US20170029457A1 - Methods for the synthesis of functionalized nucleic acids - Google Patents

Methods for the synthesis of functionalized nucleic acids Download PDF

Info

Publication number
US20170029457A1
US20170029457A1 US15/222,910 US201615222910A US2017029457A1 US 20170029457 A1 US20170029457 A1 US 20170029457A1 US 201615222910 A US201615222910 A US 201615222910A US 2017029457 A1 US2017029457 A1 US 2017029457A1
Authority
US
United States
Prior art keywords
alkyl
aryl
alkenyl
alkynyl
alkylene
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/222,910
Inventor
Gregory L. Verdine
Meena
Naoki Iwamoto
David Charles Donnell Butler
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Wave Life Sciences Pte Ltd
Original Assignee
Wave Life Sciences Pte Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Wave Life Sciences Pte Ltd filed Critical Wave Life Sciences Pte Ltd
Priority to US15/222,910 priority Critical patent/US20170029457A1/en
Publication of US20170029457A1 publication Critical patent/US20170029457A1/en
Assigned to WAVE LIFE SCIENCES LTD. reassignment WAVE LIFE SCIENCES LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BUTLER, DAVID CHARLES DONNELL, IWAMOTO, NAOKI, VERDINE, GREGORY L., ., MEENA
Priority to US15/941,494 priority patent/US10280192B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • C07H21/04Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids with deoxyribosyl as saccharide radical
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C309/00Sulfonic acids; Halides, esters, or anhydrides thereof
    • C07C309/63Esters of sulfonic acids
    • C07C309/64Esters of sulfonic acids having sulfur atoms of esterified sulfo groups bound to acyclic carbon atoms
    • C07C309/65Esters of sulfonic acids having sulfur atoms of esterified sulfo groups bound to acyclic carbon atoms of a saturated carbon skeleton
    • C07C309/66Methanesulfonates
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/04Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms
    • C07D295/10Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by doubly bound oxygen or sulphur atoms
    • C07D295/104Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by doubly bound oxygen or sulphur atoms with the ring nitrogen atoms and the doubly bound oxygen or sulfur atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings
    • C07D295/108Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by doubly bound oxygen or sulphur atoms with the ring nitrogen atoms and the doubly bound oxygen or sulfur atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings to an acyclic saturated chain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H1/00Processes for the preparation of sugar derivatives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H13/00Compounds containing saccharide radicals esterified by carbonic acid or derivatives thereof, or by organic acids, e.g. phosphonic acids
    • C07H13/02Compounds containing saccharide radicals esterified by carbonic acid or derivatives thereof, or by organic acids, e.g. phosphonic acids by carboxylic acids
    • C07H13/04Compounds containing saccharide radicals esterified by carbonic acid or derivatives thereof, or by organic acids, e.g. phosphonic acids by carboxylic acids having the esterifying carboxyl radicals attached to acyclic carbon atoms

Definitions

  • Oligonucleotides are useful in therapeutic, diagnostic, research and nanomaterials applications.
  • the use of natural sequences of DNA or RNA for therapeutics is limited because of their instability against extra and intracellular nucleases, poor cell penetration and distribution.
  • in vitro studies have shown that the properties of antisense nucleotides such as binding affinity, sequence specific binding to the complementary RNA (Cosstick and Eckstcin, 1985; LaPlanche et al., 1986; Latimer et al., 1989; Hacia et al., 1994; Mesmaeker et al., 1995), stability to nucleases are affected by the configurations of the phosphorous atoms Therefore, there is a need for modified oligonucleotides to impart stability towards ubiquitous nucleases, increase binding affinity towards complementary RNA and increase cell penetration and bio-distribution for a number of in-vitro and in-vivo applications.
  • nucleic acids comprise chiral phosphorous moieties.
  • Another embodiment provides a process for the preparation of phosphorothiotriesters of structure IIa, wherein W is O.
  • R 1 is selected from:
  • Another embodiment provides a process for the preparation of phosphorothiotriesters of structure IIIa, wherein the silylating reagent is selected from
  • silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide, trimethylsilyltriflate, chlorotrimethylsilane, or 1-(trimethylsilyl)imidazole.
  • silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide.
  • Another embodiment provides the process, wherein the H-phosphonate is covalently linked to a solid phase.
  • Another embodiment provides a process for the preparation of phosphorothiotriesters comprising non-stereorandom phosphorous linkages of structure IIIb, wherein W is O.
  • Another embodiment provides a process for the preparation of phosphorothiotriesters comprising non-stereorandom phosphorous linkages of structure IIIb, wherein R 1 is selected from:
  • Another embodiment provides a process for the preparation of phosphorothiotriesters comprising non-stereorandom phosphorous linkages of structure IIIb, wherein the silylating reagent is selected from
  • silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide, trimethylsilyltriflate, chlorotrimethylsilane, or 1-(trimethylsilyl)imidazole.
  • silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide.
  • Another embodiment provides the process, wherein the H-phosphonate is covalently linked to a solid phase.
  • Another embodiment provides the process wherein the phosphorothiotriesters of structure IIIb comprise non-stereorandom phosphorous linkages and the H-phosphonate of structure Ic comprise non-stereorandom phosphorous linkages; and W is independently selected from O, NH, or CH 2 . Another embodiment provides the process wherein W is O.
  • Another embodiment provides the process wherein R 6 is methyl.
  • bis(thiosulfonate) reagent of structure IVc is selected from:
  • Another embodiment provides a process for the preparation of phosphorothiotriesters of structure IIIa, wherein the silylating reagent is selected from
  • silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide, trimethylsilyltriflate, chlorotrimethylsilane, or 1-(trimethylsilyl)imidazole.
  • silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide.
  • Another embodiment provides the process, wherein the H-phosphonate is covalently linked to a solid phase.
  • FIG. 1 provides the 31 P NMR spectrum of Compound 100S in CD 3 CN as described in Example 6;
  • FIG. 2 provides the 31 P NMR spectrum of Compound 100S in CD 3 CN after adding BSTFA as described in Example 6;
  • FIG. 3 provides the 31 P NMR spectrum of Compound 100S in CD 3 CN after adding BSTFA, TEA and MTS as described in Example 6;
  • FIG. 4 provides the 31 P NMR spectrum of Compound 100R in CD 3 CN as described in Example 6;
  • FIG. 5 provides the 31 P NMR spectrum of Compound 100R in CD 3 CN as described in Example 6;
  • FIG. 6 provides the 31 P NMR spectrum of Compound 100R in CD 3 CN after adding BSTFA, TEA and MTS as described in Example 6.
  • C 1 -C x includes C 1 -C 2 , C 1 -C 3 . . . C 1 -C x .
  • a group designated as “C 1 -C 4 ” indicates that there are one to four carbon atoms in the moiety, i.e. groups containing 1 carbon atom, 2 carbon atoms, 3 carbon atoms or 4 carbon atoms, as well as the ranges C 1 -C 2 and C 1 -C 3 .
  • C 1 -C 4 alkyl indicates that there are one to four carbon atoms in the alkyl group, i.e., the alkyl group is selected from among methyl, ethyl, propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, and t-butyl.
  • a numerical range such as “1 to 10” refers to each integer in the given range; e.g., “1 to 10 carbon atoms” means that the group may have 1 carbon atom, 2 carbon atoms, 3 carbon atoms, 4 carbon atoms, 5 carbon atoms, 6 carbon atoms, 7 carbon atoms, 8 carbon atoms, 9 carbon atoms, or 10 carbon atoms.
  • heteroatom or “hetero” as used herein, alone or in combination, refer to an atom other than carbon or hydrogen. Heteroatoms are may be independently selected from among oxygen, nitrogen, sulfur, phosphorous, silicon, selenium and tin but are not limited to these atoms. In embodiments in which two or more heteroatoms are present, the two or more heteroatoms can be the same as each another, or some or all of the two or more heteroatoms can each be different from the others.
  • alkyl refers to a straight-chain or branched-chain saturated hydrocarbon monoradical having from one to about ten carbon atoms, or one to six carbon atoms. Examples include, but are not limited to methyl, ethyl, n-propyl, isopropyl, 2-methyl-1-propyl, 2-methyl-2-propyl, 2-methyl-1-butyl, 3-methyl-1-butyl, 2-methyl-3-butyl, 2,2-dimethyl-1-propyl, 2-methyl-1-pentyl, 3-methyl-1-pentyl, 4-methyl-1-pentyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 2,2-dimethyl-1-butyl, 3,3-dimethyl-1-butyl, 2-ethyl-1-butyl, n-butyl, isobutyl, sec-butyl, t-butyl, n
  • C 1 -C 6 alkyl or “C 1-6 alkyl” means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, 4 carbon atoms, 5 carbon atoms or 6 carbon atoms.
  • the “alkyl” is substituted. Unless otherwise indicated, the “alkyl” is unsubstituted.
  • alkenyl refers to a straight-chain or branched-chain hydrocarbon monoradical having one or more carbon-carbon double-bonds and having from two to about ten carbon atoms, or two to about six carbon atoms.
  • the group may be in either the cis or trans conformation about the double bond(s), and should be understood to include both isomers. Examples include, but are not limited to ethenyl (—CH ⁇ CH 2 ), 1-propenyl (—CH 2 CH ⁇ CH 2 ), isopropenyl [—C(CH 3 ) ⁇ CH 2 ], butenyl, 1,3-butadienyl and the like.
  • C 2 -C 6 alkenyl or “C 2-6 alkenyl”
  • alkenyl group may consist of 2 carbon atoms, 3 carbon atoms, 4 carbon atoms, 5 carbon atoms or 6 carbon atoms.
  • the “alkenyl” is substituted. Unless otherwise indicated, the “alkenyl” is unsubstituted.
  • alkynyl refers to a straight-chain or branched-chain hydrocarbon monoradical having one or more carbon-carbon triple-bonds and having from two to about ten carbon atoms, or from two to about six carbon atoms. Examples include, but are not limited to ethynyl, 2-propynyl, 2-butynyl, 1,3-butadiynyl and the like.
  • the “alkynyl” is substituted. Unless otherwise indicated, the “alkynyl” is unsubstituted.
  • heteroalkyl refers to alkyl, alkenyl and alkynyl structures respectively, as described above, in which one or more of the skeletal chain carbon atoms (and any associated hydrogen atoms, as appropriate) are each independently replaced with a heteroatom (i.e.
  • an atom other than carbon such as though not limited to oxygen, nitrogen, sulfur, silicon, phosphorous, tin or combinations thereof
  • heteroatomic group such as though not limited to —O—O—, —S—S—, —O—S—, —S—O—, ⁇ N—N ⁇ , —N ⁇ N—, —N ⁇ N—NH—, —P(O) 2 —, —O—P(O) 2 —, —P(O) 2 —O—, —S(O) 2 —, —S(O) 2 —, —SnH 2 — and the like.
  • haloalkyl refers to alkyl, alkenyl and alkynyl groups respectively, as defined above, in which one or more hydrogen atoms is replaced by fluorine, chlorine, bromine or iodine atoms, or combinations thereof.
  • two or more hydrogen atoms may be replaced with halogen atoms that are the same as each another (e.g. difluoromethyl); in other embodiments two or more hydrogen atoms may be replaced with halogen atoms that are not all the same as each other (e.g. 1-chloro-1-fluoro-1-iodoethyl).
  • Non-limiting examples of haloalkyl groups are fluoromethyl, chloromethyl and bromoethyl.
  • a non-limiting example of a haloalkenyl group is bromoethenyl.
  • a non-limiting example of a haloalkynyl group is chloroethynyl.
  • carbon chain refers to any alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl or heteroalkynyl group, which is linear, cyclic, or any combination thereof. If the chain is part of a linker and that linker comprises one or more rings as part of the core backbone, for purposes of calculating chain length, the “chain” only includes those carbon atoms that compose the bottom or top of a given ring and not both, and where the top and bottom of the ring(s) are not equivalent in length, the shorter distance shall be used in determining the chain length. If the chain contains heteroatoms as part of the backbone, those atoms are not calculated as part of the carbon chain length.
  • cycloalkyl refers to a saturated, hydrocarbon monoradical ring, containing from three to about fifteen ring carbon atoms or from three to about ten ring carbon atoms, though may include additional, non-ring carbon atoms as substituents (e.g. methylcyclopropyl).
  • a numerical range such as “C 3 -C 6 cycloalkyl” or “C 3-6 cycloalkyl”, means that the cycloalkyl group may consist of 3 carbon atoms, 4 carbon atoms, 5 carbon atoms or 6 carbon atoms, i.e., is cyclopropyl, cyclobutyl, cyclopentyl or cyclohepty, although the present definition also covers the occurrence of the term “cycloalkyl” where no numerical range is designated.
  • the term includes fused, non-fused, bridged and spiro radicals.
  • a fused cycloalkyl may contain from two to four fused rings where the ring of attachment is a cycloalkyl ring, and the other individual rings may be alicyclic, heterocyclic, aromatic, heteroaromatic or any combination thereof. Examples include, but are not limited to cyclopropyl, cyclopentyl, cyclohexyl, decalinyl, and bicyclo [2.2.1] heptyl and adamantyl ring systems. Illustrative examples include, but are not limited to the following moieties:
  • the “cycloalkyl” is substituted. Unless otherwise indicated, the “cycloalkyl” is unsubstituted.
  • non-aromatic heterocyclyl and “heteroalicyclyl” as used herein, alone or in combination, refer to a saturated, partially unsaturated, or fully unsaturated nonaromatic ring monoradicals containing from three to about twenty ring atoms, where one or more of the ring atoms are an atom other than carbon, independently selected from among oxygen, nitrogen, sulfur, phosphorous, silicon, selenium and tin but are not limited to these atoms.
  • the two or more heteroatoms can be the same as each another, or some or all of the two or more heteroatoms can each be different from the others.
  • fused, non-fused, bridged and spiro radicals include fused, non-fused, bridged and spiro radicals.
  • a fused non-aromatic heterocyclic radical may contain from two to four fused rings where the attaching ring is a non-aromatic heterocycle, and the other individual rings may be alicyclic, heterocyclic, aromatic, heteroaromatic or any combination thereof.
  • Fused ring systems may be fused across a single bond or a double bond, as well as across bonds that are carbon-carbon, carbon-hetero atom or hetero atom-hetero atom.
  • the terms also include radicals having from three to about twelve skeletal ring atoms, as well as those having from three to about ten skeletal ring atoms.
  • Attachment of a non-aromatic heterocyclic subunit to its parent molecule can be via a heteroatom or a carbon atom.
  • additional substitution can be via a heteroatom or a carbon atom.
  • an imidazolidine non-aromatic heterocycle may be attached to a parent molecule via either of its N atoms (imidazolidin-1-yl or imidazolidin-3-yl) or any of its carbon atoms (imidazolidin-2-yl, imidazolidin-4-yl or imidazolidin-5-yl).
  • non-aromatic heterocycles contain one or more carbonyl or thiocarbonyl groups such as, for example, oxo- and thio-containing groups.
  • Examples include, but are not limited to pyrrolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidino, morpholino, thiomorpholino, thioxanyl, piperazinyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, 1,2,3,6-tetrahydropyridinyl, 2-pyrrolinyl, 3-pyrrolinyl, indolinyl, 2-
  • the terms also include all ring forms of the carbohydrates, including but not limited to the monosaccharides, the disaccharides and the oligosaccharides.
  • the “non-aromatic heterocyclyl” or “heteroalicyclyl” is substituted. Unless otherwise indicated, the “non-aromatic heterocyclyl” or “heteroalicyclyl” is unsubstituted.
  • aryl refers to an aromatic hydrocarbon radical of six to about twenty ring carbon atoms, and includes fused and non-fused aryl rings.
  • a fused aryl ring radical contains from two to four fused rings where the ring of attachment is an aryl ring, and the other individual rings may be alicyclic, heterocyclic, aromatic, heteroaromatic or any combination thereof.
  • aryl includes fused and non-fused rings containing from six to about twelve ring carbon atoms, as well as those containing from six to about ten ring carbon atoms.
  • a non-limiting example of a single ring aryl group includes phenyl; a fused ring aryl group includes naphthyl, phenanthrenyl, anthracenyl, azulenyl; and a non-fused bi-aryl group includes biphenyl.
  • the “aryl” is substituted. Unless otherwise indicated, the “aryl” is unsubstituted.
  • heteroaryl refers to an aromatic monoradicals containing from about five to about twenty skeletal ring atoms, where one or more of the ring atoms is a heteroatom independently selected from among oxygen, nitrogen, sulfur, phosphorous, silicon, selenium and tin but not limited to these atoms and with the proviso that the ring of said group does not contain two adjacent O or S atoms.
  • the two or more heteroatoms can be the same as each another, or some or all of the two or more heteroatoms can each be different from the others.
  • heteroaryl includes fused and non-fused heteroaryl radicals having at least one heteroatom.
  • heteroaryl also includes fused and non-fused heteroaryls having from five to about twelve skeletal ring atoms, as well as those having from five to about ten skeletal ring atoms. Bonding to a heteroaryl group can be via a carbon atom or a heteroatom.
  • an imidazole group may be attached to a parent molecule via any of its carbon atoms (imidazol-2-yl, imidazol-4-yl or imidazol-5-yl), or its nitrogen atoms (imidazol-1-yl or imidazol-3-yl).
  • a heteroaryl group may be further substituted via any or all of its carbon atoms, and/or any or all of its heteroatoms.
  • a fused heteroaryl radical may contain from two to four fused rings where the ring of attachment is a heteroaromatic ring and the other individual rings may be alicyclic, heterocyclic, aromatic, heteroaromatic or any combination thereof.
  • a non-limiting example of a single ring heteroaryl group includes pyridyl; fused ring heteroaryl groups include benzimidazolyl, quinolinyl, acridinyl; and a non-fused bi-heteroaryl group includes bipyridinyl.
  • heteroaryls include, without limitation, furanyl, thienyl, oxazolyl, acridinyl, phenazinyl, benzimidazolyl, benzofuranyl, benzoxazolyl, benzothiazolyl, benzothiadiazolyl, benzothiophenyl, benzoxadiazolyl, benzotriazolyl, imidazolyl, indolyl, isoxazolyl, isoquinolinyl, indolizinyl, isothiazolyl, isoindolyloxadiazolyl, indazolyl, pyridyl, pyridazyl, pyrimidyl, pyrazinyl, pyrrolyl, pyrazinyl, pyrazolyl, purinyl, phthalazinyl, pteridinyl, quinolinyl, quinazolinyl, quinoxalinyl, triazolyl,
  • the “heteroaryl” is substituted. Unless otherwise indicated, the “heteroaryl” is unsubstituted.
  • heterocyclyl refers collectively to heteroalicyclyl and heteroaryl groups.
  • the number of carbon atoms in a heterocycle is indicated (e.g., C 1 -C 6 heterocycle)
  • at least one non-carbon atom must be present in the ring.
  • Designations such as “C 1 -C 6 heterocycle” refer only to the number of carbon atoms in the ring and do not refer to the total number of atoms in the ring.
  • 4-6 membered heterocycle refers to the total number of atoms that are contained in the ring (i.e., a four, five, or six membered ring, in which at least one atom is a carbon atom, at least one atom is a heteroatom and the remaining two to four atoms are either carbon atoms or heteroatoms).
  • a four, five, or six membered ring in which at least one atom is a carbon atom, at least one atom is a heteroatom and the remaining two to four atoms are either carbon atoms or heteroatoms.
  • those two or more heteroatoms can be the same or different from one another.
  • Non-aromatic heterocyclic groups include groups having only three atoms in the ring, while aromatic heterocyclic groups must have at least five atoms in the ring. Bonding (i.e.
  • heterocycle attachment to a parent molecule or further substitution) to a heterocycle can be via a heteroatom or a carbon atom.
  • heterocyclyl is substituted.
  • heterocycyl is unsubstituted.
  • halogen halo or halide as used herein, alone or in combination refer to fluoro, chloro, bromo and/or iodo.
  • the compounds, or their pharmaceutically acceptable salts may contain one or more asymmetric centers and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, such as (R)- or (S)-.
  • the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both Z and E geometric isomers (e.g., cis or trans).
  • Z and E geometric isomers e.g., cis or trans
  • all possible isomers, as well as their racemic and optically pure forms, and all tautomeric forms are also intended to be included.
  • stereoisomer refers to the relationship between two or more compounds made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not superimposable.
  • enantiomer refers to two stereoisomers that are nonsuperimposeable mirror images of one another. It is contemplated that the various stereoisomers of the compounds disclosed herein, and mixtures thereof, are within the scope of the present disclosure and specifically includes enantiomers.
  • a “tautomer” refers to a compound wherein a proton shift from one atom of a molecule to another atom of the same molecule is possible.
  • the compounds presented herein may exist as tautomers. In solutions where tautomerization is possible, a chemical equilibrium of the tautomers will exist. The exact ratio of the tautomers depends on several factors, including temperature, solvent, and pH. Some examples of tautomeric equilibrium are shown below.
  • non-stereorandom phosphorous linkage(s) refers to a chiral phosphorous atom in the phosphodiester, or other isosteric linkage type, internucleotide linkage.
  • the handedness of chirality at phosphorous is independently selected at each phosphorous atom.
  • the oligonucleotide described herein is a pure diastereomer.
  • the oligonucleotide is greater that 95% diastereomeric purity.
  • the oligonucleotide is greater that 90% diastereomeric purity.
  • “Optional” or “optionally” means that a subsequently described event or circumstance may or may not occur and that the description includes instances when the event or circumstance occurs and instances in which it does not.
  • “optionally substituted alkyl” means that the alkyl radical may or may not be substituted and that the description includes both substituted aryl radicals and aryl radicals having no substitution.
  • Natural nucleic acids have a phosphate backbone; artificial nucleic acids may contain other types of backbones, but contain the same bases.
  • nucleotide refers to a monomeric unit of a polynucleotide that consists of a heterocyclic base, a sugar, and one or more phosphate groups.
  • the naturally occurring bases (guanine, (G), adenine (A), cytosine (C), thymine (T), and uracil (U)) are derivatives of purine or pyrimidine, though it should be understood that naturally and non-naturally occurring base analogs are also included.
  • the naturally occurring sugar is the pentose (five-carbon sugar) deoxyribose (which forms DNA) or ribose (which forms RNA), though it should be understood that naturally and non-naturally occurring sugar analogs are also included.
  • Nucleic acids are linked via phosphate bonds to form nucleic acids, or polynucleotides, though many other linkages are known in the art (such as, though not limited to phosphorothioates, boranophosphates and the like).
  • Artificial nucleic acids include PNAs (peptide nucleic acids), phosphothionates, and other variants of the phosphate backbone of native nucleic acids.
  • nucleoside refers to a moiety wherein a nucleobase or a modified nucleobase is covalently bound to a sugar or modified sugar.
  • sugar refers to a monosaccharide in closed and/or open form.
  • Sugars include, but are not limited to, ribose, deoxyribose, pentofuranose, pentopyranose, and hexopyranose moieties.
  • modified sugar refers to a moiety that can replace a sugar.
  • the modified sugar mimics the spatial arrangement, electronic properties, or some other physicochemical property of a sugar.
  • nucleic acid and “polynucleotide” as used herein refer to a polymeric form of nucleotides of any length, either ribonucleotides (RNA) or deoxyribonucleotides (DNA). These terms refer to the primary structure of the molecules and, thus, include double- and single-stranded DNA, and double- and single-stranded RNA. These terms include, as equivalents, analogs of either RNA or DNA made from nucleotide analogs and modified polynucleotides such as, though not limited to, methylated and/or capped polynucleotides.
  • RNA ribonucleotides
  • DNA deoxyribonucleotides
  • RNA poly- or oligo-ribonucleotides
  • DNA poly- or oligo-deoxyribonucleotides
  • RNA or DNA derived from N-glycosides or C-glycosides of nucleobases and/or modified nucleobases
  • nucleic acids derived from sugars and/or modified sugars and nucleic acids derived from phosphate bridges and/or modified phosphorous-atom bridges.
  • the term encompasses nucleic acids containing any combinations of nucleobases, modified nucleobases, sugars, modified sugars, phosphate bridges or modified phosphorous atom bridges.
  • Examples include, and are not limited to, nucleic acids containing ribose moieties, the nucleic acids containing deoxy-ribose moieties, nucleic acids containing both ribose and deoxyribose moieties, nucleic acids containing ribose and modified ribose moieties.
  • the prefix poly- refers to a nucleic acid containing about 1 to about 10,000 nucleotide monomer units and wherein the prefix oligo- refers to a nucleic acid containing about 1 to about 200 nucleotide monomer units.
  • nucleobase refers to the parts of nucleic acids that are involved in the hydrogen-bonding that binds one nucleic acid strand to another complementary strand in a sequence specific manner.
  • the most common naturally-occurring nucleobases are adenine (A), guanine (G), uracil (U), cytosine (C), and thymine (T).
  • modified nucleobase refers to a moiety that can replace a nucleobase.
  • the modified nucleobase mimics the spatial arrangement, electronic properties, or some other physicochemical property of the nucleobase and retains the property of hydrogen-bonding that binds one nucleic acid strand to another in a sequence specific manner.
  • a modified nucleobase can pair with all of the five naturally occurring bases (uracil, thymine, adenine, cytosine, or guanine) without substantially affecting the melting behavior, recognition by intracellular enzymes or activity of the oligonucleotide duplex.
  • chiral reagent refers to a compound that is chiral or enantiopure and can be used for asymmetric induction in nucleic acid synthesis.
  • chiral ligand or “chiral auxiliary” refers to a moiety that is chiral or enantiopure and controls the stereochemical outcome of a reaction.
  • condensation reagent refers to a reagent that activates a less reactive site and renders it more susceptible to attack by a nucleophile.
  • blocking group refers to a group that transiently masks the reactivity of a functional group.
  • the functional group can be subsequently unmasked by removal of the blocking group.
  • moiety refers to a specific segment or functional group of a molecule. Chemical moieties are often recognized chemical entities embedded in or appended to a molecule.
  • solid support refers to any support which enables synthetic mass production of nucleic acids and can be reutilized at need.
  • the term refers to a polymer, that is insoluble in the media employed in the reaction steps performed to synthesize nucleic acids, and is derivatized to comprise reactive groups.
  • linking moiety refers to any moiety optionally positioned between the terminal nucleoside and the solid support or between the terminal nucleoside and another nucleoside, nucleotide, or nucleic acid.
  • a “DNA molecule” refers to the polymeric form of deoxyribonucleotides (adenine, guanine, thymine, or cytosine) in its either single stranded form or a double-stranded helix. This term refers only to the primary and secondary structure of the molecule, and does not limit it to any particular tertiary forms. Thus, this term includes double-stranded DNA found, inter alia, in linear DNA molecules (e.g., restriction fragments), viruses, plasmids, and chromosomes.
  • linear DNA molecules e.g., restriction fragments
  • viruses e.g., plasmids, and chromosomes.
  • sequences can be described herein according to the normal convention of giving only the sequence in the 5′ to 3′ direction along the non-transcribed strand of DNA (i.e., the strand having a sequence homologous to the mRNA).
  • an “antisense” nucleic acid molecule comprises a nucleotide sequence which is complementary to a “sense” nucleic acid encoding a protein, e.g., complementary to the coding strand of a double-stranded cDNA molecule, complementary to an mRNA sequence or complementary to the coding strand of a gene. Accordingly, an antisense nucleic acid molecule can hydrogen bond to a sense nucleic acid molecule.
  • a “complementary DNA” or “cDNA” includes recombinant polynucleotides synthesized by reverse transcription of mRNA and from which intervening sequences (introns) have been removed.
  • nucleic acids comprise chiral phosphorous moieties.
  • Another embodiment provides a process for the preparation of phosphorothiotriesters of structure IIIa, wherein W is O.
  • R 1 is selected from:
  • Another embodiment provides a process for the preparation of phosphorothiotriesters of structure IIIa, wherein the silylating reagent is selected from
  • silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide, trimethylsilyltriflate, chlorotrimethylsilane, or 1-(trimethylsilyl)imidazole.
  • silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide.
  • Another embodiment provides the process, wherein the H-phosphonate is covalently linked to a solid phase.
  • Another embodiment provides a process for the preparation of phosphorothiotriesters comprising non-stereorandom phosphorous linkages of structure IIIb, wherein W is O.
  • Another embodiment provides a process for the preparation of phosphorothiotriesters comprising non-stereorandom phosphorous linkages of structure IIIb, wherein R 1 is selected from:
  • Another embodiment provides a process for the preparation of phosphorothiotriesters comprising non-stereorandom phosphorous linkages of structure IIIb, wherein the silylating reagent is selected from
  • silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide, trimethylsilyltriflate, chlorotrimethylsilane, or 1-(trimethylsilyl)imidazole.
  • silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide.
  • Another embodiment provides the process, wherein the H-phosphonate is covalently linked to a solid phase.
  • Another embodiment provides the process wherein the phosphorothiotriesters of structure IIIb comprise non-stereorandom phosphorous linkages and the H-phosphonate of structure Ic comprise non-stereorandom phosphorous linkages; and W is independently selected from O, NH, or CH 2 . Another embodiment provides the process wherein W is O.
  • Another embodiment provides the process wherein R 6 is methyl.
  • bis(thiosulfonate) reagent of structure IVc is selected from:
  • Another embodiment provides a process for the preparation of phosphorothiotriesters of structure IIIa, wherein the silylating reagent is selected from
  • silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide, trimethylsilyltriflate, chlorotrimethylsilane, or 1-(trimethylsilyl)imidazole.
  • silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide.
  • Another embodiment provides the process, wherein the H-phosphonate is covalently linked to a solid phase.
  • Oligonucleotides have several pharmaceutical properties which can be improved through the application of prodrug strategies.
  • oligonucleotides are rapidly degraded by nucleases and exhibit poor cellular uptake through the cytoplasmic cell membrane (Poijarvi-Virta et al., Curr. Med. Chem. (2006), 13(28); 3441-65; Wagner et al., Med. Res. Rev. (2000), 20(6):417-51; Peyrottes et al., Mini Rev. Med. Chem.
  • the steps of reacting a molecule comprising an achiral H-phosphonate moiety and a nucleoside comprising a 5′-OH moiety to form a condensed intermediate can occur without isolating any intermediates.
  • the steps of reacting a molecule comprising an achiral H-phosphonate moiety and a nucleoside comprising a 5′-OH moiety to form a condensed intermediate occurs is a one-pot reaction.
  • a molecule comprising an achiral H-phosphonate moiety, condensing reagent, chiral reagent, and compound comprising a free nucleophilic moiety are added to the reaction mixture at different times.
  • a molecule comprising an achiral H-phosphonate moiety, condensing reagent, and chiral reagent are present in the same reaction vessel or same pot.
  • a molecule comprising an achiral H-phosphonate moiety, condensing reagent, chiral reagent, and compound comprising a free nucleophilic moiety are present in the same reaction or same pot. This allows the reaction to be performed without isolation of intermediates and eliminates time-consuming steps, resulting in an economical and efficient synthesis.
  • the achiral H-phosphonate, condensing reagent, chiral amino alcohol, 5′-OH nucleoside are present at the same time in a reaction.
  • the formation of the chiral intermediate for condensation is formed in situ and is not isolated prior to the condensation reaction.
  • a molecule comprising an achiral H-phosphonate moiety has been activated by reaction with a condensing reagent, chiral reagent in a different reaction vessel from that used when reacting the chiral intermediate with the compound comprising a free 5′-OH moiety.
  • the synthesis of the nucleic acid is performed in solution. In other embodiments, the synthesis of the nucleic acid is performed on solid phase.
  • the reactive groups of a solid support may be unprotected or protected.
  • oligonucleotide synthesis a solid support is treated with various reagents in several synthesis cycles to achieve the stepwise elongation of a growing oligonucleotide chain with individual nucleotide units.
  • the nucleoside unit at the end of the chain which is directly linked to the solid support is termed “the first nucleoside” as used herein.
  • the first nucleoside is bound to the solid support via a linker moiety, i.e.
  • the linker stays intact during the synthesis cycles performed to assemble the oligonucleotide chain and is cleaved after the chain assembly to liberate the oligonucleotide from the support.
  • Solid supports for solid-phase nucleic acid synthesis include the supports described in, e.g., U.S. Pat. Nos. 4,659,774, 5,141,813, 4,458,066; Caruthers U.S. Pat. Nos. 4,415,732, 4,458,066, 4,500,707, 4,668,777, 4,973,679, and 5,132,418; Andrus et al. U.S. Pat. Nos. 5,047,524, 5,262,530; and Koster U.S. Pat. No. 4,725,677 (reissued as Re34,069).
  • the solid phase is an organic polymer support.
  • the solid phase is an inorganic polymer support.
  • the organic polymer support is polystyrene, aminomethyl polystyrene, a polyethylene glycol-polystyrene graft copolymer, polyacrylamide, polymethacrylate, polyvinylalcohol, highly cross-linked polymer (HCP), or other synthetic polymers, carbohydrates such as cellulose and starch or other polymeric carbohydrates, or other organic polymers and any copolymers, composite materials or combination of the above inorganic or organic materials.
  • the inorganic polymer support is silica, alumina, controlled poreglass (CPG), which is a silica-gel support, or aminopropyl CPG.
  • solid supports include fluorous solid supports (see e.g., WO/2005/070859), long chain alkylamine (LCAA) controlled pore glass (CPG) solid supports (sce e.g., S. P. Adams, K. S. Kavka, E. J. Wykes, S. B. Holder and G. R. Galluppi, J. Am. Chem. Soc., 1983, 105, 661-663; G. R. Gough, M. J. Bruden and P. T. Gilham, Tetrahedron Lett., 1981, 22, 4177-4180).
  • Membrane supports and polymeric membranes see e.g.
  • a membrane can be chemically functionalized for use in nucleic acid synthesis.
  • a linker or spacer group attached to the membrane may be used to minimize steric hindrance between the membrane and the synthesized chain.
  • suitable solid supports include those generally known in the art to be suitable for use in solid phase methodologies, including, for example, glass sold as PrimerTM 200 support, controlled pore glass (CPG), oxalyl-controlled pore glass (see, e.g., Alul, et al., Nucleic Acids Research, 1991, 19, 1527), TentaGel Support—an aminopolyethyleneglycol derivatized support (see, e.g., Wright, et al., Tetrahedron Lett., 1993, 34, 3373), and Poros-a copolymer of polystyrene/divinylbenzene.
  • CPG controlled pore glass
  • oxalyl-controlled pore glass see, e.g., Alul, et al., Nucleic Acids Research, 1991, 19, 1527
  • TentaGel Support an aminopolyethyleneglycol derivatized support (see, e.g., Wright, et al., Tetrahe
  • the solid support material can be any polymer suitably uniform in porosity, has sufficient amine content, and sufficiently flexible to undergo any attendant manipulations without losing integrity.
  • suitable selected materials include nylon, polypropylene, polyester, polytetrafluoroethylene, polystyrene, polycarbonate, and nitrocellulose.
  • Other materials can serve as the solid support, depending on the design of the investigator. In consideration of some designs, for example, a coated metal, in particular gold or platinum can be selected (see e.g., US publication No. 20010055761).
  • a nucleoside is anchored to a solid support which is functionalized with hydroxyl or amino residues.
  • the solid support is derivatized to provide an acid labile trialkoxytrityl group, such as a trimethoxytrityl group (TMT).
  • TMT trimethoxytrityl group
  • a diglycoate linker is optionally introduced onto the support.
  • a linking moiety or linker is optionally used to connect the solid support to the compound comprising a free nucleophilic moiety.
  • Suitable linkers are known such as short molecules which serve to connect a solid support to functional groups (e.g., hydroxyl groups) of initial nucleosides molecules in solid phase synthetic techniques.
  • the linking moiety is a succinamic acid linker, or a succinate linker (—CO—CH 2 —CH 2 —CO—), or an oxalyl linker (—CO—CO—).
  • the linking moiety and the nucleoside are bonded together through an ester bond.
  • the linking moiety and the nucleoside are bonded together through an amide bond.
  • the linking moiety connects the nucleoside to another nucleotide or nucleic acid.
  • Suitable linkers are disclosed in, for example, Oligonucleotides And Analogues A Practical Approach, Ekstein, F. Ed., IRL Press, N.Y., 1991, Chapter 1.
  • a linker moiety is used to connect the compound comprising a free nucleophilic moiety to another nucleoside, nucleotide, or nucleic acid.
  • the linking moiety is a phosphodiester linkage.
  • the linking moiety is an H-phosphonate moiety.
  • the linking moiety is an X-phosphonate moiety.
  • the solvent is acetonitrile, pyridine, or NMP.
  • the solvent is acetone, acetontrile, NMP, ethyl acetate, THF, dioxane, DMF, DMSO, DCM, chloroform, pyridine, 2,6-lutidine, HMPA, HMPT, DMA, glyme, diglyme, sulfone, methyl tert-butyl ether, or combinations thereof.
  • the solvent is a polar, aprotic organic solvent.
  • the solvent is anhydrous.
  • Acidification to remove blocking groups is accomplished by a Br ⁇ nsted acid or Lewis acid. In some embodiments, acidification is used to remove R 1 blocking groups.
  • Useful Br ⁇ nsted acids are carboxylic acids, alkylsulfonic acids, arylsulfonic acids, phosphoric acid and its derivatives, phosphonic acid and its derivatives, alkylphosphonic acids and their derivatives, arylphosphonic acids and their derivatives, phosphinic acid, dialkylphosphinic acids, and diarylphosphinic acids which have a pKa (25° C. in water) value of ⁇ 0.6 (trifluoroacetic acid) to 4.76 (acetic acid) in an organic solvent or water (in the case of 80% acetic acid).
  • the concentration of the acid (1 to 80%) used in the acidification step depends on the acidity of the acid. Consideration to the acid strength must be taken into account as strong acid conditions will result in depurination/depyrimidination, wherein purinyl or pyrimidinyl bases are cleaved from ribose ring.
  • acidification is accomplished by a Lewis acid in an organic solvent.
  • Useful Lewis acids are ZnX 2 wherein X is Cl, Br, I, or CF 3 SO 3 .
  • the acidifying comprises adding an amount of a Br ⁇ nsted or Lewis acid effective to convert the condensed intermediate into the compound of Formula 4 without removing purine moieties from the condensed intermediate.
  • Acids that are useful in the acidifying step also include, but are not limited to 10% phosphoric acid in an organic solvent, 10% hydrochloric acid in an organic solvent, 1% trifluoroacetic acid in an organic solvent, 3% dichloroacetic acid in an organic solvent or 80% acetic acid in water.
  • concentration of any Br ⁇ nsted or Lewis acid used in the process is selected such that the concentration of the acid does not exceed a concentration that causes cleavage of the nucleobase from the sugar moiety.
  • acidification comprises adding 1% trifluoroacetic acid in an organic solvent. In some embodiments, acidification comprises adding about 0.1% to about 8% trifluoroacetic acid in an organic solvent. In other embodiments, acidification comprises adding 3% dichloroacetic acid in an organic solvent. In other embodiments, acidification comprises adding about 0.1% to about 10% dichloroacetic acid in an organic solvent. In yet other embodiments, acidification comprises adding 3% trichloroacetic acid in an organic solvent. In yet other embodiments, acidification comprises adding about 0.1% to about 10% trichloroacetic acid in an organic solvent. In some embodiments, acidification comprises adding 80% acetic acid in water.
  • acidification comprises adding about 50% to about 90%, or about 50% to about 80%, about 50% to about 70%, about 50% to about 60%, about 70% to about 90% acetic acid in water.
  • the acidification comprises the further addition of cation scavengers to the acidic solvent.
  • the cation scavengers can be triethylsilane or triisopropylsilane.
  • R 1 is deblocked prior to the step of acidifying the condensed intermediate.
  • R 1 is deblocked by acidification, which comprises adding 1% trifluoroacetic acid in an organic solvent.
  • R 1 is deblocked by acidification, which comprises adding 3% dichloroacetic acid in an organic solvent. In some embodiments, R 1 is deblocked by acidification, which comprises adding 3% trichloroacetic acid in an organic solvent.
  • hydroxyl or amino moieties which are located on nucleobases or sugar moieties are routinely blocked with blocking (protecting) groups (moieties) during synthesis and subsequently deblocked.
  • a blocking group renders a chemical functionality of a molecule inert to specific reaction conditions and can later be removed from such functionality in a molecule without substantially damaging the remainder of the molecule (see e.g., Green and Wuts, Protective Groups in Organic Synthesis, 2 nd Ed., John Wiley & Sons, New York, 1991).
  • amino groups can be blocked with nitrogen blocking groups such as phthalimido, 9-fludrenylmethoxycarbonyl (FMOC), triphenylmethylsulfenyl, t-BOC, 4,4′-dimethoxytrityl (DMTr), 4-methoxytrityl (MMTr), 9-phenylxanthin-9-yl (Pixyl), trityl (Tr), or 9-(p-methoxyphenyl)xanthin-9-yl (MOX).
  • Nitro blocking groups such as phthalimido, 9-fludrenylmethoxycarbonyl (FMOC), triphenylmethylsulfenyl, t-BOC, 4,4′-dimethoxytrityl (DMTr), 4-methoxytrityl (MMTr), 9-phenylxanthin-9-yl (Pixyl), trityl (Tr), or 9-(p-methoxyphenyl)xanthin
  • Hydroxy groups can be protected such as tetrahydropyranyl (THP), t-butyldimethylsilyl (TBDMS), 1-[(2-chloro-4-methyl)phenyl]-4-methoxypiperidin-4-yl (Ctmp), 1-(2-fluorophenyl)-4-methoxypiperidin-4-yl (Fpmp), 1-(2-chloroethoxy)ethyl, 3-methoxy-1,5-dicarbomethoxypentan-3-yl (MDP), bis(2-acetoxyethoxy)methyl (ACE), triisopropylsilyloxymethyl (TOM), 1-(2-cyanoethoxy)ethyl (CEE), 2-cyanoethoxymethyl (CEM), [4-(N-dichloroacetyl-N-methylamino)benzyloxy]methyl, 2-cyanoethyl (CN), pivaloyloxymethyl (PivOM), levunyloxymethyl (
  • hydroxyl blocking groups are acid-labile groups, such as the trityl, monomethoxytrityl, dimethoxytrityl, trimethoxytrityl, 9-phenylxanthin-9-yl (Pixyl) and 9-(p-methoxyphenyl)xanthin-9-yl (MOX).
  • Chemical functional groups can also be blocked by including them in a precursor form. Thus an azido group can be considered a blocked form of an amine as the azido group is easily converted to the amine.
  • R 2 is a blocking group
  • removal of the blocking group at R 2 is orthogonal to the removal of the blocking group at R 1 .
  • the blocking groups at R 1 and R 2 remain intact during the synthesis steps and are collectively removed after the chain assembly.
  • the R 2 blocking group are removed simultaneously with the cleavage of the nucleic acids from the solid support and with the removal of the nucleobase blocking groups.
  • the blocking group at R 1 is removed while the blocking groups at R 2 and nucleobases remain intact.
  • Blocking groups at R 1 are cleavable on solid supports with an organic base such as a primary amine, a secondary amine, or a mixture thereof. Deblocking of the R 1 position is commonly referred to as front end deprotection.
  • the nucleobase blocking groups are cleavable after the assembly of the respective nucleic acid with an acidic reagent.
  • one or more of the nucleobase blocking groups is cleavable under neither acidic nor basic conditions, e.g. cleavable with fluoride salts or hydrofluoric acid complexes.
  • one or more of the nucleobase blocking groups are cleavable after the assembly of the respective nucleic acid in the presence of base or a basic solvent, and wherein the nucleobase blocking group is stable to the conditions of the front end deprotection step with amines.
  • blocking groups for nucleobases are not required. In other embodiments, blocking groups for nucleobases are required. In yet other embodiments, certain nucleobases require blocking group while other nucleobases do not require blocking groups. In embodiments where the nucleobases are blocked, the blocking groups are either completely or partially removed under conditions appropriate to remove the blocking group at the front end.
  • R 1 can denote OR a , wherein R a is acyl, and Ba denotes guanine blocked with an acyl group including, but not limited to isobutyryl, acetyl or 4-(tert-butylphenoxy)acetyl. The acyl groups at R 1 and Ba will be removed or partially removed during the same deblocking step.
  • Oligonucleoside phosphorothioates have shown therapeutic potential (Stein et al., Science (1993), 261:1004-12; Agrawal et al., Antisence Res. and Dev. (1992), 2:261-66; Bayever et al., Antisense Res. and Dev. (1993), 3:383-390). Oligonucleoside phosphorothioates prepared without regard to the stereochemistry of the phosphorothioate exist as a mixture of 2 n diastereomers, where n is the number of internucleotide phosphorothioates linkages. The chemical and biological properties of these diastereomeric phosphorothioates can be distinct.
  • the nucleobase Ba utilized in the compounds and methods described herein is a natural nucleobase or a modified nucleobase derived from natural nucleobases.
  • examples include, but are not limited to, uracil, thymine, adenine, cytosine, and guanine having their respective amino groups protected by acyl protecting groups, 2-fluorouracil, 2-fluorocytosine, 5-bromouracil, 5-iodouracil, 2,6-diaminopurine, azacytosine, pyrimidine analogs such as pseudoisocytosine and pseudouracil and other modified nucleobases such as 8-substituted purines, xanthine, or hypoxanthine (the latter two being the natural degradation products).
  • R 8 is a linear or branched alkyl, aryl, aralkyl, or aryloxylalkyl group having 1 to 15 carbon atoms, including, by way of example only, a methyl, isopropyl, phenyl, benzyl, or phenoxymethyl group; and each of R 9 and R 10 represents a linear or branched alkyl group having 1 to 4 carbon atoms.
  • Modified nucleobases also include expanded-size nucleobases in which one or more benzene rings has been added. Nucleic base replacements described in the Glen Research catalog (www.glenresearch.com); Krueger A T et al, Acc. Chem. Res., 2007, 40, 141-150; Kool, E T, Acc. Chem. Res., 2002, 35, 936-943; Benner S. A., et al., Nat. Rev. Genet., 2005, 6, 553-543; Romesberg, F. E., et al., Curr. Opin. Chem. Biol., 2003, 7, 723-733; Hirao, I., Curr. Opin. Chem. Biol., 2006, 10, 622-627, are contemplated as useful for the synthesis of the nucleic acids described herein. Some examples of these expanded-size nucleobases are shown below:
  • modified nucleobases also encompass structures that are not considered nucleobases but are other moieties such as, but not limited to, corrin- or porphyrin-derived rings.
  • Porphyrin-derived base replacements have been described in Morales-Rojas, H and Kool, E T, Org. Lett., 2002, 4, 4377-4380. Shown below is an example of a porphyrin-derived ring which can be used as a base replacement:
  • modified nucleobases also include base replacements such as those shown below:
  • Modified nucleobases which are fluorescent are also contemplated.
  • Non-limiting examples of these base replacements include phenanthrene, pyrene, stillbene, isoxanthine, isozanthopterin, terphenyl, terthiophene, benzoterthiophene, coumarin, lumazine, tethered stillbene, benzo-uracil, and naphtho-uracil, as shown below:
  • modified nucleobases can be unsubstituted or contain further substitutions such as heteroatoms, alkyl groups, or linking moieties connected to fluorescent moieties, biotin or avidin moieties, or other protein or peptides.
  • Modified nucleobases also include certain ‘universal bases’ that are not nucleobases in the most classical sense, but function similarly to nucleobases.
  • One representative example of such a universal base is 3-nitropyrrole.
  • nucleosides can also be used in the process disclosed herein and include nucleosides that incorporate modified nucleobases, or nucleobases covalently bound to modified sugars.
  • nucleosides that incorporate modified nucleobases include 4-acetylcytidine; 5-(carboxyhydroxylmethyl)uridine; 2′-O-methylcytidine; 5-carboxymethylaminomethyl-2-thiouridine; 5-carboxymethylaminomethyluridine; dihydrouridine; 2′-O-methylpseudouridine; beta,D-galactosylqueosine; 2′-O-methylguanosine; N 6 -isopentenyladenosine; 1-methyladenosine; 1-methylpseudouridine; 1-methylguanosine; 1-methylinosine; 2,2-dimethylguanosine; 2-methyladenosine; 2-methylguanosine; N 7 -methylguanosine; 3-methyl-cytidine
  • nucleosides include 6′-modified bicyclic nucleoside analogs that have either (R) or (S)-chirality at the 6′-position and include the analogs described in U.S. Pat. No. 7,399,845.
  • nucleosides include 5′-modified bicyclic nucleoside analogs that have either (R) or (S)-chirality at the 5′-position and include the analogs described in US Patent Application Publication No. 20070287831.
  • the nucleobases or modified nucleobases comprises biomolecule binding moieties such as antibodies, antibody fragments, biotin, avidin, streptavidin, receptor ligands, or chelating moieties.
  • Ba is 5-bromouracil, 5-iodouracil, or 2,6-diaminopurine.
  • Ba is modified by substitution with a fluorescent or biomolecule binding moiety.
  • the substituent on Ba is a fluorescent moiety.
  • the substituent on Ba is biotin or avidin.
  • nucleotides The most common naturally occurring nucleotides are ribose sugars linked to the nucleobases adenosine (A), cytosine (C), guanine (G), and thymine (T) or uracil (U). Also contemplated are modified nucleotides wherein the phosphate group or the modified phosphorous atom moieties in the nucleotides can be linked to various positions of the sugar or modified sugar. As non-limiting examples, the phosphate group or the modified phosphorous-atom moiety can be linked to the 2′, 3′, 4′ or 5′ hydroxyl moiety of a sugar or modified sugar. Nucleotides that incorporate the modified nucleobases described above can also be used in the process disclosed herein. In some embodiments, nucleotides or modified nucleotides comprising an unprotected —OH moiety are used in the process disclosed herein.
  • modified sugars can also be incorporated in the nucleic acids disclosed herein.
  • the modified sugars contain one or more substituents at the 2′ position including one of the following: F; CF 3 , CN, N 3 , NO, NO 2 , O-, S-, or N-alkyl; O-, S-, or N-alkenyl; O-, S- or N-alkynyl; or O-alkyl-O-alkyl, O-alkyl-N-alkyl or N-alkyl-O-alkyl wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted C 1 -C 10 alkyl or C 2 -C 10 alkenyl and alkynyl.
  • substituents include, and are not limited to, O(CH 2 ) n OCH 3 , and O(CH 2 ) n NH 2 , wherein n is from 1 to about 10, MOE, DMAOE, DMAEOE. Also contemplated herein are modified sugars described in WO 2001/088198; and Martin et al., Helv. Chim. Acta, 1995, 78, 486-504.
  • modified sugars comprise substituted silyl groups, an RNA cleaving group, a reporter group, a fluorescent label, an intercalator, a group for improving the pharmacokinetic properties of a nucleic acid, or a group for improving the pharmacodynamic properties of a nucleic acid, and other substituents having similar properties.
  • the modifications may be made at the at the 2′, 3′, 4′, 5′, or 6′ positions of the sugar or modified sugar, including the 3′ position of the sugar on the 3′-terminal nucleotide or in the 5′ position of the 5′-terminal nucleotide.
  • Modified sugars also include sugar mimetics such as cyclobutyl or cyclopentyl moieties in place of the pentofuranosyl sugar.
  • Representative United States patents that teach the preparation of such modified sugar structures include, but are not limited to, U.S. Pat. Nos. 4,981,957; 5,118,800; 5,319,080; and 5,359,044.
  • Some modified sugars that are contemplated include:
  • modified sugars include glycerol, which form glycerol nucleic acid (GNA) analogues.
  • GNA glycerol nucleic acid
  • One example of a GNA analogue is shown below and is described in Zhang, R et al., J. Am. Chem. Soc., 2008, 130, 5846-5847; Zhang L, et al., J. Am. Chem. Soc., 2005, 127, 4174-4175 and Tsai C H et al., PNAS, 2007, 14598-14603:
  • modified sugars include hexopyranosyl (6′ to 4′), pentopyranosyl (4′ to 2′), pentopyranosyl (4′ to 3′), or tetrofuranosyl (3′ to 2′) sugars.
  • Hexopyranosyl (6′ to 4′) sugars contemplated include:
  • Pentopyranosyl (4′ to 2′) sugars contemplated include:
  • Pentopyranosyl (4′ to 3′) sugars contemplated include:
  • Tetrofuranosyl (3′ to 2′) sugars contemplated include:
  • X is selected from S, Se, CH 2 , N-Me, N-Et or N-iPr.
  • modified sugars and sugar mimetics can be prepared by methods known in the art, including, but not limited to: A. Eschenmoser, Science (1999), 284:2118; M. Bohringer et al, Helv. Chim. Acta (1992), 75:1416-1477; M. Egli et al, J. Am. Chem. Soc. (2006), 128(33):10847-56; A. Eschenmoser in Chemical Synthesis: Gnosis to Prognosis, C. Chatgilialoglu and V. Sniekus, Ed., (Kluwer Academic, Netherlands, 1996), p. 293; K.-U. Schoning et al, Science (2000), 290:1347-1351; A.
  • protective groups are used to block some or all reactive moieties and prevent such groups from participating in chemical reactions until the protective group is removed.
  • each protective group is removable by a different means.
  • Protective groups that are cleaved under totally disparate reaction conditions fulfill the requirement of differential removal.
  • protective groups are removed by acid, base, and/or hydrogenolysis.
  • Groups such as trityl, dimethoxytrityl, acetal and t-butyldimethylsilyl are acid labile and are used in certain embodiments to protect carboxy and hydroxy reactive moieties in the presence of amino groups protected with Cbz groups, which are removable by hydrogenolysis, and/or Fmoc groups, which are base labile.
  • carboxylic acid and hydroxy reactive moieties are blocked with base labile groups such as, but not limited to, methyl, ethyl, and acetyl in the presence of amines blocked with acid labile groups such as t-butylcarbamate or with carbamates that are both acid and base stable but hydrolytically removable.
  • hydroxy reactive moieties are blocked with hydrolytically removable protective groups such as the benzyl group, while amine groups capable of hydrogen bonding with acids are blocked with base labile groups such as Fmoc.
  • carboxylic acid reactive moieties are protected by conversion to simple ester compounds, or they are, in yet another embodiment, blocked with oxidatively-removable protective groups such as 2,4-dimethoxybenzyl, while co-existing amino groups are blocked with fluoride labile silyl or carbamate blocking groups.
  • Allyl blocking groups are useful in the presence of acid- and base-protecting groups since the former are stable and can be subsequently removed by metal or pi-acid catalysts.
  • an allyl-blocked hydroxy groups can be deprotected with a Pd(O)-catalyzed reaction in the presence of acid labile t-butylcarbamate or base-labile acetate amine protecting groups.
  • Yet another form of protecting group is a resin to which a compound or intermediate is attached. As long as the residue is attached to the resin, that functional group is blocked and cannot react. Once released from the resin, the functional group is available to react.
  • blocking/protecting groups useful in the synthesis of the compounds described herein are, by way of example only:
  • protecting groups useful to protect nucleotides during synthesis include base labile protecting groups and acid labile protecting groups.
  • Base labile protecting groups are used to protect the exocyclic amino groups of the heterocyclic nucleobases. This type of protection is generally achieved by acylation. Three commonly used acylating groups for this purpose are benzoyl chloride, phenoxyacetic anhydride, and isobutyryl chloride. These protecting groups are stable to the reaction conditions used during nucleic acid synthesis and are cleaved at approximately equal rates during the base treatment at the end of synthesis.
  • the 5′-protecting group is trityl, monomethoxy trityl, dimethoxytrityl, trimethoxytrityl, 2-chlorotrityl, DATE, TBTr, 9-phenylxanthine-9-yl (Pixyl), or 9-(p-methoxyphenyl)xanthine-9-yl (MOX).
  • thiol moieties are incorporated in the compounds described herein and are protected.
  • the protecting groups include, but are not limited to, pixyl, trityl, benzyl, p-methoxybenzyl (PMB), or tert-butyl (t-Bu).
  • Pivaloyl chloride (4.96 ml, 40.3 mmol) was added dropwise via syringe to an ice cold DCM solution (50 mL) of 2-(hydroxymethyl)phenol (5 g, 40.3 mmol) and triethylamine (5.61 ml, 40.3 mmol).
  • An ice-cold solution of the crude pivalate ester was treated with triethylamine (6.74 ml, 48.4 mmol) and 50 mL DCM.
  • Methanesulfonyl chloride (3.43 ml, 44.3 mmol) was then added slowly (5 min) via syringe and the resulting mixture was warmed to r.t. The mixture was poured onto ice and the organic layer was separated then washed with sat NaHCO 3 (aq), dried (MgSO 4 ), filtered and reduced to 10.5 g crude pale yellow oil.
  • Compound 23 is prepared according to a literature method (Journal of Medicinal Chemistry, 50(23), 5568-5570; 2007.)
  • Compound 30 is prepared according to a literature method (Tetrahedron, 42(2), 601-7; 1986.)
  • Compound 31 is prepared from compound 30 according to a patent procedure (US 20090181444)
  • Compound 33 is prepared from compound 32 according to a patent procedure (US 20090181444)
  • compound 100 is converted to compound 107 by a procedure analogous to that described for compound 101.
  • compound 100 is converted to compound 108 by a procedure analogous to that described for compound 101.
  • compound 100 is converted to compound 109 by a procedure analogous to that described for compound 101.
  • compound 100 is converted to compound 110 by a procedure analogous to that described for compound 101.
  • Compound 110 is converted to compound 210 by a procedure analogous to that described for compound 201.
  • compound 100 is converted to compound 111 by a procedure analogous to that described for compound 101.
  • Compound 111 is converted to compound 211 by a procedure analogous to that described for compound 201.
  • compound 100 is converted to compound 112 by a procedure analogous to that described for compound 101.
  • Compound 112 is converted to compound 212 by a procedure analogous to that described for compound 201.
  • compound 100 is converted to compound 113 by a procedure analogous to that described for compound 101.
  • compound 100 is converted to compound 114 by a procedure analogous to that described for compound 101.
  • compound 100 is converted to compound 115 by a procedure analogous to that described for compound 101.
  • Compound 100 is converted to compound 152 by a procedure analogous to that described for compound 101.
  • compound 152 is converted to compound 153 by a procedure analogous to that described for compound 151.
  • Compound 100 is converted to compound 154 by a procedure analogous to that described for compound 101.
  • Compound 303 is prepared by sulfurization of compound 300 on support followed by cleavage.
  • ACN 450 ⁇ L
  • BSTFA 50 ⁇ L
  • compound 12 (20 mg) are added to compound 300 (1 ⁇ mol) which is shaken for 18 h.
  • the CPG is collected by filtration resuspended in 20% PrNH 2 in dry CH 3 CN (2 mL) and shaken for 16 h at rt. Solvents were removed under reduced pressure and the residue is purified by RPHPLC to provide pure compound 303.

