) £3.44,3.27. ESI-LCMS: m/z 539.0 [M + H]+.
114
EXAMPLE 3
Préparation of Compound (3a)
HCÎ F
P3-1
TBSd 'F
P3-2
ΗΟ-^\χΟχ/Ν HO—/ °
TBSd F
P3-3
TBDPSO->^OyN^o
ΤΒϋρεο-χ,Ογ^θ *
TBSd F
P3-4
TBSd F
P3-5
TBSd F
P3-8
=<7 ·ο
TBsd F
P3-7
TBDPSCT>^OyN^o
TBsd F
P3-8
Hd F
3a [0248] Préparation of (P3-2V To a solution of P3-1 (100.0 g, 406.5 mmol) in Siyridine (750 mL) was added DMTrCl (164.9 g, 487.8 mmol). The solution was stirred at R.T. br 15 hours. MeOH (300 mL) was added, and the mixture was concentrated to dryness under educed pressure. The residue was dissolved in EtOAc and washed with water. The organic layer was dried over Na2SO4 and concentrated. The residue was dissolved in DCM (500 mL).
Imidazole (44.3 g, 650.4 mmol) and TBSC1 (91.9 g, 609.8 mmol) was added. The reaction mixture was stirred at R.T. for 14 hours. The reaction solution was washed with NaHCOj and brine. The organic layer was dried over Na2SO4, and concentrated to give the crude as a light yellow solid. The crude (236.4 g, 356.6 mmol) was dissolved in 80% HOAc aq. solution (500mL). The mixture was stirred at R.T. for 15 hours. The mixture was diluted with EtOAc j urifîed by silica gel column chromatography (1-2% MeOH in DCM) to give P3-2 (131.2 g,
89.6%) as a light yellow solid.*H NMR (DMSO-dd. 400 MHz) δ 11.39 (s, IH), 7.88 (d, J= 7.2
Hz, IH),5.89(dd,Jt= 18.0Ηζ,Λ=2.0Hz, IH), 5.64(d,J=8.0Hz, IH),5.21 (dd,J/=J2 = 7.2 ώζ,ΙΗ), 5.18-5.03 (m, IH), 4.37-4.29 (m, IH), 3.86 (dd, Ji = J2= 3.2 Hz, 3H), 3.78-3.73 (m, ] H), 3.51-3.56 (m, IH), 3.31 (s, IH), 0.89 (s, 9H), 0.11 (s, 6H); ESI-MS: m/z 802 [M + H]+.
[0249] Préparation of (P3-3): To a solution of P3-2 (131.2 g, 364.0 mmol) în anhydrous CH3CN (1200 mL) was added IBX (121.2 g, 432.8 mmol) at R.T. The reaction
115 mixture was refluxed for 3 hours and then cooled to O’C. The precipitate was filtered-off, and tie filtrate was concentrated to give the crude aldéhyde (121,3 g) as a yellow solid. The rldehyde was dissolved in 1,4-dioxane (1000 mL). 37% CH2O (81.1 mL, 1.3536 mol) and 2M NaOH aq. solution (253.8 mL, 507.6 mmol) were added. The mixture was stirred at R.T. for 2 l ours and then neutralized with AcOH to pH - 7. To the solution were added EtOH (400 mL) and NaBH» (51.2 g, 1.354 mol). The mixture was stirred at R.T. for 30 minutes. The mixture was quenched with saturated aq. NH4CI and extracted with EA. The organic layer was dried c ver Na2SO4 and concentrated. The residue was purified by silica gel column chromatography 1-3% MeOH in DCM) to give P3-3 (51.4 g, 38.9 %) as a white solid.
[0250] Préparation of (P3-4V To a solution of P3-3 (51.4 g, 131.6 mmol) in anhydrous DCM (400 mL) were added pyridine (80 mL) and DMTrCl (49.1 g,144.7 mmol) at ( °C. The reaction was stirred at R.T. for 14 hours, and then treated with MeOH (30 mL). The solvent was removed, and the residue was purified by silica gel column chromatography (1-3% MeOH in DCM) to give a mono-DMTr protected intermediate as a yellow foam (57.4 g, 62.9%). To the intermediate (57.4 g, 82.8 mmol) in CH2C12 (400 mL) was added imidazole (8.4 g, 124.2 rimot) and TBDPSCt (34.1 g, 124.2 mmol). The mixture was stirred at R.T. for 14 hours. The frecipitate was filtered off, and the filtrate was washed with brine and dried over Na2SO4. The solvent was removed to give the residue (72.45 g) as a white solid. The solid was dissolved in 80% HOAc aq. solution (400 mL). The mixture was stirred at R.T. for 15 hours. The mixture was diluted with EtOAc and washed with NaHCOj solution and brine. The organic layer was cried overNa2SO4 and purified by silica gel column chromatography (1-2% MeOH in DCM) to give P3-4 (37.6 g, 84.2%) as a white solid. *H NMR (CDjOD, 400 MHz) <57.76 (d, J- 4.0 Hz, IH), 7.70 (dd, J/= 1.6 Ηζ,Λ = 8.0 Hz, 2H), 7.66-7.64 (m, 2H), 7.48-7.37 (m, 6H), 6.12 (dd, J, = 2.8 Hz, J2= 16.8 Hz, IH), 5.22 (d, J = 8.0 Hz, 1H).5.20~5.05 (m, lH),4.74(dd,J/= 5.6 Hz, J2 = 17.6 Hz, IH), 4.16 (d, J= 12.0 Hz, 1 H), 3.87-3.80 (m, 2H), 3.56 (d,J= 12.0 Hz, IH), 1.16 (s, 9 H), 0.92 (s, 9H), 0.14 (s, 6H).
[0251] Préparation of (P3-5): To a solution of P3-4 (11.8 g, 18.8 mmol) in anhydrous DCM (100 mL) was added Dess-Martin periodinane (16.3 g, 37.6 mmol) at O’C 1 nder nitrogen. The reaction was stirred R.T. for 2.5 hours. Water (100 mL) was added, and the riixture was then filtered. The filtrate was washed with saturated aq. NaHCOj and concentrated. The crude residue was purified by silica gel column chromatography (20% EtOAc in hexane) to give P3-5 as a white solid (10.1 g, 86.0%).
[0252] Préparation of (P3-6): To a mixture of methyltriphenylphosphonium bromide (15.7 g, 48.5 mmol) in anhydrous THF (100 mL) was added n-BuLi (19.4 mL, 48.48
116
nmol) at -78°C under nitrogen. The reaction was stirred at 0°C for 30 minutes. A solution of *3-5 (10.1 g, 16.2 mmol) in anhydrous THF (70 mL) was added dropwise at 0°C under nitrogen. The réaction was stirred at R.T. for 1.5 hours. The reaction was quenched by NH4CI and extracted with EtOAc. The crude product was purified by silica gel column chromatography E‘0% EtOAc in hexane) to give P3-6 as a white solid (8.3 g, 82.2%).’H NMR (CDCI3, 400
Hz)<58.16(s, 1 H), 8.81 (d,J=8.0Hz, 1 H), 7.58-7.67 (m, 4H), 7.37-7.46 (m, 6H), 6.17(d,J=
6.0 Hz, IH), 5.91 (dd, J/ = 10.8 Hz, Λ = 17.6 Hz, IH), 5.42 (d, J= 17.6 Hz, IH), 5.22-5.30 (m, ::H), 4.60-4.84 (m, 2H), 3.69 (dd, J2 = 11.6 Hz, J2 = 21.2 Hz, 2H), 1.10 (s, 9H), 0.91 (s, IH), 0.12 (d, J =8.0 Hz, 6H).
[0253] Préparation of (P3-7): To a solution of P3-6 (6.3 g, 10.09 mmol) in î nhydrous CH3CN (50 mL) were added TPSC1 (6.1 g, 20.2 mmol), DMAP (2.5 g, 20.2 mmol) tnd NEt3 (3 mL) at R.T. The reaction was stirred at R.T. for 2 hours. NH4OH (25 mL) was t dded, and the reaction was stirred for 1 hour. The mixture was diluted with DCM (150 mL) tnd washed with water, 0.1 M HCl and saturated aq. NaHCOj. The solvent was removed, and t le crude product was purified by silica gel column chromatography (2% MeOH in DCM) to i ;ive P3-7 as a yellow solid (5.9 g, 93.6%).
(0254] Préparation of (P3-81: To a solution of P3-7 (5.9 g, 9.5 mmol) in MeOH (10 riL) was added Pd/C (1.5 g) at R.T. The reaction was stirred at R.T. for 2 hours under H2 ( jalloon). The mixture was filtered, and the filtrate was concentrated in vacuo to give P3-8 as a white solid (5.4 g, 91.3%).
[0255] Préparation of (3a): To a solution of P3-8 (5.4 g, 8.6 mmol) in MeOH (60 mL) was added NH4F (10.0 g), and the reaction mixture was refluxed ovemight. After cooling t » R.T., the mixture was filtered, and the filtrate was concentrated. The crude product was purified by silica gel column chromatography (10% MeOH in DCM) to give compound 3a as a vhite solid (1.6 g, 67.8%), *H NMR (CDjOD, 400 M Hz) <J8.08 (d, J= 7.6 Hz, I H), 6.07 (dd, J/ = 3.2 Hz, J2 = 15.6 Hz, IH), 5.88 (d, J = 7.2 Hz, IH), 5.04 (ddd, Ji = 32 Hz, J2 = 5.2 Hz, Λ =
5-4.0 Hz, IH), 4.45 (dd, Jt = 5.2 Hz, J2 =17.2 Hz, IH), 3.76 (d, J= 12.0 Hz, IH), 3.57 (d, J =
12.0 Hz, IH), 1.78-1.85 (m, IH), 1.58-1.67 (m, IH), 0.95 (t, J= 7.6 Hz, 3H); ESI-MS: m/z 274 [M + H]+, 547 [2M + Hf.
117
EXAMPLE 4
Préparation of Compound (4a)
TBDPSO
P3-7 [0256] To a solution of P3-7 (280 mg, 0.45 mmol) in MeOH (10 mL) was added ISIH4F (1.0 g) at R.T. The reaction mixture was refluxed for 5 hours. After cooling to R.T., the ί31ixture was filtered, and the filtrate was concentrated. The crude product was purified by silica el column chromatography (10% MeOH in DCM) to give compound 4a as a white solid (82 , 67.2% 1.6 g, 67.8%). lH NMR (CDjOD, 400 M Hz) J 8.11 (d, J= 7.6 Hz, IH), 5.99-6.08 2H), 5.88 (d, J= 7.6 Hz, IH), 5.47 (dd,J/ = 1.2 Hz, J2 = 17.2 Hz, IH), 5.26 (dd,Λ = 1.6 Hz, =112 Hz, IH), 4.97 (d, J= 5.2 Hz, 0.5H),4.82 (d, J= 7.6 Hz, 0.5H), 4.52 (dd, Jt = 5.2 Hz, J2 = 23.2 Hz, IH), 3.65 (d, J= 12.4 Hz, 1H),3.54 (d, J= 12.4 Hz, 1 H); ESI-MS: m/z 272 [M + H]+, 543[2M + H]+.
EXAMPLE 5
Préparation of Compound (5a)
TBDPS
TBSÔ [0257] Préparation of (P5-I): To a solution of P3-6 (600 mg, 0.96 mmol) in MeOH 0 mL) was added 10% Pd/C (320 mg) at R.T. The mixture was stirred under H2 balloon at .T. for 3 hours. The reaction mixture was filtered, and the filtrate was concentrated to give P5(540 mg, 89.8 %) as a colorless solid. The crude product was used directly for the next step without purification.
[0258] Préparation of (5a): To a solution of P5-1 (540 mg, 0.86 mmol) in MeOH (3 mL) was added NH4F (1.2 g, 32.4 mmol) R.T. The mixture was refluxed for 30 hours. The solid was removed by filtration, and the filtrate was concentrated. The residue was purification by silica gel column chromatography (2.5%-9%McOH in DCM) to give compound 5a (190 mg,
0.6%) as a colorless solid. ’H NMR (CDjOD, 400 MHz) δ 8.05 (d, 8.0 Hz, IH), 6.09 (dd,
118
Jz =4.0 Hz, J2 =14.8 Hz, IH), 5.04-5.20 (m ,1H), 4.42 (dd, Jt = 5.2 Hz, J2 = 13.6 Hz, IH), 3.71 (d, J= 11.6Hz, IH), 3.57(d, J= 12.0Hz, IH), 1.61-1.82(m,2H), 0.94(t, J= 7.2Hz,3H).
EXAMPLE 6
Préparation of Compound (6a) rC TBSO-XzO>.N^_ TBSO—'''VJ “
TBSd F
P6-1
NH, ô
HÔ 'F
6a rt
TBSO t
P3-3 [0259] Préparation of (P6-1): To a solution of P3-3 (800 mg, 2.05 mmol) in anhydrous DCM (15 mL) were added imidazole (558 mg, 8.2 mmol), TBSC1 (1.2 g, 8.2 mmol) and AgNO3 (700 mg, 4.1 mmol) at R.T. The reaction mixture was stirred at R.T. ovemight. Îtie mixture was filtered, and the filtrate was washed with brine and concentrated in vacuo. The siduc was purifîed by column chromatography on silica gel to give P6-1 as a white solid (950 g, 79.2%).
[0260] Préparation of (6a): To a solution of P6-1 (600 mg, 0.97 mmol) in anhydrous CH3CN (18 mL) was added DMAP (239 mg, 2.91 mmol), NEt3 (294 mg, 2.91 mmol) andTPSC1 (879mg, 2.91 mmol) atR.T. The reaction wasstirredat R.T. for 1 hour. NH4OH(9 mL) was added, and the reaction was stirred for 3 hours. The mixture was diluted with EtOAc (200 mL) and washed with water, 0.1 M HCl and saturated aq. NaHCO3. The organic layer was separated, dried and concentrated to give a crude residue. The crude residue was purified by cjlumn chromatography on silica gel to give the product as a white solid (500 mg, 83.3%). The solid was treated with NH4F (1.0 g) in MeOH (20 mL) at refluxed température for 5 hours. The mixture was filtered, and the filtrate was concentrated in vacuo. The residue was purifîed by c jlumn chromatography on silica gel (15% MeOH in DCM) to give compound 6a as a white solid (132 mg, 59.3%).*H NMR (DMSO-r/d, 400 MHz) â7.89 (d, J= 7.6 Hz, IH), 7.22 (d, J =
18.8 Hz, 2H), 6,09 (dd, Jt = 4.4 Hz, J2 = 14.8 Hz, IH), 5.73 (d, J= 5.2 Hz, 1H), 5.52 (d, J = 5.6 Fz, 1 H),5.12 (ζ J =4.8 Hz, IH), 4.90-5.06 (m, IH),4.50 (t, J =6.0Hz, IH),4.27-4.33 (m, IH), 3 [1m + h]+.
(dd, Jt = 5.2 Hz, J2 = 12,0 Hz, IH), 3.47-3.58 (m, 3H); ESI-MS: m/z 276 [M + Hf, 551 !0
119
EXAMPLE 7
Préparation of Compound (7a)
TBDPSO CH . . TBSÔ' ?
P7-1 nh4f
HÔ [0261] Préparation of (P7-1): A mixture of P3-4 (1.60 g, 2.5 mmol), PPhj (1.3 g, .0 mmol) and CCU (0.76g, 5.0 mmol) in DCE (20 mL) was heated to 130°C under microwave irradiation under N2 for 40 mins. After cooled to R.T., the solvent was removed, and the residue v as purified on a silica gel column (PE/EA = 50/1 to 10/1) to give P7-1 (1.1 g, 68.8%) as a v hite solid.
[0262] Préparation of (P7-2); P7-1 (0.80 g, 1.3 mmol), DMAP (0.3 g, 2.6 mmol), T PSC! (0.8 g, 2.6 mmol) and EtjN (0.3 g, 2.6 mmol) were dissolved in MeCN (30 mL). The n.ixture was stirred at R.T. for 14 hours. NHj in THF (saturated at 0°C, 100 mL) was added to the mixture, and the mixture was stirred at R.T. for 2 hours. The solvent was removed, and the residue was purified by column (DCM/MeOH = 100:1 to 50:1) to give P7-2 (0.63 g, 78.8%) as a white solid.
[0263] Préparation of (7a): To a solution of P7-2 (0.63 g, 0.98 mmol) in MeOH 0 mL) was added NH4F (0.3 g), and the reaction was refluxed for 12 hours. The réaction was ci >o!ed to R.T., and the precipitate was filtered off. The filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography (10% MeOH in DCM) to give ompound 7a as a white solid (153 mg, 53.5%). *H NMR (CD3OD, 400 M Hz) £8.05 (d, J= 7.2 F z, iH), 6.14 (dd, Λ = 3.6 Hz, Λ =15.2 Hz, IH), 5.92 (d, J = 7.2 Hz, IH), 5.15 (ddd, Λ = 4.0
E z, J2 = 5.2 Hz, Ji = 53.6 Hz, IH), 4.57 (dd, Λ = 4.8 Hz, J2 = 15.2 Hz, IH), 3.93 (d, J- 11.6
E z, IH), 3.75-3.84 (m, 3H); ES1-MS: m/z 294 [M + H]+, 587[2M + H]+.
120
EXAMPLE «
Préparation of Compound (8a)
[0264] To a solution of P7-1 (630 mg, 0.5 mmol) in MeOH (10 mL) was added NH^F (0.1 g), and the reaction was refluxed for 12 hours. The mixture was filtered, and the liltrate was concentrated in vacuo. The crude product was purified by silica gel column chromatography (10% MeOH in DCM) to give compound 8a as a white solid (153 mg, 53.5%). 'H NMR (CDjOD, 400 M Hz) J7.99 (d, 8.0 Hz, IH), 6.17 (dd, J, = 4.4 Hz, J2 =14.4 Hz,
H), 5.70 (d, J = 8.0 Hz, IH), 5.22 (ddd, Jt = J2 = 4.8 Hz, Jj = 53.2 Hz, IH), 4.55 (dd, Jt ~ 5.2 Hz, J2 = 12.4 Hz, IH), 3.88 (d, J= 12.0 Hz, IH), 3.76-3.79 (m, 3H); Negative-ESl-MS: m/z i:93[M-H]-.
EXAMPLE 9
Préparation of Compound (9a)
[0265] Préparation of (P9-1): A mixture of P3-4 (3.2 g, 5.0 mmol), PhjP (5.2 g, 20 runot), iodine (2.60 g, 10.2 mmol) and imidazote (1.4 g, 20mmot) in anhydrous THF (40 mL) was stirred at 80°C for 14 hours. The reaction was cooled to R.T. and quenched with saturated aq. NaîSîOî. The solution was extracted with EA. The organic layer was dried over Na2SO4 a!nd concentrated. The residue was purified by silica gel column chromatography (20-50% EA il PE) to give P9-1 (1.6 g, 68.2%) as a white solid.
[0266] Préparation of (P9-2): A mixture of P9-1 (1.4 g, 0.2 mmol), EtjN (40 mg, 0L4mmol) and Pd/C in EtOH (20 mL) was stirred at R.T. under H2 (balloon) ovemight. The
121 precipitate was filtered off, and the filtrate was concentrated. The residue was purified on a silica gel column (20%-50% EtOAc in PE) to give P9-2 as a white solid (l.l g, 78%). *HNMR. (CDCh,400MHz) <58.11 (brs, lH), 7.76 (d, J= 8.0 Hz, 1H), 7.39-7.67 (m, 10H), 6.18 (dd, J, =
3.2 Hz, J2 = 14.4 Hz, 1H), 526-5.30 (m, 1H), 4.86 (m, 1H), 4.42 (dd, Jt = 5.2 Hz, J2 = 152 Hz, 1H), 3.81 (d, J = 11.2 Hz, 1 H), 3.58 (d, J= 11.2 Hz, 1 H), 1.16 (s, 3H), 1.11 (s, 9H), 0.91 (s, 9H), 0.13 (s, 3 H), 0.08 (s, 3H).
[0267] Préparation of (P9-3): P9-2 (650 mg, 1.1 mmol), DMAP (270 mg, 22 mmol), TPSC1 (664 mg, 2.2 mol) and EtjN (222 mg, 22 mmol) were dîssolved in MeCN (20 mL). The mixture was stirred at R.T. for 14 hours. The reaction was added NH3 in THF (saturated at 0°C), and the mixture was stirred at R.T. for 2 hours. The solvent was removed, aid the residue was purified on a silica gel column (1-10% MeOH in DCM) to give P9-3 (430 mg, crude) as a light yellow syrup.
[0268] Préparation of (9a): A mixture of P9-3 (430 mg, 0.7 mmol) and NH|F (97 mg, 2. Immol) in MeOH (10 mL) was refluxed for 14 hours. The solvent was removed, and the Îsidue was purified on a silica get column (5%-l 0% MeOH in DCM) to give compound 9a as a hite solid (64.8 mg, 35.4%). *H NMR (CD3OD, 400 MHz) <58.10 (d, J= 7.6 Hz, 1H), 6.03 (dd, =2.0 Hz, Λ = 16.8 Hz, 1H), 5.87 (d, J= 7.6 Hz, 1H), 4.98 (m, 1H), 4.37 (dd, J/ = 52 Hz, Λ s
21.6 Hz, 1H), 3.59(dd, Jt = 12.0 Hz, J2 = 28.4 Hz,2H), 123 (d,.7=0.8 Hz,3H).
EXAMPLE 10
Préparation of Compound (10a)
TBDPSO
[0269] To a stirred solution of P9-2 (400 mg, 0.65 mmol) in MeOH (20 mL) was a ided NH4F (52 mg, 1.5 mmol). The mixture was refluxed ovemight. The solvent was removed, and the residue was purified on a silica get column (5-10% MeOH in DCM) to give compound 10a (140 mg, 82.4%) as a white solid. 1
4 Hz, 1H), 6.06 (dd, Jt = 2.8 Hz, J2 = 16.4 Hz, 1H), 5.67 (d, J = 8.0 Hz, 1H), 5.08 (m, 1H), 4 37 (d, J, = 5.2 Hz, J2 = 18.8 Hz, 1H), 3.59 (dd, Jt =12.0 Hz, J2 = 26.4 Hz, 2H), 1.23 (s, 3H).
Ï5
H NMR. (CDjOD, 400 MHz) J 8.05 (d, J =
ES1-TOF-MS: m/z 283 [M + Naf.
122
TBDPSO
P3-5
EXAMPLE 11
Préparation of Compound (lia)
P11-2
P11-1
[0270) Préparation of (PI 1-1Ί: To a solution of P3-5 (2.1 g, 3.5 mmol) in snhydrous THF (25 mL) was added cthynylmagnesium bromide (5.1 mmol) at -78°C. The r saction was stirred at 0°C for 3 hours. The reaction was quenched with saturated aq. NH4CI (10 riL). The mixture was diluted with EtOAc (200 mL) and washed with water and brine. The c rganic layer was dried and concentrated to give a residue. The residue was purified by column chromatography on silica gel (eluting with DCM: MeOH = 60:1) to give PI 1-1 as a white solid ( Ï70 mg, 83.3%).
[0271] Préparation of (Pll-21): PI 1-1 (870 mg, 1.34 mmol) was dissolved in anhydrous DCM (12 mL), and methyl chloroformate (2.3 mL) and pyridine (2.5 mL) were aidded at R.T. The reaction mixture was stirred at R.T. for 1 hour. The mixture was diluted with DCM and washed with saturated aq. NaHCOj. The organic layer was separated, dried and concentrated to give a residue. The residue was purified by column chromatography on silica el (eluting with PE: EtOAc = 8: 1) to give a crude product as a white solid (830 mg, 88.4%).
To a mixture of Pd2(dba)3 (55 mg, 0.06 mmol) in anhydrous DMF (12 mL) was added P(nBu)3 (35 mg, 0.17 mmol) and HCOONH4 (108 mg, 1.7 mmol) at R.T. under nitrogen. The reaction mixture was stirred at R.T. for 30 min. A solution of the crude product (830 mg, 1.16 mmol) in anhydrous DMF (16 mL) was added, and the reaction mixture was stirred at 70°C for 3 hours. The reaction was diluted with EtOAc and washed with brine. The organic layer was separated, dried and concentrated to give a residue. The residue was purifîed by column chromatography 01 silica gel (eluting with PE: EtOAc = 9: 1) to give PI 1-2 as a white solid (510 mg, 67.6%).^ b MR (CD3OD, 400 M Hz) 7.61-7.75 (m, 5H), 7.36-7.47 (m, 6H), 6.04 (d, J= 18.8 Hz, IH), 5 (t, J= 6.8 Hz, IH), 5.21 (dd, Jt « 1.2 Hz, J2 = 7.2 Hz, IH), 5.10 (q, J, = 5.2 Hz, J2 = 53.6
123 ' :, IH), 4.80-4.92 (m, IH), 4.59-4.79 (m, 2H), 3.86 (d, 12.0 Hz, IH), 3.75 (d, 12.0 Hz,
H), 1.09 (s, 9H), 0.92 (d, J= 4.4 Hz, 9H), 0.15 (t, 4.0 Hz, 6H).
[0272] Préparation of fPll-3): To a solution of Pi 1-2 (490 mg, 0.77 mmol) in anhydrous MeCN (15 mL) was added TPSCI (700 mg, 2.31 mmol), DMAP (282 mg, 2.31 mmol) and TEA (234 mg, 2.31 mmol) at R.T. The réaction mixture was stirred at room température for 1 hour. Then NH4OH (8 mL) was added and the réaction mixture was stirred for another 4 hours. The mixture was diluted with EtOAc and washed with water, 1.0 M aq. HCl and saturated aq. NaHCO3.The organic layer was separated and dried, concentrated to give the residue which was purified by HPLC séparation (MeCN and 0.1% HCOOH in water) to give Pll-3 as a white solid (190 mg, 38.8%).lH NMR (CD3OD, 400 MHz) Î7.88 (d, J= 7.2 Hz, IH), 7.63-7.70 (m, 4H), 7.37-7.48 (m, 6H), 6.12 (d, J= 18.4 Hz, IH), 5.49 (d, 7.6 Hz, IH),
5.34 (t, J= 6.8 Hz, IH), 4.84-5.01 (m, 2H), 4.66-4.78 (m, 2H), 3.89 (d, J= 11.6 Hz, IH), 3.75 (d, J= 11.6 Hz, IH), 1.10 (s, 9H), 0.91 (d, J= 3.2 Hz, 9H), 0.13 (t, J= 5.2 Hz, 6H).
[0273] Préparation of fl la): To a solution of PI 1-3 (130 mg, 0.21 mmol) in MeOH (! J mL) was added NHjF (1 g), and the reaction mixture was refluxed for 6 hours. The mixture was fiitered, and the filtrate was concentrated in vacuo. The residue was purified by column c iromatography on silica gel (eluting with DCMtMeOH - 13:1) to give compound lia as a hite solid (47 mg, 79.1%)?H NMR (CD3OD, 400 MHz) 8.07 (d, J= 7.6 Hz, IH), 6.05 (dd, = 1.2 Ηζ,Λ= 16.8 Hz, IH), 5.86 (d, J = 7.6 Hz, IH), 5.40 (dd,J/ = J2 = 6.8 Hz, IH), 4.8799 (m, 3H), 4.46-4.80 (m, IH), 3.75 (d, J= 12.4 Hz, IH), 3.68 (d, 12.4 Hz, IH); ESI-MS:
Wl
4* nVz 284.02 [M + H]\ 567.08 [2M + H]+.
EXAMPLE 12
Préparation of Compound fl 2a) ri
TBDPSO'XzO^'i OTBSd 'F
P3-4
Ô°H
TBDPSO-A^OyN _ V 'F
P12-1
NH,
TBDPSO-VDyNn
P12-2
NH, ό
d 'f
12a
124 [0274J Préparation of (P12-1): To a solution of P3-4 (500 mg, 0.8 mmol) in i .nhydrous toluene (12 mL) was added DAST (0.3 mL, 2 mmol) at -65°C under nitrogen. The reaction mixture was stirred at R.T. for 2 hours. The reaction was quenched with saturated aq. NaHCOj and extracted with EtOAc. The organic layer was separated, dried and concentrated to give the residue. The residue was purified by column chromatography on silica gel (eluting with PE: EtOAc = 9: 1) to give P12-1 as a yellow solid (170 mg, 42.5%). *H NMR (CD3OD, 400 MHz) £7.66 (dd, J, = 1.6 Hz, J2 = 18.0 Hz, 4H), 7.54 (d, J = 7.6 Hz, IH), 7.35-7.47 (m, 6H), ( .59 (dd,Λ = 5.6 Hz, J2 = 14.0 Hz, IH), 5.78 (d, J= 7.6 Hz, IH), 5.05-5.24 (m, 2H), 4.93 (d, -.6 Hz, IH), 4.57 (d, J =7.6 Hz, IH), 3.93-4.00 (m, 2H), 1.07 (d, J =2.4 Hz, 9H).
[0275J Préparation of (PI 2-2): To a solution of P12-1 (100 mg, 0.2 mmol) in s nhydrous MeCN (5 mL) was added TPSC1 ( 182 mg, 0.6 mmol), DMAP (68 mg, 0.6 mmol) and TEA (61 mg, 0.6 mmol) at R.T. under nitrogen. The reaction mixture was stirred at R.T. for 1 hour. NH4OH (3mL) was added, and the reaction was stirred for 2 hours. The mixture was d layer was separated, dried and concentrated to give a residue. The residue was purified by c mg, 96%).
iluted with EtOAc and washed with water, 1.0 M HCl and saturated aq. NaHCOj. The organic jlumn chromatography on silica gel (DCM:McOH = 50:1) to give P12-2 as a yellow solid (96 [0276|
Préparation of (12a): To a solution of P12-2 (96 mg, 0.2 mmol) in MeOH ( > mL) was added NH4F (500 mg) at R.T. The reaction was refluxed for 3 hours. The mixture vas filtered, and the residue was purified by RP HPLC (MeCN and 0.1% HCOOH in water) to give compound 12a as a white solid (25 mg, 48.7%). *H NMR (CDjOD, 400 MHz) £7.85 (d, J
7.6 Hz, IH), 6.59 (dd, J, = 5.2 Hz, J2 = 12.8 Hz, IH), 6.04 (d, 7.6 Hz, IH), 5.10-5.26 (m, : fl), 4.79-4.90 (m, IH), 4.57 (d, J = 7.6 Hz, IH), 3.82 (d, J= 12.4 Hz, IH), 3.76 (dd, Jt = 1.6 F z, J2 = 12.4 Hz, IH); ESI-MS: m/z 257.9 [M + H]+, 514.8 [2M + H]*.
EXAMPLE 13
Préparation of Compound (13a)
NHMMTr ô
MMTrÔ 'F
P13-1
[0277J Préparation of (P 13-1): To a solution of compound 3a (700 mg, 2.56 mmol) ir anhydrous pyridine (5 mL) were added TBDPSC1 (2.8 g, 10.24 mmol), imidazole (522 mg, 7.58 mmol) and AgNOj (870 mg, 5.12 mmol) at R.T. under N2. The reaction mixture was
125
stirred at R.T. for 3 hours. The mixture was diluted with MeOH and filtered. The mixture was concentrated, and the residue was purified by column chromatography on silica gel (eluting with DCM: MeOH = 80:1 — 40:1) to give the crude intermediate as a yellow solid (1.05 g, 80.8%)?H NMR (DMSO-c/5,400 MHz) 81.15 (d,J=1.6 Hz, IH), 7.61-7.65 (m, 4H), 7.41-7.50 (m, 7H), i>.02 (dd, Jt - 2.8 Hz, J2 = 17.2 Hz, IH), 5.69 (d, J= 6.0 Hz, IH), 5.56 (d, J= 7.6 Hz, IH), 4.965.11 (m, IH), 4.37-4.46 (m, IH), 3.82 (d, J= 10.8 Hz, IH), 3.62 (d, J= 10.8 Hz, IH), 1.70-1.78 ( m, IH), 1.53-1.59 (m, IH), 1.02 (s, 9H),0.79 (t, J = 7.6 Hz, 3H). To a solution ofthe crude intermediate (1.0 g, 1.96 mmol) in anhydrous DCM (15 mL) were added sym-collidine (1.4 g,
1.76 mmol), AgNO3 (1.0 g, 5.88 mmol) and MMTrCl (4.8 g, 15.6 mmol) at R.T. under N2. ' 'he reaction mixture was stirred at R.T. ovemight. The mixture was filtered and concentrated. ' he residue was purified by column chromatography on silica gel (eluting with PE:EtOAc=2: 1) to give crude full protected intermediates as a white solid(l. 1 g, 53.1%). To a solution of the Jrude intermediate (600 mg, 0.57 mmol) in THF (5 mL) was added TBAF (446 mg, 1.71 mmol)) î t R.T. The reaction was stirred at 40~50°C ovemight The crude product was purified by column chromatography on silica gel eluted with PE:EtOAc = 3:2 to give crude P13-1 (350 mg, ' 5.1%) as a yellow solid.
[0278] Préparation of (13a): To a solution of P13-1 (300 mg, 0.37 mmol) in CH3CN (2.5 mL) were added NM1 (2.5 mL) and a solution of phenyl(isopropoxy-L-alanmyl) phosphorochloridate (2.55 g, 7.4 mmol) in CHjCN (2.5 mL) at R.T. under N2. The reaction mixture was stirred at R.T. for 3 hours. The mixture was concentrated in vacuo. The residue was purified by column chromatography on silica gel (PE:EtOAc = 1:1) to give crude product as a yellow oïl (500 mg, 81%). The crude product was further treated with 80% HCOOH (70 mL) ai R.T. ovemight. The mixture was concentrated in vacuo, and the crude product was purified
Z5 by RP HPLC (MeCN and 0.1% HCOOH in water) to give compound 13a as a white solid (a mixture of two P isomers, 86 mg, 40.3% two steps). ’H NMR (CDjOD, 400 MHz) 81.15, 7.71 ( !d, 7.6 Hz, IH), 7.33-7.38 (m, 2H), 7.19-7.26 (m, 3H), 6.02-6.10 (m, IH), 5.87, 5.82 (2d, J = 7.6 Hz, IH), 4.99-5.02 (m, 0.5H), 4.72-4.82 (m, 1.5H), 4.14-4.43 (m, 3H), 3.89-3.94 (m,
H),1.68-1.81 (m, 6H), 1.51-1.56 (m, IH), 1.30-1.43 (m, 8H), 0.96-1.01 (m, 3H); ESI-MS: m/z 532.93 [M + H]+.
128
MMTrÔ
P14-1
MMTrÔ
P13-1
EXAMPLE 14
Préparation of Compound (14a)
[0279J Préparation of (P14-1): To a stirred solution of P13-1 (451 mg, 0.55 mmol) and NMI (ImL) in anhydrous acetonitrile (2 mL) was added dropwise a solution of 2-chloro-8methyl-4H-benzo[d][l,3,2]dioxaphosphinine (855 mg, 4.2 mmol) in acetonitrile (0.2 mL) at 0°C under N2. The mixture was stirred at R.T. for 2 hours. Solution of I2 (3.2 g, 12.6 mmol), pvridine (9 mL), H2O(3 mL) and DCM(3 mL) was added. The reaction mixture was stirred for 3 ) mins. The reaction was quenched with NaS2O3 solution and extracted with EA. The organic li.yer was dried over Na2SO4 and concentrated. The residue was purified by column on silica gel (PE: EA = 1:1 to 1:2) to give P14-1 (205 mg, 37%) as a white solid.
[0280J Préparation of (14a): P14-1 (205 mg, 0.21 mmol) was dissolved in 80% F COOH aq. solution, and the mixture was stirred at R.T. for 16 hours. The solvent was removed, and the residue was purified by RP HPLC (HCOOH System) to give compound 14a as mixture of 2 P-isomers (24 mg, 18%). ’H NMR (CDjOD, 400 MHz) <5 7.60, 7.53 (2d, J = 8.0 z, IH), 7.21-7.25 (m, IH), 7.02-7.12 (m, 2H), 5.95. 5.87 (2dd, Jt = 2.4 Hz, J2 = 18.0 Hz, IH), 71, 5.69 (2d, 8.0 Hz, IH), 5.38-5.53 (m, 2H), 5.06, 5.04 (2ddd, Ji = 2.4 Hz, J2 = 5.6 Hz, Jj
54.0 Hz, IH), 4.32-4.49 (m, 2H), 2.26 (d, J= 3.6 Hz, 3H), 1.83-1.92 (m, IH), 1.64-1.72 (m, i), 0.96, 0.93 (2t, J = 7.6 Hz, 3H). 31P NMR (CD3OD, 162 MHz) J-8.22, -8.50; ESI-LCMS:
iri/z 456 [M + H]+.
127
EXAMPLE 15
Préparation of Compound (15a)
[0281] Stepl. Préparation of (P15-1): To a mixture of P3-8 (2.2 g, 2.5 mmol), AgNOj (844 mg, 5.0 mmol) and collidine (907 mg, 7.5 mmol) in anhydrous DCM (10 mL) was added MMTrCl (1.54 g, 5.0 mmol) under N2. The reaction mixture was stirred at R.T. ovemight. The reaction mixture was filtered through a Buchner Funnel. The filtrate was vashed with saturated NaHCOj solution and brine. The organic layer was separated, dried over mhydrous Na2SO4 and filtered. The filtrate was concentrated to dryness. The residue was îurified by column on silica gel (PE:EA « 10:1 to 1:2) to give the intermediate (2.3 g, 84%), «vhich was dissolved in a solution of TBAF in THF (IM, 2.6 mL) under N2. The reaction mixture was stirred at R.T. ovemight. The residue was dissolved in EA (200 mL) and washed El*dth water and brine. The organic layer was separated, dried over anhydrous Na2SO4 and Itered. The filtrate was concentrated to dryness, and the residue was purified by column on lica gel (DCM/MeOH = 100:1 to 30:1) to give P15-1 as a white foam (1.3 g, 94%).
[0282] Préparation of (15a): To a stirred solution of P15-1 (300 mg, 0.55 mmol) and proton sponge (235 mg, 1.1 mmol) in anhydrous MeCN (9 mL) was added with a solution of POCI3 (169 mg, 1.1 mmol) in MeCN (1 mL) via syringe at 0°C. The mixture was stirred at R.T. for 40 mins. A mixture of (S)-cyclohexyl 2-aminopropanoate hydrochloride (525 mg, 2.55 mmol) and TEA (0.1 mL) was added at 0°C. The mixture was warmed to R.T. and stirred for 3 hours. The reaction mixture was quenched with saturated NaHCOj, and extracted with EA ( 100 mL x 2). The combined organic layers was dried over Na2SO4, concentrated and purified by silica gel column (1~4% MeOH in DCM) to give the crade product (400 mg, 78.15%) as a yellow solid. The crade product was treated with 80% HCOOH (50mL) at R.T. for 16 hours. The solvent was removed, and the residue was purified by RP HPLC to give compound 15a as a white solid (40 mg, 14%). ’H NMR (MeOD, 400 MHz) δ 7.82 (d, J= 7.6 Hz, 1H), 6.09 (dd, J, = 2.8 Hz, J2 = 14.0 Ηζ,ΙΗ), 5.98 (d, J = 7.6 Hz, 1H), 5.04 (ddd, J, = 3.2 Hz, J2 = 5.6 Hz, J3 =
53.6 Hz, 1H), 4.71-4.77 (m, 2H), 4.45 (dd, Λ = 5.6 Hz, J2 = 12.4 Hz, 1H), 4.14A18 (m, IH), 3.97-4.01 (m, 1H), 3.84-3.92 (m, 2H), 1.31-1.87 (m, 28H), 0.99 (t, J= 7.2 Hz, 3H). P NMR (CDjOD, 162 MHz) δ 13.94; ESI-LCMS: m/z 660 [M + H]+.
128
EXAMPLE 16
Préparation of Compound (16a)
[0283] To a stirred solution of compound 4a (150 mg, 0.56 mmol) in anhydrous THF i 3 mL) was added dropwise a solution of r-BuMgCl (1.2 mL, IM in THF) at -78°C. The i nixture was stirred at 0°C for 30 min and re-cooled to -78°C. A solution of phenyl(isopropoxy,-alaninyl) phosphorochloridate (312 mg, 1.2 mmol) in THF (1.0 mL) was added dropwise. After addition, the mixture was stirred at 25°C for 16 hours. The reaction was quenched with 1COOH (80% aq.) at 0°C. The solvent was removed, and the residue was purified on silica gel OCM:MeOH = 50:1 to 10:1) to give compound 16a as a white solid (24,0 mg, 15 %).*H NMR (MeOD, 400 MHz) 57.76 (d, J = 7.2 Hz, IH), 7.17-7.38 (m, 5H), 6.01-6.08 (m, 2H), 5.81 (d, J = 7.6 Hz, IH), 5.54-5.58 (m, IH), 5.35-5.38 (m, IH), 4.92-4.97 (m, 2H), 4.45-4.52 (m, IH), 4.08-4.19 (m, 2H), 3.88-3.92 (m, IH), 1.28-1.33 (m, 3H), 1.20-1.22 (m, 6H); 3IP NMR (CDjOD, 162 MHz) δ 7.36; ESI-LCMS: m/z 541.0[M + H]+.
EXAMPLE 17 Préparation of Compound (17a)
[0284] Préparation of (P17-1): To a solution of P3-7 (1.4 g, 2.3 mmol) în MeOH (50 mL) was added NH4F (8.0 g) at R.T. The reaction mixture was refluxed ovemight. After cooling to R.T., the mixture was filtered, and the filtrate was concentrated. The crude product
129
was purified by silica gel column chromatography (10% MeOH in DCM) to give P17-1 as a vhite solid (410 mg, 77.8%).
[0285] Préparation of (P17): To a stirred solution of P17-1 (60 mg, 0.19 mmol) in i nhydrous THF (3 mL) was added dropwise a solution of i-BuMgCl (0.38 mL, IM in THF) at 8°C. The mixture was stirred at 0°C for 30 min and re-cooled to -78°C. A solution of nhenyl(isopropoxy-L-alaninyl) phosphorochloridate (104 mg, 0.4 mmol) in THF (0.5 mL) was t dded dropwise. After addition, the mixture was stirred at 25°C for 16 hours. The reaction was < uenched with HCOOH (80% aq.) at 0°C. The solvent was removed, and the residue was purified on silica gel (DCM:MeOH “ 50:1 to 10:1) to give compound 17a as a white solid ( a mixture of two P isomers, 11.0 mg, 11 %). ’H NMR (MeOD, 400 MHz) δΊ.ΊΧ (2d, J= 8.0 Hz, 1 H), 7.17-7.37 (m, 5H), 5.98-6.07 (m, 2H), 5.61,5.68 (2d, 8.0 Hz, IH), 5.53-5.58 (m, IH),
f.35-5.40 (m, IH), 5.08-5.10 (m, IH), 4.93-4.99 (m, IH), 4.52-4.53 (m, IH), 4.16-4.21 (m, IH), < .06-4.11 (m, IH), 3.86-3.94 (m, IH), 1.28-1.34 (m, 3H), 1.20-1.22 (m, 6H). 3,P NMR (MeOD, 162 MHz) £3.72,3.45. ESI-LCMS: m/z 542.0 [M + Hf.
TBDPSO
EXAMPLE 18 Préparation of Compound (18a)
TBDPS (P18-1):
TBSÔ F
P18-1
Xn
TBDPSCr\^O^N_^o 'f
P18-2
P18-3 [0286] Préparation solution of i;hloromethyl)triphenylphosphonium chioride (2.1 g, 6.0 mmol) in anhydrous THF (10 mL) was added dropwise n-BuLi (4.6 mL, 6.0 mmol) at -70°C under nitrogen. The reaction was stirred at Î0°C for 50 mins. A solution of compound P3-9 (950 mg, 1.5 mmol) in anhydrous THF (5 mL) as added at -70°C, and the réaction was stirred at 0°C for 3 hours. The réaction was quenched r saturated aq. NH4CI and extracted with EtOAc. The organic layer was separated, dried and concentrated to give a residue. The residue was purified by column chromatography on silica gel (eluting with PE.EtOAc = 6:1) to give P18-1 as a yellow gum (900 mg, 91.2%).
130 [0287] Préparation of (P18-2): To a solution of compound P18-1 (600 mg, 0.91 rimol) in anhydrous THF (18 mL) was added dropwise n-BuLi (4.7 mL, 10.9 mmol) at -70°C vnder nitrogen. The reaction was stirred at -70°C for 3 hours. The reaction was quenched by iturated aq. NH4CI and extracted with EtOAc. The organic layer was separated, dried and concentrated to give a residue. The residue was purified by column chromatography on silica gel (eluting with PE:EtOAc = 8:1-5:1) to give P18-2 as a white solid (300 mg, 53.0%).
[0288] Préparation of (P 18-3): To a solution of P18-2 (300 mg, 0.44 mmol) in MeOH (10 mL) was added NH4F (1.0 g) at R.T. The reaction was refluxed for 3 hours. After cooling R.T., the mixture was filtered, and the filtrate was concentrated in vacuo. The residue was purified by column chromatography on silica gel (eluting with DCM:MeOH = 50:1-30:1) to give P18-3 as a white solid (135 mg, 78.1%).*H NMR (CDjOD, 400 MHz) £7.84 (d, J= 8.0 Hz, IH), 6.06 (dd, J/ = 1.6 Hz, J2 =19.6 Hz, IH), 5.67 (d, J= 8.4 Hz, IH), 5.18-5.03 (m, IH), 4.50 (dd, Λ = 5.2 Hz, J2 =21.6 Hz, IH), 3.85 (d, J= 12.4 Hz, IH), 3.72 (d, J= 12.4 Hz, IH), 3 .09 (s, IH).
[0289] Préparation of (18a): To a solution of P18-3 (130 mg, 0.5 mmol) in anhydrous THF (4 mL) was added dropwise t-BuMgCl (1.0 mL, 1.0 mmol) at -70°C under nitrogen. The reaction was stirred at R.T. for 30 mins. A solution of phenyl(isopropoxy-Laninyl) phosphorochloridate in anhydrous THF(1M, 0.8 mL, 0.78 mmol) was added at -70°C, !0 a
aid the reaction mixture was stirred at R.T. for 5 hours. The reaction was quenched by
F COOH, and the mixture was concentrated in vacuo. The residue was purified by column c iromatography on silica gel (DCM:MeOH = 60:1) to give compound 18a as a white solid (a lixture of two P isomers, 25 mg, 7.7%). *H NMR (CD3OD, 400 MHz) δ7.64, 7.60 (2d, J= 7.6 z, IH), 7.32-7.36 (m, 2H), 7.16-7.25 (m, 3H), 5.95-6.01 (m, IH), 5.67, 5.62 (2d, J= 8.0 Hz, IH), 5.10-5.25 (m, IH), 4.93-4.97 (m, IH), 4.49-4.59 (m, IH), 4.33-4.42 (m, IH), 4.24-4.29 (m, IH), 3.86-3.94 (m, IH), 3.25,3.22 (2s, IH), 1.28-1.34 (m, 3H), 1.20-1.23 (m, 6H); ESI-MS: m/z 5^40.2 [M + H]+.
131
EXAMPLE 19
NHMMTr
Hô' V
P15-1
Préparation of Compound i 19a)
O [0290] Préparation of ΓΡ19-1): P15-2(1.2 g, 2.2 mmol) was dissolved in dry i cetonitrile (20 mL), and 0.45 M tetrazole (24.0 mL, 11.0 mmol) and 3(bis(diisopropylamino)phosphinooxy)propanenitrile (1.13 g, 3.74 mmol) was added. The reaction mixture was stirred for 1 hour under N2 at R.T. TBDPH (2,7 mL, 15 mmol) was added, î nd the mixture was stirred for 1 hour. The réaction was quenched by Na2S20j solution and c xtracted with EA. The organic layer was dried over Na2SC>4 and concentrated. The residue a/as purified by column on silica gel (DCM:MeOH = 100:1 to 40:1) to give P19-1 as a white solid (759 mg, 52%).