Abstract

Described herein are methods for the synthesis of derivatives of thiosulfonate reagents. Said reagents have utility for the synthesis of phosphorothiotriesters from H-phosphonates in a stereospecific fashion.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims priority to U.S. provisional application Ser. No. 61/509,526, filed Jul. 19, 2011, the entirety of which is hereby incorporated herein by reference.
  • BACKGROUND OF THE INVENTION
  • Oligonucleotides are useful in therapeutic, diagnostic, research and nanomaterials applications. The use of natural sequences of DNA or RNA for therapeutics is limited because of their instability against extra and intracellular nucleases, poor cell penetration and distribution. Additionally, in vitro studies have shown that the properties of antisense nucleotides such as binding affinity, sequence specific binding to the complementary RNA (Cosstick and Eckstcin, 1985; LaPlanche et al., 1986; Latimer et al., 1989; Hacia et al., 1994; Mesmaeker et al., 1995), stability to nucleases are affected by the configurations of the phosphorous atoms Therefore, there is a need for modified oligonucleotides to impart stability towards ubiquitous nucleases, increase binding affinity towards complementary RNA and increase cell penetration and bio-distribution for a number of in-vitro and in-vivo applications.
  • SUMMARY OF THE INVENTION
  • Described herein are methods for the synthesis of novel functionalized nucleic acids and nucleic acid prodrugs. In some embodiments, the nucleic acids comprise chiral phosphorous moieties.
  • One embodiment provides a process for the preparation of phosphorothiotriesters of structure IIIa comprising the steps of:
      • i) reacting an H-phosphonate of structure Ia with an silylating reagent to provide a silyloxyphosphonate; and
      • ii) reacting the silyloxyphosphonate with a thiosulfonate reagent of structure IIa to provide a phosphorothiotriester of structure IIIa;
      • wherein,
      • the H-phosphonate of structure Ia has the following structure:
  • Figure US20170029457A1-20170202-C00001
      • wherein,
      • W is independently selected from O, S, NH, or CH2;
      • R3 is —OH, —SH, —NRdRd, —N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —P(O)(Re)2, —HP(O)(Re), —ORa or —SRc;
      • Y1 is O, NRd, S, or Se;
      • Ra is a blocking group;
      • Rc is a blocking group;
      • each instance of Rd is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, —P(O)(Re)2, or —HP(O)(Re);
      • each instance of Re is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2—, alkenyl-Y2—, alkynyl-Y2—, aryl-Y2—, or heteroaryl-Y2—, or a cation which is Na+1, Li+1, or K+1;
      • Y2 is O, NRd, or S;
      • each instance of R4 is independently hydrogen, —OH, —SH, —NRdRd, —N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —ORb, or —SRc, and Rb is a blocking group;
      • each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;
      • R5 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid; and
      • n is between 1 and about 200; and
      • the thiosulfonate reagent of structure IIa has the following structure:
  • Figure US20170029457A1-20170202-C00002
  • wherein,
      • X is alkyl, cycloalkyl, or heteroaryl;
      • R is alkyl, alkenyl, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, or R1-R2;
      • R1 is selected from —S-alkenylene-, —S-alkylene-, —S-alkylene-aryl-alkylene-, —S—CO-aryl-alkylene-, or —S—CO-alkylene-aryl-alkylene-;
      • R2 is selected from heterocyclo-alkylene-S—, heterocyclo-alkenylene-S—, aminoalkyl-S—, or (alkyl)4N-alkylene-S—;
      • and the phosphorothiotriester of structure IIIa has the following structure:
  • Figure US20170029457A1-20170202-C00003
      • wherein,
      • W is independently selected from O, S, NH, or CH2;
      • R is alkyl, alkenyl, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, or R1-R2;
      • R1 is selected from —S-alkenylene-, —S-alkylene-, —S-alkylene-aryl-alkylene-, —S—CO-aryl-alkylene-, or —S—CO-alkylene-aryl-alkylene-;
      • R2 is selected from heterocyclo-alkylene-S—, heterocyclo-alkenylene-S—, aminoalkyl-S—, or (alkyl)4N-alkylene-S—;
      • R3 is —OH, —SH, —NRdRd, —N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —P(O)(Re)2, —HP(O)(Re), —ORa or —SRc;
      • Y1 is O, NRd, S, or Se;
      • Ra is a blocking group;
      • Rc is a blocking group;
      • each instance of Rd is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, —P(O)(Re)2, or —HP(O)(Re);
      • each instance of Re is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2—, alkenyl-Y2—, alkynyl-Y2—, aryl-Y2—, or heteroaryl-Y2—, or a cation which is Na+1, Li+1, or K+1;
      • Y2 is O, NRd, or S;
      • each instance of R4 is independently hydrogen, —OH, —SH, —NRdRd, —N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —ORb, or —SRc, and Rb is a blocking group;
      • each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;
      • R5 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid; and
      • n is between 1 and about 200.
  • Another embodiment provides a process for the preparation of phosphorothiotriesters of structure IIa, wherein W is O.
  • Another embodiment provides a process for the preparation of phosphorothiotriesters of structure IIIa, wherein R1 is selected from:
  • Figure US20170029457A1-20170202-C00004
      • R2 is selected from:
  • Figure US20170029457A1-20170202-C00005
  • Another embodiment provides a process for the preparation of phosphorothiotriesters of structure IIIa, wherein the silylating reagent is selected from
      • 1,1,3,3-tetramethyl-1,3-diphenyldisilazane;
      • 1,3-dimethyl-1,1,3,3-tetraphenyldisilazane;
      • 1-(trimethylsilyl)imidazole;
      • N-trimethylsilyl-N-methyl trifluoroacetamide;
      • bis(dimethylamino)dimethylsilane;
      • bromotrimethylsilane;
      • chlorodimethyl(pentafluorophenyl)silane;
      • chlorotriethylsilane;
      • chlorotriisopropylsilane;
      • chlorotrimethylsilane;
      • dichlorodimethylsilane;
      • hexamethyldisilazane;
      • N,N′-bis(trimethylsilyl)urea;
      • N,N-bis(trimethylsilyl)methylamine;
      • N,N-dimethyltrimethylsilylamine;
      • N,O-bis(trimethylsilyl)acetamide;
      • N,O-bis(trimethylsilyl)carbamate;
      • N,O-bis(trimethylsilyl)trifluoroacetamide;
      • N-methyl-N-(trimethylsilyl)trifluoroacetamide;
      • N-methyl-N-trimethylsilylacetamide;
      • N-methyl-N-trimethylsilylheptafluorobutyramide;
      • N-tert-butyldimethylsilyl-N-methyltrifluoroacetamide;
      • N-methyl-N-trimethylsilylheptafluorobutyramide;
      • trimethylsilyltriflate;
      • triethylsilyltriflate;
      • triisopropylsilyltriflate; or
      • tert-butyldimethylsilyltriflate.
  • Another embodiment provides the process, wherein the silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide, trimethylsilyltriflate, chlorotrimethylsilane, or 1-(trimethylsilyl)imidazole.
  • Another embodiment provides the process, wherein the silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide.
  • Another embodiment provides the process, wherein the H-phosphonate is covalently linked to a solid phase.
  • One embodiment provides a process for the preparation of phosphorothiotriesters comprising non-stereorandom phosphorous linkages of structure IIIb comprising the steps of:
      • i) reacting a H-phosphonate comprising non-stereorandom phosphorous linkages of structure Ib with an silylating reagent to provide a silyloxyphosphonate; and
      • ii) reacting the silyloxyphosphonate with a thiosulfonate reagent of structure IIb to provide a phosphorothiotriester comprising non-stereorandom phosphorous linkages of structure IIIb;
      • wherein,
      • the H-phosphonate comprising non-stereorandom phosphorous linkages of structure Ib has the following structure:
  • Figure US20170029457A1-20170202-C00006
      • wherein,
      • W is independently selected from O, NH, or CH2;
      • R3 is —OH, —SH, —NRdRd, —N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —P(O)(Re)2, —HP(O)(Re), —ORa or —SRc;
      • Y1 is O, NRd, S, or Se;
      • Ra is a blocking group;
      • Rc is a blocking group;
      • each instance of Rd is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, —P(O)(Re)2, or —HP(O)(Re);
      • each instance of Re is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2—, alkenyl-Y2—, alkynyl-Y2—, aryl-Y2—, or heteroaryl-Y2—, or a cation which is Na+1, Li+1, or K+1;
      • Y2 is O, NRd, or S;
      • each instance of R4 is independently hydrogen, —OH, —SH, —NRdRd, —N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —ORb, or —SRe, and Rb is a blocking group;
      • each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;
      • R5 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid; and
      • n is between 1 and about 200; and
      • the thiosulfonate reagent of structure IIb has the following structure:
  • Figure US20170029457A1-20170202-C00007
  • wherein,
      • X is alkyl, cycloalkyl, aryl, or heteroaryl;
      • R is alkyl, alkenyl, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, or R1-R2;
      • R1 is selected from —S-alkenylene-, —S-alkylene-, —S-alkylene-aryl-alkylene-, —S—CO-aryl-alkylene-, or —S—CO-alkylene-aryl-alkylene-;
      • R2 is selected from heterocyclo-alkylene-S—, heterocyclo-alkenylene-S—, aminoalkyl-S—, or (alkyl)4N-alkylene-S—;
      • and the chiral phosphorothiotriester comprising non-stereorandom phosphorous linkages of structure IIIb has the following structure:
  • Figure US20170029457A1-20170202-C00008
      • wherein,
      • W is independently selected from O, NH, or CH2;
      • R is alkyl, alkenyl, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, or R1-R2;
      • R1 is selected from —S-alkenylene-, —S-alkylene-, —S-alkylene-aryl-alkylene-, —S—CO-aryl-alkylene-, or —S—CO-alkylene-aryl-alkylene-;
      • R2 is selected from heterocyclo-alkylene-S—, heterocyclo-alkenylene-S—, aminoalkyl-S—, or (alkyl)4N-alkylene-S—;
      • R3 is —OH, —SH, —NRdRd, —N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —P(O)(Re)2, —HP(O)(Re), —ORa or —SRc;
      • Y1 is O, NRd, S, or Se;
      • Ra is a blocking group;
      • Rc is a blocking group;
      • each instance of Rd is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, —P(O)(Re)2, or —HP(O)(Re);
      • each instance of Rc is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2—, alkenyl-Y2—, alkynyl-Y2—, aryl-Y2—, or heteroaryl-Y2—, or a cation which is Na+1, Li+1, or K+1;
      • Y2 is O, NRd, or S;
      • each instance of R4 is independently hydrogen, —OH, —SH, —NRdRd, —N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —ORb, or —SRc, and Rb is a blocking group;
      • each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;
      • R5 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid; and
      • n is between 1 and about 200.
  • Another embodiment provides a process for the preparation of phosphorothiotriesters comprising non-stereorandom phosphorous linkages of structure IIIb, wherein W is O.
  • Another embodiment provides a process for the preparation of phosphorothiotriesters comprising non-stereorandom phosphorous linkages of structure IIIb, wherein R1 is selected from:
  • Figure US20170029457A1-20170202-C00009
  • and
      • R2 is selected from:
  • Figure US20170029457A1-20170202-C00010
  • Another embodiment provides a process for the preparation of phosphorothiotriesters comprising non-stereorandom phosphorous linkages of structure IIIb, wherein the silylating reagent is selected from
      • 1,1,3,3-tetramethyl-1,3-diphenyldisilazane;
      • 1,3-dimethyl-1,1,3,3-tetraphenyldisilazane;
      • 1-(trimethylsilyl)imidazole;
      • N-trimethylsilyl-N-methyl trifluoroacetamide;
      • bis(dimethylamino)dimethylsilane;
      • bromotrimethylsilane;
      • chlorodimethyl(pentafluorophenyl)silane;
      • chlorotriethylsilane;
      • chlorotriisopropylsilane;
      • chlorotrimethylsilane;
      • dichlorodimethylsilane;
      • hexamethyldisilazane;
      • N,N′-bis(trimethylsilyl)urea;
      • N,N-bis(trimethylsilyl)methylamine;
      • N,N-dimethyltrimethylsilylamine;
      • N,O-bis(trimethylsilyl)acetamide;
      • N,O-bis(trimethylsilyl)carbamate;
      • N,O-bis(trimethylsilyl)trifluoroacetamide;
      • N-methyl-N-(trimethylsilyl)trifluoroacetamide;
      • N-methyl-N-trimethylsilylacetamide;
      • N-methyl-N-trimethylsilylheptafluorobutyramide;
      • N-tert-butyldimethylsilyl-N-methyltrifluoroacetamide;
      • N-methyl-N-trimethylsilylheptafluorobutyramide;
      • trimethylsilyltriflate;
      • triethylsilyltriflate;
      • triisopropylsilyltriflate; or
      • tert-butyldimethylsilyltriflate.
  • Another embodiment provides the process, wherein the silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide, trimethylsilyltriflate, chlorotrimethylsilane, or 1-(trimethylsilyl)imidazole.
  • Another embodiment provides the process, wherein the silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide.
  • Another embodiment provides the process, wherein the H-phosphonate is covalently linked to a solid phase.
  • One embodiment provides a process for the preparation of phosphorothiotriesters of structure IIIc comprising the steps of:
      • i) reacting a H-phosphonate of structure Ic with an silylating reagent to provide a silyloxyphosphonate;
      • ii) reacting the silyloxyphosphonate with a bis(thiosulfonate) reagent of structure IVc to provide a phosphorothiotriester comprising a thiosulfonate group of structure Vc;
      • iii) reacting the phosphorothiotriester comprising a thiosulfonate group of structure Vc with a nucleophile of structure VIc to provide the phosphorothiotriesters of structure IIIc;
      • wherein,
      • the H-phosphonate of structure Ic has the following structure:
  • Figure US20170029457A1-20170202-C00011
      • wherein,
      • W is independently selected from O, S, NH, or CH2;
      • R3 is —OH, —SH, —NRdRd, —N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, P(O)(Re)2, —HP(O)(Re), —ORa or —SRc;
      • Y1 is O, NRd, S, or Se;
      • Ra is a blocking group;
      • Rc is a blocking group;
      • each instance of Rd is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, —P(O)(Re)2, or —HP(O)(Re);
      • each instance of Re is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2—, alkenyl-Y2—, alkynyl-Y2—, aryl-Y2—, or heteroaryl-Y2—, or a cation which is Na+1, Li+1, or K+1;
      • Y2 is O, NRd, or S;
      • each instance of R4 is independently hydrogen, —OH, —SH, —NRdRd, —N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —ORb, or —SRc, and Rb is a blocking group;
      • each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;
      • R5 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid; and
      • n is between 1 and about 200; and
      • the bis(thiosulfonate) reagent of structure IVc has the following structure:
  • Figure US20170029457A1-20170202-C00012
  • wherein,
      • X is alkylene, alkenylene, arylene, or heteroarylene;
      • each R6 is independently alkyl, cycloalkyl, aryl, or heteroaryl;
      • the nucleophile of structure VIc has the following structure:
      • R7—SH, wherein R7 is selected from alkyl, alkenyl, aryl, heterocyclo, aminoalkyl, or (heterocyclo)alkyl;
      • and phosphorothiotriesters of structure IIIc has the following structure:
  • Figure US20170029457A1-20170202-C00013
      • wherein,
      • W is independently selected from O, S, NH, or CH2;
      • R is R7—S—S—X—
      • R7 is alkyl, alkenyl, aryl, heterocyclo, aminoalkyl, or (heterocyclo)alkyl;
      • X is alkylene, alkenylene, arylene, or heteroarylene;
      • R3 is —OH, —SH, —NRdRd, —N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —P(O)(Re)2, —HP(O)(Re), —ORa or —SRc;
      • Y1 is O, NRd, S, or Se;
      • Ra is a blocking group;
      • Rc is a blocking group;
      • each instance of Rd is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, —P(O)(Re)2, or —HP(O)(Re);
      • each instance of Re is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2—, alkenyl-Y2—, alkynyl-Y2—, aryl-Y2—, or heteroaryl-Y2—, or a cation which is Na+1, L+1, or K+1; Y2 is O, NRd, or S;
      • each instance of R4 is independently hydrogen, —OH, —SH, —NRdRd, —N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —ORb, or —SRc, and Rb is a blocking group;
      • each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;
      • R5 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid;
      • n is between 1 and about 200; and
      • wherein the phosphorous linkages of the H-phosphonate of structure Ic, the phosphorothiotriester comprising a thiosulfonate group of structure Vc, and the phosphorothiotriesters of structure IIIc may optionally comprise non-stereorandom phosphorous linkages.
  • Another embodiment provides the process wherein the phosphorothiotriesters of structure IIIb comprise non-stereorandom phosphorous linkages and the H-phosphonate of structure Ic comprise non-stereorandom phosphorous linkages; and W is independently selected from O, NH, or CH2. Another embodiment provides the process wherein W is O.
  • Another embodiment provides the process wherein R6 is methyl.
  • Another embodiment provides the process wherein bis(thiosulfonate) reagent of structure IVc is selected from:
  • Figure US20170029457A1-20170202-C00014
  • Another embodiment provides the process wherein the nucleophile of structure VIc has the following structure:
  • Figure US20170029457A1-20170202-C00015
  • Another embodiment provides a process for the preparation of phosphorothiotriesters of structure IIIa, wherein the silylating reagent is selected from
      • 1,1,3,3-tetramethyl-1,3-diphenyldisilazane;
      • 1,3-dimethyl-1,1,3,3-tetraphenyldisilazane;
      • 1-(trimethylsilyl)imidazole;
      • N-trimethylsilyl-N-methyl trifluoroacetamide;
      • bis(dimethylamino)dimethylsilane;
      • bromotrimethylsilane;
      • chlorodimethyl(pentafluorophenyl)silane;
      • chlorotriethylsilane;
      • chlorotriisopropylsilane;
      • chlorotrimethylsilane;
      • dichlorodimethylsilane;
      • hexamethyldisilazane;
      • N,N′-bis(trimethylsilyl)urea;
      • N,N-bis(trimethylsilyl)methylamine;
      • N,N-dimethyltrimethylsilylamine;
      • N,O-bis(trimethylsilyl)acetamide;
      • N,O-bis(trimethylsilyl)carbamate;
      • N,O-bis(trimethylsilyl)trifluoroacetamide;
      • N-methyl-N-(trimethylsilyl)trifluoroacetamide;
      • N-methyl-N-trimethylsilylacetamide;
      • N-methyl-N-trimethylsilylheptafluorobutyramide;
      • N-tert-butyldimethylsilyl-N-methyltrifluoroacetamide;
      • N-methyl-N-trimethylsilylheptafluorobutyramide;
      • trimethylsilyltriflate;
      • triethylsilyltriflate;
      • triisopropylsilyltriflate; or
      • tert-butyldimethylsilyltriflate.
  • Another embodiment provides the process, wherein the silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide, trimethylsilyltriflate, chlorotrimethylsilane, or 1-(trimethylsilyl)imidazole.
  • Another embodiment provides the process, wherein the silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide.
  • Another embodiment provides the process, wherein the H-phosphonate is covalently linked to a solid phase.
  • INCORPORATION BY REFERENCE
  • All publications and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The novel features of the invention are set forth with particularity in the appended claims. A better understanding of the features and advantages of the present invention will be obtained by reference to the following detailed description that sets forth illustrative embodiments, in which the principles of the invention are utilized, and the accompanying drawings of which:
  • FIG. 1 provides the 31P NMR spectrum of Compound 100S in CD3CN as described in Example 6;
  • FIG. 2 provides the 31P NMR spectrum of Compound 100S in CD3CN after adding BSTFA as described in Example 6;
  • FIG. 3 provides the 31P NMR spectrum of Compound 100S in CD3CN after adding BSTFA, TEA and MTS as described in Example 6;
  • FIG. 4 provides the 31P NMR spectrum of Compound 100R in CD3CN as described in Example 6;
  • FIG. 5 provides the 31P NMR spectrum of Compound 100R in CD3CN as described in Example 6; and
  • FIG. 6 provides the 31P NMR spectrum of Compound 100R in CD3CN after adding BSTFA, TEA and MTS as described in Example 6.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Unless otherwise stated, the following terms used in this application, including the specification and claims, have the definitions given below. It must be noted that, as used in the specification and the appended claims, the singular forms “a” “an” and “the” include plural referents unless the context clearly dictates otherwise. Unless otherwise indicated, conventional methods of mass spectroscopy, NMR, HPLC, protein chemistry, biochemistry, recombinant DNA techniques and pharmacology are employed. In this application, the use of “or” or “and” means “and/or” unless stated otherwise. Furthermore, use of the term “including” as well as other forms, such as “include”, “includes” and “included” is not limiting.
  • Certain Chemical Terminology
  • Unless otherwise noted, the use of general chemical terms, such as though not limited to “alkyl,” “amine,” “aryl,” are unsubstituted.
  • As used herein, C1-Cx includes C1-C2, C1-C3 . . . C1-Cx. By way of example only, a group designated as “C1-C4” indicates that there are one to four carbon atoms in the moiety, i.e. groups containing 1 carbon atom, 2 carbon atoms, 3 carbon atoms or 4 carbon atoms, as well as the ranges C1-C2 and C1-C3. Thus, by way of example only, “C1-C4 alkyl” indicates that there are one to four carbon atoms in the alkyl group, i.e., the alkyl group is selected from among methyl, ethyl, propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, and t-butyl. Whenever it appears herein, a numerical range such as “1 to 10” refers to each integer in the given range; e.g., “1 to 10 carbon atoms” means that the group may have 1 carbon atom, 2 carbon atoms, 3 carbon atoms, 4 carbon atoms, 5 carbon atoms, 6 carbon atoms, 7 carbon atoms, 8 carbon atoms, 9 carbon atoms, or 10 carbon atoms.
  • The terms “heteroatom” or “hetero” as used herein, alone or in combination, refer to an atom other than carbon or hydrogen. Heteroatoms are may be independently selected from among oxygen, nitrogen, sulfur, phosphorous, silicon, selenium and tin but are not limited to these atoms. In embodiments in which two or more heteroatoms are present, the two or more heteroatoms can be the same as each another, or some or all of the two or more heteroatoms can each be different from the others.
  • The term “alkyl” as used herein, alone or in combination, refers to a straight-chain or branched-chain saturated hydrocarbon monoradical having from one to about ten carbon atoms, or one to six carbon atoms. Examples include, but are not limited to methyl, ethyl, n-propyl, isopropyl, 2-methyl-1-propyl, 2-methyl-2-propyl, 2-methyl-1-butyl, 3-methyl-1-butyl, 2-methyl-3-butyl, 2,2-dimethyl-1-propyl, 2-methyl-1-pentyl, 3-methyl-1-pentyl, 4-methyl-1-pentyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 2,2-dimethyl-1-butyl, 3,3-dimethyl-1-butyl, 2-ethyl-1-butyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, isopentyl, neopentyl, tert-amyl and hexyl, and longer alkyl groups, such as heptyl, octyl and the like. Whenever it appears herein, a numerical range such as “C1-C6 alkyl” or “C1-6 alkyl”, means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, 4 carbon atoms, 5 carbon atoms or 6 carbon atoms. In one embodiment, the “alkyl” is substituted. Unless otherwise indicated, the “alkyl” is unsubstituted.
  • The term “alkenyl” as used herein, alone or in combination, refers to a straight-chain or branched-chain hydrocarbon monoradical having one or more carbon-carbon double-bonds and having from two to about ten carbon atoms, or two to about six carbon atoms. The group may be in either the cis or trans conformation about the double bond(s), and should be understood to include both isomers. Examples include, but are not limited to ethenyl (—CH═CH2), 1-propenyl (—CH2CH═CH2), isopropenyl [—C(CH3)═CH2], butenyl, 1,3-butadienyl and the like. Whenever it appears herein, a numerical range such as “C2-C6 alkenyl” or “C2-6 alkenyl”, means that the alkenyl group may consist of 2 carbon atoms, 3 carbon atoms, 4 carbon atoms, 5 carbon atoms or 6 carbon atoms. In one embodiment, the “alkenyl” is substituted. Unless otherwise indicated, the “alkenyl” is unsubstituted.
  • The term “alkynyl” as used herein, alone or in combination, refers to a straight-chain or branched-chain hydrocarbon monoradical having one or more carbon-carbon triple-bonds and having from two to about ten carbon atoms, or from two to about six carbon atoms. Examples include, but are not limited to ethynyl, 2-propynyl, 2-butynyl, 1,3-butadiynyl and the like. Whenever it appears herein, a numerical range such as “C2-C6 alkynyl” or “C2-6 alkynyl”, means that the alkynyl group may consist of 2 carbon atoms, 3 carbon atoms, 4 carbon atoms, 5 carbon atoms or 6 carbon atoms. In one embodiment, the “alkynyl” is substituted. Unless otherwise indicated, the “alkynyl” is unsubstituted.
  • The terms “heteroalkyl”, “heteroalkenyl” and “heteroalkynyl” as used herein, alone or in combination, refer to alkyl, alkenyl and alkynyl structures respectively, as described above, in which one or more of the skeletal chain carbon atoms (and any associated hydrogen atoms, as appropriate) are each independently replaced with a heteroatom (i.e. an atom other than carbon, such as though not limited to oxygen, nitrogen, sulfur, silicon, phosphorous, tin or combinations thereof), or heteroatomic group such as though not limited to —O—O—, —S—S—, —O—S—, —S—O—, ═N—N═, —N═N—, —N═N—NH—, —P(O)2—, —O—P(O)2—, —P(O)2—O—, —S(O)—, —S(O)2—, —SnH2— and the like.
  • The terms “haloalkyl”, “haloalkenyl” and “haloalkynyl” as used herein, alone or in combination, refer to alkyl, alkenyl and alkynyl groups respectively, as defined above, in which one or more hydrogen atoms is replaced by fluorine, chlorine, bromine or iodine atoms, or combinations thereof. In some embodiments two or more hydrogen atoms may be replaced with halogen atoms that are the same as each another (e.g. difluoromethyl); in other embodiments two or more hydrogen atoms may be replaced with halogen atoms that are not all the same as each other (e.g. 1-chloro-1-fluoro-1-iodoethyl). Non-limiting examples of haloalkyl groups are fluoromethyl, chloromethyl and bromoethyl. A non-limiting example of a haloalkenyl group is bromoethenyl. A non-limiting example of a haloalkynyl group is chloroethynyl.
  • The term “carbon chain” as used herein, alone or in combination, refers to any alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl or heteroalkynyl group, which is linear, cyclic, or any combination thereof. If the chain is part of a linker and that linker comprises one or more rings as part of the core backbone, for purposes of calculating chain length, the “chain” only includes those carbon atoms that compose the bottom or top of a given ring and not both, and where the top and bottom of the ring(s) are not equivalent in length, the shorter distance shall be used in determining the chain length. If the chain contains heteroatoms as part of the backbone, those atoms are not calculated as part of the carbon chain length.
  • The term “cycloalkyl” as used herein, alone or in combination, refers to a saturated, hydrocarbon monoradical ring, containing from three to about fifteen ring carbon atoms or from three to about ten ring carbon atoms, though may include additional, non-ring carbon atoms as substituents (e.g. methylcyclopropyl). Whenever it appears herein, a numerical range such as “C3-C6 cycloalkyl” or “C3-6 cycloalkyl”, means that the cycloalkyl group may consist of 3 carbon atoms, 4 carbon atoms, 5 carbon atoms or 6 carbon atoms, i.e., is cyclopropyl, cyclobutyl, cyclopentyl or cyclohepty, although the present definition also covers the occurrence of the term “cycloalkyl” where no numerical range is designated. The term includes fused, non-fused, bridged and spiro radicals. A fused cycloalkyl may contain from two to four fused rings where the ring of attachment is a cycloalkyl ring, and the other individual rings may be alicyclic, heterocyclic, aromatic, heteroaromatic or any combination thereof. Examples include, but are not limited to cyclopropyl, cyclopentyl, cyclohexyl, decalinyl, and bicyclo [2.2.1] heptyl and adamantyl ring systems. Illustrative examples include, but are not limited to the following moieties:
  • Figure US20170029457A1-20170202-C00016
  • and the like.
  • In one embodiment, the “cycloalkyl” is substituted. Unless otherwise indicated, the “cycloalkyl” is unsubstituted.
  • The terms “non-aromatic heterocyclyl” and “heteroalicyclyl” as used herein, alone or in combination, refer to a saturated, partially unsaturated, or fully unsaturated nonaromatic ring monoradicals containing from three to about twenty ring atoms, where one or more of the ring atoms are an atom other than carbon, independently selected from among oxygen, nitrogen, sulfur, phosphorous, silicon, selenium and tin but are not limited to these atoms. In embodiments in which two or more heteroatoms are present in the ring, the two or more heteroatoms can be the same as each another, or some or all of the two or more heteroatoms can each be different from the others. The terms include fused, non-fused, bridged and spiro radicals. A fused non-aromatic heterocyclic radical may contain from two to four fused rings where the attaching ring is a non-aromatic heterocycle, and the other individual rings may be alicyclic, heterocyclic, aromatic, heteroaromatic or any combination thereof. Fused ring systems may be fused across a single bond or a double bond, as well as across bonds that are carbon-carbon, carbon-hetero atom or hetero atom-hetero atom. The terms also include radicals having from three to about twelve skeletal ring atoms, as well as those having from three to about ten skeletal ring atoms. Attachment of a non-aromatic heterocyclic subunit to its parent molecule can be via a heteroatom or a carbon atom. Likewise, additional substitution can be via a heteroatom or a carbon atom. As a non-limiting example, an imidazolidine non-aromatic heterocycle may be attached to a parent molecule via either of its N atoms (imidazolidin-1-yl or imidazolidin-3-yl) or any of its carbon atoms (imidazolidin-2-yl, imidazolidin-4-yl or imidazolidin-5-yl). In certain embodiments, non-aromatic heterocycles contain one or more carbonyl or thiocarbonyl groups such as, for example, oxo- and thio-containing groups. Examples include, but are not limited to pyrrolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidino, morpholino, thiomorpholino, thioxanyl, piperazinyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, 1,2,3,6-tetrahydropyridinyl, 2-pyrrolinyl, 3-pyrrolinyl, indolinyl, 2H-pyranyl, 4H-pyranyl, dioxanyl, 1,3-dioxolanyl, pyrazolinyl, dithianyl, dithiolanyl, dihydropyranyl, dihydrothienyl, dihydrofuranyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, 3-azabicyclo[3.1.0]hexanyl, 3-azabicyclo[4.1.0]heptanyl, 3H-indolyl and quinolizinyl. Illustrative examples of heterocycloalkyl groups, also referred to as non-aromatic heterocycles, include:
  • Figure US20170029457A1-20170202-C00017
  • and the like.
  • The terms also include all ring forms of the carbohydrates, including but not limited to the monosaccharides, the disaccharides and the oligosaccharides. In one embodiment, the “non-aromatic heterocyclyl” or “heteroalicyclyl” is substituted. Unless otherwise indicated, the “non-aromatic heterocyclyl” or “heteroalicyclyl” is unsubstituted.
  • The term “aryl” as used herein, alone or in combination, refers to an aromatic hydrocarbon radical of six to about twenty ring carbon atoms, and includes fused and non-fused aryl rings. A fused aryl ring radical contains from two to four fused rings where the ring of attachment is an aryl ring, and the other individual rings may be alicyclic, heterocyclic, aromatic, heteroaromatic or any combination thereof. Further, the term aryl includes fused and non-fused rings containing from six to about twelve ring carbon atoms, as well as those containing from six to about ten ring carbon atoms. A non-limiting example of a single ring aryl group includes phenyl; a fused ring aryl group includes naphthyl, phenanthrenyl, anthracenyl, azulenyl; and a non-fused bi-aryl group includes biphenyl. In one embodiment, the “aryl” is substituted. Unless otherwise indicated, the “aryl” is unsubstituted.
  • The term “heteroaryl” as used herein, alone or in combination, refers to an aromatic monoradicals containing from about five to about twenty skeletal ring atoms, where one or more of the ring atoms is a heteroatom independently selected from among oxygen, nitrogen, sulfur, phosphorous, silicon, selenium and tin but not limited to these atoms and with the proviso that the ring of said group does not contain two adjacent O or S atoms. In embodiments in which two or more heteroatoms are present in the ring, the two or more heteroatoms can be the same as each another, or some or all of the two or more heteroatoms can each be different from the others. The term heteroaryl includes fused and non-fused heteroaryl radicals having at least one heteroatom. The term heteroaryl also includes fused and non-fused heteroaryls having from five to about twelve skeletal ring atoms, as well as those having from five to about ten skeletal ring atoms. Bonding to a heteroaryl group can be via a carbon atom or a heteroatom. Thus, as a non-limiting example, an imidazole group may be attached to a parent molecule via any of its carbon atoms (imidazol-2-yl, imidazol-4-yl or imidazol-5-yl), or its nitrogen atoms (imidazol-1-yl or imidazol-3-yl). Likewise, a heteroaryl group may be further substituted via any or all of its carbon atoms, and/or any or all of its heteroatoms. A fused heteroaryl radical may contain from two to four fused rings where the ring of attachment is a heteroaromatic ring and the other individual rings may be alicyclic, heterocyclic, aromatic, heteroaromatic or any combination thereof. A non-limiting example of a single ring heteroaryl group includes pyridyl; fused ring heteroaryl groups include benzimidazolyl, quinolinyl, acridinyl; and a non-fused bi-heteroaryl group includes bipyridinyl. Further examples of heteroaryls include, without limitation, furanyl, thienyl, oxazolyl, acridinyl, phenazinyl, benzimidazolyl, benzofuranyl, benzoxazolyl, benzothiazolyl, benzothiadiazolyl, benzothiophenyl, benzoxadiazolyl, benzotriazolyl, imidazolyl, indolyl, isoxazolyl, isoquinolinyl, indolizinyl, isothiazolyl, isoindolyloxadiazolyl, indazolyl, pyridyl, pyridazyl, pyrimidyl, pyrazinyl, pyrrolyl, pyrazinyl, pyrazolyl, purinyl, phthalazinyl, pteridinyl, quinolinyl, quinazolinyl, quinoxalinyl, triazolyl, tetrazolyl, thiazolyl, triazinyl, thiadiazolyl and the like, and their oxides, such as for example pyridyl-N-oxide. Illustrative examples of heteroaryl groups include the following moieties:
  • Figure US20170029457A1-20170202-C00018
  • and the like.
  • In one embodiment, the “heteroaryl” is substituted. Unless otherwise indicated, the “heteroaryl” is unsubstituted.
  • The term “heterocyclyl” as used herein, alone or in combination, refers collectively to heteroalicyclyl and heteroaryl groups. Herein, whenever the number of carbon atoms in a heterocycle is indicated (e.g., C1-C6 heterocycle), at least one non-carbon atom (the heteroatom) must be present in the ring. Designations such as “C1-C6 heterocycle” refer only to the number of carbon atoms in the ring and do not refer to the total number of atoms in the ring. Designations such as “4-6 membered heterocycle” refer to the total number of atoms that are contained in the ring (i.e., a four, five, or six membered ring, in which at least one atom is a carbon atom, at least one atom is a heteroatom and the remaining two to four atoms are either carbon atoms or heteroatoms). For heterocycles having two or more heteroatoms, those two or more heteroatoms can be the same or different from one another. Non-aromatic heterocyclic groups include groups having only three atoms in the ring, while aromatic heterocyclic groups must have at least five atoms in the ring. Bonding (i.e. attachment to a parent molecule or further substitution) to a heterocycle can be via a heteroatom or a carbon atom. In one embodiment, the “heterocyclyl” is substituted. Unless otherwise indicated, the “heterocycyl” is unsubstituted.
  • The terms “halogen”, “halo” or “halide” as used herein, alone or in combination refer to fluoro, chloro, bromo and/or iodo.
  • The compounds, or their pharmaceutically acceptable salts may contain one or more asymmetric centers and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, such as (R)- or (S)-. When the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both Z and E geometric isomers (e.g., cis or trans). Likewise, all possible isomers, as well as their racemic and optically pure forms, and all tautomeric forms are also intended to be included.
  • A “stereoisomer” refers to the relationship between two or more compounds made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not superimposable. The term “enantiomer” refers to two stereoisomers that are nonsuperimposeable mirror images of one another. It is contemplated that the various stereoisomers of the compounds disclosed herein, and mixtures thereof, are within the scope of the present disclosure and specifically includes enantiomers.
  • A “tautomer” refers to a compound wherein a proton shift from one atom of a molecule to another atom of the same molecule is possible. The compounds presented herein may exist as tautomers. In solutions where tautomerization is possible, a chemical equilibrium of the tautomers will exist. The exact ratio of the tautomers depends on several factors, including temperature, solvent, and pH. Some examples of tautomeric equilibrium are shown below.
  • Figure US20170029457A1-20170202-C00019
  • The term “non-stereorandom phosphorous linkage(s)” as used herein refers to a chiral phosphorous atom in the phosphodiester, or other isosteric linkage type, internucleotide linkage. For embodiments comprising more than one phosphorous internucleotide linkage, the handedness of chirality at phosphorous is independently selected at each phosphorous atom. In one embodiment, the oligonucleotide described herein is a pure diastereomer. In another embodiment, the oligonucleotide is greater that 95% diastereomeric purity. In another embodiment, the oligonucleotide is greater that 90% diastereomeric purity.
  • “Optional” or “optionally” means that a subsequently described event or circumstance may or may not occur and that the description includes instances when the event or circumstance occurs and instances in which it does not. For example, “optionally substituted alkyl” means that the alkyl radical may or may not be substituted and that the description includes both substituted aryl radicals and aryl radicals having no substitution.
  • Certain Nucleic Acid Terminology
  • Natural nucleic acids have a phosphate backbone; artificial nucleic acids may contain other types of backbones, but contain the same bases.
  • The term “nucleotide” as used herein refers to a monomeric unit of a polynucleotide that consists of a heterocyclic base, a sugar, and one or more phosphate groups. The naturally occurring bases, (guanine, (G), adenine (A), cytosine (C), thymine (T), and uracil (U)) are derivatives of purine or pyrimidine, though it should be understood that naturally and non-naturally occurring base analogs are also included. The naturally occurring sugar is the pentose (five-carbon sugar) deoxyribose (which forms DNA) or ribose (which forms RNA), though it should be understood that naturally and non-naturally occurring sugar analogs are also included. Nucleic acids are linked via phosphate bonds to form nucleic acids, or polynucleotides, though many other linkages are known in the art (such as, though not limited to phosphorothioates, boranophosphates and the like). Artificial nucleic acids include PNAs (peptide nucleic acids), phosphothionates, and other variants of the phosphate backbone of native nucleic acids.
  • The term “nucleoside” refers to a moiety wherein a nucleobase or a modified nucleobase is covalently bound to a sugar or modified sugar.
  • The term “sugar” refers to a monosaccharide in closed and/or open form. Sugars include, but are not limited to, ribose, deoxyribose, pentofuranose, pentopyranose, and hexopyranose moieties.
  • The term “modified sugar” refers to a moiety that can replace a sugar. The modified sugar mimics the spatial arrangement, electronic properties, or some other physicochemical property of a sugar.
  • The terms “nucleic acid” and “polynucleotide” as used herein refer to a polymeric form of nucleotides of any length, either ribonucleotides (RNA) or deoxyribonucleotides (DNA). These terms refer to the primary structure of the molecules and, thus, include double- and single-stranded DNA, and double- and single-stranded RNA. These terms include, as equivalents, analogs of either RNA or DNA made from nucleotide analogs and modified polynucleotides such as, though not limited to, methylated and/or capped polynucleotides. The terms encompass poly- or oligo-ribonucleotides (RNA) and poly- or oligo-deoxyribonucleotides (DNA); RNA or DNA derived from N-glycosides or C-glycosides of nucleobases and/or modified nucleobases; nucleic acids derived from sugars and/or modified sugars; and nucleic acids derived from phosphate bridges and/or modified phosphorous-atom bridges. The term encompasses nucleic acids containing any combinations of nucleobases, modified nucleobases, sugars, modified sugars, phosphate bridges or modified phosphorous atom bridges. Examples include, and are not limited to, nucleic acids containing ribose moieties, the nucleic acids containing deoxy-ribose moieties, nucleic acids containing both ribose and deoxyribose moieties, nucleic acids containing ribose and modified ribose moieties. The prefix poly- refers to a nucleic acid containing about 1 to about 10,000 nucleotide monomer units and wherein the prefix oligo- refers to a nucleic acid containing about 1 to about 200 nucleotide monomer units.
  • The term “nucleobase” refers to the parts of nucleic acids that are involved in the hydrogen-bonding that binds one nucleic acid strand to another complementary strand in a sequence specific manner. The most common naturally-occurring nucleobases are adenine (A), guanine (G), uracil (U), cytosine (C), and thymine (T).
  • The term “modified nucleobase” refers to a moiety that can replace a nucleobase. The modified nucleobase mimics the spatial arrangement, electronic properties, or some other physicochemical property of the nucleobase and retains the property of hydrogen-bonding that binds one nucleic acid strand to another in a sequence specific manner. A modified nucleobase can pair with all of the five naturally occurring bases (uracil, thymine, adenine, cytosine, or guanine) without substantially affecting the melting behavior, recognition by intracellular enzymes or activity of the oligonucleotide duplex.
  • The term “chiral reagent” refers to a compound that is chiral or enantiopure and can be used for asymmetric induction in nucleic acid synthesis.
  • The term “chiral ligand” or “chiral auxiliary” refers to a moiety that is chiral or enantiopure and controls the stereochemical outcome of a reaction.
  • In a condensation reaction, the term “condensing reagent” refers to a reagent that activates a less reactive site and renders it more susceptible to attack by a nucleophile.
  • The term “blocking group” refers to a group that transiently masks the reactivity of a functional group. The functional group can be subsequently unmasked by removal of the blocking group.
  • The term “moiety” refers to a specific segment or functional group of a molecule. Chemical moieties are often recognized chemical entities embedded in or appended to a molecule.
  • The term “solid support” refers to any support which enables synthetic mass production of nucleic acids and can be reutilized at need. As used herein, the term refers to a polymer, that is insoluble in the media employed in the reaction steps performed to synthesize nucleic acids, and is derivatized to comprise reactive groups.
  • The term “linking moiety” refers to any moiety optionally positioned between the terminal nucleoside and the solid support or between the terminal nucleoside and another nucleoside, nucleotide, or nucleic acid.
  • A “DNA molecule” refers to the polymeric form of deoxyribonucleotides (adenine, guanine, thymine, or cytosine) in its either single stranded form or a double-stranded helix. This term refers only to the primary and secondary structure of the molecule, and does not limit it to any particular tertiary forms. Thus, this term includes double-stranded DNA found, inter alia, in linear DNA molecules (e.g., restriction fragments), viruses, plasmids, and chromosomes. In discussing the structure of particular double-stranded DNA molecules, sequences can be described herein according to the normal convention of giving only the sequence in the 5′ to 3′ direction along the non-transcribed strand of DNA (i.e., the strand having a sequence homologous to the mRNA).
  • As used herein, an “antisense” nucleic acid molecule comprises a nucleotide sequence which is complementary to a “sense” nucleic acid encoding a protein, e.g., complementary to the coding strand of a double-stranded cDNA molecule, complementary to an mRNA sequence or complementary to the coding strand of a gene. Accordingly, an antisense nucleic acid molecule can hydrogen bond to a sense nucleic acid molecule.
  • As used herein, a “complementary DNA” or “cDNA” includes recombinant polynucleotides synthesized by reverse transcription of mRNA and from which intervening sequences (introns) have been removed.
  • Synthetic Methods for the Preparation Novel Functionalized Nucleic Acids and Nucleic Acid Prodrugs
  • Described herein are methods for the synthesis of novel functionalized nucleic acids and nucleic acid prodrugs. In some embodiments, the nucleic acids comprise chiral phosphorous moieties.
  • One embodiment provides a process for the preparation of phosphorothiotriesters of structure IIIa comprising the steps of:
      • i) reacting an H-phosphonate of structure Ia with an silylating reagent to provide a silyloxyphosphonate; and
      • ii) reacting the silyloxyphosphonate with a thiosulfonate reagent of structure IIa to provide a phosphorothiotriester of structure IIIa;
      • wherein,
      • the H-phosphonate of structure Ia has the following structure:
  • Figure US20170029457A1-20170202-C00020
      • wherein,
      • W is independently selected from O, S, NH, or CH2;
      • R3 is —OH, —SH, —NRdRd, —N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —P(O)(Re)2, —HP(O)(Re), —ORa or —SRc;
      • Y1 is O, NRd, S, or Se;
      • Ra is a blocking group;
      • Rc is a blocking group;
      • each instance of Rd is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, —P(O)(Re)2, or —HP(O)(Re);
      • each instance of Rc is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2—, alkenyl-Y2—, alkynyl-Y2—, aryl-Y2—, or heteroaryl-Y2—, or a cation which is Na+1, Li+1, or K+1;
      • Y2 is O, NRd, or S;
      • each instance of R4 is independently hydrogen, —OH, —SH, —NRdRd, —N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —ORb, or —SRc, and Rb is a blocking group;
      • each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;
      • R5 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid; and
      • n is between 1 and about 200; and
      • the thiosulfonate reagent of structure IIa has the following structure:
  • Figure US20170029457A1-20170202-C00021
  • wherein,
      • X is alkyl, cycloalkyl, or heteroaryl;
      • R is alkyl, alkenyl, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, or R1-R2;
      • R1 is selected from —S-alkenylene-, —S-alkylene-, —S-alkylene-aryl-alkylene-, —S—CO-aryl-alkylene-, or —S—CO-alkylene-aryl-alkylene-;
      • R2 is selected from heterocyclo-alkylene-S—, heterocyclo-alkenylene-S—, aminoalkyl-S—, or (alkyl)4N-alkylene-S—;
      • and the phosphorothiotriester of structure IIIa has the following structure:
  • Figure US20170029457A1-20170202-C00022
      • wherein,
      • W is independently selected from O, S, NH, or CH2;
      • R is alkyl, alkenyl, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, or R1-R2;
      • R1 is selected from —S-alkenylene-, —S-alkylene-, —S-alkylene-aryl-alkylene-, —S—CO-aryl-alkylene-, or —S—CO-alkylene-aryl-alkylene-;
      • R2 is selected from heterocyclo-alkylene-S—, heterocyclo-alkenylene-S—, aminoalkyl-S—, or (alkyl)4N-alkylene-S—;
      • R3 is —OH, —SH, —NRdRd, —N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —P(O)(Re)2, —HP(O)(Re), —ORa or —SRc;
      • Y1 is O, NRd, S, or Se;
      • Ra is a blocking group;
      • Rc is a blocking group;
      • each instance of Rd is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, —P(O)(Re)2, or —HP(O)(Re);
      • each instance of Re is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2—, alkenyl-Y2—, alkynyl-Y2—, aryl-Y2—, or heteroaryl-Y2—, or a cation which is Na+1, Li+1, or K+1;
      • Y2 is O, NRd, or S;
      • each instance of R4 is independently hydrogen, —OH, —SH, —NRdRd, —N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —ORb, or —SRc, and Rb is a blocking group;
      • each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;
      • R5 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid; and
      • n is between 1 and about 200.
  • Another embodiment provides a process for the preparation of phosphorothiotriesters of structure IIIa, wherein W is O.
  • Another embodiment provides a process for the preparation of phosphorothiotriesters of structure IIIa, wherein R1 is selected from:
  • Figure US20170029457A1-20170202-C00023
  • and
      • R2 is selected from:
  • Figure US20170029457A1-20170202-C00024
  • Another embodiment provides a process for the preparation of phosphorothiotriesters of structure IIIa, wherein the silylating reagent is selected from
      • 1,1,3,3-tetramethyl-1,3-diphenyldisilazane;
      • 1,3-dimethyl-1,1,3,3-tetraphenyldisilazane;
      • 1-(trimethylsilyl)imidazole;
      • N-trimethylsilyl-N-methyl trifluoroacetamide;
      • bis(dimethylamino)dimethylsilane;
      • bromotrimethylsilane;
      • chlorodimethyl(pentafluorophenyl)silane;
      • chlorotriethylsilane;
      • chlorotriisopropylsilane;
      • chlorotrimethylsilane;
      • dichlorodimethylsilane;
      • hexamethyldisilazane;
      • N,N′-bis(trimethylsilyl)urea;
      • N,N-bis(trimethylsilyl)methylamine;
      • N,N-dimethyltrimethylsilylamine;
      • N,O-bis(trimethylsilyl)acetamide;
      • N,O-bis(trimethylsilyl)carbamate;
      • N,O-bis(trimethylsilyl)trifluoroacetamide;
      • N-methyl-N-(trimethylsilyl)trifluoroacetamide;
      • N-methyl-N-trimethylsilylacetamide;
      • N-methyl-N-trimethylsilylheptafluorobutyramide;
      • N-tert-butyldimethylsilyl-N-methyltrifluoroacetamide;
      • N-methyl-N-trimethylsilylheptafluorobutyramide;
      • trimethylsilyltriflate;
      • triethylsilyltriflate;
      • triisopropylsilyltriflate; or
      • tert-butyldimethylsilyltriflate.
  • Another embodiment provides the process, wherein the silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide, trimethylsilyltriflate, chlorotrimethylsilane, or 1-(trimethylsilyl)imidazole.
  • Another embodiment provides the process, wherein the silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide.
  • Another embodiment provides the process, wherein the H-phosphonate is covalently linked to a solid phase.
  • One embodiment provides a process for the preparation of phosphorothiotriesters comprising non-stereorandom phosphorous linkages of structure IIIb comprising the steps of:
      • i) reacting a H-phosphonate comprising non-stereorandom phosphorous linkages of structure Ib with an silylating reagent to provide a silyloxyphosphonate; and
      • ii) reacting the silyloxyphosphonate with a thiosulfonate reagent of structure IIb to provide a phosphorothiotriester comprising non-stereorandom phosphorous linkages of structure IIb;
      • wherein,
      • the H-phosphonate comprising non-stereorandom phosphorous linkages of structure Ib has the following structure:
  • Figure US20170029457A1-20170202-C00025
      • wherein,
      • W is independently selected from O, NH, or CH2;
      • R3 is —OH, —SH, —NRdRd, —N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —P(O)(Re)2, —HP(O)(Re), —ORa or —SRc;
      • Y1 is O, NRd, S, or Se;
      • Ra is a blocking group;
      • Rc is a blocking group;
      • each instance of Rd is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, —P(O)(Re)2, or —HP(O)(Re);
      • each instance of Re is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2—, alkenyl-Y2—, alkynyl-Y2—, aryl-Y2—, or heteroaryl-Y2—, or a cation which is Na+1, Li+1, or K+; Y2 is O, NRd, or S;
      • each instance of R4 is independently hydrogen, —OH, —SH, —NRdRd, —N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —ORb, or —SRc, and Rb is a blocking group;
      • each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;
      • R5 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid; and
      • n is between 1 and about 200; and
      • the thiosulfonate reagent of structure IIb has the following structure:
  • Figure US20170029457A1-20170202-C00026
  • wherein,
      • X is alkyl, cycloalkyl, aryl, or heteroaryl;
      • R is alkyl, alkenyl, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, or R1-R2;
      • R1 is selected from —S-alkenylene-, —S-alkylene-, —S-alkylene-aryl-alkylene-, —S—CO-aryl-alkylene-, or —S—CO-alkylene-aryl-alkylene-; R2 is selected from heterocyclo-alkylene-S—, heterocyclo-alkenylene-S—, aminoalkyl-S—, or (alkyl)4N-alkylene-S—;
      • and the chiral phosphorothiotriester comprising non-stereorandom phosphorous linkages of structure IIIb has the following structure:
  • Figure US20170029457A1-20170202-C00027
      • wherein,
      • W is independently selected from O, NH, or CH2;
      • R is alkyl, alkenyl, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, or R1-R2;
      • R1 is selected from —S-alkenylene-, —S-alkylene-, —S-alkylene-aryl-alkylene-, —S—CO-aryl-alkylene-, or —S—CO-alkylene-aryl-alkylene-;
      • R2 is selected from heterocyclo-alkylene-S—, heterocyclo-alkenylene-S—, aminoalkyl-S—, or (alkyl)4N-alkylene-S—;
      • R3 is —OH, —SH, —NRdRd, —N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —P(O)(Re)2, —HP(O)(Re), —ORa or —SRc;
      • Y1 is O, NRd, S, or Se;
      • Ra is a blocking group;
      • Rc is a blocking group;
      • each instance of Rd is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, —P(O)(Re)2, or —HP(O)(Re);
      • each instance of Re is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2—, alkenyl-Y2—, alkynyl-Y2—, aryl-Y2—, or heteroaryl-Y2—, or a cation which is Na+1, Li+1, or K+1;
      • Y2 is O, NRd, or S;
      • each instance of R4 is independently hydrogen, —OH, —SH, —NRdRd, —N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —ORb, or —SRc, and Rb is a blocking group;
      • each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;
      • R5 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid; and
      • n is between 1 and about 200.
  • Another embodiment provides a process for the preparation of phosphorothiotriesters comprising non-stereorandom phosphorous linkages of structure IIIb, wherein W is O.
  • Another embodiment provides a process for the preparation of phosphorothiotriesters comprising non-stereorandom phosphorous linkages of structure IIIb, wherein R1 is selected from:
  • Figure US20170029457A1-20170202-C00028
      • R2 is selected from:
  • Figure US20170029457A1-20170202-C00029
  • Another embodiment provides a process for the preparation of phosphorothiotriesters comprising non-stereorandom phosphorous linkages of structure IIIb, wherein the silylating reagent is selected from
      • 1,1,3,3-tetramethyl-1,3-diphenyldisilazane;
      • 1,3-dimethyl-1,1,3,3-tetraphenyldisilazane;
      • 1-(trimethylsilyl)imidazole;
      • N-trimethylsilyl-N-methyl trifluoroacetamide;
      • bis(dimethylamino)dimethylsilane;
      • bromotrimethylsilane;
      • chlorodimethyl(pentafluorophenyl)silane;
      • chlorotriethylsilane;
      • chlorotriisopropylsilane;
      • chlorotrimethylsilane;
      • dichlorodimethylsilane;
      • hexamethyldisilazane;
      • N,N′-bis(trimethylsilyl)urea;
      • N,N-bis(trimethylsilyl)methylamine;
      • N,N-dimethyltrimethylsilylamine;
      • N,O-bis(trimethylsilyl)acetamide;
      • N,O-bis(trimethylsilyl)carbamate;
      • N,O-bis(trimethylsilyl)trifluoroacetamide;
      • N-methyl-N-(trimethylsilyl)trifluoroacetamide;
      • N-methyl-N-trimethylsilylacetamide;
      • N-methyl-N-trimethylsilylheptafluorobutyramide;
      • N-tert-butyldimethylsilyl-N-methyltrifluoroacetamide;
      • N-methyl-N-trimethylsilylheptafluorobutyramide;
      • trimethylsilyltriflate;
      • triethylsilyltriflate;
      • triisopropylsilyltriflate; or
      • tert-butyldimethylsilyltriflate.
  • Another embodiment provides the process, wherein the silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide, trimethylsilyltriflate, chlorotrimethylsilane, or 1-(trimethylsilyl)imidazole.
  • Another embodiment provides the process, wherein the silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide.
  • Another embodiment provides the process, wherein the H-phosphonate is covalently linked to a solid phase.
  • One embodiment provides a process for the preparation of phosphorothiotriesters of structure IIIc comprising the steps of:
      • i) reacting a H-phosphonate of structure Ic with an silylating reagent to provide a silyloxyphosphonate;
      • ii) reacting the silyloxyphosphonate with a bis(thiosulfonate) reagent of structure IVc to provide a phosphorothiotriester comprising a thiosulfonate group of structure Vc;
      • iii) reacting the phosphorothiotriester comprising a thiosulfonate group of structure Vc with a nucleophile of structure VIc to provide the phosphorothiotriesters of structure IIIc;
      • wherein,
      • the H-phosphonate of structure Ic has the following structure:
  • Figure US20170029457A1-20170202-C00030
      • wherein,
      • W is independently selected from O, S, NH, or CH2;
      • R3 is —OH, —SH, —NRdRd, —N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, P(O)(Re)2, —HP(O)(Re), —ORa or —SRc;
      • Y1 is O, NRd, S, or Se;
      • Ra is a blocking group;
      • Rc is a blocking group;
      • each instance of Rd is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, —P(O)(Re)2, or —HP(O)(Re);
      • each instance of Re is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2—, alkenyl-Y2—, alkynyl-Y2—, aryl-Y2—, or heteroaryl-Y2—, or a cation which is Na+1, Li+1, or K+1;
      • Y2 is O, NRd, or S;
      • each instance of R4 is independently hydrogen, —OH, —SH, —NRdRd, —N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —ORb, or —SRc, and Rb is a blocking group;
      • each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;
      • R5 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid; and
      • n is between 1 and about 200; and
      • the bis(thiosulfonate) reagent of structure IVc has the following structure:
  • Figure US20170029457A1-20170202-C00031
  • wherein,
      • X is alkylene, alkenylene, arylene, or heteroarylene;
      • each R6 is independently alkyl, cycloalkyl, aryl, or heteroaryl;
      • the nucleophile of structure VIc has the following structure:
      • R7—SH, wherein R7 is selected from alkyl, alkenyl, aryl, heterocyclo, aminoalkyl, or (heterocyclo)alkyl;
      • and phosphorothiotriesters of structure IIIc has the following structure:
  • Figure US20170029457A1-20170202-C00032
      • wherein,
      • W is independently selected from O, S, NH, or CH2;
      • R is R7—S—S—X—
      • R7 is alkyl, alkenyl, aryl, heterocyclo, aminoalkyl, or (heterocyclo)alkyl;
      • X is alkylene, alkenylene, arylene, or heteroarylene;
      • R3 is —OH, —SH, —NRdRd, —N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —P(O)(Re)2, —HP(O)(Re), —ORa or —SRc;
      • Y1 is O, NRd, S, or Se;
      • Ra is a blocking group;
      • Rc is a blocking group;
      • each instance of Rd is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, —P(O)(Re)2, or —HP(O)(Re);
      • each instance of Re is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2—, alkenyl-Y2—, alkynyl-Y2—, aryl-Y2—, or heteroaryl-Y2—, or a cation which is Na+1, Li+1, or K+1;
      • Y2 is O, NRd, or S;
      • each instance of R4 is independently hydrogen, —OH, —SH, —NRdRd, —N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —ORb, or —SRc, and Rb is a blocking group;
      • each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;
      • R5 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid;
      • n is between 1 and about 200; and
      • wherein the phosphorous linkages of the H-phosphonate of structure Ic, the phosphorothiotriester comprising a thiosulfonate group of structure Vc, and the phosphorothiotriesters of structure IIIc may optionally comprise non-stereorandom phosphorous linkages.
  • Another embodiment provides the process wherein the phosphorothiotriesters of structure IIIb comprise non-stereorandom phosphorous linkages and the H-phosphonate of structure Ic comprise non-stereorandom phosphorous linkages; and W is independently selected from O, NH, or CH2. Another embodiment provides the process wherein W is O.
  • Another embodiment provides the process wherein R6 is methyl.
  • Another embodiment provides the process wherein bis(thiosulfonate) reagent of structure IVc is selected from:
  • Figure US20170029457A1-20170202-C00033
  • Another embodiment provides the process wherein the nucleophile of structure VIc has the following structure:
  • Figure US20170029457A1-20170202-C00034
  • Another embodiment provides a process for the preparation of phosphorothiotriesters of structure IIIa, wherein the silylating reagent is selected from
      • 1,1,3,3-tetramethyl-1,3-diphenyldisilazane;
      • 1,3-dimethyl-1, 1,3,3-tetraphenyldisilazane;
      • 1-(trimethylsilyl)imidazole;
      • N-trimethylsilyl-N-methyl trifluoroacetamide;
      • bis(dimethylamino)dimethylsilane;
      • bromotrimethylsilane;
      • chlorodimethyl(pentafluorophenyl)silane;
      • chlorotriethylsilane;
      • chlorotriisopropylsilane;
      • chlorotrimethylsilane;
      • dichlorodimethylsilane;
      • hexamethyldisilazane;
      • N,N′-bis(trimethylsilyl)urea;
      • N,N-bis(trimethylsilyl)methylamine;
      • N,N-dimethyltrimethylsilylamine;
      • N,O-bis(trimethylsilyl)acetamide;
      • N,O-bis(trimethylsilyl)carbamate;
      • N,O-bis(trimethylsilyl)trifluoroacetamide;
      • N-methyl-N-(trimethylsilyl)trifluoroacetamide;
      • N-methyl-N-trimethylsilylacetamide;
      • N-methyl-N-trimethylsilylheptafluorobutyramide;
      • N-tert-butyldimethylsilyl-N-methyltrifluoroacetamide;
      • N-methyl-N-trimethylsilylheptafluorobutyramide;
      • trimethylsilyltriflate;
      • triethylsilyltriflate;
      • triisopropylsilyltriflate; or
      • tert-butyldimethylsilyltriflate.
  • Another embodiment provides the process, wherein the silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide, trimethylsilyltriflate, chlorotrimethylsilane, or 1-(trimethylsilyl)imidazole.
  • Another embodiment provides the process, wherein the silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide.
  • Another embodiment provides the process, wherein the H-phosphonate is covalently linked to a solid phase.
  • Modified Oligonucleotides
  • Oligonucleotides have several pharmaceutical properties which can be improved through the application of prodrug strategies. In particular, oligonucleotides are rapidly degraded by nucleases and exhibit poor cellular uptake through the cytoplasmic cell membrane (Poijarvi-Virta et al., Curr. Med. Chem. (2006), 13(28); 3441-65; Wagner et al., Med. Res. Rev. (2000), 20(6):417-51; Peyrottes et al., Mini Rev. Med. Chem. (2004), 4(4):395-408; Gosselin et al., (1996), 43(1):196-208; Bologna et al., (2002), Antisense & Nucleic Acid Drug Development 12:33-41). In one example, Vives et al., (Nucleic Acids Research (1999), 27(20):4071-76) found that tert-butyl SATE pro-oligonucleotides displayed markedly increased cellular penetration compared to the parent oligonucleotide. Described herein are methods for the synthesis of modified oligonucleotides or pronucleotides.
  • Reaction Conditions and Reagents Used in the Methods of the Invention. Conditions
  • The steps of reacting a molecule comprising an achiral H-phosphonate moiety and a nucleoside comprising a 5′-OH moiety to form a condensed intermediate can occur without isolating any intermediates. In some embodiments, the steps of reacting a molecule comprising an achiral H-phosphonate moiety and a nucleoside comprising a 5′-OH moiety to form a condensed intermediate occurs is a one-pot reaction. In an embodiment, a molecule comprising an achiral H-phosphonate moiety, condensing reagent, chiral reagent, and compound comprising a free nucleophilic moiety are added to the reaction mixture at different times. In another embodiment, a molecule comprising an achiral H-phosphonate moiety, condensing reagent, and chiral reagent are present in the same reaction vessel or same pot. In another embodiment, a molecule comprising an achiral H-phosphonate moiety, condensing reagent, chiral reagent, and compound comprising a free nucleophilic moiety are present in the same reaction or same pot. This allows the reaction to be performed without isolation of intermediates and eliminates time-consuming steps, resulting in an economical and efficient synthesis. In specific embodiments, the achiral H-phosphonate, condensing reagent, chiral amino alcohol, 5′-OH nucleoside are present at the same time in a reaction. In a further embodiment, the formation of the chiral intermediate for condensation is formed in situ and is not isolated prior to the condensation reaction. In another embodiment, a molecule comprising an achiral H-phosphonate moiety has been activated by reaction with a condensing reagent, chiral reagent in a different reaction vessel from that used when reacting the chiral intermediate with the compound comprising a free 5′-OH moiety.
  • Synthesis on Solid Support
  • In some embodiments, the synthesis of the nucleic acid is performed in solution. In other embodiments, the synthesis of the nucleic acid is performed on solid phase. The reactive groups of a solid support may be unprotected or protected. During oligonucleotide synthesis a solid support is treated with various reagents in several synthesis cycles to achieve the stepwise elongation of a growing oligonucleotide chain with individual nucleotide units. The nucleoside unit at the end of the chain which is directly linked to the solid support is termed “the first nucleoside” as used herein. The first nucleoside is bound to the solid support via a linker moiety, i.e. a diradical with covalent bonds to both the polymer of the solid support and the nucleoside. The linker stays intact during the synthesis cycles performed to assemble the oligonucleotide chain and is cleaved after the chain assembly to liberate the oligonucleotide from the support.
  • Solid supports for solid-phase nucleic acid synthesis include the supports described in, e.g., U.S. Pat. Nos. 4,659,774, 5,141,813, 4,458,066; Caruthers U.S. Pat. Nos. 4,415,732, 4,458,066, 4,500,707, 4,668,777, 4,973,679, and 5,132,418; Andrus et al. U.S. Pat. Nos. 5,047,524, 5,262,530; and Koster U.S. Pat. No. 4,725,677 (reissued as Re34,069). In some embodiments, the solid phase is an organic polymer support. In other embodiments, the solid phase is an inorganic polymer support. In some embodiments, the organic polymer support is polystyrene, aminomethyl polystyrene, a polyethylene glycol-polystyrene graft copolymer, polyacrylamide, polymethacrylate, polyvinylalcohol, highly cross-linked polymer (HCP), or other synthetic polymers, carbohydrates such as cellulose and starch or other polymeric carbohydrates, or other organic polymers and any copolymers, composite materials or combination of the above inorganic or organic materials. In other embodiments, the inorganic polymer support is silica, alumina, controlled poreglass (CPG), which is a silica-gel support, or aminopropyl CPG. Other useful solid supports include fluorous solid supports (see e.g., WO/2005/070859), long chain alkylamine (LCAA) controlled pore glass (CPG) solid supports (sce e.g., S. P. Adams, K. S. Kavka, E. J. Wykes, S. B. Holder and G. R. Galluppi, J. Am. Chem. Soc., 1983, 105, 661-663; G. R. Gough, M. J. Bruden and P. T. Gilham, Tetrahedron Lett., 1981, 22, 4177-4180). Membrane supports and polymeric membranes (see e.g. Innovation and Perspectives in Solid Phase Synthesis, Peptides, Proteins and Nucleic Acids, ch 21 pp 157-162, 1994, Ed. Roger Epton and U.S. Pat. No. 4,923,901) are also useful for the synthesis of nucleic acids. Once formed, a membrane can be chemically functionalized for use in nucleic acid synthesis. In addition to the attachment of a functional group to the membrane, the use of a linker or spacer group attached to the membrane may be used to minimize steric hindrance between the membrane and the synthesized chain.
  • Other suitable solid supports include those generally known in the art to be suitable for use in solid phase methodologies, including, for example, glass sold as Primer™ 200 support, controlled pore glass (CPG), oxalyl-controlled pore glass (see, e.g., Alul, et al., Nucleic Acids Research, 1991, 19, 1527), TentaGel Support—an aminopolyethyleneglycol derivatized support (see, e.g., Wright, et al., Tetrahedron Lett., 1993, 34, 3373), and Poros-a copolymer of polystyrene/divinylbenzene.
  • Surface activated polymers have been demonstrated for use in synthesis of natural and modified nucleic acids and proteins on several solid supports mediums. The solid support material can be any polymer suitably uniform in porosity, has sufficient amine content, and sufficiently flexible to undergo any attendant manipulations without losing integrity. Examples of suitable selected materials include nylon, polypropylene, polyester, polytetrafluoroethylene, polystyrene, polycarbonate, and nitrocellulose. Other materials can serve as the solid support, depending on the design of the investigator. In consideration of some designs, for example, a coated metal, in particular gold or platinum can be selected (see e.g., US publication No. 20010055761). In one embodiment of oligonucleotide synthesis, for example, a nucleoside is anchored to a solid support which is functionalized with hydroxyl or amino residues. Alternatively, the solid support is derivatized to provide an acid labile trialkoxytrityl group, such as a trimethoxytrityl group (TMT). Without being bound by theory, it is expected that the presence of the trialkoxytrityl protecting group will permit initial detritylation under conditions commonly used on DNA synthesizers. For a faster release of oligonucleotide material in solution with aqueous ammonia, a diglycoate linker is optionally introduced onto the support.
  • Linking Moiety
  • A linking moiety or linker is optionally used to connect the solid support to the compound comprising a free nucleophilic moiety. Suitable linkers are known such as short molecules which serve to connect a solid support to functional groups (e.g., hydroxyl groups) of initial nucleosides molecules in solid phase synthetic techniques. In some embodiments, the linking moiety is a succinamic acid linker, or a succinate linker (—CO—CH2—CH2—CO—), or an oxalyl linker (—CO—CO—). In other embodiments, the linking moiety and the nucleoside are bonded together through an ester bond. In other embodiments, the linking moiety and the nucleoside are bonded together through an amide bond. In further embodiments, the linking moiety connects the nucleoside to another nucleotide or nucleic acid. Suitable linkers are disclosed in, for example, Oligonucleotides And Analogues A Practical Approach, Ekstein, F. Ed., IRL Press, N.Y., 1991, Chapter 1.
  • A linker moiety is used to connect the compound comprising a free nucleophilic moiety to another nucleoside, nucleotide, or nucleic acid. In some embodiments, the linking moiety is a phosphodiester linkage. In other embodiments, the linking moiety is an H-phosphonate moiety. In yet other embodiments, the linking moiety is an X-phosphonate moiety.
  • Solvents for Synthesis
  • Synthesis of the nucleic acids is performed in an aprotic organic solvent. In some embodiments, the solvent is acetonitrile, pyridine, or NMP. In some embodiments, the solvent is acetone, acetontrile, NMP, ethyl acetate, THF, dioxane, DMF, DMSO, DCM, chloroform, pyridine, 2,6-lutidine, HMPA, HMPT, DMA, glyme, diglyme, sulfone, methyl tert-butyl ether, or combinations thereof. In some embodiments, the solvent is a polar, aprotic organic solvent. In some embodiments, the solvent is anhydrous.
  • Acidification Conditions to Remove Blocking Groups.
  • Acidification to remove blocking groups is accomplished by a Brønsted acid or Lewis acid. In some embodiments, acidification is used to remove R1 blocking groups. Useful Brønsted acids are carboxylic acids, alkylsulfonic acids, arylsulfonic acids, phosphoric acid and its derivatives, phosphonic acid and its derivatives, alkylphosphonic acids and their derivatives, arylphosphonic acids and their derivatives, phosphinic acid, dialkylphosphinic acids, and diarylphosphinic acids which have a pKa (25° C. in water) value of −0.6 (trifluoroacetic acid) to 4.76 (acetic acid) in an organic solvent or water (in the case of 80% acetic acid). The concentration of the acid (1 to 80%) used in the acidification step depends on the acidity of the acid. Consideration to the acid strength must be taken into account as strong acid conditions will result in depurination/depyrimidination, wherein purinyl or pyrimidinyl bases are cleaved from ribose ring.
  • Figure US20170029457A1-20170202-C00035
  • In some embodiments, acidification is accomplished by a Lewis acid in an organic solvent. Useful Lewis acids are ZnX2 wherein X is Cl, Br, I, or CF3SO3.
  • In some embodiments, the acidifying comprises adding an amount of a Brønsted or Lewis acid effective to convert the condensed intermediate into the compound of Formula 4 without removing purine moieties from the condensed intermediate.
  • Acids that are useful in the acidifying step also include, but are not limited to 10% phosphoric acid in an organic solvent, 10% hydrochloric acid in an organic solvent, 1% trifluoroacetic acid in an organic solvent, 3% dichloroacetic acid in an organic solvent or 80% acetic acid in water. The concentration of any Brønsted or Lewis acid used in the process is selected such that the concentration of the acid does not exceed a concentration that causes cleavage of the nucleobase from the sugar moiety.
  • In some embodiments, acidification comprises adding 1% trifluoroacetic acid in an organic solvent. In some embodiments, acidification comprises adding about 0.1% to about 8% trifluoroacetic acid in an organic solvent. In other embodiments, acidification comprises adding 3% dichloroacetic acid in an organic solvent. In other embodiments, acidification comprises adding about 0.1% to about 10% dichloroacetic acid in an organic solvent. In yet other embodiments, acidification comprises adding 3% trichloroacetic acid in an organic solvent. In yet other embodiments, acidification comprises adding about 0.1% to about 10% trichloroacetic acid in an organic solvent. In some embodiments, acidification comprises adding 80% acetic acid in water. In some embodiments, acidification comprises adding about 50% to about 90%, or about 50% to about 80%, about 50% to about 70%, about 50% to about 60%, about 70% to about 90% acetic acid in water. In some embodiments, the acidification comprises the further addition of cation scavengers to the acidic solvent. In specific embodiments, the cation scavengers can be triethylsilane or triisopropylsilane. In some embodiments, R1 is deblocked prior to the step of acidifying the condensed intermediate. In some embodiments, R1 is deblocked by acidification, which comprises adding 1% trifluoroacetic acid in an organic solvent. In some embodiments, R1 is deblocked by acidification, which comprises adding 3% dichloroacetic acid in an organic solvent. In some embodiments, R1 is deblocked by acidification, which comprises adding 3% trichloroacetic acid in an organic solvent.
  • Removal of Blocking Moieties or Groups
  • Functional groups such as hydroxyl or amino moieties which are located on nucleobases or sugar moieties are routinely blocked with blocking (protecting) groups (moieties) during synthesis and subsequently deblocked. In general, a blocking group renders a chemical functionality of a molecule inert to specific reaction conditions and can later be removed from such functionality in a molecule without substantially damaging the remainder of the molecule (see e.g., Green and Wuts, Protective Groups in Organic Synthesis, 2nd Ed., John Wiley & Sons, New York, 1991). For example, amino groups can be blocked with nitrogen blocking groups such as phthalimido, 9-fludrenylmethoxycarbonyl (FMOC), triphenylmethylsulfenyl, t-BOC, 4,4′-dimethoxytrityl (DMTr), 4-methoxytrityl (MMTr), 9-phenylxanthin-9-yl (Pixyl), trityl (Tr), or 9-(p-methoxyphenyl)xanthin-9-yl (MOX). Carboxyl groups can be protected as acetyl groups. Hydroxy groups can be protected such as tetrahydropyranyl (THP), t-butyldimethylsilyl (TBDMS), 1-[(2-chloro-4-methyl)phenyl]-4-methoxypiperidin-4-yl (Ctmp), 1-(2-fluorophenyl)-4-methoxypiperidin-4-yl (Fpmp), 1-(2-chloroethoxy)ethyl, 3-methoxy-1,5-dicarbomethoxypentan-3-yl (MDP), bis(2-acetoxyethoxy)methyl (ACE), triisopropylsilyloxymethyl (TOM), 1-(2-cyanoethoxy)ethyl (CEE), 2-cyanoethoxymethyl (CEM), [4-(N-dichloroacetyl-N-methylamino)benzyloxy]methyl, 2-cyanoethyl (CN), pivaloyloxymethyl (PivOM), levunyloxymethyl (ALE). Other representative hydroxyl blocking groups have been described (see e.g., Beaucage et al., Tetrahedron, 1992, 46, 2223). In some embodiments, hydroxyl blocking groups are acid-labile groups, such as the trityl, monomethoxytrityl, dimethoxytrityl, trimethoxytrityl, 9-phenylxanthin-9-yl (Pixyl) and 9-(p-methoxyphenyl)xanthin-9-yl (MOX). Chemical functional groups can also be blocked by including them in a precursor form. Thus an azido group can be considered a blocked form of an amine as the azido group is easily converted to the amine. Further representative protecting groups utilized in nucleic acid synthesis are known (see e.g. Agrawal et al., Protocols for Oligonucleotide Conjugates, Eds., Humana Press, New Jersey, 1994, Vol. 26, pp. 1-72).
  • Various methods are known and used for removal of blocking groups from the nucleic acids. In some embodiments, all blocking groups are removed. In other embodiments, the blocking groups are partially removed. In yet other embodiments, reaction conditions can be adjusted to remove blocking groups on certain moieties. In certain embodiments where R2 is a blocking group, removal of the blocking group at R2 is orthogonal to the removal of the blocking group at R1. The blocking groups at R1 and R2 remain intact during the synthesis steps and are collectively removed after the chain assembly. In some embodiments, the R2 blocking group are removed simultaneously with the cleavage of the nucleic acids from the solid support and with the removal of the nucleobase blocking groups. In specific embodiments, the blocking group at R1 is removed while the blocking groups at R2 and nucleobases remain intact. Blocking groups at R1 are cleavable on solid supports with an organic base such as a primary amine, a secondary amine, or a mixture thereof. Deblocking of the R1 position is commonly referred to as front end deprotection.
  • In an embodiment, the nucleobase blocking groups, if present, are cleavable after the assembly of the respective nucleic acid with an acidic reagent. In another embodiment, one or more of the nucleobase blocking groups is cleavable under neither acidic nor basic conditions, e.g. cleavable with fluoride salts or hydrofluoric acid complexes. In yet another embodiment, one or more of the nucleobase blocking groups are cleavable after the assembly of the respective nucleic acid in the presence of base or a basic solvent, and wherein the nucleobase blocking group is stable to the conditions of the front end deprotection step with amines.
  • In some embodiments, blocking groups for nucleobases are not required. In other embodiments, blocking groups for nucleobases are required. In yet other embodiments, certain nucleobases require blocking group while other nucleobases do not require blocking groups. In embodiments where the nucleobases are blocked, the blocking groups are either completely or partially removed under conditions appropriate to remove the blocking group at the front end. For example, R1 can denote ORa, wherein Ra is acyl, and Ba denotes guanine blocked with an acyl group including, but not limited to isobutyryl, acetyl or 4-(tert-butylphenoxy)acetyl. The acyl groups at R1 and Ba will be removed or partially removed during the same deblocking step.
  • Stereochemistry of Oligonucleoside Phosphorothioate Linkages
  • Oligonucleoside phosphorothioates have shown therapeutic potential (Stein et al., Science (1993), 261:1004-12; Agrawal et al., Antisence Res. and Dev. (1992), 2:261-66; Bayever et al., Antisense Res. and Dev. (1993), 3:383-390). Oligonucleoside phosphorothioates prepared without regard to the stereochemistry of the phosphorothioate exist as a mixture of 2n diastereomers, where n is the number of internucleotide phosphorothioates linkages. The chemical and biological properties of these diastereomeric phosphorothioates can be distinct. For example, Wada et al (Nucleic Acids Symposium Series No. 51 p. 119-120; doi:10.1093/nass/nrm060) found that stereodefined-(Rp)-(Ups)9U/(Ap)9A duplex showed a higher Tm value than that of natural-(Up)9U/(Ap)9A and stereodefined-(Sp)-(Ups)9U did not form a duplex. In another example, in a study by Tang et al., (Nucleosides Nucleotides (1995), 14:985-990) stereopure Rp-oligodeoxyribonucleoside phosphorothioates were found to possess lower stability to nucleases endogenous to human serum that the parent oligodeoxyribonucleoside phosphorothioates with undefined phosphorous chirality.
  • Nucleobases and Modified Nucleobases
  • The nucleobase Ba utilized in the compounds and methods described herein is a natural nucleobase or a modified nucleobase derived from natural nucleobases. Examples include, but are not limited to, uracil, thymine, adenine, cytosine, and guanine having their respective amino groups protected by acyl protecting groups, 2-fluorouracil, 2-fluorocytosine, 5-bromouracil, 5-iodouracil, 2,6-diaminopurine, azacytosine, pyrimidine analogs such as pseudoisocytosine and pseudouracil and other modified nucleobases such as 8-substituted purines, xanthine, or hypoxanthine (the latter two being the natural degradation products). The modified nucleobases disclosed in Chiu and Rana, RNA, 2003, 9, 1034-1048, Limbach et al. Nucleic Acids Research, 1994, 22, 2183-2196 and Revankar and Rao, Comprehensive Natural Products Chemistry, vol. 7, 313, are also contemplated as Ba moieties of the compounds and methods described herein.
  • Compounds represented by the following general formulae are also contemplated as modified nucleobases:
  • Figure US20170029457A1-20170202-C00036
  • In the formulae above, R8 is a linear or branched alkyl, aryl, aralkyl, or aryloxylalkyl group having 1 to 15 carbon atoms, including, by way of example only, a methyl, isopropyl, phenyl, benzyl, or phenoxymethyl group; and each of R9 and R10 represents a linear or branched alkyl group having 1 to 4 carbon atoms.
  • Modified nucleobases also include expanded-size nucleobases in which one or more benzene rings has been added. Nucleic base replacements described in the Glen Research catalog (www.glenresearch.com); Krueger A T et al, Acc. Chem. Res., 2007, 40, 141-150; Kool, E T, Acc. Chem. Res., 2002, 35, 936-943; Benner S. A., et al., Nat. Rev. Genet., 2005, 6, 553-543; Romesberg, F. E., et al., Curr. Opin. Chem. Biol., 2003, 7, 723-733; Hirao, I., Curr. Opin. Chem. Biol., 2006, 10, 622-627, are contemplated as useful for the synthesis of the nucleic acids described herein. Some examples of these expanded-size nucleobases are shown below:
  • Figure US20170029457A1-20170202-C00037
  • Herein, modified nucleobases also encompass structures that are not considered nucleobases but are other moieties such as, but not limited to, corrin- or porphyrin-derived rings. Porphyrin-derived base replacements have been described in Morales-Rojas, H and Kool, E T, Org. Lett., 2002, 4, 4377-4380. Shown below is an example of a porphyrin-derived ring which can be used as a base replacement:
  • Figure US20170029457A1-20170202-C00038
  • Other modified nucleobases also include base replacements such as those shown below:
  • Figure US20170029457A1-20170202-C00039
  • Modified nucleobases which are fluorescent are also contemplated. Non-limiting examples of these base replacements include phenanthrene, pyrene, stillbene, isoxanthine, isozanthopterin, terphenyl, terthiophene, benzoterthiophene, coumarin, lumazine, tethered stillbene, benzo-uracil, and naphtho-uracil, as shown below:
  • Figure US20170029457A1-20170202-C00040
  • The modified nucleobases can be unsubstituted or contain further substitutions such as heteroatoms, alkyl groups, or linking moieties connected to fluorescent moieties, biotin or avidin moieties, or other protein or peptides. Modified nucleobases also include certain ‘universal bases’ that are not nucleobases in the most classical sense, but function similarly to nucleobases. One representative example of such a universal base is 3-nitropyrrole.
  • Other nucleosides can also be used in the process disclosed herein and include nucleosides that incorporate modified nucleobases, or nucleobases covalently bound to modified sugars. Some examples of nucleosides that incorporate modified nucleobases include 4-acetylcytidine; 5-(carboxyhydroxylmethyl)uridine; 2′-O-methylcytidine; 5-carboxymethylaminomethyl-2-thiouridine; 5-carboxymethylaminomethyluridine; dihydrouridine; 2′-O-methylpseudouridine; beta,D-galactosylqueosine; 2′-O-methylguanosine; N6-isopentenyladenosine; 1-methyladenosine; 1-methylpseudouridine; 1-methylguanosine; 1-methylinosine; 2,2-dimethylguanosine; 2-methyladenosine; 2-methylguanosine; N7-methylguanosine; 3-methyl-cytidine; 5-methylcytidine; N6-methyladenosine; 7-methylguanosine; 5-methylaminoethyluridine; 5-methoxyaminomethyl-2-thiouridine; beta,D-mannosylqueosine; 5-methoxycarbonylmethyluridine; 5-methoxyuridine; 2-methylthio-N6-isopentenyladenosine; N-((9-beta,D-ribofuranosyl-2-methylthiopurine-6-yl)carbamoyl)threonine; N-((9-beta,D-ribofuranosylpurine-6-yl)-N-methylcarbamoyl)threonine; uridine-5-oxyacetic acid methylester; uridine-5-oxyacetic acid (v); pseudouridine; queosine; 2-thiocytidine; 5-methyl-2-thiouridine; 2-thiouridine; 4-thiouridine; 5-methyluridine; 2′-O-methyl-5-methyluridine; and 2′-O-methyluridine.
  • In some embodiments, nucleosides include 6′-modified bicyclic nucleoside analogs that have either (R) or (S)-chirality at the 6′-position and include the analogs described in U.S. Pat. No. 7,399,845. In other embodiments, nucleosides include 5′-modified bicyclic nucleoside analogs that have either (R) or (S)-chirality at the 5′-position and include the analogs described in US Patent Application Publication No. 20070287831.
  • In some embodiments, the nucleobases or modified nucleobases comprises biomolecule binding moieties such as antibodies, antibody fragments, biotin, avidin, streptavidin, receptor ligands, or chelating moieties. In other embodiments, Ba is 5-bromouracil, 5-iodouracil, or 2,6-diaminopurine. In yet other embodiments, Ba is modified by substitution with a fluorescent or biomolecule binding moiety. In some embodiments, the substituent on Ba is a fluorescent moiety. In other embodiments, the substituent on Ba is biotin or avidin.
  • Modified Sugars of the Nucleotide/Nucleoside.
  • The most common naturally occurring nucleotides are ribose sugars linked to the nucleobases adenosine (A), cytosine (C), guanine (G), and thymine (T) or uracil (U). Also contemplated are modified nucleotides wherein the phosphate group or the modified phosphorous atom moieties in the nucleotides can be linked to various positions of the sugar or modified sugar. As non-limiting examples, the phosphate group or the modified phosphorous-atom moiety can be linked to the 2′, 3′, 4′ or 5′ hydroxyl moiety of a sugar or modified sugar. Nucleotides that incorporate the modified nucleobases described above can also be used in the process disclosed herein. In some embodiments, nucleotides or modified nucleotides comprising an unprotected —OH moiety are used in the process disclosed herein.
  • In addition to the ribose moiety described in Schemes 1-4b, other modified sugars can also be incorporated in the nucleic acids disclosed herein. In some embodiments, the modified sugars contain one or more substituents at the 2′ position including one of the following: F; CF3, CN, N3, NO, NO2, O-, S-, or N-alkyl; O-, S-, or N-alkenyl; O-, S- or N-alkynyl; or O-alkyl-O-alkyl, O-alkyl-N-alkyl or N-alkyl-O-alkyl wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted C1-C10 alkyl or C2-C10 alkenyl and alkynyl. Examples of substituents include, and are not limited to, O(CH2)nOCH3, and O(CH2)nNH2, wherein n is from 1 to about 10, MOE, DMAOE, DMAEOE. Also contemplated herein are modified sugars described in WO 2001/088198; and Martin et al., Helv. Chim. Acta, 1995, 78, 486-504. In some embodiments, modified sugars comprise substituted silyl groups, an RNA cleaving group, a reporter group, a fluorescent label, an intercalator, a group for improving the pharmacokinetic properties of a nucleic acid, or a group for improving the pharmacodynamic properties of a nucleic acid, and other substituents having similar properties. The modifications may be made at the at the 2′, 3′, 4′, 5′, or 6′ positions of the sugar or modified sugar, including the 3′ position of the sugar on the 3′-terminal nucleotide or in the 5′ position of the 5′-terminal nucleotide.
  • Modified sugars also include sugar mimetics such as cyclobutyl or cyclopentyl moieties in place of the pentofuranosyl sugar. Representative United States patents that teach the preparation of such modified sugar structures include, but are not limited to, U.S. Pat. Nos. 4,981,957; 5,118,800; 5,319,080; and 5,359,044. Some modified sugars that are contemplated include:
  • Figure US20170029457A1-20170202-C00041
  • Other non-limiting examples of modified sugars include glycerol, which form glycerol nucleic acid (GNA) analogues. One example of a GNA analogue is shown below and is described in Zhang, R et al., J. Am. Chem. Soc., 2008, 130, 5846-5847; Zhang L, et al., J. Am. Chem. Soc., 2005, 127, 4174-4175 and Tsai C H et al., PNAS, 2007, 14598-14603:
  • Figure US20170029457A1-20170202-C00042
      • wherein X is as defined herein. Another example of a GNA derived analogue, flexible nucleic acid (FNA) based on the mixed acetal aminal of formyl glycerol, is described in Joyce G F et al., PNAS, 1987, 84, 4398-4402 and Heuberger B D and Switzer C, J. Am. Chem. Soc., 2008, 130, 412-413, and is shown below:
  • Figure US20170029457A1-20170202-C00043
  • Other non-limiting examples of modified sugars include hexopyranosyl (6′ to 4′), pentopyranosyl (4′ to 2′), pentopyranosyl (4′ to 3′), or tetrofuranosyl (3′ to 2′) sugars.
  • Hexopyranosyl (6′ to 4′) sugars contemplated include:
  • Figure US20170029457A1-20170202-C00044
  • Pentopyranosyl (4′ to 2′) sugars contemplated include:
  • Figure US20170029457A1-20170202-C00045
  • Pentopyranosyl (4′ to 3′) sugars contemplated include:
  • Figure US20170029457A1-20170202-C00046
  • Tetrofuranosyl (3′ to 2′) sugars contemplated include:
  • Figure US20170029457A1-20170202-C00047
  • Other modified sugars contemplated include:
  • Figure US20170029457A1-20170202-C00048
  • Further contemplated are the sugar mimetics illustrated below wherein X is selected from S, Se, CH2, N-Me, N-Et or N-iPr.
  • Figure US20170029457A1-20170202-C00049
    Figure US20170029457A1-20170202-C00050
  • The modified sugars and sugar mimetics can be prepared by methods known in the art, including, but not limited to: A. Eschenmoser, Science (1999), 284:2118; M. Bohringer et al, Helv. Chim. Acta (1992), 75:1416-1477; M. Egli et al, J. Am. Chem. Soc. (2006), 128(33):10847-56; A. Eschenmoser in Chemical Synthesis: Gnosis to Prognosis, C. Chatgilialoglu and V. Sniekus, Ed., (Kluwer Academic, Netherlands, 1996), p. 293; K.-U. Schoning et al, Science (2000), 290:1347-1351; A. Eschenmoser et al, Helv. Chim. Acta (1992), 75:218; J. Hunziker et al, Helv. Chim. Acta (1993), 76:259; G. Otting et al, Helv. Chim. Acta (1993), 76:2701; K. Groebke et al, Helv. Chim. Acta (1998), 81:375; and A. Eschenmoser, Science (1999), 284:2118.
  • Blocking Groups