[0291] Préparation of (P19-2): P19-1 (750 mg, 1.14 mmol) was dissolved in s iturated NH3 in MeOH solution. The mixture was stirred for 2 hours at R.T. The solution was concentrated to dryness to give crude P19-2 as a yeliow solid (662 mg, 100%). JH NMR ( 3MSO-rf5,400 MHz) <58.60 (s,lH), 8.28 (s, IH), 7.48 (d, J= 7.6 Hz, IH), 7.12-7.29 (m, 12H),
6.83 (d, 8.8 Hz, 2H), 6.29 (d, J= 7.6 Hz, IH), 5.88 (d, J= 8.8 Hz, IH), 5.10 (d, J=4.8 Hz,
IH), 4.42-4.45 (m, IH), 3.72 (s, 3H), 1.64-1.91 (m, 2H), 1.10-1.13 ( m, 2H), 0.83-0.86 (m, 3H). 3 P NMR (CDjOD, 400 MHz) <5-4.48; Negative-ESI-LCMS: m/z 606 [M - H].
[0292] Préparation of ÎP19-3): P19-2 (292 mg, 0.47 mmol) was co-evaporated with pyridine twice and dissolved in anhydrous DMF (0.5 mL). DIPEA (1.2 mL) was added and
Îltowed by 2,2-dimethyt-propionic acid iodomethyl ester (680 mg, 2.8 mmol). The reaction ixture was stirred at R.T. under N2 for 16 hours. The réaction was quenched by NaiSiOj solution and extracted with EA. The organic layer was dried over Na2SO4 and concentrated.
The residue was purified by column on silica gel (DCM:McOH = 100:1 to 30:1) to give P19-3 as a white solid (95 mg, 30%).
132
10293] Préparation of (19a): P19-3 (95 mg, 0.13 mmol) was dissolved in a 80%
HCOOH aq. solution, and the mixture was stirred at R.T. for 16 hours. The solvent was Acmoved, and the residue was purified by RP HPLC (MeCN and 0.1% HCOOH in water) to give compound 19a as a white solid (10 mg, 17%). ’H NMR (CDjOD, 400 MHz) <77.69 (d, J= 7.2 Hz, IH), 5.91 (d, J= 7.6 Hz, IH), 5.84 (d, 22.0 Hz, IH), 5.73 (d, 14.0 Hz, 2H), 5.52 (d, J
- 5.2 Hz, IH), 5.13-5,22 (m, IH), 4.53-4.61 (m, IH), 4.31 (d, J= 9.6 Hz, IH), 1.92-2.08 (m, 2H), 1.23 (s, 9H), 1.03-1.07 (m, 3H); 31P NMR (CDjOD, 162 MHz) <7-7.93; ESI-LCMS: m/z <50[M + H]+.
EXAMPLE 20
Préparation of Compound (20a)
P20-a
P20-6
TBSO' 'F
P20-7
Hd r
20a [0294| Préparation of (P20-1): To a stirred suspension of P3-1 (20.0 g, 81.3mmol), iraidazole (15.9 g, 234.0 mmol), PPhj (53.5 g, 203.3 mmol) and pyridine (90 mL) in anhydrous
THF (360 mL) was added dropwise a solution of I2 (41.3 g, 162.6mmot) in THF (350 mL) at 'C. After addition, the mixture was warmed to R.T. and stirred for 14 hours. The solution was q icnched with aq. Na2S20j (150 mL) and extracted with EA The organic layer was dried over
133
Na2SO4 and concentrated. The residue was purified on a silica get column (DCM:MeOH = : 00:1 to 10:1) to afford P20-1 as a white solid (22.1 g, 76.4%). *H NMR (CDjOD, 400 MHz) δ >0
7.70 (d, J= 8.0 Hz, IH), 5.88 (dd, Jt = 1.6 Hz, J2 = 20.8 Hz, IH), 5.71 (d, J= 8.4 Hz, IH), 5.24 fdd, Jt = 2.0 Hz, J2 = 5.2 Hz, IH), 5.10 (dd, J, = 2.0 Hz, J2 = 5.2 Hz IH), 3.78-3.83 (m, IH), 7.61-3.65 (m, IH), 3.44 (dd, Jt = J2 = 6.0 Hz, IH).
[0295] Préparation of (P20-2): To a stirred solution of P20-1 (22.1 g, 62.1 mmol) in anhydrous THF (200 mL) was added dropwisc DBU (142 g, 93.1 mmol) in THF (50 mL) at i )°C over 10 mins. The mixture was stirred at 60°C for 6 hours. The reaction was quenched with aq. NaHCOj (200 mL) and extracted with EA. The organic layer was washed with brine and dried over Na2SO4. The solvent was removed, and the residue was purified on a silica gel :olumn (MeOH:DCM = 1/100 to 1/30) to afford P20-2 as a white solid (8.7 g, 61.5%). ’H NMR 'CDjOD, 400 MHz) Ô 7.51 (d, 8.0 Hz, IH), 6.05 (dd, Jt =1.2 Hz, J2 = 17.2 Hz, IH), 5.73 (d,
J = 8.0 Hz, IH), 5.26 (dd, J/ = 1.2 Hz,Λ = 4.8 Hz, 1 H), 5.13 (dd, Jt = 12 Hz, J2 = 4.8 Hz, IH), 1.63 (dd, Jt =2.0 Hz, J2 = 3.2 Hz, lH),4.41(dd,J/=J2 = 2.0Hz, IH).
[0296] Préparation of (P20-3): To a stirred solution of P20-2 (3.2 g, 14.0 mmol) in inhydrous pyridine(l 0 mL) and DCM (100 mL) was added dropwise a solution of TBSC1 (4.2 g, 28.0 mmol)at 0°C. Stirring was continued at R.T. for 18 hours. The mixture was diluted with DCM. The organic layer was washed with brine and dried over Na2SO4. The solvent was removed, and the residue was purified on a silica gel column (10% MeOH in DCM) to afford P20-3 as a white solid (3.4 g, 70.8%).
[0297] Préparation of (P20-4): To a stirred solution of NaHCOj in H2O (250 mL) and acetone (200 mL) was added oxone (30.0 x 4 g) at 0°C. The mixture was warmed to R.T., and the distillate was coliected at -78°C (120 mL) under slightiy reduced pressure to give a solution of DMDO in acetone. To a stirred solution of P20-3 (250.0 mg, 0.7 mmol) in DCM (20 mL) were added a DMDO (120 mL) solution at -40°C and MgSO«. The mixture was warmed to R.T. and then stirred for 2 hours. The solution was filtrated, and the filtrate was used for the next-step directly.
[0298] Préparation of(P20-5): To a stirred solution of P20-4 (500.0 mg, 1.4 mmol) in anhydrous DCM (50 mL) was added allyl-trimethyl-silane (760.0mg, 6.7mmol) and SnCU (12 g, 45 mmol) at -40°C. The mixture was warmed and stirred at 0°C for 1 hour. The reaction was quenched with saturated NaHCOj and extracted with DCM. The organic layer was dried over Na2SO4 and concentrated. The residue was purified on a silica gel column (20-50% EA in
PE) to give P20-5 as a white foam (120 mg, 41%). lH NMR (CDjOD, 400 MHz) δ 8.01 (d, J=
8.4 Hz, IH), 6.12 (dd, Jt = 3.6 Hz, J2 = 152 Hz, IH), 5.87-5.96 (m,lH), 5.71 (d, J = 8.4 Hz,
134 ® 1Η), 5.06-5.22 (m, 3H), 4.60 (dd, J, = 5.6 Hz, J2 - 14.4 Hz, IH), 3.72 (d, J= 11.6 Hz, IH), 3.48 (d, J= 11.6 Hz, IH), 2.62-2.67 (m, IH), 2.23-2.29 (m, 1 H); ESI-LCMS: m/z = 422[M + Na]+.
[0299] Préparation of (P20-6): To a stirred solution ofP20-5 (270.0 mg, 0.7 mmol) ira dry DCM were added imidazole (400.0mg, 5.9mmol) and TBSC1 (390.0 mg, 2.6 mmol) at ÎT. The mixture was stirred at R.T. for 18 hours. The solution was diluted with EA. The lvent was washed with brine and dried over Na2SO4. The solvent was removed, and the sidue was purified on a silica gel column (20-40% EA in PE) to afford compound P20-6 as a white foam (280 mg, 80.7%). ESI-LCMS: m/z 537 [M + Na]+.
[0300] Préparation of (P20-7): To a stirred solution of P20-6 (280.0 mg, 0.5 mmol) in dry MeCN were added TPSCl (350.0 mg, 1.2 mmol), NEt] (400.0 mg, 4.0 mmol) and □MAP (270.0 mg, 2.2 mmol) at R.T. The mixture was stirred at R.T. for 18 hours. The jolution was quenched with ammonium. The organic layer was washed with brine and dried over Na2SO4. The solvent was removed, and the residue was purified by TLC (using EA) to ifford compound P20-7 as a white foam (240.0 mg, 85.7%). ESI-LCMS: m/z 514 [M + H]+.
[0301] Préparation of (P20-8): To a stirred solution of P20-7 (270.0 mg, 0.5 mmol) in dry DCM were added AgNCh (1.5 g, 8.8mmol), MMTrCl (450.0 mg, 1.5 mmol) and collidine £00.0 mg, 4.1 mmol) at R.T. The mixture was stirred at R.T. for 18 hours. The solution was diluted with DCM. The organic layer was washed with brine and dried over Na2SC>4. The solvent was removed, and the residue was purified on a silica gel column (20-40% EA in PE) to afford compound P20-8 as a white foam (300 mg, 81.6%). ESI-LCMS: m/z 786 [M + H]+, [0302[ Préparation of (20a): To a stirred solution of P20-8 (170.0 mg, 0.3 mmol) in dry MeOH was added NH4F (300.0 mg, 8.1 mmol), and the mixture was refluxed for 24 hours. The solvent was removed under reduced pressure, and the residue was purified on a silica gel column (2-5% MeOH ïn DCM) to give the crude product. The crude product was further purified by RP HPLC (water and 0.1% HCOOH in MeCN) to afford compound 20a as a white solid (47.0 mg, 49.8%). ’H NMR (CD3OD, 400 MHz) <5 8.13 (d, J= 8.4 Hz, IH), 6.12 (dd, Jt =
3.2 Hz, J2~ 12.0 Hz, IH), 5.87-5.97 (m, 2H),4.98-5.14 (m, 3H), 4.45 (dd, 5.2 Hz, J2= 17.6 Hz, IH), 3.71 (d, J= 11.6 Hz, IH), 3.54 (d, J= 11.6 Hz, IH), 2.54-2.59 (m, IH), 2.33-2.39 (m, IH); ESI-LCMS: m/z 286 [M + H]*.
135
EXAMPLE 21
Préparation of Compound (21a)
P20-3
NHMMTr
TBS(J 'F
P21-1
[0303] Préparation of ΓΡ21-1): To a stirred solution of P20-8 (250.0 mg, 0.3 mmol) i i MeOH was added Pd/C (500.0 mg), and the mixture was stirred under H2 (balloon) for 18 liours at R.T. The reaction was filtered, and the solvent removed under reduced pressure. The residue was purified by prep. TLC (30% EtOAc in PE) to afford P21-1 as a white foam (210.0 mg, 84.0%).
[0304] Préparation of (P21-2): To a stirred solution of P21-1 (210.0 mg, 0.3 mmol) ii i dry THF was added TBAF (1 mL, Immol), and the mixture was stirred at R.T. for 18 hours. Ïtie solvent was removed under reduced pressure, and the residue was purified by prep. TLC , 0% EtOAc in PE) to give compound 21a as a white foam (111.2 mg, 74.6%). *H NMR ( )MSO-J5,400 MHz) 58.49 (s, IH), 7.75 (d, 6.8 Hz, IH), 6.83-7.32 (m, 14H), 6.25 (d, J = .6 Hz, IH), 5.95 (dd, J, = 4.8 Hz, J2 = 14.8 Hz, IH), 5.48 (d, 5.6 Hz, IH), 4.86-5.15 (m,
, 4.15-4.21 (m, IH), 3.72 (s, 3H), 3.38-3.49 (m, 2H), 1.24-1.58 (m, 4H), 0.84 (t, J- 7.2 Hz, ; ESI-MS: m/z 560 [M + H]+.
[0305] Préparation of (P2D: Compound P21-2 (81 mg) was dissolved in a mixture (5 mL) of formic acid (80%) and water (20%). The resuiting solution was stirred at R.T. for 3 hours and then concentrated. The residue was co-evaporated with methanol/toluene three times. (Chromatography on silica ge! with 5- ! 2% methanol in DCM gave a mixture of two compounds, which was dissolved in methanol with a drop of concentrated aqueous ammonia and c mcentrated. The residue was purified on silica gel with 5-12% methanol in DCM to give compound 21a (27 mg) as a white solid; 1
H NMR (CDjOD, 400 MHz) 58.05 (d, J = 7.6 Hz,
T), 6.06 (dd, Ji = 2.8 Hz, Λ = 16 Hz, IH), 5.87 (d ,7= 7.6 Hz, IH), 5.10 (dd, J- 3.2, 5.2 Hz,
I.5H), 4.96 (dd, 3.2, 5.2 Hz, 0.5H), 4.42 (dd, J= 5.6, 17.2 Hz, IH), 3.67 (dd, J= 11.6, 76 Hz,
136
2H), 1.70-1.79 (m, IH), 1.31-1.61 (m, m, 3H), 0.94 (t, J= 6.8 Hz, 3H). MS: m/z 417 [M + 2methylheptylamïne]+.
EXAMPLE 22
Préparation of Compound (22a)
NHBz
P22-3
P22-4 [0306] Préparation of (Ρ22-1Ί: To a solution of P20-2 (523 g, 23.1 mmol) in t nhydrous MeOH (50 mL) was added PbCO3(12.7 g, 46.3 mmol) at R.T. A solution of I2 (11.7 ί, 46.3 mmol) in MeOH (10 mL) was then added dropwise at 0°C. The reaction mixture was stirred at R.T. for ovemight The reaction was quenched with Na2S2O3 and dissolved in EA. The organic layer was dried over Na2SO4 and concentrated. The residue was purifîed by column (DCM/MeOH = 100/1 to 20/1) to give P22-1 as a white solid (5.6 g, 71.8%). ’H NMR (CD3OD, 400 MHz) J7.67 (d, 8.0 Hz, IH), 5.88 (dd, Jt=J2 = 7.6 Hz, IH), 5.73 (d, J= 8.0
Iz, IH), 5.24 (dd, J2 = 4.4 Hz, J2 = 6.4 Hz, IH), 5.11 (dd, Ji = 6.4 Hz, J2 = 6.0 Hz, 1 H); 4.65 ( id, Jt = 20.0 Hz, J2 = 20.4 Hz, IH), 3.67 (d, J= 11.6 Hz, IH), 3.54 (d, J= 11.6 Hz, IH), 3.43 (3,3H).
[0307] Préparation of (P22-2): To a stirred solution of P22-1 (5.6 g, 14.5 mmol) in a ihydrous pyridine (20 mL) was added dropwise BzCl (2.9 g, 20.9 mmol) at 0°C. The mixture as stirred at R.T. for 10 hours. The reaction was quenched with H2O, and the solution was w
c jncentrated. The residue was dissolved in EA and washed with saturated NaHCCh. The o c ïlumn (20—40% EA in PE) to give P22-2 as a white foam (4.9 g, 74.2%).
rganic layer was dried over Na2SO4 and concentrated. The residue was purifîed on a silica gel
10308] Préparation of (P22-3): P22-2 (4.9 g, 10.0 mmol), BzONa (14.4 g, 100 nmol) and 15-crown-5 (22.0 g, 100 mmol) were suspended in DMF (200 mL). The mixture was stirred at 60-70°C for 3 days. The precipitate was removed by filtration, and the filtrate was
137
diluted with EA. The solvent was washed with brine and dried over Na2SÜ4. The solvent was removed, and the residue was purified on a silica gel column (20-60% EA in PE) to afford P22b as a white foam (2.3 g, 47.9%).
[0309] Préparation of (P22-4): P22-3 (2.3 g, 4.8 mmol), DMAP (1.2 g, 9.6 mmol), ' TSC1 (2.9 g, 9.6 mmol) and EtjN (0.97 g, 9.6 mmol) were suspended in MeCN (10 mL). The mixture was stirred at R.T. for 14 hours. NHj in THF (saturated at 0°C, 100 mL) was added to t le mixture, and the mixture stirred at R.T. for 2 hours. The solvent was removed, and the residue was purified by column (DCM/MeOH = 100:1 to 50:1) to give the crude product (1.2 g), 'lie crude product was dissolved in pyridine, and BzCl (0.42 g, 3.0 mmol) was added. The mixture was stirred at R.T. for 16 hours and quenched with water. The solvent was removed, înd the residue was purified on a silica gel column (PE:EA = 2:1 to 1:1) to give P22-4 as a x zhitc foam (460 mg, 31%).
[0310] Préparation of (22a): P22-4 (0.46 g, 0.8 mmol) was dissolved in saturated riethanolic ammonia (100 mL), and the mixture was stirred at R.T. for 14 hours. The solvent was removed, and the residue was dissolved in H2O and washed with DCM. The aqueous phase was lyophilized and further purified by prep. HPLC (0.1% formic acid in water/acetonitrile) to give compound 22a as a white solid (145 mg, 78.9 %). lH NMR (CD3OD, 400 MHz) «Î7.88 (d,
7.6 Hz, IH), 6.03 (d, J = 18.4 Hz, 1H), 5.87 (d, J= 7.6 Hz, IH), 4.86-5.00 (m, IH), 4.49 (id, = 23.2 Hz, J2 = 22.8 Hz, IH), 3.90 (d, J= 12.0 Hz, IH), 3.66 (d, J= 12.0 Hz, IH), 3.41 ( i, 3H); ESI-MS: m/z 276 [M + H] +.
138
EXAMPLE 23
Préparation of Compound (23a)
TBDPSO-VzCU^
DMTrO—'''V-Z -----*
TBScf F
P23-2 P23-3
P23-1
P23-5
NHMMTr
P23-6
P23-7 23a [0311) Préparation of (P23-2): To a solution of P23-1 (3.1 g, 4.5 mmol) in DMF <30 mL) was added anhydrous K2CO3 (1.24 g, 9.03 mmol) and PMBC1 (1.40 g, 9.03 mmol). rhe mixture was stirred at ambient température ovemight. The reaction was quenched with water and extracted with EA. The organic layer was concentrated, and the residue was purified an a silica gel column (PE:EA - 10:1 to 4:1) to give the intermediate as a white solid (2.36 g, 74.8%). ‘H NMR (CDClj, 400 MHz) <57.29-7.88 (m, 23H), 6.83-6.98 (m, 6H), 6.35-6.45 (m, IH), 4.51-5.50 (m, 6H), 3.89-3.95 (m, 9H), 3.66-3.71 (m, 2H),3.03 (d, J=11.2Hz, IH), 1.21 (s, 9H), 0.89 (m, 9H), 0.01-0.11 (m, 6H). The intermediate was used in the next step.
[0312| To a stireed solution of the intermediate (11.0 g, 10.47 mmol) in anhydrous THF (100 mL) was added TBAF (8.20 g, 31.42 mmol) at R.T., and the mixture was stirred at R.T. for 5 hours. The solution was removed, and the residue was purified on a silica gel column (PE: EA=5:1 to 1 :l) to give a second intermediate as a white solid (5.99 g, 82%).
[0313] To a stirred solution of the second intermediate (500 mg, 0.716 mmol) in anhydrous DMF (10 mL) was added NaH (51.5 mg, 2.14 mmol) and BnBr (365 mg, 2.14 mmol) dropwise at O’C. The mixture was stirred at R.T. for ovemight. The solution was quenched with water and extracted with EA. The concentrated organic phase was purified on a silica gel column (PE:EA = 10:1 to 4: l) to give a third intermediate as a white solid (496 mg, 79%).
139
I5
[0314] The third intermediate (2.5 g, 2.84 mmol) was dissolved in 80% HOAc (25 mL) at R.T., and the mixture was stirred at R.T. for ovemight. The reaction was quenched with MeOH, and the solvent was removed. The crude was purified on a silica gel column (PE:EA = 5:1 to 1:1) to give P23-2 as a white solid (1.2 g, 73%).
[0315] Préparation of (P23-3): To a stirred solution of DAST (1.39 g, 8.68 mmol) in anhydrous toluene (15 mL) was added dropwise a solution of P23-2 (1.0 g, 1.73 mmol) at 78°C. The mixture was stirred at -78°C for 30 mins. The solution was heated to 60qC gradually ind then stirred ovemight. The mixture was poured into saturated Na2CO3 solution. The concentrated organic phase was purified on a silica gel column (PE:EA = 10:1 to 4:1) to give P23-3 as a whitesolid (449 mg, 45%). ’HNMR (CD3OD, 400 MHz) <57.87 (d, J= 8.4 Hz, IH), 7.27-7.37 (m, 12H), 6.82-6.84 (m, 2H), 6.14 (dd, J= 16.8,2.0Hz, IH), 5.18-5.50 (m, 4H), 4.96 (s, 2H), 4.45-4.88 (m, 7H), 3.67-3.89 (m, 5H).
[0316] Préparation of (P23-4): A mixture of P23-3 (1.20 g, 2.07 mmol) and CAN (3.41 g, 6.23 mmol) in a solution of MeCN:Water (3:1, 10 mL) was stirred at R.T. ovemight. Brine (10 mL) was added, and the mixture was extracted with EA. The combined organic extracts were dried and evaporated under reduced pressure. The residue was purification by chromatography on silica gel (PE:EA = 10:1 to 2:1) to give P23-4 as a yellow solid (475 mg, 49,8%).
[0317] Préparation of (P23-5): To a stirred solution ofP23-4 (550 mg,210 mmol) in anhydrous MeCN (10 mL) were added TPSC1 (725 mg, 2.40 mmol), DMAP (293 mg, 2.40 mmol) and TEA (242 mg, 2.40 mmol) at R.T., and the mixture was stirred at R.T. ovemight. NH4OH (25 mL) was added, and the mixture was stirred for 2 hours. The solvent was removed, and the residue was purified on a silica gel column (PE:EA = 8:1 to 2:1) to give P23-5 as a white solid (700 mg crude).’H NMR (CDjOD, 400 MHz) <57.86 (d, J = 8.4 Hz, IH), 7.27-7.36 (m, 10H), 6.13 (dd, Jt = 17.2 Hz, J2 = 2.0 Hz, IH), 5.48-5.53 (m, IH), 5.11-5.26 (m, IH), 4.444.74 (m, 7H), 3.89 (dd, J, = 10.4 Hz, J2 = 2.0 Hz, IH), 3.69 (dd,Λ = 10.8 Hz, J2 =1.6 Hz, IH).
[0318] Préparation of (P23-6): To a stirred solution of P23-5 (1.0 g, 2.18 mmol) in anhydrous DCM (15 mL) was added MMTrCl (2.02 g, 6.56 mmol) and AgNO3 (1.11 g, 6.56 mmol) at R.T., and the mixture was stirred at R.T. ovemight. The solid was filtered off and washed with DCM. The filtrate was washed with brine and dried over Na2SÜ4. The organic phase was concentrated, and the residue was purified on a silica gel column (PE:EA = 8:1 to 2:1) to give P23-6 as a white solid (520 mg, 41%).
]0319] Préparation of (P23-7): To a stirred solution of P23-6 (520 mg, 0.713 mmol) in acetone were added ammonium formate (2.0 g, 31.7 mmol, in portions) and 10%
140 palladium on carbon (1.0 g). The mixture was refluxed for 12 hours. The catalyst was filtered Îf and washed with solvent. The filtrate was added EA and washed with brine. The mcentrated organic phase was purified by column chromatography (DCM:MeOH = 100:1 to !:l)and prep. TLC to give P23-7 as a white solid (270 mg, 69.0%).*H NMR (CDjOD, 400 Hz) <58.54 (s, IH), 7.73 (d, J= 7.6 Hz, IH), 7.13-7.32 (m, 12H), 6.83 (d, J= 8.4 Hz, 2H), 6.29 , J = 7.6 Hz, 1 H), 5,99-6.04 (m, IH), 5.82 (d, J =5.6 Hz, 1H),5.39 (t,J=5.2Hz, lH),5.09(t, .r= 5.2 Hz, IH),4.32-4.58 (m, 3H), 3.54-3.72 (m, 5H). ESI-MS: m/z549.6 [M + H]+.
[0320] Préparation of f23a): P23-7 (130 mg, 0.236 mmol) was dissolved in 80% ICOOH (20 mL) at R.T., and the mixture was stirred at 50°C for 12 hours. The solvent was i emoved, and the residue was co-evaporated with toluene twice. The residue was re-dissolved n MeOH (20 mL) at 60°C and stirring was continued for 48 hours. The solvent was removed, ind the residue was purified by column chromatography (DCM:MeOH = 100:1 to 10:1) to give :ompound 23a as a white solid (45 mg, 69.0%).’HNMR(CDjOD, 400 MHz) δ8.00 (d, J= 7.6 iz, IH), 6.13 (dd, J, = 16.0 Hz, J2 = 4.0 Hz, IH), 5.89 (d, J = 7.6 Hz, IH), 5.18-5.21 (m, IH), 5.05-5.07 (m, IH), 4.60(s, IH),4.51-4.57 (m, 2H), 3.84 (dd, J, =12.0Ηζ,Λ-2.0 Hz, IH), 3.75 (dd, Jt = 12.0 Hz, J2 = 2.0 Hz, IH). ESI-MS: m/z 277.8 [M + H]+, 554.8 [2M + H]+.
141
P24-4
EXAMPLE 24 Préparation of Compound (24a)
TBDPSO
TBDPSO
P24-9
HMMTr
TBDPS
Hd r
P24-12
TBsd
P24-11
TBDPS HO—
TBSO* ΐ
P24-7
TBSÔ 'F
P24-8
TBSCf ΐ
P24-10 [0321) Préparation of (P24-2): To a solution of P24-1 (30.0 g, 100.0 mmol) in pyridine (300 mL) was added BzCl (56.0 g, 400 mmol) at 25°C. The mixture was stirred at 25°C for 15 hours. The mixture was concentrated and purified by column chromatography (PE:EA « 20:1 to 2:1) to give crude P24-2 (55.0 g, 81 %).
[0322) Préparation of (P24-3): P24-2(55.0 g, 92 mmol) was dissolved in 80%
HOAc aq. solution, and the mixture was refluxed for 14 hours. The solvent was removed under reduced pressure, and the residue was co-evaporated with toluene. The residue was purified on a silica gel column (PE/EA = 4:1 to 2:1) to give P24-3 as a white solid (39.2 g, 83%).
142 [0323] Préparation of (P24-4): P24-3 (39.2 g, 83 mmol) was dissolved in saturated nethanolic ammonia, and the resulting solution was stirred at R.T. for 15 hours. The solvent was removed, and the residue was purified on a silica gel column (DCM/MeOH = 50:1 to 20:1) tDgiveP24-4 (21.0 g, 95.8%).
[0324] Préparation of (P24-5): To a solution of P24-4 (21.0 g, 79.5 mmol) in pyridine (250 mL) was added DMTrCl (28.2 g, 83.5 mmol) at 0°C. The solution was stirred at
LT. for 15 hours. The reaction was quenched with MeOH and concentrated to dryness under i educed pressure. The residue was dissolved in EtOAc and washed with water. The organic ayer was dried over Na2SO4 and concentrated. The residue was dissolved in DCM (300 mL). midazole (13.6 g, 200 mmol) and TBSC1 (30.0 g, 200 mmol) were added. The reaction mixture vas stirred at R.T. for 12 hours. The réaction mixture was washed with NaHCOj and brine. The organic Iayer was dried over Na2SO4 and concentrated. The residue (48.5 g, 79.5 mmol) was dissolved in 80% HOAc aq. solution (400 mL). The mixture was stirred at R.T. for 20 iouts. The mixture was diluted with EtOAc and washed with NaHCOj solution and brine. The Drganic layer was dried over Na2SO4 and purified by silica gel column chromatography (1-2% MeOH in DCM) to give P24-5 as a white solid (21.0 g, 70%). *H NMR (400 MHz, MeOD) â
7.83 (d, J= 8.0 Hz, 1H), 6.14 (dd, J/ = 6.0 Hz, J2 = 10.0 Hz, 1H), 5.73 (d, J= 8.4 Hz, 1H), 4.384.46 (m, 1H), 3.89-3.91 (m, 1H), 3.88 (dd, Jt = 2.8 Hz, J2 = 5.2 Hz, 1H), 3.72 (dd, J, = 2.8 Hz, J2 = 5.2 Hz, 1H), 0.93 (s, 9H), 0.15 (m, 6H). ESI-MS: m/z 379.1 [M + H]+.
[0325] Préparation of (P24-6): To a solution of P24-5 (21.0 g, 55.6 mmol) in anhydrous CH3CN (200 mL) was added IBX (17.1 g, 61.1 mmol) at R.T. The réaction mixture was rcfluxed for 1 hour and then cooled to 0°C. The precipitate was filtered off, and the filtrate was concentrated to give the aldéhyde as a yellow solid (21.0 g, 55.6 mmol). To a solution of the aldéhyde (21.0 g, 55.6 mmol) in dioxane (200 mL) were added 37% CH2O (22.2 mL, 222.4 mmol) and 2N NaOH aq. solution (55.6 mL, 111.2 mmoi). The mixture was stirred at R.T. for 2 hours and then neutralized with AcOH to pH = 7. To the reaction were added EtOH (50 mL) and NaBH4 (12.7 g, 333.6 mmol). The mixture was stirred at R.T. for 30 mins. The reaction was quenched with saturated aq. NH4C1. extracted with EA. The organic layer was dried over Na2SÛ4 and concentrated. The residue was purified by silica gel column chromatography (1-3% MeOH in DCM) to give P24-6 as a white solid (13.5 g, 59.5%).
[0326] Préparation of (P24-7): To a solution of P24-6 (13.5 g, 33.1 mmol) in DCM (100 mL) were added pyridine (20 mL) and DMTrCl (11.2 g, 33.1 mmol) at 0°C. The solution was stirred at 25 °C for 3 hours, and then treated with MeOH (30 mL). The solvent was removed, and the residue was purified by silica gel column chromatography (DCM:MeOH 143
300:1 to 100:1) to give a residue. The residue was dissolved in anhydrous pyridine (150 mL)
and TBDPSC1 (16,5 g, 60 mmol) and AgNOj (10.2 g, 60 mmol) were added. The mixture was stirred at 25°C for 15 hours, and then filtered and concentrated. The mixture was dissolved in îtOAc and washed with brine. The organic layer was dried over Na2SO4. Purified by silica gel column chromatography (DCM:MeOH = 300:1 to 100:1) gave the product as a yellow solid
16.2 g, 85.3%). The solid was dissolved in 80% HOAc aq. solution (400 mL). The mixture vas stirred at R.T. for 15 hours. The mixture was diluted with EtOAc and washed with ' 4aHCO3 solution and brine. The organic layer was dried over Na2SO4 and purified by silica gel column chromatography (DCM:MeOH = 200:1 to 50:1) to give P24-7 as a white solid (9.5 g, : 16.5%).‘H NMR (CD3OD, 400 MHz) 57.39-7.70 (m, 11Η), 6.34-6.38 (m, IH). 5.12 (d, J= 8.0 Hz, IH), 4.79 (dd, J} = 10.0 Hz, J2 = 16.0 Hz, IH), 4.14 (dd, Jt = 1.6 Hz, J2 = 11.6 Hz, IH), 3.48-3.84 (m, 2H), 3.49 (dd, J/ » 1.6 Hz, J2 = 11.6 Hz, 1H),1.12 (s, 9H). 0.92 (s, 9H). 0.16 (s, 3H).
[0327] Préparation of (P24-8): To a solution of P24-7 (6.0 g, 9.3 mmol) in anhydrous DCM (80 mL) was added Dess-Martin periodinane (7.9 g, 18.6 mmol) at 0°C under nitrogen. The reaction was stirred at R.T. for 1 hour. The solvent was removed in vacuo, and i he residue was triturated with diethyl ether (50 mL). The mixture was filtered through a pad of 14gSÛ4, and the organic solvent was stirred with an equal volume of Na2S2O3.5H2O in saturated : 'iaHCCh (50 mL) until the organic layer became clear (approx. 10 min). The organic layer was separated, washed with brine, and dried over MgSC>4. After concentration in vacuo, P24-8 was obtained as a red solid (5.8 g.98%).
[0328] Préparation of (P24-9): To a mixture of methyltriphenylphosphonium bromide (9.6 g, 27.0 mmol) in anhydrous THF (60 mL) was added n-BuLi (10.8 mL, 27.0 i nmol) at -70°C under nitrogen. The reaction was stined at 0°C for 30 mins. A solution of P2411 (5.8 g, 9.0 mmol) in anhydrous THF (20 mL) was added dropwise at 0°C under nitrogen. The i eaction was stirred at R.T. for 12 hours. The reaction was quenched with NH4CI and extracted vith EtOAc. The organic layer was separated, dried and concentrated, and the residue was ])urified by silica gel column chromatography (DCM:MeOH = 300:1 to 100:1) to give P24-9 as 1 l white solid (3.0 g, 51%).
[0329] Préparation of (P24-10): To a solution of P24-9 (2.9 g, 4.5 mmol) in anhydrous MeOH (20 mL) was added Pd/C (1.4 g) at 25°C under hydrogen atmosphère. The mixture was stirred at 25°C for 1 hour. The solution was filtered, evaporated to dryness and purified on a silica gel column (DCM:MeOH =3 00:1 to 100:1) to give P24-10 as a white solid
12.3 g, 79.3%).
144
[0330] Préparation of (P24-11): To a solution of P24-10 (1.0 g, 1.55 mmol) in anhydrous CHjCN (20 mL) were added TPSC1 (940 mg, 3.1 mmol), DMAP (380 mg, 3.1 mmol) and NEtj (470 mg, 4.6 mmol) at R.T. The reaction was stirred at R.T. for 5 hours. NH4OH (8 mL) was added, and the reaction was stirred for 1 hour. The mixture was diluted with DCM 6150 mL) and washed with water, 0.1 M HCl and saturated aq. NaHCOj. The solvent was removed, and the residue was purified by silica gel column chromatography (PE:EA = 10:1 to : :1) to give the crude product as a yellow solid (900 mg, 90 %). To a solution of the crude product in DCM (10 mL) were added MMTrCl (930 mg, 3.0 mmol), AgNOj (510 mg, 3.0 mmol) and colliding (720 mg, 6.0 mmol) at R.T. The reaction was stirred for 12 hours at R.T. {lie reaction was filtered, concentrated and purified by silica gel column chromatography
DCM:MeOH=200:l to 50:l)to give P24-11 as a yellow solid (1.1 g, 77.6%).
[0331| Préparation of (P24-12): To a solution of P24-11 (1.1 g, 1.2 mmol) in VleOH (40 mL) was added NH4F (1.0 g, 30 mmol) at 25°C and stirred at 70°C for 15 hours. The solution was filtered and evaporated to dryness, and the residue was purified by silica gel rolumn (DCM:MeOH = 200:1 to 20:1) to give P24-12 as a white solid (450 mg, 66.6%). ’H MR (400 MHz, MeOD) £8.58 (s, IH), 7.62 (d, J= 7.6 Hz, IH), 7.13-7.30 (m, 12H), 6.83-6.85 ;m, 2H), 6.29 (d, 7.6 Hz, IH), 6.18 (d, J= 6.0 Hz, IH), 5.94 (t, J= 8.0 Hz, IH), 5.22 (t, J =
5.2 Hz, IH), 4.28-4.37 (m, IH), 3.72 (s, 3H), 3.57-3.62 (m, IH), 1.39-1.60 (m, 2H), 0.79-0.84 (m, 3H). ESI-LCMS: m/z 563.6 [M + H]+.
[0332| Préparation of (24a): P24-12 (250 mg, 0.44 mmol) was dissolved in 80% HCOOH in HjO (6.0 g) at 25°C. The mixture was stirred at 35°C for 15 hours. The solution was evaporated to dryness, dissolved in MeOH (30 mL) and stirred at 60°C for 12 hours. The solution was evaporated to dryness and purified by silica gel column chromatography (DCM:MeOH = 100:1 to 100:1) to give compound 24a as a white solid (125.6 mg, 97%). ’H NMR (400 MHz, MeOD) £7.91 (d, J = 7.6 Hz, 1 H), 6.19 (t, J = 7.6 Hz, IH), 5.90 (d, J= 7.2 Hz, IH), 4.47 (t, J=13.6 Hz, IH), 3.67 (d, J= 12.0 Hz, IH), 3.52 (d, J= 12.0 Hz, IH), 1.73-1.82 (m, IH), 1.53-1.63 (m, IH), 095 (t, J =7.6Hz, 3H). ESI-LCMS: m/z 291.9 [M + H]+.
145
EXAMPLE 25
Préparation of Compound (25a)
P25-3 P25*4
P25-7 P25-8
25a [0333] Préparation of (P25-2): To a solution of P25-1 (20.0 g, 70.16 mmol) in anhydrous pyridine (200 mL) was added imidazole (19.08 g, 280.7 mmol) and TBSC1 (42.10 g, 280.7 mmol) at 25°C. The solution was stirred at 25°C for 15 hours, and then concentrated to dryness under reduced pressure. The residue was washed with EtOAc to give the crude product as a white solid (36.4 g). The crude product was dissolved in THF (150 mL) and H2O (100 mL), and then HOAc (300 mL) was added. The solution was stirred at 80°C for 13 hours. The reaction was cooled to R.T., and the mixture was concentrated to dryness under reduced pressure. The residue was dissolved washed with EtOAc and dried to give P25-2 as a white solid (31.2 g, 60.9%).
[0334] Préparation of (P25-31: To a stirred solution of P25-2 (31.2 g, 78.2 mmol) in anhydrous pyridine (300 mL) was added Ac2O (11.96 g, 117.3 mmol). The mixture was stirred at 25°C for 18 hours. MMTrCl (72.3 g, 234.6 mmol) and AgNO3 (39.9 g, 234.6 mmol)
146
vere then added. The solution was stirred at 25°C for 15 hours. And H2O was added to quench i he reaction. The solution was concentrated to dryness under reduced pressure. The residue was dissolved in EtOAc and washed with water. The organic layer was dried over Na2SÛ4 and : ïltered. The fiitrate was concentrated in vacuo to give a residue. The residue was purified by i itica gel (DCM:MeOH - 200:1 to 50:1) to give the product. The product was dissolved in NHs/MeOH (300 mL), and the mixture was stirred at 25°C for 20 hours. The solvent was removed, and the residue was purified on a silica gel column (DCM:MeOH = 100:1 to 50:1) to 1 ïive P25-3 as a yellow solid (28.6 g, 86.5 %). *H NMR (400 MHz, MeOD) <58.01 (s, IH), 7.237.35(m, 12H), 6.85-6.87 (m, 2H), 5.60 (dd, Jt = 11.2 Hz, J2 = 5.6 Hz, IH), 4.78-4.94 (m, IH), 4.44 (dd, J/ = 8.0 Hz, J2 = 4.8 Hz, IH), 3.78 (s, 3H), 3.60-3.63 (m, IH), 3.50 (dd, Jt = 32.0 Hz, 12.0 Hz, 2H), 3.32 (s, 3H), 0.94 (s, 9H), 0.12-0.14 (m, 6H).
[0335] Préparation of (P25-41: To a solution of P25-3 (7.24 g, 10.79 mmol) in anhydrous CHjCN (100 mL) was added IBX (3.93 g, 14.03 mmol) at 20°C. The reaction mixture was refluxed at 90°C for 1 hour. The reaction was filtered, and the fiitrate was < :oncentrated to give the aldéhyde as a yellow solîd (7.1 g). To a solution of the aldéhyde (7.1 g, .0.6 mmol) in dioxane (80 mL) was added 37% CH2O (4.2 mL, 42.4 mmol) and 2N NaOH aq. solution (8.0 mL, 15.9 mmol). The mixture was stirred at 25°C for 2 hours and then neutralized vith AcOH to pH = 7. To reaction was added EtOH (30 mL) and NaBfy (2.4 g, 63.6 mmol), he reaction was then stirred for 30 mins. The mixture was quenched with saturated aq. NH4CL The mixture was extracted with EA, and the organic layer was dried over Na2SO4. The solvent vas removed, and the residue was purified by silica gel column chromatography (DCMiMeOH = 200:1 to 50:1) to give P25-4 as a yellow solid (4.86 g, 65.4%).
[0336] Préparation of (P25-5): To a solution of P25-4 (3.8 g, 5.4 mmol) in DCM 40 mL) were added pyridine (10 mL) and DMTrCl (1.8 g, 5.4 mmol) at 0°C. The solution was itirred at 25°C for I hour. The reaction mixture was treated with MeOH (15 mL) and :oncentrated. The residue was purified by silica gel column chromatography (DCM:MeOH = 200:1 to 50:1) to give the mono-DMTr protected intermediate as a yellow solid (3.6 g, 66.4 %). To a solution of the intermediate in anhydrous pyridine (30 mL) were added TBDPSC1 (2.96 g,
10.8 mmol) and AgNOj (1.84 g, 10.8 mmol). The mixture was stirred at 25°C for 15 hours. The nixture was filtered and concentrated, and then dissolved in EtOAc and washed with brine. The jrganic layer was dried over Na2SO4, and then concentrated. The residue was purified by silica gel column chromatography (DCM:MeOH = 200:1 to 50:1) to give the pure intermediate as a white solid (3.8 g, 85.1%). To a solution of the intermediate (3.6 g, 2.9 mmol) in anhydrous DCM (50 mL) was added C12CHCOOH (1.8 mL) in anhydrous DCM (18 mL) at -78°C. The
147
mixture was stirred at -10°C for 30 mins. The mixture was quenched with saturated aq.
NaHCOj and extracted with DCM. The organic layer was dried over Na2SO4, and then purified by silica gel column chromatography (DCM:MeOH = 200:1 to 50:1) to give P25-5 as a white iolid(2.2 g, 80.7%).
[0337] Préparation of (P25-6): P25-5 (2.2 g, 2.3 mol) was added to a suspension of Dess-Martin periodinane (2.5 g, 5.8 mol) in anhydrous CH2CI2 (30 mL) at 25°C. The mixture was stirred at 25°C for 4 hours. The solvent was removed in vacuo, and the residue triturated with diethyl ether (30 mL). The mixture was filtered through a pad of MgSOj. The organic solvent was stirred with an equal volume of Na2S2Oj.5H2O in saturated NaHCOj (30 mL) until t le organic layer became clear (approx. 10 min). The organic layer was separated, washed with brine, and dried over MgSCL. The solvent was removed in vacuo to give P25-6 as a yellow solid (2.1 g, 95%). · [0338] Préparation of (P25-7): To a stirred solution of methyl-triphenylphosphonium bromîde (2.3 g , 6.6 mmol) in anhydrous THF (30 mL) was added dropwise nI-luLi (2.6 mL, 6.6 mmol, 2.5 M in THF) at -78°C over 1 minute. Stirring was continued at 0°C /or 1 hour. P25-6 (2.1 g, 2.2 mmol) was added to the mixture, and then stirred at 25°C for 15 hours. The reaction was quenched with saturated NH4CI (50 mL). The mixture was extracted mth EtOAc. The combined organic phase was dried with Na2SO4, filtered and evaporated to aryness to give a light yellow oil. The oil was purified by column chromatography (DCM:MeOH = 200:1 to 50:1) to give P25-7 as a white solid (1.6 g, 76%).
[0339] Préparation of (P25-8): To a solution of P25-7 (1.6 g, 1.7 mmol) in MeOH (50 mL) was added NH4F (1.5 g, 40 mmol), and the mixture was stirred at 70°C for 15 hours. ' lie solution was filtered and evaporated to dryness. The residue was purified by silica gel column (DCM:MeOH = 200:1 to 20:1) to give P25-8 as a white solid (450 mg, 49%). ’H NMR (400 MHz, MeOD) £7.95 (s, IH), 7.21-7.33 (m, 12H), 6.82-6.84 (m, 2H), 5.92 (dd, J/ = 11.2 Hz, J2 = 17.6 Hz, IH), 5.55-5.59 (m, IH), 5.18-5.31 (m, 2H), 4.54-4.68 (m, IH), 4.26-4.33 (m,
H), 3.76 (s, 3H), 3.43 (dd, 12.4 Hz, J2= 36.4 Hz, 2H). ESI-LCMS: m/z 584.1 [M + H]+.
[0340] Préparation of (25a): P25-8 (130 mg, 0.22 mmol) was dissolved in 80% ] 1COOH and the mixture was stirred at 25°C for 1 hour. Then the solution was evaporated to aryness. The residue was dissolved in MeOH (30 mL) and stirred at 60°C for 12 hours. Then the solution was evaporated to dryness, and the residue was washed by EtOAc to give P25 as a white solid (52.3 mg, 76%)?H NMR (400 MHz, MeOD)£8.03 (s, IH), 6.17 (dd, Jt = 32 Hz, J2 » 16.8 Hz, IH), 6.03 (dd, Jt = 11.2 Hz, J2 = 17.2 Hz, IH), 5.50 (dd, J, =1.6 Hz, J2 = 17.2 Hz,
148
: H), 523-5.38 (m, 2H),4.76 (dd, Jt = 4.8 Hz, J2 = 18.0 Hz, IH), 3.60 (dd, J2 = 12.0 Hz, J2 = < 4.8 Hz, 2H). ESI-MS: m/z 334.1 [M + Na]+.
EXAMPLE 26
Préparation of Compound (26a)
P25-6
TBDPS
P26-1
TBDPS
0=''·
TBSÔ
P26-2
P26-3
26a [0341] Préparation of (P26-1): To a stirred solution of P25-6 (2.1 g, 2.2 mmol) in ])yridine was added HONH2.HCt (0.61 g, 8.8 mmol) at 25°C. The mixture was stirred at 25°C ‘or 2 hours. The mixture was concentrated, and the residue was purified by column < :hromatography (DCM:MeOH = 200:1 to 50: t) to give P26-1 as a white solid (1.8 g, 83%).
[0342] Préparation of (P26-2): To a stirred solution of P26-1 (1.4 g, 1.47 mmol) in DCM were added TEA (0.44 g, 4.4 mmol) and methanesulfonyl chloride (0.34 g, 2.9 mmol) at )°C. The mixture was stirred at 25°C for 1 hour. The mixture was quenched with saturated aq. 'iaHCOj and extracted with DCM. The organic phase was dried with Na2SO4, filtered and evaporated. The residue was purified by column chromatography (DCM:MeOH = 200:1 to 50:1) to give P26-2 as a white solid (1.1 g,79%).
[0343] Préparation of (P26-3): To a solution of P26-2 (1.1 g, 1.18 mmol) in MeOH mL) was added NH4F (1.5 g, 40 mmol), and the mixture was stirred at 70°C for 15 hours.
rhe solution was filtered and evaporated to dryness. The residue was purified by silica gel :olumn (DCM:MeOH = 200:1 to 20:1) to give P26-3 as a white solid (400 mg, 71%). ‘H NMR [400 MHz, MeOD) £7.80 (s, IH), 7.20-7.32 (m, 12H), 6.86-6.88 (m, 2H), 5.82 (dd, J/ = 2.0 Hz,
J2=20.0Hz, IH), 4.51-4.66 (m, IH), 3.94 (dd, J, = 5.2 Ηζ,Λ= 20.8 Hz, IH), 3.78 (s, 3H),3.56 [dd, J/= 12.4 Hz, = 42.0 Hz, 2H). ES1-LCMS: m/z 583.1 [M + H]+.