  • In the reactions described, it is necessary in certain embodiments to protect reactive functional groups, for example hydroxy, amino, thiol or carboxy groups, where these are desired in the final product, to avoid their unwanted participation in the reactions. Protecting groups are used to block some or all reactive moieties and prevent such groups from participating in chemical reactions until the protective group is removed. In one embodiment, each protective group is removable by a different means. Protective groups that are cleaved under totally disparate reaction conditions fulfill the requirement of differential removal. In some embodiments, protective groups are removed by acid, base, and/or hydrogenolysis. Groups such as trityl, dimethoxytrityl, acetal and t-butyldimethylsilyl are acid labile and are used in certain embodiments to protect carboxy and hydroxy reactive moieties in the presence of amino groups protected with Cbz groups, which are removable by hydrogenolysis, and/or Fmoc groups, which are base labile. In other embodiments, carboxylic acid and hydroxy reactive moieties are blocked with base labile groups such as, but not limited to, methyl, ethyl, and acetyl in the presence of amines blocked with acid labile groups such as t-butylcarbamate or with carbamates that are both acid and base stable but hydrolytically removable.
  • In another embodiment, hydroxy reactive moieties are blocked with hydrolytically removable protective groups such as the benzyl group, while amine groups capable of hydrogen bonding with acids are blocked with base labile groups such as Fmoc. In another embodiment, carboxylic acid reactive moieties are protected by conversion to simple ester compounds, or they are, in yet another embodiment, blocked with oxidatively-removable protective groups such as 2,4-dimethoxybenzyl, while co-existing amino groups are blocked with fluoride labile silyl or carbamate blocking groups.
  • Allyl blocking groups are useful in the presence of acid- and base-protecting groups since the former are stable and can be subsequently removed by metal or pi-acid catalysts. For example, an allyl-blocked hydroxy groups can be deprotected with a Pd(O)-catalyzed reaction in the presence of acid labile t-butylcarbamate or base-labile acetate amine protecting groups. Yet another form of protecting group is a resin to which a compound or intermediate is attached. As long as the residue is attached to the resin, that functional group is blocked and cannot react. Once released from the resin, the functional group is available to react.
  • Typically blocking/protecting groups useful in the synthesis of the compounds described herein are, by way of example only:
  • Figure US20170029457A1-20170202-C00051
  • Representative protecting groups useful to protect nucleotides during synthesis include base labile protecting groups and acid labile protecting groups. Base labile protecting groups are used to protect the exocyclic amino groups of the heterocyclic nucleobases. This type of protection is generally achieved by acylation. Three commonly used acylating groups for this purpose are benzoyl chloride, phenoxyacetic anhydride, and isobutyryl chloride. These protecting groups are stable to the reaction conditions used during nucleic acid synthesis and are cleaved at approximately equal rates during the base treatment at the end of synthesis.
  • In some embodiments, the 5′-protecting group is trityl, monomethoxy trityl, dimethoxytrityl, trimethoxytrityl, 2-chlorotrityl, DATE, TBTr, 9-phenylxanthine-9-yl (Pixyl), or 9-(p-methoxyphenyl)xanthine-9-yl (MOX).
  • In some embodiments, thiol moieties are incorporated in the compounds described herein and are protected. In some embodiments, the protecting groups include, but are not limited to, pixyl, trityl, benzyl, p-methoxybenzyl (PMB), or tert-butyl (t-Bu).
  • Other protecting groups, plus a detailed description of techniques applicable to the creation of protecting groups and their removal are described in Greene and Wuts, Protective Groups in Organic Synthesis, 3rd Ed., John Wiley & Sons, New York, N.Y., 1999, and Kocienski, Protective Groups, Thieme Verlag, New York, N.Y., 1994, which are incorporated herein by reference for such disclosure.
  • The examples provided below further illustrate and exemplify the compounds of the present invention and methods of preparing such compounds. It is to be understood that the scope of the present invention is not limited in any way by the scope of the following examples and preparations.
  • EXAMPLES Example 1 Synthesis of Methanethiosulfonate Reagents
  • Figure US20170029457A1-20170202-C00052
    Figure US20170029457A1-20170202-C00053
    Figure US20170029457A1-20170202-C00054
    Figure US20170029457A1-20170202-C00055
  • Compound 2:
  • A solution of (Z)-but-2-ene-1,4-diol (0.93 ml, 11.3 mmol) and triethylamine (3.3 ml, 24 mmol) in DCM (50 mL) was added in a dropwise fashion to a stirring ice cold solution of methanesulfonyl chloride (1.9 ml, 24 mmol) in DCM (50 mL). After stirring for 0.5 h at r.t. the mixture was poured onto ice and extracted. The organic layer was collected, dried (MgSO4), filtered and reduced to 2.66 g, 96% of compound 2, which was judged by NMR to be sufficiently pure for direct use in the next step of the reaction.
  • 1H NMR (399 MHz, CDCl3) δ 5.94 (ddd, J=5.4, 4.1, 1.3 Hz, 2H), 4.83 (dd, J=4.1, 1.3 Hz, 4H), 3.04 (s, 6H); 13C NMR 128.34, 64.38, 38.27; MS (ESI+ve): calc (M+NH4): 262.04. found: 262.05. Rf=0.3 (1:1 EtOAc/hexane).
  • Compound 3:
  • A solution of sodium methanesulfonothioate (1.51 g, 11.3 mmol) in MeOH (20 ml) was treated with neat (Z)-but-2-ene-1,4-diyl dimethanesulfonate (1.25 g, 5.12 mmol) at r.t. After 5 min, precipitation was observed to occur. After 36 h, the mixture was partitioned between water and DCM. The organic layer was separated, dried (MgSO4), filtered and reduced to afford a colorless oil. Column chromatography (ISCO) gave the pure product as a pale colorless oil. Column chromatography gave pure compound 3 (0.89 g, 63%) as a colorless oil.
  • 1H NMR (399 MHz, CDCl3) δ 5.84 (ddd, J=6.6, 5.1, 1.5 Hz, 2H), 3.92 (dd, J=5.1, 1.5 HZ, 4H), 3.33 (s, 6H); 13C NMR 128.1, 51.47, 33.13; MS (ESI+ve): calc (M+NH4): 294.00. found: 294.04. Rf=0.4 (1:1 EtOAc/hexane).
  • Compound 4:
  • Under argon atmosphere, morpholine (10 g, 115 mmol) was added to ethylene sulfide (15 g, 250 mmol) in a round bottom flask. The reaction was stirred for 7 hrs and was directly loaded on to a silica gel column. The column was washed with DCM first and then 2% MeOH/DCM was used to obtain compound 4 (15.3 g, 91%) as colorless oil.
  • 1H NMR (399 MHz, CDCl3) δ 3.67-3.59 (m, 4H), 2.63-2.52 (m, 2H), 2.51-2.45 (m, 2H), 2.44-2.34 (m, 4H); MS (ESI+ve): calc (M+H)+=148.07. found: 148.1.
  • Compound 5:
  • A DCM solution (1 mL) of 2-morpholinoethanethiol (0.21 g, 1.44 mmol) was added dropwise via syringe to a stirring solution compound 3 (0.40 g, 1.44 mmol) in DCM (10 mL) at r.t. Immediately after addition, the TLC was checked, to reveal rapid formation of product and some quantity of dimer. After 0.5 h, the mixture was partitioned by addition of water. Upon extraction, the organic layer was separated then dried (MgSO4), filtered and reduced in vacuo. Column chromatography gave compound 5 (0.29 g, 58%) as colorless oil.
  • 1H NMR (399 MHz, CDCl3) δ 5.78 (m, 2H), 3.92 (d, J=7.3 Hz, 2H), 3.70 (t, J=4.7 Hz, 4H), 3.46 (d, J=5.5 Hz, 2H), 3.31 (s, 3H), 2.84 (dd, J=7.8, 6.7 Hz, 2H), 2.66 (dd, J=7.8, 6.7, 2H), 2.48 (t, J=4.6 Hz, 4H); 13C NMR 130.35, 126.27, 66.97, 58.20, 53.67, 51.52, 36.22, 35.16, 33.67; MS (ESI+ve): calc (M+H): 344.05. found: 344.06. Rf=0.3 (EtOAc).
  • Compound 5b:
  • A DCM solution (1 mL) of compound 4b (395 mg, 1.085 mmol) was added dropwise via syringe to a stirring DCM (15 mL) solution compound 3 (300 mg, 1.085 mmol) at r.t. After 1 h, the resulting solution was partitioned by addition of water. Upon extraction, the organic layer was separated then dried (MgSO4), filtered and reduced in vacuo. Column chromatography gave compound 5b as a colorless oil (0.35 g, 58%). 1H NMR (399 MHz, CDCl3) δ 5.83-5.70 (m, 2H), 5.35-5.21 (dt, J=26.0, 9.3 Hz, 2H), 5.16-5.07 (m, 1H), 4.59-4.54 (d, J=9.5 Hz, 1H), 4.29-4.23 (m, 1H), 4.23-4.18 (m, 1H), 3.99-3.88 (dd, J=6.7, 1.2 Hz, 2H), 3.80-3.72 (ddd, J=10.1, 4.6, 2.6 Hz, 1H), 3.64-3.56 (m, 1H), 3.50-3.43 (m, 1H), 3.31 (s, 3H), 2.09 (s, 3H), 2.03 (s, 6H), 2.00 (s, 3H); 13C NMR (100 MHz, CDCl3) δ 170.68, 170.30, 169.51, 169.30, 129.43, 127.14, 87.73, 76.49, 73.89, 69.16, 67.99, 61.99, 51.64, 35.89, 33.58, 20.95, 20.80, 20.74, 20.71; MS (ESI+ve): calc (M+NH4 +): 578.07. found: 577.96. Rf=0.5 (1:1 EtOAc/hexane).
  • Compound 6:
  • An ice cold solution of (Z)-but-2-ene-1,4-diol (0.93 ml, 11.3 mmol) and triethylamine (1.6 mL, 11.5 mmol) in DCM (50 ml) was treated dropwise via syringe with pivaloyl chloride (1.4 ml, 11.4 mmol) over 2 min. After 1 h, TLC showed good reaction.
  • The resulting mixture was partitioned by addition of water. Upon extraction, the organic layer was separated then dried (MgSO4), filtered and reduced in vacuo. This crude compound was found: by TLC (Rf=0.6, 1:1 EtOAc/hexane) to contain no starting diol and was used crude to prepare the mesylate. The crude material was taken up in DCM (50 ml) containing triethylamine (1.7 mL, 12 mmol) and cooled on an ice bath. Methanesulfonyl chloride (0.98 ml, 12.66 mmol) was added dropwise via syringe over 2 min. TLC immediately after addition indicated complete consumption of starting material. The resulting mixture was partioned by addition of water. Upon extraction, the organic layer was separated then dried (MgSO4), filtered and reduced in vacuo. Column chromatography gave pure compound 6, 1.48 g, 52%, as a colorless oil.
  • 1H NMR (399 MHz, CDCl3) δ 5.89-5.75 (m, 2H), 4.89-4.84 (d, J=5.7 Hz, 2H), 4.68-4.63 (d, J=5.9 Hz, 2H), 3.03 (s, 3H), 1.19 (s, 9H); 13C NMR (100 MHz, CDCl3) δ 178.28, 130.61, 126.11, 65.08, 59.65, 38.84, 38.21, 27.25; MS (ESI+ve): calc (M+NH4): 268.12. found: 268.20. Rf=0.3 (20% EtOAc/hexane).
  • Compound 7:
  • A MeOH (10 ml) solution of sodium methanesulfonothioate (0.63 g, 4.70 mmol) and (Z)-4-(methylsulfonyloxy)but-2-enyl pivalate (1.00 g, 4.00 mmol) was stirred at r.t. for 18 h with formation of a white precipitate (after 10 min). The resulting mixture was partitioned by addition of water and DCM. Upon extraction into DCM, the organic layer was separated then dried (MgSO4), filtered and reduced in vacuo. Column chromatography gave compound 7, 0.83 g, 78% as a colorless oil.
  • 1H NMR (399 MHz, CDCl3) δ 5.82-5.73 (m, 2H), 4.73-4.66 (m, 2H), 3.95-3.87 (m, 2H), 3.32 (s, 3H), 1.19 (s, 9H); 13C NMR (100 MHz, CDCl3) δ 178.35, 129.37, 127.32, 59.50, 51.44, 38.84, 33.61, 27.28; MS (ESI+ve): calc (M+NH4): 284.10. found: 284.19. Rf=0.4 (20% EtOAc/hexane).
  • Compound 9:
  • Pivaloyl chloride (0.60 g, 5.0 mmol) was added in a dropwise fashion to a stirring solution of S-2-hydroxyethyl methanesulfonothioate (0.65 g, 4.16 mmol) in DCM (20 ml). After 2 h at r.t. the resulting mixture with white precipitate was partitioned with water. The organic layer was separated, dried (Ns2SO4), filtered and reduced to an oil. Column gave compound 9 as a colorless oil (0.45 g, 45%). 1H NMR (399 MHz, CDCl3) δ 4.39-4.34 (t, J=6.3 Hz, 2H), 3.44-3.39 (t, J=6.3 Hz, 2H), 3.36 (s, 3H), 1.20 (s, 9H); 13C NMR (100 MHz, CDCl3) δ 62.10, 51.11, 38.96, 35.19, 27.24; MS (ESI+ve): calc (M+NH4): 158.08. found: 158.04. Rf=0.3 (20% EtOAc/hexane).
  • Compound 11:
  • Pivaloyl chloride (4.96 ml, 40.3 mmol) was added dropwise via syringe to an ice cold DCM solution (50 mL) of 2-(hydroxymethyl)phenol (5 g, 40.3 mmol) and triethylamine (5.61 ml, 40.3 mmol). An ice-cold solution of the crude pivalate ester was treated with triethylamine (6.74 ml, 48.4 mmol) and 50 mL DCM. Methanesulfonyl chloride (3.43 ml, 44.3 mmol) was then added slowly (5 min) via syringe and the resulting mixture was warmed to r.t. The mixture was poured onto ice and the organic layer was separated then washed with sat NaHCO3 (aq), dried (MgSO4), filtered and reduced to 10.5 g crude pale yellow oil.
  • Column (ISCO) gave pure 11 5.45 g, 47%.
  • 1H NMR (399 MHz, CDCl3) δ 7.53-7.46 (dd, 7.7, 1.8 Hz, 1H), 7.46-7.40 (dt, 7.7, 1.8 Hz, 1H), 7.32-7.24 (t, 7.7 Hz, 1H), 7.13-7.06 (d, 7.7 Hz, 1H), 5.21 (s, 2H), 2.79 (s, 3H), 1.40 (s, 9H); 13C NMR (100 MHz, CDCl3) δ 177.05, 150.06, 131.18, 131.07, 126.35, 125.94, 123.21, 66.88, 39.48, 38.82, 27.30, 27.26. MS (ESI+ve): calc (M+NH4): 304.12. found: 303.99. Rf=0.4 (20% EtOAc/hexane).
  • Compound 12:
  • A MeOH (20 mL) solution of sodium methanesulfonothioate (0.825 g, 6.15 mmol) was treated with 2-((methylsulfonyloxy)methyl)phenyl pivalate (1.76 g, 6.15 mmol) at r.t. and left to stir for 18 h. The mixture was partitioned between water and DCM. The organic layer was separated, dried (MgSO4), filtered and reduced to afford a colorless oil. Column chromatography gave pure compound 12 as a pale colorless oil, 0.754 g, 41%.
  • 1H NMR (399 MHz, CDCl3) δ 7.48-7.44 (dd, J=7.7, 1.7 Hz, 1H), 7.39-7.34 (td, J=7.8, 1.7 Hz, 1H), 7.25-7.20 (td, J=7.6, 1.2 Hz, 1H), 7.10-7.06 (dd, J=8.2, 1.2 Hz, 1H), 4.29 (s, 2H), 2.90 (s, 3H), 1.39 (s, 9H); 13C NMR (100 MHz, CDCl3) δ 176.69, 149.59, 131.17, 129.85, 127.41, 126.18, 123.40, 51.43, 39.47, 36.01, 27.30; MS (ESI+ve): calc (M+NH4): 320.10. found: 320.09. Rf=0.4 (20% EtOAc/hexane).
  • Compound 14:
  • Chloromethyl pivalate (0.478 ml, 3.32 mmol) was added to a stirring mixture of sodium iodide (0.050 g, 0.33 mmol) and sodium methanesulfonothioate (0.445 g, 3.32 mmol) in acetone (7 ml) at r.t. After 24 h, TLC showed good conversion to product. The solvent was removed, and the residue was partitioned between water and DCM. The organic layer was separated and dried (MgSO4), filtered and reduced to afford a colorless oil. Column chromatography gave pure 14 as a slightly pink solid, 0.41 g, 55%.
  • 1H NMR (399 MHz, CDCl3) δ 5.67 (s, 2H), 3.39 (s, 3H), 1.24 (s, 9H); 13C NMR (100 MHz, CDCl3) δ 177.35, 67.84, 52.20, 38.93, 27.05. Rf=0.5 (20% EtOAc/hexane).
  • Compound 16:
  • Prepared from 15 and NaMTS as described previously: U.S. Pat. No. 3,484,473 1H NMR (399 MHz, CDCl3) δ 4.86 (s, 2H), 3.45 (s, 6H); 13C NMR (100 MHz, CDCl3) δ 52.15, 41.50.
  • Compound 18:
  • Prepared from 17 and NaMTS as described previously: Chem. Pharm. Bull. Vol. 12(11) p. 1271, 1964.
  • 1H NMR (399 MHz, CDCl3) δ 3.55 (s, 4H), 3.40 (s, 6H); 13C NMR (100 MHz, CDCl3) δ 50.67, 35.96.
  • Compound 19:
  • A DCM solution (1 mL) of 2-morpholinoethanethiol (0.17 g, 1.2 mmol) was added dropwise via syringe to a stirring solution of compound 18 (300 mg, 1.2 mmol) in DCM (10 mL) at r.t. Immediately after addition, the TLC was checked, to reveal rapid formation of product and some dimer. After 0.5 h, the mixture was partitioned by addition of NaHCO3. Upon extraction, the organic layer was separated then dried (MgSO4), filtered and reduced in vacuo. Column chromatography gave pure 19 (0.20 g, 53%) as a colorless oil. 1H NMR (399 MHz, CDCl3) δ 3.73-3.67 (t, J=4.7 Hz, 4H), 3.51-3.46 (m, 2H), 3.35 (s, 3H), 3.07-3.01 (m, 2H), 2.88-2.83 (m, 2H), 2.69-2.63 (m, 2H), 2.52-2.43 (t, J=4.6 Hz, 4H); 13C NMR (100 MHz, CDCl3) δ 66.96, 57.91, 53.58, 50.79, 37.66, 36.10, 35.52; MS (ESI+ve): calc (M+H): 318.03. found: 318.04. Rf=0.3 (EtOAc).
  • Compound 21:
  • Compound 20 is converted to compound 21 by a procedure analogous to that described for compound 11.
  • Compound 22:
  • Compound 21 is converted to compound 22 by a procedure analogous to that described for compound 12.
  • Compound 23:
  • Compound 23 is prepared according to a literature method (Journal of Medicinal Chemistry, 50(23), 5568-5570; 2007.)
  • Compound 24:
  • An ice-cold pyridine solution (10 mL) of compound 23 (1 mmol) is treated successively, in a dropwise fashion with acetyl chloride (1 mmol), then after 5 min with MsCl (1.1 mmol). The solution is warmed to room temperature then the solvent is removed. The residue is dissolved in EtOAc, washed with water, dried (MgSO4), filtered and reduced in vacuo. Purification by column chromatography affords pure compound 24.
  • Compound 25:
  • Compound 24 is converted to compound 25 by a procedure analogous to that described for compound 12.
  • Compound 27:
  • Compound 26 is converted to compound 27 by a procedure analogous to that described for compound 14.
  • Compound 29:
  • Compound 28 is converted to compound 29 by a procedure analogous to that described for compound 14.
  • Compound 30:
  • Compound 30 is prepared according to a literature method (Tetrahedron, 42(2), 601-7; 1986.)
  • Compound 31:
  • Compound 31 is prepared from compound 30 according to a patent procedure (US 20090181444)
  • Compound 33:
  • Compound 33 is prepared from compound 32 according to a patent procedure (US 20090181444)
  • Compound 36:
  • An ice-cold DCM (20 mL) solution of compound 34 (1 mmol) is treated with NEt3 (1 mmol) followed by the dropwise addition of TMS-Cl (1.1 mmol). After 1 h, the solution is washed with water, dried (MgSO4), filtered and reduced in vacuo. The crude TMS protected material is redissolved in THF (10 mL), whereon PPh3 (1.2 mmol), compound 35 (1.2 mmol), then DEAD (1.2 mmol, dropwise) are added in succession. After stirring at r.t. for 18 h, the solvent is removed under vacuum, the residue is redissolved in DCM, the solution of which is washed with water, dried (MgSO4), filtered and reduced in vacuo. Purification by column chromatography affords pure compound 36.
  • Compound 37:
  • A THF (10 mL) solution of compound 36 (0.5 mmol) is treated with TBAF (1 mmol of a 1M solution in THF), with monitoring by TLC. On completion of TMS cleavage, the solvent is removed under vacuum, the residue is redissolved in DCM, the solution of which is washed with water, dried (MgSO4), filtered and reduced in vacuo. The crude alcohol is redissolved in pyridine (5 mL), and TsCl (0.55 mmol) is added. After 18 h at r.t., the solvent is removed, the residue is redissolved in DCM, the solution of which is washed with water, dried (MgSO4), filtered and reduced in vacuo. Purification by column chromatography affords pure compound 37.
  • Compound 38:
  • Compound 37 is converted to compound 38 by a procedure analogous to that described for compound 12.
  • Compound 40:
  • An ice-cold DCM (20 mL) solution of compound 39 (1 mmol) is treated with NEt3 (1 mmol) followed by the dropwise addition of TMS-Cl (1.1 mmol). After 1 h, the solution is washed with water, dried (MgSO4), filtered and reduced in vacuo. The crude TMS protected material is redissolved in THF (10 mL), whereon PPh3 (1.2 mmol), potassium p-toluenethiosulfonate (KTTS, 1.2 mmol), anhydrous ZnCl2 (1 mmol) then DEAD (1.2 mmol, dropwise) are added in succession. After stirring at r.t. for 18 h, the solvent is removed under vacuum, the residue is redissolved in DCM, the solution of which is washed with water, dried (MgSO4), filtered and reduced in vacuo. Purification by column chromatography affords pure compound 40.
  • Compound 41:
  • A THF (10 mL) solution of compound 40 (0.5 mmol) is treated with TBAF (1 mmol of a 1M solution in THF), with monitoring by TLC. On completion of TMS cleavage, the solvent is removed under vacuum, the residue is redissolved in DCM, the solution of which is washed with water, dried (MgSO4), filtered and reduced in vacuo. The crude alcohol is redissolved in THF (10 mL), whereon PPh3 (1.2 mmol), compound 35 (1.2 mmol), then DEAD (1.2 mmol, dropwise) are added in succession. After stirring at r.t. for 18 h, the solvent is removed under vacuum, the residue is redissolved in DCM, the solution of which is washed with water, dried (MgSO4), filtered and reduced in vacuo. Purification by column chromatography affords pure compound 40.
  • Compound 42:
  • Compound 41 is converted to compound 42 by a procedure analogous to that described for compound 14.
  • Example 2 Thioalkylation of H-Phosphonates to Provide Phosphorothiotriesters in Solution Phase
  • Figure US20170029457A1-20170202-C00056
    Figure US20170029457A1-20170202-C00057
    Figure US20170029457A1-20170202-C00058
  • Compound 100:
  • The synthetic procedure for Di-DMTr H-phosphonate TT dimer (100) has been previously described (Froehler, Brian C.; Ng, Peter G.; Matteucci, Mark D., Nucleic Acids Research (1986), 14(13), 5399-5407; Garegg, Per J.; Lindh, Ingvar; Regberg, Tor; Stawinski, Jacek; Stroemberg, Roger; Henrichson, Christina Tetrahedron Letters (1986), 27(34), 4051-4054).
  • Compound 101:
  • Compound 100, mixture of diastereomers (200 mg, 0.176 mmol) was dissolved in ACN (6 mL) then trimethylsilyl 2,2,2-trifluoro-N-(trimethylsilyl)acetamidate (227 mg, 0.882 mmol) was added. A solution of (Z)—S-4-((2-morpholinoethyl)disulfanyl)but-2-enyl methanesulfonothioate (121 mg, 0.353 mmol) in ACN (2 mL) was then added, over the course of 1 h in 3 approximately equal portions, with monitoring by TLC and HPLC/MS. After 3 h, the resulting solution was partitioned by addition of water. Upon extraction, the organic layer was separated then dried (MgSO4), filtered and reduced in vacuo. Column chromatography gave compound 101 as a white foam, 225 mg, 91%.
  • 1H NMR (399 MHz, CDCl3) δ 9.72 (d, br, 1H), 9.27, (d, br, 1H), 7.53 (dd, J=25.0, 1 Hz, 1H), 7.42, (t, J=7.0 Hz, 2H), 7.37-7.16 (m, 17H), 6.83 (m, 8H), 6.43-6.28 (m, 2H), 5.63-5.42 (m, 2H), 5.21 (q, J=7.1 Hz, 1H), 4.27 (m, br, 1H), 3.94 (m, br, 2H), 3.77 (m, 12H), 3.74-3.60 (m, 6H), 3.51-3.22 (m, 5H), 2.82-2.76 (m, 2H), 2.68-2.60 (m, 2H), 2.59-2.46 (m, 5H), 2.44-2.33 (m, 2H), 2.03-1.88 (m, 1H), 1.84 (m, 3H), 1.75-1.66 (m, 1H), 1.48-1.32 (dd, J=11.8, 1.2 Hz, 3H); 13C NMR (100 MHz, CDCl3) δ 164.10, 164.07, 164.00, 163.94, 159.14, 159.10, 150.80, 150.78, 150.75, 150.63, 145.09, 144.30, 144.27, 136.31, 136.27, 136.22, 136.18, 135.95, 135.82, 135.43, 135.35, 135.33, 135.24, 135.22, 130.52, 130.43, 130.40, 129.49, 129.30, 128.54, 128.43, 128.39, 127.64, 127.57, 113.78, 113.76, 113.73, 113.67, 112.05, 111.56, 87.77, 87.66, 87.58, 85.77, 85.59, 84.63, 84.51, 74.42, 74.33, 67.02, 66.95, 63.63, 63.49, 58.27, 58.23, 55.60, 55.58, 53.69, 53.62, 39.48, 39.26, 39.18, 35.88, 35.61, 35.43, 35.36, 28.18, 12.83, 12.79, 12.02, 11.95.; 31P NMR (162 MHz, CDCl3) δ 29.25, 29.12; MS (ESI+ve): calc (M+H): 1398.46. found: 1398.64. Rf=0.4 (5% MeOH/DCM).
  • Compound 201:
  • Compound 101 (0.150 g, 0.107 mmol) was stirred with 3% TCA/DCM (10 mL) over 10 min. TLC and HPLC/MS showed that the reaction was complete. 10 mL of MeOH was added and stirring continued for 2 min. Solvents were evaporated and the residue was purified by column chromatography to give compound 201 (85 mg, 100%) as a white solid.
  • 1H NMR (399 MHz, CD3OD) δ 7.78 (dd, J=7.2, 1.3 Hz, 1H), 7.53 (d, J=1.3 Hz, 1H), 6.33-6.27 (m, 2H), 5.83-5.70 (m, 2H), 5.25-5.19 (m, 1H), 4.47-4.30 (m, 3H), 4.27-4.22 (m, 1H), 4.11-4.05 (m, 1H), 3.89-3.82 (t, J=4.8 Hz, 4H), 3.85 (m, 2H), 3.76-3.70 (ddd, J=15.5, 7.2, 1.7 Hz, 2H), 3.52 (dd, J=7.3, 3.7 Hz, 2H), 3.28-3.19 (br, 2H), 3.16-3.05 (br, 4H), 3.05-2.98 (ddd, J=9.8, 5.5, 2.0 Hz, 2H), 2.62-2.52 (tdd, J=11.5, 5.7, 1.9 Hz, 1H), 2.47-2.36 (m, 1H), 2.33-2.28 (m, 2H), 1.92-1.87 (m, 6H); 31P NMR (162 MHz, CD3OD) δ 30.22, 30.19; MS (ESI+ve): calc (M+H): 794.20. found: 794.18. Rf=0.3 (10% MeOH/DCM).
  • Compound 102:
  • Compound 100 (400 mg, 0.352 mmol) was converted to compound 102 by a procedure analogous to that described for compound 101 (417 mg, 90%).
  • 1H NMR (399 MHz, CDCl3) δ 9.17 (d, J=6.0 Hz, 1H), 9.13-9.00 (d, J=25.7 Hz, 1H), 7.58-7.49 (dd, J=26.3, 1.5 Hz, 1H), 7.45-7.40 (ddd, J=8.0, 5.2, 1.3 Hz, 2H), 7.40-7.18 (m, 17H), 6.87-6.81 (m, 8H), 6.44-6.30 (m, 2H), 5.65-5.53 (m, 1H), 5.53-5.44 (m, 1H), 5.26-5.16 (quintet, J=6.4 Hz, 1H), 4.61-4.54 (m, 2H), 4.30-4.24 (m, 1H), 4.19-4.13 (m, 1H), 3.97-3.88 (m, 2H), 3.80-3.72 (m, 12H), 3.69-3.57 (m, 1H), 3.54-3.30 (m, 5H), 2.61-2.49 (dt, J=14.4, 5.4 Hz, 1H), 2.44-2.32 (m, 1H), 2.02-1.91 (dt, J=12.5, 5.4 Hz, 1H), 1.85-1.80 (dd, J=5.0, 1.3 Hz, 3H), 1.76-1.63 (m, 1H), 1.43-1.36 (dd, J=10.2, 1.2 Hz, 3H), 1.19-1.14 (d, J=2.0 Hz, 8H); 13C NMR (100 MHz, CDCl3) δ 178.22, 178.17, 163.82, 163.80, 163.75, 158.92, 158.88, 150.52, 150.43, 144.90, 144.88, 144.10, 144.05, 136.11, 136.08, 136.05, 136.01, 135.59, 135.28, 135.16, 135.03, 135.01, 130.30, 130.23, 130.19, 130.16, 128.69, 128.64, 128.59, 128.39, 128.34, 128.23, 128.21, 128.17, 127.42, 127.34, 113.54, 113.45, 111.85, 111.82, 111.41, 111.36, 87.59, 87.43, 87.37, 85.47, 85.33, 84.43, 84.29, 84.08, 84.00, 83.92, 74.24, 67.36, 63.38, 63.26, 59.42, 55.37, 39.22, 38.77, 27.94, 27.24, 12.57, 11.80, 11.74; 31P NMR (162 MHz, CDCl3) δ 29.23, 28.97; MS (ESI+ve): calc (M+H): 1338.51. found: 1338.84. Rf=0.5 (5% MeOH/DCM).
  • Compound 202:
  • Compound 102 (200 mg, 0.151 mmol) was converted to compound 202 by a procedure analogous to that described for compound 101 (105 mg, 97%).
  • 1H NMR (399 MHz, CD3OD) δ 7.81-7.75 (dd, J=8.2, 1.3 Hz, 1H), 7.57-7.51 (dd, J=8.2, 1.3 Hz, 1H), 6.33-6.23 (m, 2H), 5.85-5.75 (m, 1H), 5.75-5.66 (m, 1H), 5.26-5.19 (m, 1H), 4.72-4.66 (m, 2H), 4.47-4.30 (m, 3H), 4.27-4.20 (m, 1H), 4.11-4.04 (m, 1H), 3.83-3.76 (m, 2H), 3.74-3.64 (m, 2H), 2.62-2.51 (m, 1H), 2.45-2.35 (td, J=8.7, 6.5 Hz, 1H), 2.32-2.24 (m, 2H), 1.93-1.82 (m, 6H), 1.20-1.15 (d, J=2.1 Hz, 9H); 13C NMR (126 MHz, CD3OD) δ 179.65, 166.28, 152.30, 152.28, 152.22, 137.90, 137.81, 137.79, 130.07, 130.04, 129.26, 129.24, 111.93, 111.88, 111.87, 87.26, 87.22, 86.96, 86.90, 86.76, 86.54, 86.12, 86.07, 85.98, 85.92, 85.88, 85.82, 80.54, 80.49, 80.46, 80.41, 71.84, 71.67, 68.71, 68.66, 68.45, 68.40, 62.58, 62.50, 60.72, 40.51, 40.44, 39.70, 39.52, 39.48, 28.67, 28.64, 28.61, 27.53, 12.64, 12.48; 31P NMR (162 MHz, CDCl3) δ 29.23, 28.97; MS (ESI+ve): calc (M+H): 717.22. found: 717.23. Rf=0.5 (10% MeOH/DCM).
  • Compound 103:
  • Compound 100 (400 mg, 0.352 mmol) was converted to compound 103 by a procedure analogous to that described for compound 101 (379 mg, 83%).
  • 1H NMR (399 MHz, CDCl3) δ 9.48 (s, 1H), 9.41-9.29 (m, 1H), 7.60-7.48 (dd, J=9.0, 1.0 Hz, 1H), 7.46-7.40 (dt, J=6.9, 1.2 Hz, 2H), 7.39-7.17 (m, 17H), 6.89-6.79 (m, 8H), 6.44-6.31 (m, 2H), 5.27-5.20 (t, J=6.5 Hz, 1H), 4.30-4.24 (t, J=6.1 Hz, 1H), 4.19-4.15 (m, 2H), 4.13-4.07 (t, J=7.1 Hz, 1H), 3.99-3.90 (m, 2H), 3.79-3.74 (m, 12H), 3.70-3.58 (m, 1H), 3.51-3.43 (td, J=8.8, 7.2, 2.3 Hz, 1H), 3.40-3.32 (m, 1H), 3.02-2.85 (m, 2H), 2.61-2.49 (dt, J=18.5, 7.0 Hz, 1H), 2.47-2.33 (m, 1H), 1.98-1.90 (dt, J=10.2, 5.0 Hz, 1H), 1.85-1.81 (m, 3H), 1.74-1.62 (td, J=14.2, 7.1 Hz, 1H), 1.42-1.36 (m, 3H), 1.19-1.13 (d, J=4.9 Hz, 9H); 31P NMR (162 MHz, CDCl3) δ 29.36, 29.18; 13C NMR (126 MHz, CDCl3) δ 177.97, 177.89, 163.94, 163.91, 163.90, 163.86, 158.91, 158.87, 150.63, 150.54, 150.53, 150.50, 144.88, 144.85, 144.10, 144.04, 136.09, 135.99, 135.52, 135.50, 135.24, 135.16, 135.12, 135.04, 135.00, 130.31, 130.29, 130.20, 130.16, 130.13, 128.34, 128.20, 128.18, 128.14, 127.39, 127.31, 124.89, 113.55, 113.52, 113.43, 111.84, 111.38, 87.58, 87.42, 87.36, 85.30, 84.98, 84.95, 84.40, 84.33, 84.27, 83.98, 83.91, 83.84, 79.31, 79.27, 78.88, 78.84, 74.16, 74.08, 67.56, 67.50, 67.46, 67.41, 63.33, 63.24, 62.79, 62.75, 55.34, 39.21, 39.16, 39.04, 39.00, 38.85, 38.82, 29.95, 29.92, 29.66, 29.63, 27.17, 12.53, 11.80, 11.72; MS (ESI+ve): calc (M+H): 1312.69. found: 1312.49. Rf=0.4 (5% MeOH/DCM).
  • Compound 203:
  • Compound 103 (200 mg, 0.154 mmol) was converted to compound 203 by a procedure analogous to that described for compound 201 (103 mg, 98%).
  • 1H NMR (399 MHz, CD3OD) δ 7.80-7.76 (dd, J=8.2, 1.2 Hz, 1H), 7.55-7.51 (dd, 7.1, 1.2 Hz, 1H), 6.32-6.24 (m, 2H), 5.26-5.19 (m, 1H), 4.46-4.20 (m, 6H), 4.10-4.05 (m, 1H), 3.82-3.78 (dd, J=6.5, 3.2 Hz, 2H), 3.22-3.14 (ddd, J=16.6, 7.0, 5.8 Hz, 2H), 2.61-2.51 (tdd, J=13.0, 5.9, 2.1 Hz, 1H), 2.46-2.37 (ddd, J=14.3, 8.3, 6.0 Hz, 1H), 2.31-2.26 (t, J=5.8 Hz, 2H), 1.91-1.86 (dt, J=11.0, 1.2 Hz, 6H), 1.21-1.17 (m, 9H); 31P NMR (162 MHz, CD3OD) δ 30.15; 13C NMR (100 MHz, CD3OD) δ 179.45, 179.42, 166.29, 152.31, 152.29, 152.23, 137.82, 137.80, 137.78, 111.91, 111.88, 87.21, 87.17, 86.94, 86.87, 86.63, 86.52, 86.11, 86.06, 85.92, 85.84, 85.77, 80.67, 80.60, 80.49, 80.43, 71.79, 71.64, 68.80, 68.74, 68.58, 68.52, 64.11, 64.07, 64.02, 62.54, 62.44, 40.48, 40.43, 39.81, 39.71, 39.68, 39.52, 39.47, 30.74, 30.72, 30.68, 27.52, 12.65, 12.50; MS (ESI+ve): calc (M+H): 691.21. found: 691.09. Rf=0.5 (10% MeOH/DCM).
  • Compound 104:
  • Compound 100 (400 mg, 0.352 mmol) was converted to compound 104 by a procedure analogous to that described for compound 101 (451 mg, 94%).
  • 1H NMR (399 MHz, CDCl3) δ 9.17-9.01 (m, 2H), 7.51-7.46 (dd, J=7.8, 1.5 Hz, 1H), 7.45-7.38 (m, 2H), 7.37-7.09 (m, 19H), 7.01-6.90 (m, 2H), 6.87-6.78 (m, 8H), 6.39-6.27 (m, 2H), 5.15-5.01 (m, 1H), 4.20-4.13 (m, 1H), 3.96-3.90 (m, 1H), 3.90-3.83 (m, 2H), 3.80-3.68 (m, 14H), 3.52-3.20 (m, 3H), 2.45-2.16 (m, 2H), 2.01-1.88 (ddd, J=23.3, 13.6, 5.6 Hz, 1H), 1.85-1.79 (dd, J=9.3, 1.2 Hz, 3H), 1.69-1.53 (m, 1H), 1.40-1.31 (m, 12H); 13C NMR (100 MHz, CDCl3) δ 176.46, 176.37, 163.84, 163.78, 158.90, 158.87, 150.52, 150.50, 150.43, 149.38, 149.28, 144.95, 144.88, 144.16, 144.10, 136.13, 136.11, 136.09, 136.03, 135.57, 135.49, 135.37, 135.26, 135.21, 135.08, 135.04, 130.83, 130.74, 130.29, 130.21, 130.16, 129.51, 129.49, 129.40, 129.36, 129.35, 129.31, 128.38, 128.35, 128.27, 128.23, 128.19, 128.14, 127.39, 127.33, 126.05, 125.94, 122.94, 122.86, 113.53, 113.42, 111.77, 111.73, 111.39, 111.28, 87.55, 87.52, 87.37, 87.32, 85.33, 84.95, 84.90, 84.29, 84.20, 84.00, 83.92, 83.87, 83.79, 79.05, 79.00, 74.29, 74.24, 67.31, 67.24, 67.17, 67.11, 63.37, 55.37, 55.35, 39.37, 39.32, 39.15, 39.10, 38.64, 30.51, 30.41, 30.36, 27.28, 27.24, 12.59, 12.51, 11.75, 11.67; 31P NMR (162 MHz, CDCl3) δ 29.12, 28.49; MS (ESI+ve): calc (M+NH4): 1374.51. found: 1374.74. Rf=0.4 (5% MeOH/DCM).
  • Compound 204:
  • Compound 104 (200 mg, 0.147 mmol) was converted to compound 204 by a procedure analogous to that described for compound 201 (98 mg, 88%).
  • 1H NMR (399 MHz, CD3OD) δ 7.77-7.73 (m, 1H), 7.51-7.43 (m, 2H), 7.38-7.31 (m, 1H), 7.25-7.19 (ddd, J=9.2, 5.4, 1.6 Hz, 1H), 7.08-7.02 (ddd, J=8.0, 3.8, 1.3 Hz, 1H), 6.28-6.17 (m, 2H), 5.10-5.01 (m, 1H), 4.30-4.16 (m, 3H), 4.11-4.03 (m, 3H), 4.03-3.97 (d, J=5.3 Hz, 2H), 3.74-3.63 (m, 2H), 2.48-2.11 (m, 5H), 1.90-1.82 (m, 6H), 1.43-1.36 (d, J=3.4 Hz, 9H); 13C NMR (100 MHz, CD3OD) δ 178.05, 166.26, 152.25, 152.19, 150.78, 137.80, 137.76, 132.13, 132.09, 130.61, 130.56, 127.24, 124.10, 111.92, 111.84, 111.79, 87.14, 87.09, 86.80, 86.71, 86.50, 85.98, 85.95, 85.92, 85.87, 85.83, 85.75, 80.55, 80.48, 80.32, 80.27, 71.97, 71.73, 68.67, 68.61, 68.35, 68.29, 62.51, 62.42, 40.41, 40.36, 40.32, 39.66, 39.64, 39.35, 39.29, 31.08, 31.04, 27.61, 12.68, 12.65, 12.49; 31P NMR (162 MHz, CD3OD) δ 29.54, 29.29; MS (ESI+ve): calc (M+H): 753.22. found: 753.12. Rf=0.5 (10% MeOH/DCM).
  • Compound 105:
  • Compound 100 (200 mg, 0.176 mmol) was converted to compound 105 by using compound 14 in a procedure analogous to that described for compound 101 (158 mg, 70%).
  • 1H NMR (400 MHz, CDCl3) δ 7.46-7.39 (m, 2H) 7.38-7.16 (m, 18H), 6.90-6.77 (m, 8H), 6.43-6.27 (m, 1H), 5.39-5.18 (m, 2H), 4.31-4.23 (dd, J=12.0, 6.2 Hz, 1H), 4.20-4.12 (m, 1H), 3.98-3.86 (m, 1H), 3.82-3.70 (m, 12H), 3.69-3.52 (m, 1H), 3.50-3.43 (td, J=9.9, 8.9, 2.7 Hz, 1H), 3.41-3.29 (ddd, J=17.2, 10.8, 2.5 Hz, 1H), 2.59-2.49 (m, 1H), 2.44-2.30 (m, 1H), 2.03-1.93 (m, 1H), 1.86-1.79 (d, J=2.9 Hz, 3H), 1.75-1.67 (m, 4H), 1.43-1.36 (d, 3H), 1.16-1.08 (d, J=9.3 Hz, 9H); 31P NMR (162 MHz, CDCl3) δ 28.14, 27.81 (two diastereomers). MS (ESI+ve): calc (M+H): 1281.4. found: 1281.1 (M+H)+ and 1298.6 (M+NH4)+.
  • Compound 205:
  • Compound 105 (137 mg, 0.107 mmol) was converted to compound 205 by a procedure analogous to that described for compound 201 (66 mg, 91%).
  • 1H NMR (399 MHz, CD3OD) δ 7.83-7.76 (m, 1H), 7.56-7.50 (m, 1H), 6.34-6.22 (m, 2H), 5.51-5.43 (m, H), 5.28-5.20 (qt, 0.1=7.8, 1.8 Hz, 1H), 4.47-4.31 (m, 3H), 4.29-4.21 (m, 1H), 4.10-4.05 (m, 1H), 3.87-3.73 (dd, J=7.6, 3.1 Hz, 2H), 2.62-2.50 (tdd, J=16.9, 5.7, 1.9 Hz, 1H), 2.45-2.36 (m, 1H), 2.32-2.25 (ddd, J=6.9, 5.4, 1.5 Hz, 3H), 1.92-1.84 (m, 6H), 1.22-1.18 (d, J=5.3 Hz, 9H); 31P NMR (162 MHz, CD3OD) δ 28.71, 28.42 (two diastereomers). MS (ESI+ve): calc (M+H): 677.2. found: 677.2 (M+H)+, 694.2 (M+NH4)+.
  • Compound 106:
  • Compound 100 (405 mg, 0.357 mmol) was converted to compound 106 by using compound 19 and following a procedure analogous to that described for compound 101 (0.35 g, 71%). 1H NMR (399 MHz, CDCl3) δ 9.97-9.42 (m, 2H), 7.58-7.47 (m, 1H), 7.46-7.39 (m, 2H), 7.39-7.13 (m, 17H), 6.87-6.78 (m, 8H), 6.44-6.29 (dtd, J=20.4, 9.2, 4.7 Hz, 2H), 5.27-5.16 (dt, J=14.7, 7.3 Hz, 1H), 4.30-4.22 (m, 1H), 4.22-4.12 (m, 1H), 4.02-3.90 (q, J=3.8, 3.4 Hz, 2H), 3.80-3.73 (m, 12H), 3.72-3.65 (m, 5H), 3.51-3.43 (m, 1H), 3.40-3.31 (m, 1H), 3.14-2.93 (m, 2H), 2.85-2.72 (m, 4H), 2.67-2.59 (m, 2H), 2.57-2.34 (m, 6H), 1.97-1.87 (td, J=13.7, 13.1, 5.7 Hz, 1H), 1.84 (s, 3H), 1.73-1.61 (td, J=14.1, 6.8 Hz, 1H), 1.42-1.37 (d, J=6.7 Hz, 3H); 13C NMR (100 MHz, CDCl3) δ 163.97, 163.94, 163.91, 158.88, 158.84, 150.64, 150.60, 150.52, 144.86, 144.83, 144.09, 144.04, 136.06, 136.04, 135.95, 135.93, 135.54, 135.19, 135.09, 135.03, 134.99, 130.28, 130.17, 130.13, 128.29, 128.17, 128.14, 127.38, 127.31, 113.51, 113.42, 111.82, 111.79, 111.44, 111.38, 87.53, 87.38, 87.33, 85.29, 85.26, 84.89, 84.85, 84.41, 84.36, 84.29, 84.25, 83.88, 83.85, 83.80, 83.76, 79.28, 79.23, 78.72, 78.67, 74.04, 67.53, 67.46, 67.37, 67.29, 66.77, 63.33, 63.21, 57.84, 55.34, 53.41, 53.34, 39.23, 39.09, 39.01, 38.92, 38.55, 38.51, 38.46, 38.42, 35.64, 35.59, 30.35, 30.30, 30.26, 12.60, 11.79, 11.74; 31P NMR (162 MHz, CDCl3) δ 29.30, 29.14; MS (ESI+ve): calc (M+H): 1372.44. found: 1372.79. Rf=0.4 (5% MeOH/DCM).
  • Compound 206:
  • Compound 106 (200 mg, 0.146 mmol) was converted to compound 206 by a procedure analogous to that described for compound 201 (110 mg, 98%).
  • 1H NMR (399 MHz, CD3OD) δ 7.83-7.75 (dd, J=7.6, 1.4 Hz, 1H), 7.56-7.48 (d, J=1.6 Hz, 1H), 6.35-6.23 (m, 2H), 5.27-5.20 (m, 1H), 4.48-4.31 (m, 3H), 4.28-4.21 (dd, J=9.7, 2.1 Hz, 1H), 4.11-4.04 (t, J=4.0 Hz, 1H), 3.97-3.84 (br, 4H), 3.83-3.77 (dd, J=6.0, 3.2 Hz, 2H), 3.43-3.36 (m, 2H), 3.29-3.18 (m, 6H), 3.11-3.00 (m, 4H), 2.62-2.51 (tdd, J=11.7, 5.7, 1.7 Hz, 1H), 2.47-2.38 (ddd, J=14.3, 8.4, 6.0 Hz, 1H), 2.38-2.25 (q, J=5.3, 4.8 Hz, 2H), 1.91 (s, 3H), 1.88 (s, 3H); 31P NMR (162 MHz, CD3OD) δ 30.19, 30.12; 13C NMR (100 MHz, CD3OD) δ 166.28, 166.24, 166.23, 152.32, 152.27, 152.24, 138.05, 138.00, 137.77, 137.75, 112.08, 112.03, 111.97, 111.94, 87.28, 87.24, 87.01, 86.96, 86.62, 86.51, 86.10, 86.06, 85.76, 85.68, 71.73, 71.51, 68.91, 68.58, 68.51, 65.44, 62.60, 62.50, 57.50, 53.50, 40.25, 40.16, 39.64, 39.57, 39.20, 39.16, 39.06, 32.56, 32.55, 31.04, 31.00, 12.73, 12.69, 12.52; MS (ESI+ve): calc (M+H): 768.18. found: 768.14. Rf=0.3 (10% MeOH/DCM).
  • Compound 107:
  • Using compound 22 in place of compound 5, compound 100 is converted to compound 107 by a procedure analogous to that described for compound 101.
  • Compound 207:
  • Compound 107 is converted to compound 207 by a procedure analogous to that described for compound 201.
  • Compound 108:
  • Using compound 25 in place of compound 5, compound 100 is converted to compound 108 by a procedure analogous to that described for compound 101.
  • Compound 208:
  • Compound 108 is converted to compound 208 by a procedure analogous to that described for compound 201.
  • Compound 109:
  • Using compound 27 in place of compound 5, compound 100 is converted to compound 109 by a procedure analogous to that described for compound 101.
  • Compound 209:
  • Compound 109 is converted to compound 209 by a procedure analogous to that described for compound 201.
  • Compound 110:
  • Using compound 29 in place of compound 5, compound 100 is converted to compound 110 by a procedure analogous to that described for compound 101.
  • Compound 210:
  • Compound 110 is converted to compound 210 by a procedure analogous to that described for compound 201.
  • Compound 111:
  • Using compound 31 in place of compound 5, compound 100 is converted to compound 111 by a procedure analogous to that described for compound 101.
  • Compound 211:
  • Compound 111 is converted to compound 211 by a procedure analogous to that described for compound 201.
  • Compound 112:
  • Using compound 33 in place of compound 5, compound 100 is converted to compound 112 by a procedure analogous to that described for compound 101.
  • Compound 212:
  • Compound 112 is converted to compound 212 by a procedure analogous to that described for compound 201.
  • Compound 113:
  • Using compound 38 in place of compound 5, compound 100 is converted to compound 113 by a procedure analogous to that described for compound 101.
  • Compound 213:
  • Compound 113 is converted to compound 213 by a procedure analogous to that described for compound 201.
  • Compound 114:
  • Using compound 41 in place of compound 5, compound 100 is converted to compound 114 by a procedure analogous to that described for compound 101.
  • Compound 214:
  • Compound 114 is converted to compound 214 by a procedure analogous to that described for compound 201.
  • Compound 115:
  • Using compound 43 in place of compound 5, compound 100 is converted to compound 115 by a procedure analogous to that described for compound 101.
  • Compound 215:
  • Compound 115 is converted to compound 215 by a procedure analogous to that described for compound 201.
  • Example 3 Alternative Synthesis of Phosphorothiotriesters Using
  • Figure US20170029457A1-20170202-C00059
    Figure US20170029457A1-20170202-C00060
    Figure US20170029457A1-20170202-C00061
  • Compound 150:
  • Compound 100 (300 mg, 0.264 mmol) was converted to compound 150 by a procedure analogous to that described for compound 101 (170 mg, 50%).
  • 1H NMR (399 MHz, CDCl3) δ 9.34-9.30 (s, 1H), 9.28-9.17 (d, J=30.6 Hz, 1H), 7.57-7.47 (m, 1H), 7.47-7.40 (m, 2H), 7.38-7.18 (m, 17H), 7.18-7.07 (d, =1.4 Hz, 1H), 6.88-6.77 (dd, J=9.0, 1.5 Hz, 8H), 6.44-6.34 (ddd, J=15.6, 8.9, 5.4 Hz, 1H), 6.32-6.21 (ddd, J=18.9, 8.5, 5.9 Hz, 1H), 5.27-5.19 (q, J=5.9 Hz, 1H), 4.46-4.33 (m, 2H), 4.31-4.16 (m, 2H), 4.03-3.91 (m, 2H), 3.81-3.67 (m, 12H), 3.54-3.46 (m, 1H), 3.42-3.34 (m, 1H), 3.34-3.25 (d, J=20.2 Hz, 3H), 2.64-2.53 (td, J=13.4, 5.4 Hz, 1H), 2.47-2.34 (dq, J=19.9, 6.5, 5.9 Hz, 1H), 1.99-1.91 (m, 1H), 1.85-1.80 (t, J=1.5 Hz, 3H), 1.78-1.65 (tt, J=14.1, 7.5 Hz, 111), 1.44-1.37 (dd, J=7.3, 1.2 Hz, 3H); 13C NMR (100 MHz, CDCl3) δ 171.27, 163.83, 163.80, 158.95, 158.93, 158.90, 150.64, 150.53, 150.46, 150.38, 144.91, 144.88, 144.09, 144.02, 136.00, 135.98, 135.94, 135.81, 135.11, 135.04, 134.98, 134.97, 130.34, 130.27, 130.20, 128.30, 128.23, 128.20, 127.46, 127.36, 113.59, 113.56, 113.48, 111.95, 111.38, 87.60, 87.47, 87.43, 86.03, 85.83, 84.44, 84.34, 83.81, 79.82, 79.58, 73.99, 73.91, 67.85, 67.78, 63.31, 63.20, 55.39, 51.77, 51.70, 39.16, 38.99, 38.90, 37.21, 37.16, 37.12, 37.05, 12.63, 12.57, 11.85, 11.80; 31P NMR (162 MHz, CDCl3) δ 26.15, 25.60; MS (ESI+ve): calc (M+H): 1308.37. found: 1308.70. Rf=0.5 (5% MeOH/DCM).
  • Compound 151:
  • A DCM (5 mL) solution of compound 150 (150 mg, 0.116 mmol) was treated with 2-morpholinoethanethiol (17 mg, 0.116 mmol) at r.t. with monitoring by TLC. After 0.5 h, the mixture was washed with NaHCO3, extracting 5× into DCM. The organic extracts were dried (MgSO4), filtered and reduced. Column chromatography gave compound 151 as a colorless solid foam (81 mg, 51%).
  • 1H NMR (399 MHz, CDCl3) δ 9.68-9.54 (m, 1H), 9.44 (s, 1H), 7.59-7.48 (m, 1H), 7.47-7.40 (m, 2H), 7.40-7.13 (m, 17H), 6.90-6.76 (ddd, J=9.3, 4.4, 2.7 Hz, 8H), 6.45-6.27 (m, 2H), 5.32-5.22 (dd, J=8.5, 5.7 Hz, 1H), 4.34-4.25 (m, 1H), 4.23-4.14 (m, 1H), 4.07-3.89 (m, 2H), 3.79-3.74 (m, 12H), 3.74-3.65 (m, 6H), 3.51-3.33 (m, 2H), 2.90-2.79 (dd, J=14.2, 7.6 Hz, 2H), 2.73-2.55 (m, 3H), 2.55-2.34 (m, 6H), 2.02-1.91 (m, 1H), 1.87-1.81 (dd, J=4.9, 1.2 Hz, 3H), 1.77-1.66 (ddd, J=14.2, 8.7, 6.4 Hz, 1H), 1.41-1.35 (dd, J=6.6, 1.2 Hz, 3H); 13C NMR (100 MHz, CDCl3) δ 163.97, 163.93, 163.88, 158.90, 158.86, 158.71, 150.64, 150.59, 150.53, 150.50, 144.92, 144.88, 144.13, 144.08, 136.11, 136.07, 136.03, 136.00, 135.73, 135.60, 135.22, 135.14, 135.08, 135.04, 135.02, 130.32, 130.30, 130.23, 130.18, 128.33, 128.19, 128.17, 127.39, 127.33, 113.56, 113.52, 113.45, 111.85, 111.82, 111.38, 111.29, 87.56, 87.41, 87.38, 85.71, 85.35, 84.91, 84.38, 84.27, 84.22, 84.05, 83.97, 83.85, 83.78, 79.36, 79.11, 79.05, 74.25, 74.07, 67.39, 66.88, 66.79, 63.27, 57.80, 55.36, 53.55, 53.51, 53.40, 43.06, 40.72, 40.54, 39.25, 39.16, 39.01, 35.91, 12.64, 12.60, 11.78, 11.74; 31P NMR (162 MHz, CDCl3) δ 27.76, 27.46; MS (ESI+ve): calc (M+H): 1358.43. found: 1358.74. Rf=0.4 (5% MeOH/DCM).
  • Compound 251:
  • Compound 151 (75 mg, 0.055 mmol) was converted to compound 251 by a procedure analogous to that described for compound 201 (10 mg, 24%). MS (ESI+ve): calc (M+H): 754.17. found: 754.19. Rf=0.3 (10% MeOH/DCM).
  • Compound 152:
  • Compound 100 is converted to compound 152 by a procedure analogous to that described for compound 101.
  • Figure US20170029457A1-20170202-C00062
    Figure US20170029457A1-20170202-C00063
  • Compound 153:
  • Using 1-Thio-β-D-glucose tetraacetate in place of compound 4, compound 152 is converted to compound 153 by a procedure analogous to that described for compound 151.
  • Compound 253:
  • Compound 153 is converted to compound 253 by a procedure analogous to that described for compound 201.
  • Compound 154:
  • Compound 100 is converted to compound 154 by a procedure analogous to that described for compound 101.
  • Compound 155:
  • Compound 154 is converted to compound 155 by a procedure analogous to that described for compound 151.
  • Compound 255:
  • Compound 155 is converted to compound 255 by a procedure analogous to that described for compound 201.
  • Example 4 Thioalkylation of H-Phosphonates to Provide Phosphorothiotriesters in Solid Phase
  • Compound 300:
  • Synthesis of (Rp)-CAGT-H-phosphonate-oxalyl linker-CPG was carried out on an Applied Biosystems 394 DNA/RNA synthesizer according to the reported methods (Journal of American Chemical Society 2008, 130, 16031-16037; Angewandte Chemie International Edition 2009, 48, 496-499).
  • Compound 301:
  • (Sp)-CAGT-phosphorothioate (R═H): (Rp)-CAGT-H-phosphonate-oxalyl linker-CPG was treated by 0.2 M Beaucage Reagent/CH3CN—BSA (9:1, v/v), stirred for 1 h at rt, then washed successively with CS2 and acetonitrile and dried under reduced pressure. The resultant CPG was treated with 2 mL of 28% aqueous NH3 and stirred for 18 h at rt. After removal of NH3 under reduced pressure, the resulting product was analyzed by LC/MS and HPLC.
  • Compound 302:
  • (Sp)-CAGT-S-methyl phosphorothiotriester (R=Me): BSTFA (50 μL, 188 μmol) and acetonitrile (500 μL) were added to (Rp)-CAGT-H-phosphonate-oxalyl linker-CPG (14.7 mg, 1 μmol) then the mixture was shaken for 20 min at rt. S-methyl methane sulfonothioate (20 μL, 212 μmol) and NEt3 (50 μL) were added and shaking was continued for 1 h at rt. The CPG was washed with CH3CN then dried in vacuo. 20% PrNH2 in dry CH3CN (2 mL) was added to the CPG and the mixture was stirred for 16 h at rt. Solvents were removed under reduced pressure and CH3CN was added to the mixture. The CPG was removed by filtration and the filtrate was concentrated under reduced pressure. CH3CN/DMSO/0.5 M AA buffer (1:1:1, v/v/v) was added, the mixture was stirred for 16 h at rt, then analyzed by LC/MS and HPLC.
  • Compound 303:
  • Compound 303 is prepared by sulfurization of compound 300 on support followed by cleavage. ACN (450 μL), BSTFA (50 μL) and compound 12 (20 mg) are added to compound 300 (1 μmol) which is shaken for 18 h. The CPG is collected by filtration resuspended in 20% PrNH2 in dry CH3CN (2 mL) and shaken for 16 h at rt. Solvents were removed under reduced pressure and the residue is purified by RPHPLC to provide pure compound 303.
  • Example 5 Thioalkylation of H-Phosphonates to Provide Phosphorothiotriesters in Solution Phase
  • Figure US20170029457A1-20170202-C00064
    Figure US20170029457A1-20170202-C00065
    Figure US20170029457A1-20170202-C00066
  • Compound 305:
  • Compound 300 (0.5 μmol) was taken up in ACN (125 μL) then BSTFA (62 μL) was added and the mixture was shaken for 20 min. PrNH2 (125 μL) was added and the vial was rotated for 18 h. After filtration and washing with 1 mL ACN, the solvent was removed in vacuo and the residue was co-evaporated 3× with toluene to provide crude compound 304. The residue was redissolved in pyridine (375 μL) and treated with BSTFA for (16 μl, 60.0 μmol) followed by compound 9 (7.2 mg, 30.0 μmol) with stirring under Ar. After 2 h at r.t. the solvent was removed and the residue was treated with MeOH (0.125 mL) for 1 h, then AA (0.5 M, 0.125 mL) was added and the mixture was stirred at r.t. for 2 h. The product was purified by RPHPLC to provide compound 305.
  • Compound 303:
  • Substituting compound 12 for compound 9, compound 303 was prepared by a procedure analogous to that described for compound 305.
  • Compound 306:
  • Substituting compound 12 for compound 14, compound 306 was prepared by a procedure analogous to that described for compound 305.
  • Compound 307:
  • Substituting compound 12 for compound 29, compound 307 is prepared by a procedure analogous to that described for compound 305.
  • Compound 308:
  • Substituting compound 12 for compound 31, compound 308 is prepared by a procedure analogous to that described for compound 305.
  • Compound 309:
  • Substituting compound 12 for compound 38, compound 309 is prepared by a procedure analogous to that described for compound 305.
  • Example 6 Stereoselective Thioalkylation of H-Phosphanates
  • Objective: To demonstrate that the reaction of MTS reagents to H-phosphonate to generate phosphorothio triester is stereospecific. 31P NMR was used to trace the changes during the course of the reaction.
  • Figure US20170029457A1-20170202-C00067
  • Experimental procedure: In an NMR tube was added compound 100S 5′-O-(4,4′-dimethoxytrityl)thymidin-3′-yl 3′-O-(4,4′-dimethoxytrityl)thymidin-5′-yl H-phosphonate (20 mg, 18 mol) in 0.8 mL CD3CN and the 31P NMR spectrum was recorded. BSTFA (17 μL, 176 μmol) was added to same NMR tube and after 5 min 31P NMR spectrum was recorded again. Triethylamine (49 μL, 352 μmol) and S-methyl methanethiosulfonate (22 μL, 88 μmol) were added to same NMR tube and 31P NMR spectrum was recorded immediately.
  • The same procedure was repeated for Rp isomer (compound 100R). The 31P NMR spectrum recorded for the starting material, intermediate and the product show that the stereochemistry at phosphorus atom is retained during the reaction.
  • While preferred embodiments of the present invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the embodiments of the invention described herein may be employed in practicing the invention. It is intended that the following claims define the scope of the invention and that methods and structures within the scope of these claims and their equivalents be covered thereby.

Claims (18)

1. A process for the preparation of phosphorothiotriesters of structure IIIa comprising the steps of:
i) reacting an H-phosphonate of structure Ia with an silylating reagent to provide a silyloxyphosphonate; and
ii) reacting the silyloxyphosphonate with a thiosulfonate reagent of structure IIa to provide a phosphorothiotriester of structure IIIa;
wherein,
the H-phosphonate of structure Ia has the following structure:
Figure US20170029457A1-20170202-C00068
wherein,
W is independently selected from O, S, NH, or CH2;
R3 is —OH, —SH, —NRdRd, —N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —P(O)(Re)2, —HP(O)(Re), —ORa or —SRc;
Y1 is O, NRd, S, or Se;
Ra is a blocking group;
Rc is a blocking group;
each instance of Rd is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, —P(O)(Re)2, or —HP(O)(Re);
each instance of Re is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2—, alkenyl-Y2—, alkynyl-Y2—, aryl-Y2—, or heteroaryl-Y2—, or a cation which is Na+1, Li+1, or K+1;
Y2 is O, NRd, or S;
each instance of R4 is independently hydrogen, —OH, —SH, —NRdRd, —N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —ORb, or —SRc, and Rb is a blocking group;
each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;
R5 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid; and
n is between 1 and about 200; and
the thiosulfonate reagent of structure IIa has the following structure:
Figure US20170029457A1-20170202-C00069
wherein,
X is alkyl, cycloalkyl, or heteroaryl;
R is alkyl, alkenyl, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, or R1-R2;
R1 is selected from —S-alkenylene-, —S-alkylene-, —S-alkylene-aryl-alkylene-, —S—CO-aryl-alkylene-, or —S—CO-alkylene-aryl-alkylene-;
R2 is selected from heterocyclo-alkylene-S—, heterocyclo-alkenylene-S—, aminoalkyl-S—, or (alkyl)4N-alkylene-S—;
and the phosphorothiotriester of structure IIIa has the following structure:
Figure US20170029457A1-20170202-C00070
wherein,
W is independently selected from O, S, NH, or CH2;
R is alkyl, alkenyl, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, or R1-R2;
R1 is selected from —S-alkenylene-, —S-alkylene-, —S-alkylene-aryl-alkylene-, —S—CO-aryl-alkylene-, or —S—CO-alkylene-aryl-alkylene-;
R2 is selected from heterocyclo-alkylene-S—, heterocyclo-alkenylene-S—, aminoalkyl-S—, or (alkyl)4N-alkylene-S—;
R3 is —OH, —SH, —NRdRd, —N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —P(O)(Re)2, —HP(O)(Re), —ORa or —SRc;
Y1 is O, NRd, S, or Se;
Ra is a blocking group;
Rc is a blocking group;
each instance of Rd is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, —P(O)(Re)2, or —HP(O)(Re);
each instance of Re is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2—, alkenyl-Y2—, alkynyl-Y2—, aryl-Y2—, or heteroaryl-Y2—, or a cation which is Na+1, Li+1, or K+1;
Y2 is O, NRd, or S;
each instance of R4 is independently hydrogen, —OH, —SH, —NRdRd, —N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —ORb, or —SRc, and Rb is a blocking group;
each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;
R5 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid; and
n is between 1 and about 200.
2. A process for the preparation of phosphorothiotriesters comprising non-stereorandom phosphorous linkages of structure IIIb comprising the steps of:
i) reacting a H-phosphonate comprising non-stereorandom phosphorous linkages of structure Ib with an silylating reagent to provide a silyloxyphosphonate; and
ii) reacting the silyloxyphosphonate with a thiosulfonate reagent of structure IIb to provide a phosphorothiotriester comprising non-stereorandom phosphorous linkages of structure IIIb;
wherein,
the H-phosphonate comprising non-stereorandom phosphorous linkages of structure Ib has the following structure:
Figure US20170029457A1-20170202-C00071
wherein,
W is independently selected from O, NH, or CH2;
R3 is —OH, —SH, —NRdRd, —N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —P(O)(Re)2, —HP(O)(Re), —ORa or —SRc;
Y1 is O, NRd, S, or Se;
Ra is a blocking group;
Rc is a blocking group;
each instance of Rd is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, —P(O)(Re)2, or —HP(O)(Re);
each instance of Re is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2—, alkenyl-Y2—, alkynyl-Y2—, aryl-Y2—, or heteroaryl-Y2—, or a cation which is Na+1, Li+1, or K+1;
Y2 is O, NRd, or S;
each instance of R4 is independently hydrogen, —OH, —SH, —NRdRd, —N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —ORb, or —SRc, and Rb is a blocking group;
each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;
R5 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid; and
n is between 1 and about 200; and
the thiosulfonate reagent of structure IIb has the following structure:
Figure US20170029457A1-20170202-C00072
wherein,
X is alkyl, cycloalkyl, aryl, or heteroaryl;
R is alkyl, alkenyl, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, or R1-R2;
R1 is selected from —S-alkenylene-, —S-alkylene-, —S-alkylene-aryl-alkylene-, —S—CO-aryl-alkylene-, or —S—CO-alkylene-aryl-alkylene-;
R2 is selected from heterocyclo-alkylene-S—, heterocyclo-alkenylene-S—, aminoalkyl-S—, or (alkyl)4N-alkylene-S—;
and the chiral phosphorothiotriester comprising non-stereorandom phosphorous linkages of structure IIIb has the following structure:
Figure US20170029457A1-20170202-C00073
wherein,
W is independently selected from O, NH, or CH2;
R is alkyl, alkenyl, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, or R1-R2;
R1 is selected from —S-alkenylene-, —S-alkylene-, —S-alkylene-aryl-alkylene-, —S—CO-aryl-alkylene-, or —S—CO-alkylene-aryl-alkylene-;
R2 is selected from heterocyclo-alkylene-S—, heterocyclo-alkenylene-S—, aminoalkyl-S—, or (alkyl)4N-alkylene-S—;
R3 is —OH, —SH, —NRdRd, —N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —P(O)(Re)2, —HP(O)(Re), —ORa or —SRc;
Y1 is O, NRd, S, or Se;
Ra is a blocking group;
Rc is a blocking group;
each instance of Rd is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, —P(O)(Re)2, or —HP(O)(Re);
each instance of Re is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2—, alkenyl-Y2—, alkynyl-Y2—, aryl-Y2—, or heteroaryl-Y2—, or a cation which is Na+1, Li+1, or K+1;
Y2 is O, NRd, or S;
each instance of R4 is independently hydrogen, —OH, —SH, —NRdRd, —N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —ORb, or —SRc, and Rb is a blocking group;
each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;
R5 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid; and
n is between 1 and about 200.
3. The process of claim 1, wherein W is O.
4. The process of claim 1, wherein R1 is selected from:
Figure US20170029457A1-20170202-C00074
and
R2 is selected from:
Figure US20170029457A1-20170202-C00075
5. The process of claim 1, wherein the silylating reagent is selected from
1,1,3,3-tetramethyl-1,3-diphenyldisilazane;
1,3-dimethyl-1,1,3,3-tetraphenyldisilazane;
1-(trimethylsilyl)imidazole;
N-trimethylsilyl-N-methyl trifluoroacetamide;
bis(dimethylamino)dimethylsilane;
bromotrimethylsilane;
chlorodimethyl(pentafluorophenyl)silane;
chlorotriethyl silane;
chlorotriisopropylsilane;
chlorotrimethylsilane;
dichlorodimethylsilane;
hexamethyldisilazane;
N,N′-bis(trimethylsilyl)urea;
N,N-bis(trimethylsilyl)methylamine;
N,N-dimethyltrimethylsilylamine;
N,O-bis(trimethylsilyl)acetamide;
N,O-bis(trimethylsilyl)carbamate;
N,O-bis(trimethylsilyl)trifluoroacetamide;
N-methyl-N-(trimethylsilyl)trifluoroacetamide;
N-methyl-N-trimethylsilylacetamide;
N-methyl-N-trimethylsilylheptafluorobutyramide;
N-tert-butyldimethylsilyl-N-methyltrifluoroacetamide;
N-methyl-N-trimethylsilylheptafluorobutyramide;
trimethylsilyltriflate;
triethylsilyltriflate;
triisopropylsilyltriflate; or
tert-butyldimethylsilyltriflate.
6. The process of claim 5, wherein the silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide, trimethylsilyltriflate, chlorotrimethylsilane, or 1-(trimethyl silyl)imidazole.
7. The process of claim 6, wherein the silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide.
8. The process of claim 1, wherein the H-phosphonate is covalently linked to a solid phase.
9. A process for the preparation of phosphorothiotriesters of structure IIIc comprising the steps of:
i) reacting a H-phosphonate of structure Ic with an silylating reagent to provide a silyloxyphosphonate;
ii) reacting the silyloxyphosphonate with a bis(thiosulfonate) reagent of structure IVc to provide a phosphorothiotriester comprising a thiosulfonate group of structure Vc;
iii) reacting the phosphorothiotriester comprising a thiosulfonate group of structure Vc with a nucleophile of structure VIc to provide the phosphorothiotriesters of structure IIIc;
wherein,
the H-phosphonate of structure Ic has the following structure:
Figure US20170029457A1-20170202-C00076
wherein,
W is independently selected from O, S, NH, or CH2;
R3 is —OH, —SH, —NRdRd, —N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, P(O)(Re)2, —HP(O)(Re), —ORa or —SRc;
Y1 is O, NRd, S, or Se;
Ra is a blocking group;
Rc is a blocking group;
each instance of Rd is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, —P(O)(Re)2, or —HP(O)(Re);
each instance of Re is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2—, alkenyl-Y2—, alkynyl-Y2—, aryl-Y2—, or heteroaryl-Y2—, or a cation which is Na+1, Li+1, or K+1;
Y2 is O, NRd, or S;
each instance of R4 is independently hydrogen, —OH, —SH, —NRdRd, —N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —ORb, or —SRc, and Rb is a blocking group;
each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;
R5 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid; and
n is between 1 and about 200; and
the bis(thiosulfonate) reagent of structure IVc has the following structure:
Figure US20170029457A1-20170202-C00077
wherein,
X is alkylene, alkenylene, arylene, or heteroarylene;
each R6 is independently alkyl, cycloalkyl, aryl, or heteroaryl;
the nucleophile of structure VIc has the following structure:
R7—SH, wherein R7 is selected from alkyl, alkenyl, aryl, heterocyclo, aminoalkyl, or (heterocyclo)alkyl;
and phosphorothiotriesters of structure IIIc has the following structure:
Figure US20170029457A1-20170202-C00078
wherein,
W is independently selected from O, S, NH, or CH2;
R is R7—S—S—X—
R7 is alkyl, alkenyl, aryl, heterocyclo, aminoalkyl, or (heterocyclo)alkyl;
X is alkylene, alkenylene, arylene, or heteroarylene;
R3 is —OH, —SH, —NRdRd, —N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —P(O)(Re)2, —HP(O)(Re), —ORa or —SRc;
Y1 is O, NRd, S, or Se;
Ra is a blocking group;
Rc is a blocking group;
each instance of Rd is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, —P(O)(Re)2, or —HP(O)(Re);
each instance of Re is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2—, alkenyl-Y2—, alkynyl-Y2—, aryl-Y2—, or heteroaryl-Y2—, or a cation which is Na+1, Li+1, or K+1;
Y2 is O, NRd, or S;
each instance of R4 is independently hydrogen, —OH, —SH, —NRdRd, —N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —ORb, or —SRc, and Rb is a blocking group;
each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;
R5 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid;
n is between 1 and about 200; and
wherein the phosphorous linkages of the H-phosphonate of structure Ic, the phosphorothiotriester comprising a thiosulfonate group of structure Vc, and the phosphorothiotriesters of structure IIIc may optionally comprise non-stereorandom phosphorous linkages.
10. The process of claim 8, wherein the phosphorothiotriesters of structure IIIb comprise non-stereorandom phosphorous linkages and the H-phosphonate of structure Ic comprise non-stereorandom phosphorous linkages; and W is independently selected from O, NH, or CH2.
11. The process of claim 9, wherein W is O.
12. The process of claim 9, wherein R6 is methyl.
13. The process of claim 9, wherein bis(thiosulfonate) reagent of structure IVc is selected from:
Figure US20170029457A1-20170202-C00079
14. The process of claim 9, wherein the nucleophile of structure VIc has the following structure:
Figure US20170029457A1-20170202-C00080
15. The process of claim 9, wherein the silylating reagent is selected from
1,1,3,3-tetramethyl-1,3-diphenyldisilazane;
1,3-dimethyl-1,1,3,3-tetraphenyldisilazane;
1-(trimethylsilyl)imidazole;
N-trimethylsilyl-N-methyl trifluoroacetamide;
bis(dimethylamino)dimethylsilane;
bromotrimethylsilane;
chlorodimethyl(pentafluorophenyl)silane;
chlorotriethyl silane;
chlorotriisopropylsilane;
chlorotrimethylsilane;
dichlorodimethylsilane;
hexamethyldisilazane;
N,N′-bis(trimethylsilyl)urea;
N,N-bis(trimethylsilyl)methylamine;
N,N-dimethyltrimethylsilylamine;
N,O-bis(trimethylsilyl)acetamide;
N,O-bis(trimethylsilyl)carbamate;
N,O-bis(trimethylsilyl)trifluoroacetamide;
N-methyl-N-(trimethylsilyl)trifluoroacetamide;
N-methyl-N-trimethylsilylacetamide;
N-methyl-N-trimethylsilylheptafluorobutyramide;
N-tert-butyldimethylsilyl-N-methyltrifluoroacetamide;
N-methyl-N-trimethylsilylheptafluorobutyramide;
trimethylsilyltriflate;
triethylsilyltriflate;
triisopropylsilyltriflate; or
tert-butyldimethylsilyltriflate.
16. The process of claim 15, wherein the silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide, trimethylsilyltriflate, chlorotrimethylsilane, or 1-(trimethyl silyl)imidazole.
17. The process of claim 16, wherein the silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide.
18. The process of claim 17, wherein the H-phosphonate is covalently linked to a solid phase.
US15/222,910 2011-07-19 2016-07-28 Methods for the synthesis of functionalized nucleic acids Abandoned US20170029457A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US15/222,910 US20170029457A1 (en) 2011-07-19 2016-07-28 Methods for the synthesis of functionalized nucleic acids
US15/941,494 US10280192B2 (en) 2011-07-19 2018-03-30 Methods for the synthesis of functionalized nucleic acids

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201161509526P 2011-07-19 2011-07-19
PCT/US2012/046805 WO2013012758A1 (en) 2011-07-19 2012-07-13 Methods for the synthesis of functionalized nucleic acids
US201414233579A 2014-02-27 2014-02-27
US15/222,910 US20170029457A1 (en) 2011-07-19 2016-07-28 Methods for the synthesis of functionalized nucleic acids

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2012/046805 Continuation WO2013012758A1 (en) 2011-07-19 2012-07-13 Methods for the synthesis of functionalized nucleic acids
US14/233,579 Continuation US9605019B2 (en) 2011-07-19 2012-07-13 Methods for the synthesis of functionalized nucleic acids

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/941,494 Continuation US10280192B2 (en) 2011-07-19 2018-03-30 Methods for the synthesis of functionalized nucleic acids

Publications (1)

Publication Number Publication Date
US20170029457A1 true US20170029457A1 (en) 2017-02-02

Family

ID=47558410

Family Applications (3)

Application Number Title Priority Date Filing Date
US14/233,579 Active US9605019B2 (en) 2011-07-19 2012-07-13 Methods for the synthesis of functionalized nucleic acids
US15/222,910 Abandoned US20170029457A1 (en) 2011-07-19 2016-07-28 Methods for the synthesis of functionalized nucleic acids
US15/941,494 Active US10280192B2 (en) 2011-07-19 2018-03-30 Methods for the synthesis of functionalized nucleic acids

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US14/233,579 Active US9605019B2 (en) 2011-07-19 2012-07-13 Methods for the synthesis of functionalized nucleic acids

Family Applications After (1)

Application Number Title Priority Date Filing Date
US15/941,494 Active US10280192B2 (en) 2011-07-19 2018-03-30 Methods for the synthesis of functionalized nucleic acids

Country Status (16)

Country Link
US (3) US9605019B2 (en)
EP (2) EP3248982A1 (en)
JP (2) JP6128529B2 (en)
KR (1) KR20140068884A (en)
CN (2) CN103796657B (en)
AU (1) AU2012284265B2 (en)
BR (1) BR112014001244A2 (en)
CA (1) CA2842358C (en)
DK (1) DK2734208T3 (en)
ES (1) ES2626488T3 (en)
HK (1) HK1198479A1 (en)
IL (1) IL230522B (en)
MX (1) MX347361B (en)
RU (1) RU2014105311A (en)
SG (1) SG10201700554VA (en)
WO (1) WO2013012758A1 (en)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9982257B2 (en) 2012-07-13 2018-05-29 Wave Life Sciences Ltd. Chiral control
US10144933B2 (en) 2014-01-15 2018-12-04 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having immunity induction activity, and immunity induction activator
US10149905B2 (en) 2014-01-15 2018-12-11 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having antitumor effect and antitumor agent
US10160969B2 (en) 2014-01-16 2018-12-25 Wave Life Sciences Ltd. Chiral design
US10167309B2 (en) 2012-07-13 2019-01-01 Wave Life Sciences Ltd. Asymmetric auxiliary group
US10280192B2 (en) 2011-07-19 2019-05-07 Wave Life Sciences Ltd. Methods for the synthesis of functionalized nucleic acids
US10307434B2 (en) 2009-07-06 2019-06-04 Wave Life Sciences Ltd. Nucleic acid prodrugs and methods of use thereof
US10329318B2 (en) 2008-12-02 2019-06-25 Wave Life Sciences Ltd. Method for the synthesis of phosphorus atom modified nucleic acids
US10428019B2 (en) 2010-09-24 2019-10-01 Wave Life Sciences Ltd. Chiral auxiliaries
US10450568B2 (en) 2015-10-09 2019-10-22 Wave Life Sciences Ltd. Oligonucleotide compositions and methods thereof
US10479995B2 (en) 2015-07-22 2019-11-19 Wave Life Sciences Ltd. Oligonucleotide compositions and methods thereof
US10724035B2 (en) 2016-05-04 2020-07-28 Wave Life Sciences Ltd. Oligonucleotide compositions and methods thereof
US11013757B2 (en) 2016-06-03 2021-05-25 Wave Life Sciences Ltd. Oligonucleotides, compositions and methods thereof
US11407775B2 (en) 2016-03-13 2022-08-09 Wave Life Sciences Ltd. Compositions and methods for phosphoramidite and oligonucleotide synthesis
US11873316B2 (en) 2016-11-23 2024-01-16 Wave Life Sciences Ltd. Compositions and methods for phosphoramidite and oligonucleotide synthesis

Families Citing this family (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101141544B1 (en) 2009-03-13 2012-05-03 한국과학기술원 Multi-conjugate of siRNA and preparing method thereof
MX356830B (en) 2012-07-13 2018-06-15 Shin Nippon Biomedical Laboratories Ltd Chiral nucleic acid adjuvant.
AU2014346658A1 (en) * 2013-11-06 2016-06-02 Solstice Biologics, Ltd. Polynucleotide constructs having disulfide groups
US10322173B2 (en) 2014-01-15 2019-06-18 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having anti-allergic activity, and anti-allergic agent
WO2015125845A1 (en) * 2014-02-20 2015-08-27 塩野義製薬株式会社 Phosphate site modification of nucleic acid containing nitrogen-containing non-aromatic heterocycle
GB201410693D0 (en) 2014-06-16 2014-07-30 Univ Southampton Splicing modulation
RU2708237C2 (en) 2014-08-22 2019-12-05 Общество с ограниченной ответственностью "НооГен" Modified oligonucleotides and method for production thereof
US10465188B2 (en) 2014-08-22 2019-11-05 Auckland Uniservices Limited Channel modulators
SG11201702682PA (en) 2014-10-03 2017-04-27 Cold Spring Harbor Lab Targeted augmentation of nuclear gene output
DK3234131T3 (en) 2014-12-16 2020-06-29 Roche Innovation Ct Copenhagen As Method of screening for chiral toxicity
CN108026527B (en) 2015-06-15 2022-05-10 Mpeg La有限责任公司 Defined multiconjugated oligonucleotides
JPWO2016208773A1 (en) * 2015-06-26 2018-04-12 協和発酵キリン株式会社 Oligonucleotide derivatives
KR102422625B1 (en) 2015-10-09 2022-07-20 유니버시티 오브 사우스앰톤 Regulation of gene expression and screening of deregulated protein expression
CA3005245A1 (en) 2015-12-14 2017-06-22 Cold Spring Harbor Laboratory Antisense oligomers for treatment of alagille syndrome
US11096956B2 (en) 2015-12-14 2021-08-24 Stoke Therapeutics, Inc. Antisense oligomers and uses thereof
ES2903394T3 (en) 2015-12-14 2022-04-01 Cold Spring Harbor Laboratory Compositions for the treatment of retinitis pigmentosa 13
CA3005246A1 (en) 2015-12-14 2017-06-22 Cold Spring Harbor Laboratory Compositions and methods for treatment of central nervous system diseases
EP3389672A4 (en) 2015-12-14 2019-08-14 Cold Spring Harbor Laboratory Compositions and methods for treatment of liver diseases
SG11201804443UA (en) 2015-12-14 2018-06-28 Cold Spring Harbor Laboratory Antisense oligomers for treatment of autosomal dominant mental retardation-5 and dravet syndrome
WO2017155058A1 (en) * 2016-03-09 2017-09-14 学校法人東京理科大学 Nucleic acid oligomer for rna hybrid formation
PT3430141T (en) 2016-03-14 2021-02-25 Hoffmann La Roche Oligonucleotides for reduction of pd-l1 expression
MA45471A (en) 2016-04-01 2019-02-06 Avidity Biosciences Llc PHOSPHATIDYLINOSITOL-3-KINASE NUCLEIC ACIDS AND THEIR USES
MA45469A (en) 2016-04-01 2019-02-06 Avidity Biosciences Llc BETA-CATENIN NUCLEIC ACIDS AND THEIR USES
MA45470A (en) 2016-04-01 2019-02-06 Avidity Biosciences Llc KRAS NUCLEIC ACIDS AND THEIR USES
MA45349A (en) 2016-04-01 2019-02-06 Avidity Biosciences Llc EGFR NUCLEIC ACIDS AND THEIR USES
MA45328A (en) 2016-04-01 2019-02-06 Avidity Biosciences Llc NUCLEIC ACID-POLYPEPTIDE COMPOSITIONS AND USES THEREOF
SG11201906200WA (en) 2017-01-06 2019-08-27 Avidity Biosciences Llc Nucleic acid-polypeptide compositions and methods of inducing exon skipping
CN106667176A (en) * 2017-03-01 2017-05-17 戴建华 Drug pillow for treating acne
JP7152408B2 (en) * 2017-03-10 2022-10-12 キアペグ ファーマシューティカルズ アクチエボラグ releasable conjugate
EP3630199A4 (en) 2017-06-02 2021-11-10 Wave Life Sciences Ltd. Oligonucleotide compositions and methods of use thereof
JP2020524485A (en) 2017-06-02 2020-08-20 ウェイブ ライフ サイエンシズ リミテッドWave Life Sciences Ltd. Oligonucleotide composition and method of using the same
EP3642182A4 (en) 2017-06-21 2020-12-09 Wave Life Sciences Ltd. Compounds, compositions and methods for synthesis
US11597744B2 (en) 2017-06-30 2023-03-07 Sirius Therapeutics, Inc. Chiral phosphoramidite auxiliaries and methods of their use
GB201711809D0 (en) 2017-07-21 2017-09-06 Governors Of The Univ Of Alberta Antisense oligonucleotide
AU2018316159A1 (en) 2017-08-08 2020-02-06 Wave Life Sciences Ltd. Oligonucleotide compositions and methods thereof
JP6827148B2 (en) 2017-08-25 2021-02-10 ストーク セラピューティクス,インク. Antisense oligomers for the treatment of conditions and diseases
JP2021500311A (en) 2017-09-18 2021-01-07 ウェーブ ライフ サイエンシーズ リミテッド Techniques for oligonucleotide preparation
WO2019071028A1 (en) 2017-10-04 2019-04-11 Avidity Biosciences Llc Nucleic acid-polypeptide compositions and uses thereof
US11596646B2 (en) 2017-10-12 2023-03-07 Wave Life Sciences Ltd. Oligonucleotide compositions and methods thereof
US11555189B2 (en) 2017-10-18 2023-01-17 Sarepta Therapeutics, Inc. Antisense oligomer compounds
AU2018378812A1 (en) 2017-12-06 2020-07-09 Avidity Biosciences, Inc. Compositions and methods of treating muscle atrophy and myotonic dystrophy
KR102270138B1 (en) * 2018-09-17 2021-06-28 삼성에스디아이 주식회사 Composition for forming silica layer, method for manufacturing silica layer, and silica layer
IL297818A (en) 2018-12-21 2023-01-01 Avidity Biosciences Inc Anti-transferrin receptor antibodies and uses thereof
EP3980436A4 (en) 2019-06-06 2023-12-20 Avidity Biosciences, Inc. Nucleic acid-polypeptide compositions and uses thereof
CA3172111A1 (en) 2020-03-19 2021-09-23 Barbora MALECOVA Compositions and methods of treating facioscapulohumeral muscular dystrophy
EP4126066A1 (en) 2020-03-27 2023-02-08 Avidity Biosciences, Inc. Compositions and methods of treating muscle dystrophy
WO2021231107A1 (en) 2020-05-11 2021-11-18 Stoke Therapeutics, Inc. Opa1 antisense oligomers for treatment of conditions and diseases
EP4153747A2 (en) 2020-05-22 2023-03-29 Wave Life Sciences Ltd. Double stranded oligonucleotide compositions and methods relating thereto
JPWO2022230990A1 (en) * 2021-04-28 2022-11-03
AU2022345098A1 (en) 2021-09-16 2024-04-04 Avidity Biosciences, Inc. Compositions and methods of treating facioscapulohumeral muscular dystrophy
WO2023220737A2 (en) * 2022-05-13 2023-11-16 Adarx Pharmaceuticals, Inc. Oligonucleotides having a synthetic backbone and synthesis thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9605019B2 (en) * 2011-07-19 2017-03-28 Wave Life Sciences Ltd. Methods for the synthesis of functionalized nucleic acids