149
[0344] Préparation of (26a): P26-3 (200 mg, 0.34 mmol) was dissolved in 80%
1COOH aq. solution. The mixture was stirred at 25°C for 1 hour. The solution was evaporated o dryncss, dissolved in MeOH (30 mL) and stirred at 60°C for 12 hours. The solvent was lemoved, and the residue was washed by EtOAc to give compound 26a as a white solid (100.4 Aig, 95%). *HNMR(400MHz, MeOD) <57.90(s, IH), 6.34 (dd, J/ = 2.0 Hz, J2 = 19.6 Hz, IH), 1.49 (ddd, Λ = 1.6 Hz, J2 ~ 4.4 Hz, Js = 52.4 Hz, IH), 5.01 (dd, J, = 4.8 Hz, J2 = 20.8 Hz, IH), : .93 (dd, Jj - 12.4 Hz, J2 = 44.8 Hz, 2H). ESI-MS: m/z 311.1 [M + H]+.
EXAMPLE 27
Préparation of Compound (27a)
P25-6
[0345] Préparation of (P27-1): To a stirred solution of chloromethyl-triphenylîosphonium chioride (1.9 g , 5.4 mmol) in anhydrous THF (30 mL) was added dropwise nuLi (2.16 mL, 5.4 mmol, 2.5 M in THF) at -78°C over 10 mins. Stirring was continued at ’C for 2 hours. P25-6 (1.7 g, 1.8 mmol) was added, and the mixture and stirred at 25°C for 15
P
E
7S°i hiurs. The reaction was quenched with saturated NH4Ct (50 mL). The mixture was extracted v ith EtOAc. The combined organic phase was dried with Na2SO4, filtered and evaporated to dryness to give a light yeliow oil.
(DCMiMeOH = 200:1 to 50:1) to give P27-1 as a white solid (1.2 g, 70%).
[0346] Préparation of (P27-2): To a stirred solution of P27-1 (1.2 g, 1.3 mmol) in
The oit was purified by column chromatography anhydrous THF (20 mL) was added dropwise n-BuLi (8.0 mL, 20 mmol, 2.5 M in THF) at -
S°C over 10 minutes. Stirring was continued at -78°C for 4 hours. The reaction was quenched
150 with saturated NH4CI (50 mL). The mixture was extracted with EtOAc (50 x 2 mL). The combined organic phase was dried over Na2SO4f filtered and evaporated to dryness. The residue was purified by column chromatography (DCM:MeOH = 200:1 to 50:1) to give P27-2 as a white solid (1.0 g, 83%).
[0347] Préparation of (P27-3): To a solution of P27-2 (1.0 g, l.l mmol) in MeOH (40 mL) was added NH4F (1.5 g, 40 mmol), and the mixture was stirred at 70°C for 25 hours. rhe solution was filtered, and the filtrate was evaporated to dryness. The residue was purified on a silica gel column (DCM:MeOH = 200:1 to 20:1) to give P27-3 as a white solid (240 mg, J8%). *H NMR (400 MHz, MeOD) <77.85 (s, IH), 7.21-7.31 (m, 12H), 6.84-6.87 (m, 2H), 5.67 Idd, Ji =1.6 Hz, J2 = 19.2 Hz, IH), 4.47-4.62 (m, IH), 3.94 (dd, Jt = 5.2 Hz, J2 = 22.4Hz, IH), J.77 (s, 3H), 3.56 (dd, J, = 12.4 Hz, J2 = 47.2 Hz, 2H), 3.04 (s, IH). ESI-LCMS: m/z 582.1 [M hH]+.
[0348] Préparation of (27a): P27-3 (130 mg, 0.22 mmol) was dissolved in 80% iCOOH aq. solution. The mixture was stirred at 25°C for 1 hour. The solution was evaporated : o dryness. The residue was dissolved in MeOH (30 mL) and stirred at 60°C for 12 hours. The vivent was removed, and the residue was washed with EtOAc to give compound 27a as a white !olid (43.0 mg, 63%). *H NMR (400 MHz, MeOD) <77.95 (s, IH), 622 (dd, Jt = 2.4 Hz, J2 = 8.4Hz, IH), 5.49 (ddd, Jt = 2.0 Hz, J2 = 4.8 Hz, Jj = 532 Hz, IH), 4.77 (dd, Jt ~ S2 Hz, J2 = 20.0 Hz, IH), 3.79 (dd, J, = 12.4 Hz, J2 = 46.8 Hz, 2H), 3.12 (s, 3H). ESI-MS: m/z 310.1 [M + Hf.
151
EXAMPLE 28
Préparation of Compound (28a)
P28-3 P28-4
P28-6
[0349] Préparation of (P28-1): To a stirred solution of P25-1 (5.7 g. 20 mmol) ïn jnhydrous pyridine (20 mL) was added dropwise Ac2O (5,8 mL, 60 mmol) at 0°C. The mixture was stirred at R.T. for 10 hours. AgNO3 (8.5 g, 50 mmol) and MMTrCl (15.5 g, 50 mmol) were î dded. The mixture was stirred at R.T. for 10 hours. The solution was quenched with saturated NaHCOj and extracted with EA. The organic layer was dried over Na2SO4 and concentrated. ''he residue was purified on a silica gel column (DCM/MeOH = 100:1 to 50:1) to afford the intermediate as a light yellow solid (12.1 g, 93.4%). The solid was treated with saturated NH3 in MeOH at R.T. for 14 hours. The solvent was removed, and the residue was purified by silica gel column chromatography (DCM/MeOH — 80:1 to 30:1) to afford P28-1 as a white solid (9.2 g, 87.5%).
[0350] Préparation of (P28-2); To a stirred solution of P28-1 (9.2 g, 16.5mmol) în (iy THF (300 mL) were added imidazole (9.0 g, 132 mmol) and PPh3 (34.8 g, 132 mmol). A solution of I2 (26.0 g, 103 mmol) in THF (100 mL) was added dropwise under N2 at 0°C. The mixture was stirred at R.T. for 18 hours. The reaction was quenched with Na2S2O3 solution, and (he mixture was extracted with EtOAc. The organic layer was dried over Na2SO4 and
152
concentrated. The residue was purified by silica gel column chromatography (DCM/MeOH i 0:1 to 30: l)to give P28-2 as a light yellow solid (10.3 g, 93.4%).
I0351J Préparation of (P28-3): To a stirred solution of P28-2 (10.2 g, 15.3 mmol) i a dry THF (300 mL) was added DBU (4.7 g, 30.1 mmol). The mixture was stirred at 60°C for 8 fours. The solution was diluted with NaHCOj solution and extracted with EtOAc. The organic layer was dried over Na2SÛ4 and concentrated. The residue was purified by silica ge! column chromatography (ΡΕ/EtOAc = 3:1 to 1:3) to afford P28-3 as a light yellow foam (6.2 g, 75.6 %). 1H NMR (CD3OD, 400 MHz) <J7.71(s, IH), 7.23-7.76 (m, 14H), 6.74 (d, 0.8 Hz, 2H), 5.83-
5.88 (dd, Jt = 2.8 Hz, J2 = 16.0 Hz, 2H), 4.57-4.89 (m, 2H), 4.30-4.35(m, IH), 4.79 (s, 3H). ESI4S: m/z 540 [M + H]+.
[0352] Préparation of (P28-4): To a stirred solution of P28-4 (5.42 g, 10 mmol) in inhydrous CHjOH (100 mL) were added PbCOj (13.7 g, 53. Immol) followed by a solution of I2 '12.3 g, 48.9 mmol) in CH3OH (300 mL) at 0°C. The mixture was stirred at R.T. for 10 hours. The solution was quenched with a Na2S2O3 solution and extracted with DCM. The organic layer was washed with NaHCO3 solution, dried over Na2S(>4 and concentrated. The residue was purified by pre-HPLC (MeCN and 0.1% HCOOH in water) to give the pure product as a white foam (2.4 g, 34 %). The product was dissolved in dry pyridine (20 mL) and BzCl (723 mg, 5.2 mmol) was added dropwise at 0°C. The mixture was stirred at 0°C for 1 hour. The solution was quenched with NaHCO3 solution, and extracted with EtOAc. The organic layer was dried over Na2SÛ4 and concentrated. The residue was purified by silica gel column chromatography (ΡΕ/EtOAc = 5:1 to 1:1) to afford P28-4 as a white solid (2.1 g, 77.1 %).
[0353] Préparation of (P28-5): P28-4 (2.0 g, 2.5 mmol), BzONa (3.6 g, 25 mmol) and 15-crown-5 (5.5 g, 25 mmol) were suspended in DMF (50 mL). The mixture was stirred at 11O-125°C for 5 days. The precipitate was removed by filtration, and the filtrate was diluted with EA. The solution was washed with brine and dried over Na2SO4. The solvent was removed, and the residue was purified on a silica gel column (PE/EA = 10/1 to 2/1) to afford crude P28-5 as a light yellow foam ( 1.6 g, 80%).
[0354] Préparation of (P28-6): P28-5 (1.6 g, 2.0nuno!) was dissolved in methanolic ammonia (100 mL, saturated), and the mixture was stirred at R.T. for 20 hours. The solvent was removed, and the residue was purified on a silica gel column (DCM/MeOH =100:1 to 20:1) to give P28-6 as a white solid (410 mg, 34.9%). lH NMR (400 MHz, MeOD) <J7.84 (s,
IH), 7.20-7.33 (m, 12H), 6.83-6.86 (m, 2H), 5.64 (dd, Jt = 1.6 Hz, J2 = 18.4 Hz, IH), 4.46-4.62 (m, IH), 4.08 (dd, Jt= 6.0 Hz, J2= 22.0 Hz, IH), 3.76 (s, 3H), 3.58 (dd, J/= 12.4 Hz,J2= 30.4
Hz, 2H), 3.31 (s, 3H). ES1-LCMS: m/z 588.1 [M + Hf.
153 [0355] Préparation of (28a): P28-8 (200 mg, 0.34 mmol) was dissoived in 80% HCOOH and the mixture was stirred at 25°C for 1 hour. The solution was evaporated to dryness, and the residue was dissoived in MeOH (30 mL) and stirred at 60°C for 12 hours. The solvent was removed, and the residue washed with EtOAc to give compound 28a as a white solid (46.1 mg, 43%). 'H NMR (400 MHz, MeOD) £7.92 (s, IH), 6.22 (dd, Jt = 1.6 Hz, J2 =
18.8 Hz, IH),5.25 (ddd, Js = 1.6 Hz, J2 = 6.0 Hz, J3 = 54.0 Hz, 1 H), 4.89-4.91 (m, lH),3.87(d» 7=11.6 Hz, IH), 3.67 (d, J= 12.0 Hz, IH), 3.44 (s, 3H). ESI-MS: m/z 316.1 [M + H]+.
EXAMPLE 29
Préparation of Compound (29a)
O [0356] DEAD (40% in toluene, 0.15 mL, 0.33 mmol) was added to a stirred solution of triphenylphosphine (78 mg, 0.3 mmol) in anhydrous 1,4-dioxane (0.5 mL) at 0°C under argon. The mixture was warmcd up to R.T. and compound 10a (26 mg, 0.1 mmol) and bis(pivaloyloxymethyl)phosphate (98 mg, 0.3 mmol) were added. The resulting mixture was stirred at 65°C for 3 days. Diisopropylethylamine (50 pL) was added, and the mixture was stirred at 70°C for 3 days. Another reaction of the same scale was conducted separately. The two reaction mixtures were combincd and concentrated. Chromatography on silica gel with 510% methanol in DCM gave the desired product (20 mg) with a minor impurity. A second chromatography on silica gel, followed by RP HPLC with acetonitrile/water, gave the compound (2.8 mg ) as a coloriess residue; ’H NMR (CD3OD, 400 MHz) £7.65 (d, J = 8.0 Hz, 1H), 5.94 (dd,Λ = 2.4 Hz, 18.8 Hz, IH), 5.70 (d, 8.0 Hz, IH), 5.69 (d, J= 0.8 Hz, IH),
5..68 (s, IH), 5.654 (d, J= 1.2 Hz, IH), 5.650 (s, IH), 5.21 (dd, 2.0,5.2 Hz, 0.5H), 5.07 (dd, 2.0, 5.2 Hz, 0.5H), 4.42 (dd, J= 5.6, 20.8 Hz, IH), 4.14 (m, 2H), 1.223 (s, 9H), 1.220 (m, 9H); 3,P NMR (CD3OD) 4.92 (s); MS: m/z 698 [M + 2-methylheptylamine]+.
154
EXAMPLE 30
Préparation of Compound (30a)
NMMT
C,-A_7 HÔ' V
8-oh r° °γ° Ύ ει,Ν
ΒΟΡ-α, DI PEA, NT THF;0°C; 90 min
1-1
[0357] Préparation of H-2): To a solution of 1-1 (313 mg; 0.55 mmol) in THF (8 i nL) under Ar was added a solution of triethylammonium bÎs(POM)phosphate in THF (prepared rom bis(POM)phosphate (215 mg ; 1.2 equiv), THF (2 mL) and Et3N (0.1 mL; 1.3 equiv)). The resulting mixture cooled in an ice-bath. Diisopropylethyl amine (0.38 mL; 4 equiv) was added. BOP-C1 (280 mg; 2 equiv) and 3-nitro-l,2,4-triazole (125 mg; 2 equiv) was then added.
The reaction mixture was stirred at 0°C for 90 mins. The mixture was diluted with CH2CI2 (60 nL) and washed with saturated aq. NaHCO3 (2x10 mL) and brine. The combined aqueous ayers were back extracted with CH2CI2 (-20 mL). The combined organic extract was dried
Na2SO4) and evaporated. The residue purified on silica (25 g column) with CH2CI2 /i-PrOH aolvent system (2-10% gradient). Yield: 140 mg (27%).
[0358] Préparation of (30a); A solution of 1-2 (110 mg; 0.13 mmol) in 80% aq. 'ormic acid was heated at 35-37°C for 3 hours. The mixture was evaporated to give an oily ’esidue. The residue was co-evaporated 2 times with toluene. Purification on a silica gel rolumn (10 g) with CH2CI2 /MeOH solvent system (4-10% gradient) to afford compound 30a ;46 mg, 59% yield). JIP-NMR (DMSO-de): δ -4.45. MS: m/z 646 (M+46-1 ).
155
EXAMPLE 31
Préparation of Compound f31 a)
NHDMT
NHDMT
80% aq. HCOOH 35-37°C;3h
EtjN
ΒΟΡ-CI, DIPEA, NT
THF; (TC; 90 min [0359] Préparation of (2-2); To a solution of 2-1 (370 mg; 0.64 mmol) in THF (10 inL) under Ar was added triethylammonium bis(POM)phosphate (330 mg; 1.2 equiv). The mixture cooled in icc-bath, and diisopropylethyl amine (0.42 mL; 4 equiv) was added. BOP-C1 305 mg; 2 equiv) and 3-nitro-!,2,4-triazo!e (137 mg; 2 equiv) was then added. The reaction mixture was stirred at 0°C for 90 mins. The mixture was diluted with CH2C12 (50 mL) and washed with saturated aq. NaHCOj (2x10 mL) and brine. The combined aqueous layers were back extracted with CH2CI2 (—20 mL). The combined organic extract was dried (Na2SO4), evaporated, and the residue purified on silica (25 g column) with CH2C12 /i-PrOH solvent jystem (2-10% gradient). Yield: 154 mg (27%).
10360] Préparation of f31a): A solution of 2-2 (68 mg; 0.08 mmol) in 80% aq. ’ormic acid was stirred at R.T. for 3 hours. The mixture was evaporated to an oily residue. The residue was co-evaporated 2 times with toluene. Purification on a silica gel column (10 g) with CH2C12 /MeOH solvent System (4-10% gradient; target compound eluted with 8% MeOH) ifforded 31a (35 mg, 78% yield). J1P-NMR (DMSO-de): δ -4.19. MS: m/z 580 (M-l), 646 M+46-1), 550 (M-30-1).
156
EXAMPLE 32
Préparation of Compound (32a)
ΒΟΡΌ, DIPEA, MT
THF; 0°C; 90 min
[0361] To a solution of 3-1 (71 mg; 0.26 mmol) in THF (4 mL) under Ar was added méthylammonium bis(POM)phosphate (144 mg; 1.2 equiv), and the resulting mixture was cooled in an ice-bath, and diisopropylethyl amine (0,18 mL; 4 equiv) was added. BOP-C1 (132 mg; 2 equiv) and 3-nitro-l ,2,4-triazole (59 mg; 2 equiv) was then added. The reaction mixture ’ vas stirred at 0°C for 1 hour. The mixture was diluted with CH2C12 (50 mL) and washed with saturated aq. NaHCOj (2 x 10 mL) and brine. The combined aqueous layers were back extracted with CH2C12 (—20 mL). The combined organic extract was dried (Na2SO4), evaporated, and the residue was purified on silica (10 g column) with CH2Cl2/MeOH solvent System (4-10% gradient). Compound 32a was repurified by RP-HPLC (35-90%B; A: water, B: MeOH). Yield 75mg(50%). 3IP-NMR (DMSO-de): δ-4.14. MS: m/z 627 (M+46-1), 551 (M30-1).
157
EXAMPLE 33
Préparation of Compound (33a)
[0362] Préparation of (4-21: To a solution of 4-1 (0.29 g; 0.5 mmol) in MeCN (8 mL) was added 5-ethylthio-lH-tetrazole in MeCN (0.25 M; 2.4 mL; 1.2 equiv). BisSATEphosphoramidatc (0.24 g; 1.05 equiv.) in MeCN (1.5 mL) was added over 90 mins. The reaction mixture was stirred for 4 hours at R.T., and then cooled to -40°C. MCPBA (0.23 g; 2 equiv.) in CH2CI2 (3 mL) was added. The mixture was allowed to warm to R.T. and diluted with EtOAc(50 mL). The mixture was washed with 10% aq. NaHSOj (2 x 10 mL), saturated aq. NaHCOj (2 x 10 mL) and brine. The mixture was then dried (Na2SO<). The evaporated residue was purified on silica (10 g column) with CH2CI2 /MeOH solvent system (4-10% gradient) to afford 4-2 (0.26 g, 55% yield).
[0363] Préparation of (33a): A solution of 4-2 (0.21 g; 022 mmol) in 80% aq. AcOH (15 mL) was stirred 4 hours at R.T. The mixture was evaporated and purified on silica (10 g column) with CH2CI2 /MeOH solvent system (4-10% gradient). Yield: 0.13 g (90%). 3,PNMR (DMSO-de): δ -2.00. MS: m/z 686 (M+46-1).
158
EXAMPLE 34
Triethylamine was added (0.088 mL, 0.63 mmol), and the mixture was vortexed to obtain a clear solution. After addition of POCIj (0.01 mL, 0.1 mmol), the mixture was vortexed and left for 20 nin. The mixture was then centrifugated. The supematant was added to the protected nucleosîde (0.05 mmol), and the mixture was kept at ambient température for 1 hour. rris(tetrabutylammonium) hydrogen pyrophosphate (180 mg, 0.2 mmol) was added, and the mixture was kept for 2 hours at R.T. The reaction was quenched with water, evaporated, dissolved în 80% formic acid and left for 2 hours at R.T. Formic acid was evaporated, and the residue dissolved in water (5 mL) and extracted with EA (2x2 mL). The aqueous fraction was loaded onto column HiLoad 16/10 with Q Sepharose High Performance (linear gradient of NaCi from 0 to IN in 50mM TRIS-buffer (pH = 7.5)). Fractions containing the triphosphate were combined, concentrated and desalted by RP HPLC on Synergy 4 micron Hydro-RP column (Phenominex) using a lînear gradient of methanol from 0 to 20% in 50mM triethytammonium acetate bufîer (pH 7.5) for elution. The following compounds shown in Table 1 were synthesized according this procedure:
159 ' able 1 - Triphosphates obtained from Examnle 34
Compound J1PNMR Pot “P NMR PO 31pnmr ?y MS (M)
HO-P-0-Lo-Lo^O^O^° Ah Ah Ah NsYNH ho f 'NH2 34a -11.31 d -20.82 t -5.48 d 550.2
HO-P-O-P-O-P-O^O^fO'-f0 Ah Ah Ah/H HO* F 'NH2 34b -9.13 d -18.18 t -2.85 d 548.2
HO-P-O-P-O-J-OV^OKANL^f0 Ah Ah Αη/ΛΎ H^ F 'Nh2 34c -10.95 d -20.62 bs -5.37 bs 552.2
P θ θ ~ HO-P-O-p-O-P-O*Vz°xAN>^*f Ah Ah Ah/Η-Γ \^-νη H^ F NH? 34d -11.24 d -20.82 t -5.48 d 554.2
ho-B-o-LoJ-o-» Ah Ah Ah ^\-Γ N ni V nh2 34e -12.06 d -20.97 t -5.69 d 549.2
160
EXAMPLE 35
Préparation of Compound (35a) -N î
Unh +ci-p-ci o
fl
O . Il HO-p-O-p-O-Ç—O
OH Cl
I OH [0365] 1,2,4-Triazol (42 mg, 0.6 mmol) was suspended in dry CH3CN (1 mL). rriethylaminc was added (0.088 mL, 0.63 mmol), and the mixture was vortexed to obtain a clear solution. After addition of POCIj (0.01 mL, 0.1 mmol), the mixture was vortexed and left for 20 mins. The mixture was centrifùgated, and the supematant was added to the protected nucleoside (0.05 mmol). The mixture was kept at ambient température for 1 hour. Tris(tetrabutylammonium) hydrogen pyrophosphate (180 mg, 0.2 mmol) was added, and the mixture was kept for 2 hours at R.T. The reaction was quenched with water, evaporated, dissolved in ammonium hydroxide and left for 2 hours at R.T. The solvent was evaporated, and the residue dissolved in water (10 mL). The mixture was loaded onto a column HiLoad 16/10 with Q Sepharose High Performance. Séparation was done in linear gradient of NaCl from 0 to IN ïn 50mM TRIS-buffer (pH7.5). The fractions containing the product were combined, concentrated and desalted by RP HPLC on Synergy 4 micron Hydro-RP column (Phenominex). A linear gradient of methanoi from 0 to 20% in 50mM triethylammonium acetate buffer (pH 7.5) was used for clution. MS (M-l): 532.1. 31P-NMR (δ ppm): -5.12 (d), -11.31 (d) and -20.43 (0.
EXAMPLE 36
Préparation of Compounds (36a)-(36d)
HO F
N—
POCLj/PO(OMe)j pyrophosphate
I
Deoxy-2’-fluoro-4’-alkyl-cytidine (0,09 mmot) was dissolved in the mixture of DMF (5 mL) and Ν,Ν’-dimethylacetate in DMF (0.110 mL, 0.9 mmot). The reaction mixture left at R.T. ovemight. The solvent was evaporated, and the residue purified by flash chromatography in gradient of methanol in DCM from 3% to 20%. The N-Protected nucleoside was concentrated in vacuum, dried and dissolved in dry trimethylphosphate (0.7 mL). The solution was cooled to 4°C and POClj (0.017 mL, 0.18 mmol) was added. In 1 hour, tributylamine (0.102 mL, 0.3 mmol) was added at R.T. Tributylammonium pyrophosphate (156 mg, 0.34 mmot) was then added. Diy DMF (about 0.100 mL) was added to solubilize pyrophosphate. After 2 hours, the reaction was quenched with TEAB-bufîer. The product was isolated by ion-exchange chromatography on AKTA Explorer as described in Example 35. The fractions containing the product were concentrated and treated with ΝΗ»ΟΗ for 2 hours at R.T.
The product was desalted by RP HPLC as described in Example 35.
Table 2 — Triphosphates obtained from Example 36
Compound ilPNMR Pa P NMR P0 J|PNMR Py MS (MT
_,nh2 000 ZY U II II V N HO-P-O-P-O-P-O-^n/W OH OH OH 0 HÔ F 36a -11.38 bs -22.88 bs -7.62 bs 512.1
^zNHî 000 XY Il II II V N HO-p-o-P-O-P-O-^n oh oh oh 0 36b -11.49 bs -20.41 bs -5.34 bs 510.0
Compound JIPNMR Pa 3lPNMR P0 3lPNMR py MS (M-)
ήη2 ooo /Ύ ho-p-o-p-o-p-o-w 0 n^n OH OH OH ° ^HO ’f 36c -11.96 bs -22.07 t -5.66 d 508.3
>nh2 ooo ΖΎ B “ <l V N HO-P-O-P-O-P-O-^O N^N oh 6h oh P*A_/ 0 Z 4 i. ' H<5 F 36d -11.90 d -23.23 t -10.66 d 514.0
nh2 OH OH OH Z\ HO^-O-W-O-y OOO ^^\_J ° F H<? *F 36e -11.77 d -23.05 t -9.70 s 529.9
nh2 <j>H OH OH /Λ 'F 36f -11.74 d -23.37 t -10.85 d 539.2
nh2 OH OH OH /*N HO-ê-O-^-O^ojH Ooo /=*'\__/ ° —' ho-' f 36g -11.87 d -23.32 t -10.83 d 523.9
NH2 <j>H OH OH HO—P—0—P—0—P-0—o .N-< 8 8 8-DÔ*·* 'ho2 ’”f 36h -11.48 d -23.26 t -10.63 d 526.1
NHj OH OH OH HO-P-O-P-O-P-O—χ o *N-4 8 8 Oôn —] HÎ 'f 361 -11.67 d -23.22 t -10.77 d 554.1
nh2 OH OH OH HO-P-O-P-O-P-O—χ ο 8 8 8 J>ÔA 'Hdr \ 3«i -11.97 d -23.34 t -10.92 d 523.9
163
EXAMPLE 37 Préparation of Compounds (37a)
O
[0367J Compound 37a was synthesized by reaction of phosphor(tris-triazo!ide) with
Cyl-2’-deoxy-2’-fluoro-uridine as described Exemptes 34 and 35. MS (M-l): 513.1. 3IP(5 ppm): -9.43 (bs), -11.68 (d) and -23.09 (bs).
EXAMPLE 38
Préparation of Compounds (38a)
[0368] The starting nucleoside (15 mg, 0.05 mmol) was dissolved in dry t imethylphosphatc (3 mL). The solution was cooled to 4°C. POC13 (0.013 mL, 0.125 mmol) vas added, followed by pyridine (0.01 mL, 0.125 mmol). In 1 hour, tributylamine (0.035mL, 0.125 mmol) was added at R.T. followed by tributylammonium pyrophosphatc (156 mg, 0.34 mmol). Dry DMF (about 0,100 mL) was added to solubüize pyrophosphate. In 2 hours, the réaction was quenched with TEAB-buffer. The product was isotated by ion-exchange iromatography on AKTA Explorer as described in Exemple 35. The fractions containing the roduct were concentrated and treated with NH4OH for 2 hours at R.T.
The product was desalted by RP HPLC as described in Exampie 35. MS (M-l): 529.9. 31P-NMR (δ ppm): 9 42(d), -11.59(d) and -23.03(t).
164
EXAMPLE 3»
Préparation of Compound (40a)
ΒηΟ-χχΟ^^ 3ζΟ-^Λ_/ 0
Bnd F
40-4 ^NPMB ηούλ Bn(J 'F
40-2
X e NPMB
Bn<J 'F
40-5
BnO' HOa ? NPMB
BnO FBn(J >
40-7
,ΒηΟ-ΧχΟ.
BnÔ F
H(J 'F
40-10 [0369] Préparation of (40-2): To a solution of 40-1 (50.0 g, 205 mmol) in pyridine (250 mL) was added DMTrCi (75.0 g, 225.0 mmol). The solution was stirred at R.T. for 15 hours. MeOH (120 mL) was added, and the mixture was concentrated to dryness under reduced ~Lessure. The residue was dissolved in EA and washed with water. The organic layer was dried over Na2SO4 and concentrated to give the crude 5’-O-DMTr intermediate (80.52g) as a light
Pfe ove yellowsolid. The intermediatewasdissolvedinanhydrousDMF(300mL), andK2COj (80.52g, 5 i ocked FdU (101.1 g). The intermediate was treated with 80% HOAc (900 mL) at R.T.
33.2 mmol) was added followed by PMBC1 (31.7 g, 109.2 mmol). The mixture was stirred at .T. ovemight. The reaction was diluted with EA and washed with brine. The organic phase as dried over Na2SÛ4 and concentrated to give crude 5’-O-DMTr-N3-PMB FdU (98.8 g) as a light yellow solid. The solid was dissolved in DMF (300 mL), and NaH (10.42 g, 260.5 mmol) as added followed by BnBr (73.8 g, 434.2 mmol). The reaction was stirred at R.T, ovemight and then was quenched with water. The solution was diluted with EA and washed with brine.
The organic phase was dried over Na2SÛ4 and concentrated to give the crude fuily blocked FdU intermediate, which was purified on a silica gel column (PE:EA = 10:1 to 3:1) to the pure fully b
165
ovemight, and the solvent was removed. The residue was purified on a silica gel column to give
40-2 as a white foam (42.1 g, 30.2% for 4 steps).
[0370] Préparation of (40-3): To a solution of 40-2 (42.1 g, 92.6 mmol) in anhydrous CHjCN (300 mL) was added IBX (28.5 g, 121.7 mmol) at R.T. The reaction mixture was refluxed for 1 hour and then cooled to 0°C. The precipitate was filtered-off, and the filtrate was concentrated to give the crude aldéhyde (39.22 g) as a yeliow solid. To a solution of the aldéhyde (39.22 g) in 1,4-dioxane (250 mL) was added 37% CH2O (28.1 mL, 345.6 mmol) and 2N NaOH aqueous solution (86.4 mL, 172.8 mmol). The mixture was stirred at R.T. for 2 hours and then neutralized with AcOH to pH = 7. EtOH (200 mL) and NaBH4 (19.7 g, 518.6 mmol) were added, stirred at R.T. for 30 mins. The mixture was quenched with saturated aqueous MH4CI, and extracted with EA. The organic layer was dried over Na2SO4 and concentrated. rhe residue was purified by silica gel column chromatography (PE:EA= 4:1 to 2:1) to give 40-3 £5.5 g, 55.7%) as a white solid.
[0371] Préparation of (40-4): To a stirred solution of 40-3 (25.5 g, 52.5 mmol) in inhydrous pyridine (150 mL) and anhydrous CHjCN (150 mL) was added BzCl (6.6 g, 52.47 mmol) dropwise at 0°C. The mixture was stirred at R.T. for 14 hours. The reaction was Îuenched with H2O, and the solution was concentrated. The residue was dissolved in EA and rashed with saturated NaHCOj. The organic layer was dried over Na2SO4 and concentrated. he residue was purified on a silica gel column (PE/EA = 5:4) to give the mono-Bz protected intermediate (18.1 g, 60.0%) as a white foam. To a stirred solution of this intermediate (18.1 g, 2 0.68 mmol) in DMF (100 mL) were added Cs2COj (30.0 g, 92.03 mmol) and BnBr (10.4 g, 61.36 mmol). The mixture was stirred at R.T. ovemight. The reaction was quenched with saturated NH4Cl aq., extracted with EA and washed with brine. The solvent was removed to give crude 40-4 (19.3g, 95.1%) as a light yeliow solid.
I [0372] Préparation of (40-5): To a stirred solution of 40-4 (19.3 g, 28.4 mmol) in anhydrous MeOH (230 mL) was added NaOMe (24.9 g, 460 mmol) at R.T. The mixture was sirred for 1 hour. The reaction was quenched with AcOH (10 mL) and concentrated. The residue was purified on a silica gel column (PE/EA - 1/2) to afford 40-5 (11.2 g, 54.0%) as a white solid.
[0373] Préparation of (40-6): To a stirred solution of compound 40-5 (200 mg,
ÎI47 mmol) in anhydrous DCM (5 mL) was added DMP (168 mg, 0.674 mmol) at R.T. The xture was stirred at R.T. for 2 hours. The solvent was removed, and the residue was purified a silica gel column (PE:EA = 5:1 to 1:1) to give the aldéhyde crude as a light yeliow solid mg). T o a stirred solution of the aldéhyde (200 mg) in anhydrous THF (5 mL) was added
166
VieMgBr (1.0 mL, 1.01 mmol) at -78°C. The mixture was stirred at -78°C for 1 hour. The reaction was quenched with saturated NH4C1 aq .and extracted with EA. The concentrated jrganic phase was purified by column chromatography (PE: EA = 5:1 to 1:1) to give 40-6 (a nixture ofstereomers, 135 mg, 65%) as a white solid.
[0374] Préparation of (40-7): To a stirred solution of DAST (1.64 g, 10.17 mmol) in anhydrous toluene (40 mL) was added dropwise a solution of compound 40-6 (1.2 g, 2.03 mmol) at -78°C. The mixture was stirred at -78°C for 30 mins. The solution was warmed to 60°C slowly and stirring was continued ovemight. The mixture was poured into a saturated Na2COj solution. The concentrated organic phase was concentrated and purified on a silica gel column (PE:EA = 10:1 to 3:1) to afford 40-7 as a white solid (1.08 g, 83.88%). *H NMR (CDjOD, 400 MHz) £7.87 (d, J= 8.4Hz, IH), 7.27-7.37 (m, 12H), 6.82-6.84 (m, 2H), 6.14 (d, J = 46.8,2.0Hz, IH), 5.18-5.50 (m, 4H), 4.96 (s, 2H), 4.45-4.88 (m, 7H), 3.67-3.89 (m, 5H).
[0375] Préparation of(40-8): A mixture of compound 40-7 (0.91g, 1.54 mmol) and ÇAN (2.53 g, 4.61 mmol) in a 3:1 solution of MeCN:water (10 m L) was stirred at R.T. cvemight. Brine (10 mL) was added, and the mixture was extracted with EA The combined organic extracts were dried and evaporated under reduced pressure. Purification by chromatography on silica gel column with PE: EA=10:l to 2:1 afforded 40-8 as a yellow solid (J05 mg, 41,96%).
[0376] Préparation of (40-9): To a stirred solution of 40-8 (350 mg, 0.74 mmol) in aihydrous MeCN (8 mL) were added TPSC1 (449 mg, 1.48 mmol), DMAP (180 mg, 1.48 mmol) and TEA (374 mg, 3.70 mmol) at R.T. The mixture was stirred at R.T. ovemight. b H4OH (15 mL) was added, and the mixture was stirred for 2 hours. The solvent was removed, and the residue was purified on a silica gel column with PE: EA=8:1 to 1:1 to afford the crude (' 80 mg crude), which was dissoived in anhydrous DCM (10 mL). A mixture of MMTrCl (( 95mg, 2.25mmol) and AgNOj (380mg, 2.25 mmol) was added at R.T., and the mixture was stirred at R.T. ovemight. The solid was filtered off and washed with DCM. The filtrate was
I washed with brine and dried over Na2SO4. The concentrated organic phase was purified on a si ica gel column (PE:EA= 8:1 to 2:1) to afford 40-9 as a yellowsolid (460 mg, 81.33%).
[0377] Préparation of (40-10): To a stirred solution of compound 40-9 (450 mg, mmol) in acetone were added ammonium formate (1.29 g, 20.6mmol, in portions) and 10% palladium on carbon (1.0 g). The mixture was refluxed for 12 h. The catalyst was filtered off ar d washed with acetone. The filtrate was diluted with EA and washed with brine. The ccncentrated organic phase was purified by column chromatography (DCM:MeOH = 100:1 to
S: 1) to afford 40-10 as a white solid (250 mg, 72.8%). *H NMR (DMSO-d6,400 M Hz) £8.56
0.
1β7 l s, IH), 7.73 (d, J= 7.6 Hz, IH), 7.14-7.28 (m, 12H), 6.84 (d, J= 8.8 Hz, 2H), 6.30 (d, 7.6
Hz, IH), 6.03-6.08 (m, IH), 5.84 (d, 5.2 Hz, IH), 5.33-5.35 (m, IH), 4.97-5.18 (m, IH),
Λ86-4.90 (m, IH), 4.34 (d, 4.4 Hz, IH), 3.72 (s, 3H), 3.54-3.57 (m, 2H), 1.28 (dd, J, = 6.4
Hz, Ji = 25.6 Hz, 3H). ESI-MS: m/z 563.50 [M + H]+.
(0378] Préparation of (40a): 40-10 (101 mg, 0.179 mmol) was dissolved in 80% HOAc (20 mL) at R.T. The mixture was stirred at 50°C for 5 hours. The solvent was removed, rnd the residue was co-evaporated with toluene twice. The residue was purified by column chromatography (DCM:MeOH = 100:1 to 10:1) to afford 40a as a white solid (36.6 mg, 70.26%). *H NMR (CDjOD, 400 MHz) 57.98 (d, J= 7.6 Hz, IH), 6.20-6.24 (m, IH), 5.92 (d, J ” 7.2 Hz, IH), 5.17-5.30 (m, IH), 4.99-5.14 (m, IH), 4.51-4.86 (m, IH), 3.78 (d, J= 1.6 Hz, 2H), 1.35-1.43 (m, 3H). ESI-MS; m/z 291.84 [M + H]+, 582.81 [2M + H]+.
EXAMPLE 40
Préparation of Compound (41a)
TBDPSO
41-3 tBDPSO
TBDPSO-A^Oyf4-^ TBSÔ 'F
41-2
NH2 O —
HO' 'F
41a [0379] Préparation of (41-2): To a solution of 41-1 (3 g, 4.8 mmol) in anhydrous f CM (50 mL) were added BzCl (1.3 g, 9.6 mmol), DMAP (1.1 g, 9.6 mmol) and NEt3 (4 mL) at R.T. The reaction was stirred at R.T. for 2 hours. Water was added, and the reaction was stirred for another 1 hour. The mixture was diluted with DCM (150 mL) and washed with water, 0.1 M F Cl and saturated aqueous NaHCOj. The solvent was removed, and the crude product was purified by silica gel column chromatography (25% EtOAc in PE) to give 41-2 as a yellow solid (2.8 g, 80.0%).
[0380] Préparation of (41-3): A mixture of 41-2 (2.6 g, 3.6 mmol) and Pd(OAc)i mg) in DCM (50 mL) was suspended in a solution of CH2N2 in Et2O (generated by si andard procedure, 350 mL) at -78°C. The réaction was stirred to R.T. ovemight. The mixture vas quenched with HOAc, and the reaction was stirred for another 1 hour. The mixture was d luted with EtOAc (150 mL) and washed with water and saturated aqueous NaHCOî. The
168
solvent was removed, and the crude was dissolved in NHj.MeOH (sat., 100 mL). The reaction vas stirred to R.T. ovemight. The crude product was purified by silica gel column i ihromatography (25% EtOAc in PE) to give 41-3 as a yellow solid (800 mg, 35.2%).
[0381] Préparation of (41-4): To a solution of 41-3 (800 mg, 1.3 mmol) in anhydrous CH3CN (50 mL) were added TPSC1 (755 mg, 2.5 mmol), DMAP (305 mg, 2.5 mmol) 1 nd NEtj (400 mg, 4 mmol) at R.T. The reaction was stirred at R.T. for 2 hours. NH4OH (25 mL) was added, and the reaction was stirred for another 1 hour. The mixture was diluted with DCM (150 mL) and washed with water, 0.1 M HCl and saturated aqueous NaHCOj. The jolvent was removed, and the crude product was purified by silica gel column chromatography (25% EtOAc in PE) to give 41-4 as a yellow solid (340 mg, 42.5%).
[0382] Préparation of (41a): To a solution of 41-4 (200.0 mg) in MeOH (10 mL) Was added NH4F (600 mg). The réaction was refluxed for 24 hours. The solvent was removed, and the residue was purified by column chromatography on silica gel (DCM: MeOH - 15: 1) to give 41a (50.0 mg, 55.9%) as a white solid. *H NMR (CDjOD, 400 M Hz) J 8.13 (d, J= 7.6 Jz, IH), 6.01 (dd, Jt = 2.4 Hz, J2 = 15.6 Hz, 1H),5.85 (d, J = 7.6 Hz, IH), 5.04-4.89 (m,lH), 52 (dd, J/ = 5.2 Hz, J2 *= 19.6 Hz, IH), 3.66 (s, 2H), 1.00-0.94 (m, IH), 0.54-0.30 (m, 4H); ESI-MS: m/z 285.82 [M + Hf, 570.84 [2M + H]+.
169
EXAMPLE 41
Préparation of Compound (42a)
ho' 1=
42-1
ΤΤΟ-Α/λ/Tfo-'A_7 —— MMTrO' F
42*4
NHDMTr
MMTrO
MMTrO
MMTrO'
42-8
MMTrO'
NHDMTr
NHDMTr
TBSO Cl— MMTrO'
MMTrO
42-9
MMTrO
HO' 'F
42a
I0 [0383] Préparation of (42-2): To a solution of 42-1 (50 g, 203 mmol) in anhydrous pyridine (200 mL) was added TBDPSC1 (83.7 g, 304 mmol, 1.5 eq). The reaction was stirred ovemight at R.T. The solution was concentrated under reduced pressure to give a syrup, which vras partitioned between ethyl acetate and water. The organic layer was separated, washed with brine, dried over magnésium sulfate and concentrated to give the 5-OTBDPS ether as a white foam (94 g). The crude ether was dissolved in anhydrous DCM (300 mL), and silver nitrate (' >6.03 g, 388.4 mmol, 2.0 eq) and cotlidine (235 mL, 1.94 mol, 10 eq) were added. The mixture v as stirred at R.T., and MMTrCl (239.3 g, 776.8 mmol, 4 eq) was added. After being stirred overnight at R.T., the mixture was filtered through Celite and filtrate was diluted with MTBE. The solution was washed successively with IM citric acid, diluted brine and 5% sodium bicarbonate. The organic solution was dried over sodium sulfate and concentrated under vacuum to give the fully protected intermediate as a yellow foam. The crude intermediate was dissolved in anhydrous THF (250 mL) and treated with TBAF (60 g, 233 mmol, 1.2 eq). The mixture was stirred for 2 hours at R.T., and the solvent was removed under reduced pressure. T lie residue was taken into ethyl acetate and washed brine. After drying over magnésium
170 iiulfate, the solvent was removed in vacuo. The residue was purified by column chromatography
PE:EA « 5:1 to 1:1 ) to give 42-2 as a white foam (91 g, 86.4%).
[0384] Préparation of (42-3): To a solution of 42-2 (13.5 g, 26 mmol) in DCM (100 mL) was added pyridine (6.17 mL, 78 mmol, 3 eq). The solution was cooled to 0°C and DessMartin periodinane (33.8 g, 78 mmol, 3 eq) was added. The mixture was stirred for 4 hours at R.T. and quenched by the addition of a 4% Na2S20j/4% sodium bicarbonate aqueous solution (to pH 6, —150 mL). The mixture was stirred for another 15 mins. The organic layer was s eparated, washed with diluted brine and concentrated under reduced pressure. The residue was c issolved in dioxanc (100 mL), and the solution was treated with 37% aqueous formaldéhyde (21.2 g, 10 eq) and 2N aqueous sodium hydroxide (10 eq). The réaction mixture was stirred at F„T. ovemight. The reaction was quenched with saturated NH4CI (-150 mL), and the mixture was concentrated under reduced pressure. The residue was parti tioned between ethyl acetate and 5% sodium bicarbonate. The organic phase was separated, washed with brine, dried over magnésium sulfate and concentrated. The residue was purified by column chromatography (WeOH:DCM = 100:1-50:1) togive 42-3 as a white foam (9.2 g, 83.6%).
[0385] Préparation of (42-4): 42-3 (23 g, 42.0 mmol) was co-evaporated with toluene twice. The residue was dissolved in anhydrous DCM (250 mL) and pyridine (20 mL). T|he solution was cooled to -35°C. Triflic anhydride (24.9 g, 88.1 mmol, 2.1 eq) was added dropwise over 10 mins. At this température, the réaction was stirred for 40 mins and then was qluenched with water (50 mL) at 0°C. The mixture was stirred 30 mins, and extracted with EA ( 50 mL x 2). The organic phase was dried over NajSCL, and filtered through a silica gel pad. The filtrate was concentrated under reduced pressure. The residue was purified by column c iromatography (PE:EA= 100:1-1:1) to give 42-4 as a brown foam (30.0 g, 88.3%).
[0386] Préparation of(42-5): 42-4 (30 g, 36.9 mmol) was co-evaporated twice with toluene and dissolved in anhydrous DMF (150 mL). The solution was cooled to 0°C, and treated with sodium hydride (60% in minerai oil; 1.5 g, 40.6 mmol). The reaction was stirred at R.T. for 1 h. Lithiumchloride (4.6 g, 110.7 mmol, 3 eq) was added. Stirring was continued for aiihydro-chtoro compound. The mixture was taken ïnto 100 mL of half saturated ammonium chloride and ethyl acetate. The organic phase was separated, washed with diluted brine and concentrated under reduced pressure. The residue was dissolved in THF (150 mL), and the solution was treated with IN aqueous sodium hydroxide (-41 mL, 40.1 mmol, 1.1 eq). The mixture was stirred at R.T. for lh. The réaction was diluted with half saturated sodium b: carbonate (-60 mL) and extracted with EA. The organic phase was dried (magnésium sulfate) hours when LCMS indicated complété conversion of the anhydre triflate intermediate to
171 and concentrated under reduced pressure. The residue was purified by column chromatography (DCM:MeOH = 300:1-60:1) to give 42-5 as a yellow foam (18.3 g, 87.6%).
[03871 Préparation of (42-6): To a solution of 42-5 (18.3 g, 32.33 mmol) in anhydrous DCM (150 mL) was added TBSC1 (17.7 g, 64.6 mmol) and imidazole (6.6 g, 97 mmol). The reaction was stirred overnight at R.T. The reaction was diluted wîth water and îxtracted with DCM. The organic layer was separated, washed with brine, dried over Na2SC>4 ind concentrated. The residue was purified by column chromatography (DCM:MeOH = 100:1-80:1) to give 42-6 as a white foam (18.4 g, 83.7%).
[0388] Préparation of (42-7): A solution of 42-6 (18.4 g, 27.1 mmol), DMAP (6.6 g» 54.0 mmol) and TEA (5.4 g ,54.0 mmol) in MeCN (450 mL) was treated with 2,4,6riispropylbenzenesulfonyl chloride (16.3 g, 54.0 mmol). The mixture was stirred at R.T. for 3 îours. NH4OH (70 mL) was added, and the mixture was stirred for 2 hours. The solution was Eporated under reduced pressure, and the residue was purified on a silica gel column 'M/MeOH = 100:1 to 15:1) to give the crude (18.0 g). The crude was dissolved in ydrous DCM (150 mL). Collidine (8.1 g, 66.3 mmol, 2.5 eq), silver nitrate (4.5 g, 26.5 mmol, 1.0 eq) and DMTrCl (13.4 g, 39.7 mmol, 1.5 eq) were added. The reaction was stirred temight at R.T. The mixture was filtered through Celite. The filtrate was washed with brine d extracted with DCM. The organic layer was separated, dried over Na2SC>4 and ncentrated. The residue was purified by column chromatography (PE:EA = 60:1-3:1) as a yellow foam. The foam was dissolved in THF (150 mL) and TBAF (10.4 g, 39.7 mmol, 1.5 eq) was added. The réaction was stirred at R.T. After being concentrated, the mixture was washed with brine and extracted with EA. The organic layer was separated, dried over Na2SO4 and concentrated. The residue was purified by column chromatography (PE:EA =60:l-EA) to give ‘2-7 as a yellow foam (21.3 g, 92.4%).
[0389] Préparation of (42-8): To a solution of 42-7 (2.0 g, 2.3 mmol) in anhydrous
ÎCM (20 mL) was added Dess-Martin periodinane (1.95 g, 4.6 mmol) at 0°C under nitrogen. lie reaction was stirred at R.T. for 5 hours. The mixture was diluted with EtOAc (100 mL), and washed with a mixture of saturated aqueous Na2S20j and saturated aqueous NaHCOj. The crude product was purified by column chromatography on silica gel (PE: EtOAc = 2: 1) to give 42-8 (1.8 g, 90%) as a yellow solid.
[0390] Préparation of (42-9): To a solution of tetramethyl methylenediphosphonate
Î)0 mg, 1.68 mmol) in anhydrous THF (10 mL) was added NaH (84 mg, 2.1 mmol) at 0°C der nitrogen. The reaction was stirred at 0°C for 30 min. A solution of 42-8 (1.2 g, 1.4 mmol) in anhydrous THF (10 mL) was added dropwise at 0°C. The mixture was stirred at R.T. for 1 h.
172
The reaction was quenched with saturated aqueous NH4CI, and the crude product was purified jy column chromatography on silica gel (DCM: MeOH = 150:1) to give 42-9 (12 g, 882%) as i yellow solid. ‘H NMR (DMSO-d6, 400 M Hz) δ 8.51 (s, IH), 7.46-7.09 (m, 22H), 6.88-6.82 'm, 6H), 6.62 (q, Jt = 17.2 Hz, J2 = 22.4 Hz, IH), 6.12 (d, 72 Hz, IH), 5.86-5.75 (m, 2H),
5.43 (d, 25.2 Hz, IH), 4.63 (dd, Jt = 4.8 Hz, J2 - 212 Hz, IH), 4.45 (d, J= 12.0 Hz, IH),
3.94 (d, J= 12.0 Hz, IH), 3.72 (s, 9H), 3.53 (q, Jt = 112 Hz, J2 = 16.0 Hz, 6H); ES1-MS: m/z 971.59 [M + H]+.
[0391] Préparation of (42a)t A solution of 42-9 (300 mg) in 80% HOAc (26 mL) was stirred at 80-90°C for 2 h. The solvent was removed, and the crude product was purified by column chromatography on silica gel (DCM: MeOH 20:1) to give 42a (70 mg, 57%) as a white Jolid. ’H NMR (DMSO-d6, 400 M Hz) δ 7.61 (d, J = 7.6 Hz, IH), 7.35 (d, 15.2 Hz, 2H), ( .72 (q, Ji = 17.6 Hz, J2 = 24.4 Hz, IH), 623 (d, 6.0 Hz, IH), 5.99-5.85 (m, 2H), 5.74 (q, J= , 5.37-5.21 (m, IH), 4.694.61 (m, IH), 3.96 (d, J= 12.4 Hz, IH), 3.82 (d, J~ 12.0 Jz, IH), 6.72(q, J/ = 52 Hz, J2= 10.8 Hz, 6H);ES1-MS: m/z397.81 [M + H]+.
Hz, IH), Hz, IH), 6.1
EXAMPLE 42
Préparation of Compound (43a)
43-4
BnÔ
43-1 [0392] Préparation of (43-2): To a stirred solution of 43-1(3.8 g, 6.6 mmol) in arihydrous DMF (lOOmL) was added NaH (22 g) followed by CH3I (93 g, 66 mmol) at 0°C. Stirring was continued at R.T. ovemight. The reaction was quenched with saturated NH«Cl aq. Tlie mixture was diluted with EA and washed with brine. The organic layer was dried over NH2SO4 and concentrated. The residue was purified by silica gel column chromatography (PE:EA = 2:1) to give 43-2 (3.0 g, 70%) as a white solid.
[0393] Préparation of (43-3): A mixture of 43-2 (3.0 g, 5.1 mmol) and CAN (5.56
10.2 mmol) in a 3:1 solution of MeCNiWater (16 mL) was stirred at R.T. ovemight. The
173
solution was diluted with brine (10 mL ) and was extracted with EA. The combined organic extracts were dried and evaporated under reduced pressure. Purification by chromatography on silica (PE:EA =1:1) gave 43-3 as a yellow solid (1.71 g, 72%).
[0394] Préparation of (43-4): To a stirred solution of 43-3 (1.7 g, 3.6 mmol) in anhydrous MeCN (50 mL) were added TPSC1 (2.2 g, 7.2 mmol), DMAP (880 mg, 7.2 mmol) and TEA (1.1g ,10.8 mmol) at R.T. The mixture was stirred at R.T. overnight. NH4OH (25 mL) was added, and the mixture was stirred for 2 hours. The solvent was removed, and the residue was purified on a silica gel column (PE:EA = 8:1 to 2:1) to give the intermediate (1.4 g). The intermediate was dissoived in anhydrous DCM (30 mL), and MMTrCI (1.6 g, 5.2 mmol), AgNCh (1.4 g, 7.8 mmol) and collidîne (1.57 g, 13 mmol) were added. The mixture was stirred at R.T. overnight. The solid was fiitered off and washed with DCM. The filtrate was washed with brine and dried over Na2SO4. The concentrated organic phase was purified on a silica gel :olumn (PE:EA = 3:2) to give 43-4 (1.1 g, 57.9%) as a white solid.
[0395] Préparation of (43-5): To a stirred solution of 43-4 (550 mg, 0.74 mmol) in icetone were added ammonium formate (1.0 g, 15.8 mmol, in portions) and 10% palladium on ;arbon (1.0 g). The mixture was refluxed for 48 hours. The catalyst was fiitered off and washed vith the acetone. The filtrate was diluted with EA, washed with brine and dried. The Îoncentrated organic phase was purified by column chromatography (DCM:MeOH - 50:1) to îve 43-5 (330 mg, 72%).
[0396] Préparation of (43a): 43-5 (200 mg, 0.36 mmol) was dissoived in 80% ÇHiCOOH (20 mL) at R.T. The mixture was stirred at 60°C for 12 hours. The solvent was removed. The residue was purified by column chromatography (DCM:MeOH = 10:1), and the resulting solid was washed with DCM to give pure 43a as a white solid (44mg, 42%). *H NMR (CD3OD, 400 MHz) £8.02 (d, J= 7.2 Hz, IH), 6.14 (dd, Λ = 3.6 Hz, J2 = 15.2 Hz, IH), 5.88 (d, J= 7.2 Hz, 1 H), 5.10 (ddd, Λ = 4.0 Hz, J2 = 5.2 Hz, J2 = 53.6 Hz, 1 H), 4.47 (dd, Ji = 5.2 Hz, J2 = 14.8 Hz, IH), 3.84 (d, J= 12.0 Hz, IH), 3.70 (d, J= 12.0 Hz, IH), 3.58-3.64 (m, 2H), 3.36 ( j, 3H). ESI-MS: m/z 290 [M + H]+.
174
44-1
EXAMPLE 43
Préparation of Compound (44a) h e ©
1) POMO-P-O EtjNH
PO MO
ΒΟΡ-CI, DIPEA, NT
THF; r.t; 90 min
2) 80% aq. HCOOH 35°C; 30 mins.
[0397] To a solution of triethylammonium bis(POM)phosphate (0. 3 mmol, prepared 'rom 100 mg of bis(POM)phosphate and 50 gL of EtjN) in THF (3 mL) was added nucleoside ' 14-1 (150 mg; 0.26 mmol). The mixture was cooled in ice-bath. Diisopropylethyl amine (0.18 i riL; 4 equiv) was added then, followed by BOP-C1 (132 mg; 2 equiv) and 3-nitro-1^2,4-triazole ?i9 mg; 2 equiv). The réaction mixture was stirred at 0°C for 90 mins., and then diluted with
H2CI2 (30 mL) and washed with saturated aq. NaHCOj and brine. The combined aqueous 1 îyers were back extracted with CH2CI2. The combined organic extract was dried (Na2SO4),
CM
SV!
s' 'aporated, and the residue purified on silica (10 g column) with CH2C12 /i-PrOH solvent stem (3-10% gradient). The obtained mixture of products were treated for 30 mins at 35°C with 80% aq. HCOOH, and then evaporated and coevaporated with toluene. The evaporated residue was purified on silica (10 g column) with CH2C12 /MeOH solvent System (5-10% gradient) to obtain 44a (8 mg, 5%). 3IP-NMR (DMSO-di): 6 -5.07. MS: m/z = 668 (M+46-1).
175
EXAMPLE 44 Préparation of Compound (45a)
DMTO
Préparation of (45-2):
80%aq.HCOOH
r.t ; 30 min
ΒΟΡ-CI, DIPEA, NT
THF: 0°C: 90 min [0398]
To a solution of triethylammonium )is(POM)phosphate (0. 7 mmol, prepared from 233 mg of bis(POM)phosphate and 0.1 mL of itjN) in THF (8 mL) was added nucleoside 45-1 (253 mg; 0.42 mmol), followed by aiisopropylethyl amine (0.36 mL; 5 equiv), BOP-C1 (268 mg; 2.5 equiv) and 3-nitro-l,2,4i riazole (120 mg; 2.5 equiv). The reaction mixture was stirred at R.T. for 2 hours. The mixture was diluted with CH2CI2 (40 mL) and washed with saturated aq. NaHCOj and brine. The combined aqueous layers were back extracted with CH2C12. The combined organic extract was dried (Na2SO4), evaporated, and the residue was purified on silica (10 g column) with 1 lexanes/EtOAc solvent System (40-100% gradient) to yield 45a ( 180 mg, 47%).
[0399] Préparation of (45a): A solution of compound 45-2 (0.12 g; 0.13 mmol) in Î0% aq. HCOOH (8 mL) was stirred 30 mins. at R.T. The mixture was evaporated, coevaporated with toluene and purified on silica (10 g column) with CH2C12/MeOH solvent system (4-10% gradient) to yield 45a (55 mg, 70%). 31P-NMR (DMSO-de): 8 -4.36. MS: m/z 047 (M+46-1).
17β
EXAMPLE 45
Préparation of Compound (46a) [Me2CHC(O)JjO v O >-1-0
Py
NHMMT
Cl -M 0
Uo 48-2
80% aq. HCOOH
36°C; 3 h
11:
[0400] Préparation of (46-2): A mixture of 46-1 (170 mg; 0.3 mmol) in pyridine (3 >) and isobutyric anhydride (0.1 mL; 2 equiv) was stirred o/n at R.T. The mixture was Concentrated, and the residue was partitioned between EtOAc (30 mL) and saturated aq. NaHCOj. The organic layer was washed with water, brine and dried (NaîSQi). The residue was purified on silica (10 g column) with a hexanes/EtOAc solvent system (30 to 100% [ radient) to afford 46-2 (180 mg, 85%).
[0401] Préparation of (46a): A solution of 46-2 (0.18 g; 0.25 mmol) in 80% aq. ÎCOOH (5 mL) was heated for 3 hours at 36°C. The mixture was then evaporated, •evaporated with toluene and purified on silica (10 g column) with a CHîCL/MeOH solvent system (4-10% gradient) to afford 46a (75 mg, 70%). MS: m/z = 434 (M+1).
EXAMPLE 46
Préparation of Compound (47a) [MeCH3CtO)bO Py
NHDMT £F
1=0 47-2
60% aq HCOOH rt;3h
NH2
Λ0O Lo 47.
[0402]
Préparation of (47-2): 47-2 was prepared from 46-1 (274 mg, 0.46 mmol) and propyonîc anhydride (0.12 mL, 2 equiv.) in pyridine (5 mL) in the same manner as described for 46-2 (260 mg, 80%).
177
[0403] Préparation of (47a): 47-2 (120 mg, 0.2 mmol) was treated with 80% aq.
HCOOH at R.T. for 3 hours. The mixture was evaporated, coevaporated with toluene and purified on silica (10 g column) with a CH2Cl2/MeOH solvent system (4-10% gradient) to yield
47a (62 mg, 75%). MS: m/z = 404 (M-l).
EXAMPLE 47
Préparation of Compound (48a)
[0404] Préparation of (48-2): 48-2 was prepared from 46-1 (150 mg, 0.27 mmol) snd valeric anhydride (0.11 mL, 2 equiv.) in pyridine (3 mL) in the same manner as described far 46-2 (150 mg,73%).
[0405] Préparation of (48a): 48-2 (140 mg, 0.18 mmot) was treated with 80% aq.
HCOOH at R.T. for 3 hours. The mixture was evaporated and purified on silica (10 g column) with a CH2Cl2/McOH solvent system (4-10% gradient) to yield 48a (70 mg, 84%). MS: m/z = 462 (M+1).
178
EXAMPLE 48
Préparation of Compounds (49a), (50a) and (51a)
[0406] Préparation of (49-2), (50-2) and (51-2): To a solution of46-1 (1.26 g, 2.12 iimol) in pyridine (15 mL) were added n-octanoic acid (0.34 mL, t.O equiv.), DCC (60% in xylene; 0.81 mL, 1 equiv.) and DMAP (52 mg; 0.2 equiv.). The resulting mixture was stirred for 6 hours at R.T. The mixture was evaporated, and the residue partitloned between CH2C12 (100 mL) and saturated aq. NaHCOj (25 mL). The organic layer was washed with water, brine and dried (Na2SO4). The residue was treated with toluene. The solid material was filtered off, and the filtrate was purified on silica (25 g column) with a heaxanes/EtOAc solvent System (30100% gradient) to yield 49-2 (0.57 g, 32%), 50-2 (0.18 g, 12%), and 51-2 (0.2 g, 13%).
[0407] Préparation of (49a): A mixture of 49-2 (114 mg, 0.13 mmol) and 80% aq. formic acid was stirred for 3 hours at R.T. The mixture was evaporated and coevaporated with toluene and purified on silica (10 g column) with a CH2Cl2/MeOH solvent System (2-8% gradient) to yield49a(53 mg, 75%). MS: m/z = 544(M-l).
[0408] Préparation of (50a): 50a (44 mg, 75% yield) was prepared from 50-2 (104 mg, 0.14 mmol) în the same manner as described for 49a by using a 4-10% gradient of MeOH in C H2C12 for purification. MS: m/z = 418 (M-t).
[0409] Préparation of (51a): Sla (60 mg, 71% yield) was prepared from 50-2 (140 mg, 0.2 mmol) in the same manner as described for 49a by using a 4-10% gradient of MeOH in C H2C12 for purification. MS: m/z = 418 (M-l).
179
EXAMPLE 49 Préparation of Compound (52a)
[0410] Préparation of (52-2): A solution of JV-(tert-butoxycarbonyl)-L-valine (0.41 g, 1.9 mmol) and carbonytdiimidazote (0.31 g, 1.9 mmol) in THF (9 mL) was stirred at R.T. for 1.5 hours. The mixture was then stirred at 40°C for 20 mins. The mixture was added to a solution of 7a (0.42 g, 1,43 mmol) and DMAP (25 mg, 0.2 mmol) in DMF (8 mL) and TE A (4 r 1L) at 80°C. The reaction mixture was stirred at 80°C for 1 h, then cooled and concentrated.
The residue was partitioned between iert-butyl methyl ether (100 mL) and water. The organic lyer was washed with water, brine and dried (Na2SO4). The residue was purified on silica (25 g column) with a CH2Cl2/MeOH solvent system (2-10% gradient) to yield 52-2 (0.32 g, 90% in tlie mixture with 5-isomer), which was repurified by RP-HPLC (10-100% B; A: water, B:
MeOH). Yield: 0.25 g (35%).
[0411] Préparation of (52a): A solution of 52-2 (0.12 g; 0.24 mmol) in EtOAc (0.6 mL) was treated with HCl/dioxane (4 M; 0.6 mL) for 20 mins. with vigorous shaking. The white precipitate was filtered, washed with diethyl ether and dried to yield 52a as the dihydrochloride sait (95 mg; 85%). MS: m/z = 391 (M-l).
EXAMPLE 50
Préparation of Compound (53a)
N-Boc-L-Valino-OH Eî3N
DIPEA. BopQ. NT rt; 1 h
0 1) HCOOH
2) HCI/doxane
53-1 53-2 [0412] Préparation of (53-2): To a solution of N-Boc-Val-OH (0.16 g, 0.74 mmol) and EtjN (0.14 mL, 1.0 mmol) in THF was added 53-1. The resulting mixture was evaporated, coevaporated with pyridine and toluene and dissolved in THF (4 mL). DIPEA (0.38 mL, 2.2 rrmol) was added, followed by BOP-CI (0.28 g, 1.1 mmol) and 3-nitro-l,2,4-triazole (0.13 g, 1.1 mmol). The reaction mixture was stirred at R.T. for 1 h. The mixture was diluted with CH2C12
180
mL) and washed with saturated aq. NaHCOj and brine. The combined aqueous layers were jack extracted with CH2CI2. The combined organic extract was dried (Na2SO4), evaporated, and he residue was purified on silica (10 g column) with a hexanes/0.5 % EtjN/EtOAc solvent system (20-100% gradient) to yield 53-2 (0.39 g, 81%).
[0413] Préparation of (53a): A mixture of 14-2 (0.37 g, 0.33 mmol) and 80% aq. 1ICOOH (10 mL) was stirred at R.T. for 3 hours. The mixture was evaporated, and the residue 1 vas partitioned between water and CH2C12. The aqueous layer was washed with CH2CI2 and evaporated. The solid residue was suspended în EtOAc (1.5 mL) and treated with 4N HCl in dioxane (1.5 mL) with vigorous shaking. The solid was filtered, washed with diethyl ether and turified by RP-HPLC (A: 0.5N HCOOH in water, B: 0.5 N HCOOH in acetonitrile). The îsulting formic acid sait of 5’-O-valyn ester was converted into 53a dihydrochloride sait (63 îg, 40%) by suspending in EtOAc (2 mL) and treatment with 4N HCt/dîoxane (2 mL). MS: irVz= 391 (M-l).
EXAMPLE 51
Préparation of Compound (39a)
Me( atmi [0414] Préparation of (39-2): A solution of 39-1 (13 g, 1.4 mmol) in anhydrous * 'eOH (20 mL) was charged with Pd/C (1.3 g) and stirred at 25°C under hydrogen (1 atm) __losphere for 1 hour. The solution was filtered, evaporated to dryness, and purified on a silica Jet column (DCM:MeOH = 100:1 to 50:1) to give 39-2 (1.2 g, 92.3 %) as a white solid.
[0415] Préparation of (39-3): To a solution of 39-2 (1.2 g, 1.3 mmol) in MeOH (40 mL) was added NH4F (370 mg, 10 mmol) at 25°C and stirred at 60°C for 6 hours. The solution vas filtered, evaporated to dryness, and purified on a silica gel column (DCM:McOH = 200:1 to ):1) to give 39-3 as a white solid (249 mg, 30.7%). *H NMR (MeOD, 400 MHz) £7.92 (s, fl), 7.19-7.33 (m, 12H), 6.83-6.85 (m, 2H), 5.50 (dd, 4.0 Hz, Λ = 14.8 Hz, IH), 4.19-4.88 (άι, IH), 4.22 (dd, Ji =5.2 Hz,Λ = 16.0 Hz, IH), 3.76 (s, 3H), 3.41 (dd,Λ = 12.0 Hz,Λ = 36.8 H z, 2H), 1.52-1.74 (m, 2H), 0.87 (t, J= 7.6 Hz, 3H); ESI-LCMS: m/z 586.1 [M + H]+.
181
[0416] Préparation of (39a): A solution of 39-3 of 80% formic acid/20% water (3 mL) stood at RT for 2 hours, and then was concentrated to dryness. The residue was coevaporated with MeOH/toluene (3 times) and then ethyl acetate added. The suspension in ethyl acetate was heated at 70°C for 5 mins. The solvent was removed using a pipet. This washing was repeated 3 times. The resulting product (44mg) was further purified on reverse-phase HPLC using acetonitrile/water as mobile phase to give 39a (20 mg) as an off-white solid. *H NMR (DMSO, 400 MHz) £7.92 (s, IH), 10.82 br, IH), 7.96 (s, IH), 6.56 (s, 2H), 5.99 (dd, 5.0, 12.8 Hz, IH), 5.65 (d, J= 4.8 Hz, IH), 5.58,5.45 (2t, J = 5.2 Hz, 0.5H, 0.5H), 5.25 (br, IH), 1.19-4.88 (m, IH), 4.22 (dd, Jx = 5.2 Hz, J2 = 16.0 Hz, IH), 3.76 (s, 3H), 3.41 (dd, Ji = 12.0 Hz, J2 - 36.8 Hz, 2H), 1.52-1.74 (m, 2H), 0.87 (t, 7.6 Hz, 3H); ESI-LCMS: m/z 443.6 [M + 6i nethyl-2-heptylamine)]+.
EXAMPLE 52
Préparation of Compounds (55a) and (56a)
[0417] 1,2,4-Triazol (21 mg, 0.3 mmol) was dissolved in the mixture of CH3CN (0.7 mL) and EtjN (44 pL, 0.31 mmol). POCI3 (9ul, 0.1 mmol) was added, and the mixture was kept a[ R.T, for 20 mins. The white precipitate was filtered, and the filtrate added to the dry nlcleoside (28 mg, 0.05 mmol). The reaction was controlled by TLC and monitored by the disappearance of the starting nucleoside. After completion of the reaction, tetrabutylammonium sait of pyrophosphate (150 mg) was added, followed by DMF (0.5 mL) to get a homogeneous solution. After 1.5 hours at ambient température, the reaction was diluted with water (4 mL) a id extracted with DCM (2x5 mL). The combined organic extracts were evaporated, dissolved ii 5 mL of 80% HCOOH and left for 2 hours at R.T. The reaction mixture was concentrated and d stributed between water (5 mL) and DCM (5 mL). The aqueous fraction was loaded on the column HiLoad 16/10 with Q Sepharose High Performance. Séparation was done in a linear gradient of NaCl from 0 to IN in 50mM TRIS-buffer (pH7.5). Two fractions were obtained. Tie first fraction, containing the monophosphate (55a) was eluted at 70-75%B. and triphosphate (S6a) was eluted at 75-80%B. Both fractions were desalted by RP HPLC on Synergy 4 micron
Hydro-RP column (Phenominex). A linear gradient of methanol from 0 to 30% in 50mM triethylammonium acetate buffer (pH 7.5) was used for elution. The corresponding fractions were combined, concentrated and tyophilized 3 times to remove excess of buffer.
ιαζ
EXAMPLE 53
Préparation of Compounds (56b-e)
OCH
[0418J 1,2,4-Triazol (21 mg, 0.3 mmol) was dissolved in the mixture of CHjCN (0.7 r iL) and EtjN (44 gL, 0.31 mmol). POCb (9ul, 0.1 mmol) was added, and the mixture was kept at R.T. for 20 mins. The white precipitate was filtered, and the filtrate added to the dry rucleoside (28 mg, 0.05 mmol). The reaction was controlled by TLC and monitored by the disappearance of the starting nucleoside. After completion of the reaction, tetrabutylammonium s ilt of pyrophosphate (150 mg) was added followed by DMF (0.5 mL) to get a homogeneous solution. After 1.5 hours at ambient température, the reaction was diluted with water (4 mL) a îd extracted with DCM (2x5 mL). The combined organic extracts were evaporated, dissolved it. 5 mL of 80% HCOOH and left for 4 hours at 38°C. The réaction mixture was concentrated aid distributed between water (5 mL) and DCM (5 mL). The aqueous fraction was loaded on tl e column HiLoad 16/10 with Q Sepharose High Performance. Séparation was done in a linear giadient of NaCI from 0 to IN in 50 mM TRIS-buffer (pH7.5). Two fractions were obtained. T te triphosphate (56b-e) was eluted at 75-80%B. Desaltin was performed by RP HPLC on S; mergy 4 micron Hydro-RP column (Phenominex). A linear gradient of methanol from 0 to 30% in 50 mM triethylammonium acetate buffer (pH 7.5) was used for elution. The cc rresponding fractions were combined, concentrated and lyophilized 3 times to remove excess ofbufTer.
183
Table 3 - Triphosphates obtained from Example 53
Compound MS (M-l) J1PNMR Pa J P NMR PO JIPNMR py
O Ho—F>— oh° HÔ* 55a 373.00 +3.64 (s) NA NA
ooo f'f HO—p—ο—P—0—P—O-^AJwNn Ah Ah Ahc|-^C( ° Ηά zf 56a 532.95 -6.67 -6.74(d) -21.87(t) Sa a
.NHa o 0 0 ΖΎ HO-P-O-P-O-P-O^OyN^N Ah Ah Ah 0 Z hô- f 56b 526.05 -6.33 -6.47(d) -22.48(t) -11.53 -11.64(d)
>^NHl 0 0 0 ΧΎ N Il N Λ Λ M >N HO-P-O-P-0—P— Ah Ah Ah f^Q; 0 HÔ* F 56c 516.00 -632(bs) -22.45 (t) -11.64(d)
~Νίζ 0 o o n π II ' / HO-P-O-P-O-P—0-^.0^”^ Ah oh AhV^-^ HCj F 56d 524.4 -10.57 -10.67(d) -23.3 l(t) -11.31 -11.94(d)
NH2 3 i? 0 Q HO-P-O-P-O—P—O-’V.O./4^ 1 1 e v¥ T 0 OH OH 6hF7X H- HÔ* F 56e 529.8 -6.17(bs) 21.96(bs) 11.42(bs)
184
EXAMPLE54
Préparation of Compound (57a)
(0419] 2’-Deoxy-2’-fluoro-4’-C-(ethenyl)guanosÎne (25a, 31 mg, 0.1 mmol) was cissolved in dry pyridine (3 mL). Isobutyric anhydrate (50 pL, 0.3 mmol) was added. The réaction mixture was kept at ambient température. After 40 hours, isobutyric anhydrate (100 pL, 0.6 mmol) was added, and the reaction mixture was left ovemight. The pyridine was evaporated. The residue was purified by silica gel chromatography using a gradient of methanol il DCM from 3% to 10% to yield 57a (20 mg, 50%). ’H NMR (DMSO-d6) 5: 10.72 (s, IH),
7.88 (s, IH), 6.47 (s, 2H), 6.18-6.13 (dd, IH), 5.90-5.83 (dd, IH), 5.79-5.62 (m, 2H), 5.49-5.44 ( i, IH), 535-5.32 (d, IH), 428-4.25 (d, IH), 4.12-4.10 (d, IH), 2.60-2.45 (m, 2H), 1.12-1.09 (i η, 6H), 1.02-0.96 (m, 6H); m/z 452 (M+1).
185
EXAMPLE 55
Préparation of Compound (58a)
Hd 'F
58-1
Bntf 'F
58-3
Bnd 'F
58-4
58-5
584 [0420J Préparation of (58-2): To a solution of 58-1 (50.0 g, 205 mmol) in pyridine (250 mL) was added DMTrCl (75.0 g, 225.0 mmol). The solution was stirred at R.T. for 15 tours. MeOH (120 mL) was added, and the mixture was concentrated to dryness under reduced pressure. The residue was dissolved in EA and washed with water. The organic layer was dried over Na2SO4 and concentrated to give the crude DMTr protected dérivative (80.5 g, 89%) as a ght yellow solid. Dried KîCOj ( 80.52 g, 583.2 mmol) and then PMBQ (31.7 g, 1092 mmol) were added to a stirred solution of the DMTr protected dérivative (80 g, 146 mmol) in aihydrous DMF (300 mL). The stirring was continued at ambient température for ovemight.
The reaction was monitored by TLC. The mixture was diluted with EA and washed with water.
The organic layer was dried over Na2SO4 and concentrated to give 58-2 (98.8 g, 90%) as light yjllow solid.
[0421] Préparation of (58-3): NaH (10.4 g, 260.5 mmol) and BnBr (73.8 g, 4342 nmol) were added to a stirred solution of 58-2 (98.8 g, 147.9 mmol) in anhydrous DMF (300 n- L), and the stirring was continued at 25°C ovemight. The réaction was monitored by TLC.
186
The réaction was quenched with water, extracted with EA and washed with brine. The solvent was removed, and the residue was purified on silica get (PE: EA= 10:1 to 3:1) to give the Bn protected dérivative (tOI.l g, 90%) as a light yellow solid. The Bn protected dérivative (101.1
133.4 mmol) was dissolved in 80% HOAc (900 mL) at 25°C, The mixture was stirred at 25°C ivemight. The reaction was quenched with MeOH, and the solvent was removed to give the itcohol (42.1 g, 70%) as a white foam. To a solution of the atcohol (42.1 g, 92.6 mmol) in inhydrous CHjCN (300 mL) was added IBX (28.5 g, 121.7 mmol) at 25°C. The reaction i nixture was refluxed for 1 hour and then cooled to 0°C. The precipitate was filtered-off, and the : îltrate was concentrated to give 58-3 (39.2 g, 93%) as a yellow solid.
[0422] Préparation of (58-4): To a solution of 58-3 (39.2 g, 86.39 mmol) in 1,4(Îioxane (250 mL) was added 37% CH2O (28.1 mL, 345.6 mmol) and 2N NaOH aqueous iotution (86.4 mL, 172.8 mmol), The mixture was stirred at 25°C for 2 h and then neutralized xrith AcOH to pH = 7. To the reaction were added EtOH (200 mL) and NaBHi (19.7 g, 518.6 r imol). The mixture was stirred at 25°C for 30 mins. The reaction was quenched with saturated aqueous NH4CI. The mixture was extracted with EA, and the organic layer was dried over Na2SÛ4 and concentrated. The residue was purified by silica get column chromatography (PE: E A = 4:1 to 2:1) to give the diol dérivative (25.5 g, 55%) as a white solid. To a stirred solution 01 the diol dérivative (25.5 g, 52.5 mmol) în anhydrous pyridine (150 mL) and anhydrous C H3CN (150 mL) was added BzCl (6.6 g, 52.47 mmol) dropwise at 0°C. The mixture was then s irred at 25°C for 14 h. The réaction was quenched with H2O, and the solution was 0 meentrated. The residue was dissolved in EA and washed with NaHCÛ3. The organic layer w as dried over Na2SÛ4 and concentrated. The residue was purified on a silica gel column (PE/EA = 5:4) to give 58-4 (18. t g, 60%) as a white foam.
[0423] Préparation of (58-5): CS2CO3 (30.0 g, 92.0 mmol) and BnBr (10,4 g, 61.3 mmol) were added to a stirred solution of compound 58-4 (18.1g, 30.6 mmot) in anhydrous DMF (300 mL), and stirring was continued at 25°C ovemight. The reaction was quenched with NH4CI, extracted with EA and washed with brine. The solvent was removed to give the Bz pjotected dérivative (19.3 g, 95%) as a light yellow solid. To a stirred solution of the Bz protected dérivative (19.3 g, 28.4 mmol) in anhydrous MeOH (230 mL) was added NaOMe (24.9 g, 460 mmol) at 25°C for 1 h. The reaction was quenched with AcOH (10 mL) and concentrated. The residue was purified on a silica gel column (PE/EA = 1/2) to afford 58-5 (11.2 g, 54%) as a white solid.
[0424] Préparation of (58-6): To a stirred solution of58-5 (200 mg, 0.347 mmol) in anhydrous DCM (5 mL) was added DMP (168 mg, 0.674 mmot) at 25°C. The mixture was
187 stirred at 25°C for 2 h. The solvent was removed, and the residue was purified on a silica gel :olumn (PE: EA = 5:1 to 1:1) to give the aldéhyde dérivative (161 mg, 81%). To a stirred solution of the aldéhyde dérivative (200 mg, 0.348 mmol) in anhydrous THF (5 mL) was added VIcMgBr (1.0 mL, 1.01 mmol) at -78°C. The mixture was stirred at -78°C for 1 h. The reaction vas quenched with NH4CI and extracted with EA. The concentrated organic phase was purified )y column chromatography (PE: EA = 5:1 to 1:1) to give 58-6 (135 mg, 65%).
[0425] Préparation of (58-7): To a solution of 58-6 (900 mg, 1.5 mmol) in DCM vas added DMP (2.5 g, 6.0 mmol) at 0°C. After stirring at 0°C for 1 h, the mixture was quenched with NaîSiOj. The solvent was removed, and the residue was purified on a silica gel column (PE: EA = 5:1 to 1:1) to give the ketone dérivative (700 mg, 78%). To a solution ofthe Jketone dérivative (700 mg, 1.52 mmol) in MeOH was added NaBFL» in portions. After stirring t the same température for 1 h, the mixture was quenched with water. The solvent was emoved, and the residue was purified on a silica gel column (PE: EA = 5:1 to 1:1) to give 58-7 (500 mg, 71%).
[0426] Préparation of (58-8): To a stirred solution of DAST (1.39 g, 8.68 mmol) in î nhydrous toluene (15 mL) was added dropwise a solution of 58-6 (1.0 g, 1.73 mmol) at -78°C. he mixture was stirred at -78°C for 30 min. The solution was warmed to 25°C slowly and stirring continued overnight. The mixture was poured into a saturated Na2COj solution. The concentratedorganic phase was purified on a silica gel column (PE: EA=l0:l to 4:1) to give the f luoride dérivative (449 mg, 45%). A mixture of the fluoridc dérivative (120 g, 2.07 mmol) and CAN (3.41 g, 623 mmol) in a 3:1 solution of MeCN and water (10 mL) was stirred at 25°C ovemight. Brine (10 mL) was added, and the mixture extracted with EA The combined organic ex tracts were dried and evaporated under reduced pressure. Purification by chromatography on silica with PE: EA = 10:1 to 2:1 gave 58-8 as a yellow solid (475 mg, 50%).
[0427] Préparation of (58-9): To a stirred solution of 58-8 (550 mg, 210 mmol) in anhydrous MeCN (10 mL) were added TPSC1 (725 mg, 2.40 mmol), DMAP (293 mg, 2.40 mmol) and TEA (242 mg, 2.40 mmol) at 25°C. The mixture was stirred at 25°C ovemight. ί Ή4ΟΗ (25 mL) was added and stirred for 2 h. The solvent was removed, and the residue was purified on a silica gel column (DCM: MeOH = 10:1) to give 58-9 (300 mg). ’H NMR (CDjOD, 400 MHz) δ 7.70 (d, 8.4 Hz, IH), 7.25-7.36 (m, 10H), 6.13 (dd, J= 2.8, 16.8 Hz, IH), 5.40 (1,7 = 7.6 Hz, IH), 5.15 (m, IH), 4.81 (d, J= 11.6 Hz, IH), 4.40-4.52 (m, 4H), 3.82 (d, J= 8.8
F z, 7H), 3.62 (d, J= 9.6 Hz, 7H), 1.35 (dd, J= 2.8,14.4 Hz, 3H). ESI-MS: m/z 472,1 [M + H] +
188 [0428] Préparation of (58a): AIM boron trichloride solution in CH2CI2 (3.2 mL; J.2 mmol) was added dropwise to a solution of58-9 (200 mg, 0.42 mmol) in anhydrous CH2CI2 10 mL) at -78°C. The mixtûre was slowly (in 4 h) warmed to -30 °C and stirred at -30 to -20°C br 3 h. Ammonium acetate (I g) and MeOH (5 mL) were added, and the resulting mixture ; illowed to warm to ambient température. The solvent was removed, and residue purified by RP: 1PLC (0-60% B; A: 50 mM aqueous TEAA, B: 50 mM TEAA in MeOH) to yield 58a (75 mg). H NMR (CDjOD) Ô 7.97 (d, IH), 6.20 (dd, 1 H), 5.92 (d, 1 H), 5.22 (dt, 1 H), 4.98 (dq, 1 H), 4.58 (dd, 1 H), 3.73 (m, 2 H), 1.40 (dd, 3 H). l9F NMR (CDjOD) δ -205.80 (m, 1 F), -188.54 (m, F). ESI-MS: m/z 290.4 [M - H]'.
EXAMPLE 56
Préparation of Compound (59a) x, ΗΟΎχΚο_ HO'· -F
59-1
TBStJ F
59-2
TBSÔ 'F
59-3
ΗΟ-Χ,Ο N 'Z f / HO F υ
TBDPSO—\ o '__i O
Xh
TBDPSO-\x°x/N
HO-f’Yy °“
TBSd 'F Z TBSÔ V ° / HÔ F
59-4 59-5 59a [0429] Préparation of (59-2): To a solution of 59-1 (100.0 g, 406.5 mmol) in ïdine (750 mL) was added DMTrCl (164.9 g, 487.8 mmol). The solution was stirred at R.T.
h. MeOH (300 mL) was added, and the mixture was concentrated to dryness under reduced pressure. The residue was dissolved in EtOAc and washed with water. The organic layer was dried over Na2SO< and concentrated. The residue was dissolved in DCM (500 mL), To this solution were added imidazole (44.3 g, 650.4 mmol) and TBSCI (91.9 g, 609.8 mmol), The resulting réaction mixture was stirred at R.T. for 14 h. The reaction solution was washed vrith NaHCOj and brine. The organic layer was dried over Na2SO.j, and concentrated to give the c ude product as a light yellow solid. The crude product (236.4 g, 356.6 mmol) was dissolved in 8 )% HOAc aqueous solution (500 mL). The mixture was stirred at R.T. for 15 h. The mixture was diluted with EtOAc, washed with NaHCOj solution and brine. The organic layer was dried o rcr Na2SO< and purified on a silica gel column chromatography ( 1 -2% MeOH in DCM) to give 59-2 (131.2 g, 89.6%) as a light yellow solid. ‘H NMR (DMSO-d6,400 MHz) 611.39 (s, IH),
189
7.88 (d, J = 7.2 Hz, IH), 5.89 (dd, J= 18.0 Hz, 2.0 Hz, IH), 5.64 (d, J= 8.0 Hz, IH), 5.21 (dd, J^J2 = 7.2 Ηζ,ΙΗ), 5.18-5.03 (m, IH), 4.37-4.29 (m, IH), 3.86 (dd, J= 3.2 Hz, J= 3.2 Hz, 3H), 3.78-3.73 (m, IH), 3.51-3.56 (m, IH), 3.31 (s, IH), 0.89 (s, 9H), 0.11 (s, 6H); ESI-MS: m/z 802 [M + H]+.
[0430] Préparation of (59-3): To a solution of 59-2 (131.2 g, 364.0 mmol) in anhydrous CH3CN (1200 mL) was added IBX (121.2 g, 432.8 mmol) at R.T. The reaction mixture was refluxed for 3 h and then cooled to 0°C. The precipitatc was filtered-off, and the Eiltrate was concentrated to give the crude aldéhyde (121.3 g) as a yellow solid. The aldéhyde /as dissolved in 1,4-dioxane (1000 mL). 37% CH2O (81.1 mL, 1.3536 mol) and 2M NaOH iqueous solution (253.8 mL, 507.6 mmol) were added. The mixture was stirred at R.T. for 2 h md then neutralized with AcOH to pH = 7. To the solution were added EtOH (400 mL) and ^aBH4 (51.2 g, 1.354 mol). The mixture was stirred at R.T. for 30 mins and quenched with sat ; iqueous NH4C1. The mixture was extracted with EA. The organic layer was dried over Na2SO4 imd concentrated. The residue was purified by silica gel column chromatography (1-3% MeOH ί n DCM) to give 59-3 (51.4 g, 38.9 %) as a white solid.
[0431] Préparation of (59-4): To a solution of 59-3 (51.4 g, 131.6 mmol) in t nhydrous DCM (400 mL) were added pyridine (80 mL) and DMTrCl (49.1 g,144.7 mmol) at ( °C. The réaction was stirred at R.T. for 14 h, and then treated with MeOH (30 mL). The ί olvent was removed, and the residue was purified by silica gel column chromatography (1-3% MeOH in DCM) to give the mono-DMTr protected intermediate as a yellow foam (57.4 g, (2.9%). To the mono-DMTr protected intermediate (57.4 g, 82.8 mmol) in CH2C12 (400 mL) was added imidazole (8.4 g, 124.2 mmol) and TBDPSC1 (34.1 g, 124.2 mmol). The mixture was stirred at R.T. for 14 h. The precipitated was filtered off, and the filtrate was washed with brine and dried over Na2SO4. The solvent was removed to give the residue (72.45 g) as a white salid, which was dissolved in 80% HOAc aqueous solution (400 mL). The mixture was stirred at R.T. for 15 h. The mixture was diluted with EtOAc, washed with NaHCO3 solution and brine.
c (CD3OD, 400 MHz) 37.76 (d, J= 4.0 Hz, IH), 7.70 (dd, 1.6 Hz, J= 8.0 Hz, 2H), 7.66-7.64 (rn, 2H), 7.48-7.37 (m, 6H), 6.12 (dd, J = 2.8 Hz, J = 16.8 Hz, IH), 5.22 (d, J = 8.0 Hz, 1 3
The organic layer was dried over Na2SO4 and purified by silica gel column iromatography (1-2% MeOH in DCM) to give 59-4 (37.6 g, 84.2%) as a white solid. ’H NMR
H).5.20-5.05 (m, IH), 4.74 (dd, 5.6 Hz, J = 17.6 Hz, IH), 4,16 (d, J= 12.0 Hz, IH), 3.87,80 (m, 2H), 3.56 (d, 12,0 Hz, IH), 1.16 (s, 9H), 0.92 (s, 9H), 0.14 (s, 6H).
[0432] Préparation of (59-5): To a solution of 59-4 (3.0 g, 4.78 mmol) in anhydrous DCM (100 mL) was added Dcss-Martin periodinane (10,4 g, 23.9 mmol) at 0°C
190
under nitrogen. The reaction mixture was stirred at R.T. for 5 h. The mixture was poured into
NaHCOj and Na2S20j (1:1) aqueous solution. The organic layer was dried over anhydrous Na2SO4 and concentrated to give a residue. The residue was purified on a silica gel column [20% EtOAc in PE) to give the intermediate (2.5 g, 83.1 %) as a white solid.
[0433] To a mixture of bromotriphenyl(propyl)phosphorane (6.45 g, 16.8 mmol) in mhydrous THF (3 mL) was added t-BuOK (16.8 mL, 16.8 mmol) at 0°C under nitrogen. The eaction mixture was stirred at 0°C for 50 mins. A solution of the above intermediate (1.5 g, 2.4 nmol) in anhydrous THF (3 mL) was added dropwise at 0°C under nitrogen. The reaction i nixture was stirred at R.T. for 3 h. The reaction was quenched by NH4CI aqueous solution and 1 :xtracted with EtOAc. The organic layer was dried over anhydrous Na2SO4 and concentrated to ipve a residue. The residue was purified on a silica gel column (20% EtOAc in PE) to give 59-5
11.3 g, 83%) as a white solid.
[0434] Préparation of (59a); To a solution of 59-5 (300 mg, 0.45 mmol) in inhydrous CHjCN (2 mL) were added TPSC1 (341 mg, 1.13 mmol), DMAP (138 mg, 1.13 mmol) and NEtj (571 mg, 5.65 mmol) at R.T. The reaction mixture was stirred at R.T, for 2 h. NH4OH (1 mL) was added, and the reaction mixture was stirred for 1 h. The mixture was ( iluted with EA and washed with water. The organic layer was dried and concentrated to give a residue. The residue was purified on a silica gel column (2% MeOH in DCM) to give the cytidine dérivative (285 mg, 95.0%) as a white solid.
[0435] To a solution of the cytidine dérivative (280 mg, 0.43 mmol) in MeOH (10 nL) was added NH4F (1.0 g) at R.T.
The reaction mixture was refluxed for 12 h. The mixture
H NMR (CDjOD 400 MHz) .59 (m, 2H), 4.82-4.96 (m, IH), 4.42 (dd, J= 4.8 Hz, J= 24.4 Hz, IH), 3.72 (d, J = 12.4 Hz, v ras filtered, and the filtrate was concentrated. The residue was purified on a silica gel column (10% MeOH in DCM) to give 59a (81 mg, 61%) as a white solid.1
8.11 (d, 8.0 Hz, IH), 5.91 (dd, J= 1.2 Hz, J= 17.6 Hz, IH), 5.90 (d, J= 7.6 Hz, IH), 5.575
IH) 3.58 (d, J = 12.4 Hz, IH), 2.31-2.41 (m, 2H), 0.99 (ζ J= 7.6 Hz, 3H). ESI-TOF-MS: m/z
300.1 [M + H]+.
EXAMPLE 57
Préparation of Compound (60a)
TBDPSO
TBSO βΟ-1
191
[0436] Préparation of (60-1 ): To a solution of59-5 (450 mg, 0.69 mmol) in MeOH [10 mL) was added Pd/C (200 mg) at R.T. The reaction mixture was stirred R.T. for 1 h under rh (batloon). The mixture was filtered, and the filtrate was concentrated to give crude 60-1 (440 ng, 97.1%) as a white solid.
[0437] Préparation of (60a): To a solution of 60-1 (440 mg, 0.67 mmol) in anhydrous CHjCN (2 mL) were added TPSCI (510 mg, 1.68 mmol), DMAP (205 mg, 1,68 mmol) and NEtj (338 mg, 3.35 mmol) at R.T. The reaction mixture was stirred at R.T. for 2 h. ' ΊΗ^ΟΗ (1 mL) was added, and the reaction was stirred for 1 h. The mixture was diluted with ] IA and washed with water. The solvent was removed. The crude product was purified on a iitica gel column (2% MeOH in DCM) to give the cytidine dérivative (205 mg, 46.5%) as a white solid.
[0438] To a solution of the cytidine dérivative (205 mg, 0.31 mmol) in MeOH (6 mL) was added NH»F (0.6 g) at R.T. The reaction mixture was refluxed ovemight. After Jooling to R.T., the mixture was filtered. The filtrate was concentrated, and the residue was [urified on a silica get column (10% MeOH in DCM) to give 60a (59 mg, 62.8 %) as a white solid. *H NMR (CDjOD, 400 MHz) £8.09 (d, J = 7.6 Hz, IH), 6.01 (dd, J= 3.2 Hz, J= 15.6 Hz, IH), 5.89 (d, 7.2 Hz, IH), 4.95-5.12 (m, IH), 4.41 (dd, 5.2 Hz, J= \12 Hz, IH),
3.75 (d, J= 12.0 Hz, 1H)3.56 (d, 11.6 Hz, IH), 1.73-1.80 (m, IH), 1.55-1.63 (m, IH), 1.401.46 (m, 4H), 0.92 (t, 7.6 Hz, 3H). ESI-MS: m/z 301.8 [M + H]+.
EXAMPLE 58
Préparation of Compound (61a)
59-4
TBDPSO
î!·!
ar hydrous NaîSO», and concentrated to give the crude intermediate (1.5 g) as a white solid.
[0439] Préparation of (61-1): To a solution of 59-4 (1.5 g, 2.39 mmol) in anhydrous DCM (100 mL) was added Dess-Martin periodinane (5.2 g, 11.95 mmol) at 0°C ulder nitrogen. The reaction mixture was stirred at R.T. for 5 h. The mixture was poured into
NaHCOj and NajSîOj solution and washed with brine. The organic layer was dried with
192 [0440] To a solution of the crude intermediate (1.5 g, 2.39 mmol) in THF (12 mL) vas added méthylmagnésium bromidc (2.4 mL, 7.2 mmol) dropwise at 0°C. The resulting nixturc was stirred at 0°C for 2 h. After the starting material was consumed, the reaction was luenched with saturated NH4CI. The réaction mixture was extracted with DCM. The organic ayer was washed with brine, dried and concentrated to give crude 61-1 (1.5 g).
[0441] Préparation of (61-2): To a solution of 61-1 (1.5 g, 2.39 mmol) in anhydrous DCM (50 mL) was added Dess-Martin periodinane (4.5 g, 10.6 mmol). The reaction Iiixturc was stirred at R.T. overnight. The mixture was poured into NaHCOj and Na2S2O3 qucous solution. The organic tayer was separated, washed with brine, dried and concentrated to give a residue. The residue was purified on a silica gei column (10% EtOAc in PE) to give the intermediate (907 mg, 58.6%) as a white solid.