Family Cites Families (688)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2878264A (en) 1959-03-17 Substituted amino alcohols
CH372667A (en) 1957-09-26 1963-10-31 Robins Co Inc A H Process for the preparation of 3-aryl-3-pyrrolidinols
US3135766A (en) 1961-10-03 1964-06-02 Mead Johnson & Co 3-substituted-3-pyrrolidinols
US3484473A (en) * 1967-05-12 1969-12-16 Buckman Labor Inc Methylene bisesters of thiolsulfonic acids
DE1934150A1 (en) 1968-07-10 1970-01-15 Pennwalt Corp New 1-alkanoyloxy-1,2,4,5-tetrahydro-3H, 3-benzazepine
US3687808A (en) 1969-08-14 1972-08-29 Univ Leland Stanford Junior Synthetic polynucleotides
US3745162A (en) 1970-08-31 1973-07-10 Robins Co Inc A H 1,2,3,4-tetrahydroisoquinoline-2-(thio)-carboxamides
GB1448437A (en) 1973-02-24 1976-09-08 Beecham Group Ltd Diphenylpropylamines
US4022791A (en) 1975-06-03 1977-05-10 Pfizer Inc. 2-Aminomethyl-3,4-dihydronaphthalenes
GB1504424A (en) 1975-08-09 1978-03-22 Beecham Group Ltd Isoquinoline-derived aminoethers
BR7807288A (en) 1977-11-08 1979-06-12 Genentech Inc POLYNUCLEOTIDE SYNTHESIS PROCESS
DD133885B1 (en) 1978-01-04 1981-02-25 Hans Lehmann AGENTS FOR THE CONTROL OF PHYTOPATHOGENIC BACTERIA AND MUSHROOMS
US4500707A (en) 1980-02-29 1985-02-19 University Patents, Inc. Nucleosides useful in the preparation of polynucleotides
US4458066A (en) 1980-02-29 1984-07-03 University Patents, Inc. Process for preparing polynucleotides
US5132418A (en) 1980-02-29 1992-07-21 University Patents, Inc. Process for preparing polynucleotides
US4668777A (en) 1981-03-27 1987-05-26 University Patents, Inc. Phosphoramidite nucleoside compounds
US4415732A (en) 1981-03-27 1983-11-15 University Patents, Inc. Phosphoramidite compounds and processes
US4973679A (en) 1981-03-27 1990-11-27 University Patents, Inc. Process for oligonucleo tide synthesis using phosphormidite intermediates
US4542142A (en) 1982-11-22 1985-09-17 Roussel Uclaf Insecticidal cyclopropane carboxylic acid derivatives with 3-unsaturated-side chain
USRE34069E (en) 1983-08-18 1992-09-15 Biosyntech Gmbh Process for the preparation of oligonucleotides
DE3329892A1 (en) 1983-08-18 1985-03-07 Köster, Hubert, Prof. Dr., 2000 Hamburg METHOD FOR PRODUCING OLIGONUCLEOTIDES
US5118800A (en) 1983-12-20 1992-06-02 California Institute Of Technology Oligonucleotides possessing a primary amino group in the terminal nucleotide
US5643889A (en) 1984-07-11 1997-07-01 Temple University-Of The Commonwealth System Of Pennsylvania Cholesterol conjugates of 2'5'-oligoadenylate derivatives and antiviral uses thereof
FR2567892B1 (en) 1984-07-19 1989-02-17 Centre Nat Rech Scient NOVEL OLIGONUCLEOTIDES, THEIR PREPARATION PROCESS AND THEIR APPLICATIONS AS MEDIATORS IN DEVELOPING THE EFFECTS OF INTERFERONS
US5367066A (en) 1984-10-16 1994-11-22 Chiron Corporation Oligonucleotides with selectably cleavable and/or abasic sites
FR2575751B1 (en) 1985-01-08 1987-04-03 Pasteur Institut NOVEL ADENOSINE DERIVATIVE NUCLEOSIDES, THEIR PREPARATION AND THEIR BIOLOGICAL APPLICATIONS
FR2576898B1 (en) 1985-02-01 1988-01-08 Lafon Labor 3-PHENYL-TETRAHYDROPYRIDINE DERIVATIVES, METHOD OF PREPARATION AND THERAPEUTIC USE
US4659774A (en) 1985-11-01 1987-04-21 American Hoechst Corporation Support for solid-phase oligonucleotide synthesis
US4735949A (en) 1986-02-18 1988-04-05 Warner-Lambert Company Disubstituted-7-pyrrolidinonaphthyridine antibacterial agents
US4840956A (en) 1986-02-18 1989-06-20 Warner-Lambert Company Novel disubstituted-7-pyrrolidinoquinoline antibacterial agents
IL83663A0 (en) 1986-10-27 1988-01-31 Robins Co Inc A H Preparation of 3-pyrrolidinols
EP0366685B1 (en) 1987-06-24 1994-10-19 Howard Florey Institute Of Experimental Physiology And Medicine Nucleoside derivatives
EP0302349B1 (en) 1987-07-30 1993-09-29 Bar Ilan University Biologically active carboxylic acid esters
US4923901A (en) 1987-09-04 1990-05-08 Millipore Corporation Membranes with bound oligonucleotides and peptides
US5175273A (en) 1988-07-01 1992-12-29 Genentech, Inc. Nucleic acid intercalating agents
US4943629A (en) 1988-08-12 1990-07-24 American Cyanamid Company Antidiabetic alpha-substituted phosphonates
US4945158A (en) 1988-08-12 1990-07-31 American Cyanamid Company Antidiabetic phosphonates
US5262530A (en) 1988-12-21 1993-11-16 Applied Biosystems, Inc. Automated system for polynucleotide synthesis and purification
US5047524A (en) 1988-12-21 1991-09-10 Applied Biosystems, Inc. Automated system for polynucleotide synthesis and purification
JP2794461B2 (en) 1989-08-17 1998-09-03 有機合成薬品工業株式会社 Phosphoramidite compounds and solid-phase synthesis of oligoribonucleotides using the same
US5141813A (en) 1989-08-28 1992-08-25 Clontech Laboratories, Inc. Multifunctional controlled pore glass reagent for solid phase oligonucleotide synthesis
US5134066A (en) 1989-08-29 1992-07-28 Monsanto Company Improved probes using nucleosides containing 3-dezauracil analogs
CA2029273A1 (en) 1989-12-04 1991-06-05 Christine L. Brakel Modified nucleotide compounds
US5130302A (en) 1989-12-20 1992-07-14 Boron Bilogicals, Inc. Boronated nucleoside, nucleotide and oligonucleotide compounds, compositions and methods for using same
AU658134B2 (en) 1989-12-28 1995-04-06 Virginia Commonwealth University Sigma receptor ligands and the use thereof
US5852188A (en) 1990-01-11 1998-12-22 Isis Pharmaceuticals, Inc. Oligonucleotides having chiral phosphorus linkages
US5587361A (en) 1991-10-15 1996-12-24 Isis Pharmaceuticals, Inc. Oligonucleotides having phosphorothioate linkages of high chiral purity
JP2580091B2 (en) 1990-01-11 1997-02-12 アイシス・ファーマシューティカルス・インコーポレーテッド Compositions and methods for detecting and modulating RNA activity and gene expression
US6339066B1 (en) 1990-01-11 2002-01-15 Isis Pharmaceuticals, Inc. Antisense oligonucleotides which have phosphorothioate linkages of high chiral purity and which modulate βI, βII, γ, δ, Ε, ζ and η isoforms of human protein kinase C
US5587470A (en) 1990-01-11 1996-12-24 Isis Pharmaceuticals, Inc. 3-deazapurines
US5635488A (en) 1991-10-15 1997-06-03 Isis Pharmaceuticals, Inc. Compounds having phosphorodithioate linkages of high chiral purity
US5506212A (en) 1990-01-11 1996-04-09 Isis Pharmaceuticals, Inc. Oligonucleotides with substantially chirally pure phosphorothioate linkages
US5681941A (en) 1990-01-11 1997-10-28 Isis Pharmaceuticals, Inc. Substituted purines and oligonucleotide cross-linking
US5620963A (en) 1991-10-15 1997-04-15 Isis Pharmaceuticals, Inc. Oligonucleotides for modulating protein kinase C having phosphorothioate linkages of high chiral purity
US5646265A (en) 1990-01-11 1997-07-08 Isis Pharmceuticals, Inc. Process for the preparation of 2'-O-alkyl purine phosphoramidites
US5212295A (en) 1990-01-11 1993-05-18 Isis Pharmaceuticals Monomers for preparation of oligonucleotides having chiral phosphorus linkages
US5914396A (en) 1990-01-11 1999-06-22 Isis Pharmaceuticals, Inc. 2'-O-modified nucleosides and phosphoramidites
US5457191A (en) 1990-01-11 1995-10-10 Isis Pharmaceuticals, Inc. 3-deazapurines
US5459255A (en) 1990-01-11 1995-10-17 Isis Pharmaceuticals, Inc. N-2 substituted purines
US7101993B1 (en) 1990-01-11 2006-09-05 Isis Pharmaceuticals, Inc. Oligonucleotides containing 2′-O-modified purines
US5151510A (en) 1990-04-20 1992-09-29 Applied Biosystems, Inc. Method of synethesizing sulfurized oligonucleotide analogs
US5292875A (en) 1990-04-20 1994-03-08 Lynx Therapeutics, Inc. Method of synthesizing sulfurized oligonucleotide analogs
DK0531447T3 (en) 1990-05-23 2000-03-27 Isis Pharmaceuticals Inc Compositions and Methods for Modulating RNA Activity by Modifying the 5 'Cap Structure of RNA
US5386023A (en) 1990-07-27 1995-01-31 Isis Pharmaceuticals Backbone modified oligonucleotide analogs and preparation thereof through reductive coupling
ATE154246T1 (en) 1990-07-27 1997-06-15 Isis Pharmaceuticals Inc NUCLEASE RESISTANT PYRIMIDINE MODIFIED OLIGONUCLEOTIDES THAT DETECTE AND MODULATE GENE EXPRESSION
US5677437A (en) 1990-07-27 1997-10-14 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5489677A (en) 1990-07-27 1996-02-06 Isis Pharmaceuticals, Inc. Oligonucleoside linkages containing adjacent oxygen and nitrogen atoms
US5792844A (en) 1990-07-27 1998-08-11 Isis Pharmaceuticals, Inc. Oligonucleoside linkages containing adjacent nitrogen atoms
US5618704A (en) 1990-07-27 1997-04-08 Isis Pharmacueticals, Inc. Backbone-modified oligonucleotide analogs and preparation thereof through radical coupling
US5834607A (en) 1990-07-27 1998-11-10 Isis Pharmaceuticals, Inc. Amines and methods of making and using the same
US5610289A (en) 1990-07-27 1997-03-11 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogues
US6087482A (en) 1990-07-27 2000-07-11 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5223618A (en) 1990-08-13 1993-06-29 Isis Pharmaceuticals, Inc. 4'-desmethyl nucleoside analog compounds
US5688941A (en) 1990-07-27 1997-11-18 Isis Pharmaceuticals, Inc. Methods of making conjugated 4' desmethyl nucleoside analog compounds
US6121433A (en) 1990-07-27 2000-09-19 Isis Pharmaceuticals, Inc. Oligomeric compounds having nitrogen-containing linkages
WO1994022886A1 (en) 1993-03-30 1994-10-13 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5138045A (en) 1990-07-27 1992-08-11 Isis Pharmaceuticals Polyamine conjugated oligonucleotides
US5783682A (en) 1990-07-27 1998-07-21 Isis Pharmaceuticals, Inc. Oligonucleotide mimics having nitrogen-containing linkages
US5602240A (en) 1990-07-27 1997-02-11 Ciba Geigy Ag. Backbone modified oligonucleotide analogs
US5608046A (en) 1990-07-27 1997-03-04 Isis Pharmaceuticals, Inc. Conjugated 4'-desmethyl nucleoside analog compounds
US5998603A (en) 1994-09-29 1999-12-07 Isis Pharmaceuticals, Inc. 4'-desmethyl nucleoside analogs, and oligomers thereof
US5623070A (en) 1990-07-27 1997-04-22 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5378825A (en) 1990-07-27 1995-01-03 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogs
US5541307A (en) 1990-07-27 1996-07-30 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogs and solid phase synthesis thereof
US5432272A (en) 1990-10-09 1995-07-11 Benner; Steven A. Method for incorporating into a DNA or RNA oligonucleotide using nucleotides bearing heterocyclic bases
JPH04348044A (en) 1991-02-21 1992-12-03 Matsushita Electron Corp Resin sealing device for semiconductor
US5512668A (en) 1991-03-06 1996-04-30 Polish Academy Of Sciences Solid phase oligonucleotide synthesis using phospholane intermediates
US7015315B1 (en) 1991-12-24 2006-03-21 Isis Pharmaceuticals, Inc. Gapped oligonucleotides
US20020183502A1 (en) 1991-05-21 2002-12-05 Mesmaeker Alain De Backbone-modified oligonucleotide analogs and methods for using same
US6414112B1 (en) 1991-05-24 2002-07-02 Ole Buchardt Peptide nucleic acids having 2,6-diaminopurine nucleobases
JPH04348077A (en) 1991-05-24 1992-12-03 Nec Corp Thin-film transistor
WO1992022655A1 (en) 1991-06-10 1992-12-23 Lucky Limited Hepatitis c diagnostics and vaccines
US5359052A (en) 1991-08-05 1994-10-25 Polish Academy Of Sciences Chalcophospholanes useful in the synthesis of oligonucleoside phosphorothioates, phosphorodithioates and related selenates
US5646267A (en) 1991-08-05 1997-07-08 Polish Academy Of Sciences Method of making oligonucleotides and oligonucleotide analogs using phospholanes and enantiomerically resolved phospholane analogues
US6369209B1 (en) 1999-05-03 2002-04-09 Isis Pharmaceuticals, Inc. Oligonucleotides having A-DNA form and B-DNA form conformational geometry
US7119184B2 (en) 1991-08-12 2006-10-10 Isis Pharmaceuticals, Inc. Oligonucleotides having A-DNA form and B-DNA form conformational geometry
US5654284A (en) 1991-10-15 1997-08-05 Isis Pharmaceuticals, Inc. Oligonucleotides for modulating RAF kinase having phosphorothioate linkages of high chiral purity
EP0655088B1 (en) 1991-10-15 2002-07-24 Isis Pharmaceuticals, Inc. Oligonucleotides having chiral phosphorus linkages
US5661134A (en) 1991-10-15 1997-08-26 Isis Pharmaceuticals, Inc. Oligonucleotides for modulating Ha-ras or Ki-ras having phosphorothioate linkages of high chiral purity
US5607923A (en) 1991-10-15 1997-03-04 Isis Pharmaceuticals, Inc. Oligonucleotides for modulating cytomegalovirus having phosphorothioate linkages of high chiral purity
US5599797A (en) 1991-10-15 1997-02-04 Isis Pharmaceuticals, Inc. Oligonucleotides having phosphorothioate linkages of high chiral purity
US5576302A (en) 1991-10-15 1996-11-19 Isis Pharmaceuticals, Inc. Oligonucleotides for modulating hepatitis C virus having phosphorothioate linkages of high chiral purity
DE59208572D1 (en) 1991-10-17 1997-07-10 Ciba Geigy Ag Bicyclic nucleosides, oligonucleotides, processes for their preparation and intermediates
US5594121A (en) 1991-11-07 1997-01-14 Gilead Sciences, Inc. Enhanced triple-helix and double-helix formation with oligomers containing modified purines
US5484908A (en) 1991-11-26 1996-01-16 Gilead Sciences, Inc. Oligonucleotides containing 5-propynyl pyrimidines
US6235887B1 (en) 1991-11-26 2001-05-22 Isis Pharmaceuticals, Inc. Enhanced triple-helix and double-helix formation directed by oligonucleotides containing modified pyrimidines
US5359044A (en) 1991-12-13 1994-10-25 Isis Pharmaceuticals Cyclobutyl oligonucleotide surrogates
JP3131222B2 (en) 1991-12-24 2001-01-31 アイシス・ファーマシューティカルス・インコーポレーテッド 2 'modified oligonucleotide having a gap
GB9213601D0 (en) 1992-06-26 1992-08-12 Mastico Robert A Protein based delivery system
US7067497B2 (en) 1992-09-29 2006-06-27 Isis Pharmaceuticals, Inc. Modulation of telomere length by oligonucleotides having a G-core sequence
US6005107A (en) 1992-12-23 1999-12-21 Biochem Pharma, Inc. Antiviral compounds
US6444656B1 (en) 1992-12-23 2002-09-03 Biochem Pharma, Inc. Antiviral phosphonate nucleotides
DE69400208T2 (en) 1993-01-25 1996-11-28 Hybridon Inc OLIONUCLEOTIDALKYLPHOSPHONATES AND PHOSPHONOTHIOATES
WO1994022890A1 (en) 1993-03-31 1994-10-13 Sterling Winthop Inc. Novel 5'-substituted nucleosides and oligomers produced therefrom
HU9501978D0 (en) 1993-03-31 1995-09-28 Sterling Winthorp Inc Bifunctional nucleosides, oligomers thereof, and methods of making and using the same
US5955591A (en) 1993-05-12 1999-09-21 Imbach; Jean-Louis Phosphotriester oligonucleotides, amidites and method of preparation
US6015886A (en) 1993-05-24 2000-01-18 Chemgenes Corporation Oligonucleotide phosphate esters
EP0628394B1 (en) 1993-06-10 1998-08-26 Idemitsu Petrochemical Co. Ltd. Injection molding die
US5502177A (en) 1993-09-17 1996-03-26 Gilead Sciences, Inc. Pyrimidine derivatives for labeled binding partners
US5643989A (en) 1993-10-29 1997-07-01 Azdel, Inc. Fiber reinforced functionalized polyolefin composites
US5457187A (en) 1993-12-08 1995-10-10 Board Of Regents University Of Nebraska Oligonucleotides containing 5-fluorouracil
DE4435728A1 (en) 1994-01-19 1995-07-20 Boehringer Mannheim Gmbh Biotin silane compounds and binding matrix containing these compounds
US6117679A (en) 1994-02-17 2000-09-12 Maxygen, Inc. Methods for generating polynucleotides having desired characteristics by iterative selection and recombination
EP0746618B1 (en) 1994-02-22 2002-08-21 Novozymes A/S A method of preparing a variant of a lipolytic enzyme
US5596091A (en) 1994-03-18 1997-01-21 The Regents Of The University Of California Antisense oligonucleotides comprising 5-aminoalkyl pyrimidine nucleotides
AU2445795A (en) 1994-05-11 1995-12-05 Novo Nordisk A/S An enzyme with endo-1,3(4)-beta -glucanase activity
US5525711A (en) 1994-05-18 1996-06-11 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Pteridine nucleotide analogs as fluorescent DNA probes
DE69507197T2 (en) 1994-05-31 1999-05-27 Bayer Ag Aminobenzofuryl and thienyl derivatives
HRP950288A2 (en) 1994-05-31 1997-08-31 Bayer Ag Oxalylamino-benzofuran- and benzothienyl-derivatives
US6207646B1 (en) 1994-07-15 2001-03-27 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US6239116B1 (en) 1994-07-15 2001-05-29 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
CA2560114A1 (en) 1994-07-15 1996-02-01 The University Of Iowa Research Foundation Immunomodulatory oligonucleotides
CA2199464A1 (en) 1994-09-07 1996-03-14 Radhakrishnan Iyer Oligonucleotide prodrugs
US5681940A (en) 1994-11-02 1997-10-28 Icn Pharmaceuticals Sugar modified nucleosides and oligonucleotides
CA2208528A1 (en) 1994-12-22 1996-06-27 Hybridon, Inc. Synthesis of stereospecific oligonucleotide phosphorothioates
GB9501465D0 (en) * 1995-01-25 1995-03-15 King S College London Nucleoside phosphorothioate derivatives,synthesis and use thereof
EP0813539B1 (en) 1995-03-06 2006-05-24 Isis Pharmaceuticals, Inc. Improved process for the synthesis of 2'-o-substituted pyrimidines and oligomeric compounds therefrom
US6166197A (en) 1995-03-06 2000-12-26 Isis Pharmaceuticals, Inc. Oligomeric compounds having pyrimidine nucleotide (S) with 2'and 5 substitutions
EP0739983B1 (en) 1995-04-27 2009-12-30 Takara Bio Inc. Gene encoding lacto-n-biosidase
EP0824588B1 (en) 1995-05-11 2004-07-14 Applied Research Systems ARS Holding N.V. Il-6 activity inhibitor
CA2221589A1 (en) 1995-05-19 1996-11-21 Glycomed Incorporated Collection of activated glycoside compounds and their biological use
DE69609511T2 (en) 1995-05-23 2001-03-29 Hybridon Inc New synthons for stereoselective oligonucleotide synthesis
AU5871196A (en) 1995-05-23 1996-12-24 Hybridon, Inc. Methods and compounds for the synthesis of oligonucleotides and the oligonucleotides thereby produced
WO1996039154A1 (en) 1995-06-06 1996-12-12 Isis Pharmaceuticals, Inc. Oligonucleotides having phosphorothioate linkages of high chiral purity
US5932450A (en) 1995-06-07 1999-08-03 Gen-Probe Incorporated Enzymatic synthesis of oligonucleotides using digestible templates
US5795765A (en) 1995-06-29 1998-08-18 Takara Shuzo Co., Ltd. Gene encoding endoglycoceramidase
US5824503A (en) 1995-06-29 1998-10-20 Takara Shuzo Co, Ltd. Gene encoding endoglycoceramidase activator
US6017700A (en) 1995-08-04 2000-01-25 Bayer Corporation Cationic oligonucleotides, and related methods of synthesis and use
US5936080A (en) 1996-05-24 1999-08-10 Genta Incorporated Compositions and methods for the synthesis of organophosphorus derivatives
WO1997009443A1 (en) 1995-09-05 1997-03-13 Michigan State University PROCESS FOR THE ISOLATION AND PURIFICATION OF TAXOL AND TAXANES FROM TAXUS spp
US5734041A (en) 1995-10-20 1998-03-31 Mcgill University Preparation of chiral phosphorothioate oligomers
US6476216B1 (en) 1995-10-20 2002-11-05 Mcgill University Preparation of phosphorothioate oligomers
US6160109A (en) 1995-10-20 2000-12-12 Isis Pharmaceuticals, Inc. Preparation of phosphorothioate and boranophosphate oligomers
US7018793B1 (en) 1995-12-07 2006-03-28 Diversa Corporation Combinatorial screening of mixed populations of organisms
CA2246503A1 (en) 1996-02-15 1997-08-21 National Institutes Of Health Rnase l activators and antisense oligonucleotides effective to treat rsv infections
US6214805B1 (en) 1996-02-15 2001-04-10 The United States Of America As Represented By The Department Of Health And Human Services RNase L activators and antisense oligonucleotides effective to treat RSV infections
GB9604669D0 (en) 1996-03-05 1996-05-01 Ciba Geigy Ag Chemical compounds
US5824669A (en) 1996-03-22 1998-10-20 Nitromed, Inc. Nitrosated and nitrosylated compounds and compositions and their use for treating respiratory disorders
WO1997043409A2 (en) 1996-05-10 1997-11-20 Novo Nordisk A/S Method of providing novel dna sequences
US5856465A (en) 1996-05-24 1999-01-05 Polska Akademia Nauk Centrum Badan Molekularnych I Makromolekularnych Compositions and methods for the synthesis of chirally pure organophosphorus nucleoside derivatives
US5898031A (en) 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
DE19622783A1 (en) 1996-06-07 1997-12-11 Hoechst Ag Isolation of the biosynthetic genes for pseudo-oligosaccharides from Streptomyces glaucescens GLA.O and their use
JP2000514307A (en) 1996-07-16 2000-10-31 ジェン―プローブ・インコーポレーテッド Methods for detecting and amplifying nucleic acid sequences using modified oligonucleotides with increased target specific T <M>
AU717387B2 (en) 1996-07-24 2000-03-23 Rolf H. Berg Peptide nucleic acids having enhanced binding affinity, sequence specificity and solubility
WO1998007734A1 (en) 1996-08-21 1998-02-26 Hybridon, Inc. Oligonucleotide prodrugs
US6056973A (en) 1996-10-11 2000-05-02 Sequus Pharmaceuticals, Inc. Therapeutic liposome composition and method of preparation
GB9621522D0 (en) 1996-10-16 1996-12-04 Biocompatibles Ltd Synthesis of phosphorus compounds
US6172209B1 (en) 1997-02-14 2001-01-09 Isis Pharmaceuticals Inc. Aminooxy-modified oligonucleotides and methods for making same
US6639062B2 (en) 1997-02-14 2003-10-28 Isis Pharmaceuticals, Inc. Aminooxy-modified nucleosidic compounds and oligomeric compounds prepared therefrom
US6369237B1 (en) 1997-03-07 2002-04-09 President And Fellows Of Harvard College DNA glycosylase inhibitors, and uses related thereto
US6015887A (en) 1997-04-11 2000-01-18 Isis Pharmaceuticals, Inc. Chiral peptide nucleic acids and methods for preparing same
US6468983B2 (en) 1997-04-21 2002-10-22 The Cleveland Clinic Foundation RNase L activators and antisense oligonucleotides effective to treat telomerase-expressing malignancies
PL184612B1 (en) 1997-04-25 2002-11-29 Pan Method of obtaining p-chiral analoques of nucleotides
CA2291839A1 (en) 1997-05-28 1998-12-03 Peter E. Nielsen Conjugated peptide nucleic acids having enhanced cellular uptake
WO1999000377A1 (en) 1997-06-27 1999-01-07 The Procter & Gamble Company Pro-fragrance cyclic acetals
WO1999005160A2 (en) 1997-07-25 1999-02-04 Hybridon, Inc. Oligonuclotides having 3' terminal stereospecific phosphorothioates
US6767739B2 (en) 2001-07-30 2004-07-27 Isis Pharmaceuticals Inc. Antisense modulation of microsomal triglyceride transfer protein expression
GB9717158D0 (en) 1997-08-13 1997-10-22 King S College London Solution synthesis of oligonucleotides and their phosphorothioate analogues
US6383808B1 (en) 2000-09-11 2002-05-07 Isis Pharmaceuticals, Inc. Antisense inhibition of clusterin expression
US6750344B1 (en) 1997-09-05 2004-06-15 Isis Pharmaceuticals, Inc. Amine compounds and combinatorial libraries comprising same
US6794499B2 (en) 1997-09-12 2004-09-21 Exiqon A/S Oligonucleotide analogues
DE19741715A1 (en) 1997-09-22 1999-03-25 Hoechst Ag New pentopyranosyl nucleoside compounds
US6232463B1 (en) 1997-10-09 2001-05-15 Isis Pharmaceuticals, Inc. Substituted purines and oligonucleotide cross-linking
US6528640B1 (en) 1997-11-05 2003-03-04 Ribozyme Pharmaceuticals, Incorporated Synthetic ribonucleic acids with RNAse activity
US6617438B1 (en) 1997-11-05 2003-09-09 Sirna Therapeutics, Inc. Oligoribonucleotides with enzymatic activity
US6080543A (en) 1997-12-08 2000-06-27 E. & J. Gallo Winery Detection of fungal pathogens
US6582936B1 (en) 1997-12-12 2003-06-24 The Regents Of The University Of California Methods for making nucleic acids
US6248519B1 (en) 1998-03-11 2001-06-19 E & J Gallo Winery Detection of fermentation-related microorganisms
US7045610B2 (en) 1998-04-03 2006-05-16 Epoch Biosciences, Inc. Modified oligonucleotides for mismatch discrimination
AU3884199A (en) 1998-05-06 1999-11-23 Ottawa Health Research Institute Methods for the prevention and treatment of parasitic infections and related diseases using cpg oligonucleotides
DE69927495T2 (en) 1998-05-14 2006-07-06 Coley Pharmaceutical Group, Inc., Wellesley METHOD OF REGULATING HEMATOPOESIS BY USING CPG OLIGONUCLEOTIDES
US6242589B1 (en) 1998-07-14 2001-06-05 Isis Pharmaceuticals, Inc. Phosphorothioate oligonucleotides having modified internucleoside linkages
US6867294B1 (en) 1998-07-14 2005-03-15 Isis Pharmaceuticals, Inc. Gapped oligomers having site specific chiral phosphorothioate internucleoside linkages
EP1100807A1 (en) 1998-07-27 2001-05-23 University Of Iowa Research Foundation STEREOISOMERS OF CpG OLIGONUCLEOTIDES AND RELATED METHODS
DE69929444T2 (en) 1998-08-10 2006-09-28 Antigenics Inc., Woburn CPG COMPOSITIONS, SAPONIN ADJUVANTIES, AND METHOD OF USE THEREOF
WO2000023444A1 (en) 1998-10-21 2000-04-27 Abbott Laboratories 5,7-disubstituted-4-aminopyrido[2,3-d]pyrimidine compounds
US6995259B1 (en) 1998-10-23 2006-02-07 Sirna Therapeutics, Inc. Method for the chemical synthesis of oligonucleotides
US6451524B1 (en) 1998-11-25 2002-09-17 Isis Pharmaceuticals, Inc. Identification of disease predictive nucleic acids
WO2000031110A1 (en) 1998-11-25 2000-06-02 Isis Pharmaceuticals, Inc. Identification of disease predictive nucleic acids
CA2353789C (en) 1998-12-21 2012-02-14 Genencor International, Inc. Chemically modified enzymes with multiple charged variants
CA2702148C (en) 1999-01-06 2014-03-04 Genenews Inc. Method of profiling gene expression in a human subject having an infectious disease
US6265172B1 (en) 1999-02-08 2001-07-24 University Of Kentucky Diagnostic test and therapy for manganese superoxide dismutate (mNsod) associated diseases
US6121437A (en) 1999-03-16 2000-09-19 Isis Pharmaceuticals, Inc. Phosphate and thiophosphate protecting groups
US6506594B1 (en) 1999-03-19 2003-01-14 Cornell Res Foundation Inc Detection of nucleic acid sequence differences using the ligase detection reaction with addressable arrays
GB9907245D0 (en) 1999-03-29 1999-05-26 Goldsborough Andrew Cleavage of nucleic acids from solid supports
JP3072345B1 (en) 1999-03-31 2000-07-31 農林水産省家畜衛生試験場長 Swine erysipelas recombinant subunit vaccine
US5998148A (en) 1999-04-08 1999-12-07 Isis Pharmaceuticals Inc. Antisense modulation of microtubule-associated protein 4 expression
US6977245B2 (en) 1999-04-12 2005-12-20 The United States Of America As Represented By The Department Of Health And Human Services Oligodeoxynucleotide and its use to induce an immune response
US6300069B1 (en) 1999-05-03 2001-10-09 Qiagen Gmbh Generation and amplification of nucleic acids from ribonucleic acids
US6656730B1 (en) 1999-06-15 2003-12-02 Isis Pharmaceuticals, Inc. Oligonucleotides conjugated to protein-binding drugs
US6066500A (en) 1999-06-25 2000-05-23 Isis Pharmaceuticals Inc. Antisense modulation of Beta catenin expression
US6271004B1 (en) 1999-06-25 2001-08-07 Display Systems Biotech A/S Method for improved reverse transcription at high temperatures
US6414135B1 (en) 1999-07-07 2002-07-02 Isis Pharmaceuticals, Inc. C3′-methylene hydrogen phosphonate monomers and related compounds
US20030092647A1 (en) 2001-08-08 2003-05-15 Crooke Rosanne M. Antisense modulation of cholesteryl ester transfer protein expression
US6147200A (en) 1999-08-19 2000-11-14 Isis Pharmaceuticals, Inc. 2'-O-acetamido modified monomers and oligomers
US7264932B2 (en) 1999-09-24 2007-09-04 Applera Corporation Nuclease inhibitor cocktail
JP2003510282A (en) 1999-09-25 2003-03-18 ユニバーシティ オブ アイオワ リサーチ ファウンデーション Immunostimulatory nucleic acids
ATE333284T1 (en) 1999-09-27 2006-08-15 Coley Pharm Group Inc METHOD USING IMMUNOSTIMULATIVE INTERFERON INDUCED BY NUCLEIC ACIDS
US6949520B1 (en) 1999-09-27 2005-09-27 Coley Pharmaceutical Group, Inc. Methods related to immunostimulatory nucleic acid-induced interferon
US20020082227A1 (en) 1999-09-30 2002-06-27 Scott Henry Use of oligonucleotides for inhibition of complement activation
US6309836B1 (en) * 1999-10-05 2001-10-30 Marek Kwiatkowski Compounds for protecting hydroxyls and methods for their use
AU7863200A (en) 1999-10-06 2001-05-10 Quark Biotech, Inc. Method for enrichment of natural antisense messenger rna
GB9924285D0 (en) 1999-10-14 1999-12-15 Avecia Ltd Process
US20010055761A1 (en) 1999-10-29 2001-12-27 Agilent Technologies Small scale dna synthesis using polymeric solid support with functionalized regions
FR2800750B1 (en) 1999-11-05 2003-01-31 Centre Nat Rech Scient MEMBRANE PROTEINS CTL (CHOLINE TRANSPORTER LIKE) INVOLVED IN THE TRANSPORT OF CHOLINE
AU1656601A (en) 1999-11-12 2001-06-12 Isis Pharmaceuticals, Inc. Gapped oligomers having site specific chiral phosphorothioate internucleoside linkages
US6322985B1 (en) 1999-12-27 2001-11-27 Technion Research And Development Foundation Ltd. Abundant, well distributed and hyperpolymorphic simple sequence repeats in prokaryote genomes and use of same for prokaryote classification and typing
EP1244906A1 (en) 1999-12-30 2002-10-02 Cabot Corporation Sensors with improved properties
WO2001050349A1 (en) 1999-12-30 2001-07-12 Rutgers, The State University Of New Jersey Electronic document customization and transformation utilizing user feedback
US6649750B1 (en) 2000-01-05 2003-11-18 Isis Pharmaceuticals, Inc. Process for the preparation of oligonucleotide compounds
US6159697A (en) 2000-01-19 2000-12-12 Isis Pharmaceuticals, Inc. Antisense modulation of Smad7 expression
US7585847B2 (en) 2000-02-03 2009-09-08 Coley Pharmaceutical Group, Inc. Immunostimulatory nucleic acids for the treatment of asthma and allergy
US6495677B1 (en) 2000-02-15 2002-12-17 Kanda S. Ramasamy Nucleoside compounds
US6936432B2 (en) 2000-03-01 2005-08-30 Message Pharmaceuticals Bacterial RNase P proteins and their use in identifying antibacterial compounds
GB0004889D0 (en) * 2000-03-01 2000-04-19 Avecia Ltd Synthesis of oligonucleotides
JP2003528068A (en) 2000-03-17 2003-09-24 コリクサ コーポレイション New amphiphilic aldehydes and their use as adjuvants and immune effectors
MXPA02010261A (en) 2000-04-20 2003-04-25 Hoffmann La Roche Pyrrolidine and piperidine derivatives and their use for the treament of neurodegenerative disorders.
DE10019756A1 (en) 2000-04-20 2001-10-25 Bayer Ag Process for the production of superabsorbent polymers from polyacrylonitriles
US20020013287A1 (en) 2000-05-09 2002-01-31 Reliable Biopharmaceuticals, Inc. St Louis Missouri Polymeric compounds useful as prodrugs
US6492171B2 (en) 2000-05-16 2002-12-10 Isis Pharmaceuticals, Inc. Antisense modulation of TERT expression
US6815542B2 (en) 2000-06-16 2004-11-09 Ribapharm, Inc. Nucleoside compounds and uses thereof
AU2001278517A1 (en) 2000-08-03 2002-02-18 F. Hoffman-La Roche Ag Nucleic acid binding compounds containing pyrazolo(3,4-d)pyrimidine analogues of purin-2,6-diamine and their uses
US6725412B1 (en) 2000-08-15 2004-04-20 Dolby Laboratories Licensing Corporation Low latency data encoder
US6809195B1 (en) 2000-08-16 2004-10-26 Isis Pharmaceuticals, Inc. Process for the preparation of oligonucleotides
US6559279B1 (en) 2000-09-08 2003-05-06 Isis Pharmaceuticals, Inc. Process for preparing peptide derivatized oligomeric compounds
WO2002022809A2 (en) 2000-09-15 2002-03-21 Coley Pharmaceutical Gmbh PROCESS FOR HIGH THROUGHPUT SCREENING OF CpG-BASED IMMUNO-AGONIST/ANTAGONIST
EP1191097A1 (en) 2000-09-21 2002-03-27 Leids Universitair Medisch Centrum Induction of exon skipping in eukaryotic cells
GB0024752D0 (en) 2000-10-10 2000-11-22 Univ Belfast Oxidative halogenation of aromatic compounds
AU2002244337B2 (en) 2000-10-18 2005-08-11 Glaxosmithkline Biologicals S.A. Vaccines
US6372492B1 (en) 2000-10-30 2002-04-16 Isis Pharmaceuticals, Inc. Antisense modulation of talin expression
US6682889B1 (en) 2000-11-08 2004-01-27 Becton, Dickinson And Company Amplification and detection of organisms of the Chlamydiaceae family
NL1016978C2 (en) 2000-12-22 2002-06-25 Robert Jan Colenbrander Device and method for packaging and preparing food and method for manufacturing such a device.
DK1355916T3 (en) 2001-01-22 2007-05-07 Merck & Co Inc Nucleoside derivatives as inhibitors of RNA-dependent RNA viral polymerase
US8008459B2 (en) 2001-01-25 2011-08-30 Evolva Sa Concatemers of differentially expressed multiple genes
US7838287B2 (en) 2001-01-25 2010-11-23 Evolva Sa Library of a collection of cells
WO2002059294A1 (en) 2001-01-26 2002-08-01 Commonwealth Scientific And Industrial Research O Rganisation Methods and means for producing efficient silencing construct using recombinational cloning
US20050277133A1 (en) 2001-05-18 2005-12-15 Sirna Therapeutics, Inc. RNA interference mediated treatment of polyglutamine (polyQ) repeat expansion diseases using short interfering nucleic acid (siNA)
US20030207804A1 (en) 2001-05-25 2003-11-06 Muthiah Manoharan Modified peptide nucleic acids
GB0113523D0 (en) 2001-06-04 2001-07-25 Torotrak Dev Ltd An Hydraulic control circuit for a continuosly variable transmission
US20030069410A1 (en) 2001-06-14 2003-04-10 Isis Pharmaceuticals, Inc. Methods for preparing oligonucleotides having chiral phosphorothioate linkages
US20050019915A1 (en) 2001-06-21 2005-01-27 Bennett C. Frank Antisense modulation of superoxide dismutase 1, soluble expression
US20030138458A1 (en) 2001-06-29 2003-07-24 Michael Houghton HCV E1E2 vaccine compositions
CA2452458A1 (en) 2001-07-03 2003-01-16 Isis Pharmaceuticals, Inc. Nuclease resistant chimeric oligonucleotides
US7205399B1 (en) 2001-07-06 2007-04-17 Sirna Therapeutics, Inc. Methods and reagents for oligonucleotide synthesis
US6440739B1 (en) 2001-07-17 2002-08-27 Isis Pharmaceuticals, Inc. Antisense modulation of glioma-associated oncogene-2 expression
US7425545B2 (en) 2001-07-25 2008-09-16 Isis Pharmaceuticals, Inc. Modulation of C-reactive protein expression
US6455308B1 (en) 2001-08-01 2002-09-24 Isis Pharmaceuticals, Inc. Antisense modulation of serum amyloid A4 expression
US7888324B2 (en) 2001-08-01 2011-02-15 Genzyme Corporation Antisense modulation of apolipoprotein B expression
US7407943B2 (en) 2001-08-01 2008-08-05 Isis Pharmaceuticals, Inc. Antisense modulation of apolipoprotein B expression
US7227014B2 (en) 2001-08-07 2007-06-05 Isis Pharmaceuticals, Inc. Antisense modulation of apolipoprotein (a) expression
US7259150B2 (en) 2001-08-07 2007-08-21 Isis Pharmaceuticals, Inc. Modulation of apolipoprotein (a) expression
AU2002361468A1 (en) 2001-08-14 2003-03-18 The Government Of The United States Of America As Represented By The Secretary Of Health And Human S Method for rapid generation of mature dendritic cells
CA2458393A1 (en) 2001-08-24 2003-03-06 Massachusetts Institute Of Technology Reagents that facilitate the purification of compounds synthesized on a solid support
US7049122B2 (en) 2001-09-21 2006-05-23 Academia Sinica Mutant-type lipases and applications thereof
US6933288B2 (en) 2002-02-04 2005-08-23 Isis Pharmaceuticals, Inc. Pyranosyl cytosines: pharmaceutical formulations and methods
JP4348044B2 (en) 2002-02-12 2009-10-21 株式会社キラルジェン Method for producing highly stereoregular dinucleoside phosphorothioates
US20030159938A1 (en) 2002-02-15 2003-08-28 George Hradil Electroplating solution containing organic acid complexing agent
US20040149587A1 (en) 2002-02-15 2004-08-05 George Hradil Electroplating solution containing organic acid complexing agent
US8232383B2 (en) 2002-02-20 2012-07-31 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US20050096284A1 (en) 2002-02-20 2005-05-05 Sirna Therapeutics, Inc. RNA interference mediated treatment of polyglutamine (polyQ) repeat expansion diseases using short interfering nucleic acid (siNA)
JP2005525358A (en) 2002-02-28 2005-08-25 ビオタ インコーポレーティッド Nucleotide mimetics and their prodrugs
EP1485396A2 (en) 2002-02-28 2004-12-15 Biota, Inc. Nucleoside 5'-monophosphate mimics and their prodrugs
US7288376B2 (en) 2002-03-22 2007-10-30 Council Of Scientific And Industrial Research Method of detection of SP-A2 gene variants useful for prediction of predisposition to aspergillosis
US20040102394A1 (en) 2002-11-23 2004-05-27 Isis Pharmaceuticals Inc. Modulation of huntingtin interacting protein 2 expression
US7247621B2 (en) 2002-04-30 2007-07-24 Valeant Research & Development Antiviral phosphonate compounds and methods therefor
AU2003237875A1 (en) 2002-05-15 2003-12-02 Isis Pharmaceuticals, Inc. Antisense modulation of apolipoprotein b expression
US20040014957A1 (en) 2002-05-24 2004-01-22 Anne Eldrup Oligonucleotides having modified nucleoside units
AU2003241621A1 (en) 2002-05-24 2003-12-12 Isis Pharmaceuticals, Inc. Oligonucleotides having modified nucleoside units
US7507808B2 (en) 2002-12-12 2009-03-24 Isis Pharmaceuticals, Inc. Modulation of endothelial lipase expression
WO2003106477A1 (en) 2002-06-01 2003-12-24 Isis Pharmaceuticals, Inc. Oligomeric compounds that include carbocyclic nucleosides and their use in gene modulation
WO2004000351A1 (en) 2002-06-20 2003-12-31 Cytos Biotechnology Ag Packaged virus-like particles for use as adjuvants: method of preparation and use
WO2004003228A1 (en) 2002-07-01 2004-01-08 Unisearch Limited Genotyping method
CA2489174C (en) 2002-07-10 2013-02-05 Thomas Tuschl Rna-interference by single-stranded rna molecules
US20040023905A1 (en) 2002-07-31 2004-02-05 Isis Pharmaceuticals Inc. Antisense modulation of LAR expression
US20050255086A1 (en) 2002-08-05 2005-11-17 Davidson Beverly L Nucleic acid silencing of Huntington's Disease gene
US20080274989A1 (en) 2002-08-05 2008-11-06 University Of Iowa Research Foundation Rna Interference Suppression of Neurodegenerative Diseases and Methods of Use Thereof
US20050042646A1 (en) 2002-08-05 2005-02-24 Davidson Beverly L. RNA interference suppresion of neurodegenerative diseases and methods of use thereof
WO2004014933A1 (en) 2002-08-07 2004-02-19 University Of Massachusetts Compositions for rna interference and methods of use thereof
AU2003259735A1 (en) 2002-08-08 2004-02-25 Sirna Therapeutics, Inc. Small-mer compositions and methods of use
AR040996A1 (en) 2002-08-19 2005-04-27 Coley Pharm Group Inc IMMUNE STIMULATING NUCLEIC ACIDS
US7414116B2 (en) 2002-08-23 2008-08-19 Illumina Cambridge Limited Labelled nucleotides
BR0314236A (en) 2002-09-13 2005-08-09 Replicor Inc Oligonucleotide formulation, pharmaceutical composition, kit, antiviral compound, preparation of oligonucleotide and methods for selection of an antiviral oligonucleotide for use as an antiviral agent, for prophylaxis or treatment of a viral infection in a patient, for prophylactic treatment of cancer caused by oncoviruses. for identifying a compound that alters the binding of an oligonucleotide to at least one viral component, for purifying oligonucleotide binding to at least one viral component and for enriching oligonucleotides from an oligonucleotide cluster
US7572604B2 (en) 2002-10-07 2009-08-11 Isis Innovation Limited Modified carbohydrate processing enzyme
US7030230B2 (en) 2002-10-25 2006-04-18 Isis Pharmaceuticals, Inc. Process of purifying phosphoramidites
CN1753687A (en) 2002-10-29 2006-03-29 科勒制药集团股份有限公司 Use of cpg oligonucleotides in the treatment of hepatitis c virus infection
US20040147020A1 (en) 2002-11-01 2004-07-29 The Regents Of The University Of Colorado, A Body Corporate Dopamine neurons from human embryonic stem cells
US7696345B2 (en) 2002-11-05 2010-04-13 Isis Pharmaceuticals, Inc. Polycyclic sugar surrogate-containing oligomeric compounds and compositions for use in gene modulation
AU2003291721A1 (en) 2002-11-05 2004-06-03 Isis Pharmaceuticals, Inc. Phosphorous-linked oligomeric compounds and their use in gene modulation
CA2505090A1 (en) 2002-11-05 2004-05-27 Isis Pharmaceuticals, Inc. Conjugated oligomeric compounds and their use in gene modulation
CA2504929C (en) 2002-11-05 2014-07-22 Charles Allerson Compositions comprising alternating 2'-modified nucleosides for use in gene modulation
US7381527B2 (en) 2002-11-06 2008-06-03 Council Of Scientific And Industrial Research Method of detection of SP-A2 gene variants
US7511131B2 (en) 2002-11-13 2009-03-31 Genzyme Corporation Antisense modulation of apolipoprotein B expression
JP4986109B2 (en) 2002-11-13 2012-07-25 ジェンザイム・コーポレーション Antisense regulation of apolipoprotein B expression
EP2314691A3 (en) 2002-11-14 2012-01-18 Dharmacon, Inc. Fuctional and hyperfunctional siRNA
EP2216407B1 (en) 2003-03-07 2016-01-13 Alnylam Pharmaceuticals, Inc. Therapeutic compositions
WO2004080466A1 (en) 2003-03-07 2004-09-23 Ribapharm Inc. Cytidine analogs and methods of use
CA2524255C (en) 2003-03-21 2014-02-11 Academisch Ziekenhuis Leiden Modulation of exon recognition in pre-mrna by interfering with the secondary rna structure
GB0306657D0 (en) 2003-03-24 2003-04-30 Avecia Ltd Process and compounds
US7537767B2 (en) 2003-03-26 2009-05-26 Cytis Biotechnology Ag Melan-A- carrier conjugates
RU2351362C2 (en) 2003-03-26 2009-04-10 Цитос Байотекнолоджи Аг CONJUGATES OF PEPTIDE Melan-A, VIRUS-LIKE PARTICLE ANALOGUE
ITRM20030149A1 (en) 2003-04-02 2004-10-03 Giuliani Spa ANTISENSE OLIGONUCLEOTIDES (ODN) FOR SMAD7 AND THEIR USE IN THE MEDICAL FIELD
US7598227B2 (en) 2003-04-16 2009-10-06 Isis Pharmaceuticals Inc. Modulation of apolipoprotein C-III expression
US20050261237A1 (en) 2003-04-25 2005-11-24 Boojamra Constantine G Nucleoside phosphonate analogs
BRPI0409680A (en) 2003-04-25 2006-04-18 Gilead Sciences Inc anti-cancer phosphonate analogs
US7432261B2 (en) 2003-04-25 2008-10-07 Gilead Sciences, Inc. Anti-inflammatory phosphonate compounds
CN101410120A (en) 2003-04-25 2009-04-15 吉里德科学公司 Anti-inflammatory phosphonate compounds
US7452901B2 (en) 2003-04-25 2008-11-18 Gilead Sciences, Inc. Anti-cancer phosphonate analogs
WO2004096233A2 (en) 2003-04-25 2004-11-11 Gilead Sciences, Inc. Nucleoside phosphonate conjugates
US20090247488A1 (en) 2003-04-25 2009-10-01 Carina Cannizzaro Anti-inflammatory phosphonate compounds
WO2005002626A2 (en) 2003-04-25 2005-01-13 Gilead Sciences, Inc. Therapeutic phosphonate compounds
US7470724B2 (en) 2003-04-25 2008-12-30 Gilead Sciences, Inc. Phosphonate compounds having immuno-modulatory activity
WO2004096286A2 (en) 2003-04-25 2004-11-11 Gilead Sciences, Inc. Antiviral phosphonate analogs
US7407965B2 (en) 2003-04-25 2008-08-05 Gilead Sciences, Inc. Phosphonate analogs for treating metabolic diseases
US7045306B2 (en) 2003-04-28 2006-05-16 The General Hospital Corporation Method for identifying compounds in vitro that modulate the dysregulation of transcription of transcription mediated by mutant huntingtin protein
US7214491B2 (en) 2003-05-07 2007-05-08 E. I. Du Pont De Nemours And Company Δ-12 desaturase gene suitable for altering levels of polyunsaturated fatty acids in oleaginous yeasts
AU2004239114B2 (en) 2003-05-14 2008-03-13 Japan Science And Technology Agency Inhibition of the expression of huntingtin gene
AU2004253455B2 (en) 2003-06-03 2011-03-03 Eli Lilly And Company Modulation of survivin expression
KR20060016817A (en) 2003-06-20 2006-02-22 콜리 파마슈티칼 게엠베하 Small molecule toll-like receptor (tlr) antagonists
CA2533701A1 (en) 2003-07-31 2005-02-17 Isis Pharmaceuticals, Inc. Oligomeric compounds and compositions for use in modulation of small non-coding rnas
JP2005089441A (en) 2003-08-08 2005-04-07 Toudai Tlo Ltd Manufacturing method of highly stereoregular phosphorus atom-modified nucleotide analogue
JP2011088935A (en) 2003-08-08 2011-05-06 Chiralgen Ltd Optically-active nucleoside 3'-phosphoroamidite for production of phosphorus atom modified nucleotide analog
US7825235B2 (en) 2003-08-18 2010-11-02 Isis Pharmaceuticals, Inc. Modulation of diacylglycerol acyltransferase 2 expression
MXPA06001976A (en) 2003-08-21 2006-05-31 Univ Griffith Novel sulfenamides.
RU2006108577A (en) 2003-08-21 2007-09-27 Гриффит Юниверсити (Au) NEW SULFENAMIDES
CN1863813B (en) 2003-08-27 2011-03-30 生物区科学管理控股有限公司 Novel tricyclic nucleosides or nucleotides as therapeutic agents