[0442] To a mixture of bromo(methyl)triphenylphosphorane (5.0 g, 14 mmol) in i nhydrous THF (8 mL) was added t-BuOK (12.6 mL, 12.6 mmol) at 0°C under nitrogen. The mixture was stirred at R.T. for 50 mins. A solution of the above intermediate (900 mg, 1.4 runot) in anhydrous THF (4 mL) was added dropwise at 0°C under nitrogen. The reaction mixture was stirred at R.T. for 3 h. The reaction mixture was quenched with NH4CI aqueous solution and extracted with DCM. The organic layer was separated, washed with brine, dried d concentrated to give a residue. The residue was purified on a silica gel column (5% EtOAc PE) to give 61-2 (700 mg, 78.0%) as a white solid.
[0443] Préparation of (61a): To a solution of 61-2 (298 mg, 0.46 mmol) in anhydrous CHjCN (5.5 mL) were addedTPSC1 (346.5 mg, 1.14mmol), DMAP (139.6 mg, 1.14 n imol) and NEt3 (115.6 mg, 1.14 mmol) at R.T. The reaction mixture was stirred at R.T. for 2 h. h H4OH (1 mL) was added, and the mixture was stirred for another 1 h. The mixture was diluted v ith DCM and washed with water. The organic layer was separated, washed with brine, dried . id concentrated to give a residue. The residue was purified on a silica gel column (2% MeOH il 1 DCM) to give the cytidine dérivative (250 mg, 85.0%) as a white solid.
[0444] To a solution of the cytidine dérivative (250 mg, 0.39 mmol) in MeOH (10 ni) was added NH4F (1.0 g) at R.T. The reaction was refluxed for 12 h. The mixture was fiitered, and the filtrate was concentrated. The residue was purified on a silica gel column (10% N ieOH in DCM) to give 61 a (55 mg, 49%) as a white solid. lH NMR (CDjOD, 400 MHz) £8.11 (â, J= 7.6 Hz, IH), 6.21 (dd, J= 4.2 Hz, J= 14.0 Hz, IH), 5.91 (d, J= 7.6 Hz, IH), 5.10 (dt, J=
4.
3.
Hz, J= 53.6 Hz, IH), 5.13 (brs, !H), 5.00 (brs, IH), 4.46 (dd, .7=4.8 Hz, J= 11.6 Hz, IH), 83 (d, J= 11.6 Hz, IH), 3.54 (d, 11.6 Hz, IH), 1.84 (s, 3H). ESI-MS: m/z 285.9 [M + H]+.
193
EXAMPLE 59
Préparation of Compound (62a)
TBSO'
[0445] Préparation of (62-1): To a solution of 61-2 (400 mg, 0.63 mmol) in MeOH 10 mL) was added Pd/C (400 mg) at R.T. The reaction was stirred at R.T. for 5 h under H2 balloon). The mixture was filtered, and the filtrate was concentrated to give crude 62-2 (350 mg, 87%) as a white solid.
[0446] Préparation of (62a): To a solution of 62-1 (350 mg, 0.55 mmol) in inhydrous CHjCN (6 mL) were added TPSC1 (414 mg, 1.4 mmol), DMAP (166.8 mg, 1.4 mmol) and NEtj (138.1 mg, 1.4 mmol) at R.T. The réaction mixture was stirred at R.T. for 2 h.
NH4OH (1 mL) was added, and the reaction was stirred for another 1 h. The mixture was cilutcd with EA and washed with water. The organic layer was separated, dried and concentrated to give a residue. The residue was purifîed on a silica gel column (2% MeOH in DCM) to give the cytidine dérivative (300 mg, 85%) as a white solid.
[0447] To a solution of the cytidine dérivative (300 mg, 0.47mmol) in MeOH (10
The reaction mixture was refluxed ovemight.
After eboling to R.T., the mixture was filtered. The filtrate was concentrated. The crude product was p urifîcd on a silica gel column ( 10% MeOH in DCM) to give 62a (83 mg, 61 %) as a white solid. ‘H NMR (CDjOD, 400 MHz) 58.12 (d, J= 7.6 Hz, IH), 6.22 (dd, J= 6.4 Hz, J= 12.4 Hz, IH),
Î94 (d, J =-7.6 Hz, IH), 5.25 (dt, 7=5.6 Hz, J= 54.0 Hz, 1 H), 4.38 (t, J = 4.8 Hz, 1 H), 3.72 (d, = 11.6 Hz, IH), 3.67 (d. J= 11.6 Hz, IH), 2.31-2.42 (m, IH), 0.99 (2d, J = 7.2 Hz, 6H). ESIS: m/z 287.8 [M + H]+.
184
EXAMPLE 60
Préparation of Compound (63a)
MMTru
TfO TfOMMTriJ
MMTrd 'F
63-7
MMTrd
NHDMTr
MMTru
MMTrÔ
NHDMTr
MMTrtJ 'F
63*10
TBSO Cl MMTrd
63-8
MMTrÜ 'F
63-11
[0448] Préparation of (63-2): To a solution of 63-1 (50 g, 203 mmol) in anhydrous pyridine (200 mL) was added TBDPS-C1 (83.7 g, 304 mmol). The reaction was aliowed to proceed ovemight at R.T. The solution was concentrated under reduced pressure to give a residue. The residue was partitioncd between ethyl acetate and water. The organic layer was separated, washed with brine, dried over magnésium sulfate and concentrated under reduced p -essure to give 5-OTBDPS ether as a white foam (94 g).
[0449] To a solution of the 5-OTBDPS ether (94.0 g, 1942 mmol) in anhydrous ECM (300 mL) were added silver nitrate (66.03 g, 388.4 mmol) and collidinc (235 mL, 1.94 n ol). The mixture was stirred at R.T. After most of silver nitrate was dissolved (-15 min), the n ixture was cooled to 0°C. Monomethoxytrityl chloride (239.3 g, 776.8 mmol) was added as a single portion, and the mixture was stirred ovemight at R.T. The mixture was filtered through Colite, and the fiitrate was diluted with MTBE. The solution was washed successively with IM citric acid, diluted brine and 5% sodium bicarbonate. The organic solution was dried over sc dium sulfate and concentrated under vacuum to give the fully protected intermediate as a yellow foam.
195
JO [0450] The fiilly protected intermediate was dissolved in toluene (100 mL), and the solution was concentrated under reduced pressure. The residue was dissolved in anhydrous THF (250 mL) and treated with TB AF (60 g, 233 mmol). The mixture was stirred for 2 hours at R.T., and the solvent was removed under reduced pressure. The residue was taken into ethyl acetate, and the solution was washed with saturated sodium bicarbonate and brine. Afler drying over magnésium sulfate, the solvent was removed in vacuum. The residue was purified by column :hromatography (PE: EA= 5:1,1:1) to give 63-2 (91 g, 86.4%) as a white foam.
[0451] Préparation of (63-3): To a solution of 63-2 (13.5 g, 26 mmol) in DCM (100 nL) was added pyridine (6.17 mL, 78 mmol). The solution was cooled to 0°C and Dess-Martin îeriodinane (33.8 g, 78 mmol) was added as a single portion. The reaction mixture was stirred 'or 4 h at R.T. The reaction was quenched with NaîSîOj solution (4%) and sodium bicarbonate aqueous solution (4%) (the solution was adjusted to pH 6, —150 mL). The mixture was stirred 1 br 15 min. The organic layer was separated, washed with diluted brine and concentrated under reduced pressure. The residue was dissolved in dioxane (100 mL), and the solution was treated mth 37% aqueous formaldéhyde (21.2 g, 10 eq) and 2N aqueous sodium hydroxide (10 eq). ' hé reaction mixture was stirred at R.T. ovemight. After stirring for 0.5 h at R.T., the excess of s queous sodium hydroxide was neutralized with saturated with NH4C1 (--150 mL). The mixture was concentrated under reduced pressure. The residue was partitioned between ethyl acetate and 5% sodium bicarbonate. The organic phase was separated, washed with brine, dried over magnésium sulfate and concentrated. The residue was purified by column chromatography ( VieOH: DCM= 100:1-50:1) to give 63-3 (9.2 g, 83.6%) as a white foam.
[0452] Préparation of (63-4): 63-3 (23 g, 42.0 mmol) was co-evaporated with toluene twice. The residue was dissolved in anhydrous DCM (250 mL) and pyridine (20 mL). ijhe solution was cooled to -35°C. Triflic anhydride (24.9 g, 88.1 mmol) was added dropwise over 10 mins. The reaction was stirring for 40 min at -35°C. When TLC (PE: EA= 2:1 and E CM: MeOH= 15:1) showed that the reaction was complété, the reaction was quenched with water (50 mL) at 0°C. The mixture was stirred 30 mins, extracted with EA. The organic phase was dried over Na2SO< and filtered through a silica gel pad. The filtrate was concentrated under reduced pressure. The residue was purified by column chromatography (PE: EA= 100:1-1:1) to ve 63-4 (30.0 g, 88.3%) as a brown foam.
[0453] Préparation of (63-5): 63-4 (30 g, 36.9 mmol) was co-evaporated twice with toluene. The resulting bis-triflate was dissolved in anhydrous DMF (150 mL), cooled to 0°C and treated with sodium hydride (60% in minerai oil; 1.5 g, 40.6 mmol, 1.1 eq). The reaction mixture was stirred at R.T. for 1 h until TLC (DCM: MeOH = 15:1) showed the disappearance g:
of the bis-triflate and formation of the 2,5'-anhydro intermediate. Lithium chioride (4.6 g, 110.7 ΐδβ mmol, 3 eq) was added, and the stirring was continued for 2 h. The mixture was taken into 100 mL of half saturated ammonium chioride and ethyi acetate. The organic phase was separated, vashed with diluted brine and concentrated under reduced pressure to give 63-5.
[0454] Préparation of (63-6): 63-5 was dissolved in THF (150 mL), and the solution was treated with IN aqueous sodium hydroxide (—41 mL, 40.1 mmol, I.I eq). The mixture was stirred at R.T. for I h. The reaction was monitored by LCMS. The reaction was diluted with half saturated sodium bicarbonate (-60 mL) and extracted with ethyi acetate. The organic phase was dried (magnésium sulfate) and concentrated under reduced pressure. Aurification of the residue by column chromatography (DCM: MeOH= 300:1-60:1) gave 63-6 (18.3 g, 87.6%) as a yellow foam.
[0455] Préparation of (63-7): To a solution of 63-6 (18.3 g, 32.33 mmol) in t nhydrous DCM (150 mL) was added TBS-C1 (17.7 g, 64.6 mmol) and imidazole (6.6 g, 97 mmol). The reaction was allowed to proceed ovemight at R.T. The reaction was diluted with water and extracted with DCM. The organic layer was separated, washed with brine, dried over NajSCL and concentrated. Purification of the residue by column chromatography (DCM: MeOH=300:1-80:1) gave 63-7 (18.4 g, 83.7%) as a white foam.
J0456] Préparation of (63-8): A solution of 63-7 (18.4 g, 27.1 mmol), DMAP (6.6 g, 54.0 mmol) and TEA (5.4 g,54.0 mmol) in MeCN (450 mL) was treated with 2,4,6tiispropylbenzenesulfonyl chioride (TPSC1, 16.3 g, 54.0 mmol). The mixture was stirred at
F ..T. for 3 h. NH3 H2O (70 mL) was added, and the mixture was stirred for 2 h. The solution v 'as evaporated under reduced pressure, and the residue was purified on a silica gel column ( 3 CM: MeOH= 100:1 to 15:1) to give 63-8 (18.0 g) as a light yellow solid.
[0457] Préparation of (63-9): To a solution of 63-8 (18.0 g, 26.5 mmol) in . îhydrous DCM (150 mL) was added collidine (8.1 g, 66.3 mmol, 2.5 eq), silver nitrate (4.5 g, 26.5 mmol, 1.0 eq) and DMTrCl (13.4 g, 39.7 mmol, 1.5 eq). The reaction was allowed to proceed ovemight at R.T. The mixture was filtered. The filtrate was washed with brine and extracted with DCM. The organic layer was separated, dried over Na2SÛ4 and concentrated. Tjhe residue was purified by column chromatography (PE: EA= 60:1-3:1) as a yellow foam. Îie foam was dissolved in THF (150 mL), and TBAF (10.4 g, 39.7 mmol, 1.5 eq) was added.
1e reaction was allowed to proceed ovemight at R.T. The mixture was concentrated, washed th brine and extracted with EA. The organic layer was separated, dried over Na2SÛ4 and concentrated. Purification of the residue by column chromatography (PE: EA =60:1-EA) gave
63-9 (21.3 g, 92.4%) as a yellow foam.
197
[0458] Préparation of (63-10): To a solution of 63-9 (2.0 g, 2.3 mmot) in anhydrous DCM (20 mL) was added Dess-Martin periodinane (1.95 g, 4.6 mmol) at 0°C under nitrogen. The reaction was stirred at R.T. for 5 h. The mixture was diluted with EtOAc (100 mL) and washed with a mixture of saturated aqueous Na2S2O3 and saturated aqueous NaHCO3. The crude product was purified by column chromatography on silica gel (PE: EtOAc = 2: 1) to give 63-10 (1.8 g, 90%) as a yellow solid.
[0459] Préparation of (63-11): To a solution of tetramethyl methylcnediphosphonate (390 mg, 1.68 mmol) in anhydrous THF (10 mL) was added NaH (84 mg, 2.1 mmol) at 0°C under nitrogen. The reaction was stirred at 0°C for 30 min. A solution of S3-10 (1.2 g, 1.4 mmol) in anhydrous THF (10 mL) was added dropwise at 0°C. The reaction mixture was stirred at R.T. for 1 h. The reaction was quenched by saturated aqueous NH4Q, uid the crude product was purified by column chromatography on silica gel (DCM: MeOH 150: 1) to give 63-11 (1.2 g, 88.2%) as a yellow solid. *H NMR (DMSO-d6,400 M Hz) â 8.51 I s, 1H), 7.46-7.09 (m, 22H), 6.88-6.82 (m, 6H), 6.62 (q, J/ = 17.2 Hz, J2 = 22.4 Hz, 1 H), 6.12 (d, J =72 Hz, 1H), 5.86-5.75 (m, 2H), 5.43 (d, J= 25.2 Hz, 1 H), 4.63 (dd, J =4.8 Hz, J= 21.2 Hz,
H), 4.45 (d, J= 12.0 Hz, 1H), 3.94 (d, J= 12.0 Hz, 1H), 3.72 (s, 9H), 3.53 (q, J= 11.2 Hz, J= 6.0 Hz, 6H). ESI-MS: m/z 971.59 [M + H]+.
[0460] Préparation of (63a): A solution of 63-11 (1.0 g, 1.03 mmol) in 80% HOAc
Î46 mL) was stirred at 80-90°C for 2 h. The solvent was removed, and the crude product was mrified by column chromatography on silica gel (DCM: MeOH = 20:1) to give an intermediate (337 mg, 82.3%) as a white solid. The intermediate was dissolved in MeOH and wet Pd/C (300 r ng) was added. The reaction mixture was stirred under H2 (1 atm) for 1 h and then filtered. The solvent was removed, and the residue was purified on a silica gel column (DCM: MeOH= 20:1) t j give 63a (192 mg, 63.9%) as a white solid. *H NMR (CD3OD, 400 MHz) <57.60 (d, J = 7.6
Hz, 1H), 5.87 (d, J= 7.2 Hz, 1H), 5.70(dd, J= 2.0 Hz, J= 21.6 Ηζ,ΙΗ), 5.31(m, 1H), 4.67 (dd, J = 5.6 Hz, J = 19.6 Ηζ,ΙΗ), 3.80(m, 2H), 3.75 (2d, J= 2.4 Hz, 6H), 1.92-2.20 (m, 4H). J,P NMR ( 2D3OD, 162 MHz) 6 35.77. ESI-MS: m/z 400.0 [M + H]+.
198
EXAMPLE «t
Préparation of Compound (64a)
NHDMTr
MMTrO
MMTrO
NHDMTr
[0461] Préparation of (64-2): To a solution of 64-1 (1.0 g, 4.3 mmol) in THF (20 mL) was added NaH (120 mg, 3.0 mmol), and the reaction mixture was stirred at 0°C for 1 h. Selectfluor (1.2 g, 3.4 mmol) was added into the reaction mixture. The crude product was purifîed on a silica gel column and eluted with EA to give 64-2 (500 mg, 57%) as a white solid. ’H NMR (CDjOD, 400 MHz) Ô5.65 (dt, J = 14.0 Hz, J = 44.8 Hz, IH), 3.90 (d, J = 9.6 Hz, 2H).
[0462] Préparation of (64-3): To a solution of compound 64-2 (390 mg, 1.68 mmol) in anhydrous THF (10 mL) was added NaH (84 mg, 2.1 mmol) at 0°C under nitrogen. The reaction mixture was stirred at 0°C for 30 mins. A solution of 63-10 (1.2 g, 1.4 mmol) in aihydrous THF (10 mL) was added dropwise at 0°C. The reaction mixture was stirred at R.T. for 1 h. The reaction was quenched with saturated aqueous NH4CI and concentrated to give a residue. The residue was purified on a silica gel column (DCM: MeOH13150: 1) to give crude 64-3 (1.2 g, 88.2%) as a yellow solid.
[0463] Préparation of (64a): A solution of crude 64-3 (230 mg, 0.23 mmol) in 80% EOAc (3 mL) was stirred at 80-90°C for 2 h. The crude product was purified on a silica gel column (eluted with DCM: MeOH= 20:1) to give 64a (54 mg, 53.7%) as a white solid. lH n[mR (DMSO, 400 MHz) <57.69 (d, J= 7.2 Hz, IH), 7.37 (d, J= 1.6 Hz, 2H), 6.62-6.78 (m, IH), 6.40 (d, J= 5.6 Hz, IH), 6.03-6.07 (m, IH), 5.77 (d, J = 7.6 Hz, IH), 5.61-5.64 (m, IH), 5.48-5.51 (m, IH), 4.60-4.64 (m, IH), 4.38 (d, 11.6 Hz, IH), 3.98 (d,J= 11.6 Hz, IH), 3.75 (2d, J= 11.6 Hz, 6H). ESI-MS: m/z 416.3 [M + H]+.
199
EXAMPLE «2 Préparation of Compound (65a)
64-3 65a [0464] A solution of crude 64-3 (230 mg, 0.23 mmol) in 80% HOAc (3 mL) was stirred at 80-90°C for 2 h. The crude product was purified on a silica gel column (eluted with DCM: MeOH3 20:1) to give 64a (52 mg, 33.7%) as a white solid. ’H NMR (DMSO, 400 MHz) <57.59 (d, J = 7.2 Hz, IH), 7.32 (s, 2H), 6.25-6.28 (m, IH), 5.86-6.02 (m, 2H), 5.73 (s, IH), 5.31 (d, 14.0Hz, IH), 4.72(d, J = 16.4 Hz, IH), 3.90 (d, J- 10.0Hz, IH),3.73 (2d, J = 11.6 Hz, 6H).
EXAMPLE 63
Préparation of Compound (66a)
[0465] A solution of 64a (130 mg, 0.3 mmol) in EA:MeOH (5:1,20 mL) was stirred tnder H2 (15 Psi) at R.T. for 2 h. The reaction mixture was filtered and concentrated to give a rîsidue. The residue was purified on a silica gel column (DCM: MeOH3 20: 1) to give 66a (70 ng, 54%) as a white solid. *H NMR (DMSO, 400 MHz) <57.61 (d, J-7.2 Hz, IH), 5.87 (d, J= 7.2 Hz, IH), 5.58-5.80 (m, IH), 5.26-5.47 (m, 2H), 4.97-5.03 (m, IH), 5.58-5.80 (m, IH), 3.733 .94 (m, 6H), 2.33-2.59 (m, 2H). ESI-MS: m/z 418.3 [M + H]+.
200
EXAMPLE 64
Préparation of Compound (67a) θ 2 m
1 67-2 1
NHDMTr
Pn
Λ—
MMTrd F
63-10
NHDMTr
/ Cl—' MMTrO F
67-3 [0466] Préparation of (67-2): To a solution of 67-1 (2.0 g, 6.9 mmol) in THF (20 mL) was added NaH (110 mg, 2.8 mmol), and the reaction mixture was stirred at 0°C for 1 h. Selectfluor (5.0 g, 13.6 mmol) was added into the reaction mixture. The reaction was quenched with saturated NHæ! and extracted with EA. The organic layer was separated, dried and concentrated to give the crude product. The crude product was purified on a silica gel column (eluted with EA) to give 67-2 (600 mg, 28.3%) as a white solid. *H NMR (CDjOD, 400 MHz) £ .65 (dt, 14.0 Hz, 44.8 Hz, 1 H), 4.24-4.46 (m, 8H), 1.35-1.39 (m, 12H).
[0467] Préparation of (67-3): To a solution of 67-2 (2.14 g, 7.0 mmol) in anhydrous THF (10 mL) was added NaH (84 mg, 2.1 mmol) at 0°C under nitrogen. The réaction mixture was stirred at 0°C for 30 mins. A solution of 63-10 (3.0 g, 3.5 mmol) in ; ihydrous THF (10 mL) was added in dropwise at 0°C. The reaction mixture was stirred at R.T.
τ 1 h. The reaction was quenched with saturated aqueous NH4CI and concentrated to give a a
f ' residue. The residue was purified on a silica gel column (DCM: MeOH=150: 1) to give crude 67-3 (2.9 g, 79.5%) as a yellow solid.
[0468] Préparation of (67a): A solution of crude 67-3 (1.0 g, 0.98 mmol) in 80% E OAc (25 mL) was stirred at 80-90°C for 2 h. The crude product was purified on a silica gel column (eluted with DCM: MeOH= 20:1) to give 67a (133 mg, 32.5%) as a white solid. *H NMR (DMSO, 400 MHz) £7.67 (d, 7.2 Hz, IH), 7.34 (d, J= 12.8 Hz, 2H), 6.33-6.69 (m,
IH), 6.05 (d, J = 6.8 Hz, IH), 6.00-6.05 (m, IH), 5.76 (d, 7.6 Hz, IH), 5.45-5.61 (m, IH),
0-4.63 (m, IH), 4.08-4.14 (m, 5H), 1.23-1.29 (m, 6H).3,PNMR (DMSO, 162 MHz) δ 1.93,
460
1.30. ESI-MS: m/z 466.1 [M + Na]+.
201
EXAMPLE 65
Préparation of Compound (68a)
[0469] To a solution of 67a (130 mg, 0.29 mmol) in MeOH (20 mL) was stirred mder H2 (15 Psi) at R.T. for 2 h. The reaction mixture was filtered and concentrated to give a esidue. The residue was purified on a silica gel column (eluted with DCM: MeOH= 20:1) to jive a mixture of diastereomers of 68a (90 mg, 69.2%) as a white solid. *H NMR (DMSO, 400 MHz) <57.61-7.68 (m, IH), 7.28-7.38 (m, 2H), 5.89-5.95 (m, IH), 5.58-5.79 (m, 2H), 5.18-5.39 l m, 2H), 4.53-4.85 (m, IH), 4.04-4.39 (m, 4H), 3.71-3.83 (m, 2H), 2.21-2.35 (m, 2H), 1.21-1.27 (m, 6H). 3,P NMR (DMSO, 162 MHz) Ô 18.2, 18.02, 17.73, 17.56. ESI-MS: m/z 446.1 [M + H]+
EXAMPLE 66
Préparation of Compound (69a)
Xh
MMTrÔ 'F
63-4 q ^NH —
MMTrd 'F
69-3 [0470]
A
TTO-' y_7 — MMTrÔ 'F
69-1 nh2
MMTrO' F
69-2
ΜΜΤγΟ^Χ,Ο^^ MeS-'YJ — MMTrC) nh2 . Me H s°A7N^ F HÔ 'F
69-4 6ga
Préparation of (69-1): 63-4 (3.0 g, 3.69 mmol) was co-evaporated twice w th toluene. The resulting bis-triflate was dissolved in anhydrous DMF (20 mL). The solution wits cooled to 0°C and treated with sodium hydride (60% in minerai oil; 177 mg, 0.43 mmol).
Tl.e reaction was stirred at R.T. for 1 h (TLC (PE: EA =2:1) showed complété disappearance of th< : bis-triflate and clean formation of the 2’,5'-anhydro intermediate). The réaction mixture was us :d for the next step without any further woricup
202
[0471] Préparation of (69-2): To the above stirred reaction mixture was added NaSMe (9.0 g, 0.13 mmol) and 15-Crown-5 (4.87 g, 22.14 mmol) at 0°C under nitrogen. The solution was stirred at R.T. for 2 h (TLC (PE: EA= 1:1) showed the reaction was complété). The reaction was quenched with water. The mixture was extracted by EtOAc, washed with brine, and dried over MgSO«. The mixture was filtered and concentrated to give a residue. The residue was purified on a silica gel column (PE: EA= 5:2) to give 69-2 (1.23 g, 59.0%) as a white foam.
[0472] Préparation of (69-3): To a stirred solution of 69-2 (1.34 g, 2.32 mmol) in anhydrous DCM (10 mL) was added MMTrCl (1.32 g, 4.64 mmol), AgNO3 (1.17 g, 6.96 mmol) and Cotlidine (1.41 g, 11.6 mmol) at R.T. under nitrogen. The reaction mixture was stirred at
LT. for 1 h (TLC (PE: EA= 1:1) showed the reaction was complété). The mixture was filtered and concentrated. The residue was purified on a silica gel column (PE: EA= 8:1) to give 69-3 ;i .31g, 66.5%) as a white foam.
[0473] Préparation of (69-4): To a solution of 69-3 (900 mg, 1.06 mmol) in inhydrous MeCN (9 mL) was added DMAP (259 mg, 2.12 mmol), TE A (214 mg, 2.12 mmol) ind TPSC1 (640 mg, 2.12 mmol) at R.T. under nitrogen. The reaction mixture was stirred at LT. for 2 h (TLC (DCM: MeOH=t0:l) showed the reaction was complété). NH4OH (10 mL) vas added, and the reaction mixture was stirred for another 1 h (LCMS showed the reaction was * :omptete). The solution was diluted with water, extracted with EtOAc. The organic layer was washed with IM HCl, saturated NaHCO3 and brine, and dried over MgSO.4. The mixture was : ïltered and concentrated to give a residue. The residue was purified on a silica gel column 1 DCM: MeOH= 70:1) to give 69-4 (870 mg, 68.5%) as a white solid.
[0474] Préparation of (69a): 69-4 (800 mg, 0.95 mmol) was dissolved in 80% HOAc aq. (50 mL). The réaction mixture was heated to 75°C ovemight (LCMS showed the réaction was complété). The reaction mixture was concentrated and purified on a silica gel (lolumn (DCM: MeOH= 15:1) to give 69a (180 mg, 62.5%) as a white solid. *H NMR (CD3OD, ' 00 MHz) δ 8.05 (d, J = 7.2 Hz, IH), 6.11 (dd, J =3.2 Hz J = 15.6 Hz, IH), 5.87 (d, J = 7.6 Hz, IH), 5.05 (dt, J = 4.8 Hz, J = 53.6 Hz, IH), 4.47 (dd, J =5.2 Hz J= 17.6 Hz, IH), 3.83 (d, j = 12.0 Hz, 2H), 2.84 (d, J= 14.4 Hz, 2H), 2.15 (s, 3H). ESI-MS: m/z 305.8 [M + H] *
203
EXAMPLE 67
Préparation of Compound (70a)
63-5 70a [04751 To a solution of63-5 (100 g, 182.5 mmol) in MeCN (2 L) was added 6N HCl iq. (15 g). The mixture was stirred at 40°C for 7 h, and then ncutralized to pH = 5-6 with a 15% ammonia solution (-8 g). The mixture was filtered to give a solid, which was further vashed by PE to give an intermediate (32.2 g, 60%) as a white solid. To a mixture of the intermediate (32.2 g, 109.5 mmol), TEA (22.1 g, 219 mmol) and DMAP (1.34 g, 11 mmol) in MeCN (1 L) was added with isobutyric anhydrous (69.2 g, 438 mmol). The mixture was stirred ut R.T. for 3 h. The reaction was quenched by the addition of water (200 mL) and extracted ivith 2-Mc-THF (800 mL). The organic layer was washed with saturated NaHCOî and brine. ' he organic layer was dried and concentrated to give a residue, which was purified by a silica i ;el column (10% toluene in heptane) to give 70a (42.3 g, 89%) as a white solid. *H NMR (CDjOD, 400 MHz) <57.65 (d, J= 8.0 Hz, IH), 5.95 (dd, J= 2.8, 20.4 Hz, IH), 5.55-5.74 (m, ; H), 4.33-4.41 (m, 2H), 3.88 (s, 2H), 2.57-2.72 (m, 2H), 1.14-1.22 (m, 12H).
EXAMPLE 68
Préparation of Compound (71a) —MMTrÔ 'F
63-4
MMTrd 'F
71-1
Æ
ΜΜΤγΟ-\Α»ΝΛ„ -------—
Br—0
MMTrtJ 'F
71-2
204 [0476] Préparation of (71-1): To a solution of 63-4 (4.2 g, 5.17 mmol) in DMF (50 mL) at 0°C was added NaH (227 mg of 60% dispersion, 5.7 mmol). The mixture was stirred at 0°C for 2 h ,and then LiBr (1.34 g, 15.5 mmol) was added. The mixture was stined ovemight at R.T., diluted with EA (150 mL) and washed successivety with water and brine. The organic layer was dried over Na2SO4 and concentrated. The residue was purified on a silica gel column eluted with 10% EA in PE to give 71-1 as a yellow solid (2 g, 66%) [0477] Préparation of (71-2): To a solution of 71-1 (1.74 g, 2.9 mmol) in THF (20 nL) at 0°C was added IN NaOH (32 mL, 32 mmol), and the mixture was stined at 0°C for 2 h. The mixture was partitioned between EA (100 mL) and water (20 mL), and the organic layer vas dried over Na2SO4 and evaporated to dryness. The residue was purified on a silica gel i :olumn eluted wîth 20% EA in PE to give the 5dérivative as a yellow solid (1.6 g, 90%).
[0478] To a solution of 5'-OH dérivative (2.3 g, 3.76 mmol) in anhydrous DCM (20 mL) were added cotlidine (0.8 g, 6.7 mol) and MMTrCl (2.7 g, 8.7 mmol). The reaction mixture was stined at R.T. ovemight. The mixture was filtered and washed successïvely with saturated t queous NaHCOj and brine, dried over Na2SO4 and concentrated. The residue was purified on a f îlica gel column eluted with 10% EA în PE to give 71-2 as a yellow solid (2.4 g, 73%).
[0479] Préparation of(71a): To a solution of 71-2 (2.4 g, 2.72 mmol) in anhydrous ÇHjCN (30 mL) were added TPSC1 (1.65 g, 5.44 mmol), DMAP (0.663 g, 5.44 mmol) and NEtj (1.5 mL) at R.T. The mixture was stined at R.T. for 3 h, and 28% aqueous ammonia (30 mL) liras added. The mixture was stirred for 1 h. The mixture was diluted with EA (150 mL) and vashed successïvely with water, saturated aqueous NaHCOj and brine. The solvent was removed, and the residue was purified on a silica gel column eluted with 2% MeOH in DCM to give a cytidine dérivative as a yellow solid (1.5 g, 62%).
[0480] The cytidine dérivative (1.35 g, 1.5 mmol) was dissolved in 80% AcOH (40 n lL), and the mixture was stined at 60°C for 2 h. The mixture was concentrated, and the residue was purified on a silica gel column using 5% MeOH in DCM as elute to give 71a as a white solid (180 mg, 35 %). *H NMR(MeOD, 400 MHz) 58.00 (d, 7= 7.2 Hz, IH), 6.12 (dd, 7= 3.6 Hz, 7= 15.6Hz, IH), 5.88(d,7= 7.6Hz, lH),5.10(dd,7=4.8Hz,7=53.2Hz, IH), 4.59(dd, 7 = 5.2 Hz, 7= 16.4 Hz, IH), 3.95 (d, 7= 11.6 Hz, IH), 3.76 (d, 7= 11.6 Hz, IH), 3.70 (d, 7 .6Hz, IH), 3.63 (d,7= 11.2 Hz, IH); ESI-TOF-MS:m/z337.9 [M + H]\
2ϋϋ
EXAMPLE 69
Préparation of Compound (72a)
MMTrÔ 'F
72-1
MMTrd F
63-a
MMTrd F
72-3
MMTrd F Hd ï
72-4 72a [0481] Préparation of (72-1): To a solution of 63-6 (1.0 g, 1.8 mmol ) in 1, 4< ioxane (2 mL) was added TEA (3 mL) and 37% HCHO (3 mL). The reaction mixture was stirred for 10 h at 60°C. The reaction was concentrated to dryness under vacuum, and the residue was purified by column on a silica gel column (DCM: MeOH = 100:1-30:1) to give 72-1 ÎI70 mg, 45%) as a white foam. ’H NMR (DMSO-d6, 400 MHz) δ 11.4 (s, IH), 7.27-7.49 (m, JH), 6.89 (d, J= 8.8 Hz, 2H), 4.90-4.95 (m, IH), 4.58 (dd, J= 5.2 Hz, J= 23.6 Hz, IH), 3.9607 (m, 4H), 3.73 (s, 3H), 3.50-3.62 (m, IH), 3.37-3.39 (m, IH), ESI-TOF-MS: m/z 596.9 [M + r.
(0482] Préparation of (72-2): To a solution of 72-1 (430 mg, 0.72 mmol) in dioxane (2 mL) was added 30% CHjCOOH (0.7 mL) and PtO2 (290 mg). The reaction mixture as stirred under H2 (latm) at R.T. for 2 h. The mixture was filtered, and the filtrate was c mcentrated to dryness. The residue was purified on a silica gel column (DCM: MeOH = )0:1-30:1) to give 72-2 (268 mg, 64%) as a white foam. *H NMR (DMSO-d6, 400 MHz) δ
1.3 (s, IH), 7.27-7.46 (m, 13H), 6.88 (d, J= 8.8 Hz, 2H), 5.78 (d, 20.8 Hz, IH), 5.06-5.08 (tj, J= 20.8 Hz, IH), 4.49 (dd, J= 4.2 Hz , J= 24.4 Hz, IH), 3.94-4.04 (m, 2H), 3.70 (s, 3H), 3 [M + H]+.
59-3,63 (m, IH), 3.52-3,53 (m, IH), 3.34-3.40 (m, !H), 1.66 (s, 3H). ES1-TOF-MS: m/z 580.9 [0483] Préparation of (72-3): To a solution of 72-2 (260 mg, 0.45 mmol) in ar hydrous DCM (3 mL) was added AgNOj (228 mg, 1.35 mmol), collidine (223 mg, 1.8 mmol) ai d MMTrCÏ (456 mg, 1.35 mmol). The mixture was stirred at R.T. for 10 h . The reaction mixture was filtered, and the filtrate was concentrated to dryness. The residue was purified on a ica gel column (PE: EA = 50:1-3:1) to give 72-3 (303 mg, 80%) as a white foam.
[0484] Préparation of (72-4): To a solution of 72-3 (300 mg, 0.35 mmol) in anhydrous CHjCN (3 mL) was added DMAP (107 mg, 0.88 mmol), TEA ( 141 mg, 1.4 mmo!) si:
zuo and TPSCl (106 mg, 0.35 mmol) at R.T. The reaction mixture was stirred at R.T. for 4 h. NH4OH (1 mL) was added, and the mixture was stirred at R.T. for another 1 h. The solvent was removed, and the residue was partitïoned by EA and water. The organic layer was washed by srine twice, dried and concentrated to give a residue. The residue was purified on a silica gel :olumn (PE: EA = 50:1-3:1) to give 72-4 (270 mg, 90%) as a white foam.
[0485] Préparation of (72a): 72-4 (260 mg, 0.31 mmol) in 10 mL of 60% HCOOH vas stirred at R.T. for 2 h. The solvent was removed, and the residue was washed with EA to jîve 72a (3 ! mg, 32%) as a white powder. *H NMR (MeOD, 400 MHz) δ7.85 (d, J = 0.8 Hz, IH), 6.12 (dd, J= 4.0 Hz, J= 152 Hz, IH), 5.08-5.22 (m, IH), 4.58 (dd, J= 4.8 Hz, 7= 14.8 Hz, IH), 3.92 (d, J= 15.6 Hz, IH), 3.74-3.84 (m, 3H), 1.94 (d, J= 0.8 Hz, IH). ESI-TOF-MS: m/z 307.9 [M + H]+.
EXAMPLE 70 Préparation of Compound (73a)
MMTrO
73-3 [0486]
73a
Préparation of (73-1): 63-6 (600 mg, 1.06 mmol) in formic acid (5 mL, 80% là water) was stirred at R.T. ovemight. Completion of the reaction was determined by TLC ( DCM: MeOH= 10:1). The solvent was removed to give crude 73-1 (290 mg, 93.2%).
[0487] Préparation of (73-2): To a solution of 73-1 (290 mg, 0.98 mmol) în pyridine (5 mL) and acetonitrile (5 mL) was added BzCl (371 mg, 2.65 mmol). The reaction mixture was stirred at 0°C for 0.5 h. The reaction was warmed to R.T. and stirred for 2 h.
Completion of the reaction was determined by LCMS. The reaction was quenched with water ai îd extracted with EA. The organic layer was washed with brine, dried over MgSO4, filtered aiid concentrated. The residue was purified on a silica gel column (DCM: MeOH= 200:1) to gi ve 73-2 (245 mg, 49.8%) as a white solid.
207
[0488] Préparation of (73-3): To a solution of 73-2 (245 mg, 0.49 mmol) in anhydrous acetonitrile (2.5 mL) was added TPSC1 (394 mg, 0.98 mmol), DMAP (119.5 mg, 0.98 mmol) and TEA (98 mg, 0.98 mmol). The mixture was stirred at R.T. for 3 h. NH2OH HCI (68 mg, 0.98 mmol) and DBU (368 mg, 1.47 mmol) were added, and the reaction mixture was stirred at R.T. for 2 h. The reaction mixture was diluted with water and extracted with EtOAc. The combined organic layer was washed with IM HCl, saturated NaHCOj and brine, dried and concentrated. The residue was purified on a silica gel column (DCM: MeOH= 20:1) to give 733 (49 mg, 32.9%) as a white solid.
[0489] Préparation of (73a): 73-3 (49 mg, 0.1 mmol) in NHj/MeOH (30 mL) was stirred at R.T. for 2 days. The solvent was removed. The residue was purified on a silica gel :olumn (DCM: MeOH= 30:1) to give 73a (12.9 mg, 44.0%) as a white solid. ‘H NMR (DMSOi6,400 MHz) δ 10.07 (brs, IH), 9.68 (brs, IH), 7.02 (d, J= 8.0 Hz, IH), 6.06 (dd, J= 6.4 Hz, J = 13.6 Hz, IH), 5.94 (d, 7= 5.6 Hz, IH), 5.60 (d, J= 8.4 Hz, IH), 5.36 (t,7=5.2 Hz, IH), 5.16 <dt,7= 5.2 Hz,7= 53.6 Hz, IH), 431-435 (m, IH), 3.58-3.76 (m, 2H), 3.57-3.58 (m, 2H). ESITOF-MS: m/z 308.1 [M - H]+.
EXAMPLE 71
Préparation of Componnd (74a)
MMTrÔ *F
63-6 rt*
ΜΜΤγΟΆ/°ν/Ν Ci—0
MMTrÔ *F
74-1
HÔ F
74a
MMTrÔ 'F
74-2 [0490] Préparation of (74-1): To a solution of 63-6 (1.2 g, 2.12 mmol) in anhydrous DCM (20 mL) were added collîdine (750 mg, 6.51 mol) and MMTrCl (2.6 g, 8.5 nmol). The reaction mixture was stirred at R.T. ovemight. The reaction was filtered and washed successivcly with saturated aqueous NaHCOj and brine, dried over Na2SO4 and concentrated. The residue was purified on a silica gel column eluted with 10% EA ïn PE to give
7< -1 as a yellow solid (1.4 g, 72%).
208 [0491] Préparation of (74-21: To a stirred solution of74-1 (600 mg, 0.715 mmol) in anhydrous acetonitrile (6 mL) were added TPSCt (432 mg, 1.43 mmol), DMAP (174 mg, 1.43 mmol) and TEA (144 mg, 1.43 mmol). The mixture was stirred at R.T. for 2 h. Completion of the réaction was determined by TLC (DCM: MeOH= 10:1). CH3NH2 (310 mg, 10 mmol) was added dropwise at 0°C. The reaction mixture was stirred at R.T. for 2 h. The mixture was
diluted with water and extracted with EtOAc. The combined organic layer was washed with IM HCl, saturated NaHCOj and brine. The solvent was removed, and the residue was purified by irep-TLC (DCM: MeOH= 10:1) to give 74-2 (307 mg, 50.45%) as a white solid.
[0492] Préparation of (74a): 74-2 (300 mg, 0.352 mmol) in formic acid (10 mL, i0% in water) was stirred at R.T. ovemight Completion of the réaction was determined by TLC (DCM: MeOHs 10:1). The solvent was removed to dryness. The residue was dissolved in 20 mL of methanol. Ammonia (0.5 mL) was added, and the mixture was stirred at R.T. for 5 Îins. The solvent was removed, and the residue was washed with PE (5X) to give 74a (103 mg, 5.3%) as a white solid. *H NMR (DMSO-J6, 400 MHz) <57.79 (d, J= 4.8 Hz, IH), 7.72 (d, J= 2 Hz, IH), 6.10 (dd, 4.4 Hz, J= 14.8 Hz, IH), 5.97 (brs, IH), 5.73 (d, J = 7.6 Hz, IH), 39 (brs, IH), 5.08 (dt, J= 4.2 Hz, J = 53.2 Hz, IH), 4.37-4.40 (m, IH), 3.73 (s, 2H), 3.54-3.70 J» 2H), 2.73 (d, J= 4.4 Hz, 3H). ESI-TOF-MS: m/z 308.1 [M + H]+.
209
EXAMPLE 72 Préparation of Compound (75a)
HaCfHjChz-Br
75-1
75-3
HaCtHjCîn-O^j^ODMTr
---- OH
75-4
NO-Prfe h3C(h2c)17-o<V^oh crPxo^^CN _ ” OBn *
75-5
Nfl-Prh
H3C(H2C)17-Oz*s^O'PxOx'---CN
OBn
75-6
75-7
[0493] Préparation of (75-3): To a stirred solution of 75-1 (20.0 g, 151 mmol) in aihydrous THF (200 mL)was added NaH (7.8 g, 196 mmol) in portions at 0°C. The mixture v as stirred for 1 h, and 75-2 (65.0 g, 196 mmol) was added dropwise at 0°C. The mixture was irred at R.T. for 10 h. The réaction was quenched with water and extracted with EA. The faction was washed with brine, and the organic layer was concentrated to obtain crude 75-3 (72 [0494] Préparation of (75-4): Crude 75-3 (72 g, 151 mmol) was dissolved with 80% CH3COOH (300 mL) and stirred for 10 h. The solvent was removed under reduced pressure. The residue was dissolved in EA and washed with saturated NaHCO3 and brine si ccessively. The organic layer was dried over Na2SO4 and concentrated to dryness. The residue was purified on a silica gel column to give the crude intermediate, which was dissolved anhydrous pyridine (80 mL) and DCM (400 mL). A solution of DMTrCl (56.0 g, 166 mmol) DCM (150 mL) was added dropwise at 0°C. The mixture was stirred at R.T. for 10 h. The réaction mixture was concentrated to dryness, and the residue was purified by column on silica gd (PE: EA= 2:1) to give 75-4 (58.5 g, 61%).
in in
210
[0495] Préparation of (75-5): To a stirred solution of 75-4 (10.0 g, 15.5 mmol) in anhydrous DMF (80 mL) was added NaH (0.8 g, 20 mmol) at 0°C. The mixture was stirred at R.T. for 1 h, and BnBr (33.8 g, 20 mmol) was added. The reaction mixture was stirred at R.T. for 10 h. The reaction was quenched with water and extracted with EA. The reaction was washed with brine, and the organic layer was concentrated to give the crude intermediate (10.5 g, 92%) as a white foam. The crude intermediate (10.2 g, 13.8 mmol) in 80% CH3COOH (100 mL) was stirred at R.T. for 12 h. The solvent was removed. The residue was dissoived in EA, washed with saturated NaHCO3 and brine successively, dried and concentrated to give a residue. The residue was purified on a silica gel column twice (PE: EA= 3:1) to give 75-5 (4.2 g, 70%) as a white foam.
[0496] Préparation of (75-6): To a solution of 75-5 (4.0 g, 9.2 mmol) in anhydrous
CH3CN (30 mL) was added DIPEA (6.1 g, 47.6 mmol) and 2-cyanoethyl N,Ndiisopropylchlorophosphoramidite (2.8 g, 11.9 mmol). The mixture was stirred at R.T. for 2 h. The solvent was removed, and residue was partitioned by EA and saturated NaHCO3. The organic layer was dried over MgSO< and concentrated to give a residue. The residue was jurified on a silica gel column (PE: EA= 3:1) to give 75-6 (5.1g, 88 %) as a white solid.
[0497] Préparation of (75-7): To a solution of 75-6 ( t .0 g, 1.6 mmol) and 63-9 (925 ng, 1.1 mmol) in anhydrous MeCN (1 mL) was added tetrazole (12 mL, 0.45M in MeCN, 5.5 1 nmol) dropwise at R.T. After stirred for 3 h, TBDPH (0.96 mL, 5M 4.8 mmol) was added. The 1 eaction mixture was stirred at R.T. for 1 h. The mixture was diluted with EA and washed with Saturated Na2SO3 and brine, dried over anhydrous Na2SO4 and concentrated. The residue was urifîed by silica gel chromatography (PE/EA = 50:1 to 1:1) to give 75-7 (1.1 g, 73.3%) as a white solid.
[0498] Préparation of (75a): 75-7 (1.0 g, 0.7 mmol) in 60% HCOOH (3 mL) was itirred at R.T. for 12 h. The solvent was removed. The residue was dissoived in EA and washed with saturated NaHCO3 and brine successively, dried and concentrated to give a residue. yhe residue was purified twice on a silica gel column (DCM : MeOH= 30:1) to give crude 75a (510 mg, 86%) as a white foam. To a solution of crude 75a (275 mg, 0.33 mmol) in C2H3OH v ras added a few drops IN NaOH until pH~7.0. The mixture was stirred for 0.5 h. The mixture was concentrated to give a residue. The residue was purified by HPLC (MeCN and water, neutral system) to give 75a (sodium sait, 170 mg, 64%) as a white solid. lH NMR (CD3OD, 400 hllHz) £8.01 (d, 7.6 Hz, IH), 7.23-7.37 (m, 5H), 6.22 (dd, 3.6 Hz, J= 14.4 Hz, IH), 6.01 (p, 7.6 Hz, IH), 5.01-5.16 (m, IH), 4.63-4.72 (m, 2H), 4.52-4.11 (m, IH), 4.23-4.29 (m, IH),
91-4.09 (m, 3H), 3.69-3.81 (m, 3H), 3.51-3.60 (m, 2H), 3.41-3,45 (m, 2H), 1.48-1.55 (m, 2H),
211
I
1.21-1.35 (m, 32H), 0.87-0.91 (m, 3H). 31P NMR (CDjOD, 162 MHz) δ -0.223. ESI-TOF-MS: m/z 788.3 [M-H]+.
EXAMPLE 73
Préparation of Compound (76a)
Acu
AcÔ F
Ç0 mixi [0499| Préparation of (76-1): To a solution of 73-1 (4.1 g, 13.95 mmol) in pyridine
140 mL) was added AcjO (3.13 g, 30.68 mmol) at R.T., and the mixture was stirred ovemight. ' ’he mixture was concentrated, and the residue was purified on a sitica gel column (PE: EA= : :1) to give 76-1 (4.0 g, 75.9%).
[0500] Préparation of (76-2): To a solution of 76-1 (1.3 g, 3.44 mmol) in pyridine 70 mL) was added NBS (1.22 g, 6.88mmol) at R.T., and the mixture was stirred ovemight. The ixture was concentrated, and the residue was purified on a sitica gel column (PE: EA= 4:1) to ;ive 76-2 (1.43 g, 72.2%).
[05011 Préparation of (76-3): To a solution of 76-2 (770 mg, 1.68 mmol) in dioxane (10 mL) was added MeeSn2 (Ll g, 3.36 mmol) and (PPhj^PdCh (100 mg) under N2 atmosphère. The mixture was heated at 80°C for 4 h. The mixture was concentrated, and the residue was purified on a silica gel column to give an intermediate (400 mg, 43.96%). To a solution of the intermediate (330 mg, 0.61 mmol) in anhydrous MeCN (3 mL) was added Selectflour® (462 mg, 1.34 mmol) at R.T. The mixture was stirred at R.T. for 2 days. The mixture was concentrated, and the residue was purified on a sitica gel column (PE: EA= 4:1) to g ve 76-3 (100 mg, 41.5%).
[0502] Préparation of (76a): To a solution of 76-3 (100 mg, 0.25 mmol) in MeCN (7 mL) was added DMAP (62 mg, 0.51mmol), TEA (51 mg, 0.51 mmol) and TPSC1 (153 mg, 0.51 mmol). The mixture was stirred at R.T. for 0.5 h. NHj.H2O (0.75 mL) was added. The
212
mixture was stirred at R.T. for 0.5 h. The mixture was extracted with EtOAc and washed with IN HCl and brine. The organic layer was dried and concentrated. The residue was purified on a silica gel column (PE: EA= 1:1) to give an intermediate (60 mg, 60.1%). The intermediate (50 mg, 0.13 mmol) in NHj/MeOH (5 mL) was stirred at R.T. for 3 h. The mixture was :oncentrated, and the residue was purified on a silica gel column (MeOH: DCM= 1:10) to give 76a (30 mg, 76.2%). *H NMR (CDjOD, 400 MHz) <58.25 (d, 6.8 Hz, IH), 6.09 (d, 16.0
Tz, IH), 5.00 (dt, 4.0 Hz, J= 532 Hz, IH), 4.48-4.54 (m, IH), 3.73-3.95 (m, 4H). ESI-TOFMS: m/z 312.1 [M + H]+.
EXAMPLE 74
Préparation of Compound (77a)
77-1
[0503| 77-1 (680 mg, 0.8 mmol) and triphenylphosphine (312 mg, 1.2 mmol) were dissolved in the mixture of 5 mL of dioxine and 0.25 mL of dry éthanol. A solution of diisopropyl azadicarboxylate (40% w solution in toluene, 128 mmol) in 3 mL of dioxane was added, and the mixture was stirred at R.T. for 2 h. The mixture was evaporated to dryness. The residue was dissolved in 10 mL ofTHF, cooled down to 4°C and 2 équivalents ofTBAF in THF ere added. The mixture was warmed up to R.T. and the solvent was evaporated. The resulting n îcleoside was treated with 80% HCOOH at R.T. for 3 h, and then the acid was evaporated. Lolated by isocratic silica gel chromatography using mixture of DCM (950 mL), MeOH (50 nrL), and NH<OH (2.5 mL) for elution gave 77a (80mg, 30%). H’-NMR (DMSO-D6) δ: 8.06 (s, l|l), 6.41 (s, 2H), 6.11-6.06 (dd, IH), 5.98-5.89 (dd, IH), 5.65-5.64 (d, IH), 534-526 (m, 2H),
18-5.11 (m, IH), 4.58-4.50 (dt, IH), 4.42-4.36 (q, 2H), 3.50-3.28 (m, 2H), 1.30 (t, 3H). MS:
384 (M-l+HCOOH).
5.
213
EXAMPLE 75
Préparation of Compound (78a)
HÔ* >
78-1
c ’·> TBSO F
78-3
TBSCÎ V
78-2
78-4 78-5 [0504] Préparation of (78-2): To a solution of 78-1 (10.0 g, 37.17 mmol) in mhydrous pyridine (100 mL) was added imidazole (9.54 g, 140.4 mmol) and TBSC! (21.1 g, 140.4 mmol) at 25°C. The solution was stirred at 25°C for 15 h. The solution was concentrated o dryness under reduced pressure. The residue was dissolved in EtOAc (200 mL) and washed vith water and brine. The organic layer was separated, dried over anhydrous Na2SO4 and îltcrcd. The filtrate was concentrated in vacuo to give a residue. The residue was purified by a iiîlica gel column (PE/EA = 10:1 to 2:1) to give an intermediate (11.8 g, 64%). To an ice-cold lut ion of the intermediate (t 1.8 g, 23.7 mmol) in CH2C12 (150 mL) was added a solution of pi oluenesulfonic acid monohydrate (8.2 g, 47.5 mmol) in small portion under N2. The mixture 1 vas stirred at 25°C for 30 min, and then washed with saturated aq. NaHCOj. The organic layer was separated, dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated in vacuum to give a residue, which was purified by silica gel (PE/EA = 10:1 to 1:1) to give 78-2 (6.7 g, 74%) as a solid.
[0505] Préparation of (78-3): To a solution of 78-2 (6.7 g, 17.5 mmol) in inhydrous pyridine (50 mL) was added TMSCl (2.8 g, 26.2 mmol) in small portions at 0°C i nder N2. The reaction mixture was stirred at 25°C ovemight. AgNO3 (77.8 g, 510 mmol) and MMTrCl (156.8 g, 510 mmol) in anhydrous pyridine (50 mL) was added in small portions under N2. The réaction mixture was stirred at 25°C ovemight. Ammonia (30 mL) was added, and the :action mixture was stirred for 30 min. The mixture was filtered through a Buchner funnel, and r
t îe filtrate was washed with saturated NaHCO3 solution and brine. The organic layer was separated, dried over anhydrous Na2SO4, filtered and concentrated. Chromatography on silica gjel (PE:EA = 10:1 to 2:1) gave an amine protected dérivative (6.1 g, 53%). To a solution of
Z'dine (142 mg, 1.8 mmol) in anhydrous DMSO (2 mL) at 0°C was added TFA (1.3 mg, 0.9 o!) dropwise. The mixture was stirred at 25°C until a clear solution formed. The solution
214
was then added into a solution of the amine protected dérivative (l.O g, 1.5 mmol) and DCC (0.95 g, 4.6 mmol) in anhydrous DMSO at 0°C dropwise. Stirring was continued at 25°C for 10
h. Water (10 mL) was added, and the mixture was stirred at 25°C for l h. The precipitate was removed by filtration, and the filtrate was extracted with EtOAc (20 mL). The organic layer was washed with brine (20 mL) and then dried over Na2SO4. The solvent was removed, and the residue was purified on a silica gel column (EA:PE - 10:1 to 2:1) to give the aldéhyde dérivative (850 mg, 85%). To a solution of the aldéhyde dérivative (2.6 g, 4.0 mmol) in 1,4dioxane (30 mL) was added 37% CH2O (1.3 g, 16.0 mmol) and 2N NaOH aqueous solution (3.0 mL, 6.0 mmol). The mixture was stirred at 25°C for 2 h and then neutralized with AcOH to jH=7. To the reaction were added EtOH (10 mL) and NaBHi (912 mg, 24.0 mmol). The eaction was stirred for 30 mins, and then quenched with saturated aqueous NH4CI. The mixture vas extracted wîth EA, and the organic layer was dried over Na2SO4. Purification by silica gel column chromatography (EA: PE= 10:1 to 2:1) gave 78-3 (1.1 g, 40%) as a yellow solid.
[0506} Préparation of (78-4): A stirred solution of 78-3 (685 mg, 1.0 mmol) in Eydrous CH3CN (5 mL) and anhydrous pyridine (5 mL) was cooled to 0°C. BzCl (126 mg, mmol) was added, and the reaction mixture was stirred at 25°C. After 1.5 h, water (5 mL) added. The resulting mixture was extracted with DCM (2x30 mL). The combined extracts were washed with a saturated aqueous solution of NaHCOj (20 mL), dried over MgSÛ4, and hporated under reduced pressure. The residue was purified by silica gel column amatography (DCM: MeOH = 200:1 to 50:1) to give the Bz-protected dérivative (679 mg, 86%). To a stirred solution of Bz-protected dérivative (432 mg, 0.55 mmol) in anhydrous DMF (5 mL) was added imidazole (258 mg, 3.85 mmol) and TBSC1 (240.0 mg, 1.65mmol). The mixture was stirred for 15 h. Water (10 mL) was added, and the mixture was extracted with EA. 'Aie combined extracts were washed with aqueous solution of NaHCOj (60 mL) and brine (60 niL), dried over MgSOj, and evaporated under reduced pressure to give the two-TBS protected drrivativc (680 mg, 137 %). The two-TBS protected dérivative (680 mg, 0.75 mmol) was dssolved in anhydrous CH3OH (5 mL), and NaOCHj (162 mg, 3.0 mmol) was added. The reaction mixture was stirred at 35°C for 2 h. The reaction was quenched with 80 % AcOH (3 mL) and extracted with DCM (2x50 mL). The combined extracts were washed wîth aqueous solution of NaHCOj (20 mL), dried over MgSO4, and evaporated under reduced pressure. The residue was purified by silica gel column chromatography (EA: PE = 20:1 to 3:1) to give 78-4 (239 mg, 40%) as a white foam. .
[0507] Préparation of (78-5): 78-4 (239 mg, 0.30 mmol) was co-evaporated with uene three times to remove H2O. To a solution of 78-4 in DCM (5 mL) was added DMAP to
215
(182 mg, 1.50 mmol) and TfCl (69 mg, 0.45 mmol) at 0°C under N2. The mixture was stirred
0°C for 40 mins. Completion of the reaction was determined by LCMS. The mixture was concentrated to give the crude Tf-derivative (353 mg). To a solution ofthe Tf-derivative in
DMF (5 mL) was added LiCl (31 mg, 0.76 mmol) at 0°C under N2. The mixture was stirred at
25°C for 40 mins. The mixture was washed with NaHCO3 and extracted with EA. The combined organic layer was dried over Na2SÛ4 and concentrated to give crude 78-5 (268 mg) as i light yellow oil.
[0508] Préparation of (78a): To a solution of78-5 (268 mg, 0.328 mmot) in MeOH '5 mL) was added NH4F (37 mg, 0.984 mmot) at 25°C for 4 h. The solution was filtered and waporated to dryness. The residue was dissolved in HCOOH (20 mL) and H2O (4 mL) at 25°C. The mixture was stirred at 25°C for 1 h and concentrated. The mixture was dissolved in MeCN and purified by prep-HPLC to give 78a (32 mg) as a white solid. *H NMR (MeOD, 400 MHz) δ 1.33 (s, IH), 8.20 (s, 1H), 6.32 (dd, J= 5.6, 12.4 Hz, IH), 5.77 (m, LH), 4.69 (m, 1H), 3.85 (m, H). ESI-MS: m/z 317.9 [M + H]+.
EXAMPLE 76
Préparation of Compound (79a)
.. NHMMTr
^.N
784
NHMMTr _«· TBSO F
79-1
.. NHMMTr TBDPSO-^yi TBScf *F
79-2 N NHMMTr NHj ___ '·> y >
TBSO F HO F
79-3 79a [0509] Préparation of (79-1): To a solution of 78-4 (1.1 g, 1.33 mmol) in .33 mol). The mixture was stirred at 25°C for 4 h. The solvent was removed in vacuum, and anhydrous DCM (6.6 mL) at 0°C under nitrogen was added Dess-Martin periodinane (1.45 g, 3 the residue triturated with mcthyl-t-butyl ether (30 mL). The mixture was filtered through a pad or MgSO4, and the organic solvent was stirred with an equal volume of Na2S2O3 in 30 mL of saturated NaHCO3 until the organic layer became ctear (approx. 10 min). The organic layer was separated, washed with brine, and dried over MgSO4. Prior to removing the solvent în vacuum, fi e residue was purified on a silica gel column (PE: EA= 7:1) to give 79-1 (750 mg, 75%) as a white solid.
210 [0510] Préparation of (79-2): To a stirred solution of methyl-triphenylihosphonium bromide (1.74 g, 4.89 mmol) in anhydrous THF (8 mL) was added n-BuLi (1.91 i nL, 4.89 mmol, 2.5 M in THF) at -78°C dropwise. The mixture was stirred at 0°C for 1 h. 79!(750 mg, 0.81 mmol) was added, and the mixture stirred at 25°C ovemight The réaction was uenched with saturated NH4CI (30 mL), and extracted with EtOAc (2x30 mL). The combined organic phase was washed with brine, dried with MgSO^, filtered and evaporated to dryness to give a light white solid. The solid was purified by column chromatography (PE: EA = 5:1) to iive79-2 (440 mg, 60%).
[0511] Préparation of (79-3): To a solution of79-2 (440 mg, 0.48 mmol) in MeOH (8 mL) was added Pd/C (500 mg, 10%) at R.T. under hydrogen atmosphère. The mixture was stirred at R.T. for 1.5 h. The mixture was filtered, and the filtrate was concentrated to dryness. < 'rude 79-3 (365 mg, 83%) was used for the next step without further purification.
[0512] Préparation of (79a): 79-3 (365 mg, 0.40 mmol) in MeOH (50 mL) was e dded NH4F (5.6 g, 0.15 mmol), and the solution was heated to refluxed ovemight. Completion cf the reaction was determined by LCMS. The mixture was filtered, and the filtrate was concentrated to dryness. The residue was purified on a silica gel column (PE: EA = 3:1) to give t îe amine protected dérivative (173 mg, 77%) as a white solid. The amine protected dérivative (100 mg, 0.18 mmol) in formic acid (4.4 mL) was stirred at 25°C ovemight The solution was concentration to dryness, and the residue was purified on a silica gel column (PE: EA = 1:3) to give 79a (40 mg, 90%) as a white solid. ’H NMR (400MHz, CD3OD) £8.25 (s, IH), 8.09 (s, IH), 6.14 (dd, 6.0, 12.8 Hz, IH), 5.58 (tn, IH), 4.45-4.48 (m, IH), 3.60 (q, 2H), 1.66-1.74 ( η, 2H), 0.88(t, 3H); ESI-MS: m/z 297.9 [M + H]+.
EXAMPLE 77
Préparation of Compound (80a)
NHMMTr
NHMMTr
NHMMTr
H<
BnO F
80-1
BnO F
80-2
TBSO
TBSÔ F
80-3
NHMMTr
HO F
80a
[0513] Préparation of (80-1): To a solution of 78-3 (4.4 g, 6.4 mmol) in anhydrous jyridine (5 mL) and DCM ( 25 mL). A solution of DMTrCl (2.37 g, 7.04 mmol) in DCM (5 i nL) was added dropwise at 0°C under N2. After 2 h, the reaction was quenched with CH3OH and concentrated to dryness. The residue was purified on a column of silica gel (PE: EA = 00:1 to 2:1) to obtain the DMTr protected dérivative (4.3 g, 68%). The DMTr protected dérivative (2.2 g, 2.5 mmol) in IM TB AF (2.5 mL) of THF (2.5 mL) solution was stirred at i !5°C for 3 h. The solvent was removed in vacuum, and the residue was purified by column chromatography (PE/EA= 50:1 to 1:2) to give the diol dérivative (1.86 g, 96%). To a solution ( f the diol dérivative (1.3 g, 1.5 mmol) in anhydrous THF (5 mL) was added NaH (132 mg, 3.3 iimol) at 0°C. The mixture was stirred for 1 h, and TBI (276 mg, 0.75 mmol), and BnBr (558 ng, 3.3 mmol) was added. The mixture was stirred for 10 h at 25°C. The reaction was c uenched with water, and the solvent was evaporated. The mixture was extracted with EA and brine. The organic layer was dried over Na2SO4, and evaporated to afford the crude product. The product was purified by silica gel (PE/EA = 100:1 to 3:1) to afford 80-1 (1.4 g, 90%) as a white foam.
[0514] Préparation of (80-2): To a solution of 80-1 (1.3 g, 1.23 mmol) in anhydrous DCM (17 mL) was added CI2CHCOOH (1.57 g, 12.3 mmol) at -78°C. The mixture was stirred at -20-10°C for 40 mins. The reaction was quenched with saturated NaHCOj, and diluted with DCM (50 mL). The mixture was washed with brine, and the organic solution was dried over Na2SO4 and concentrated in vacuum. The residue was purified on a silica gel column (’E/EA= 100:1 to 1:1) to give 80-2 (652 mg, 70%) as a white foam.
[0515] Préparation of (80-3): To a solution of 80-2 (630 mg, 0.84 mmol) in anhydrous DCM (5 mL) was added DAST (1.35 g, 8.4 mmol) at -78°C. The mixture was g •adually warmed to 0°C. The reaction was quenched with saturated NaHCOj. The mixture was diluted with DCM (50 mL) and washed with brine. The organic solution was dried over N a2SO4 and concentrated in vacuum. The residue was purified on a silica gel column (PE/EA = )0:1 to 2:1) to give 80-3 as a white solid (302 mg, 48%).
[0516] Préparation of (80a): A mixture of 80-3 (210 mg, 0.28 mmol) and Pd(OH)2 (200 mg) in methanol (3 mL) was stirred at 0°C at 40 psi H2 for 20 h. Pd(OH)2 was filtered off, ai id the filtrate was concentrated to dryness. The residue was purified by column (DCMZMcOH = 10:1) to give 80a (12 mg). *HNMR (400MHz, CDjOD) £8.33 (s, IH), 8.20 (s, IH), 6.33 (dd, J = 6.0, 13.2 Hz, IH), 5.79 (t, J= 5.6 Hz, IH), 5.66 (t, J= 5.2 Hz, IH), 4.52-4.80 (m, 3H), 3.8082 (m, 2H). ESI-MS: m/z 302.0 [M + H]+.
an
3.
218
EXAMPLE 78
Préparation of Compoand (81a)
ua-A O. n”
zNH
\ nh3 rasd' 'f nh2
H
[0517]
Préparation of (81-2): To a solution of 81-1 (20.0 g, 70.2 mmol) in inhydrous pyridine (200 mL) was added imidazole (19.1 g, 280 mmol) and TBSCt (42.1 g, 281 nmot) at 25°C. The solution was stirred at 25°C for 15 h, and then concentrated to dryness inder reduced pressure. The residue was dissolved in EtOAc and then filtered. The filtrate was concentrated to dryness to give the TBS protected dérivative (36.4 g, 99%). The TBS protectcd dérivative (36.5 g, 71.1 mmol) was dissolved in THF (150 mL). HjO (100 mL), and then AcOH Î00 mL) were added. The solution was stirred at 80°C for 13 h. The réaction was cooled to
T., and then concentrated to dryness under reduced pressure to give 81-2 (31.2 g, 61%) as a white solid.
[0518] Préparation of (81-3): To a solution of 81-2 (31.2 g, 78.2 mmol) in anhydrous pyridine (300 mL) was added AciO (11.9 g, 117.3 mmol). The mixture was stirred at for 18 h. MMTrCl (72.3 g, 234.6 mmol) and AgNOj (39.9 g, 234.6 mmol) were added, and the solution was stirred at 25°C for 15 h. H2O was added to quench the reaction and the solution was concentrated to dryness under reduced pressure. The residue was dissolved in EtOAc and washed with water. The organic layer was dried over NaiSO< and filtered. The filtrate was concentrated in vacuum to give a residue, which was purified by silica gel (DCM:MeOH = 200:1 to 50:1) to give the MMTr protectcd amine derivative (35.2 g, 63%). T lie MMTr protected amine derivative (35.2 g, 49.3 mmol) was dissolved in NH/MeOH (300 nL). The mixture was stirred at 25”C for 20 h. The solution was evaporated to dryness, and purified by a silica gel column (DCM: MeOH = 100:1 to 50:1) to give 81-3 as a yellow solid (28.6 g. 87%).
219
[0519] Préparation of (81-4): To a solution of 81-3 (12.0 g, 17.9 mmol) in anhydrous DCM (200 mL) was added Dess-Martin periodinane (11.3 g, 26.8 mmol) at 0°C. The mixture was stirred at 0°C for 2 h, and then at R.T. for 2 h. The mixture was quenched with a saturated NaHCOj and Na2S20j solution. The organic layer was washed with brine (2X) and dried over anhydrous Na2SO4. The solvent was evaporated to give the aldéhyde (12.6 g), which was used directly in the next step. To a solution of the aldéhyde (12.6 g, 18.0 mmol) in 1,4dioxane (120 mL) was added 37% HCHO (11.6 g, 144 mmol) and 2N NaOH aqueous solution (13.5 mL, 27 mmol). The mixture was stirred at 25°C ovemight. EtOH (60 mL) and NaBfL (10.9 g, 288 mmol) were added, and the reaction was stirred for 30 mins. The mixture was quenched with saturated aqueous NH4CI, and then extracted with EA. The organic layer was dried over Na2SO4, and purified by silica gel column chromatography (DCM: MeOH = 200:1 to 50:1) to give 81-4 (7.5g, 59%) as a yellow solid.
[0520] Préparation of (81-5): To a solution of 81-4 (3.8 g, 5.4 mmol) in DCM (40 nL) was added pyridine (10 mL) and DMTrCl (1.8 g, 5.4 mmol) at 0°C. The solution was ütirred at 25°C for 1 h. MeOH (15 mL) was added, and the solution was concentrated. The iesidue was purified by silica gel column chromatography (DCM: MeOH = 200:1 to 50:1) to ;ive the MMTr protected dérivative (3.6 g, 66%) as a yellow solid. To a solution of the MMTr irotected dérivative (3.6 g, 3.6 mmol) in anhydrous pyridine (30 mL) was added TBDPSC1 (2.96 g, 10.8 mmol) and AgNO3 (1.84 g, 10.8 mmol). The mixture was stirred at 25°C for 15 h. Ί lie mixture was filtered and concentrated. The mixture was dissolved in EtOAc and washed y rith brine. The organic layer was dried over Na2SO4., and then purified by silica gel column Ïomatography (DCM: MeOH = 200:1 to 50:1) to give the TBDPS protected dérivative (3.8 g, 1%) as a solid. To a solution of the TBDPS protected dérivative (3.6 g, 2.9 mmol) in aihydrous DCM (50 mL) was added CtjCHCOOH (1.8 mL) in anhydrous DCM (18 mL). The mixture was stirred at -78°C for 1 h. CtjCHCOOH (3.6 mL) was added at -78°C. The mixture as stirred at -10°C for 30 mins. The mixture was quenched with saturated aqueous NaHCOj a îd extracted with DCM. The organic layer was dried over Na2SO4, and then purified by silica gel column chromatography (DCM: MeOH = 200:1 to 50:1) to give 81-5 (22 g, 80%).
[0521] Préparation of (81-6): To an ice cooled solution of 81-5 (800 mg, 0.85 m mot) in anhydrous DCM (20 mL) was added pyridine (336 mg, 4.25 mmol) and Tf2O (360 mg,
1.28 mmol) dropwise. The réaction mixture was stirred at 0°C for 15 mins. The reaction was quenched by ice water and stirred for 30 mins. The mixture was extracted with EtOAc, washed wth brine (50 mL) and dried over MgSO4. The solvent was evaporated to give the crude bi.î(triflate) dérivative. To the bis(triflatc) dérivative (790 mg, 0.73 mmol) in anhydrous DMF
220 (35 mL) was added LiCl (302 mg, 7.19 mmol). The mixture was heated to 40°C and stirred ovemight. Completion of the reaction was determined by LCMS. The solution was washed with brine and extracted with EtOAc. The combined organic layers were dried over MgSO4, and the residue was purified on a silica gel column (DCM/MeOH = 100:1) to give 81-6 (430 mg, 61%).
[0522] Préparation of (81a): To 81-6 (470 mg, 0.49 mmol) in MeOH (85 mL) was added NH4F (8.1 g, 5.92 mmol), and the solution was heated to reflux ovemight. The mixture was filtered, and the filtrate was concentrated to dryness. The residue was purified on a silica gel column (DCM/MeOH = 20:1) to give the diol (250 mg, 84%) as a white solid. The diol (130 mg, 0.21 mmol) in formic acid (5 mL) was stirred at 25°C ovemight. The solution was concentration to dryness, and the residue in MeOH (30 mL) was stirred at 70°C ovemight. Completion of the réaction was determined by LCMS and HPLC. The solvent was removed, md the crude product was washed with EtOAc to give 81a (58 mg, 81%) as a white solid. *H NMR (DMSO-rfi, 400 MHz) <510.73 (br, 1H), 7.98 (s, 1H), 6.58 (br, 2H), 6.08 (q, J = 4.8, 9.2 Tz, 2H), 5.64 (dt, J= 5.6,52.8 Hz, 1H), 5.40 (m, 1H), 4.52 (m, 1H), 3.80-3.82 (m, 2H), 3.64 (q, >H). ESI-MS: m/z 333.8 [M +H]+, 666.6 [2M +H]+
EXAMPLE 79
Préparation of Compound (82a)
[0523] Préparation of (82-1): To a solution of 81-4 (310 mg, 0.33 mmol) in anhydrous DCM (10 mL) was added pyridine (130 mg, 1.65 mmol) and Tf2O (139 mg, 0.49 mmol) diluted by DCM dropwise at 0°C. The mixture was stirred at 0°C for 15 mins. The réaction was quenched with ice cold water. The organic layer was separated and washed with brine. The organic layer was dried over Na2SÛ4 and evaporated to give to give the triflate dérivative (420mg crude), which was used directly in the next step. To a solution of the triflate d ïrivative (420 mg crude) in anhydrous pentan-2-one was added Nal (396 mg, 2.64 mmol). The
221 mixture was stirred at 40°C for 3 h, and then dissolved with EtOAc. The organic layer were washed with Na2S2O3 twice and washed with brine. The organic layer was dried over Na2SO4 and evaporated to give a residue. The residue was purified by a column (DCM: MeOH = 300:1 to 100:1) to give 82-1 (195 mg, 56% for two steps).
[0524] Préparation of (82-2): To a solution of 82-1 (650 mg, 0.62 mmol) in MeOH (10 mL) was added NH4F (45.8 g, 12.4 mmol). The mixture was refluxed ovemight. The mixture was filtered and evaporated to dryness. The residue was purified on a silica gel column (DCM/MeOH = 200:1 to 20:1) to give 82-2 (250 mg, 58%).
[0525] Préparation of (82-3): To a stirred solution of 82-2 (300 mg, 0.43 mmol), EtiN (217 mg, 2.15 mmol) in anhydrous MeOH (10 mL) was added 10% Pd/C (50 mg). The mixture was stirred in a hydrogénation apparatus (30 psi hydrogen) at R.T. ovemight. The ;atalyst was filtrated off, and the filtrate was evaporated to give a residue. The residue was nirified on a silica gel column (DCM/MeOH « 200:1 to 20:1) to afford 82-3 as a white solid <180 mg, 73%).
[0526] Préparation of (82a): 82-3 (110 mg, 0.19 mmol) was dissolved in HCOOH H8 g) and H2O (6 g) at 25°C, and stirred for 1 h. The solution was evaporated to dryness, dissolved in MeOH (30 mL). The mixture was stirred at 60°C for 12 h. The solution was ivaporated to dryness, and dissolved in EtOAc (50 mL). The mixture was stirred at 60°C for 1 h. The mixture was filtered and washed with EtOAc to give 82a as a white solid (45.3 mg, i 0%). *H NMR (400MHz, MeOD) <78.00 (s, IH), 6.11-6.15 (m, IH), 5.35-5.50 (m, IH), 4.53<59 (m, IH), 3.54-3.64 (m, 2H), 1.26 (s, 3H). ESI-MS: m/z 299.76 [M + 1]\ 598.66 [2M + 1]\
EXAMPLE 80
Préparation of Compound (83a)
83-3 83a [0527] Préparation, of (83-1): 81-1 (5.7 g. 20 mmol) was co-evaporated with p; rridine three times, and then dissolved in pyridine (20 mL). The mixture was cooled to 0°C ai td Ac2O (5.8 mL, 60 mmol) was added dropwise. The mixture was stirred at 25°C for 10 h,
222
and then cooied to 0°C. AgNOj (8.5 g, 50 mmol), and then MMTrCl (15.5 g, 50 mmol) were added in portions. The mixture was stirred at 25°C for 10 h. The reaction was quenched with saturated NaHCOj and extracted with EA. The organic layer was dried over Na2SÛ4 and concentrated. The residue was purified by silica gel column chromatography (DCM/MeOH = 100:1 to 50:1) to afford the Ac protected dérivative (12.1 g, 93%) as a light yellow solid. The Ac protected dérivative (12.1 g) was dissolved in methanolic NHj (saturated). The mixture was stirred at 25°C for 14 h. The solvent was removed, and the residue was purified on a silica gel column (DCM/MeOH = 80:1 to 30:1) to give 83-1 (9.2 g, 87%).
[0528] Préparation of (83-2): To a stirred solution of 83-1 (9.2 g, 16.5 mmol) in dry THF (300 mL) was added imidazole (9.0 g, 132 mmol) and PPhj (34.8 g, 132 mmol). A solution of I2 (26.0 g, 103 mmol) in THF (100 mL) was added dropwise under N2 at 0°C. The mixture was stirred at 25°C for 18 h and then quenched with a Na2S20j solution. The mixture was extracted with EtOAc. The organic layer was dried over Na2SÛ4 and concentrated. The residue was purified on a silica gel column (DCM/MeOH = 80:1 to 30:1) to give the iodide lerivative (10.3 g, 93%) as a light yellow solid. To a stirred solution of the iodide dérivative 102 g, 15.3 mmol) in dry THF (300 mL) was added DBU (4.7 g, 30.1 mmol). The mixture was ütirred at 60°C for 8 h. The solution was diluted with a NaHCOj solution and extracted with
ZtOAc. The organic layer was dried over,Na2S04 and concentrated. The residue was purified < m a silica gel column (PE/EtOAc= 3:1 to 1:3) to afford 83-2 (6.2 g, yield 76%).
[0529] Préparation of (83-3): To a stirred solution of 83-2 (5.42 g, 10 mmol) in anhydrous CHjOH (100 mL) was added PbCOj (13.7 g, 53.1 mmol). A solution of h (12.3 g, ^8.9 mmol) in CHjOH (300 mL) was added dropwise at 0°C. The mixture was stirred at 25°C for 10 h. The solution was quenched with a Na2S20j solution and extracted with DCM. The rganic layer was washed with a NaHCOj solution, dried over Na2SÛ4 and concentrated to give residue. The residue was purified by HPLC (0.1% HCOOH in water and MeCN) to give the desired methoxyl dérivative (2.4 g, 34%). To a stirred solution of the methoxyl dérivative (2.4
3.4 mmol) in dry pyridine (20 mL) was added BzCl (723 mg, 52 mmol) dropwise at 0°C. The mixture was stirred at 0°C for 1 h. The solution was quenched with a NaHCOj solution and !5-crown-5 (5.5 g, 25 mmol) were suspended in DMF (50 mL). The mixture was stirred at 110I25°C for 5 days. The precipitate was removed by filtration, and the filtrate was diluted with EÀ. The solution was washed with brine and dried over Na2SÛ4. The solvent was removed, extracted with EtOAc. The organic layer was dried over Na2SO4 and concentrated. Purified by a ùlica gel column (ΡΕ/EtOAc = 5:1 to 1:1) afforded 83-3 (2.1 g, 77%) as a white solid.
[0530] Préparation of (83a): 83-3 (2.0 g, 2.5 mmol), BzONa (3.6 g, 25 mmol) and
223 and the residue was purified on a silica gel column (PE/EA = 10/1 to 2/1) to afford the crude Bz protected dérivative (1.6 g, 80%). The Bz protected dérivative (1.6 g, 2.0 mmol) was dissolved in methanolic ammonia (100 mL), and the mixture was stirred at 25’C for 20 h. The solvent was removed, and the residue was purified by a silica gel column (DCM/MeOH = 100:1 to 20:1) to the diol dérivative as a white solid (410 mg, 35%). The diol dérivative (200 mg, 0.34 mmol) was dissolved in HCOOH (24 g) and H2O (6 g) at 25’C, and the mixture was stirred at 25°C for
h. The solution was evaporated to dryness, and dissolved in MeOH (30 mL). The mixture was stirred at 60°C for 12 h. The solution was evaporated to dryness and dissolved in EtOAc [50 mL). The mixture was stirred at 60’C for 1 h. The mixture was then filtered and washed
I with EtOAc to give 83a as a white solid (46.1 mg, 43%). *H NMR (CDjOD, 400MHz) <57.92 (s,
LH), 6.22 (dd, J= 1.6, 18.8 Hz, IH), 5.17-532 (m, IH), 4.89-4.91 (m, IH), 3.77 (m, 2H), 3.44 s, 3 H). ESI-MS: m/z 316.1 [M + H]+.
EXAMPLE 81
Préparation of Compound (84a)
Bztf 'F
84-1
84-7 84-8
[0531] Préparation of (84-2): To a stirred solution of 84-1 (100.0 g, 265.9 mmol) in dry THF (1000 mL) was added Li(O-t-Bu)jAlH (318.9 mL, 318.9 mmol) at -78’C under N2. The mixture was stirTed at -78°C for 1 h and then at R.T for 1 h. The reaction mixture was cooled to -5 O’C and quenched with ice and a saturated NH4CI solution. The mixture was extracted with EtOAc. The organic layer was dried over Na2SÛ4 and concentrated to afford the
224 »5
V-OH dérivative (100.5 g) as a white solid. To a stirred solution of the Γ-OH dérivative (100.5 g, 265.9 mmol) in dry DCM (600 mL), NEtj (110 mL) and MsCl (45.5 g, 298.0 mmol) were added dropwise at 0°C. The mixture was stirred at R.T. for 2 h. The mixture was quenched with ice water at 0°C and extracted with DCM. The organic layer was dried over Na2SO4, :oncentrated and purified on a silica gel column (PE: EA = 50:1 to 5:1) to afford 84-2 (113.4 g, yieid: 93.9%) as a white solid.
[0532] Préparation of (84-3): To a suspension of compound 6-chloro-9/7-purin-2imine (70.1 g, 414.7 mmol), HMDS (480 mL) and (NH4)2SO4 (0.8 g) was added dry DCE (400 nL). The mixture was refluxed under N2 for 18 h and then cooled to R.T. To the silylated 2umino-6-chloropurine solution was added 84-2 (78.0 g, 171.Immol) and TMSOTf (60 mL, 331.9 mmol). The mixture was refluxed ovemight, concentrated and neutralized with a NaHCOj solution, The resulting precipitate was filtered, and the filtrate was extracted with EtOAc. The organic layer was dried over Na2SO4 and concentrated. Chromatography on a î ilica gel column (PE: EA = 5:1 to 2:1) gave 84-3 (10.8 g, yieid: 11.9%) as a light yellow solid.
[0533] Préparation of (84-4): To a suspension of 84-3 (30.0 g, 56.6 mmol) in DCM (300 mL) were added MMTrCl (34.9 g, 113.2 mmol) and AgNOj (19.3 g, 1132 mmol). The réaction mixture was cooled to 0°C, and collidine (18.0 g, 150 mmol) was added. The resulting suspension was stirred at R.T. for 12 h. The suspension was filtered. The filtrate was extracted m rith DCM and washed with a NaHCOj solution. The organic layer was dried over Na2SO4 and concentrated. Purification by a silica gel column (PE: EA = 20:1 to 3:1) to give 84-4 (35.0 g, ytcld: 77.9%) as a light yellow solid. *H NMR (CDCIj. 400 MHz) δ 7.94-7.96 (m, 4H), 7.057Î58 (m, 18H), 6.62-6.67 (m, 2H), 6.55 (dd, J= 6.0 Hz, 7= 9.6 Hz, IH), 5.60-5.66 (m, IH), 4
69-4.76 (m, 2H), 4.55^.58 (m, IH), 3.64 (s, IH). ESI-MS: m/z 802 [M + HJ*.
[0534] Préparation of (84-5): To a stirred solution of 84-4 (35.0 g, 43.6 mmol) in dpr MeOH (400 mL) was added NaOMe (23.5 g, 436 mmol) and 2-mercapto-ethanol (30.6 g, 3
T col )2.4 mmol). The mixture was refluxed ovemight. The pH was adjusted to 9-10 with CO2. 1e precipitate was filtered, and the filtrate was concentrated. Purification on a silica gel côlumn (PE: EA = 10:1 to 1:1) gave pure 84-5 (24.0 g, yieid 95.7%) as a light yellow solid.
[0535] Préparation of (84-6): To a solution of 84-5 (24.0 g, 41.7 mmol) in pyridine (250 mL) was added DMTrCl (28.2 g, 83.5 mmol) at 0°C. The solution was stirred at R.T. for
h. MeOH (50 mL) was added, and the mixture was concentrated to dryness under reduced pressure. The residue was dissolved in EtOAc and washed with water. The organic layer was dried over Na2SO4, filtered, concentrated and purified by a silica gel column (DCM: MeOH =
2( 0:1 to 50:1) to give a first intermediate (27.6 g) as a yellow solid. To a solution of the first
225 intermediate (27.6 g, 31.5 mmol) in DCM (200 mL) was added imidazole (4.3 g, 63 mmol) and TBSCl (9.5 g, 63 mmol). The mixture was stirred at R.T. for 12 h. The solution was washed with NaHCOj and brine. The organic layer was dried over Na2SO4, filtered, concentrated and purified by a silica gel column (DCM: MeOH = 200:1 to 100:1) to give a second intermediate (30.2 g) as a yellow solid. To a solution of the second intermediate (30.2 g, 30.4 mmol) in anhydrous DCM (50 mL) was added C12CHCOOH (20 ml) in anhydrous DCM (500 mL). The mixture was stirred at -78°C for 1 h. C12CHCOOH (30 mL) was added at -78°C. The mixture was stirred at -20°C for 2 h. The mixture was quenched with saturated aqueous NaHCOj and extracted with DCM. The organic layer was dried over Na2SO4, and then purified by a silica gel column (DCM: MeOH = 200:1 to 30:1) to give 84-6 (18.0 g, 62.5%) as a white solid. *H NMR (400MHz, MeOD) <58.27 (s, IH), 7.16-7.38 (m, 12H), 6.79-6.83 (m, 2H), 6.42 (dd, 4.4 Hz, J = 10.0 Hz, IH), 4.54-4.62(m, IH), 3.92 (d, 8.8 Hz, 2H), 3.74 (s, 3H), 3.70-3.72 (m, IH),
0.92 (s, 9H), 0.11-0.13 (m, 6H). ESI-LCMS: m/z 690.0 [M + H]+.
[0536] Préparation of (84-7): 84-6 (7.0 g, 10.0 mmol) was added to a suspension of DMP (10.6 g, 25 mmol) in anhydrous CH2Ch (100 mL) at 0°C. The mixture was stirred at 25°C 'or 2 h. The solvent was removed in vacuo, and the residue triturated with diethyl ether (100 nL). The mixture was filtered through a pad of MgSCU The organic solvent was stirred with În equal volume of Na2S2Oj.5H2O in 100 mL of saturated NaHCOj until the organic layer ecame clear (10 min). The organic layer was separated, washed with brine, and dried over 1gSO4. The solvent was removed in vacuo to give a third intermediate as a red solid (6.5 g, 95%). To a solution of the third intermediate (6.5 g, 9.5 mmol) in 1,4-dioxane (80 mL) was î dded 37% CH2O (6.0 mL, 60 mmol) and 2N NaOH aqueous solution (9.5 mL, 19 mmol). The i lixturc was stirred at 25°C for 2 h and then neutralized with AcOH to pH 7. EtOH (30 mL) and NaBH4 (3.8 g, 100 mmol) were added, and the mixture was stirred for 30 mins. The mixture was quenched with saturated aqueous NH4CI, and then extracted with EA. The organic layer was dried over Na2SO4. Purification by a silica gel column (DCM: MeOH = 200:1 to 30:1) gave 84-7 (4.2 g, 58.3%) as a yellow solid.
[0537] Préparation of (84-8): To a solution of 84-7 (4.2 g, 5.8 mmol) in DCM (50 mL) was added pyridine (5 mL) and DMTrCl (1.9 g, 5.8 mmol) at -20°C. The solution was s irred at 0°C for 2 h. The reaction mixture was treated with MeOH (15 mL), and then o me entra tcd. The residue was purified by a silica gel column (DCM: MeOH = 200:1 to 50:1) to ve the fourth intermediate (1.3 g) as a yellow solid. To a solution of the fourth intermediate (' .3 g, 1.3 mmol) in anhydrous pyridine (15 mL) was added TBDPSCl (1.1 g, 3.9 mmol) and AgNOj (0.68 g, 4.0 mmol). The mixture was stirred at 25°C for 15 h. The mixture was filtered,
226 concentrated, dissolved in EtOAc and washed with brine. The organic layer was dried over Na2SO4. Purification by a silica gel column (DCM: MeOH = 200:1 to 100:1) gave a fîfth intermediate (1.4 g) as a solid. To a solution of the fîfth intermediate (1.4 g, l.l mmol) in anhydrous DCM (50 mL) was added ChCHCOOH (0.7 ml) in anhydrous DCM (18 mL). The 5 mixture was stirred at -78°C for 1 h. C12CHCOOH (1.5 ml) was added at -78°C, and the mixture was stirred at -20°C for 1.5 h. The mixture was quenched with saturated aqueous NaHCOj and extracted with DCM. The organic layer was dried over Na2SO4. Purification by a silica gel column (DCM: MeOH = 200:1 to 50:1) gave 84-8 (650 mg, 11.6%) as a white solid.
[0538] Préparation of (84-9): To a solution of pyridine (521 mg, 6.59 mmol) in anhydrous DMSO (5 mL) was added TFA (636 mg, 5.58 mmol) dropwise at 10°C under N2. The mixture was stirred until a clear solution formed. To this solution (0.8 mL) was added a mixture of 84-8 (650 mg, 0.68 mmol) and DCC (410 mg, 2.0 mmol) in anhydrous DMSO (5 mL) at R.T. under N2. The mixture was stirred at 20 °C ovemight. Water (30 mL) was added. The mixture was diluted with DCM (30 mL) and filtered. The filtrate was extracted with DCM.
The organic layers were washed with saturated aqueous NaHCOj, dried over Na2SÛ4 and concentrated in vacuo. The crude product was purified on a silica gel column (PE: EA = 10:1 to 1:1) to give the sixth intermediate (600 mg) as a yellow solid. To a stirred solution of Methyltriphenyl-phosphonium bromide (714 mg, 2.0 mmol) in anhydrous THF (5 mL) was added nBuLi (0.8 mL, 2.0 mmol, 2.5 M in THF) at -78°C dropwise over 1 min. Stirring was continued at 0 °C for 1 h. The sixth intermediate (600 mg, 0.63 mmol) was added to the mixture, and the mixture was stirred at 25°C for 15 h. The reaction was quenched with saturated NH4CI (20 mL) and extracted with EtOAc. The combined organic phase was dried with Na2SO4, filtered and evaporated to dryness to give a light yellow oil. The oil was purified by column chromatography (DCM: MeOH = 200:1 to 50:1) to give 84-9 (250 mg, 38.5%) as a yellow solid.
[0539] Préparation of (84-10): 84-9 (250 mg, 0.26 mmol) was dissolved in THF (5.0 mL). TBAF (131 mg, 0.5 mmol) was added at 20°C, and stirring was continued for 2 h.
The solution was evaporated to dryness. The residue was dissolved in EA (50 mL) and washed with water (2X). The solution was evaporated to dryness, and purified by a silica gel column (PE: EA = 10:1 to 12) to give 84-10 (57.6 mg, 36.9%) as a white solid. lH NMR (400MHz, 30 MeOD) δ 8.34 (s, IH), 7.15-7.38 (m, 12H), 6.79-6.82 (m, 2H), 6.44 (dd, 2.0 Hz, 10.0 Hz,
IH), 6.01 (dd, J= 11.2 Hz, J= 17.6 Hz, IH), 5.51 (dd, J= 1.6 Hz, J= 17.2 Hz, IH), 5.35 (dd, J=
1.6 Hz, J= 17.2 Hz, IH), 4.68-4.76 (m, IH), 3.74 (s, 3H), 3.63(dd, 2.0 Hz, J= 12.8 Hz, IH)
3.52(dd, J= 2.0 Hz, J= 12.8 Hz, IH). ESI-LCMS: m/z 602.0 [M + H]+.
227 [0540] Préparation of(84a): A solution of 84-10 (27 mg) in 1.5 mL of 80% formic acid stood at R.T. for 4.5 h and then concentrated to dryness. The residue was mixed with water and lyophilized. MeOH (1.5 mL) and TEA (0.1 mL) were added, and the mixture was concentrated. The precipitate from MeOH and EtOAc was filtered and washed with EtOAc to give 84 (9.3 mg) as a slightly-amber solid. ’H NMR (CD3OD, 400 MHz) <58.44 (s, IH), 6.57 (d, J= 10.8 Hz, IH), 6.05 (dd, J = 17.6 Hz, 10.8 Hz, IH), 5.45 (dd, J= 17.6 Hz, J= 1.6 Hz, IH), 5.37 (dd, J= 10.8 Hz, 1.6 Hz, IH), 4.78 (dd, J= 18.4 Hz, 17.2 Hz, IH), 3.67 (d, J= 12.4 Hz, IH), 3.56 (dd, J= 12.4 Hz, 2.0 Hz, IH); ESI-MS: m/z 328.4 [M -Hf.
EXAMPLE 82
Préparation of Compound (85a)
«5-2 85a [0541] Préparation of (85-2): A mixture of 85-1 (200 mg; 0.22 mmol) in pyridine (2.5 mL) and isobutyric anhydride (44 gL; 1.2 equiv) was stirred R.T. ovemight. The mixture was concentrated, and the residue partitioned between EtOAc (50 mL) and water. The organic layer was washed with IN citric acid, water, saturated aqueous NaHCOj and brine. The mixture was dried with Na2SO4. The solvent was evaporated and the residue was purified on a silica column (10 g column) using hexanes/EtOAc (30 to 100% gradient) to give 85-2 (0.16 g, 75%).
[0542] Préparation of (85a): A solution of 85-2 (0.16 g; 0.16 mmol) in 80% aq. HCOOH (5 mL) was stirred at R.T. for 3 h. The solvent was evaporated and then co-evaporated with toluene. Purification on a silica column (10 g column) with CH2C12 /MeOH (4-10% gradient) gave 85a (43 mg, 74%). ’H-NMR (DMSO-d^): δ 7.75 (d, 1 H), 7.33 (d, 2 H), 6.07 (dd, 1 H), 5.75 (d, 1 H), 5.55 (dd, 1 H), 5.43 (dt, 1 H), 5.43 (t, 1 H), 3.79 (dd, 2 H), 3.63 (ddd, 2 H), 2.64 (sept, 1 H), 1.12 (d, 6 H). MS: m/z = 362.1 [M+1]
228
EXAMPLE 83
Préparation of Compound (86a)
NHDMT
MMTd' V
NHDMT
MMTtJ 'F
86-2
88-1 [0543] Préparation of (86-2): 86-2 was prepared using a similar procedure for preparing 85-2 with the following: 86-1 (220 mg; 0.22 mmol), (2.5 mL), isobutyric anhydride (0.13 mL; 3.6 equiv), EtOAc (30 mL), and hexanes/EtOAc (30 to 100% gradient) to give 86-2 (175 mg, 85%).