JP4731324B2 (en) 2003-08-28 2011-07-20 武 今西 N-O bond cross-linked novel artificial nucleic acid
JP4616175B2 (en) 2003-09-02 2011-01-19 株式会社キラルジェン Method for producing 5'-phosphitylated monomer and H-phosphonate oligonucleotide derivative
JP4580870B2 (en) 2003-09-02 2010-11-17 株式会社キラルジェン Method for producing ribonucleotide or ribonucleotide derivative
US20050053981A1 (en) 2003-09-09 2005-03-10 Swayze Eric E. Gapped oligomeric compounds having linked bicyclic sugar moieties at the termini
CN100393209C (en) 2003-09-09 2008-06-11 杰龙公司 Modified oligonucleotides for telomerase inhibition
US20050074801A1 (en) 2003-09-09 2005-04-07 Monia Brett P. Chimeric oligomeric compounds comprising alternating regions of northern and southern conformational geometry
US8680063B2 (en) 2003-09-12 2014-03-25 University Of Massachusetts RNA interference for the treatment of gain-of-function disorders
ES2485848T3 (en) 2003-09-12 2014-08-14 University Of Massachusetts RNA interference for the treatment of disorders related to function gain
GB0323968D0 (en) 2003-10-13 2003-11-19 Glaxosmithkline Biolog Sa Immunogenic compositions
US8188254B2 (en) 2003-10-30 2012-05-29 Coley Pharmaceutical Gmbh C-class oligonucleotide analogs with enhanced immunostimulatory potency
US20050239102A1 (en) 2003-10-31 2005-10-27 Verdine Gregory L Nucleic acid binding oligonucleotides
US7846436B2 (en) 2003-11-28 2010-12-07 Chemgenes Corporation Oligonucleotides and related compounds
WO2005063983A1 (en) 2003-12-29 2005-07-14 Galapagos Genomics N.V. Modulators of bone homeostasis identified in a high-throughput screen
WO2005070859A1 (en) 2004-01-27 2005-08-04 Takeshi Wada Fluorous supports and processes for production of oligonucleotide derivatives with the same
US20050176045A1 (en) 2004-02-06 2005-08-11 Dharmacon, Inc. SNP discriminatory siRNA
US20080221303A1 (en) 2004-02-18 2008-09-11 Jehoshua Katzhendler Method for the Preparation of Peptide-Oligonucleotide Conjugates
JP3976742B2 (en) 2004-02-27 2007-09-19 江守商事株式会社 Immunostimulatory oligonucleotides that induce interferon alpha
WO2005085272A1 (en) 2004-03-05 2005-09-15 Takeshi Wada Boranophosphate monomer and process for producing oligonucleotide derivative therefrom
WO2005092909A1 (en) 2004-03-25 2005-10-06 Toudai Tlo, Ltd. Processes for producing ribonucleotide analogue with high stereoregularity and deoxyribonucleotide analogue
US20050244869A1 (en) 2004-04-05 2005-11-03 Brown-Driver Vickie L Modulation of transthyretin expression
AU2005230684B2 (en) 2004-04-05 2011-10-06 Alnylam Pharmaceuticals, Inc. Process and reagents for oligonucleotide synthesis and purification
TWI350168B (en) 2004-05-07 2011-10-11 Incyte Corp Amido compounds and their use as pharmaceuticals
EP1753881A2 (en) 2004-05-27 2007-02-21 The Government of the United States of America as represented by The Secretary of the Department of Health and Human Services Differential expression of molecules associated with acute stroke
US7759318B1 (en) 2004-05-28 2010-07-20 Isis Pharmaceuticals, Inc. Identification of novel pathways, genes and promoter motifs regulating adipogenesis
AU2005252663B2 (en) 2004-06-03 2011-07-07 Isis Pharmaceuticals, Inc. Double strand compositions comprising differentially modified strands for use in gene modulation
EP2500430B1 (en) 2004-06-28 2017-03-08 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
PT1786472E (en) 2004-08-10 2013-03-06 Genzyme Corp Antisense modulation of apolipoprotein b expression
ATE556085T1 (en) 2004-08-26 2012-05-15 Nippon Shinyaku Co Ltd PHOSPHORAMIDITE COMPOUND AND METHOD FOR PRODUCING OLIGO-RNA
US20070066551A1 (en) 2004-09-07 2007-03-22 Keefe Anthony D Aptamer medicinal chemistry
US7884086B2 (en) 2004-09-08 2011-02-08 Isis Pharmaceuticals, Inc. Conjugates for use in hepatocyte free uptake assays
WO2006031461A2 (en) 2004-09-09 2006-03-23 Isis Pharmaceuticals, Inc. Pyrrolidinyl groups for attaching conjugates to oligomeric compounds
EP2397563A3 (en) 2004-09-17 2012-07-18 Isis Pharmaceuticals, Inc. Enhanced antisense oligonucleotides
US20060089325A1 (en) 2004-10-13 2006-04-27 Sanjay Bhanot Antisense modulation of PTP1B expression
US7622451B2 (en) 2004-11-03 2009-11-24 University Of Kansas Novobiocin analogues as neuroprotective agents and in the treatment of autoimmune disorders
US8212012B2 (en) 2004-11-03 2012-07-03 University Of Kansas Novobiocin analogues having modified sugar moieties
EP1807440B1 (en) 2004-11-03 2020-02-19 The University of Kansas Novobiocin analogues as anticancer agents
US9120774B2 (en) 2004-11-03 2015-09-01 University Of Kansas Novobiocin analogues having modified sugar moieties
US8212011B2 (en) 2004-11-03 2012-07-03 University Of Kansas Novobiocin analogues
KR100721928B1 (en) 2004-11-05 2007-05-28 주식회사 바이오씨에스 Pharmaceutical composition for treating or preventing dermatitis comprising CpG oligodeoxynucleotide
EP1657307A1 (en) 2004-11-16 2006-05-17 Immunotech S.A. Oligonucleotides that induce the secretion of GM-CSF
AU2005313883B2 (en) 2004-12-09 2011-03-31 Alnylam Pharmaceuticals, Inc. Compositions and methods for inducing an immune response in a mammal and methods of avoiding an immune response to oligonucleotide agents such as short interfering RNAs
US9809824B2 (en) 2004-12-13 2017-11-07 The United States Of America, Represented By The Secretary, Department Of Health And Human Services CpG oligonucleotide prodrugs, compositions thereof and associated therapeutic methods
WO2006066260A2 (en) 2004-12-17 2006-06-22 Thiosense, Inc. Compositions of and methods for producing phosphorus-chiral monomers and oligomers
US20070099851A1 (en) 2004-12-30 2007-05-03 Linn Gregory S Stable analogues of ribose-1-phosphate and methods for treating diabetes and other metabolic disorders
US20060183763A1 (en) 2004-12-31 2006-08-17 Pfizer Inc Novel pyrrolidyl derivatives of heteroaromatic compounds
KR100980898B1 (en) 2005-01-28 2010-09-07 권형주 Oligonucleotides derived from mycobacterium for stimulating immune function, treating immune-related diseases, atopic dermatitis and/or protecting normal immune cell
CA2598992A1 (en) 2005-02-24 2006-08-31 Coley Pharmaceutical Group, Inc. Immunostimulatory oligonucleotides
WO2006137953A1 (en) 2005-04-01 2006-12-28 The Regents Of The Univerisity Of California Phosphono-pent-2-en-1-yl nucleosides and analogs
SI1877070T1 (en) 2005-05-05 2009-06-30 Antisense Pharma Gmb H Therapeutic use of tgf-beta2 antisense oligonucleotides
US20060257912A1 (en) 2005-05-06 2006-11-16 Medtronic, Inc. Methods and sequences to suppress primate huntington gene expression
US7902352B2 (en) 2005-05-06 2011-03-08 Medtronic, Inc. Isolated nucleic acid duplex for reducing huntington gene expression
DK1910395T3 (en) 2005-06-23 2013-02-04 Isis Pharmaceuticals Inc Compositions and Methods for Modulating SMN2 Splicing
US9133517B2 (en) 2005-06-28 2015-09-15 Medtronics, Inc. Methods and sequences to preferentially suppress expression of mutated huntingtin
EP2316967A1 (en) 2005-06-28 2011-05-04 Medtronic, Inc. Methods and sequences to preferentially suppress expression of mutated huntingtin gene.
EP1948600B1 (en) 2005-07-05 2014-04-16 The President & Fellows Of Harvard College Liver targeted conjugates
JP4984634B2 (en) 2005-07-21 2012-07-25 ソニー株式会社 Physical information acquisition method and physical information acquisition device
WO2007016379A2 (en) 2005-07-28 2007-02-08 Id-Fish Technology, Inc. Method for improving cell permeability to foreign particles
EP2338991B1 (en) 2005-08-29 2017-01-18 Regulus Therapeutics Inc. Methods for use in modulating MIR-122a
EP1937312B1 (en) 2005-08-30 2016-06-29 Ionis Pharmaceuticals, Inc. Chimeric oligomeric compounds for modulation of splicing
US7700567B2 (en) 2005-09-29 2010-04-20 Supergen, Inc. Oligonucleotide analogues incorporating 5-aza-cytosine therein
US20070077993A1 (en) 2005-09-30 2007-04-05 Midgley Timothy M Method and apparatus for collecting user game play data and crediting users in a gaming environment
AU2006304205C1 (en) 2005-10-12 2012-11-15 Idera Pharmaceuticals, Inc. Immune regulatory oligonucleotide (IRO) compounds to modulate toll-like receptor based immune response
US9308252B2 (en) 2005-10-27 2016-04-12 Cook Biotech, Inc. Extracellular matrix materials as vaccine adjuvants for diseases associated with infectious pathogens or toxins
CA2627134A1 (en) 2005-10-28 2007-05-03 Tosoh Corporation Method of preparing carotenoid synthesizing microorganism and method of producing carotenoids
CA2627025A1 (en) 2005-10-28 2007-05-03 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of huntingtin gene
KR20080075107A (en) 2005-11-11 2008-08-14 화이자 인코포레이티드 Combinations and methods of using an immunomodulatory oligodeoxynucleotide
WO2007064291A1 (en) 2005-11-30 2007-06-07 Jyoti Chattopadhyaya Method and compounds for rna synthesis
EP1957507B1 (en) 2005-12-02 2018-10-24 Ionis Pharmaceuticals, Inc. Antibacterial 4,5-substituted aminoglycoside analogs having multiple substituents
US8076303B2 (en) 2005-12-13 2011-12-13 Spring Bank Pharmaceuticals, Inc. Nucleotide and oligonucleotide prodrugs
CA2634381C (en) 2005-12-21 2011-03-08 Pfizer Products Inc. Carbonylamino pyrrolopyrazoles, potent kinase inhibitors
SI2161038T1 (en) 2006-01-26 2014-05-30 Isis Pharmaceuticals, Inc. Compositions and their uses directed to Huntingtin
DE602007009487D1 (en) 2006-01-27 2010-11-11 Isis Pharmaceutical Inc 6-MODIFIED BICYCLIC NUCLEIC ACID ANALOGUE
DK1991678T4 (en) 2006-02-15 2020-10-19 Rechtsanwalt Thomas Beck COMPOSITIONS AND PROCEDURES FOR OLIGONUCLEOTIDE FORMULATIONS
JP4713514B2 (en) 2006-02-20 2011-06-29 エフ.ホフマン−ラ ロシュ アーゲー Improved labeling reagent
US8383660B2 (en) 2006-03-10 2013-02-26 Pfizer Inc. Dibenzyl amine compounds and derivatives
US9040674B2 (en) 2006-03-31 2015-05-26 Applied Biosystems, Llc Reagents useful for synthesizing rhodamine-labeled oligonucleotides
WO2007118222A2 (en) 2006-04-06 2007-10-18 Ibis Biosciences, INC Compositions for the use in identification of fungi
KR101176697B1 (en) 2006-04-20 2012-08-23 에프. 호프만-라 로슈 아게 Diazepan derivatives modulators of chemokine receptors
ES2396453T3 (en) 2006-04-24 2013-02-21 Sigma Alimentos, S.A. De C.V. Method for multiple and simultaneous detection and quantification of pathogens by polymerase chain reaction in real time
EP2018436A2 (en) 2006-04-25 2009-01-28 Immune Disease Institute Inc. Targeted delivery to leukocytes using non-protein carriers
GB0608838D0 (en) 2006-05-04 2006-06-14 Novartis Ag Organic compounds
EP2015758B1 (en) 2006-05-05 2014-04-02 Isis Pharmaceuticals, Inc. Compounds and methods for modulating expression apob
WO2007131232A2 (en) 2006-05-05 2007-11-15 Isis Pharmaceuticals, Inc. Compositions and their uses directed to ptpr alpha
US20090012120A1 (en) 2006-05-10 2009-01-08 Board Of Trustees Of Michigan State University Synthesis of N-heterocycles, beta-amino acids, and allyl amines via aza-payne mediated reaction of ylides and hydroxy aziridines
CN101490074B (en) 2006-05-11 2013-06-26 Isis制药公司 5'-modified bicyclic nucleic acid analogs
US7666854B2 (en) 2006-05-11 2010-02-23 Isis Pharmaceuticals, Inc. Bis-modified bicyclic nucleic acid analogs
WO2007139190A1 (en) 2006-05-31 2007-12-06 Toray Industries, Inc. Immunostimulatory oligonucleotide and pharmaceutical application thereof
US8097596B2 (en) 2006-06-30 2012-01-17 Lakewood-Amedex, Inc. Compositions and methods for the treatment of muscle wasting
EP2046993A4 (en) 2006-07-07 2010-11-17 Univ Massachusetts Rna silencing compositions and methods for the treatment of huntington's disease
CN101506368B (en) 2006-07-12 2017-02-08 加利福尼亚大学董事会 Transducible delivery of nucleic acids by reversible phosphotriester charge neutralization protecting groups
CA2659155A1 (en) 2006-07-20 2008-01-24 Amgen Inc. Substituted azole aromatic heterocycles as inhibitors of 11.beta.-hsd-1
GB0614947D0 (en) 2006-07-27 2006-09-06 Isis Innovation Epitope reduction therapy
AU2007281082A1 (en) 2006-08-04 2008-02-07 Isis Pharmaceuticals, Inc. Compositions and methods for the modulation of JNK proteins
AT504194B1 (en) 2006-09-07 2008-07-15 Oesterr Rotes Kreuz BACTERIA DETECTION
US8138330B2 (en) 2006-09-11 2012-03-20 Sigma-Aldrich Co. Llc Process for the synthesis of oligonucleotides
MX2009003398A (en) 2006-09-27 2009-08-12 Coley Pharm Gmbh Cpg oligonucleotide analogs containing hydrophobic t analogs with enhanced immunostimulatory activity.
ES2526295T5 (en) 2006-10-18 2021-05-04 Ionis Pharmaceuticals Inc Antisense compounds
CA2667816A1 (en) 2006-10-23 2008-05-02 Irm Llc Compositions and methods for inhibiting cathepsin proteases
EP2104738A2 (en) 2006-10-26 2009-09-30 Coley Pharmaceuticals GmbH Oligoribonucleotides and uses thereof
FR2908414B1 (en) 2006-11-13 2012-01-20 Centre Nat Rech Scient IMMOBILIZATION OF MEMBRANE PROTEINS ON A SUPPORT THROUGH AN AMPHIPHILE MOLECULE
US8754107B2 (en) 2006-11-17 2014-06-17 Abbvie Inc. Aminopyrrolidines as chemokine receptor antagonists
EP2102340A2 (en) 2006-11-27 2009-09-23 Isis Pharmaceuticals, Inc. Methods for treating hypercholesterolemia
US8093222B2 (en) 2006-11-27 2012-01-10 Isis Pharmaceuticals, Inc. Methods for treating hypercholesterolemia
CA2671883A1 (en) 2006-12-12 2008-06-19 Idera Pharmaceuticals, Inc. Synthetic agonists of tlr9
UY30892A1 (en) 2007-02-07 2008-09-02 Smithkline Beckman Corp AKT ACTIVITY INHIBITORS
EP2125852B1 (en) 2007-02-15 2016-04-06 Ionis Pharmaceuticals, Inc. 5'-substituted-2'-f modified nucleosides and oligomeric compounds prepared therefrom
AU2008230886B9 (en) 2007-03-24 2014-09-04 Kastle Therapeutics, Llc Administering antisense oligonucleotides complementary to human apolipoprotein B
US7960353B2 (en) 2007-05-10 2011-06-14 University Of Kansas Novobiocin analogues as neuroprotective agents and in the treatment of autoimmune disorders
CA2723672C (en) 2007-05-11 2019-09-03 Adynxx, Inc. Gene expression and pain
WO2008143343A1 (en) * 2007-05-24 2008-11-27 Kyorin Pharmaceutical Co., Ltd. Mutilin derivative having heterocyclic aromatic ring carboxylic acid structure in substituent at 14-position
GB0710186D0 (en) 2007-05-29 2007-07-04 Texas Instr Denmark PWM loop with minimum allasing error property
CA2688321A1 (en) 2007-05-30 2008-12-11 Isis Pharmaceuticals, Inc. N-substituted-aminomethylene bridged bicyclic nucleic acid analogs
RU2471781C2 (en) 2007-06-05 2013-01-10 НСАБ, Филиаль аф НьюроСёрч Свиден АБ, Сверийе Novel disubstituted phenylpyrrolidines as modulators of cortical catecholaminergic neurotransmission
EP2173760B2 (en) 2007-06-08 2015-11-04 Isis Pharmaceuticals, Inc. Carbocyclic bicyclic nucleic acid analogs
EP2164865A2 (en) 2007-06-13 2010-03-24 Hochschule Mannheim Compounds for the modulation of huntingtin aggregation, methods and means for identifying such compounds
ATE462787T1 (en) 2007-06-18 2010-04-15 Commissariat Energie Atomique REVERSIBLE SIRNA-SILENCING OF A MUTATED AND ENDOGENOUS HUNTINGTON WILD-TYPE AND ITS APPLICATION TO THE TREATMENT OF HUNTINGTON'S DISEASE
GB0712494D0 (en) 2007-06-27 2007-08-08 Isis Innovation Substrate reduction therapy
CA2692579C (en) 2007-07-05 2016-05-03 Isis Pharmaceuticals, Inc. 6-disubstituted bicyclic nucleic acid analogs
CA2692161C (en) 2007-07-09 2015-09-29 Idera Pharmaceuticals, Inc. Stabilized immune modulatory rna (simra) compounds
TWI413530B (en) 2007-07-20 2013-11-01 Kao Corp Organopolysiloxane
CA2694718A1 (en) 2007-07-31 2009-02-05 University Of Saskatchewan Genetic variation in pro-melanin-concentrating hormone gene affects carcass traits in cattle
US7812003B2 (en) 2007-08-02 2010-10-12 Safe Stephen H Antisense microRNA and uses therefor
AU2008286735A1 (en) 2007-08-15 2009-02-19 Idera Pharmaceuticals, Inc. Toll like receptor modulators
MX2010003606A (en) 2007-10-01 2010-07-02 Isis Pharmaceuticals Inc Antisense modulation of fibroblast growth factor receptor 4 expression.
KR100886139B1 (en) 2007-11-13 2009-02-27 주식회사 삼천리제약 Method for preparing oligonucleotide
TW200930375A (en) 2007-12-21 2009-07-16 Exelixis Inc Benzofuropyrimidinones
TWI340765B (en) 2007-12-26 2011-04-21 Ind Tech Res Inst Oligonucleotide sequences and dna chip for identifying filamentous microorganisms and the identification method thereof
CN101911676A (en) 2008-01-15 2010-12-08 联发科技股份有限公司 Multimedia presenting system, multimedia processing apparatus thereof, and method for presenting video and audio signals
WO2009089659A1 (en) 2008-01-18 2009-07-23 Shanghai Targetdrug Co., Ltd. Pyrollidine-based compounds
CA2711587A1 (en) 2008-02-04 2009-08-13 Galapagos Nv Target sequences and methods to identify the same, useful in treatment of neurodegenerative diseases
JP2009190983A (en) 2008-02-12 2009-08-27 Tokyo Institute Of Technology Oligonucleotide derivative
US8426378B2 (en) 2008-03-21 2013-04-23 Isis Pharmaceuticals, Inc. Oligomeric compounds comprising tricyclic nucelosides and methods for their use
US20110130440A1 (en) 2008-03-26 2011-06-02 Alnylam Pharmaceuticals, Inc. Non-natural ribonucleotides, and methods of use thereof
GB2471806B (en) 2008-04-03 2012-12-19 Spring Bank Pharmaceuticals Inc Compositions and methods for treating viral infections
DE112009001327A5 (en) 2008-04-04 2011-03-31 Universität Hamburg Process for the stereoselective synthesis of phosphorus compounds
WO2009124238A1 (en) 2008-04-04 2009-10-08 Isis Pharmaceuticals, Inc. Oligomeric compounds comprising neutrally linked terminal bicyclic nucleosides
WO2009135322A1 (en) 2008-05-09 2009-11-12 The Universtity Of British Columbia Methods and compositions for the treatment of huntington's disease
US8679750B2 (en) 2008-05-09 2014-03-25 The University Of British Columbia Methods and compositions for the treatment of Huntington'S disease
EP2300102B1 (en) 2008-05-15 2014-01-08 Dynavax Technologies Corporation Immunostimulatory sequences containing CpG for use in the treatment of allergic rhinitis
US8541387B2 (en) 2008-05-22 2013-09-24 Isis Pharmaceuticals, Inc. Modulation of SMRT expression
EP2291200A4 (en) 2008-05-22 2012-05-30 Isis Pharmaceuticals Inc Methods for modulating expression of rbp4
WO2009143391A2 (en) 2008-05-22 2009-11-26 Isis Pharmaceuticals, Inc Methods for modulation expression of creb
WO2009148605A2 (en) 2008-06-04 2009-12-10 Isis Pharmaceuticals, Inc. Methods for treating hypercholesterolemia
EP2320895A2 (en) 2008-07-03 2011-05-18 Exelixis, Inc. Cdk modulators
WO2010030849A1 (en) 2008-09-12 2010-03-18 University Of Louisville Research Foundation, Inc. Compositions and methods for treating cancer,inhibiting proliferation, and inducing cell death
WO2010030858A1 (en) 2008-09-15 2010-03-18 Enanta Pharmaceuticals, Inc. 4'-allene-substituted nucleoside derivatives
EP3067359A1 (en) 2008-09-23 2016-09-14 Scott G. Petersen Self delivering bio-labile phosphate protected pro-oligos for oligonucleotide based therapeutics and mediating rna interference
EP2356129B1 (en) 2008-09-24 2013-04-03 Isis Pharmaceuticals, Inc. Substituted alpha-l-bicyclic nucleosides
WO2010036696A1 (en) 2008-09-24 2010-04-01 Isis Pharmaceuticals, Inc. Cyclohexenyl nucleic acid analogs
AU2009302385B2 (en) 2008-10-07 2015-12-03 President And Fellows Of Harvard College Telomerase inhibitors and methods of use thereof
CA2738957A1 (en) 2008-10-22 2010-04-29 Quark Pharmaceuticals, Inc. Methods for treating eye disorders
US8987435B2 (en) 2008-10-24 2015-03-24 Isis Pharmaceuticals, Inc. Oligomeric compounds and methods
CN102264374B (en) 2008-10-24 2015-01-07 Isis制药公司 5' and 2' bis-substituted nucleosides and oligomeric compounds prepared therefrom
US9394333B2 (en) 2008-12-02 2016-07-19 Wave Life Sciences Japan Method for the synthesis of phosphorus atom modified nucleic acids
WO2010072831A1 (en) 2008-12-23 2010-07-01 Girindus America, Inc Sulfurizing reagents and their use for oligonucleotides synthesis
WO2010080953A1 (en) 2009-01-08 2010-07-15 Isis Pharmaceuticals, Inc. Transgenic murine model of human lipoprotein metabolism, hypercholesterolemia and cardiovascular disease
KR20100087540A (en) 2009-01-28 2010-08-05 삼성전자주식회사 Ink composition for inkjet recording
WO2010091301A1 (en) 2009-02-06 2010-08-12 Isis Pharmaceuticals, Inc. Oligomeric compounds and excipients
US8202974B2 (en) 2009-02-10 2012-06-19 Idera Pharmaceuticals, Inc. Synthetic RNA-based agonists of TLR7
JP5766617B2 (en) 2009-02-20 2015-08-19 ユニバーシティ・オブ・カンザス Novobiocin analogs having modified sugar moieties
EP2403863B1 (en) 2009-03-02 2013-08-28 Alnylam Pharmaceuticals Inc. Nucleic acid chemical modifications
WO2010107838A1 (en) 2009-03-16 2010-09-23 Isis Pharmaceuticals, Inc. Targeting apolipoprotein b for the reduction of apolipoprotein c-iii
US8653254B2 (en) 2009-03-31 2014-02-18 Takeda Pharmaceutical Company Limited Process for producing nucleoside
WO2010118263A1 (en) 2009-04-08 2010-10-14 University Of Massachusetts Single-nucleotide polymorphism (snp) targeting therapies for the treatment of huntington's disease
WO2010120262A1 (en) 2009-04-14 2010-10-21 Smith Holdings, Llc Methods and compositions for the treatment of medical conditions involving cellular programming
WO2010129853A2 (en) 2009-05-07 2010-11-11 The Regents Of The University Of California TRANSDUCIBLE DELIVERY OF NUCLEIC ACIDS USING MODIFIED dsRNA BINDING DOMAINS
EP2438079A4 (en) 2009-06-01 2013-05-22 Univ California Nucleic acid delivery compositions and methods of use thereof
EP2437753B1 (en) 2009-06-05 2016-08-31 Infectious Disease Research Institute Synthetic glucopyranosyl lipid adjuvants and vaccine compositions containing them
LT3449926T (en) 2009-06-17 2020-01-27 Biogen Ma Inc. Compositions and methods for modulation of smn2 splicing in a subject
JP5670097B2 (en) 2009-06-19 2015-02-18 花王株式会社 Two-layer separated hair cosmetic
US20120108800A1 (en) 2009-06-23 2012-05-03 Shumpei Murata Method for synthesizing nucleic acid
WO2011005761A1 (en) 2009-07-06 2011-01-13 Ontorii, Inc Novel nucleic acid prodrugs and methods use thereof
WO2011005764A1 (en) 2009-07-06 2011-01-13 Ada Technologies, Inc. Electrochemical device and method for long-term measurement of hypohalites
WO2011005860A2 (en) 2009-07-07 2011-01-13 Alnylam Pharmaceuticals, Inc. 5' phosphate mimics
US20110009477A1 (en) 2009-07-08 2011-01-13 Idera Pharmaceuticals, Inc. Oligonucleotide-based compounds as inhibitors of toll-like receptors
EP2458005A1 (en) 2009-07-23 2012-05-30 Galaxy Pharma Inc. Fgf21 cis-element binding substance
WO2011015573A1 (en) 2009-08-03 2011-02-10 Galapagos Nv Molecular targets and compounds, and methods to identify the same, useful in the treatment of neurodegenerative diseases
WO2011015572A1 (en) 2009-08-03 2011-02-10 Galapagos Nv Molecular targets and compounds, and methods to identify the same, useful in the treatment of neurodegenerative diseases
UA107360C2 (en) 2009-08-05 2014-12-25 Biogen Idec Inc Bicyclic aryl sphingosine 1-phosphate analogs
EP2462153B1 (en) 2009-08-06 2015-07-29 Isis Pharmaceuticals, Inc. Bicyclic cyclohexose nucleic acid analogs
US8927553B2 (en) 2009-08-10 2015-01-06 Daljit Singh Dhanoa Deuterium-enriched alkyl sulfonamides and uses thereof
NZ599032A (en) 2009-09-11 2014-09-26 Isis Pharmaceuticals Inc Modulation of huntingtin expression
EP2479182B8 (en) 2009-09-16 2016-07-13 Wave Life Sciences Japan, Inc. Novel protecting group for synthesizing rna and derivative thereof
WO2011038288A1 (en) 2009-09-25 2011-03-31 Isis Pharmaceuticals, Inc. Modulation of ttc39 expression to increase hdl
WO2011045702A1 (en) 2009-10-15 2011-04-21 Pfizer Inc. Pyrrolo[2,3-d] pyrimidine compounds
TWI475051B (en) 2009-11-18 2015-03-01 Kao Corp Organic polysiloxane
JP5809408B2 (en) 2009-11-25 2015-11-10 花王株式会社 Hair cosmetics
WO2011075560A1 (en) 2009-12-17 2011-06-23 Merck Sharp & Dohme Corp. Aminopyrimidines as syk inhibitors
JP2013515953A (en) 2009-12-28 2013-05-09 アチラ ラボズ プライベート リミテッド Diagnostic gel composition and method for producing said diagnostic gel composition
AU2011203986C1 (en) 2010-01-08 2015-03-05 Ionis Pharmaceuticals, Inc. Modulation of angiopoietin-like 3 expression
US8750507B2 (en) 2010-01-25 2014-06-10 Cisco Technology, Inc. Dynamic group creation for managed key servers
WO2011097643A1 (en) 2010-02-08 2011-08-11 Isis Pharmaceuticals, Inc. Selective reduction of allelic variants
AU2011213563B2 (en) 2010-02-08 2015-12-24 Ionis Pharmaceuticals, Inc. Selective reduction of allelic variants
SG182471A1 (en) 2010-02-10 2012-08-30 Glaxosmithkline Llc 6-amino-2-{ [ (1s)-1-methylbutyl] oxy}-9-[5-(1-piperidinyl)-7,9-dihydro-8h-purin-8-one maleate
US8859755B2 (en) 2010-03-05 2014-10-14 Chiralgen, Ltd. Method for preparing ribonucleoside phosphorothioate
WO2011127175A1 (en) 2010-04-06 2011-10-13 Isis Pharmaceuticals, Inc. Modulation of cd130 (gp130) expression
JP2013527148A (en) 2010-04-07 2013-06-27 アイシス ファーマシューティカルズ, インコーポレーテッド Regulation of CETP expression
WO2011133871A2 (en) 2010-04-22 2011-10-27 Alnylam Pharmaceuticals, Inc. 5'-end derivatives
EP2625186B1 (en) 2010-04-28 2016-07-27 Ionis Pharmaceuticals, Inc. 5' modified nucleosides and oligomeric compounds prepared therefrom
WO2011139702A2 (en) 2010-04-28 2011-11-10 Isis Pharmaceuticals, Inc. Modified nucleosides and oligomeric compounds prepared therefrom
WO2011139911A2 (en) 2010-04-29 2011-11-10 Isis Pharmaceuticals, Inc. Lipid formulated single stranded rna
EP2563921B1 (en) 2010-04-30 2016-11-23 Cellectis Method for modulating double-strand break-induced homologous recombination
GB201008902D0 (en) 2010-05-27 2010-07-14 Imp Innovations Ltd Membrane enhanced polymer sythesis
WO2012030683A2 (en) 2010-08-31 2012-03-08 Merck Sharp & Dohme Corp. Novel single chemical entities and methods for delivery of oligonucleotides
WO2012039448A1 (en) 2010-09-24 2012-03-29 株式会社キラルジェン Asymmetric auxiliary group
US9238812B2 (en) 2010-09-30 2016-01-19 Lsip, Llc Agent for suppressing expression of dominant mutant gene
KR101381048B1 (en) 2010-10-20 2014-04-14 씨제이제일제당 (주) A microorganism producing O-phosphoserine and the method of producing L-cysteine or derivatives thereof from O-phosphoserine using the same
EP3327125B1 (en) 2010-10-29 2020-08-05 Sirna Therapeutics, Inc. Rna interference mediated inhibition of gene expression using short interfering nucleic acids (sina)
AU2011325956B2 (en) 2010-11-12 2016-07-14 The General Hospital Corporation Polycomb-associated non-coding RNAs
US8822671B2 (en) 2010-11-30 2014-09-02 The University Of Tokyo 2'-O-modified RNA
US20140050778A1 (en) 2010-12-28 2014-02-20 University Of Rochester Nucleic acid binding compounds, methods of making, and use thereof
WO2012109395A1 (en) 2011-02-08 2012-08-16 Isis Pharmaceuticals, Inc. Oligomeric compounds comprising bicyclic nucleotides and uses thereof
CA2827380C (en) 2011-02-12 2020-08-25 University Of Iowa Research Foundation Therapeutic nucleic acids
US9353371B2 (en) 2011-05-02 2016-05-31 Ionis Pharmaceuticals, Inc. Antisense compounds targeting genes associated with usher syndrome
FR2975600B1 (en) 2011-05-24 2013-07-05 Assist Publ Hopitaux De Paris AGENTS FOR THE TREATMENT OF TUMORS
AU2012283994A1 (en) 2011-07-19 2014-03-06 University Of Idaho Embodiments of a probe and method for targeting nucleic acids
DK2742136T3 (en) 2011-08-11 2017-11-20 Ionis Pharmaceuticals Inc GAPMER COMPOUNDS INCLUDING 5 'MODIFIED DEOXYRIBONUCLEOSIDES IN GAP AND APPLICATIONS THEREOF
WO2013033223A1 (en) 2011-08-29 2013-03-07 Isis Pharmaceuticals, Inc. Methods and compounds useful in conditions related to repeat expansion
US20140080896A1 (en) 2011-08-30 2014-03-20 The Regents Of The University Of California Identification of small molecules that facilitate therapeutic exon skipping
WO2013030588A1 (en) 2011-08-31 2013-03-07 The University Of Manchester Method for diagnosing a neurodegenerative disease
US20140255936A1 (en) 2011-09-09 2014-09-11 Mayo Foundation For Medical Education And Research Detecting frontotemporal dementia and amyotrophic lateral sclerosis
ES2832531T3 (en) 2011-11-30 2021-06-10 Sarepta Therapeutics Inc Oligonucleotides for the treatment of repeat expansion diseases
CA2895077A1 (en) 2011-12-12 2013-06-20 Beverly Packard In vivo delivery of oligonucleotides
AU2012353330B2 (en) 2011-12-16 2018-04-19 National University Corporation Tokyo Medical And Dental University Chimeric double-stranded nucleic acid
CN102675386B (en) 2011-12-24 2014-07-02 河南科技大学 Method for separating and purifying gentiamarin
WO2013134558A1 (en) 2012-03-07 2013-09-12 The Texas A & M University System Cancer treatment targeting non-coding rna overexpression
CA2866381C (en) 2012-03-13 2020-03-24 Gilead Sciences, Inc. 2'- substituted carba-nucleoside analogs for antiviral treatment
KR20130114435A (en) 2012-04-09 2013-10-17 삼성전자주식회사 Biomolecule detection apparatus including a plurality of electrode
DK2841578T3 (en) 2012-04-23 2017-07-31 Biomarin Tech Bv RNA modulating oligonucleotides with improved properties for the treatment of neuromuscular diseases
MX355708B (en) 2012-05-22 2018-04-27 Idenix Pharmaceuticals Llc D-amino acid compounds for liver disease.
SG11201407967TA (en) 2012-05-30 2015-01-29 Hokkaido System Science Co Ltd Oligonucleotide synthesis method using highly dispersible liquid-phase support
CN104684893B (en) 2012-07-13 2016-10-26 日本波涛生命科学公司 Asymmetric auxiliary group
RU2015104762A (en) 2012-07-13 2018-08-31 Уэйв Лайф Сайенсес Лтд. CHIRAL CONTROL
MX356830B (en) 2012-07-13 2018-06-15 Shin Nippon Biomedical Laboratories Ltd Chiral nucleic acid adjuvant.
US10086081B2 (en) 2012-08-06 2018-10-02 Alnylam Pharmaceuticals, Inc. Carbohydrate conjugated RNA agents and process for their preparation
WO2014028739A1 (en) 2012-08-15 2014-02-20 Isis Pharmaceuticals, Inc. Method of preparing oligomeric compounds using modified capping protocols
EP3459549B1 (en) 2012-10-12 2022-04-06 Ionis Pharmaceuticals, Inc. Selective antisense compounds and uses thereof
JP6570447B2 (en) 2012-10-15 2019-09-04 アイオーニス ファーマシューティカルズ, インコーポレーテッドIonis Pharmaceuticals,Inc. Composition for modulating C9ORF72 expression
EP2906697A4 (en) 2012-10-15 2016-06-22 Ionis Pharmaceuticals Inc Methods for monitoring c9orf72 expression
ES2762326T5 (en) 2012-10-15 2023-04-27 Ionis Pharmaceuticals Inc Methods to modulate the expression of C9ORF72
EP2725029A1 (en) 2012-10-29 2014-04-30 Laboratoire Biodim New antibacterial compounds and biological applications thereof
JP2015535261A (en) 2012-10-29 2015-12-10 コクリスタル ファーマ,インコーポレイテッド Pyrimidine nucleotides and their monophosphate prodrugs for the treatment of viral infections and cancer
WO2014069520A1 (en) 2012-10-31 2014-05-08 武田薬品工業株式会社 New modified nucleic acid
EP2920307B1 (en) 2012-11-15 2018-05-02 Roche Innovation Center Copenhagen A/S Anti apob antisense conjugate compounds
MX2015006289A (en) 2012-11-26 2015-12-03 Roche Innovation Ct Copenhagen As Compositions and methods for modulation of fgfr3 expression.
US9211300B2 (en) 2012-12-19 2015-12-15 Idenix Pharmaceuticals Llc 4′-fluoro nucleosides for the treatment of HCV
RU2649367C2 (en) 2013-01-30 2018-04-02 Ф. Хоффманн-Ля Рош Аг Lna oligonucleotide carbohydrate conjugates
WO2014118272A1 (en) 2013-01-30 2014-08-07 Santaris Pharma A/S Antimir-122 oligonucleotide carbohydrate conjugates
EP2951304B1 (en) 2013-02-04 2020-07-08 Ionis Pharmaceuticals, Inc. Selective antisense compounds and uses thereof
CA2900238A1 (en) 2013-02-22 2014-08-28 Sirna Therapeutics, Inc. Short interfering nucleic acid (sina) molecules containing a 2' internucleoside linkage
CA2901983A1 (en) 2013-03-01 2014-09-04 National University Corporation Tokyo Medical And Dental University Chimeric single-stranded antisense polynucleotides and double-stranded antisense agent
CA2907511A1 (en) 2013-03-28 2014-10-02 Syngenta Participations Ag Methods of controlling neonicotinoid resistant pests
CA2907750A1 (en) 2013-03-28 2014-10-02 Syngenta Participations Ag Methods of controlling neonicotinoid resistant pests
BR112015027377B1 (en) 2013-05-01 2023-01-10 Ionis Pharmaceuticals, Inc OLIGOMERIC COMPOUNDS WITH CONJUGATE GROUPS, COMPOSITIONS COMPRISING THE SAID COMPOUNDS AND USES THEREOF
EP3015467A4 (en) 2013-05-24 2016-11-02 Ajinomoto Kk Morpholino oligonucleotide manufacturing method
WO2014188001A1 (en) 2013-05-24 2014-11-27 Santaris Pharma A/S Oligonucleotide modulators of b-cell cll/lymphoma 11a (bcl11a) and uses thereof
ES2700277T3 (en) 2013-05-30 2019-02-14 Univ Nat Corp Tokyo Medical & Dental Double chain agents for the delivery of therapeutic oligonucleotides
WO2014203518A1 (en) 2013-06-16 2014-12-24 National University Corporation Tokyo Medical And Dental University Double-stranded antisense nucleic acid with exon-skipping effect
US20160122761A1 (en) 2013-06-21 2016-05-05 Isis Pharmaceuticals, Inc. Compositions and methods for modulation of target nucleic acids
PL3013959T3 (en) 2013-06-27 2020-05-18 Roche Innovation Center Copenhagen A/S Antisense oligomers and conjugates targeting pcsk9
TWI772856B (en) 2013-07-19 2022-08-01 美商百健Ma公司 Compositions for modulating tau expression
US10894963B2 (en) 2013-07-25 2021-01-19 Exicure, Inc. Spherical nucleic acid-based constructs as immunostimulatory agents for prophylactic and therapeutic use
US10435430B2 (en) 2013-07-31 2019-10-08 Ionis Pharmaceuticals, Inc. Methods and compounds useful in conditions related to repeat expansion
CN105579442A (en) 2013-09-06 2016-05-11 先正达参股股份有限公司 Insecticidal compounds
WO2015051169A2 (en) 2013-10-02 2015-04-09 Moderna Therapeutics, Inc. Polynucleotide molecules and uses thereof
EA201690675A1 (en) 2013-10-03 2016-08-31 Модерна Терапьютикс, Инк. POLYNUCLEOTES ENCODING THE RECEPTOR OF LOW DENSITY LIPOPROTEINS
WO2015051366A2 (en) 2013-10-04 2015-04-09 Novartis Ag Novel formats for organic compounds for use in rna interference
EP4166667A3 (en) 2013-10-11 2023-08-02 Ionis Pharmaceuticals, Inc. Compositions for modulating c9orf72 expression
US20160237432A1 (en) 2013-10-14 2016-08-18 Ionis Pharmaceuticals, Inc. Methods for modulating expression of c9orf72 antisense transcript
EP3058068B1 (en) 2013-10-14 2019-04-24 Ionis Pharmaceuticals, Inc. Compositions for modulating expression of c9orf72 antisense transcript
NZ719477A (en) 2013-11-11 2022-05-27 Sangamo Therapeutics Inc Methods and compositions for treating huntington’s disease
CN105722980A (en) 2013-11-14 2016-06-29 罗氏创新中心哥本哈根有限公司 APOB antisense conjugate compounds
US20150167017A1 (en) 2013-12-13 2015-06-18 Moderna Therapeutics, Inc. Alternative nucleic acid molecules and uses thereof
US10322173B2 (en) 2014-01-15 2019-06-18 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having anti-allergic activity, and anti-allergic agent
US10144933B2 (en) 2014-01-15 2018-12-04 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having immunity induction activity, and immunity induction activator
EP3095459A4 (en) 2014-01-15 2017-08-23 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having antitumor effect and antitumor agent
ES2917473T3 (en) 2014-01-16 2022-07-08 Wave Life Sciences Ltd chiral design
WO2015143078A1 (en) 2014-03-18 2015-09-24 University Of Massachusetts Raav-based compositions and methods for treating amyotrophic lateral sclerosis
EP3137476B1 (en) 2014-04-28 2019-10-09 Ionis Pharmaceuticals, Inc. Linkage modified oligomeric compounds
US9382540B2 (en) 2014-05-01 2016-07-05 Isis Pharmaceuticals, Inc Compositions and methods for modulating angiopoietin-like 3 expression
US11110154B2 (en) 2014-05-08 2021-09-07 Sangamo Therapeutics, Inc. Methods and compositions for treating Huntington's Disease
EP3146051B8 (en) 2014-05-20 2019-11-27 University of Iowa Research Foundation Huntington's disease therapeutic compounds
US20160017327A1 (en) 2014-07-11 2016-01-21 The Johns Hopkins University Phosphorodiamidate morpholino oligomers (pmos) and their use in suppression of mutant huntingtin expression and attenuation of neurotoxicity
JP2017524357A (en) 2014-07-16 2017-08-31 モデルナティエックス インコーポレイテッドModernaTX,Inc. Chimeric polynucleotide
EP2982758A1 (en) 2014-08-04 2016-02-10 Centre Hospitalier Universitaire Vaudois (CHUV) Genome editing for the treatment of huntington's disease
US10252974B2 (en) 2014-08-07 2019-04-09 Takeda Pharmaceutical Company Limited Cationic lipid
WO2016024205A1 (en) 2014-08-15 2016-02-18 Pfizer Inc. Oligomers targeting hexanucleotide repeat expansion in human c9orf72 gene
WO2016037191A1 (en) 2014-09-05 2016-03-10 Health Research, Inc. Use of huntingtin-derived plasmids and peptides for active immunization as a huntington's disease (hd) therapeutic
CN113846101A (en) 2014-11-17 2021-12-28 阿尔尼拉姆医药品有限公司 Apolipoprotein C3(APOC3) iRNA compositions and methods of use thereof
EP3220921A1 (en) 2014-11-19 2017-09-27 Roche Innovation Center Copenhagen A/S Lna chiral phosphorothioates
DK3234131T3 (en) 2014-12-16 2020-06-29 Roche Innovation Ct Copenhagen As Method of screening for chiral toxicity
BR112017013573A2 (en) 2014-12-24 2018-03-06 Uniqure Ip Bv double stranded rna, dna sequence, expression cassette, gene therapy vector, and host cell.
US9688707B2 (en) 2014-12-30 2017-06-27 Ionis Pharmaceuticals, Inc. Bicyclic morpholino compounds and oligomeric compounds prepared therefrom
WO2016112132A1 (en) 2015-01-06 2016-07-14 Ionis Pharmaceuticals, Inc. Compositions for modulating expression of c9orf72 antisense transcript
WO2016127002A1 (en) 2015-02-04 2016-08-11 Bristol-Myers Squibb Company Lna oligonucleotides with alternating flanks
JP6772156B2 (en) 2015-02-04 2020-10-21 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company How to choose a therapeutic molecule
EP3253875B1 (en) 2015-02-04 2020-01-08 H. Hoffnabb-La Roche Ag Tau antisense oligomers and uses thereof
SG10201907396XA (en) 2015-02-10 2019-09-27 Genzyme Corp VARIANT RNAi
CA2976445A1 (en) 2015-02-13 2016-08-18 Alnylam Pharmaceuticals, Inc. Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
EP3262174A4 (en) 2015-02-23 2018-10-17 Ionis Pharmaceuticals, Inc. Compounds and methods for increasing antisense activity
WO2016141236A1 (en) 2015-03-03 2016-09-09 Ionis Pharmaceuticals, Inc. Compositions for modulating mecp2 expression
WO2016145142A1 (en) 2015-03-10 2016-09-15 Emory University Nucleotide and nucleoside therapeutics compositions and uses related thereto
WO2016154096A1 (en) 2015-03-20 2016-09-29 Ionis Pharmaceuticals, Inc. Modulation of smggds expression
RS60762B1 (en) 2015-04-03 2020-10-30 Univ Massachusetts Oligonucleotide compounds for targeting huntingtin mrna
WO2016164896A2 (en) 2015-04-10 2016-10-13 Ionis Pharmaceuticals, Inc. Modulation of smn expression
US10407678B2 (en) 2015-04-16 2019-09-10 Ionis Pharmaceuticals, Inc. Compositions for modulating expression of C9ORF72 antisense transcript
KR102104163B1 (en) 2015-04-16 2020-04-23 아이오니스 파마수티컬즈, 인코포레이티드 Composition for modulating C9ORF72 expression
WO2016209862A1 (en) 2015-06-23 2016-12-29 Alnylam Pharmaceuticals, Inc. Glucokinase (gck) irna compositions and methods of use thereof
EP3313989A4 (en) 2015-06-29 2018-12-05 Ionis Pharmaceuticals, Inc. Modified crispr rna and modified single crispr rna and uses thereof
IL256622B2 (en) 2015-07-10 2023-09-01 Ionis Pharmaceuticals Inc Modulators of diacyglycerol acyltransferase 2 (dgat2)
WO2017011286A1 (en) 2015-07-10 2017-01-19 Alnylam Pharmaceuticals, Inc. Insulin-like growth factor binding protein, acid labile subunit (igfals) and insulin-like growth factor 1 (igf-1) irna compositions and methods of use thereof
CA2989970A1 (en) 2015-07-17 2017-01-26 Alnylam Pharmaceuticals, Inc. Multi-targeted single entity conjugates
MA43072A (en) 2015-07-22 2018-05-30 Wave Life Sciences Ltd COMPOSITIONS OF OLIGONUCLEOTIDES AND RELATED PROCESSES
US20180216114A1 (en) 2015-07-27 2018-08-02 Alnylam Pharmaceuticals, Inc. Xanthine dehydrogenase (xdh) irna compositions and methods of use thereof
SG10201912341SA (en) 2015-07-31 2020-02-27 Alnylam Pharmaceuticals Inc TRANSTHYRETIN (TTR) iRNA COMPOSITIONS AND METHODS OF USE THEREOF FOR TREATING OR PREVENTING TTR-ASSOCIATED DISEASES
DK3341479T3 (en) 2015-08-24 2020-02-24 Roche Innovation Ct Copenhagen As LNA-G Process
KR20180054640A (en) 2015-08-25 2018-05-24 알닐람 파마슈티칼스 인코포레이티드 METHODS AND COMPOSITIONS FOR THE TREATMENT OF PROGNOSTIC PROTEIN CONDUCTORS Subtilillocexcis (PCSK9) gene-related disorders
US10889813B2 (en) 2015-09-02 2021-01-12 Alnylam Pharmaceuticals, Inc. Programmed cell death 1 ligand 1 (PD-L1) iRNA compositions and methods of use thereof
EP3350328A1 (en) 2015-09-14 2018-07-25 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting patatin-like phospholipase domain containing 3 (pnpla3) and methods of use thereof
WO2017059411A1 (en) 2015-10-01 2017-04-06 Memorial Sloan-Kettering Cancer Center Inhibitors of menaquinone biosynthesis
US10874686B2 (en) 2015-10-01 2020-12-29 Memorial Sloan-Kettering Cancer Center Anthranilyl-adenosinemonosulfamate analogs and uses thereof
KR20180073584A (en) 2015-10-02 2018-07-02 로슈 이노베이션 센터 코펜하겐 에이/에스 Oligonucleotide joining method
WO2017062862A2 (en) 2015-10-09 2017-04-13 Wave Life Sciences Ltd. Oligonucleotide compositions and methods thereof
EP3183347A4 (en) 2015-10-17 2018-04-18 Lifesplice Pharma LLC Splice modulating oligonucleotides and methods of use thereof
WO2017068087A1 (en) 2015-10-22 2017-04-27 Roche Innovation Center Copenhagen A/S Oligonucleotide detection method
WO2017067970A1 (en) 2015-10-22 2017-04-27 Roche Innovation Center Copenhagen A/S In vitro toxicity screening assay
US11260073B2 (en) 2015-11-02 2022-03-01 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating C90RF72
EP3374498A1 (en) 2015-11-12 2018-09-19 H. Hoffnabb-La Roche Ag Standardized neuronal cell assays from primate species
MA43822A (en) 2016-03-13 2018-11-28 Wave Life Sciences Ltd COMPOSITIONS AND SYNTHESIS OF PHOSPHORAMIDITE AND OLIGONUCLEOTIDES
PT3430141T (en) 2016-03-14 2021-02-25 Hoffmann La Roche Oligonucleotides for reduction of pd-l1 expression
CN108779139A (en) 2016-03-18 2018-11-09 罗氏创新中心哥本哈根有限公司 The L-LNA- guanosine monomers of acyl group protection
WO2017165489A1 (en) 2016-03-23 2017-09-28 Emory University Antiviral agents for treating zika and dengue virus infections
WO2017180835A1 (en) 2016-04-13 2017-10-19 Ionis Pharmaceuticals, Inc. Methods for reducing c9orf72 expression
KR102468177B1 (en) 2016-04-14 2022-11-16 에프. 호프만-라 로슈 아게 Trityl-mono-GalNAc Compounds and Uses Thereof
MA45290A (en) 2016-05-04 2019-03-13 Wave Life Sciences Ltd PROCESSES AND COMPOSITIONS OF BIOLOGICALLY ACTIVE AGENTS
MA45270A (en) 2016-05-04 2017-11-09 Wave Life Sciences Ltd COMPOSITIONS OF OLIGONUCLEOTIDES AND RELATED PROCESSES
CN109311925B (en) 2016-05-12 2022-06-03 罗氏创新中心哥本哈根有限公司 Enhanced coupling of sterically defined oxaazaphospholane phosphoramidite monomers to nucleosides or oligonucleotides
CN109072234B (en) 2016-05-13 2022-11-29 豪夫迈·罗氏有限公司 Protein-based sample collection matrices and devices
WO2017198775A1 (en) 2016-05-18 2017-11-23 Eth Zurich Stereoselective synthesis of phosphorothioate oligoribonucleotides
WO2017210647A1 (en) 2016-06-03 2017-12-07 Wave Life Sciences Ltd. Oligonucleotides, compositions and methods thereof
CN109310765A (en) 2016-06-20 2019-02-05 领先基因生物技术股份有限公司 Antibody-drug conjugates
US20190264267A1 (en) 2016-07-25 2019-08-29 Wave Life Sciences Ltd. Phasing
MA46905A (en) 2016-11-23 2019-10-02 Wave Life Sciences Ltd COMPOSITIONS AND SYNTHESIS OF PHOSPHORAMIDITES AND OLIGONUCLEOTIDES