[0544] Préparation of (86a): 86a was prepared using a similar procedure for preparing 85a with the following: 86-2 (117 mg; 0.13 mmol), 80% aq. HCOOH (4 mL) and CH2C12 /MeOH (4-10% gradient) to give 86a (36 mg, 77%). ’H-NMR (DMSO-d«): 8 7.58 (d, 1 H), 7.29 (d, 2 H), 6.00 (s, 1 H), 5.73 (d, 1 H), 5.24 (ddd, 1 H), 4.55 (dd, 1 H), 4.22 (dd, 2 H), 3.80 (dd, 2 H), 2.58 (sept, 1 H), 1.08,1.07 (2d, 6 H). MS: m/z = 364 [M+l].
[0545] Préparation of (87-2): 87-2 was prepared using a similar procedure for preparing 46-2 with the following: 87-1 (178 mg, 0.3 mmol), hexanoic anhydride (0.14 mL, 2 equiv.), pyridine (3 mL) to give 87-2. (120 mg, 50%).
[0546] Préparation of (87a): 87a was prepared using a similar procedure for preparing 85a with the following: 87-2 (120 mg, 0.15 mmoi), 80% aq. HCOOH and CH2CI2 /MeOH (4-10% gradient) to give 87a (62mg, 85%). ’H-NMR (CDCh): 8 8.2 (br, 1 H), 7.42 (d,
H), 6.8 (br, 1 H), 6.03 (d, 1 H), 5.77 (dd, I H), 5.64 (dd, 1 H), 5.51 (ddd, t H), 4,43 (dd, 2 H),
3,82 (dd, 2 H), 2.4! (m, 2 H), 2.33 (m, 2 H), 1.64 (m, 4 H), 1.31 (m, 8 H), 0.82 (m, 6 H). MS:
m/z = 488 [M-1].
229
EXAMPLE 85
Préparation of Compound (88a)
DMTO
NHDMT
[0547] Préparation of (88-2): 88-2 was prepared using a similar procedure for preparing 85-2 with the following: 85-1 (220 mg; 0.24 mmol), pyridine (3 mL), dodecanoyc anhydride (0.12 g; 1.3 equiv), EtOAc (50 mL) and hexanes/EtOAc (25 to 80% gradient) to give 88-2 (022 g, 85%).
[0548] Préparation of (88a): 88a was prepared using a similar procedure for preparing 85a with the following: 88-2 (0.19 g; 0.17 mmol), 80% aq. HCOOH (5 mL) and CH2CI2 /MeOH (4-10% gradient) to give 88a (66 mg, 82%). ’H-NMR (DMSO-de): δ 7.77 (d, 1 H), 7.35 (d, 2 H), 6.07 (dd, 1 H), 5.77 (d, 1 H), 5.60 (dd, 1 H), 5.55 (ddd, 1 H), 5.43 (t, 1 H), 3.78 (dd, 2 H), 3.65 (ddd, 2 H), 2.41 (m, 2 H), 1.56 (m, 2 H), 1.24 (m, 16 H), 0.85 (m, 3 H). MS: m/z = 474 [M-l].
EXAMPLE 86
Préparation of Compounds (89a) and (90a)
[0549] Préparation of (89-2): To a solution of 89-1 (175 mg; 0.18 mmol) in MeCN (2.5 mL) at 0°C was added TMSBr (0.28 mL; 10 equiv.). The mixture was stirred at R.T. for 1 h, evaporated and treated with water. The obtained white solid was fiitered, dried and washed
230 with CH2CI2. The white solid was then dissoived in NMP (2 mL) and treated with DIPEA (94 gL; 3 equiv.) and pivaloyloxymethyliodide (84 gL; 3 equiv.). The mixture was stirred at R.T. for I day, and then partitioned between water (20 mL) and /ert-butyl methyl ether (TBME; 60 mL). The organic layer was washed with saturated aqueous NaHCOj, water and brine. The combined aqueous washings were back extracted with TBME (2 x 20 mL). The combined organic extract was dried and purified on a silica column (10 g column) with CH2CI2 /i-PrOH (2-10% gradient) to give 89-2 (42 mg, 26%).
[0550] Préparation of (89a): A solution of 89-2 in 80% aq. HCOOH was stirred at R.T. for 3 h. The solvent was evaporated and then co-evaporated with toluene. Purification on a silica column (10 g column) with CH2CI2 /MeOH (4-15% gradient) gave 89a (17 mg, 74%). ‘H-NMR (CDjOD): δ 7.47 (d, I H), 6. 28 (dd, 1 H), 6.04 (dd, 1 H), 5.77-5.71 (m, 2 H), 5.53 (m, 4 H), 5.18 (ddd, 1 H), 5.60 (dd, 1 H), 3. 77 (dd, 2 H), 1.08 (m, 18 H). 3,P-NMR (CDjOD): δ 17.64. MS: m/z = 598 [M+1], [0551] Préparation of (90a): A mixture of 89a (12 mg; 0.02 mmol) in EtOH (1 mL) and Pd/C (10%; 2.5 mg) was stirred ovemight under an atmospheric pressure of hydrogen. The mixture was filtered through a Celite pad. The solvent was evaporated and the product was purified on a silica column (10 g column) with CH2CI2 /MeOH (4-17% gradient) to give 90a (6 mg, 50%). *H-NMR (CDjOD): δ 7.51 (d, 1 H), 5.79 (d, 1 H), 5.65-5.54 (m, 5 H), 5.20 (ddd, 1 H), 5.60 (dd, 1 H), 3. 70 (dd, 2 H), 2.17-2.06 (m, 1 H), 2.02-1.87 (m, 3 H), 1.13 (m, 18 H). 3,PNMR (CDjOD): δ 33.16. MS: m/z = 600 [M+1].
231
EXAMPLE 87
Préparation of Compound (91a)
-OH r
Oy° r
86-1
91-2 vo·
Htf >
91a ^ο^ο^οΛο or τ
[0552] Préparation of (91-2): bis(isopropyloxycarbonyloxymethyl)phosphate bis(POC)phosphate and 0.1 mL of EtjN) in THF (2 mL) was added 86-1 (100 mg; 0.11 mmol), followed by diisopropylethyl amine (0.19 mL; 10 equiv), BOP-C1 (140 mg; 5 equiv) and 3-nitro1,2,4-triazole (63 mg; 5 equiv). The mixture was stirred at R.T. for 90 mins., and then diluted with CH2C12 (30 mL). The mixture was washed with saturated aqueous NaHCOj and brine. The mixture was dried with Na2SO«. The solvent was evaporated, and the residue was purified on a silica column (10 g column) with hexanes/EtOAc (40-100% gradient) to give 91-2 (117 mg, 90%).
[0553] Préparation of (91a): 91a was prepared using a similar procedure for preparing 85a with the following: 91-2 (87 mg; 0.07 mmol), 80% aq. HCOOH (5 mL) and CH2CI2 /MeOH (4-15% gradient) to give 91a (36 mg, 85%). ’H-NMR (CDjCN): δ 7.67 (dd, 1 H), 6.35 (dd, 1 H), 6.1 (br, 2 H), 5.82 (d, 1 H), 5.62 (m, 4 H), 5.22 (dm, 1 H), 4.98 (br, IH), 4.89 (m, 2 H), 4.49 (d, 1 H), 4.34 (m, 2 H), 3.88 (dd, 2H), 129 (d, 6 H), 1,28 (d, 6 H); 3IP-NMR (CDjCN): δ -4.49. MS: m/z = 606 [M+1].
To a solution of triethylammonium (0.33mmol, prepared from 110 mg of
232
EXAMPLE 88
Préparation of Compound (92a) o
,0.
92·
0^0 Hdf 'F [0554] Préparation of (92-2) and (92-3): To a solution of triethylammonium bis(POM)phosphate (0.48 mmol, prepared from 176 mg of bis(POM)phosphate and 0.15 mL of Et3N) in THF (2 mL) was added 92-1 (150 mg; 0.18 mmol) followed by diisopropylethyl amine (0.31 mL; 10 equiv), BOP-Ct (229 mg; 5 equiv), and 3-nitro-l,2,4-triazole (103 mg; 5 equiv). The mixture was stirred at R.T. for 90 mins., and then diluted with CH2C12 (30 mL). The mixture was washed with saturated aqueous NaHCOj and brine. The mixture was dried with Na2SO4. The solvent was evaporated, and the residue was purified on a silica column (10 g column) with CH2CI2 /i-PrOH (2-10% gradient) to obtain 92-2 (44 mg, 21%) and 92-3 (73 mg, 28%).
[0555] Préparation of (92a): A mixture of 92-2 and 92-3 (73 mg and 44 mg) and
80% aq. HCOOH (3 mL) was heated for 30 mins., at 35°C. The solvent was evaporated and then coevaporated with toluene. The solvent was evaporated, and the residue was purified on a silica column (10 g column) with CH2C12 /MeOH (4-10% gradient) to obtain 92a (40 mg, 75%).
’H-NMR (DMSO-D6); δ 10.6 (br, 1 H), 7.76 (s, 1 H), 6.44 (br, 2 H), 5.99 (dd, 1 H), 5.83 (d, 1
233 • H), 5.53-5.27 (2 m, 6 H), 4.39 (dt, 1 H), 4.04 (m, 2 H), 1.17 (s, 3 H), 1.06,1.08 (2 s, 18 H). 31PNMR (DMSO-de): δ-4.09. MS: m/z = 608 [M+1].
EXAMPLE 89
Préparation of Compound (93a)
o
[0556] Préparation of (93-2) and (93-3): 93-2 and 93-3 (68 mg and 80 mg, respectively) were prepared in the same manner from 93-1 (200 mg; 0.23 mmol) and bis(POM) 5 phosphate (230 mg) with DIPEA (0.4 mL), BopCl (290 mg), and 3-nitro-l,2,4-triazole (130 mg) in THF (3 mL) as 92-2 and 92-3 from 92-1.
[0557] Préparation of (93a): 93-2 and 93-3 (68 mg and 80 mg, respectively) were converted into 93 (42 mg) with formic acid in the same manner as 92 from 92-2 and 92-3. *HNMR (DMSO-De): 5 7.73 (s, 1 H), 6.46 (br, 2 H), 6.04 (dd, 1 H), 5.91 (dd, 1 H), 5.87 (d, 1 H), 5.48 (d, 4 H), 5.33 (m, 1 H), 5.24 (ddd, 1 H), 4.60 (dt, 1 H), 4.07 (m, 2 H), 1.07, 1.06, 1.05 (4 s, 18 H). 3,P-NMR (DMSO-de): δ -4.37. MS: m/z = 620 [M+1].
234
EXAMPLE 90
Préparation of Compound (94a)
O
NH ^NH2 o
[0558]
To a solution of 93a (53 mg; 0.09 mmol) in EtOH (2 mL) was added 10%
Pd/C (10 mg). The mixture stirred under hydrogen at atmospheric pressure for 1 h. The mixture was filtered through a Celite pad, and the filtrate evaporated. Purification on a silica column (10 g column) with CHiClj /MeOH (4-11% gradient) yielded 94a (45 mg, 81%). ’H-NMR (DMSOD6): δ 10.6 (br, 1 H), 7.81 (s, 1 H), 6.4 (br, 2 H), 5.97 (dd, 1 H), 5.85 (d, 1 H), 5.60-5.44 (m, 5 H), 4.37 (m, 1 H), 4.11 (ddd, 2 H), 1.66 (m, 2 H), 1.09,1.06 (2 s, 18 H), 0.81 (7,3 H); 3,P-NMR (DMSO-de): δ -4.10. MS: m/z = 622 [M+1].
EXAMPLE 91
Préparation of Compounds (95a) and (96a)
[0559] Préparation of (95-1): To a solution of 5-Amino-2H-[l,2,4]triazin-3-one (180 mg, 1.5 mmol) in HMDS was added a catalytic amount of (NHO4SO4. The mixture was heated to reflux for 5 h. HMDS was evaporated to give a crude product. To a solution of the crude product in anhydrous CH3CN was added 70a (220 mg, 0.5 mmol) and TMSOTf (0.45 mL,
2.5 mmol). The mixture was heated to reflux for 24 h in a sealed tube. The reaction was
235 quenched with NaHCÛ3 and diluted with EA. The organic solvent was removed, and the residue was purified by prep-TLC first, and the by RP-HPLC (0.5% HCOOH in water and
MeCN) to give the pure 95-1 (100 mg, 46%).
[0560] Préparation of (95-2): To a solution of 95-1 (80 mg, 0.18 mmol) in anhydrous CH3CN was added 1,2,4-triazole (911 mg, 11.7 mmol) and TEA (1.45 g, 14.4 mmol). The mixture was cooled to 0°C and POCI3 was added. The reaction mixture was stirred at 25°C for 24 h. The solvent was evaporated and partitioned with EA and water. The organic layer was concentrated to give the crude 95-2 (80 mg, 90%).
[0561] Préparation of (95a): 95-2 (90 mg, 0.18 mmol) was dissolved in 20 mL of saturated THF ammonia. The resulting solution was stirred at 25°C for 2 h. The solvent was removed, and the residue was purified on a silica gel column (EA: PE = 6:1) to give 95a as a white solid (70 mg, 70%).
[0562] Préparation of (96a): 95a (70 mg, 0.16 mmol) was dissolved in 20 mL of saturated MeOH ammonia. The resulting solution was stirred at 25°C for 2 h. The solvent was removed, and the residue was purified by RP-HPLC (0.5% HCOOH in water and MeCN) to give 96a (5 mg, 11%) as a white solid. 'H NMR (CD3OD, 400 MHz) £7.57 (s, IH), 6.35 (dd, J = 3.6 Hz, .7 = 15.6 Hz, IH), 5.45-5.47 (m, IH), 4.70 (dd, 4.8 Hz, 16.2 Hz, IH), 3.83 (s, 2H), 3.71 (d, J= 1.6 Hz, 2H). ESI-TOF-MS: m/z 295.1 [M + H]+.
EXAMPLE 92
Préparation of Compounds (97a-g)
HOMO R2 [0563] Dry nucleoside (0.05 mmol) was dissolved in a mixture of DMF (3 mL) and DMA-DMF (0.04 mL, 0.1 mmol). The reaction was kept at ambient température for 4 h and then evaporated to dryness. The residue was dissolved in a mixture of PO(OMe)3 (0.7 mL) and pyridine (0.3 mL). The mixture was evaporated in vacuum for 15 min. at 42°C, than cooled down to R.T. N-Methylimidazole (0.009 mL, 0.11 mmol) was added followed by POCI3 (9μΙ, 0.11 mmol). The mixture was kept at R.T. for 20-40 mins. The reaction was controlled by
236
LCMS and monitored by the appearance of the corresponding nucleoside 5'-monophosphate.
After completion of the reaction, tetrabutylammonium sait of pyrophosphate (150 mg) was added, followed by DMF (0.5 mL) to get a homogeneous solution. After 1.5 h at ambient température, the reaction was diluted with water (10 mL). The mixture was loaded on the column HiLoad 16/10 with Q Sepharose High Performance, and séparation was done in a linear gradient of NaCl from 0 to IN în 50mM TRIS-buffer (pH7.5). The triphosphate (97a-f) was eluted at 75-80%B. The corresponding fractions were concentrated. The residue was dissolved in 5% ammonium hydroxide, kept for 15 min. at R.T. and concentrated. Desalting was achieved by RP HPLC on Synergy 4 micron Hydro-RP column (Phenominex). A linear gradient of fq methanol from 0 to 30% in 50mM triethylammonium acetate buffer (pH 7.5) was used for elution. The corresponding fractions were combined, concentrated and lyophilized 3 times to remove excess of buffer.
Table 4 - Triphosphates obtained from Examnle 92
Compound MS (M-l) JlPNMR Pa PNMR Ρβ J1PNMR Ργ
NH, î î s rt HO- -O-^O-P-OV OH OH ÔHO-<C/ ° / 4 >► HO F 97a 528.0 -6.71 -6.82(d) -21.43(t) -11.35 -11.47(d)
NHj M θ î ΗΟ-Ρ-Ο-Ρ-Ο-β-Ο^ο OH OH ÔHp—Ά_/ ° / 4 », HO F 97b 544.0 -6.25(bs) -21.45(bs) -11.44 -11.56(d)
nh2 il « » ΗΟ-Ρ-ο-β-Ο-Ρ-Ο-. 0 M OH OH OH /r\_/ b/ho* ►f 97c 575.7 -8.86 -9.00(d) -22.95(t) -11.81 -11.94(d)
\-zNHî OH oh oh ho-A-o-A-o-Ç-o-< on-4 8 o 0 HO4 *F 97d 545.9 -9.41 -9.44(d) -23.04(1) -12.00 -12.13(d)
RFÇ «oXJUL 0 i S Oô- Λ ~ClHO* Έ 97e 552.1 -10.32 -10.44(d) -23.26(t) -11.84 -11.96(d)
237
Compound MS (M-l) JiPNMR Pa J‘PNMR P0 J‘P NMR PY
ΝΗχ ΟΟΟ HO-P-O-P-O-3-O-V-<\>N4, ÔH éH ° Hu F 97f 508.4 -8.30 (bs) -22.72(bs) -11.51 -11.63(d)
O O O Ηθ-β-0-β-Ο-β-Ο r¥ ÔH ÔH ÔhV^°S>N’V'F| HO T 22s 550.1 -9.17 -9.29 (d) -23.04 (t) -11.97 -12.09(d)
EXAMPLE 93 Préparation of Compounds (98a-e) and (99a) ]0564] Dry nucleoside (0.05 mmol) was dissolved in a mixture of PO(OMe)3 (0.7 mL) and pyridine (0.3 mL). The mixture was evaporated in vacuum for 15 mins. at 42°C, than cooled down to R.T. N-Methylimidazole (0.009 mL, 0.11 mmol) was added followed by POClj (9μ1, 0.11 mmol). The mixture was kept at R.T. for 20-40 mins. The reaction was controlled by 5 LCMS and monitored by the appearance of the corresponding nucleoside 5’-monophosphate.
After completion of the reaction, tetrabutylammonium sait of pyrophosphate (150 mg) was added, followed by DMF (0.5 mL) to get a homogeneous solution. After 1.5 h at ambient température, the reaction was diluted with water (10 mL) and loaded on the column HiLoad 16/10 with Q Sepharose High Performance. Séparation was done in a linear gradient of NaCl 10 from 0 to IN in 50mM TRIS-buffer (pH7.5). The triphosphate (98a-e) was eluted at 75-80%B.
The corresponding fractions were concentrated. Desalting was achieved by RP HPLC on Synergy 4 micron Hydro-RP column (Phenominex). A linear gradient of methanol from 0 to 30% in 50mM triethylammonium acetate buffer (pH 7.5) was used for elution. The corresponding fractions were combined, concentrated and lyophilized 3 times to remove excess 15 of buffer.
Table 5 - Compounds obtained from Example 93
Compound MS (M-l) JlPNMR Pa J1PNMR Ρβ J1P NMR Py
0 ° Ο ,βΝ Q HO-P-O-P -O -P-OA/Û^N éH éH hK f H0 F NH2 98a 538.0 -5.21 -5.33(d) -20.56(0 -11.09 -11.20(0
238
Compound MS (M-l) 3lPNMR Pa 31PNMR P0 JIPNMR Ργ
JXL· 0 ί 4 ci—J NV κσ p 98b 556.2 -10.85(bs) -23.1l(bs) -11.76 -11.88(d)
N 0 O o ho-p-o-p-o-p-o-m0W/N ÔH ÔH *f 98c 540.4 -8.86(bs) -23.84(t) -11.68 -11.80(d)
N jNHî o o o V** HO-P-O-P-O-P-O-* o N-< / ÔH ÔH ÔH_N**' HO4 *F 98d 536.0 -9.35 -9.47(d) -23.05(t) -11.60 -11.72(d)
A y y y r\ M TT’-fews= xtf > 98e 545.9 -10.54 -10.66 -2326 -11.80 -11.93(d)
o -lar1 HO* *F 99a 357.2 1.42(s) NA NA
EXAMPLE 94 Préparation of Compound (100a)
100-1 100-2
100a [0565] Préparation of (100-2): To an ice-cold solution of 100-1 (22 mg; 0.055 mmol) in acetonitrile (0.5 mL) was added TMSBr (80 gL; 10 equiv.). The resulting mixture was stirred at R.T. for 1 h. The mixture was concentrated, and the residue was partitioned between water and diethyl ether. The aqueous layer was washed with Et2O, neutralized with triethylammonium bicarbonate buffer and lyophilized to yield the triethylammonium sait of 100-
2.
239
[0566] Préparation of (100a): 100-2 was rendered anhydrous by cocvaporating with pyridine and toluene. Anhydrous 100-2 was dissolved in HMPA (t mL) and 1,1carbonytdiimidazole (32 mg; 0.2 mmol) was added. The mixture was stirred at R.T. for 6 h. A solution of tetrabutylammonium pyrophosphate (0.22 g; -0.2 mmot) in DMF (2 mL) was added.
The mixture was stirred ovemight at R.T. The mixture was diluted with triethylammonium acetate buffer and purified by RP-HPLC with a gradient 0-60% B (A: 50 mM aqueous TEAA, B: 50mM TEAA in MeOH) and repurified by RP-HPLC with a gradient 0-30% B to give 100a. 3,P-NMR (D2O): δ 3.22 (d, IP), -8.21 (br, 1 P), -22.91 (br, 1 P). MS: m/z = 528 (M-l).
EXAMPLE 95 Préparation of Compound (100b)
100-3 100-4 [0567] Préparation of (100-4): 100-4 was prepared from 100-3 (54 mg; 0.13 mmol) in acetonitrile (1.3 mL) with TMSBr (0.18 mL) using a similar procedure as described for the préparation of 100-2.
[0568] Préparation of (100b): 100b was prepared from 100-4 in HMPA (2 mL) with CDI (84 mg) and tetrabutylammonium pyrophosphate (0.5 g) in DMF (2 mL) using a U similar procedure as described for the préparation of 100a. 31P-NMR (D2O): δ 17.90 (d, IP),9.00 (d, 1 P), -22.91 (t, 1 P). MS: m/z = 530 (M-l).
EXAMPLE 96 Préparation of Compound (100c)
HO' F
100-8
Hd >
100-6
100c [0569] Préparation of (100-6): 100-6 was prepared from 100-5 (40 mg; 0.09 mmol) in acetonitrile (1 mL) with TMSBr (0.1 mL) using a similar procedure as described for the préparation of 100-2.
240 [0570] Préparation of (100c): 100c was prepared from 100-6 in HMPA (1.5 mL) with CDI (50 mg) and tetrabutylammonium pyrophosphate (0.3 g) using a similar procedure as described for the préparation of 100a. 3lP-NMR (D2O): 5 -7.13 (br, !P), -10.14 (d, 1 P), -22.84 (br, 1 P). l’F-NMR (D2O): δ -117.53 (dd, 1 F), -197.8 (m, 1 F). MS: m/z = 545.5 (M-l).
EXAMPLE 97
Préparation of Compounds (lOOd) and (100e)
Hd 'F H(J 'F
100-7 100-8
[0571] Préparation of (100-8): To an ice-co!d solution of diastereomers 100-7 (35 mg; 0.08 mmol) in acetonitrile (1 mL) was added TMSBr (0.1 mL; 10 equiv.). The resulting mixture was stirred overnight at R.T. and then concentrated. The residue was partitioncd between water and CH2C!2. The aqueous layer was washed with CH2C12, neutralized with triethylammonium bicarbonate buffer and lyophilizcd to yield the triethylammonium sait of 1008.
[0572] Préparation of (IQOd) and (100e): 100-8 was rendered anhydrous by cocvaporating with pyridine and toluene. Anhydrous 100-8 was dissoived in DMF (1.5 mL) and CDI (54 mg; 0.3 mmol) was added. The mixture was stirred at R.T, for 7 h. A solution of tetrabutylammonium pyrophosphate (0.3 g; -0.3 mmol) in DMF (4 mL) was added. The mixture was stirred at R.T for 3 days. The mixture was diluted with triethylammonium acetate buffer. Two consecutive RP-HPLC purifications with a gradient 0-60% B (A: 50 mM aqueous TEAA, B: 50mM TEAA in MeOH) and 0-40% B gave lOOd and 100e as single diastereomers. lOOd: 3lP-NMR (D2O): 6 4.28 (dd, IP), -6.37 (d, 1 P), -22.36 (t, 1 P). MS: m/z = 548.1 (M-l). 100e: 3lP-NMR (D2O): δ 4.13 (dd, IP), -6.38 (d, 1 P), -22.46 (t, 1 P). MS: m/z = 548.1 (M-l).
241
EXAMPLE 98
Préparation of Compound (101a)
TBDPSO
59-4
TBDPS
[0573] Préparation of (101-1): To a solution of 59-4 (IJ g, 2.39 mmol) in anhydrous DCM (100 mL) was added Dcss-Martin periodinane (5.2 g, 11.95 mmol) at 0°C under nitrogen. The mixture was stirred at R.T. for 5 h. The mixture was poured into NaHCOj and NaîSîOjaq. Solution. The organic layer was washed with brine, dried over with anhydrous NaîSO4, and concentrated to dryness to give the crude 101-1 (1.5 g) as a white solid, which was used for the next step without further purification.
[0574] Préparation of (101-2): To a mixture of bromo(isobutyl)triphenylphosphorane (4.8 g, 12.03 mmol) in anhydrous THF (8 mL) was added t-BuOK(l 12 mL, 112 mmol) at 0°C under nitrogen. The mixture was stirred at R.T. for 1 h. A solution of 101-1 (1.0 g, 1.6 mmol) in anhydrous THF (4 mL) was added dropwise at 0°C. The mixture was stirred at R.T. for 3 h. The reaction was quenched with a NHiCl aq. solution and extracted with DCM. The organic layer was dried and concentrated to give a residue, which was purified by silica gel column chromatography (5% EtOAc in PE) to give 101-2 (793 mg, 74.4%) as a white solid.
[0575] Préparation of (101-3): To a solution of 101-2 (364 mg, 0.547 mmol) in anhydrous CHjCN (6 mL) were added TPSC1 (414 mg, 1.37 mmol), DMAP (167 mg, 1.37 mmol) and NEtj (138 mg, 1.37 mmol) at R.T. The mixture was stirred at R.T. for 2 h. NH4OH (6 mL) was added, and the mixture was stirred for another 1 h. The mixture was diluted with DCM and washed with a NaHCOj aq. solution. The organic layer was separated and concentrated to give a residue, which was purified by silica gel column chromatography (2% MeOH in DCM) to give 101-3 (347 mg, 95.0%) as white solid.
242
[0576] Préparation of(101a): To a solution of27-3 (347 mg, 0.52 mmol) in MeOH (10 mL) was added NH4F (1.5 g) at R.T. The reaction mixture was refluxed for 12 h, and then filtered. The fiitrate was concentrated in vacuo, and the residue was purified by silica gel column chromatography (10% MeOH in DCM) to give 101a (87 mg, 53%) as a white solid. *H NMR (CD3OD, 400MHz) J8.ll (d, J= 7.6 Hz, IH), 6.03 (dd, J= 1.2, 17.6 Hz, IH), 5.88 (d, J = 7.2 Hz, IH), 6.03 (dd, J= 1.6, 11.6 Hz, IH), 5.39 (d, J= 10.8 Hz, 1H),4.88 (dd, J= 3.2,60.0 Hz, IH), 4.41 (dd, J = 4.8, 24.4 Hz, IH), 3.70 (d, J= 12.4 Hz, IH), 3.57 (d, J= 12.0 Hz, IH), 3.08-3.14 (m, IH), 0.94-0.98 (m, 6H). ESI-MS: m/z626.9 [2M + H]+.
101-2
EXAMPLE 99 Préparation of Compound (102a)
102-3 102-4 102a 10 [0577] Préparation of (1Û2-1): To a solution of 101-2 (1.0 g, 1.5 mmol) in MeOH (20 mL) was added NH4F (6 g) at R.T., and the mixture was refluxed ovemight. After cooting to R.T., the mixture was filtered, and the fiitrate was concentrated. The residue was purified by silica gel column chromatography (8 % MeOH in DCM) to give 102-1 (400 mg, 85%) as a white solid.
[0578] Prcparation_of (102-2): To a solution of 102-1 (400 mg, 127 mmol) in
MeOH (10 mL) was added Pd/C (400 mg) at R.T. The mixture was stirred at R.T. under a baltoon of H2 for 1.5 h. The mixture was filtered, and the fiitrate was concentrated in vacuo to give 102-2 (400 mg, 99 %) as a white solid.
[0579] Préparation of (102-3): To a solution of 102-2 (400 mg, 126 mmol) in anhydrous DMF (5 mL) were added imidazole (968 mg, 142 mmol), and TBSC1 (1.5 g, 10.0 mmol) at R.T. The mixture was stirred at 50°C ovemight. The mixture was diluted with DCM and washed with a NaHCO3 aq. solution. The organic layer was dried and concentrated. The
243 residue was purifîed by silica gel column chromatography (10% EA in PE) to give 102-3 (676 mg, 98 %) as a white solid.
[0580] Préparation of (102-4): To a solution of 102-3 (676 mg, 1.24 mmol) in anhydrous CH3CN (6 mL) were added TPSC! (941 mg, 13.11 mmol), DMAP (379 mg, 3.11 mmol) and NEt3 (314 mg, 3.11 mmol) at R.T. The reaction was stirred at R.T. for 3 h. ΝΗ,ΟΗ (1 mL) was added, and the reaction was stirred for 4 h. The mixture was diluted with DCM and washed with a NaHCO3 solution. The organic layer was dried and concentrated. The residue was purifîed by silica gel column chromatography (2% MeOH in DCM) to give 102-4 (450 mg, 67%) as a white solid.
[0581] Préparation of (102a): To a solution of 102-4 (450 mg, 0.83 mmol) in MeOH (10 mL) was added NH«F (2 g) at R.T. The reaction mixture was refluxed ovemlght, After cooling to R.T., the mixture was filtered, and the filtrate was concentrated. The residue was purifîed by silica gel column chromatography (8 % MeOH in DCM) to give 102a (166.6 mg, 64%) as a white solid. *H NMR (CD3OD, 400MHz) <58.09 (d, J= 7.6 Hz, IH), 6.07 (d, J=
3.6 Hz, IH), 6.05 (d, 2.8 Hz, IH), 5.89 (d, J= 7.6 Hz, IH), 5.03 (dd, J= 5.2, 57.2 Hz, IH),
4.41 (dd, J= 4.2, 17.2 Hz, IH), 3.74 (d, J= 12.0 Hz, IH), 3.54 (d, 12.0 Hz, IH), 1.23-1.78 (m, 5H), 0.90 (d, J= 6.4 Hz, 6H). ESI-MS: m/z 631.1 [2M + H]+.
EXAMPLE 100 Préparation of Compound (103a)
MMTrÔ F
103-2
103-1
Acd 'F H(5 F
103-3 103a [0582] Préparation of (103-2): 103-1 (3.8 g, 6.9 mmol) in 80% AcOH aq. was stirred at 50°C for 4 h. The mixture was concentrated to give a residue, which was purified by silica gel column chromatography (5% MeOH in DCM) to give the uridine dérivative (1.5 g,
244
78.2%) as a white solid. To a solution of the uridine dérivative (1.5 g, 5.4 mmol) in Py (10 mL) was added Ac2O (1.38 g, 13.5 mmol) at R.T. The mixture was stirred at R.T. for 12 h. The mixture was concentrated to give a residue, which was purified by silica gel column chromatography (20% EA in PE) to give 103-2 (1.3 g, 68%) as a white solid.
[0583] Préparation of (103-3): To a solution of N-(5-fluoro-2-hydroxy-l,2dihydropyrimidin-4-yl)benzamide (0.5 g, 2.1 mmol) in anhydrous PhCl (5 mL) was added ammonium sulfate (6 mg, 0.043 mmol), followed by HMDS (0.7 g, 4.3 mmol). The mixture was heated to 130°C for 8 h. The mixture was concentrated under vacuum to 2 mL, and then cooled to 0°C. TMSOTf (310 mg, 1.4 mmol) was then added. After stirring for 10 min at 0°C, 103-2 (150 mg, 0.4 mmol) in PhCl (5 mL) was added. The mixture was stirred at 130°C for 10 h. The mixture was concentrated, and the residue was re-dissolved in DCM (10 mL), washed with water (5 mL) and saturated NaHCOj. The organic layer was dried over Na2SO4, evaporated to dryness and the crude product was purified by silica gel column chromatography (60% PE in EA) to give 103-3 (30 mg, 16%) as a white solid.
[0584] Préparation of (103a): A solution of 103-3 (150 mg, 0.34 mmol) in NHj/McOH (10 mL) was stirred at R.T. for 3 h. The mixture was concentrated, and the residue was purified by HPLC séparation (0.1% HCOOH in water and MeCN) to give 103a (60 mg, 60%) as a white solid. lH NMR (CDjOD, 400MHz) <78.28 (d, J= 6.8 Hz, IH), 6.10 (dd, J = 2.0, 15.2 Hz, IH), 4.99-5.15 (m, IH), 4.62-4.65 (m, IH), 4.49-4.55 (m, 2H), 3.89 (dd, J= 1.6, 12.0 Hz, IH), 3.75 (dd, J= 1.2,12.0 Hz, IH). ESI-MS: m/z 613,1 [2M + Naf.
EXAMPLE 101 Préparation of Compound (104a)
103-3 104-1 104a [0585] Préparation of (104-1): 103-3 (150 mg, 0.31 mmol) was dissolved in 80% aqueous acetic acid (3 mL). The solution was heated to reflux for 2 h. The mixture was cooled to ambient température and diluted with water (5 mL), neutralized to pH>7 with saturated
NaHCOj and extracted with EA. The organic layer was dried and evaporated to dryness. The residue was purified by silica gel column chromatography (50% EA in PE) to give 104-1 (80 mg, 70%) as a white solid.
245 [0586] Préparation of f 104a): 104-1 (80 mg, 0.22 mmol) in saturated NHj/MeOH (10 mL) was stirred at R.T. for 3 h. The mixture was concentrated, and the residue was purified by silica gel column chromatography (5% MeOH in DCM) to give 104a (40 mg, 60%) as a white solid. *H NMR (CDjOD, 400MHz) £8.30 (d, 6.8 Hz, IH), 6.18 (dd, 4.0,14,0 Hz,
IH), 5.13-5.65 (m, IH), 4.52-4.56 (m, IH), 3.980-3.95 (m, 2H), 3.76 (s, 3H). ESI-MS: m/z
319.1 [M + Na]+.
EXAMPLE 102 Préparation of Compound (105a)
105a
[0587] Préparation of (105-2): To a solution of triethylammonium bis(isopropyloxycarbonyloxymethyl)phosphate (0. 065 mmol, prepared from 22 mg of bis(POC)phosphate and EtjN) in THF was added 105-1 (31 mg; 0.05 mmol). The resulting 10 mixture evaporated, and the residue was rendered anhydrous by coevaporation with pyridine, followed by toluene. The anhydrous evaporated residue was dissoived THF (1 mL) and cooled in an ice-bath. To the solution was added diisopropylethyl amine (35 pL; 4 equiv), followed by
BOP-Cl (25 mg; 2 equiv) and 3-nitro-1,2,4-triazole (11 mg; 2 equiv). The mixture was stirred at
0°C for 90 min. The mixture was diluted with CHjCh, washed with saturated aq. NaHCOj and brine, and dried with Na 2 S O4. The evaporated residue was purified on silica (10 g column) with a CH2CI2 /i-PrOH solvent system (3-10% gradient) to give 105-2 (13 mg, 28%).
24β [0588J Préparation of (105a): A solution of 105-2 (13 mg; 0.014 mmol) in 80% aq. HCOOH (2 mL) was stirred at R. T. for 3 h. The mixture was evaporated and then coevaporated with toluene. The product was purified on silica (10 g column) with a CH2Cl2/MeOH solvent system (4-15% gradient) to give 105a (7 mg, 78%). ’H-NMR (DMSO-de): δ 7.52 (d, 1 H), 7.28, 7.24 (2 br s, 2 H) 5.92 (dd, 1 H), 5.74 (d, l H), 5.69 (d, 1 H), 5.62 (d, 4 H), 4.97 (ddd, 1 H), 4.82 (m, 2 H), 4.38 (dt, 1 H), 4.07 (m, 2 H), 1.23 (m, 12 H), 1.04 (m, IH), 0.37 (m, 4 H). 3IP-NMR (DMSO-de): δ -4.51.19F-NMR (DMSO-de): δ -199.23 (dt). MS: m/z = 598.4 (M+l).
EXAMPLE 103 Préparation of Compound (106a) lu 0 -^O^O^O-P-OH
NHDMT
“Ai/ ° f 5· HO F OyO y0 ei3n
ΒΟΡ-CI, DIPEA, NT; THF
106-1
NHDMT
10589] Préparation of (106-1): 106-1 (15 mg; 30% yield) was prepared in the same manner from 43-5 (32 mg; 0.057 mmol) and bis(POC)phosphate (24 mg) with DIPEA (40 pL), BopCl(29 mg) and3-nitro-l,2,4-triazole (13 mg) as 105-2 from 105-1.
[0590] Préparation of (106a): 106-1 (15 mg) was converted in formic acid to 106a (8 mg; 78% yield) in the same manner as 105-2 to 105a. ’H-NMR (DMSO-de): δ 7.55 (d, 1 H), 7.32,7.27 (2 br s, 2 H) 6.06 (dd, 1 H), 5.84 (d, 1 H), 5.73 (d, 1 H), 5.61 (d, 4 H), 5.08 (ddd, 1 H),
4.83 (m, 2 H), 4.36 (m, 1 H), 4.21 (dd, H), 4.16 (dd, 1 H), 3.56 (d, I H), 3.49 (d, 1 H), 3.28 (s, 3 H), 1,25,1.24 (2 d, 12 H). 3IP-NMR (DMSO-de): δ -4.45. MS: m/z = 602.4 (M+l).
247
EXAMPLE 104
Préparation of Compound (107a)
[0591] Préparation of (107-1): 107-1 (30 mg; 30% yield) was prepared in the same manner from 40-10 (65 mg; 0.115 mmol) and bis(POC)phosphate (49 mg) with DIPEA (80 pL), BopCl (58 mg) and 3-nitro-l,2,4-triazole (26 mg) as 105-2 from 105-1.
[0592] Préparation of (106a): 107-1 (30 mg) was converted in formic acid to 107a (15 mg; 73% yield) in the same manner as 105-2 to 105a. *H-NMR (DMSO-de): 8 7.60 (d, 1
H), 7.36,7.32 (2 br s, 2 H) 6.02 (m, 2 H), 5.74 (d, 1 H), 5.62 (m, 4 H), 5.17 (ddd, 1 H), 4.99 (dq, 1 H), 4.83 (m, 2 H), 4.61 (m, 1 H), 4,19 (m, 2 H), 1,40 (dd, 3 H), 1.24, 1.23 (2 d, 12 H). 3,PNMR (DMSO-de): 8 -4.52.19F-NMR (DMSO-de): 8 -185.92 (m, 1 F), -200.48 (d, I F). MS: m/z = 604.3 (M+1).
EXAMPLE 105
Préparation of Compound (108a)
248
[0593] To a solution of 4’-ethyl-2'-fluorocytidine (50 mg, 0.183 mmol) in DMF (l mL) were added DCC (l 13 mg, 0.55 mmol), isobutyric acid (48.5 μΐ, 0.55 mmol) and DMAP (22 mg, 0.183 mmol). The mixture was stirred at R.T. ovemight The mixture was filtered, and the filtrate was concentrated with a rotary evaporator until half of its original volume was achieved. EA was added to the mixture. The mixture was washed with water, followed by brine. The mixture was dried over anhydrous Na2SO4 and concentrated in vacuo to give a residue, which was purified by silica gel with DCM/ MeOH=95:5 to give 108a (40.8 mg, 54%) as a white solid. lH NMR (DMSO-J6,400 MHz) <5 7.67 (d, J= 7.2 Hz, 1H), 7.34 (br s, 2H),
5.85, 5.8 (2d, J = 21.2, 22 Hz, 1H), 5.72 (d, J= 7.6 Hz, 1H), 5.55-5.41 (m, 2H), 4.1 (q, 2H),
2.68-2.52 (m, 2H), 1.77-1.64 (m, 2H), 1.13, 1.14 (2s, 2 x3H), 1.09-1.07 (m, 6H), 0.96 (t, 7.6
Hz, 3H); MS m/z 414 (M-H4), 829 (2M+H4).
EXAMPLE 106
Préparation of Compound (109a)
[0594] 3’,5’-diacetylnucleoside (36 mg, 1 mmol) was dissolved in methanol saturated with NH4OH and kept ovemight at R.T. The solvent was evaporated, and the product isolated by column chromatography in gradient of methanol in DCM from 0 to 15% on a 10g 15 Biotage cartridge. The product was 109a obtained (20 mg, 73%). lH-NMR (DMSO-de): δ 11.4 (s, 1H), 11.84-11.82 (d, 1H); 6.10-6.05 (m, 1H), 5.95-5.83 (d, 1H), 5.71 (s, 1H), 5.65-5.63 (d, 1H), 5.37-3.36 (t, 1H), 5.26-5.20 (t, 1H), 5.11-5.07 (t, 1H), 4.56-4.55 (m, 1H), 4.46^.33 (m, 2H), 3.58-3.56 (m, 2H). MS 277.2 (M-H).
249
EXAMPLE 107
Préparation of Compound (110a)
[0595] Préparation of (110-1): To a solution of 70a (6.55 g, 2.1 mmol) and the benzoyt protected base moiety (2.3 g, 5.3 mmol) in PhCl (50 mL) was added TMSOTf (3.6 g,
16.1 mmol). After addition, the mixture was heated to 140°C for 8 h. The mixture was cooled to R.T., and evaporated to give a residue. The residue was rc-dissolved in DCM and washed 5 with saturated NaHCOj and brine. The organic layer was dried and concentrated to give a residue, which was purified by silica get column (40% EA in PE) to give 110-1 (300 mg, 10%) as a white solid.
[0596] Préparation of (110a): 110-1 (300 mg, 0.55 mmol) in 80% aqueous acetic acid (5 mL) was heated to reflux for 2 h. The mixture was cooled to ambient température and 10 diluted with water (5 mL), and then extracted with EA. The organic layer was washed with saturated NaHCOj and brine. The mixture was dried and concentrated to give a residue, which was purified by silica gel column (10% EA in PE) to give the protected uridine dérivative (180 mg, 70%) as a white solid. The protected uridine dérivative (180 mg, 0.4 mmol) in saturated NHj/MeOH (10 mL) was stirred at R.T. for 3 h. The mixture was concentrated to give a 15 residue, which was purified by préparative HPLC (0.1% HCOOH in water and MeCN) to give
110a (80 mg, 60%) as a white solid. ’H NMR (CDjOD, 400MHz) <58.31 (d, J = 6.8 Hz, IH), 6.17 (dd, 7= 4.0, 14.0 Hz, IH), 5.13-527 (m, IH), 4.52-4.56 (m, iH), 3.92 (dd,7= 12.0, 58.8 Hz, 2H). ESI-TOF-MS: m/z 334.7 [M + Naf.
250
EXAMPLE 108
Préparation of Compound (111a)
111-1 111-2 111-3 1114
[0597] Préparation of (111-2): Compound 111-1 (30.0 g, 0.1 mol) was suspended in anhydrous pyridine (300 mL) and stirred at room température (R.T.) for 1 hour. The suspension was cooled to 0°C and TMSCt (27.3 g, 0.25 mmol) was added dropwise. After addition was complété, the mixture was warmed to R.T. and stirred for 30 min. The mixture 5 was then re-cooled to 0°C and BzCl (15.5 g, 0.11 mot) was added dropwise. The mixture was warmed to R.T. and stirred ovemight. The reaction was cooled to 0°C and quenched with H2O. Aqueous ammonia was added, and the reaction was stirred at R.T, for 2 hours. The solution was concentrated and the residue was taken up into ethyl acetate (EA) and H2O. The aqueous phase was extracted with EA several times, and the combined organic layers were dried over Na2SO4 1 q and concentrated. The residue was purified on a silica get column to give compound 111-2 as a white solid (28.2 g, 76%). ESI-LCMS: m/z=368 [M+Na]+.
[0598] Préparation of (111-3): To a stirred suspension of compound 111-2 (18.4 g, 50 mmol), PPhj (22.3 g, 85 mmol) and pyridine (25 mL) in anhydrous THF (300 mL) was added a solution of I2 (19.05 g, 75 mmol) in THF (80 mL) dropwise at 0°C. After addition, the mixture 15 was warmed to R.T. and stirred for 60 hours. The precipitate was removed by filtration, and the filtrate was concentrated. The residue was dissolved in dichloromethane (DCM) and washed with saturated Na2S20j aqueous solution and then brine. The organic layer was dried over Na2SO4 and concentrated. The residue was purified on a silica get column to afford compound 111-3 (16.4 g, 69%). ESI-LCMS: m/z=478 [M+H]+.
2q [0599] Préparation of (111-4): To a stirred solution of compound 111-3 (17.0 g,
35.6 mmol) in anhydrous dimethylformamide (DMF) (300 mL) was added dropwise a solution of t-BuOK (10.0 g, 89.1 mmol) in DMF (120 mL) at 0°C over 20 min. Stirring was continued at
0°C for 45 min, and then concentrated hydrochloric acid (4.5 mL) was added. A pH value of 8-9 was achieved by adding a saturated NaHCOj solution. The precipitate was removed by
251 filtration, and the filtrate was diluted with ethyl acetate. The solution was washed with brine and dried over Na2SO4. The solvent was removed, and the residue was purified on a silica gel column to afford compound 111-4 as a white solid (8.6 g, 69%). ESI-LCMS: m/z=35O [M+H]+.
[0600] Préparation of (111-5): To a stirred solution of Bn EtjNCl (37.4 g, 0.16 mol) in MeCN (600 mL) was added NaNj (10.8 g, 0.16 mol). The mixture was sonîcated for 20 min, and then stirred at R.T. for 16 hours. The solution was filtrated into a solution of compound 111-4 (11.5 g, 32.9 mmol) and N-methylmorpholine (3.5 g) in anhydrous THF (200 mL). The mixture was cooled to 0°C and a solution of Ij (33.6 g, 0.14 mol) in THF (100 mL) was added dropwise. Stirring was continued at 0-10°C for 20 hours. N-Acetyl cystein was added until no gas evolved. Saturated Na2S20j aq. was added until a light yellow solution was achieved. The solution was concentrated and then diluted with EA. The organic phase was washed with brine and dried over Na2SO4. The solvent was removed, and the residue was purified on a silica gel column to give compound 111-5 (14.7 g, 84%). ESI-LCMS: m/z=519 [M+H]+.
[0601] Préparation of (111-6): To a stirred solution of compound 111-5 (12.5 g,
24.8 mmol) in anhydrous pyridine (200 mL) was added BzCl (4.3 g, 30 mmol) dropwise at 0°C. The mixture was then stirred at R.T. for 10 hours. The reaction was quenched with H2O, and the solution was concentrated. The residue was dissolved in EA and washed with saturated NaHCOj. The organic layer was dried over Na2SO4 and concentrated. The residue was purified on a silica gel column to give compound 111-6 as a white foam (11.2 g). ESI-LCMS: m/z=623 [M+H]+.
[0602] Préparation of (111-7): Compound 111-6 (9.43 g, 15.2 mmol), BzONa (21.9 g, 152 mmol) and 15-crown-5 (33.4 g, 152 mmol) were suspended in 200 mL DMF. The mixture was stirred at 60-70°C for 3 days. The precipitate was removed by filtration, and the filtrate was diluted with EA. The solvent was washed with brine and dried over Na2SO4. The solvent was removed, and the residue was purified on a silica gel column to afford compound 111-7 as a white foam (4.4 g, 46%). ESI-LCMS: m/z=617 [M+H]+.
[0603] Préparation of (1 lia): Compound 111-7 (4.4 g, 7.13 mmol) was dissolved in 100 mL of saturated methanolic ammonia, and the resulting solution was stirred at R.T. for 14 hours. The solvent was removed, and the residue was purified on a silica gel column (DCM/MeOH = 30:1 to 10:1) to give 11 la as a whitesolid (1.9 g, 88%). *H NMR (CDjOD, 400 M Hz) δ 7.70 (d, J= 7.6 Hz, IH), 6.40 (t, 7.2 Hz, IH), 5.93 (d, J= 7.6 Hz, IH), 4.50 (t, J=
13.2 Hz, IH), 3.88 (dd, Jt = 12.0 Hz, J2 = 26.8 Hz, 2H); ESI-MS: m/z=3O5 [M+H]\ 609 [2M+HJ*.
252
EXAMPLE 109 Prenaratlon of Compound (112a)
nh2 rt H(5 'F 112-1 NHBz —. — Hd 'F 112-2 NHBz rt -'Ύ/Λ - Ht? > 112-3
__NHBz HÔ *F 112-4 __NHBz rt HO'’ V 112-5 __NHBz Πν - Βζό F 112-6
IVHBz rt Bzd F 112-7 nh2 Oj — HO' 'F 112a
[0604| Préparation of (112-2): To a stirred solution of compound 112-1 (21.0 g, 85.7 mmol) in DMF (100 mL) was added benzoyl anhydride (9.66 g, 87 mmol) in portions. The mixture was stirred at R.T. ovemight. The solvent was removed under reduced pressure, and the residue was triturated with CH2CI2 to give compound 112-2 as a white solid (29.90 g, 100%).
[06051 Préparation of (112-3): To a stirred suspension of compound 112-2 (10.0 g,
28.65 mmol), PPh3 (15.01 g, 57.30 mmol) and pyridine (20 mL) in anhydrous THF (100 mL) was added dropwise a solution of I2 (14.55 g, 57.30 mmol) in THF (50 mL) at 0°C. After addition, the mixture was warmed to R.T. and stirred for 14 hours. The reaction was quenched with saturated aqueous Na2S2O3 (150 mL) and extracted with EA (100 mL, 3 times). The organic layer was dried over Na2SO4 and concentrated. The residue was purified on a silica gel column (DCM/MeOH = 100:1 to 50:1) to afford compound 112-3 (4.61 g, 35.1%) as a white solid.
[0606| Préparation of (112-4): To a stirred solution of compound 112-3 (4.6 g,
10.02 mmol) in anhydrous DMF ( 100 mL) was added dropwise a suspension of t-BuOK (3.36 g,
30.06 mmol) in DMF (20 mL) at 0°C over 10 min. The mixture was stirred at R.T. for 2 hours.
The mixtures was then quenched with saturated aqueous NH4CI (50 mL), and extracted with
THF and EA. The organic layer was washed with brine and dried over Na2SO4. The solvent
253 was removed, and the residue was purified on a silica gel column (MeOH/DCM = 1/100 to 1/30) to afford compound 112-4 as white solid (3.30 g, 99.6%).
[0607] Préparation of (112-5): To a stirred solution of BnEtjNCl (11.69 g, 50.2 mmol) in MeCN (50 mL) was added NaN3 (326 g, 50.2 mmol). The mixture was sonicated for 20 min and then stirred at R.T. for 16 hours. The solution was filtratcd into a solution of compound 112-4 (3.31 g, 10.02 mmol) and NMM (5.02 g, 50.2 mmol) in anhydrous THF (80 mL). The mixture was cooled to 0°C, and a solution of I2 (12.5 g, 50.2 mmol) in THF (40 mL) was added dropwise. Stirring was continued at 0-10°C for 20 hours. N-Acetyl cystein was added until no gas evolved. Saturated aqueous Na2S2O3 was added unti! a light yellow solution achieved. The solution was concentrated and then diluted with EA. The organic phase was washed with brine and dried over Na2SÛ4. The solvent was removed, and the residue was purified on a silica gel column (PE:EA:DCM = 1:1:1) to give compound 112-5 (14.7 g, 84%) as a white foam. 'H NMR (CD3OD, 400 MHz) δ 11.41 (s, 1 H), 8.19 (d, J « 7.2 Hz, 1 H), 8.00 (d, J = 7.2 Hz, IH), 7.62-7.66 (m, IH), 7.50-7.54 (m, 2H), 7.39 (d, J= 7.2 Hz, IH), 6.44 (d, J = 6.8 Hz, IH), 6.13 (d, J= 20.4 Hz, IH), 5.36-5.41 (m, IH), 4.70-4.76 (m, IH), 3.72 (dd, J, = 17.6 Hz,J2= 11.6 Hz, 2H).
[0608] Préparation of(112-6): To a stirred solution ofcompound 112-5 (3.6 g, 7.20 mmol) in anhydrous pyridine (80 mL) was added BzCl (1.31 g, 9.36 mmol) dropwise at 0°C. The mixture was stirred at R.T. for 10 hours. The reaction was quenched with HiO, and the solution was concentrated. The residue was dissolved in EA and washed with saturated aqueous NaHCO3. The organic layer was dried over Na2SO4 and concentrated. The residue was purified on a silica gel column (PE/EA = 10/1 to 1/1) to give compound 112-6 (3.2 g, 73.7%) as a pale yellow foam.
[0609] Préparation of (112-7): Compound 112-6 (2.0 g, 3.31 mmol), BzONa (4.76 g, 33.1 mmol) and 15-crown-5 (7.28 g, 33.1 mmol) were suspended in DMF (100 mL). The mixture was stirred at 60-70°C for 3 days. The precipitate removed by filtration, and the filtrate was diluted with EA. The solution was washed with brine and dried over NaiSO^. The solvent was removed, and the residue was purified on a silica gel column (PE/EA = 4/1 to 2/1) to afford compound 112-7 as a light yellow foam (1.0 g, 50.7%).
[0610] Préparation of (112a): Compound 112-7 (0.5 g, 0.84 mmol) was dissolved in methanolic ammonia (30 mL), and the mixture was stirred at R.T. for 14 hours. The solvent was removed, and the residue was purified on a silica gel column (DCM/MeOH = 30:1 to 10:1) to give 112a as white solids (0.11 g, 41.8%). ’H NMR (CD3OD, 400 MHz) S 7.83 (d, J = 7.6
Hz, IH), 6.10 (dd, Jt = 19.6 Hz, J2 = 1.6 Hz, IH), 5.94 (d, J= 7.6 Hz, IH), 5.10 (ddd, J, = 53.6
254 φ Hz,5.2 Hz,Λ = 1.2 Hz, IH), 4.57 (t, J= 5.2 Hz, IH), 3.82 (dd, J, = 38.0 Ηζ,Λ= 12.4 Hz,
2H); ESI-MS: m/z=287 [M + H]+, 573 [2M+H]+.
EXAMPLE 110
Préparation of Compoiind (113a)
113-1 113-2
113-3 113a [0611] Préparation of (113-2): To a stirred solution of compound 113-1 (4.6 g, i 6.2 mmol) in anhydrous pyridine (40 mL) was added BzCi (7.3 g, 51.8 mmol) dropwise at 0°C. The mixture was stirred at R.T. for 14 hours. The réaction was quenched with H2O and the solution 5 was concentrated. The residue was dissolved in EA and washed with saturated NaHCOj. The organic layer was dried over Na2SO4 and concentrated. The residue was purified on a silica gel column (PE/EA = 10/1 to 1/1) to give compound 113-2 (7.4 g, 84.1%).
[0612] Préparation of (113-3): Compound 113-2 (7.4 g, 12.4 mmol), DMAP (3.1 g,
24.8 mmol), TPSC1 (7.5 g, 24.8 mol) and EtjN (2.5 g, 24.8 mmol) were suspended in MeCN (50 jQ mL). The mixture was stirred at R.T. for 14 hours. The solvent was removed, and the residue was dissolved in NHj (200 mL) in THF. The mixture was stirred at R.T. for 2 hours. The solvent was removed, and the residue was purified on a silica gel column (DCM/MeOH = 100:1 to 50:1) to give the crude product. The crude product was dissolved in anhydrous pyridine (50 mL), and BzCl (1.7g, 12.2 mmol) was added dropwise at 0°C. The mixture was stirred at R.T.
for 14 hours. The reaction was quenched with H2O, and the solution was concentrated. The residue was dissolved in EA and washed with saturated NaHCOj. The organic layer was dried over Na2SO4 and concentrated. The residue was purified on a silica gel column (PE/EA = 10/1 to 1/1) to give compound 113-3 as a white foam (4.2 g, 48.4%).
[0613] Préparation of (113a): Compound 113-3 (4.2 g, 6.0 mmol) was dissolved in ,q 200 mL of saturated methanolic ammonia, and the mixture was stirred at R.T. for 14 hours. The solvent was removed and then water added. The aqueous mixture was washed with DCM several times and lyophilized to give 113a as a white solid (1.5 g, 88%). *H NMR (CDjOD, 400
255 • MHz) Ô7.74 (d, J= 7.2 Hz, IH), 6.43 (d, J= 5.6 Hz, IH), 5.87 (d, J= 7.6 Hz, IH), 4.39 (dd, J{ “ 2.4 Hz, J2 = 5.6 Hz, IH), 4.15 (d, J= 5.6 Hz, IH), 3.80 (s, IH). ESI-MS: m/z=285 [M+H]+.
EXAMPLE 111 RSV Antiviral Assays [0614] CPE réduction assays are performed as described by Sidwell and Huffinan et al., Appl Microbiol, (1971) 22(5):797-801 with slight modifications. HEp-2 cells (ATCC) at a 5 concentration of 6000 cell/weil are infected with RSV Long strain (ATCC) at a multiplicity of infection (mo.i.) of 0.01, and each of the test compounds are provided to duplicate wetls at final concentrations starting from 100 μΜ using i/3 stepwise dilutions. For each compound, two welts are set aside as uninfected, untreated cell contrats (CC), and two wetls per test compound receive virus only as a control for virus réplication (VC). The assay is stopped after 6 days, <10 before ait of the cells in the virus-infected untreated contrai wetls exhibited signs of virus cytopathology (giant cell formation, syncytia). At the end of the incubation, 20 μΐ of cell counting kit-8 reagent (CCK-8, Dojindo Molecular Technologies, Inc.) are added to each well. After 4 hour incubation, the absorbance is measured in each well according to manufacturer^ instruction, and the 50% effective concentration (ECso) is calculated by using régression 1 g analysis, based on the mean O.D. at each concentration of compound.
[0615] RT-PCR based assays were performed in HEp-2 cells (ATCC: CCL-23) at a concentration of 20000 cell/well were plated in 96 well plates and incubated ovemight. Each of the test compounds were 1/3 serially diluted and dosed to HEp-2 cells in duplicates. The highest final concentration for each compound was 100 uM. After 24 hour compound pre-incubation, 20 RSV A2 (ATCC: VR-1540) at MOI of 0.1 was added. Two wells per compound were set aside as uninfected, untreated cell contrais (CC), and four wells per test compound received virus only as a control for virus réplication (VC). The assay was stopped 4 days after virus infection and conditioned media was removed for viral RNA isolation. The quantifies of the RSV virus were measured by reai-time PCR using a set of RSV spécifie primera and probe. The data was 25 analyzed with Prism software with EC50 defined as drug concentration that reduced the viral load 50% from the viral control (VC).
[0616] Standard RSV polymerase assays were conducted in the presence of 3 pL extract of RSV-infected cells in a réaction buffer containing 50mM tris-acetate pH 8, !20mM Kacetate, 4.5mM MgCt2, 5% glycerol, 2mM EDTA, 50ug/mL BSA, and 3mM DTT. Varying jq concentration of test compounds were used to înitiate RNA synthesis for 120 mins at 30°C„ and radioactive 33P GTP (15 uCi) was used as tracer. The reaction was stopped by adding 50 mM
EDTA, and RNA samples were purified through G-50 size exclusion spin columns and phénol256 chloroform extraction. The radio-labelcd RNA products were resolved by ctectrophorcsis on a 6% polyacrylamide TBE gel, and visuatized and quantitated after being exposed on a phosphorlmager screen. Polymerase inhibition experiments (IC50) were conducted the same way in the presence of increasing concentration of test compounds.
[0617] Compounds of Formula (I), Formula (II) and Formula (ΠΙ) are active in the assay as noted in Tables 6 and 7. In Table 6, ‘A* indicates an ECjo < 2 μΜ, ‘B’ indicates an ECjo of >2 μΜ and <10 μΜ and ‘C* indicates an EC50 > 10 μΜ and < 50 μΜ. In Table 7, ‘A’ indicates an ECso < 1 μΜ, *B* indicates an ECso of >1 μΜ and < 10 μΜ and ‘C’ indicates an ECîo> 10 μΜ and< 100 μΜ.
Table 6 - Activity of compounds as determined bv RSV polymerase assay
No. ECm No. ECso No. EC50 No. ECOT No. EC50
35a A 36i B 56c A 97b A 97g A
36a A 36j B 56da A 97c A 98b A
36c A 56a B 56e A 97d A 98c A
36e A 56a B 97a A
Table 7 - Activity of compounds as determined bv RT-PCR assav
No. ECso No. EC50 No. ECso No. ECso No. ECso No. ECso
la C 14a A 28a B 48a B 8ta B 106a C
2a C 20a B 30a A 50a A 82a A 108a B
3a A 21a A 31a B 52a A 83a B -
4a C 22a C 33a A 58a C 85a A -
7a A 23a A 39a B 69a A 86a A -
9a C 25a C 41a B 7ta A 87a A -
lia B 26a B 46a B 73a C 92a C -
13a C 27a B 45a C 76a A 105a C -
EXAMPLE î 12 Influenza Antiviral Assay [0618] Human tung carcinoma A549 cells (ATCC, Manassas, VA) were plated at a density of 5 x 104 celts/mL (5 x 103 cells/wcll) in assay media (Ham’s Fl2 media supplemented with 0.3% FBS, 1% penicinin/streptomycin (ail Mediatech, Manassas, VA) and 1% DMSO (Sigma-Aldrich, St Louis, MO)) in black 96-wcll plates. After 24 hours, seriatly diluted test
257
compounds were added to ce ils and incubated for an additional 24 hours. Cells were infected with 250 IU/well of Influenza strain A/WSN/33 (H1N1) (Virapur, San Diego CA) and incubated for 20 hours at 37°C, 5% CO2. The cell culture supematant was aspirated off and 50 pL of 25 pM 2’-(4-Methylumbelliferyl)-a-D-N-acctylneuraminic acid (Sigma-Aldrich) dissolved in 33 mM MES, pH 6.5 (Emeratd Biosystems, Bainbridge Istand, WA) was added to the cells. After incubation for 45 mins at 30°C, reactions were stopped by addition of 150 pL stop solution (100 mM glycine, pH 10.5, 25% éthanol, ail Sigma-Aldrich). Fluorescence was measured with excitation and émission filters of 355 and 460 nm, respectively, on a Victor X3 multi-label plate reader (Perkin Elmer, Waltham, MA). Cytotoxicity of uninfected parallel cultures was determined by addition of 100 pLof CellTiter-Glo®reagent (Promega, Madison, Wl), and incubation for 10 mins at R.T. Luminescence was measured on a Victor X3 multi-label plate reader.
[0619] Compounds of Formula (I), Formula 01) and Formula 011) are active in the assay as notcd in Table 8, where ‘A’ indicatcs an EC30 < 20 pM, *B’ indicates an ECjo of >20 15 pM and < 100 μΜ and *C’ indicates an ECjo > 100 pM and < 250 μΜ.
Table 8 - Activity of compounds
No. % Inhibition No. % Inhibition
la C 20a C
2a C 21a C
3a C 22a C
4a C 23a C
6a C 25a A
7a C 26a C
9a C 27a B
12a C 28a C
I6a C 30a C
17a C 31a C
18a C 39a B
EXAMPLE 113 Influenza Pol Assay [0620] Recombinant influenza polymerase trimer is obtained as described (Aggarwal
S. et al., PLoS ONE 2010). Standard RNA polymcrization assays are conducted in the presence
258
of 0,15 uM enzyme, U uM 50-mer oligonucleotide template, 400 uM AG primer and varying concentration of the test compounds are incubated together for 40 minutes at 30°C. Radioactive
P GTP are used as the tracer and the radio-labeled RNA products are resol ved by electrophoresis on a 15% polyacrylamide TBE gel, and is visualîzcd and quantitated after being 5 exposed on a phosphorlmager screen. Polymciase inhibition expérimenta (ICjo) are conducted the same way in the presence of increasing concentration of test compounds.
EXAMPLE 114 Parainfluenza vlrus-3 (PIV-3) RT-PCR Assavs [0621] RT-PCR based assays were performed in A549 cells (ATCC: CCL-185). A549 cells at a concentration of 20,000 cells/well were plated in 96 well plates and incubated overnight. Each of the test compounds were 1/3 serially diluted and dosed to A549 cells in 10 duplicates. The highest final concentration for each compound was 100 uM. After 24 hour compound pre-incubation, human parainfluenza virus 3 (hPIV3, ATCC: VR-93) at multiplicity of infection (MOI) of 0.1 was added. Two wells per compound were set aside as uninfected, untreated cell controls (CC), and four wells per test compound received virus only as a control for virus réplication (VC). The assay was stopped 7 days after virus infection and conditioned 5 media was removed for viral RNA isolation. The quantities of the hPIV3 virus were measured by real-time PCR using a set of hPIV3 spécifie primera and probe. The data was analyzed with Prism software with ECso defined as drug concentration that reduced the viral load 50% from the viral control (VC). The results are provided in Table 9 and show Compounds of Formula (I), Formula (II) and Formula (III) are active against PIV-3, where ‘A’ indicates an ECso < 20 μΜ, *B* indicates an EC50 of>20 μΜ and < 100 μΜ and ‘C* indicates an EC50 > 100 μΜ.
Table 9
No. Εϋ«(μΜ) No. ECjo (jiM) NO. EC«(uM)
3a A 22a C 71a C
6a C 23a C 111a A
7a A 40a C 112a A
9a C 43a c 113a C
20a C 69a A
EXAMPLE 115 Parainfluenza vlrus-3 (PIV-3) Enhanced Green Fluorescent Protein (eGFP) Assay [0622] HPIV3-eGFP based assays were performed in A549 cells (ATCC: CCL-185).
A549 cells at a concentration of 20,000 cells/well were plated in 96 well plates and incubated overnight. Each of the test compounds were 1/3 serially diluted and dosed to A549 cells in
259 duplicates. The highest final concentration for each compound was 100 uM. After 24 hour compound pre-incubation, hPIV3-eGFP, (Roth et at., Antiviral Res. (2009) 82(1):12-21.) at
MOI of 0.1 was added. Two wetls per compound were set aside as uninfected, untreated cell contrais (CC), and foin* wells per test compound received virus only as a control for virus réplication (VC). The assay was stopped 3 days after virus infection by removing the conditioned media and adding RIPA lysis buffer at 50 ul/well. The eGFP fluorescence signal in the lysis buffer was detected in a Victor 3 plate reader. The data was analyzed with Prism software with EC» defined as drug concentration that reduced the viral load 50% from the viral control (VC). The results are provided in Table 10 and show Compounds of Formula (I), 10 Formula 01) and Formula 011) are active against PIV-3, where ‘A’ indicates an EC» < 20 μΜ, ‘B’ indicates an ECso of >20 μΜ and < 100 μΜ and ’C’ indicates an ECso > 100 μΜ.
Table 10
No. EC50 (μΜ) No. EQoOiM)
3a A 40a C
4a C 43a C
6a C 69a B
7a A 71a C
9a C Ilia A
20a C 112a A
22a C 113a C
23a C
EXAMPLE 116 Parainfluenza virus-3 (PIV-3) Plaque Assay [0623] MA-104 cells were grown in 24-welI plates to a confluency of 90% in the presence of minimal essential medium (MEM) supplemented with 10% fêtai bovine sérum and antibiotics (C-EMEM). The cells were then washed twice with non-complete minimal essential medium (NC-EMEM). Test articles were dissolved in DMSO to a stock concentration of 10 15 mM.
[0624] An aliquot of 0.5mL of the test article at various concentrations was then inoculated in triplicate wells and incubated for 60 minutes at 37°C with 5% CO2 for the diffusion of test article into MA-104 cells. After the incubation period, a stock of human PIV type 3 was thawed and diluted with NC-EMEM to achieve a viral concentration of IO4 pfu/mL.
2q An aliquot of 0.1 mL was then inoculated into ail the wells except for the négative and test article toxîcîty control wells. Upon infection, the plates were incubated for 72 hours at 37°C at
5% CO2. After incubation, the plates were examined under microscopy to record cytotoxicity.
260
The supematants coliected for viral quantification using a standard plaque assay using MA-104 cells as the indicator cells.
[0625] To perform the plaques assay, MA-104 cells were grown to confluence in 24well plates. The cells were washed with serum-free medium prior to inoculation of duplicate wells with serial 10-fold dilutions of supematant sample. After 1 hour incubation at 37°Ç, the samples were aspirated and 1.0 mL of methyl cellulose overlay media was added to each well. After 6 days of culture, the cells were fixed and stained with 0.06% ciystal violet in 1% glutaraldehyde and viral plaques enumerated. The data was analyzed with Prism software with EQo defined as drug concentration that reduced the viral load 50% from the viral control (VÇ). Table 11 provides the results and show Compounds of Formula (I), Formula (II) and Formula (ΠΙ) are active against PIV-3, where ‘A’ indicates an EC50 < 20 μΜ.
Table 11
No. EC<o (μΜ)
3a A
7a A
111a A
EXAMPLE 117 Human metanneumovfrus (hMPV) TCIDct Assay [0626] LLC-MK2 cells were grown in 24-weIl plates to a confluency of 90% in the presence of minimal essential medium (MEM) supplemented with 10% fêtai bovine sérum and antibiotics (C-EMEM). The cells were then washed twice wîth non-complete minimal essential medium (NC-EMEM). Test articles were dissoived in DMSO to a stock concentration of 10 mM.
[0627] An aliquot of 0.5mL of the test article at various concentrations was then inoculated in triplicate wells and incubated for 60 minutes at 37°C with 5% CO2 for the diffusion of test article into LLC-MK2 cells. After the incubation period, a stock of human metapneumovirus was thawed and diluted with NC-EMEM to achieve a viral concentration of 104 pfu/mL. An aliquot of 0.1 mL was then inoculated into ail the wells except for the négative and test article toxicity control wells. Upon infection, the plates were incubated for 7 days at 37°C at 5% CO2. After incubation, the plates were examîned under microscopy to record cytotoxicity. The supematants coliected for viral quantification using a standard TCIDÎO assay using LLC-MK2 cells as the indicator cells. The data was analyzed with Prism software with ECÎO defined as drug concentration that reduced the viral load 50% from the viral control (VC). The results of the assay are shown in Table 12 and show Compounds of Formula (I), Formula soi 17135
(Π) and Formula (III) are active against human metapneumovirus, where ‘A’ indicates an ECîo < 20 μΜ.
Table !2
No. ECW (μΜ)
3a A
7a A
111a A
[0628] Although the foregoing has been described in some detail by way of illustrations and examples for purposes of clarity and understanding, it will be understood by 5 those of skill in the art that numerous and various modifications can be made without departing from the spirit of the présent disclosure. Therefore, it should be clearly understood that the forms disclosed herein are illustrative only and are not intended to limit the scope of the présent disclosure, but rather to also cover ail modification and alternatives coming with the true scope and spirit of the invention.
262

Claims (52)

  1. AMENDED CLAIMS
    1. Use of an effective amount of a compound selected from Formula (I), Formula (II), and Formula (III), or a pharmaceutically acceptable sait of the foregoing, in the préparation of a medicine for ameliorating or treating a viral infection caused by a virus selected from a henipavirus, a morbillivirus, a respirovirus, a rubulavirus and a metapneumovirus, wherein the compound is 5 selected from Formula (I), Formula (II), and Formula (III) has one of the following structures:
    (ΙΠ) wherein:
    B1a Bîb and BÎC are independently an optionally substituted heterocyclic base or an optionally substituted heterocyclic base with a protected amino group;
    R1a is selected from the group consisting of hydrogen, an optionally substituted acyl, an optionally substituted O-linked amino acid.
    263
    AMENDED CLAIMS when the dashed line (---) of Formula (I) is a single bond, R2* is CH2, and R3A is
    O (oxygen);
    when the dashed line (--) of Formula (I) is absent, R2* is selected from the group consisting of an optionally substituted Cm alkyl, an optionally substituted C24S alkenyl, an optionally substituted Cm alkynyl, an optionally substituted C3.6 cycloalkyl, an optionally substituted -Ο-Cm alkyl, an optionally substituted alkenyl, an optionally substituted -O-Cj^ alkynyl and cyano, and R3A is selected from the group consisting of OH, -OC(=O)R‘,a and an optionally substituted O-linked amino acid;
    R18 is selected from the group consisting of O, OH, , an optionally substituted Nlinked amino acid and an optionally substituted N-linked amino acid ester dérivative;
    R1C and R2C are independently selected from the group consisting of O', OH, an optionally substituted Cm alkoxy.
    , an optionally substituted N-linked amino acid and an optionally substituted N-linked amino acid ester dérivative; or n and R2C is 0' or OH;
    Rlcis
    R2b and R3C are independently selected from the group consisting of an optionally substituted Cm alkyl, an optionally substituted C» alkenyl, an optionally substituted Cm
    AMENDED CLAIMS alkynyl, an optionally substituted C3.6 cycloalkyl, an optionally substituted -O-Cu alkyl, an optionally substituted -O-C3-6 alkenyl, an optionally substituted -O-C3^ alkynyl and cyano;
    R40 is selected from the group consisting of OH, -OC(=O)R,,c and an optionally substituted O-linked amino acid;
    R4A, R3b and RJC are independently a halogen;
    R5A, R4b and R60 are independently hydrogen or halogen;
    R6A, R7A and R8A are independently selected from the group consisting of absent, hydrogen, an optionally substituted Ci.24 alkyl, an optionally substituted C2.24 alkenyl, an optionally substituted C2.24 alkynyl, an optionally substituted C3.5 cycloalkyl, an optionally substituted C3.6 cycloalkenyl, an optionally substituted aryl, an optionally substituted heteroaryl, an optionally substituted aiyl(Cj^ alkyl), an optionally substituted *(CR,5AR,6A)|rO-Ci.24 alkyl, an optionally substituted *-{CR17AR18A)(rO-Ci.24 alkenyl.
    R6A and R7A are taken together to form a moiety selected from the group consisting of an optionally substituted and an optionally substituted • 265
    AMENDED CLAIMS wherein the oxygens connected to R6A and R7A, the phosphorus and the moiety form a sixmembered to ten-membered ring system;. .
    R9A is independently selected from the group consisting of an optionally substituted Ci-24 alkyl, an optionally substituted C2.24 alkenyl, an optionally substituted C2.24 alkynyl, an 5 optionally substituted C3.6 cycloalkyl, an optionally substituted C3.6 cycloalkenyl,
    NR3OAR31A, an optionally substituted N-linked amino acid and an optionally substituted Nlinked amino acid ester dérivative;
    R10A and R,ia are independently an optionally substituted N-linked amino acid or an optionally substituted N-linked amino acid ester dérivative;
    10 RI2A, R13a and RI4A are independently absent or hydrogen;
    each R13A, each R16A, each RI7A and each Rl8A are independently hydrogen, an optionally substituted C1.24 alkyl or alkoxy;
    R19A r20A r22A r23A r5B( r«B^ R8B^ R9B, R9C RIOC R12C RI3C independently selected from the group consisting of hydrogen, an optionally substituted C1.24 alkyl and an 15 optional! y substi tuted aryl ;
    R2IA, R24A, R7B, Riob, Rllc and R14C are independently selected from the group consisting of hydrogen, an optionally substituted C(.24 alkyl, an optionally substituted aryl, an optionally substituted -O-C1.24 alkyl and an optionally substituted -O-aryl;
    R23*, R29A, R1ib and R,sc are independently selected from the group consisting of 20 hydrogen, an optionally substituted C1.24 alkyl and an optionally substituted aryl;
    RI6C, R,7C and R18C are independently absent or hydrogen;
    R26A and R27* are independently -ON or an optionally substituted substituent selected from the group consisting of C2-8 organylcarbonyl, C2-8 alkoxycarbonyl and C2-8 org anylaminocarbonyl;
    25 R28A is selected from the group consisting of hydrogen, an optionally substituted Cj.
    24-alkyl, an optionally substituted C2.24 alkenyl, an optionally substituted C2-24 alkynyl, an optionally substituted C34 cycloalkyl and an optionally substituted C34cycloalkenyl;
    R30a and R3IA are independently selected from the group consisting of hydrogen, an optionally substituted Ci.24-alkyl, an optionally substituted C2.24 alkenyl, an optionally
    266
    AMENDED CLAIMS substituted C2.24 alkynyl, an optionally substituted C^ cycloalkyl and an optionally substituted C3-6 cycloalkenyl;
    for Formula (III),---is a single bond or a double bond;
    when ----- is a single bond, each R7C and each R8C is independently hydrogen or halogen; and when---is a double bond, each R7C is absent and each Rsc is independently hydrogen or halogen;
    R”a and Rc are independently an optionally substituted C|.24-alkyl; m and n are independently 0 or 1 ;
    p and q are independently selected from the group consisting of 1,2 and 3;
    r is 1 or 2;
    Z1A, Z2A, Z3A, Z4*, ZlB, ZÎB and Z,c are independently O or S; and provided that when the dashed line (----) of Formula (I) is absent; R,A is
    R9A wherein R8A is an unsubstituted Cm alkyl or phenyl optionally parasubstituted with a halogen or methyl and R9A is methyl ester, ethyl ester, isopropyl ester, nbutyl ester, benzyl ester or phenyl ester of an amino acid selected from the group consisting of glycine, alanine, valine, leucine, phenylalanine, tryptophan, méthionine and proline; R3A is OH; R4A is fluoro; RSA is fluoro or hydrogen; and B,A is an unsubstituted uracil; then R2* cannot be -OCH3;
    provided that when the dashed line (----) of Formula (I) is absent; R1A is H; R3A is
    OH; R4A is fluoro; R5A is fluoro; and B1A is an unsubstituted cytosine; then R2* cannot be allenyl;
    provided that when the dashed line (---) of Formula (I) is absent; R,A is H; R3A is
    OH; R4A is fluoro; RSA is hydrogen; and BIA is an unsubstituted thymine; then R2* cannot be C] alkyl substituted with an N-amido; and provided that when the dashed line (---) of Formula (I) is absent; RIA is H; R3A is
    OH; R4A is fluoro; RJA is fluoro; and B,A is an unsubstituted cytosine; then R2* cannot be ethynyl.
    267
    AMENDED CLAIMS
  2. 2. The use of Claim 1, wherein the virus is a henipavirus, a morbillivinis and/or a rubulavirus.
  3. 3. The use of Claim 2, wherein the henipavirus is a nipahvirus; wherein the morbillivinis is the measles virus; and/or wherein the rubulavirus is the mumps virus.
  4. 4. The use of Claim 1, wherein the virus is a respirovirus.
  5. 5. The use of Claim 4, wherein the respirovirus is human parainfluenza virus 1.
  6. 6. The use of Claim 4, wherein the respirovirus is human parainfluenza virus 3.
  7. 7. The use of Claim 1, wherein the virus is a rubulavirus.
  8. 8. The use of Claim 7, wherein the rubulavirus is human parainfluenza virus 2 and/or human parainfluenza virus 4.
  9. 9. The use of Claim 1, wherein the virus is a metapneumovîrus.
  10. 10. The use of Claim 9, wherein the metaphenumovirus is human metapneumovirus.
  11. 11. The use of any one of Claims 1-10, wherein the compound is a compound of
    Formula (I).
  12. 12.
    The use of Claim 11, wherein R1A is
    Rt(W-P—
    R11A .
  13. 13. The use of Claim 12, wherein R6A and R7A are both hydrogen or absent.
  14. 14. The use of Claim 12, wherein one of R6A and R7A is hydrogen, and the other of R6A and R7a is selected from the group consisting of an optionally substituted Cj.24 alkyl, an optionally substituted C224 alkenyl, an optionally substituted C2.24 alkynyl, an optionally substituted cycloalkyl, an optionally substituted cydoalkenyl, an optionally substituted aryl, an optionally substituted heteroaryl and an optionally substituted aryl(CM alkyl); wherein both R6A and R7A are independently selected from the group consisting of an optionally substituted C1.24 alkyl, an optionally substituted C2.24 alkenyl, an optionally substituted C2.24 alkynyl, an optionally substituted C3.6 cycloalkyl, an optionally substituted Cj^ cydoalkenyl, an optionally substituted aryl, an optionally substituted heteroaryl and an optionally substituted aryl(C|^ alkyl); wherein at least one
    268
    AMENDED CLAIMS of R6A and R7A are selected from the group consisting of and the other of R6A and R7A is selected from the group consisting of absent, hydrogen, an optionally substituted C1-24 alkyl, an optionally substituted C2.24 aikenyl, an optionally substituted C2.24 alkynyl, an optionally substituted C3-6 cycloalkyl, an optionally substituted C3^ cycloalkenyl, an optionally substituted aryl, an optionally substituted heteroaryl and an optionally substituted aryl(Ct-e alkyl); wherein both R6A and
    R7A are independently selected from the group consisting of optionally substituted C1.24 alkyl; wherein R6A and R7A are both an optionally substituted C2.24 aikenyl; wherein R6A and R7A are both *-<CR,5AR,6A)rO-Ci.24 alkyl; wherein R6A and R7A are both *-{CR,7AR,8A)<rO-C2.24 aikenyl; wherein R6A and R7A are both an optionally substituted aryl; or wherein R6A and R7A are both an optionally substituted aryl(Ct^ alkyl).
    AMENDED CL AIMS
    The use of Claim 12, wherein R6A and R ; wherein R6A wherein
    R25A; wherein R6A and R7A are both wherein R and ; or wherein R6A and R7A can be taken together to form a *
    «
    5 moiety selected from the group consisting of an optionally substituted and an optionally substituted , wherein the oxygens connected to ReA and R7A, the phosphorus and the moiety form a six-membered to ten-membered ring system.
  15. 16. The use of Claim 15, wherein group consisting of:
    is selected from the
    270
    AMENDED CLAIMS wherein R is an optionally substituted aryl, an optionally substituted heteroaryl or an optionally substituted heterocydy!.
  16. 17. The use of any one of Claîms 12-16, wherein Z,A is O.
  17. 18. The use of any one of Claims 12-16, wherein ZlA is S.
  18. 19. The use of Claim 12, wherein R8A is selected from the group consisting of absent, hydrogen, an optionally substituted C].24 alkyl, an optionally substituted C2.24 alkenyl, an optionally substituted C2.24 alkynyl, an optionally substituted Cj^ cycloalkyi and an optionally substituted Cj^
    271
    AMENDED CLAIMS cycloalkenyl; and R9A is independently selected from the group consisting of an optionally substituted Ci-j4 alkyl, an optionally substituted C2.24 alkenyl, an optionally substituted C2.24 alkynyl, an optionally substituted C34 cycloalkyl and an optionally substituted C34 cycloalkenyl; wherein R8A is hydrogen, and R9A is NR30AR3tA, wherein R30 and R31 are independently selected from the group consisting of hydrogen, an optionally substituted C1.24 alkyl, an optionally substituted C2-24 alkenyl, an optionally substituted C2.24 alkynyl, an optionally substituted C34 cycloalkyl and an optionally substituted C34 cycloalkenyl; wherein R8A is absent or hydrogen; and R9a is an optionally substituted N-linked amino acid or an optionally substituted N-linked amino acid ester dérivative; or wherein R8A is an optionally substituted aryl; and R9A is an optionally substituted N-linked amino acid or an optionally substituted N-linked amino acid ester dérivative.
  19. 20. The use of Claim 12, wherein R9A is selected from the group consisting of alanine, asparagine, aspartate, cysteine, glutamate, glutamine, glycine, proline, serine, tyrosine, arginine, histidïne, isoleucine, leucine, lysine, méthionine, phenylalanine, threonine, tryptophan, valine and ester dérivatives thereof; or wherein R9A is selected from the group consisting of alanine isopropyl ester, alanine cyclohexyl ester, alanine neopentyl ester, valine isopropyl ester and leucine isopropyl ester; or wherein R9A has the structure ? wherein R33a is selected from the group consisting of hydrogen, an optionally substituted Ci^-alkyl, an optionally substituted C34 cycloalkyl, an optionally substituted aryl, an optionally substituted aiyl(C)4 alkyl) and an optionally substituted haloalkyl; R34A is selected from the group consisting of hydrogen, an optionally substituted C^ alkyl, an optionally substituted C14 haloalkyl, an optionally substituted C34 cycloalkyl, an optionally substituted Ce aryl, an optionally substituted Cio aryl and an optionally substituted aryl(C[4 alkyl); and R3SA is hydrogen or an optionally substituted Ci^-alkyl; or R34A and * »
    R are taken together to form an optionally substituted C34 cycloalkyl.
  20. 21. The use of Claim 20, wherein RMA is an optionally substituted Ci^-alkyl; R35A is hydrogen; and/or R33A îs an optionally substituted C14 alkyl, an optionally substituted C34 cycloalkyl or an optionally substituted benzyl,
  21. 22. The use of any one of Claims 12 or 19-21, wherein Z2A is O.
  22. 23. The use of any one of Claims 12 or 19-21, wherein Z2A is S.
    272
    AMENDED CLAIMS
  23. 24. The use of Claim 12, wherein R,0A and R1,A are both an optionally substituted Nlinked amino acid or an optionally substituted N-linked amino acid ester dérivative; wherein R10A and R,IA are independently selected from selected from the group consisting of alanine, asparagine, aspartate, cysteine, glutamate, glutamine, glycine, proline, serine, tyrosine, arginine, histidine, isoleucine, leucine, lysine, méthionine, phenylalanine, threonine, tryptophan, valine and ester dérivatives thereof; wherein R,0A and R,IA are independently selected from the group consisting of alanine isopropyl ester, alanine cyclohexyl ester, alanine neopentyl ester, valine isopropyl ester and leucine isopropyl ester; or wherein R,0A and R,,A are independently hâve the structure wherein R is selected from the group consisting of hydrogen, an optionally substituted CM-alkyl, an optionally substituted Cj^ cycloalkyl, an optionally substituted aryl, an optionally substituted aryI(CM alkyl) and an optionally substituted haloalkyl; R37A is selected from the group consisting of hydrogen, an optionally substituted Cm alkyl, an optionally substituted C,^ haloalkyl, an optionally substituted C34 cycloalkyl, an optionally substituted Ce aryl, an optionally substituted Cio aryl and an optionally substituted aryî(CM alkyl); and R38A is hydrogen or an optionally substituted CM-alkyl; or R37A and R38A are taken together to form an optionally substituted C34 cycloalkyl.
  24. 25. The use of Claim 24, wherein R37A is an optionally substituted CM-alkyl; R38A is hydrogen and/or R36A is an optionally substituted Cm alkyl, an optionally substituted C3.6 cycloalkyl or an optionally substituted benzyl.
  25. 26. The use of any one of Claims 12 or 24-25, wherein Z3A is O.
  26. 27. The use of any one of Claims 12 or 24-25, wherein Z3A is S.
  27. 28.
    The use of Claim 12, wherein R6A is absent or hydrogen; wherein Rl2A, RI3A and Rl4A are independently absent or hydrogen.
  28. 29. The use of Claim 28, wherein m is 0, and R12A and Rl3A are independently absent or hydrogen; or wherein m is 1, and R12A, RI3A and Rl4A are independently absent or hydrogen.
    273
    AMENDED CLAIMS
  29. 30. The use of Claim 11, wherein R1 Ais H.
  30. 31. The use of Claim 11, wherein RlA is an optionally substituted acyl.
  31. 32. The use of Claim 31, wherein the optionally substituted acyl is -C(=O)R39A, wherein R is selected from the group consisting of an optionally substituted Ci.» alkyl, an optionally substituted C2.12 alkenyl, an optionally substituted C2.11 alkynyl, an optionally substituted C3.8 cycloalkyl, an optionally substituted C3g cycloalkenyl, an optionally substituted Ce-io aryl, an optionally substituted heteroaryl, an optionally substituted heterocyclyl, an optionally substituted arylfCM alkyl), an optionally substituted heteroaryl(CM alkyl) and an optionally substituted heterocyclyl (Cm alkyl).
  32. 33. The use of Claim 32, wherein R39A is an unsubstituted C].j2 alkyl.
  33. 34. The use of Claim 11, wherein R1A is an optionally substituted O-linked amino acid;
    or wherein R1A is θ NH2 ( wherein R40A is selected from the group consisting of hydrogen, an optionally substituted Cm alkyl, an optionally substituted Cm haloalkyl, an optionally substituted C3.6 cycloalkyl, an optionally substituted Ce aryl, an optionally substituted Cio aryl and an optionally substituted aryl(CM alkyl); and R4|A is hydrogen or an optionally substituted Cmalkyl; or R40* and R41A are taken together to form an optionally substituted Cm cycloalkyl.
  34. 35. The use of Claim 34, wherein R40A is an optionally substituted CM-alkyl; and/or R41A is hydrogen.
  35. 36. The use of any one of Claims 11-35, wherein B1A is selected from the group consisting of:
    274
    AMENDED CLAIMS wherein:
    R*2 is selected from the group consisting of hydrogen, halogen and NHR12, wherein 5 Rn is selected from the group consisting of hydrogen, -C(=O)RK2 and -Cf^JOR12;
    Rb2 is halogen or NHRW2, wherein RW2 is selected from the group consisting of hydrogen, an optionally substituted Cm alkyl, an optionally substituted Cm alkenyl, an optionally substituted Cj-g cycloalkyl, -C(=O)Rm2 and -C(=O)ORN2;
    Ra is hydrogen or NHR02, wherein R02 is selected from the group consisting of 10 hydrogen, -C(=O)RK and -C(=O)ORQ2;
    R02 is selected from the group consisting of hydrogen, halogen, an optionally substituted Cm alkyl, an optionally substituted Cm alkenyl and an optionally substituted Cm alkynyl;
    Rm is selected from the group consisting of hydrogen, hydroxy, an optionally 15 substituted Cm alkyl, an optionally substituted C3.8 cycloalkyl, -C(=O)RR2 and -C(=O)ORS2;
    Rr îs selected from the group consisting of hydrogen, halogen, an optionally substituted CMalkyl, an optionally substituted Cm alkenyl and an optionally substituted Cm alkynyl;
    Y2 and Y3 are independently N or CRn, wherein Rn is selected from the group
    20 consisting of hydrogen, halogen, an optionally substituted CM-alkyl, an optionally substituted CM-alkeny! and an optionally substituted CM-alkynyl;
    R02 is an optionally substituted Cm alkyl;
    AMENDED CLAIMS
    R30 is hydrogen or NHR72, wherein R12 is independently selected from the group consisting of hydrogen, -C(=O)RU2 and -C(=O)ORV2; and R!Q ru RM2f RN2t rQ2 rw rs2, rü2 rv2 independentiy seïected from the group consisting of Cm alkyl, C2-e alkenyl, Cm alkynyl, Cm cycloalkyl, Cm 5 cycloalkenyl, Ce-ίο aryl, heteroaryl, heteroalicyclyl, aryI(CM alkyl), heteroaryl(CM alkyl) and heteroalicyclyl (Cm alkyl).
  36. 39. The use of Claim 36, wherein BlA is
  37. 40. The use of any one of Claims 11-39, wherein the dashed line (----) is absent, R2* is selected from the group consisting of an optionally substituted Cm alkyl, an optionally substituted
    Cm alkenyl, an optionally substituted Cm alkynyl, an optionally substituted -Ο-Cm alkyl, an optionally substituted -Ο-Cm alkenyl, an optionally substituted -Ο-Cm alkynyl and cyano, and
    276
    AMENDED CLAIMS
    R3A is selected from the group consisting of OH, -OC(=O)RA and an optionally substituted Olinked amino acid.
  38. 41. The use of Claim 40, wherein R2* is an optionally substituted C^ alkyl.
  39. 42. The use of Claim 41, wherein R2* is a halogen substituted C^ alkyl, a hydroxy substituted Ci-β alkyl, an alkoxy substituted Cj^ alkyl or a sulfenyl substituted Ci< alkyl.
  40. 43. The use of Claim 41, wherein R2* is a halogen substituted C]^ alkyl.
  41. 44. The use of Claim 40, wherein R2* is an optionally substituted C2-6 alkenyl, an optionally substituted C2< alkynyl, an optionally substituted C3< cycloalkyl, an optionally substituted -O-Ci_6 alkyl, an optionally substituted -O-C3^ alkenyl, an optionally substituted -OC3-6 alkynyl or cyano.
  42. 45. The use of any one of Claims 11-44, wherein R3A is OH.
  43. 46. The use of any one of Claims 11-44, wherein R3A is -OC(=O)R a, wherein R A is an optionally substituted Ci_g alkyl.
  44. 47. The use of any one of Claims 11 -44, wherein R3A is O-linked amino acid.
  45. 48. The use of Claim 47, wherein the O-linked amino acid is selected from the group consisting of alanine, asparagine, aspartate, cysteine, glutamate, glutamine, glycine, proline, serine, tyrosine, arginine, histidine, isoleucine, leucine, lysine, methionïne, phenylalanine, threonine.
    tryptophan and valine; or wherein R3A is θ NH2 , wherein R42A is selected from the group consisting of hydrogen, an optionally substituted Ci^ alkyl, an optionally substituted Ci-6 haloalkyl, an optionally substituted C3^ cycloalkyl, an optionally substituted Ce aryl, an optionally substituted Cio aryl and an optionally substituted aryl(Ci^ alkyl); and R43A is hydrogen or an optionally substituted Ci-4-alkyl; or R42A and R43A are taken together to form an optionally substituted C3_6 cycloalkyl.
  46. 49. The use of Claim 48, wherein R42A is an optionally substituted Ci^-alkyl; and/or R43A is hydrogen.
  47. 50. The use of any one of Claims 11-49, wherein R3A is hydrogen.
  48. 51. The use of any one of Claims 11 -49, wherein R3A is halogen.
  49. 52. The use of any one of Claims 11-51, wherein R4A is fluoro.
    AMENDED CLAIMS
  50. 53. The use of any one of Claims 1-10, wherein the compound is a compound of Formula (II).
  51. 54. The use of any one of Claims 1-10, wherein the compound is a compound of Formula (III).
  52. 55. The use of Claim 11, wherein the compound of Formula (I) is selected from the group consisting of:
OA1201400433 2012-03-21 2013-03-19 Substituted nucleosides, nucleotides and analogs thereof OA17135A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US61/613,836 2012-03-21
US13/721,988 2012-12-20

Publications (1)

Publication Number Publication Date
OA17135A true OA17135A (en) 2016-03-28

Family

ID=

Similar Documents

Publication Publication Date Title
AU2018203423B2 (en) Substituted nucleosides, nucleotides and analogs thereof
AU2013235220B2 (en) Substituted nucleosides, nucleotides and analogs thereof
US10485815B2 (en) Substituted nucleosides, nucleotides and analogs thereof
USRE48171E1 (en) Substituted nucleosides, nucleotides and analogs thereof
OA17135A (en) Substituted nucleosides, nucleotides and analogs thereof
OA17044A (en) Substituted nucleosides, nucleotides and analogs thereof.
NZ627179B2 (en) Substituted nucleosides, nucleotides and analogs thereof