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9605019B2 (en) * 2011-07-19 2017-03-28 Wave Life Sciences Ltd. Methods for the synthesis of functionalized nucleic acids

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10329318B2 (en) 2008-12-02 2019-06-25 Wave Life Sciences Ltd. Method for the synthesis of phosphorus atom modified nucleic acids
US10307434B2 (en) 2009-07-06 2019-06-04 Wave Life Sciences Ltd. Nucleic acid prodrugs and methods of use thereof
US10428019B2 (en) 2010-09-24 2019-10-01 Wave Life Sciences Ltd. Chiral auxiliaries
US10280192B2 (en) 2011-07-19 2019-05-07 Wave Life Sciences Ltd. Methods for the synthesis of functionalized nucleic acids
US10167309B2 (en) 2012-07-13 2019-01-01 Wave Life Sciences Ltd. Asymmetric auxiliary group
US9982257B2 (en) 2012-07-13 2018-05-29 Wave Life Sciences Ltd. Chiral control
US10590413B2 (en) 2012-07-13 2020-03-17 Wave Life Sciences Ltd. Chiral control
US10149905B2 (en) 2014-01-15 2018-12-11 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having antitumor effect and antitumor agent
US10144933B2 (en) 2014-01-15 2018-12-04 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having immunity induction activity, and immunity induction activator
US10160969B2 (en) 2014-01-16 2018-12-25 Wave Life Sciences Ltd. Chiral design
US10479995B2 (en) 2015-07-22 2019-11-19 Wave Life Sciences Ltd. Oligonucleotide compositions and methods thereof
US10450568B2 (en) 2015-10-09 2019-10-22 Wave Life Sciences Ltd. Oligonucleotide compositions and methods thereof
US11407775B2 (en) 2016-03-13 2022-08-09 Wave Life Sciences Ltd. Compositions and methods for phosphoramidite and oligonucleotide synthesis
US10724035B2 (en) 2016-05-04 2020-07-28 Wave Life Sciences Ltd. Oligonucleotide compositions and methods thereof
US11013757B2 (en) 2016-06-03 2021-05-25 Wave Life Sciences Ltd. Oligonucleotides, compositions and methods thereof
US11873316B2 (en) 2016-11-23 2024-01-16 Wave Life Sciences Ltd. Compositions and methods for phosphoramidite and oligonucleotide synthesis

Also Published As

Publication number Publication date
CN103796657B (en) 2017-07-11
US10280192B2 (en) 2019-05-07
EP2734208A4 (en) 2015-03-18
JP2017141272A (en) 2017-08-17
CN107365339A (en) 2017-11-21
EP3248982A1 (en) 2017-11-29
MX2014000716A (en) 2014-05-22
EP2734208B1 (en) 2017-03-01
AU2012284265A1 (en) 2014-02-06
KR20140068884A (en) 2014-06-09
CA2842358C (en) 2020-07-14
RU2014105311A (en) 2015-08-27
ES2626488T3 (en) 2017-07-25
CA2842358A1 (en) 2013-01-24
WO2013012758A1 (en) 2013-01-24
EP2734208A1 (en) 2014-05-28
IL230522A0 (en) 2014-03-31
IL230522B (en) 2018-02-28
JP6128529B2 (en) 2017-05-17
BR112014001244A2 (en) 2017-02-21
CN103796657A (en) 2014-05-14
US20140194610A1 (en) 2014-07-10
US20180222936A1 (en) 2018-08-09
SG10201700554VA (en) 2017-03-30
JP6490133B2 (en) 2019-03-27
ES2626488T8 (en) 2017-08-14
JP2014522862A (en) 2014-09-08
DK2734208T3 (en) 2017-06-19
HK1198479A1 (en) 2015-05-08
MX347361B (en) 2017-04-12
US9605019B2 (en) 2017-03-28
AU2012284265B2 (en) 2017-08-17

Similar Documents

Publication Publication Date Title
US10280192B2 (en) Methods for the synthesis of functionalized nucleic acids
US9695211B2 (en) Method for the synthesis of phosphorus atom modified nucleic acids
KR101885383B1 (en) Novel nucleic acid prodrugs and methods of use thereof
JP4865544B2 (en) Method for producing highly stereoregular ribonucleotide analogs and deoxyribonucleotide analogs
CN103889999A (en) Ionic tags for synthesis of oligoribonucleotides
AU2015255202B2 (en) Novel nucleic acid prodrugs and methods of use thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: WAVE LIFE SCIENCES LTD., SINGAPORE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:VERDINE, GREGORY L.;., MEENA;IWAMOTO, NAOKI;AND OTHERS;SIGNING DATES FROM 20161213 TO 20161214;REEL/FRAME:041373/0649

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION