WO1995018136A1 - Backbone modified oligonucleotide analogs and solid phase synthesis thereof - Google Patents

Backbone modified oligonucleotide analogs and solid phase synthesis thereof Download PDF

Info

Publication number
WO1995018136A1
WO1995018136A1 PCT/US1994/014883 US9414883W WO9518136A1 WO 1995018136 A1 WO1995018136 A1 WO 1995018136A1 US 9414883 W US9414883 W US 9414883W WO 9518136 A1 WO9518136 A1 WO 9518136A1
Authority
WO
WIPO (PCT)
Prior art keywords
substituted
group
alkyl
oligonucleotide
carbon atoms
Prior art date
Application number
PCT/US1994/014883
Other languages
French (fr)
Inventor
Phillip Dan Cock
Yogesh S. Sanghvi
François MORVAN
Original Assignee
Isis Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Isis Pharmaceuticals, Inc. filed Critical Isis Pharmaceuticals, Inc.
Priority to EP95906115A priority Critical patent/EP0737201A4/en
Publication of WO1995018136A1 publication Critical patent/WO1995018136A1/en

Links

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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/04Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F7/00Compounds containing elements of Groups 4 or 14 of the Periodic System
    • C07F7/02Silicon compounds
    • C07F7/08Compounds having one or more C—Si linkages
    • C07F7/18Compounds having one or more C—Si linkages as well as one or more C—O—Si linkages
    • C07F7/1804Compounds having Si-O-C linkages
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/06Pyrimidine radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/06Pyrimidine radicals
    • C07H19/10Pyrimidine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/16Purine radicals

Definitions

  • This invention relates to the synthesis of nuclease resistant oligonucleotide analogs which are useful for therapeutics, diagnostics and as research reagents.
  • the invention relates to solid-phase synthetic methods wherein amine-terminated synthons are coupled with aldehyde-terminated synthons to produce hydroxylamino- containing and/or hydrazino-containing covalent linkages.
  • Classical therapeutics generally has focused upon interactions with proteins in an effort to moderate their disease causing or disease potentiating functions. Recently, however, attempts have been made to moderate the production of proteins by interactions with the molecules (i.e., intracellular RNA) that direct their synthesis. These interactions have involved hybridization of complementary "antisense" oligonucleotides or certain analogs thereof to RNA. Hybridization is the sequence-specific hydrogen bonding of oligonucleotides or oligonucleotide analogs to RNA or to single stranded DNA. By interfering with the production of proteins, it has been hoped to effect therapeutic results with maximum effect and minimal side effects.
  • oligonucleotides and oligonucleotide analogs like other therapeutics, depends on a number of factors that influence the effective concentration of these agents at specific intracellular targets.
  • One important factor for oligonucleotides is the stability of the species in the presence of nucleases. It is unlikely that unmodified oligonucleotides will be useful therapeutic agents because they are rapidly degraded by nucleases. Modification of oligonucleotides to render them resistant to nucleases therefore is greatly desired.
  • oligonucleotides Modification of oligonucleotides to enhance nuclease resistance generally has taken place on the phosphorus atom of the sugar-phosphate backbone. Phosphorothioates, methyl phosphonates, phosphoramidates and phosphorotriesters have been reported to confer various levels of nuclease resistance. Phosphate-modified oligonucleotides, however, generally have suffered from inferior hybridization properties. See, e . g. , Cohen, J.S., ed. Oligonucleotides : Antisense Inhibi tors of Gene Expression, (CRC Press, Inc., Boca Raton FL, 1989) .
  • Another key factor is the ability of antisense compounds to traverse the plasma membrane of specific cells involved in the disease process.
  • Cellular membranes consist of lipid-protein bilayers that are freely permeable to small, nonionic, lipophilic compounds and are inherently impermeable to most natural metabolites and therapeutic agents. See, e . g. , Wilson, Ann . Rev. Biochem . 1978, 47, 933.
  • the biological and antiviral effects of natural and modified oligonucleotides in cultured mammalian cells have been well documented. It appears that these agents can penetrate membranes to reach their intracellular targets.
  • modified oligonucleotides and oligonucleotide analogs are internalized less readily than their natural counterparts.
  • the activity of many previously available antisense oligonucleotides has not been sufficient for practical therapeutic, research or diagnostic purposes.
  • Two other serious deficiencies of prior art compounds designed for antisense therapeutics are inferior hybridization to intracellular RNA and the lack of a defined chemical or enzyme-mediated event to terminate essential RNA functions.
  • Modifications to enhance the effectiveness of the antisense oligonucleotides and overcome these problems have taken many forms. These modifications include base ring modifications, sugar moiety modifications and sugar-phosphate backbone modifications. Prior sugar-phosphate backbone modifications, particularly on the phosphorus atom, have effected various levels of resistance to nucleases. However, while the ability of an antisense oligonucleotide to bind to specific DNA or RNA with fidelity is fundamental to antisense methodology, modified phosphorus oligonucleotides have generally suffered from inferior hybridization properties.
  • the present invention provides novel compounds that mimic and/or modulate the activity of wild-type nucleic acids.
  • the compounds contain a selected nucleoside sequence which is specifically hybridizable with a targeted nucleoside sequence of single stranded or double stranded DNA or RNA.
  • At least a portion of the compounds of the invention has structure I :
  • L 1 -L 2 -L 3 -L 4 is CHa-R A -NRi-CHa, CHa-NR ⁇ A -CHa, R A -NR ⁇
  • each R x and R 2 are, independently, the same or different and are H; alkyl or substituted alkyl having 1 to about 10 carbon atoms; alkenyl or substituted alkenyl having 2 to about 10 carbon atoms; alkynyl or substituted alkynyl having 2 to about 10 carbon atoms; alkaryl, substituted alkaryl, aralkyl, or substituted aralkyl having 7 to about 14 carbon atoms; alicyclic; heterocyclic; a reporter molecule; an RNA cleaving group; a group for improving the pharmacokinetic properties of an oligonucleotide; or a group for improving the pharmacodynamic properties of an • ⁇ oligonucleotide;
  • B x is a nucleosidic base; n is an integer greater than 0; Q is 0, S, CH 2 , CHF or CF 2 ; X is H; OH; alkyl or substituted alkyl having 1 to about 10 carbon atoms; alkaryl, substituted alkaryl, aralkyl, or substituted aralkyl having 7 to about 14 carbon atoms; F; Cl; Br; CN; CF 3 ; 0CF 3 ; OCN; 0-alkyl; S-alkyl; N-alkyl; O- alkenyl; S-alkenyl; N-alkenyl; S0CH 3 ; S0 2 CH 3 ; 0N0 2 ; N0 2 ; N 3 ; NH 2 ; heterocycloalkyl; heterocycloalkaryl; aminoalkylamino; polyalkylamino or substituted silyl; an RNA cleaving group; a group for improving the pharmacokinetic properties of an
  • the compounds of the invention are prepared by coupling preselected 3' -functionalized and 4' -functionalized nucleosides and/or oligonucleotides under conditions effective to form the above-noted L 1 -L 2 -L 3 -L 4 linkages.
  • a 3'-C-formyl nucleoside or oligonucleotide synthon is reacted with a support-bound 5'- hydroxylamino or 5'-hydrazino nucleoside or oligonucleotide synthon.
  • a 4'-C-formyl synthon is reacted with a support-bound 3' -methylhydroxylamino or 3' - methylhydrazino synthon.
  • Z- L and Y 2 are selected such that
  • Figures 1 and 2 show exemplary compounds used in accordance with the invention.
  • Figure 3 is a synthetic scheme depicting a preferred method for preparation of oxime-linked oligonucleotide analogs.
  • nucleoside refers to a unit made up of a heterocyclic base and its sugar.
  • nucleotide refers to a nucleoside having a phosphate group on its 3' or 5 ' sugar hydroxyl group. Thus nucleosides, unlike nucleotides, have no phosphate group.
  • Oligonucleotide refers to a plurality of joined nucleotide units formed in a specific sequence from naturally occurring bases and pentofuranosyl groups joined through a sugar group by native phosphodiester bonds. This term refers to both naturally occurring and synthetic species formed from naturally occurring subunits.
  • oligonucleotide analogs that is, compounds which function like oligonucleotides but which have non-naturally occurring portions. Oligonucleotide analogs can have altered sugar moieties, altered base moieties or altered inter-sugar linkages. For the purposes of this invention, an oligonucleotide analog having non-phosphodiester bonds, i.e., an altered inter-sugar linkage, is considered to be an "oligonucleoside. " The term "oligonucleoside” thus refers to a plurality of nucleoside units joined by linking groups other than native phosphodiester linking groups.
  • oligomers is intended to encompass oligonucleotides, oligonucleotide analogs or oligonucleosides.
  • reference is made to a series of nucleosides or nucleoside analogs that are joined via either natural phosphodiester bonds or other linkages, including the four atom linkers of this invention.
  • linkage generally is from the 3' carbon of one nucleoside to the 5' carbon of a second nucleoside
  • the term “oligomer” can also include other linkages such as 2' -5' linkages.
  • Oligonucleotide analogs also can include other modifications consistent with the spirit of this invention, particularly modifications that increase nuclease resistance.
  • modifications that increase nuclease resistance For example, when the sugar portion of a nucleoside or nucleotide is replaced by a carbocyclic moiety, it is no longer a sugar.
  • other substitutions such a substitution for the inter-sugar phosphorodiester linkage are made, the resulting material is no longer a true nucleic acid species. All such compounds are considered to be analogs.
  • reference to the sugar portion of a nucleic acid species shall be understood to refer to either a true sugar or to a species taking the structural place of the sugar of wild type nucleic acids.
  • reference to inter-sugar linkages shall be taken to include moieties serving to join the sugar or sugar analog portions in the fashion of wild type nucleic acids.
  • This invention concerns modified oligonucleotides, i.e., oligonucleotide analogs or oligonucleosides, and methods for effecting the modifications.
  • modified oligonucleotides and oligonucleotide analogs exhibit increased chemical and/or enzymatic stability relative to their naturally occurring counterparts.
  • Extracellular and intracellular nucleases generally do not recognize and therefore do not bind to the backbone-modified compounds of the invention.
  • the neutral or positively charged backbones of the present invention can be taken into cells by simple passive transport rather than by complicated protein-mediated processes.
  • Another advantage of the invention is that the lack of a negatively charged backbone facilitates sequence specific binding of the oligonucleotide analogs or oligonucleosides to targeted RNA, which has a negatively charged backbone and will repel similarly charged oligonucleotides. Still another advantage of the present invention is it presents sites for attaching functional groups that initiate cleavage of targeted RNA.
  • the modified internucleoside linkages of this invention are intended to replace naturally-occurring phosphodiester-5' -methylene linkages with four atom linking groups to confer nuclease resistance and enhanced cellular uptake to the resulting compound.
  • Preferred linkages have structure CHa-R A -NRi-CHa, CE 2 -NR 1 -R h -CU 2 , R A -NR ⁇ C ⁇ -C ⁇ , CH 2 -CH 2 - NR ⁇ R A , CHz-CHa-R A -NRi, or NR ⁇ A -CHa-CHa where R A is 0 or NR 2 .
  • linkages are prepared by functionalizing the sugar moieties of two nucleosides which ultimately are to be adjacent to one another in the selected sequence.
  • an "upstream” synthon such as structure III is modified at its terminal 3' site
  • a “downstream” synthon such as structure II is modified at its terminal 4' site.
  • the invention provides efficient syntheses of oligonucleosides via intermolecular reductive coupling on a solid support.
  • B x can be nucleosidic bases selected from adenine, guanine, uracil, thymine, cytosine, 2-aminoadenosine or 5- methylcytosine, although other non-naturally occurring species can be employed to provide stable duplex or triplex formation with, for example, DNA.
  • Representative bases are disclosed in U.S. Patent No. 3,687,808 (Merigan, et al . ) , which is incorporated herein by reference.
  • Q can be S, CH 2 , CHF CF 2 or, preferably, 0. See, e . g. , Bellon, et al . , Nucleic Acids Res . 1993, 21 , 1587.
  • X can be H, OH, alkyl, substituted alkyl, alkaryl or aralkyl, F, Cl, Br, CN, CF 3 , 0CF 3 , OCN, O-alkyl, S-alkyl, N-alkyl, O-alkenyl, S-alkenyl, N-alkenyl, SOCH 3 , S0 2 CH 3 , ON0 2 , N0 2 , N 3 , NH 2 , heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, polyalkylamino or substituted silyl, an RNA cleaving group, a group for improving the pharmacokinetic properties of an oligonucleotide, or a group for improving the pharmacodynamic properties of an oligonucleotide.
  • alkyl denote branched and straight chain hydrocarbyl residues, including alkyl groups having one or more 3 H and/or 1 C atoms. It is preferred that X is H or OH, or, alternatively F, O-alkyl or O-alkenyl, especially where Q is 0. Preferred alkyl and alkenyl groups have from 1 to about 10 carbon atoms.
  • Solid supports e . g. , (P) ) according to the invention include any of those known in the art for polynucleotide synthesis, including controlled pore glass
  • CPG oxalyl-controlled pore glass
  • TentaGel Support an aminopolyethyleneglycol derivatized support (see, e . g. , Wright, et al . , Tetrahedron Letters 1993, 34 , 3373) or Poros -- a copolymer of polystyrene/divinylbenzene. Attachment and cleavage of nucleosides and oligonucleosides can be effected via standard procedures. See, e . g. , Pon, R.T.
  • solid support further includes any linkers (e . g. , long chain alkyl amines and succinyl residues) used to bind a growing oligonucleoside to a stationary phase such as CPG.
  • linkers e . g. , long chain alkyl amines and succinyl residues
  • Y can be OH, OR HP , CH 2 0H, or CH 2 0R HP where R HP is a hydroxyl protecting group.
  • R HP is a hydroxyl protecting group.
  • a wide variety of hydroxyl protecting groups can be employed in the methods of the invention. See, e . g. , Beaucage, et al . , Tetrahedron 1992, 12 , 2223. In general, protecting groups render chemical functionality inert to specific reaction conditions, and can be appended to and removed from such functionality in a molecule without substantially damaging the remainder of the molecule.
  • hydroxyl protecting groups include phthalimido, t-butyldimethylsilyl (TBDMS) , t- butyldiphenylsilyl (TBDPS) , fluorenylmethoxycarbonyl (FMOC) , and other hydroxyl protecting groups.
  • the invention is also directed to methods for the preparation of oligonucleosides with modified inter-sugar linkages. These modifications may be effected using solid supports which may be manually manipulated or used in conjunction with a DNA synthesizer using methodology commonly known to those skilled in DNA synthesizer arts . Generally, the procedure involves functionalizing the sugar moieties of two nucleosides which will be adjacent to one another in the selected sequence. In a 5' to 3' sense, an "upstream” synthon such as structure II is modified at its terminal 3' site, while a “downstream” synthon such as structure III is modified at its terminal 5' site.
  • linkages can be formed by selecting a 3'-C-formyl derivatized compound as the upstream synthon and a 5' -aminohydroxy derivatized compound as the downstream synthon. Coupling then is effected to provide, for example, a dinucleoside having an oxime linkage.
  • the oxime is present as E/Z isomers.
  • the oxime nitrogen atom is adjacent to a carbon atom on the 3' end of the upstream nucleoside.
  • Dinucleosides having the oxime nitrogen adjacent to a carbon atom on the 5' or downstream nucleoside are synthesized utilizing a 4'-C-formyl derivatized compound as the upstream synthon and a 3'-deoxy- 3' -aminohydroxymethyl derivatized compound as the downstream synthon, again providing E/Z isomers.
  • the oxime linked compound can be incorporated directly into an oligomer and/or can be reduced to a corresponding hydroxylamino linked species.
  • Reduction of oxime linked dinucleosides either as the dinucleoside or as a dinucleoside moiety in an oligomer with sodium cyanoborohydride yields the corresponding hydroxylamino linked compounds.
  • Hydroxylamino linked compounds can be alkylated at the amino moiety of the hydroxylamino linkage to yield a corresponding N-alkylamino linkage.
  • 3'-C-f ⁇ rmyl derivatized nucleosides can be formed via several synthetic pathways.
  • the presently preferred method utilizes a stereoselective intermolecular radical C-C bond formation reaction, ( see, e . g. , U.S. Application Serial No. 08/040,903, filed March 31, 1993) .
  • a 3'-0- phenylthiocarbonate ester derivative of thymidine reacted with j ⁇ -tri-n-butylstannylstyrene and AIBN to provide a 3'-C- styryl derivative, which on oxidative cleavage gave a 3'-C- formyl thymidine derivative.
  • 3'-C-formyl nucleotide analog a radical carbonylation of the corresponding 3 ' -deoxy-3 ' -iodo nucleoside gives the 3'-C-formyl nucleotide analog.
  • the iodo compound is treated with CO, 2,2' -azobisisobutrylonitrile (AIBN) , and tris (trimethylsilyl) silane (TTMS) .
  • AIBN 2,2' -azobisisobutrylonitrile
  • TTMS tris (trimethylsilyl) silane
  • 3'-C-formyl derivatized compounds can be synthesized from either a 3' -deoxy-3' -cyano sugar or nucleoside.
  • Both 4'-C- formyl (also identified as 5'-aldehydo) and 3'-C-formyl group can be blocked in a facile manner utilizing O- methylaminobenzenthiol as a blocking group.
  • the 4'- and 3'- C-formyl groups can be deblocked with silver nitrate oxidation.
  • An alternate method of 3'-C-formyl nucleoside synthesis employs l-O-methyl-3' -deoxy-3' -O-methylaminobenzene thiol-5' -O-trityl-S-D-erythro-pento furanoside, which serves as a precursor for any 3' -deoxy-3' -C-formyl nucleoside.
  • the l-O-methyl-3 ' -deoxy-3' -O-methyl amino benzenethiol-5' -O- trityl- ⁇ -D-erythro-pentofuranoside is reacted with an appropriate base utilizing standard glycosylation conditions and then deblocked to yield the nucleoside.
  • a 3' -deoxy-3' -cyano nucleoside is prepared from either the corresponding 3' -deoxy-3' -iodo nucleoside or by glycosylation with l-O-methyl-3' -deoxy-3' -O-cyano-5' -0- trityl- ⁇ -D-ervthro-pentofuranoside.
  • Oligonucleosides according to the invention linked by hydrazines, hydroxylamines and other linking groups, can be protected by a dimethoxytrityl group at the 5' -hydroxyl and activated for coupling at the 3'-hydroxyl with cyanoethyldiisopropyl-phosphite moieties.
  • oligonucleosides are linked with the units of a specified DNA sequence utilizing normal phosphodiester bonds.
  • the resulting oligonucleotide analog or oligomer has a "mixed" backbone containing both phosphodiester linkages and four atom linkages of the inventions.
  • a sequence-specific 15-mer oligonucleotide can be synthesized to have seven hydroxylamine, hydrazine or other type linked dinucleosides attached via alternating phosphodiester linkages.
  • Such a structure will provide increased solubility in water compared to fully modified oligomers, which may contain linkages of the invention.
  • Oligonucleosides containing a uniform backbone linkage can be synthesized by use of CPG-solid support and standard nucleic acid synthesizing machines such as Applied Biosystems Inc. 380B and 394 and Milligen/Biosearch 7500 and 8800s.
  • the initial nucleoside (number 1 at the 3 '-terminus) is attached to a solid support such as controlled pore glass .
  • each new nucleoside is attached either by manual manipulation or by the automated synthesizer system.
  • One preferred method, shown in Figure 1 employs a solid support to which has been bound a downstream synthon having a protected 5' site.
  • the 5' site preferably is protected with a phthalimido group.
  • DHMO 1,2-dihydro-4- hydroxy-2-methyl-l-oxaphthalazine
  • the 5' site of the downstream synthon can be liberated with any nucleophilic base having a pKa from about 7.7 to about 10.0.
  • the selected base should not appreciably react with any linker used to bind a growing oligonucleoside to a stationary phase.
  • Representative bases include hydrazine and methylhydrazine.
  • the 5' site preferably is liberated with 3% methylhydrazine in methylene chloride and washed with methylene chloride:methanol.
  • the aminohydroxyl group at the 5' position of the upstream synthon also is protected with a phthalimido group to yield a 5' -phthalimido protected 3'- deoxy-3' -C-formyl nucleoside, which is reacted with the downstream synthon in, for example, 2.5% acetic acid in methylene chloride.
  • Deprotection at the 5' position and washing liberates the next 5' -aminohydroxy reaction site.
  • the cycle is repeated with the further addition of upstream synthon until the desired sequence is constructed.
  • Each nucleoside of this sequence is connected with oxime linkages.
  • the terminal nucleoside of the desired oligonucleoside preferably is added to the sequence as a 5'-OTBDMS blocked 3' -deoxy-3' -C-formyl nucleoside.
  • the oxime linked oligonucleoside can be removed from the support with, for example, ammonium hydroxide. If an aminohydroxyl linked oligonucleoside is desired, the oxime linkages are reduced with sodium cyanoborohydride in acetic acid. Alternately reduction can be accomplished while the oxime linked oligonucleoside is still attached to the support.
  • Free amino groups produced upon reduction can be alkylated with, for example, acetone and sodium cyanoborohydride in acetic acid.
  • the alkylation step can be used to introduce other, useful, functional molecules on the macromolecule.
  • useful functional molecules include but are not limited to reporter molecules, RNA cleaving groups, groups for improving the pharmacokinetic properties of an oligonucleotide, and groups for improving the pharmacodynamic properties of an oligonucleotide.
  • Such molecules can be attached to or conjugated to the macromolecule via attachment to the nitrogen atom in the backbone linkage. Alternatively, such molecules can be attached to pendent groups extending from the 2' position of the sugar moiety of one or more of the nucleosides of the macromolecules.
  • pendent groups extending from the 2' position enhance binding or hybridization of the compound to a target nucleic acid.
  • Dimeric pairs of compounds of the invention linked via MMI [methylene (methylimino) ] linkages were studies by NMR. While we do not wish to be bound by theory, the sugar moieties of RNA and DNA oligonucleotides normally are located in one of two major conformations, either the C2'-endo conformation (DNA like or southern conformation) or the C3'-endo conformation (RNA like or northern conformation) .
  • RNA character imparted by the MMI linkage might contribute to the +0.2 °C increase in Tm per modification for MMI oligomers as compared to normal phosphodiester oligonucleotides.
  • RNA like characteristic were imparted to the dimers.
  • the compounds of this invention can be used in diagnostics, therapeutics, and as research reagents and kits.
  • the oligonucleotide analog is administered to an animal suffering from a disease modulated by some protein. It is preferred to administer to patients suspected of suffering from such a disease an amount of oligonucleotide analog that is effective to reduce the symptomology of that disease.
  • One skilled in the art can determine optimum dosages and treatment schedules for such treatment regimens .
  • RNA or DNA portion which is to be modulated be preselected to comprise that portion of DNA or RNA which codes for the protein whose formation or activity is to be modulated.
  • the targeting portion of the composition to be employed is, thus, selected to be complementary to the preselected portion of DNA or RNA, that is to be an antisense oligonucleotide for that portion.
  • the compounds of the invention hybridize to HIV mRNA encoding the tat protein, or to the TAR region of HIV mRNA. In another preferred embodiment, the compounds mimic the secondary structure of the TAR region of HIV mRNA, and by doing so bind the tat protein.
  • Other preferred compounds complementary sequences for herpes, papilloma and other viruses are also preferred.
  • RNA for research and diagnostic purposes. Such selective, strong binding is accomplished by interacting such RNA or DNA with compositions of the invention which are resistant to degradative nucleases and which hybridize more strongly and with greater fidelity than known oligonucleotides or oligonucleotide analogs.
  • CPG beads were washed thoroughly with dichloromethane methanol (3 x 50 ml, 1:1, v/v) and cloudy solution was decanted. Finally, CPG beads 2 were filtered off and washed with ethyl ether and dried under vacuum for 6 hours.
  • the CPG beads were filtered off and washed thoroughly with pyridine (30 ml) , dichloromethane (30 ml) and ether (30 ml) . The CPG beads then were dried under vacuum for 2 hours . Then, the CPG beads were shaken with piperidine (10 ml) for 15 minutes, filtered off and washed with dichloromethane (30 ml) ether (30 ml) and dried under vacuum for 4 hours. The resulting beads were suspended in glacial acetic acid (10 ml) and sonicated for 30 minutes. The suspension was diluted with methanol dichloromethane (3 x 30 ml, 1:1 v/v) and cloudy solution was decanted.
  • Example 2A The procedure of Example 2A was repeated except that 5' -O-phthalimido-2' -deoxyadenosine 4 . was used in place of 5' -O-phthalimidothymidine.
  • the loading was 36 ⁇ mol of 4 . per gram of CPG.
  • Example 2A The procedure of Example 2A was repeated except that 5' -O-phthalimido-N-2-isobutyryl-) -6-diphenylcarbamoyl- 2' -deoxyguanosine 5 was used in place of 5'-0- phthalimidothymidine.
  • the loading was 38 ⁇ mol of 5 . per gram of CPG.
  • Example 2A The procedure of Example 2A was repeated except that 5' -0-phthalimido-N-4-benzoyl-2' -deoxycytidine 6 . was used in place of 5' -O-phthalimidothymidine. The loading was 32 ⁇ mol of 6 . per gram of CPG.
  • Example 2A The procedure of Example 2A is repeated except that 5' -0-phthalimido-5-methyl-N-4-benzoyl-2' -deoxycytidine 7 . is used in place of 5' -O-phthalimidothymidine.
  • EXAMPLE 3 The procedure of Example 2A is repeated except that 5' -0-phthalimido-5-methyl-N-4-benzoyl-2' -deoxycytidine 7 . is used in place of 5' -O-phthalimidothymidine.
  • 5' -O-Phthalimidothymidine-3' -O-succinyl-CPG (7 . , 28 mg, l ⁇ mol of nucleoside) was packed into a small column and connected on an ABI DNA synthesizer model 380B.
  • Compound 7 . was treated with a solution 3% N-methyl hydrazine in dichloromethane: methanol (9:1 v/v) for 120 seconds with a 300 second wait step to give 5' -0-amino-thymidine-3' -0- succinyl-CPG.
  • Step 2 The CPG was washed with dichloromethane for 240 seconds.
  • the CPG was washed with dichloromethane for 150 seconds.
  • Steps 1-4 were repeated ten times.
  • the 5-terminal nucleotide e . g. , 5'-t- butyldiphenylsilyl-3' -aldehyde-3' -deoxy-thymidine 18 .
  • the excess of 18 was removed by washing step 4.
  • the resulting CPG beads (with oligomer) were transferred into a small flask and 1.0 ml of glacial acetic acid was added. NaCNBH 3 (15 mg) was added and the mixture was shaken and sonicated for .15 minutes. Formaldehyde (20% in water, lOO ⁇ l) was added, the mixture was sonicated for 15 minutes and NaCNBH 3 (2 x 15mg) was added with 15 minutes sonication between each addition. The resulting beads were washed with methanol dichloromethane (1:1, v/v, 15 ml) . H. Step 8.
  • Oligomer was cleaved from the solid support by treatment with 30% ammonia for 2 hours at room temperature. The ammonia then was evaporated. I . Step 9 .
  • T.T oxime dimer was prepared as described in Example 3. Thus, twice repeating the steps 1-4 provided T.T oxime dimer. This dimer was further extended by repeating steps 1-4 using a 5'0-FMOC (9- fluorenylmethoxycarbonyl) protected nucleoside 22_ instead of nucleoside 12 . during step 3. This modification provides a convenient way of switching to standard phosphoramidite chemistry without removing the CPG from automated DNA synthesizer. The resulting T.T.T oxime trimer, still attached to CPG, was treated with a solution of 5% piperidine in acetonitrile for two minutes with a flow rate of 1.6 ml/minute.
  • a three minute waiting step and a three minute acetonitrile wash step following the piperidine treatment removed the 5'-0-FMOC group completely (see, e . g. , Ma et al . Biopolymers 1989, 28, 965-993) .
  • T * T.T oxime trimer bearing a free 5' -OH group now was ready for a standard phosphoramidite cycle of DNA synthesis.
  • Thymidine was incorporated as the last residue utilizing a procedure generally in accordance with Oligonucleotide Synthesis, M.J. Gait, ed., IRL Press, Oxford, 1984, to furnish a tetramer.
  • the two oxime linkages of the tetramer were reduced and methylated following
  • T.T.T oxime trimer was prepared as described in Example 4.
  • the last thymidine nucleoside residue was incorporate utilizing standard phosphoramidite chemistry, followed sulfurization with Beaucage reagent (see, Beaucage et al . , Tetrahedron 1992, 48, 2223-2312) to provide a CPG attached tetramer.
  • the CPG was taken off the DNA synthesizer and treated in a manner described in steps 7 and 8 of Example 3.
  • the product was purified by reverse phase HPLC as described in step 10 of Example 3.
  • the 5'-0-trityl off phosphorothioate oligonucleotide (T s C s G s C s T s G s G s T s G s A s G s TsT s T s C) was synthesized on a DNA synthesizer in l ⁇ M scale on a CPG support utilizing standard phosphoramidite chemistry, (see, Zon in Protocols For Oligonucleotides and Analogs, pages 165-189, S. Agrawal, Humana Press, Totowa, NJ, 1993) .
  • coupling was performed with the 5' -O-phthalimido-3' -O-phosphoramidite derivative of thymidine 2$.
  • the terminal TBDPS group was then removed by TBAF treatment as described in step 9 of Example 3.
  • the crude, chimeric oligomer was then purified by HPLC in a manner described in step 10 of Example 3.
  • the purified material had a retention time of 22 . 1 minutes .
  • the chromatographic peak was broad due to a mixture of phosphorothioate diastereomers .
  • Example 6 The synthesis of the phosphorothioate portion is accomplished by standard phosphoramidite chemistry protocol as described in Example 6. The resulting chimeric oligomer with a 3' -MMI tail/cap is then purified by HPLC. Alternatively, the procedure described in Example 5 can be employed to prepare chimeric oligomers of any desired length and base composition.
  • Synthesis of 3' - and 5' -end capped chimeric oligomers is accomplished by combination of the methods described in Examples 3 and 5.
  • 5'-0-DMT blocked oligomer T* ⁇ *T*C is synthesized by incorporating appropriate building blocks (compounds ⁇ , 2J3 and 13.) following the protocol described in Example 3 (steps 1-6) .
  • the 5'-0-DMT group is removed from the CPG-attached oligomer utilizing standard acidic deprotection.
  • a phosphorothioate chain of desired length is synthesized onto the oxime tetramer.
  • the 5'-cap of the MMI tetramer is attached following the protocol of Example 6(A) to furnish the title compound.
  • the process consists of two steps on an automated DNA synthesizer ( e . g. , ABI 380 B) and two steps off the synthesizer.
  • the oligomers were cleaved from the solid support (CPG) by ammonia treatment (55°C, 5 hours) and the resulting solution was concentrated in vacuo . The residue was diluted with 300 ⁇ l of water and the samples were analyzed by HPLC. A reverse phase C18 column (5 ⁇ m, 5.0 X 0.46 cm) and a short (1 cm) guard column were used with a flow-rate of
  • Each dimer bears a 5'-0-TBDPS group, which was left on the molecule to increase lipophilicity during HPLC analysis.
  • EXAMPLE 8 Synthesis Of 5' -O-Phthalimido-2' -deoxynucleoside Derivatives. A. 5' -O-Phthalimidothymidine, 3 .
  • the 5' -O-phthalimidothymidine was prepared by the procedure described in the Example 6 of Application Serial No. 08/040,903.
  • 2' -Deoxyadenosine was conveniently transformed into 5' -0-tert-butyldiphenylsilyl-2' ,3' -dideoxy-3' -C- styryladenosine in 60% yield utilizing the radical reaction described in Example 82 of Application Serial No. 08/040,903.
  • the 5'-0-TBDPS group of the latter compound was deblocked to provide 2', 3' -dideoxy-3' -C-styryladenosine in 91% yield, mp 215-217°C.
  • Oxidative cleavage (0 s 0 4 /NaI0 4 ) of the styryl group of the latter compound furnished the title compound in 55% yield as a white powder.
  • 3 '-C-styryl nucleoside derivatives of 2' -deoxycytidine, 2' -deoxyadenosine, and 2'- deoxyguanosine are transformed into 3' -C-formyl-5' -O-FMOC- derivatives of 2' -deoxycytidine 2JL, 2' -deoxyadenosine 2_4, and 2' -deoxyguanosine .25 . , respectively.
  • Luminescence Spectrophotometer PERKIN ELMER LS50B
  • N-Hydroxyphthalimide (10 g, 59.5 mmoles) was dissolved in anhydrous CH 2 C1 2 (300 ml) and N-methyl hydrazine (9.7 ml) was added to the stirred solution. The solution turned red and then yellow. When all the starting material had disappeared, the solvent was evaporated and the resulting oil was crystallized from anhydrous ethanol as fine needles.
  • Analysis Protocol A Neutral conditions The spectrometer was turned on 0.5 hour prior to the measurement . A blank run was performed using spectrophotometer grade methanol.
  • 1,2-Dihydro-4-hydroxy-2- methyl-1-oxophthalazine (0.234 mg) was then dissolved in methanol (100 ml) and the sample analyzed on the spectrophotometer. The excitation wavelength was chosen at 298 nm. and the corresponding emission was observed at 400 nm. The following concentrations ( ⁇ Mol) of 1,2-Dihydro-4- hydroxy-2-methyl-1-oxophthalazine were tested:
  • Example 11 (A) The procedure of Example 11 (A) was repeated except that the oxophthalazine sample was basified by addition of N- methyl hydrazine (1% volume) . Basification results in the decay of emission intensity by 25 units and moves the excitation wavelength to 485 nm.
  • Example 11 (A) The procedure of Example 11 (A) was repeated except that the oxophthalazine sample was acidified with glacial acetic acid (0.1 ml) . The intensities of the emission was enhanced considerably and the values are summarized below.
  • MMI dimers in which one of the starting nucleosides bears an amide protecting group on the base for example i ⁇ -benzoyl-S- methylcytosine
  • the oxime was stirred overnight with methanolic ammonia, and the resulting solution concentrated to provide the crude product.
  • the so obtained intermediate, base unprotected oxime dimer was dissolved in glacial acetic acid (0.1 M) , placed in an ice bath, and sodium cyanoborohydride (3 x 2 eq) was added over 15 minutes. The mixture was allowed to warm to room temperature over 1 hour, placed in an ice bath, and aqueous formaldehyde (20 eq, 20%) was added in one portion.
  • the silylated MMI dimer was dissolved in dry THF (0.1 M) and cooled in an ice bath. A solution of tetrabutylammonium fluoride in THF (1 M, 1.5 eq per silyl group) was added dropwise over 5 minutes. The solution was stirred at 0 °C until the reaction was complete as judged by TLC (1-2 hours) , at which point silica (5 g/mmol) was added, and the mixture carefully concentrated. The silica was applied to a column packed in 5% MeOH/CH 2 Cl 2 , and eluted with 5% to 10% MeOH/CH 2 Cl 2. The appropriate fractions were combined, concentrated, and the residue azeotroped with EtOAc to provide the silylated product .
  • reaction mixture was then directly loaded onto a column packed with 25% EtOAc/hexane + 0.1% Et 3 N, and eluted with a stepwise gradient to EtOAc + 0.1% Et 3 N.
  • Fractions containing only the product were pooled and concentrated to yield a hard foam, which was lyophilized from dry 1,4-dioxane to afford the phosphoramidite as a fine white powder.
  • the crude oxime dimer was then evaporated and dissolved in (1/1 : v/v) mixture of water and 1,4-dioxane with NaI0 4 (0.503 mmole, 2.2 eq, ) osmium tetroxide (4% solution in water, 0.229 mmole, O.leq.), 4-methyl-morpholine-N-oxide (0.332 mmole, 1.45 eq.) before being stirred in the dark for 4 hours. After work-up (same as described in the example 9 above) , the residue was chromatographed on silica gel column with 5% MeOH/CH 2 Cl 2 to yield 36% (0.082 mmole) of a white foam.
  • 5' -O-phthalimido-3' -deoxy-3' -C-formyl-5-methyl-i ⁇ -benzoyl- cytidine (0.150 mmole, leq.) and 5' -O-amino-3' -O-TBDPS- thymidine (0.15 mmole, leq.) in CH 2 C1 2 were coupled following the general procedure described above.
  • the MMI tetramer was purified by silica gel column chromatography using 3% MeOH/CH 2 Cl 2 to yield 56% over four synthetic steps (oxime coupling, debenzoylation, reductive methylation and rebenzoylation) of the title compound.
  • MMI tetramer (0.113 mmole, leq.) was dimethoxytritylated following the general procedure described above. MMI tetramer was purified by silica gel column chromatography. Elution with 5% MeOH/CH 2 Cl 2 , pooling and evaporation furnished 81% yield of the desired compound.
  • Tetameric MMI containing oligonucleotides correspond to chimeric oligonucleotides where several phosphodiesters and/or phosphorothioates linkages have been replaced by at least three consecutive methylene (methyl) imino (MMI) internucleosidic link.
  • MMI methylene (methyl) imino
  • MMI tetramer (or longer chain) containing chimeric oligonucleotides can be - synthesized using standard phosphoramidite chemistry.
  • the chimeric oligomers were synthesized attached to CPG (Controlled Pore Glass) solid support and the assembling was performed on an automated DNA synthesizer such as Millipore Expedite or Applied Biosystem 380B.
  • the initial deoxynucleoside or MMI tetramer determining the 3'-end of the MMI containing chimeric oligonucleotide was loaded on to the CPG solid support via a succinyl linkage following the procedure described in Example 2 above (for example: 5' -0-DMTr-T*T*T*C-0- (succinyl-CPG-NMe 2 ) -3' MMI was loaded onto the CPG providing a loading of 22.67 mmole/g) .
  • the chain elongation of chimeric oligomers was performed in a similar manner as described for the MMI phosphoramidite Dimers.
  • Table 1 Chimeric Oligomers Containing T*T*T*C and ⁇ *T* M ⁇ C*T MMI Tetramers.
  • the aqueous layer was slowly diluted to a total volume of 350 mL with cold water, at whic point solid began forming.
  • the ether was decanted, an additional portion ether was added, stirred, and decanted, and the process repeated.
  • the solution was now diluted to a total volume of 500 mL with water, and allowed to stand on ice for several hours.
  • the reaction mixture was cooled to room temperature and diluted with CH 2 C1 2 (50 ml) .
  • the CH 2 C1 2 suspension was transferred onto the top of a prepacked silica gel (CH 2 C1 2 ) column.
  • Elution with CH 2 C1 2 /CH 2 C1 2 :MeOH (9:1, v/v) furnished the desired product as homogenous material .
  • Appropriate fractions were pooled and concentrated to provide 0.6 g (29%) of the title compound (contaminated with 20% of the 3'-0-methyl isomer) and 0.61 g (31%) of the unreacted starting material.
  • Triethylamine (25 mL) and water (25 mL) were added and the mixture was concentrated to a small volume, diluted with EtOAc (500 mL) and extracted with 5% aqueous sodium bicarbonate (2 x 200 mL) , water (100 mL) , brine (100 mL) , then dried over magnesium sulfate, filtered and concentrated to afford 10.7 g (106%) of a foam. This material was azeotroped with dry 1,4-dioxane, then dissolved in 160 mL of the same solvent .
  • reaction mixture was then coevaporated with toluene (3 x 50 ml) under vacuum.
  • the reaction was complete (by TLC) by the third coevaporation.
  • NaCNBH 3 (3 x 250 mg, 12 mmol) was added to the stirred reaction mixture in small portions (fume-hood) .
  • the stirring was continued for 30 minutes and additional amount of NaCNBH 3 (3 x 250 mg, 12 mmol) was added in a similar manner.
  • reaction mixture was poured into ice-water (250 ml) and extracted with CH 2 C1 2 (2 x 250 ml) .
  • CH 2 C1 2 layer was washed with water (2 x 250 ml) and dried MgS0 4 ) .
  • the solvent was removed and the residue purified by silica gel column chromatography. Elution with a gradient of CH 2 Cl 2 -CH 2 Cl 2 :MeOH (95:5, v/v) provided the desired product as homogenous material. Appropriate fractions were pooled and concentrated to furnish 5.08 g (50%) of the 3', 5' -protected MMI dimer.
  • DMT-0-T*T-OH (1.78 g, 2.19 mmol) was phosphitylated according to the general procedure, except that the organic layer was extracted with 5% aqueous sodium bicarbonate, dried, filtered, and concentrated prior to chromatography (1% MeOH/CH 2 Cl 2 + 0.1% Et 3 N) . This resulted in contamination with an impurity, and additional chromatography (1:1 CH 2 Cl 2 /EtOAc + 0.1% Et 3 N to 2% MeOH in 1:1 CH 2 Cl 2 /EtOAc + 0.1% Et 3 N) was necessary to completely purify the product.
  • MMI methyl imino
  • the chimeric oligonucleotides containing MMI dimers were synthesized utilizing standard phosphoramidite chemistry.
  • the chimeric oligomers were synthesized attached to CPG (Controlled Pore Glass) solid support and the assembling was performed on an automated DNA synthesizer such as
  • the initial deoxynucleoside or MMI dimer determining the 3'-end of the MMI containing chimeric oligonucleotide was loaded on to the CPG solid support via a succinyl linkage following the procedure described in Example 2 above.
  • Step 4 Capping:
  • Step 5- Isolation And Purification At the end of the automated synthesis, MMI containing oligonucleotides were concomitantly deprotected and cleaved from the solid support by treatment with concentrated aqueous ammonia solution (30%) at 55 °C for 10 hours. The ammonia solution was then evaporated and the full length oligonucleotides were separated from the failure sequences on reverse phase HPLC (column: RCM Waters 8 X 10 Bondapack HC18HA, flow rate 2 ml/minutes, gradient 5% to 25% CH,CN in TEAA 0.05 N pH 7.00 over 25 minutes) .
  • the flask containing the mixture was then equipped with a magnetic stir bar, a rubber septum, and an argon inlet and dissolved in 7.5 mL CH 2 C1 2 and diluted with 7.5 mL MeOH.
  • Bromocresol green indicator solution in MeOH was added dropwise to give a more notable yellow color to the solution and a catalytic amount of AcOH was also added at this time.
  • the mixture was then equilibrated in an ice water bath. At ice bath temperature, NaCNBH 3 (0.850 g, 13.52 mmol) was added in four approximately equal portions over a period of 4 h. Over this time, HOAc was added dropwise upon the appearance of blue-green color to restore the original gold-yellow tint and maintain the pH of the mixture.
  • the propypyreneyl T-T dimer of Example 17 was incorporated into a oligomer of the sequence Gp 0 G po Ap 0 T*Tp 0 Gp 0 Gp 0 Tp 0 C in a lO ⁇ mol synthesis using the standard protocols above.
  • the oligomer was further shown by CGE to be full length material free of short mer sequences.
  • PROCEDURE 1 Nuclease Resistance A. Evaluation of the resistance of oligonucleotide- mimicking macromolecules to serum and cytoplasmic nucleases.
  • Oligonucleotide-mimicking macromolecules of the invention can be assessed for their resistance to serum nucleases by incubation of the oligonucleotide-mimicking macromolecules in media containing various concentrations of fetal calf serum or adult human serum. Labelled oligonucleotide-mimicking macromolecules are incubated for various times, treated with protease K and then analyzed by gel electrophoresis on 20% polyacrylamine-urea denaturing gels and subsequent autoradiography. Autoradiograms are quantitated by laser densitometry.
  • cytoplasmic nucleases an HL 60 cell line can be used. A post-mitochondrial supernatant is prepared by differential centrifugation and the labelled macromolecules are incubated in this supernatant for various times. Following the incubation, macromolecules are assessed for degradation as outlined above for serum nucleolytic degradation. Autoradiography results are quantitated for evaluation of the macromolecules of the invention. It is expected that the macromolecules will be completely resistant to serum and cytoplasmic nucleases.
  • oligonucleotide- mimicking macromolecules to specific endo- and exo-nucle- ases.
  • Evaluation of the resistance of natural oligonucleotides and oligonucleotide-mimicking macromolecules of the invention to specific nucleases i.e., endonucleases, 3',5'-exo-, and 5' , 3' -exonucleases
  • the oligonucleotide-mimicking macromolecules are incubated in defined reaction buffers specific for various selected nucleases.
  • urea is added and analysis on 20% polyacrylamide gels containing urea is done. Gel products are visualized by staining with Stains All reagent (Sigma Chemical Co.) . Laser densitometry is used to quantitate the extent of degradation.
  • the effects of the macromolecules linkage are determined for specific nucleases and compared with the results obtained from the serum and cytoplasmic systems. As with the serum and cytoplasmic nucleases, it is expected that the oligonucleotide- mimicking macromolecules of the invention will be completely resistant to endo- and exo-nucleases.
  • PROCEDURE 2 5-Lipoxygenase Analysis and Assays A. Therapeutics
  • an animal suspected ' of having a disease characterized by excessive or abnormal supply of 5-lipoxygenase is treated by administering the macromolecule of the invention.
  • Persons of ordinary skill can easily determine optimum dosages, dosing methodologies and repetition rates. Such treatment is generally continued until either a cure is effected or a diminution in the diseased state is achieved. Long term treatment is likely for some diseases.
  • the oligonucleotide-mimicking macromolecules of this invention will also be useful as research reagents when used to cleave or otherwise modulate 5-lipoxygenase mRNA in crude cell lysates or in partially purified or wholly purified RNA preparations.
  • This application of the invention is accomplished, for example, by lysing cells by standard methods, optimally extracting the RNA and then treating it with a composition at concentrations ranging, for instance, from about 100 to about 500 ng per 10 Mg of total RNA in a buffer consisting, for, example, of 50 mm phosphate, pH ranging from about 4-10 at a temperature from about 30° to about 50° C.
  • the cleaved 5-lipoxygenase RNA can be analyzed by agarose gel electrophoresis and hybridization with radiolabeled DNA probes or by other standard methods.
  • the oligonucleotide-mimicking macromolecules of the invention will also be useful in diagnostic applications, particularly for the determination of the expression of specific mRNA species in various tissues or the expression of abnormal or mutant RNA species.
  • the macromolecules target a abnormal mRNA by being designed complementary to the abnormal sequence, they would not hybridize to normal mRNA.
  • Tissue samples can be homogenized, and RNA extracted by standard methods.
  • the crude homogenate or extract can be treated for example to effect cleavage of the target RNA.
  • the product can then be hybridized to a solid support which contains a bound oligonucleotide complementary to a region on the 5' side of the cleavage site. Both the normal and abnormal 5' region of the mRNA would bind to the solid support. The 3' region of the abnormal RNA, which is cleaved, would not be bound to the support and therefore would be separated from the normal mRNA.
  • Targeted mRNA species for modulation relates to 5- lipoxygenase; however, persons of ordinary skill in the art will appreciate that the present invention is not so limited and it is generally applicable.
  • the inhibition or modulation of production of the enzyme 5-lipoxygenase is expected to have significant therapeutic benefits in the treatment of disease.
  • an assay or series of assays is required. D. In Vitro Assays
  • the cellular assays for 5-lipoxygenase preferably use the human promyelocytic leukemia cell line HL-60. These cells can be induced to differentiate into either a monocyte like cell or neutrophil like cell by various known agents. Treatment of the cells with 1.3% dimethyl sulfoxide, DMSO, is known to promote differentiation of the cells into neutrophils. It has now been found that basal HL-60 cells do not synthesize detectable levels of 5-lipoxygenase protein or secrete leukotrienes (a downstream product of 5-lipoxygenase) . Differentiation of the cells with DMSO causes an appearance of 5-lipoxygenase protein and leukotriene biosynthesis 48 hours after addition of DMSO.
  • DMSO dimethyl sulfoxide
  • 5-lipoxygenase protein synthesis can be utilized as a test system for analysis of oligonucleotide- mimicking macromolecules which interfere with 5-lipoxygenase synthesis in these cells.
  • a second test system for oligonucleotide-mimicking macromolecules makes use of the fact that 5-lipoxygenase is a "suicide" enzyme in that it inactivates itself upon reacting with substrate.
  • Treatment of differentiated HL-60 or other cells expressing 5 lipoxygenase, with 10 ⁇ M A23187, a calcium ionophore promotes translocation of 5-lipoxygenase from the cytosol to the membrane with subsequent activation of the enzyme.
  • a direct effect which oligonucleotide-mimicking macromolecules can exert on intact cells and which can be easily be quantitated is specific inhibition of 5-lipoxygenase protein synthesis.
  • cells can be labelled with 35 S-methionine (50 ⁇ Ci/mL) for 2 hours at 37° C to label newly synthesized protein.
  • Cells are extracted to solubilize total cellular proteins and 5-lipoxygenase is immunoprecipitated with 5-lipoxygenase antibody followed by elution from protein A Sepharose beads.
  • the immunoprecipitated proteins are resolved by SDS-polyacrylamide gel electrophoresis and exposed for autoradiography.
  • the amount of immunopre ⁇ cipitated 5-lipoxygenase is quantitated by scanning densitometry.
  • a predicted result from these experiments would be as follows.
  • the amount of 5-lipoxygenase protein immuno- precipitated from control cells would be normalized to 100%.
  • Treatment of the cells with 1 ⁇ M, 10 ⁇ M, and 30 ⁇ M of the macromolecules of the invention for 48 hours would reduce immunoprecipitated 5-lipoxygenase by 5%, 25% and 75% of control, respectively.
  • Cytosolic proteins are incubated with 10 ⁇ M 14 C-arachidonic acid, 2mM ATP, 50 ⁇ M free calcium, 100 ⁇ g/ml phosphatidylcholine, and 50 mM bis-Tris buffer, pH 7.0, for 5 min at 37° C.
  • the reactions are quenched by the addition of an equal volume of acetone and the fatty acids extracted with ethyl acetate.
  • the substrate and reaction products are separated by reverse phase HPLC on a Novapak C18 column (Waters Inc., Millford, MA) . Radioactive peaks are detected by a Beckman model 171 radiochromatography detector. The amount of arachidonic acid converted into di-HETE's and mono-HETE's is used as a measure of 5-lipoxygenase activity.
  • a predicted result for treatment of DMSO differentiated HL-60 cells for 72 hours with effective the macromolecules of the invention at 1 ⁇ M, 10 ⁇ M, and 30 ⁇ M would be as follows. ' Control cells oxidize 200 pmol arachidonic acid/ 5 min/ 10 6 cells. Cells treated with 1 ⁇ M, 10 ⁇ M, and 30 ⁇ M of an effective oligonucleotide-mimicking macromolecule would oxidize 195 pmol, 140 pmol, and 60 pmol of arachidonic acid/ 5 min/ 10 s cells respectively.
  • a quantitative competitive enzyme linked immunosorbant assay (ELISA) for the measurement of total 5-lipoxygenase protein in cells has been developed. Human 5-lipoxygenase expressed in E.
  • coli and purified by extraction, Q-Sepharose, hydroxyapatite, and reverse phase HPLC is used as a standard and as the primary antigen to coat microtiter plates. 25 ng of purified 5-lipoxygenase is bound to the microtiter plates overnight at 4°C. The wells are blocked for 90 min with 5% goat serum diluted in 20 mM Tris-HCL buffer, pH 7.4, in the presence of 150 mM NaCl (TBS) .
  • Cell extracts (0.2% Triton X- 100, 12,000 x g for 30 min.) or purified 5-lipoxygenase were incubated with a 1:4000 dilution of 5-lipoxygenase polyclonal antibody in a total volume of 100 ⁇ L in the microtiter wells for 90 min.
  • the antibodies are prepared by immunizing rabbits with purified human recombinant 5-lipoxygenase.
  • the wells are washed with TBS containing 0.05% tween 20 (TBST) , then incubated with 100 ⁇ L of a 1:1000 dilution of peroxidase conjugated goat anti-rabbit IgG (Cappel Laboratories, Malvern, PA) for 60 min at 25°C.
  • the wells are washed with TBST and the amount of peroxidase labelled second antibody determined by development with tetramethylbenzidine.
  • Predicted results from such an assay using a 30 mer oligonucleotide-mimicking macromolecule at 1 ⁇ M, 10 ⁇ M, and 30 ⁇ M would be 30 ng, 18 ng and 5 ng of 5-lipoxygenase per 10 6 cells, respectively with untreated cells containing about 34 ng 5-1ipoxygenase.
  • a net effect of inhibition of 5-lipoxygenase biosynthesis is a diminution in the quantities of leukotrienes released from stimulated cells.
  • DMSO-differentiated HL-60 cells release leukotriene B4 upon stimulation with the calcium ionophore A23187.
  • Leukotriene B4 released into the cell medium can be quantitated by radioimmunoassay using commercially available diagnostic kits (New England Nuclear, Boston, MA) .
  • Leukotriene B4 production can be detected in HL-60 cells 48 hours following addition of DMSO to differentiate the cells into a neutrophil- like cell. Cells (2 x 10 5 cells/mL) will be treated with increasing concentrations of the macromolecule for 48-72 hours in the presence of 1.3% DMSO.
  • the cells are washed and resuspended at a concentration of 2 x 10 6 cell/mL in Dulbecco's phosphate buffered saline containing 1% delipidated bovine serum albumin.
  • Cells are stimulated with 10 ⁇ M calcium ionophore A23187 for 15 min and the quantity of LTB4 produced from 5 x 10 5 cell determined by radioimmunoassay as described by the manufacturer. Using this assay the following results would likely be obtained with an oligonucleotide-mimicking macromolecule directed to the 5-LO mRNA.
  • Cells will be treated for 72 hours with either 1 ⁇ M, 10 ⁇ M or 30 ⁇ M of the macromolecule in the presence of 1.3% DMSO.
  • the quantity of LTB 4 produced from 5 x 10 5 cells would be expected to be about 75 pg, 50 pg, and 35 pg, respectively with untreated differentiated cells producing 75 pg LTB 4 .
  • the edematous response is quantitated by measurement of ear thickness and wet weight of a punch biopsy. Measurement of leukotriene B 4 produced in biopsy specimens is performed as a direct measurement of 5- lipoxygenase activity in the tissue. Oligonucleotide-mimicking macromolecules will be applied topically to both ears 12 to 24 hours prior to administration of arachidonic acid to allow optimal activity of the compounds. Both ears are pretreated for 24 hours with either 0.1 ⁇ mol, 0.3 ⁇ mol, or 1.0 ⁇ mol of the macromolecule prior to challenge with arachidonic acid. Values are expressed as the mean for three animals per concentration.
  • Inhibition of polymorphonuclear cell infiltration for 0.1 ⁇ mol, 0.3 ⁇ mol, and 1 ⁇ mol is expected to be about 10%, 75% and 92% of control activity, respectively. Inhibition of edema is expected to be about 3%, 58% and 90%, respectively while inhibition of leukotriene B 4 production would be expected to be about 15%, 79% and 99%, respectively.
  • PROCEDURE 3 Solid Support Biochemistry Reagents
  • Oligomers of the present invention also can advantageously be used as solid-phase biochemistry reagents (see, e . g. , "Solid-Phase Biochemistry - Analytical and Synthetic Aspects” , W. H. Scouten, ed. , John Wiley & Sons, New York, 1983) , notably in solid-phase biosystems, especially bioassays or solid-phase techniques which concerns diagnostic detection/quantitation or affinity purification of complementary nucleic acids ( see, e. g. , "Affinity Chromatography - A Practical Approach” , P. D. G. Dean, W. S. Johnson and F. A.
  • PROCEDURE 4 Investigation of mutant ⁇ -amyloid precursor protein ( ⁇ APP)
  • Point mutations in the gene encoding ⁇ -amyloid have been suggested in familial Alzheimer's disease (FAD) .
  • FAD familial Alzheimer's disease
  • MMI oligomers are used to probe for such point mutation expressed proteins.
  • a MMI containing oligomer of the selected sequence coding for the protein of interest is synthesized as above.
  • a fluorescein label is added to the oligomer using fluorescein phosphoramidite (Cat. No. 10-1963-95, Glen Research Corp., Sterling VA) . This amidite is attached in the normal manner as the last phosphate residue added.
  • the oligomers can be labeled with other reporter molecules (acridine or psoralen, both also available from Glen Research or rhodamine) before or after oligomer synthesis.
  • Labeled oligomers are contacted with tissue or cell samples suspected of abnormal ⁇ APP expression under conditions in which specific hybridization can occur, and the sample is washed to remove unbound oligomers. Label remaining in the sample indicates bound oligonucleotide and is quantitated using a fluorimeter, fluorescence microscope or other routine means.
  • Tissue or cell samples suspected of expressing a point mutation in the ⁇ APP gene are incubated with a fluorescein- labeled oligomer which is targeted to the mutant codon 717, codon 670 or codon 671 of ⁇ APP mRNA.
  • An identical sample of cells or tissues is incubated with a second labeled oligomer which is targeted to the same region of normal ⁇ APP mRNA, under conditions in which specific hybridization can occur, and the sample is washed to remove unbound oligomer. Label remaining in the sample indicates bound oligomer and can be quantitated using a fluorimeter or other routine means.
  • mutant ⁇ APP is indicated if the first sample binds labeled oligomer and the second sample does not bind fluorescent label. Double labeling can also be used with the oligomers to specifically detect expression of mutant ⁇ APP.
  • a single tissue sample is incubated with a rhodamine-labeled oligomer which is targeted to codon 717, codon 670 or codon 671 of mutant ⁇ APP mRNA and a fluorescein-labeled oligomer which is targeted to the translation initiation site of mutant ⁇ APP mRNA, under conditions in which specific hybridization can occur.
  • the sample is washed to remove unbound oligomer and labels are detected by and fluori etry with appropriate filters.
  • the presence of mutant ⁇ APP is indicated if the sample does not bind rhodamine-labeled oligomer but does retain the fluorescein label.
  • PROCEDURE 5 Detection of mutant H-ras gene expression
  • MMI oligomers are labeled after synthesis with fluorescein or other fluorescent tag as illustrated above.
  • the oligomers are labeled with other reporter molecules before or after oligomer synthesis.
  • Labeled oligomers are contacted with tissue or cell samples suspected of abnormal ras expression under conditions in which specific hybridization can occur, and the sample is washed to remove unbound oligomer. Label remaining in the sample indicates bound oligomer and is quantitated using a fluorimeter, fluorescence microscope or other routine means.
  • Tissue or cell samples suspected of expressing a point mutation in the H-ras gene are incubated with a fluorescein- labeled oligomer which is targeted to the mutant codon 12, codon 13 or codon 61 of H-ras mRNA.
  • An identical sample of cells or tissues is incubated with a second labeled oligomer which is targeted to the same region of normal H-ras mRNA, under conditions in which specific hybridization can occur, and the sample is washed to remove unbound oligomer. Label remaining in the sample indicates bound oligomer and can be quantitated using a fluorimeter or other routine means .
  • the presence of mutant H-ras is indicated if the first sample binds labeled oligomer and the second sample does not bind fluorescent label.
  • Double labeling can also be used with oligomers to specifically detect expression of mutant ras.
  • a single tissue sample is incubated with a rhodamine-labeled oligomer which is targeted to codon 12, codon 13 or codon 61 of mutant H-ras mRNA and a fluorescein-labeled oligomer which is targeted to the translation initiation site of ras mRNA, under conditions in which specific hybridization can occur.
  • the sample is washed to remove unbound oligomer and labels are detected by and fluorimetry with appropriate filters.
  • the presence of mutant ras is indicated if the sample does not bind rhodamine-labeled oligomer but does retain the fluorescein label .
  • PROCEDURE 6 Effect of MMI-containing antisense oligonucleotides on PKC- ⁇ mRNA levels: A549 cells were treated with compounds ID No.s 9495 and 9496 as well as a phosphorothioate oligonucleotide standard of the same sequence as compound ID No. 9495 above, at doses from 100 to 400 nM for four hours in the presence of the cationic lipids DOTMA/DOPE, washed and allowed to recover for an additional 20 hours. Total RNA was extracted and 20 ⁇ g of each was resolved on 1.2% gels and transferred to nylon membranes.
  • the MMI compounds exhibited greater activity than the test standard.
  • the high specific binding of the test compounds to the PKC- ⁇ sequence can also be used to distinguish PKC- ⁇ mRNA from other mRNA of other PKC isozymes such as the ⁇ , T and ⁇ isozymes.
  • PROCEDURE 7 Northern blot analysis of ras expression in vivo
  • Kingston, R.E. in Current Protocols in Molecular Biology, (F.M. Ausubel, R. Brent, R.E. Kingston, D.D. Moore, J.A. Smith, J.G. Seidman and K. Strahl, eds.), John Wiley and Sons, NY.
  • the ras probe was a Sall-Nhel fragment of a cDNA clone of the activated (mutant) H-ras mRNA having a GGC-to-GTC mutation at codon-12.
  • the control probe was G3PDH. Blots were prehybridized for 15 minutes at 68 °C with the QuickHyb hybridization solution (Stratagene, La Jolla, CA) .
  • the heat- denatured radioactive probe (2.5 x 10 s counts/2 ml hybridization solution) mixed with 100 ⁇ l of 10 mg/ml salmon sperm DNA was added and the membrane was hybridized for 1 hour at 68 °C.
  • the blots were washed twice for 15 minutes at room temperature in 2x SSC/0.1% SDS and once for 30 minutes at 60 °C with 0.1XSSC/0.1%SDS. Blots were autoradiographed and the intensity of signal was quantitated using an ImageQuant Phosphorlmager (Molecular Dynamics, Sunnyvale, CA) . Northern blots were first hybridized with the ras probe, then stripped by boiling for 15 minutes in 0.Ix SSC/0.1%SDS and rehybridized with the control G3PDH probe to check for correct sample loading.
  • the MMI-containing oligomers had IC 50 s of approximately 100 nM, 90 nM and 120 nM, respectively. All were more active in this assay than a known-active phosphorothioate oligonucleotide of the same sequence that was used as the test standard. This standard phosphorothioate oligonucleotide had C50 of approximately 150 nM.

Abstract

Compounds and methods for preparing oligonucleotide analogs are provided. In preferred embodiments, the methods involve solid-phase coupling of synthons bearing either 3'-electrophilic groups and 5'-nucleophilic groups or 5'-electrophilic groups and 3'-nucleophilic groups to form neutral, achiral oligomers.

Description

BACKBONE MODIFIED OLIGONUCLEOTIDE ANALOGS AND SOLID PHASE SYNTHESIS THEREOF
CROSS REFERENCE TO RELATED APPLICATIONS
This application is a continuation-in-part of U.S. Serial No. 08/174,379, filed December 28, 1993, which is a continuation-in-part of U.S. Serial No. 08/040,903, filed March 31, 1991, which is a continuation-in-part of PCT/US92/04294, filed May 21, 1992, and of U.S. Serial No. 903,160, filed June 24, 1992, which are continuations-in-part of U.S. Serial No. 703,619 filed May 21, 1991, which is a continuation-in-part of U.S. Serial No. 566,836 filed on August 13, 1990 and U.S. Serial No. 558,663 filed on July 27, 1990. This application also is related to the subject matter disclosed and claimed in the following patent applications filed herewith by the present inventors: U.S. Serial No.
08/39,979, filed March 30, 1993; U.S. Serial No. 08/40,526, filed March 31, 1993; and U.S. Serial No. 08/40,933, filed March 31, 1993. Each of these patent applications are assigned to the assignee of this application and are incorporated by reference herein.
FIELD OF THE INVENTION
This invention relates to the synthesis of nuclease resistant oligonucleotide analogs which are useful for therapeutics, diagnostics and as research reagents. In particular, the invention relates to solid-phase synthetic methods wherein amine-terminated synthons are coupled with aldehyde-terminated synthons to produce hydroxylamino- containing and/or hydrazino-containing covalent linkages. BACKGROUND OF THE INVENTION
It is well known that most of the bodily states in mammals, including most disease states, are effected by proteins. Proteins, either acting directly or through their enzymatic functions, contribute in major proportion to many diseases in animals and man.
Classical therapeutics generally has focused upon interactions with proteins in an effort to moderate their disease causing or disease potentiating functions. Recently, however, attempts have been made to moderate the production of proteins by interactions with the molecules (i.e., intracellular RNA) that direct their synthesis. These interactions have involved hybridization of complementary "antisense" oligonucleotides or certain analogs thereof to RNA. Hybridization is the sequence-specific hydrogen bonding of oligonucleotides or oligonucleotide analogs to RNA or to single stranded DNA. By interfering with the production of proteins, it has been hoped to effect therapeutic results with maximum effect and minimal side effects. The pharmacological activity of antisense oligonucleotides and oligonucleotide analogs, like other therapeutics, depends on a number of factors that influence the effective concentration of these agents at specific intracellular targets. One important factor for oligonucleotides is the stability of the species in the presence of nucleases. It is unlikely that unmodified oligonucleotides will be useful therapeutic agents because they are rapidly degraded by nucleases. Modification of oligonucleotides to render them resistant to nucleases therefore is greatly desired.
Modification of oligonucleotides to enhance nuclease resistance generally has taken place on the phosphorus atom of the sugar-phosphate backbone. Phosphorothioates, methyl phosphonates, phosphoramidates and phosphorotriesters have been reported to confer various levels of nuclease resistance. Phosphate-modified oligonucleotides, however, generally have suffered from inferior hybridization properties. See, e . g. , Cohen, J.S., ed. Oligonucleotides : Antisense Inhibi tors of Gene Expression, (CRC Press, Inc., Boca Raton FL, 1989) .
Another key factor is the ability of antisense compounds to traverse the plasma membrane of specific cells involved in the disease process. Cellular membranes consist of lipid-protein bilayers that are freely permeable to small, nonionic, lipophilic compounds and are inherently impermeable to most natural metabolites and therapeutic agents. See, e . g. , Wilson, Ann . Rev. Biochem . 1978, 47, 933. The biological and antiviral effects of natural and modified oligonucleotides in cultured mammalian cells have been well documented. It appears that these agents can penetrate membranes to reach their intracellular targets. Uptake of antisense compounds into a variety of mammalian cells, including HL-60, Syrian Hamster fibroblast, U937, L929, CV-1 and ATH8 cells has been studied using natural oligonucleotides and certain nuclease resistant analogs, such as alkyl triesters and methyl phosphonates. See, e . g. , Miller, et al . , Biochemistry 1977, 16, 1988; Marcus-Sekura, et al . , Nucleic Acids Research 1987, 15, 5749; and Loke, et al . , Top. Microbiol . Immunol . 1988, 141 , 282. Often, modified oligonucleotides and oligonucleotide analogs are internalized less readily than their natural counterparts. As a result, the activity of many previously available antisense oligonucleotides has not been sufficient for practical therapeutic, research or diagnostic purposes. Two other serious deficiencies of prior art compounds designed for antisense therapeutics are inferior hybridization to intracellular RNA and the lack of a defined chemical or enzyme-mediated event to terminate essential RNA functions.
Modifications to enhance the effectiveness of the antisense oligonucleotides and overcome these problems have taken many forms. These modifications include base ring modifications, sugar moiety modifications and sugar-phosphate backbone modifications. Prior sugar-phosphate backbone modifications, particularly on the phosphorus atom, have effected various levels of resistance to nucleases. However, while the ability of an antisense oligonucleotide to bind to specific DNA or RNA with fidelity is fundamental to antisense methodology, modified phosphorus oligonucleotides have generally suffered from inferior hybridization properties.
Replacement of the phosphorus atom has been an alternative approach in attempting to avoid the problems associated with modification on the pro-chiral phosphate moiety. For example, Matteucci, Tetrahedron Letters 1990, 31 , 2385 disclosed the replacement of the phosphorus atom with a methylene group. However, this replacement yielded unstable compounds with nonuniform insertion of formacetal linkages throughout their backbones. Cormier, et al . , Nucleic Acids Research 1988, 16, 4583, disclosed replacement of phosphorus with a diisopropylsilyl moiety to yield homopolymers having poor solubility and hybridization properties. Stirchak, et al . , Journal of Organic Chemistry 1987, 52, 4202 disclosed replacement of phosphorus linkages by short homopolymers containing carbamate or morpholino linkages to yield compounds having poor solubility and hybridization properties. Mazur, et al . , Tetrahedron 1984, 40, 3949, disclosed replacement of a phosphorus linkage with a phosphonic linkage yielded only a homotrimer molecule. Goodchild, Bioconjugate Chemistry 1990, 1, 165, disclosed ester linkages that are enzymatically degraded by esterases and, therefore, are not suitable for antisense applications.
The limitations of available methods for modification of the phosphorus backbone have led to a continuing and long felt need for other modifications which provide resistance to nucleases and satisfactory hybridization properties for antisense oligonucleotide diagnostics and therapeutics. OBJECTS OF THE INVENTION
It is an object of the invention to provide oligonucleotide analogs for diagnostic, research, and therapeutic use. It is a further object of the invention to provide oligonucleotide analogs capable of forming duplex or triplex structures.
It is a further object of the invention to provide oligonucleotide analogs having enhanced cellular uptake. Another object of the invention is to provide oligonucleotide analogs having greater efficacy than unmodified antisense oligonucleotides.
It is yet another object of the invention to provide methods for synthesis and use of oligonucleotide analogs.
These and other objects will become apparent to persons of ordinary skill in the art from a review of the present specification and the appended claims.
SUMMARY OF THE INVENTION The present invention provides novel compounds that mimic and/or modulate the activity of wild-type nucleic acids. In general, the compounds contain a selected nucleoside sequence which is specifically hybridizable with a targeted nucleoside sequence of single stranded or double stranded DNA or RNA. At least a portion of the compounds of the invention has structure I :
Figure imgf000008_0001
wherein:
L1-L2-L3-L4 is CHa-RA-NRi-CHa, CHa-NR^A-CHa, RA-NR^
CH2-CH2, CHa-CHa- Ri-RA, CHa-CHa-RA-NR^ or NR^A-CHa-CHa,- RA is 0 or NR2; each Rx and R2 are, independently, the same or different and are H; alkyl or substituted alkyl having 1 to about 10 carbon atoms; alkenyl or substituted alkenyl having 2 to about 10 carbon atoms; alkynyl or substituted alkynyl having 2 to about 10 carbon atoms; alkaryl, substituted alkaryl, aralkyl, or substituted aralkyl having 7 to about 14 carbon atoms; alicyclic; heterocyclic; a reporter molecule; an RNA cleaving group; a group for improving the pharmacokinetic properties of an oligonucleotide; or a group for improving the pharmacodynamic properties of an ■ oligonucleotide;
Bx is a nucleosidic base; n is an integer greater than 0; Q is 0, S, CH2, CHF or CF2; X is H; OH; alkyl or substituted alkyl having 1 to about 10 carbon atoms; alkaryl, substituted alkaryl, aralkyl, or substituted aralkyl having 7 to about 14 carbon atoms; F; Cl; Br; CN; CF3; 0CF3; OCN; 0-alkyl; S-alkyl; N-alkyl; O- alkenyl; S-alkenyl; N-alkenyl; S0CH3; S02CH3; 0N02; N02; N3; NH2; heterocycloalkyl; heterocycloalkaryl; aminoalkylamino; polyalkylamino or substituted silyl; an RNA cleaving group; a group for improving the pharmacokinetic properties of an oligonucleotide; or a group for improving the pharmacodynamic properties of an oligonucleotide.
The compounds of the invention are prepared by coupling preselected 3' -functionalized and 4' -functionalized nucleosides and/or oligonucleotides under conditions effective to form the above-noted L1-L2-L3-L4 linkages. In certain embodiments, a 3'-C-formyl nucleoside or oligonucleotide synthon is reacted with a support-bound 5'- hydroxylamino or 5'-hydrazino nucleoside or oligonucleotide synthon. In other embodiments, a 4'-C-formyl synthon is reacted with a support-bound 3' -methylhydroxylamino or 3' - methylhydrazino synthon. In still further embodiments, linkages having structure CH=N-RA-CH2, CH2-CH=N-RA, CH2-RA- N=CH, or RA-N=CH-CH2 where RA is 0 or NR-L are formed by coupling synthons having structures II and III:
Figure imgf000009_0001
II wherein:
Z-L and Y2 are selected such that
(i) Zj is C(0)H and Y2 is CH2RANH2; or (ii) Z± is CH2RANH2 and Y2 is C(0)H; or
(iii) Z is CH2C(0)H and Y2 is RANH2; or (iv) Zx is RANH2 and Y2 is H(0)CCH2; Y1 is OH, 0RHP, CH2OH, or CH2ORHP where RHP is a hydroxyl protecting group; (P) is a solid support; each Lc is, independently, a covalent linkage having structure CH=N-RA-CH2, CH2-CH=N-RA, CH2-RA-N=CH, RA-N=CH-CH2; CH2-NH-RA-CH2, CH2-CH2-NH-RA, CH2-RA-NH-CH2, RA-NH-CH2-CH2, 0- P(0)20-CH2, or O-P(S) (0)0-CH2; and n is 0-200.
BRIEF DESCRIPTION OF THE DRAWINGS
The numerous objects and advantages of the present invention may be better understood by those skilled in the art by reference to the accompanying figures, in which:
Figures 1 and 2 show exemplary compounds used in accordance with the invention.
Figure 3 is a synthetic scheme depicting a preferred method for preparation of oxime-linked oligonucleotide analogs.
DETAILED DESCRIPTION OF THE INVENTION
The term "nucleoside" as used in connection with this invention refers to a unit made up of a heterocyclic base and its sugar. The term "nucleotide" refers to a nucleoside having a phosphate group on its 3' or 5 ' sugar hydroxyl group. Thus nucleosides, unlike nucleotides, have no phosphate group. "Oligonucleotide" refers to a plurality of joined nucleotide units formed in a specific sequence from naturally occurring bases and pentofuranosyl groups joined through a sugar group by native phosphodiester bonds. This term refers to both naturally occurring and synthetic species formed from naturally occurring subunits.
The compounds of the invention generally can be viewed as "oligonucleotide analogs", that is, compounds which function like oligonucleotides but which have non-naturally occurring portions. Oligonucleotide analogs can have altered sugar moieties, altered base moieties or altered inter-sugar linkages. For the purposes of this invention, an oligonucleotide analog having non-phosphodiester bonds, i.e., an altered inter-sugar linkage, is considered to be an "oligonucleoside. " The term "oligonucleoside" thus refers to a plurality of nucleoside units joined by linking groups other than native phosphodiester linking groups. The term "oligomers" is intended to encompass oligonucleotides, oligonucleotide analogs or oligonucleosides. Thus, in speaking of "oligomers" reference is made to a series of nucleosides or nucleoside analogs that are joined via either natural phosphodiester bonds or other linkages, including the four atom linkers of this invention. Although the linkage generally is from the 3' carbon of one nucleoside to the 5' carbon of a second nucleoside, the term "oligomer" can also include other linkages such as 2' -5' linkages.
Oligonucleotide analogs also can include other modifications consistent with the spirit of this invention, particularly modifications that increase nuclease resistance. For example, when the sugar portion of a nucleoside or nucleotide is replaced by a carbocyclic moiety, it is no longer a sugar. Moreover, when other substitutions, such a substitution for the inter-sugar phosphorodiester linkage are made, the resulting material is no longer a true nucleic acid species. All such compounds are considered to be analogs. Throughout this specification, reference to the sugar portion of a nucleic acid species shall be understood to refer to either a true sugar or to a species taking the structural place of the sugar of wild type nucleic acids. Moreover, reference to inter-sugar linkages shall be taken to include moieties serving to join the sugar or sugar analog portions in the fashion of wild type nucleic acids.
This invention concerns modified oligonucleotides, i.e., oligonucleotide analogs or oligonucleosides, and methods for effecting the modifications. These modified oligonucleotides and oligonucleotide analogs exhibit increased chemical and/or enzymatic stability relative to their naturally occurring counterparts. Extracellular and intracellular nucleases generally do not recognize and therefore do not bind to the backbone-modified compounds of the invention. In addition, the neutral or positively charged backbones of the present invention can be taken into cells by simple passive transport rather than by complicated protein-mediated processes. Another advantage of the invention is that the lack of a negatively charged backbone facilitates sequence specific binding of the oligonucleotide analogs or oligonucleosides to targeted RNA, which has a negatively charged backbone and will repel similarly charged oligonucleotides. Still another advantage of the present invention is it presents sites for attaching functional groups that initiate cleavage of targeted RNA.
The modified internucleoside linkages of this invention are intended to replace naturally-occurring phosphodiester-5' -methylene linkages with four atom linking groups to confer nuclease resistance and enhanced cellular uptake to the resulting compound. Preferred linkages have structure CHa-RA-NRi-CHa, CE2-NR1-Rh-CU2 , RA-NR^C^-C^, CH2-CH2- NR^RA, CHz-CHa-RA-NRi, or NR^A-CHa-CHa where RA is 0 or NR2.
Generally, these linkages are prepared by functionalizing the sugar moieties of two nucleosides which ultimately are to be adjacent to one another in the selected sequence. In a 4' to 3' sense, an "upstream" synthon such as structure III is modified at its terminal 3' site, while a "downstream" synthon such as structure II is modified at its terminal 4' site. More specifically, the invention provides efficient syntheses of oligonucleosides via intermolecular reductive coupling on a solid support.
Bx can be nucleosidic bases selected from adenine, guanine, uracil, thymine, cytosine, 2-aminoadenosine or 5- methylcytosine, although other non-naturally occurring species can be employed to provide stable duplex or triplex formation with, for example, DNA. Representative bases are disclosed in U.S. Patent No. 3,687,808 (Merigan, et al . ) , which is incorporated herein by reference.
Q can be S, CH2, CHF CF2 or, preferably, 0. See, e . g. , Bellon, et al . , Nucleic Acids Res . 1993, 21 , 1587.
X can be H, OH, alkyl, substituted alkyl, alkaryl or aralkyl, F, Cl, Br, CN, CF3, 0CF3, OCN, O-alkyl, S-alkyl, N-alkyl, O-alkenyl, S-alkenyl, N-alkenyl, SOCH3, S02CH3, ON02, N02, N3, NH2, heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, polyalkylamino or substituted silyl, an RNA cleaving group, a group for improving the pharmacokinetic properties of an oligonucleotide, or a group for improving the pharmacodynamic properties of an oligonucleotide. It is intended that the term "alkyl" denote branched and straight chain hydrocarbyl residues, including alkyl groups having one or more 3H and/or 1 C atoms. It is preferred that X is H or OH, or, alternatively F, O-alkyl or O-alkenyl, especially where Q is 0. Preferred alkyl and alkenyl groups have from 1 to about 10 carbon atoms.
Solid supports ( e . g. , (P) ) according to the invention include any of those known in the art for polynucleotide synthesis, including 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 Letters 1993, 34 , 3373) or Poros -- a copolymer of polystyrene/divinylbenzene. Attachment and cleavage of nucleosides and oligonucleosides can be effected via standard procedures. See, e . g. , Pon, R.T. in Protocols for Oligonucleotides and Analogs, Agrawal, S., ed. , Humana Press, Totowa, NJ, 1993. As used herein, the term solid support further includes any linkers ( e . g. , long chain alkyl amines and succinyl residues) used to bind a growing oligonucleoside to a stationary phase such as CPG.
Y can be OH, ORHP, CH20H, or CH20RHP where RHP is a hydroxyl protecting group. A wide variety of hydroxyl protecting groups can be employed in the methods of the invention. See, e . g. , Beaucage, et al . , Tetrahedron 1992, 12 , 2223. In general, protecting groups render chemical functionality inert to specific reaction conditions, and can be appended to and removed from such functionality in a molecule without substantially damaging the remainder of the molecule. Representative hydroxyl protecting groups include phthalimido, t-butyldimethylsilyl (TBDMS) , t- butyldiphenylsilyl (TBDPS) , fluorenylmethoxycarbonyl (FMOC) , and other hydroxyl protecting groups.
It is preferred that the oligonucleotide analogs of the invention comprise from about 5 to about 50 subunits having the given structure (i.e., n = 5-50) . While each subunit of the oligonucleotide analogs can have repetitive structure I, such need not be the case. For example, the subunits can have alternating or more random structures.
The invention is also directed to methods for the preparation of oligonucleosides with modified inter-sugar linkages. These modifications may be effected using solid supports which may be manually manipulated or used in conjunction with a DNA synthesizer using methodology commonly known to those skilled in DNA synthesizer arts . Generally, the procedure involves functionalizing the sugar moieties of two nucleosides which will be adjacent to one another in the selected sequence. In a 5' to 3' sense, an "upstream" synthon such as structure II is modified at its terminal 3' site, while a "downstream" synthon such as structure III is modified at its terminal 5' site.
More specifically, certain linkages can be formed by selecting a 3'-C-formyl derivatized compound as the upstream synthon and a 5' -aminohydroxy derivatized compound as the downstream synthon. Coupling then is effected to provide, for example, a dinucleoside having an oxime linkage. In this instance, the oxime is present as E/Z isomers. The oxime nitrogen atom is adjacent to a carbon atom on the 3' end of the upstream nucleoside. Dinucleosides having the oxime nitrogen adjacent to a carbon atom on the 5' or downstream nucleoside are synthesized utilizing a 4'-C-formyl derivatized compound as the upstream synthon and a 3'-deoxy- 3' -aminohydroxymethyl derivatized compound as the downstream synthon, again providing E/Z isomers. In both instances the oxime linked compound can be incorporated directly into an oligomer and/or can be reduced to a corresponding hydroxylamino linked species. Reduction of oxime linked dinucleosides either as the dinucleoside or as a dinucleoside moiety in an oligomer with sodium cyanoborohydride yields the corresponding hydroxylamino linked compounds. Hydroxylamino linked compounds can be alkylated at the amino moiety of the hydroxylamino linkage to yield a corresponding N-alkylamino linkage.
3'-C-fσrmyl derivatized nucleosides can be formed via several synthetic pathways. The presently preferred method utilizes a stereoselective intermolecular radical C-C bond formation reaction, ( see, e . g. , U.S. Application Serial No. 08/040,903, filed March 31, 1993) . Thus, a 3'-0- phenylthiocarbonate ester derivative of thymidine reacted with jβ-tri-n-butylstannylstyrene and AIBN to provide a 3'-C- styryl derivative, which on oxidative cleavage gave a 3'-C- formyl thymidine derivative. In a similar manner, a radical carbonylation of the corresponding 3 ' -deoxy-3 ' -iodo nucleoside gives the 3'-C-formyl nucleotide analog. The iodo compound is treated with CO, 2,2' -azobisisobutrylonitrile (AIBN) , and tris (trimethylsilyl) silane (TTMS) . Alternately, 3'-C-formyl derivatized compounds can be synthesized from either a 3' -deoxy-3' -cyano sugar or nucleoside. Both 4'-C- formyl (also identified as 5'-aldehydo) and 3'-C-formyl group can be blocked in a facile manner utilizing O- methylaminobenzenthiol as a blocking group. The 4'- and 3'- C-formyl groups can be deblocked with silver nitrate oxidation.
An alternate method of 3'-C-formyl nucleoside synthesis employs l-O-methyl-3' -deoxy-3' -O-methylaminobenzene thiol-5' -O-trityl-S-D-erythro-pento furanoside, which serves as a precursor for any 3' -deoxy-3' -C-formyl nucleoside. The l-O-methyl-3 ' -deoxy-3' -O-methyl amino benzenethiol-5' -O- trityl-β-D-erythro-pentofuranoside is reacted with an appropriate base utilizing standard glycosylation conditions and then deblocked to yield the nucleoside. In yet another method, a 3' -deoxy-3' -cyano nucleoside is prepared from either the corresponding 3' -deoxy-3' -iodo nucleoside or by glycosylation with l-O-methyl-3' -deoxy-3' -O-cyano-5' -0- trityl-β-D-ervthro-pentofuranoside. Oligonucleosides according to the invention, linked by hydrazines, hydroxylamines and other linking groups, can be protected by a dimethoxytrityl group at the 5' -hydroxyl and activated for coupling at the 3'-hydroxyl with cyanoethyldiisopropyl-phosphite moieties. These compounds can be inserted into any desired sequence by standard, solid phase, automated DNA synthesis techniques. One of the most popular processes is the phosphoramidite technique ( see, e . g. , Beaucage, et al. , Tetrahedron 1992, 48, 2223 and references cited therein) , wherein a nucleoside or oligonucleotide having a free hydroxyl group is reacted with a protected 2' -cyanoethyl phosphoramidite monomer or oligomer in the presence of a weak acid to form a phosphite-linked structure. Oxidation of the phosphite linkage followed by hydrolysis of the cyanoethyl group yields the desired phosphate or phosphorothioate linkage. In one embodiment of the invention, protected oligonucleosides are linked with the units of a specified DNA sequence utilizing normal phosphodiester bonds. The resulting oligonucleotide analog or oligomer has a "mixed" backbone containing both phosphodiester linkages and four atom linkages of the inventions. In this manner, a sequence-specific 15-mer oligonucleotide can be synthesized to have seven hydroxylamine, hydrazine or other type linked dinucleosides attached via alternating phosphodiester linkages. Such a structure will provide increased solubility in water compared to fully modified oligomers, which may contain linkages of the invention.
Oligonucleosides containing a uniform backbone linkage can be synthesized by use of CPG-solid support and standard nucleic acid synthesizing machines such as Applied Biosystems Inc. 380B and 394 and Milligen/Biosearch 7500 and 8800s. The initial nucleoside (number 1 at the 3 '-terminus) is attached to a solid support such as controlled pore glass . In sequence specific order, each new nucleoside is attached either by manual manipulation or by the automated synthesizer system. One preferred method, shown in Figure 1, employs a solid support to which has been bound a downstream synthon having a protected 5' site. The 5' site preferably is protected with a phthalimido group. This permits the extent of nucleoside loading on the support to be determined by UV/visible spectrophotometry, as treatment with 3% N- methylhydrazine in methylene chloride yields 1,2-dihydro-4- hydroxy-2-methyl-l-oxaphthalazine (DHMO) , which has λmax = 304 nm and e= 6,000 1 cm"1 mol"1. In addition, emission spectroscopy has been utilized to determine the percent deblocking of the 5' -O-phthalimido group by measuring the release of DHMO. ( see, e . g. , Example 11). The 5' site of the downstream synthon can be liberated with any nucleophilic base having a pKa from about 7.7 to about 10.0. The selected base should not appreciably react with any linker used to bind a growing oligonucleoside to a stationary phase.
Representative bases include hydrazine and methylhydrazine. The 5' site preferably is liberated with 3% methylhydrazine in methylene chloride and washed with methylene chloride:methanol. The aminohydroxyl group at the 5' position of the upstream synthon also is protected with a phthalimido group to yield a 5' -phthalimido protected 3'- deoxy-3' -C-formyl nucleoside, which is reacted with the downstream synthon in, for example, 2.5% acetic acid in methylene chloride. Deprotection at the 5' position and washing liberates the next 5' -aminohydroxy reaction site. The cycle is repeated with the further addition of upstream synthon until the desired sequence is constructed. Each nucleoside of this sequence is connected with oxime linkages. The terminal nucleoside of the desired oligonucleoside preferably is added to the sequence as a 5'-OTBDMS blocked 3' -deoxy-3' -C-formyl nucleoside. The oxime linked oligonucleoside can be removed from the support with, for example, ammonium hydroxide. If an aminohydroxyl linked oligonucleoside is desired, the oxime linkages are reduced with sodium cyanoborohydride in acetic acid. Alternately reduction can be accomplished while the oxime linked oligonucleoside is still attached to the support. Free amino groups produced upon reduction can be alkylated with, for example, acetone and sodium cyanoborohydride in acetic acid. The alkylation step can be used to introduce other, useful, functional molecules on the macromolecule. Such useful functional molecules include but are not limited to reporter molecules, RNA cleaving groups, groups for improving the pharmacokinetic properties of an oligonucleotide, and groups for improving the pharmacodynamic properties of an oligonucleotide. Such molecules can be attached to or conjugated to the macromolecule via attachment to the nitrogen atom in the backbone linkage. Alternatively, such molecules can be attached to pendent groups extending from the 2' position of the sugar moiety of one or more of the nucleosides of the macromolecules. Examples of such other useful functional groups are provided by United States patent application serial number 782,374, filed Oct. 24, 1991, entitled Derivatized Oligonucleotides Having Improved Uptake & Other Properties, assigned to the same assignee as this application, herein incorporated by reference, and in other of the above-referenced patent applications.
In certain preferred compounds of the invention, pendent groups extending from the 2' position enhance binding or hybridization of the compound to a target nucleic acid. Dimeric pairs of compounds of the invention linked via MMI [methylene (methylimino) ] linkages were studies by NMR. While we do not wish to be bound by theory, the sugar moieties of RNA and DNA oligonucleotides normally are located in one of two major conformations, either the C2'-endo conformation (DNA like or southern conformation) or the C3'-endo conformation (RNA like or northern conformation) . While other conformations are possible, such as 04-endo, generally the greatest percentage of the sugar moieties of the nucleoside molecules will be in one or the other of the C3' - endo or C2'-endo conformation. In this study the two sugar moieties, the 3' sugar and the 5' sugar, in a dimer pair were analyzed as to the percentage of the sugar moiety located in either the C2'-endo conformation (DNA like or southern conformation) or the C3'-endo conformation (RNA like or northern conformation) . Again while we do not wish to be bound by theory, it has generally been found that for binding to RNA, the C3'-endo conformation is preferred since, in identical sequences, RNA-RNA binding is generally stronger than RNA-DNA binding which in turn is stronger than DNA-DNA binding. In this study, for T-T dimers linked by normal phosphodiester linkages 35% of the 5' sugar and 28% of the 3' sugar were determined by NMR to be in the C3'-endo conformation. Thus for the phosphodiester linkage, both of the 3' and the 5' sugar are very DNA like, existing in the C2'-endo conformation. In contrast, in a T-T dimer linked by MMI linkages of the invention, 68% of the 5' sugar and 31% of the 3' sugar were determined to be in the C3'-endo conformation. Again while we do not wish to be bound by theory, this increase in RNA character imparted by the MMI linkage might contribute to the +0.2 °C increase in Tm per modification for MMI oligomers as compared to normal phosphodiester oligonucleotides. When we substituted a 2'-0- methyl modified sugar for the 3' deoxy sugar in our MMI linked T-T dimers further RNA like characteristic were imparted to the dimers. Now 66% of the 5' sugar was in the C3'-endo conformation while 65% of the 3' sugar of the T-T2,.0. methyl dimer pair resided in the C3'-endo conformation. When a 2'-0-fluoro modified sugar was substituted for the 3' deoxy sugar of our MMI T-T dimer pair, the percent of C3'-endo for the 3' sugar increased to 92% while the 5' sugar increased to 70%. Thus in the T-T2,.p dimer pair, the 2'-modified sugar imparted even greater RNA like characteristics even though neither sugar moiety contained a RNA 2'-hydroxyl group.
These increasing RNA like characteristics are reflected in increased hybridization as illustrated by their Tms shown in Tables 3 and 4 of the Examples of this specification.
The compounds of this invention can be used in diagnostics, therapeutics, and as research reagents and kits. For therapeutic use the oligonucleotide analog is administered to an animal suffering from a disease modulated by some protein. It is preferred to administer to patients suspected of suffering from such a disease an amount of oligonucleotide analog that is effective to reduce the symptomology of that disease. One skilled in the art can determine optimum dosages and treatment schedules for such treatment regimens .
It is preferred that the RNA or DNA portion which is to be modulated be preselected to comprise that portion of DNA or RNA which codes for the protein whose formation or activity is to be modulated. The targeting portion of the composition to be employed is, thus, selected to be complementary to the preselected portion of DNA or RNA, that is to be an antisense oligonucleotide for that portion.
In accordance with one preferred embodiment of this invention, the compounds of the invention hybridize to HIV mRNA encoding the tat protein, or to the TAR region of HIV mRNA. In another preferred embodiment, the compounds mimic the secondary structure of the TAR region of HIV mRNA, and by doing so bind the tat protein. Other preferred compounds complementary sequences for herpes, papilloma and other viruses .
It is generally preferred to administer the therapeutic agents in accordance with this invention internally such as orally, intravenously, or intramuscularly. Other forms of administration, such as transdermally, topically, or intralesionally may also be useful. Inclusion in suppositories may also be useful. Use of pharmacologically acceptable carriers is also preferred for some embodiments . This invention is also directed to methods for the selective binding of RNA for research and diagnostic purposes. Such selective, strong binding is accomplished by interacting such RNA or DNA with compositions of the invention which are resistant to degradative nucleases and which hybridize more strongly and with greater fidelity than known oligonucleotides or oligonucleotide analogs. Additional objects, advantages, and novel features of this invention will become apparent to those skilled in the art upon examination of the following examples thereof, which are not intended to be limiting, wherein parts and percents are by weight unless otherwise indicated. EXAMPLE 1 Capping Of Free Amino Group Of Succinyl-CPG, 2
To a suspension of succinyl-CPG [1 , (10.0 g) , . prepared according to the procedure of Damha, et al . , Nucleic Acids Res . 1990, 18, 3813-21] , in glacial acetic acid (50 ml) , 20% formaldehyde in water (1.0 ml) was added. The resulting mixture was shaken for 15 minutes. Sodium cyanoborohydride (180 mg) was added in two portions with 5 minutes shaking between each addition. Addition of 20% formaldehyde in water followed by a double reduction with sodium cyanoborohydride was repeated once. The CPG beads were washed thoroughly with dichloromethane methanol (3 x 50 ml, 1:1, v/v) and cloudy solution was decanted. Finally, CPG beads 2 were filtered off and washed with ethyl ether and dried under vacuum for 6 hours.
EXAMPLE 2 CPG Loading
A. 5' -O-Phthalimido-3' -O- (succinyl-CPG-NMe2) - thymidine, 8. 5 * -O-Phthalimidothymidine (3./ 0.8 mmol.), dimethylaminopyridine (0.2 mmol.) , 2. (2.0 g) , triethylamine (160μl) and DEC (4.0 mmol.) were shaken together in dry pyridine (24 ml) for 66 hours. Pentachlorophenol (1.0 mmol) was added and the resulting mixture was shaken for 24 hours. The CPG beads were filtered off and washed thoroughly with pyridine (30 ml) , dichloromethane (30 ml) and ether (30 ml) . The CPG beads then were dried under vacuum for 2 hours . Then, the CPG beads were shaken with piperidine (10 ml) for 15 minutes, filtered off and washed with dichloromethane (30 ml) ether (30 ml) and dried under vacuum for 4 hours. The resulting beads were suspended in glacial acetic acid (10 ml) and sonicated for 30 minutes. The suspension was diluted with methanol dichloromethane (3 x 30 ml, 1:1 v/v) and cloudy solution was decanted. Then, the CPG beads were filtered off washed with dichloromethane (30 ml) and ether (30 ml) and dried under vacuum over P205 for 12 hours, to yield to 8.. The loading was 35 μmol of 3. per gram of CPG.
B. 5' -O-Phthalimido-3' -O- (suσcinyl-CPG-NMe2) -2' - deoxyadenosine, 9.
The procedure of Example 2A was repeated except that 5' -O-phthalimido-2' -deoxyadenosine 4. was used in place of 5' -O-phthalimidothymidine. The loading was 36 μmol of 4. per gram of CPG.
C. 5' -0-Phthalimido-3' -0- (succinyl-CPG-NMe2) -2'- isobutyryl-0-6-diphenylcarbamoyl-2' -deoxyguanosine, 10
The procedure of Example 2A was repeated except that 5' -O-phthalimido-N-2-isobutyryl-) -6-diphenylcarbamoyl- 2' -deoxyguanosine 5 was used in place of 5'-0- phthalimidothymidine. The loading was 38 μmol of 5. per gram of CPG.
D. 5'-0-Phthalimido-3'-0- (succinyl-CPG-NMe2) -N-4- protected-2' -deoxycytidine, 11
The procedure of Example 2A was repeated except that 5' -0-phthalimido-N-4-benzoyl-2' -deoxycytidine 6. was used in place of 5' -O-phthalimidothymidine. The loading was 32 μmol of 6. per gram of CPG. E. 5'-0-Phthalimido-3'-0- (succinyl-CPG-NMe2) -5- methyl-N-4-protected-2/ -deoxycytidine, 12
The procedure of Example 2A is repeated except that 5' -0-phthalimido-5-methyl-N-4-benzoyl-2' -deoxycytidine 7. is used in place of 5' -O-phthalimidothymidine. EXAMPLE 3
Synthesis of Homothymidylate 3' -De(oxyphosphinico) -3' - [methylene(methylimino) ] backbone, T12 MMI A. Step 1.
5' -O-Phthalimidothymidine-3' -O-succinyl-CPG (7., 28 mg, lμmol of nucleoside) was packed into a small column and connected on an ABI DNA synthesizer model 380B. Compound 7. was treated with a solution 3% N-methyl hydrazine in dichloromethane: methanol (9:1 v/v) for 120 seconds with a 300 second wait step to give 5' -0-amino-thymidine-3' -0- succinyl-CPG.
B. Step 2. The CPG was washed with dichloromethane for 240 seconds.
C. Step 3.
5' -O-Phthalimido-3' -C-aldehydo-3' -deoxythymidine 13. in a 0.1 M solution of dichloromethane mixed with a 5% glacial acetic acid in dichloromethane (1:1, v/v) was passed through the column for 15 seconds (20 eq.) with a 600 minute wait step.
D. Step 4.
The CPG was washed with dichloromethane for 150 seconds.
E. Step 5.
Steps 1-4 were repeated ten times.
F. Step 6.
The 5-terminal nucleotide, e . g. , 5'-t- butyldiphenylsilyl-3' -aldehyde-3' -deoxy-thymidine 18., was incorporated following step 3. The excess of 18 was removed by washing step 4.
G. Step 7.
The resulting CPG beads (with oligomer) were transferred into a small flask and 1.0 ml of glacial acetic acid was added. NaCNBH3 (15 mg) was added and the mixture was shaken and sonicated for .15 minutes. Formaldehyde (20% in water, lOOμl) was added, the mixture was sonicated for 15 minutes and NaCNBH3 (2 x 15mg) was added with 15 minutes sonication between each addition. The resulting beads were washed with methanol dichloromethane (1:1, v/v, 15 ml) . H. Step 8.
Oligomer was cleaved from the solid support by treatment with 30% ammonia for 2 hours at room temperature. The ammonia then was evaporated. I . Step 9 .
The dried residue was dissolved in tetrahydrofuran (THF) /dioxane (1.0 ml, 1:1 v/v) . Tetrabutylammonium fluoride (TBAF) solution (1.0 M) in THF (40μl) was added and the mixture was stirred for 16 hours. The resulting suspension was diluted with triethyl ammonium acetate (pH 7.0, 500 μl of 50 mM solution) and was concentrated under vacuum. j. Step iθ.
Purification of the oligomer of step 9 was effected by HPLC. A reverse phase Supelcosil LC-18 (5 μm, 5.0 x 0.46 cm) column and a precolumn (5 μm, 1.0 x 0.46 cm) were used for purifications. The optimal separation was achieved by elution (1 ml/minute) with ammonium acetate buffer (pH 5.9) mixed with 15% acetonitrile and increasing the polarity with 50% of acetonitrile in 25 minutes. The desired T12 oligomer was eluted (retention time = 28.8 minutes) with isocratic 1:1 acetonitrile and ammonium acetate. EXAMPLE 4
Synthesis Of Mixed Phosphodiester And MMI Linked Oligomer: 5'-TPT*T*T-3' [p = 3'-0-P(0)2-0-5'; . = 3'CH2-N (CH3)-0-5/]
CPG-loaded T.T oxime dimer was prepared as described in Example 3. Thus, twice repeating the steps 1-4 provided T.T oxime dimer. This dimer was further extended by repeating steps 1-4 using a 5'0-FMOC (9- fluorenylmethoxycarbonyl) protected nucleoside 22_ instead of nucleoside 12. during step 3. This modification provides a convenient way of switching to standard phosphoramidite chemistry without removing the CPG from automated DNA synthesizer. The resulting T.T.T oxime trimer, still attached to CPG, was treated with a solution of 5% piperidine in acetonitrile for two minutes with a flow rate of 1.6 ml/minute. A three minute waiting step and a three minute acetonitrile wash step following the piperidine treatment removed the 5'-0-FMOC group completely ( see, e . g. , Ma et al . Biopolymers 1989, 28, 965-993) .
The T*T.T oxime trimer bearing a free 5' -OH group now was ready for a standard phosphoramidite cycle of DNA synthesis. Thymidine was incorporated as the last residue utilizing a procedure generally in accordance with Oligonucleotide Synthesis, M.J. Gait, ed., IRL Press, Oxford, 1984, to furnish a tetramer. The two oxime linkages of the tetramer were reduced and methylated following
NaCNBH3/AcOH/HCHO treatments as described in step 7 of Example 3. The resulting tetramer was cleaved from the CPG following step 8 of Example 3. The crude tetramer was purified by HPLC in a similar protocol as described in step 10 of Example 7. The title compound was eluted (retention time = 26.4 minutes) over 30 minutes with an acetonitrile gradient of 5% to 25% mixed with buffer (triethylammonium acetate, pH 5.9) . The product was further characterized by mass spectra (calculated 1614.57; observed 1614.25) and capillary gel electrophoresis (CE; 10% polyacrylamide, 45 cm X 75 μm capillary, 30 KV/30°C) as clean material (single peak at 5.6 minutes retention time on CE) . EXAMPLE 5 Synthesis Of Mixed Phosphorothioate And MMI Linked Oligomer: 5'-TpsT.T.T-3' [ps = 3' -O-P(O) (S) -0-5' ; * = 3'-CH2- N(CH3) -O- 5']
T.T.T oxime trimer was prepared as described in Example 4. The last thymidine nucleoside residue was incorporate utilizing standard phosphoramidite chemistry, followed sulfurization with Beaucage reagent ( see, Beaucage et al . , Tetrahedron 1992, 48, 2223-2312) to provide a CPG attached tetramer. The CPG was taken off the DNA synthesizer and treated in a manner described in steps 7 and 8 of Example 3. The product was purified by reverse phase HPLC as described in step 10 of Example 3. The title compound was eluted (retention time = 24.5 and 25.6 minutes) over 30 minutes as a mixture of diastereoisomers (due to chiral P=S) with an acetonitrile gradient of 5% to 30% mixed with buffer (triethylammonium acetate, pH 5.9) . The product was further characterized by mass spectra and CE. EXAMPLE 6
Synthesis Of Chimeric Oligomers
A. 5' -MMI Capped phosphorothioate: T.T.Cme*TsCsGsCsTBGsGsTsGsAsGsTsTsTsC [. = 3' -CH2-N(CH3) -0-5' ; C e = 5-methylcytosine; s = 3' -0-P(0) (S) -0-5']
The 5'-0-trityl off phosphorothioate oligonucleotide (TsCsGsCsTsGsGsTsGsAsGsTsTsTsC) was synthesized on a DNA synthesizer in lμM scale on a CPG support utilizing standard phosphoramidite chemistry, (see, Zon in Protocols For Oligonucleotides and Analogs, pages 165-189, S. Agrawal, Humana Press, Totowa, NJ, 1993) . Next, coupling was performed with the 5' -O-phthalimido-3' -O-phosphoramidite derivative of thymidine 2$. via standard procedures to provide a protected 5'-0-amino end of the oligomer. The 5' -O-phthalimido group was deprotected generally in accordance with the procedure of step 1 of Example 3. The CPG was then washed with dichloromethane for 240 seconds. N-4-benzoyl-3' -deoxy-3' -C-formyl-5' -0- phtalimido-5-methylcytosine 7 was then coupled with the oligomer on the support following the procedure of step 3 of Example 3. The CPG was washed with dichloromethane for 150 seconds. Oxime coupling was continued one more time following steps 1-4 of Example 3 with thymidine derivative 13. Lastly, 5' -O-tert-butyl-diphenylsilyl protected thymidine 18 was incorporate utilizing steps 3-4 of Example 3. The resulting oligomer was cleaved from the support by treatment with ammonium hydroxide (2 hours, room temperature) and then heated at 55°C for 6 hours. The solution was concentrated and the residue dissolved in glacial acetic acid (2 ml) . The oxime linkages were then reduced with NaCNBH3 (15 mg) and methylated in the manner described in step 7 of Example 3.
The terminal TBDPS group was then removed by TBAF treatment as described in step 9 of Example 3. The crude, chimeric oligomer was then purified by HPLC in a manner described in step 10 of Example 3. The purified material had a retention time of 22 . 1 minutes . The chromatographic peak was broad due to a mixture of phosphorothioate diastereomers .
B . 3 ' - MMI Capped Phosphorothioate : GSTSTSCSTSCBGBCSTBGSGBTB-GSABGBT.T.T.C [s = 3 ' -0-P (=0) (s) -0- 5 ' ; * = 3 ' CH2 N (CH3) -0- 5 ' ]
Synthesis of a tetrameric τ*T*T*C building block with MMI internucleosidic linkages is achieved by the procedure described in Example 23 of Application PCT/US92/04294, filed May 21, 1993, utilizing solution phase stepwise coupling. This building block is protected with a 5'-0-DMT group and is attached to CPG via standard procedures (see, Pon, R.T. in Protocols for Oligonucleotides and Analogs, pages 465-496, Agrawal, S. ed., Humana Press, Totowa, NJ, 1993) . Thus, 5' -0-DMT-T*T*T*C-CPG is packed into a 1 μM column and attached to a DNA synthesizer. The synthesis of the phosphorothioate portion is accomplished by standard phosphoramidite chemistry protocol as described in Example 6. The resulting chimeric oligomer with a 3' -MMI tail/cap is then purified by HPLC. Alternatively, the procedure described in Example 5 can be employed to prepare chimeric oligomers of any desired length and base composition.
C. 3' - and 5' -MMI Capped Phosphorothioate: T.T.C*TsCBGBCBTBGB-GBTBGBAaGsT.T.T.C [. = 3' -CH2-N(CH3) -0-5' ; s = 3'-0-P(0) (S)0-5']
Synthesis of 3' - and 5' -end capped chimeric oligomers is accomplished by combination of the methods described in Examples 3 and 5. 5'-0-DMT blocked oligomer T*τ*T*C is synthesized by incorporating appropriate building blocks (compounds ϋ, 2J3 and 13.) following the protocol described in Example 3 (steps 1-6) . The 5'-0-DMT group is removed from the CPG-attached oligomer utilizing standard acidic deprotection. Next, a phosphorothioate chain of desired length is synthesized onto the oxime tetramer. The 5'-cap of the MMI tetramer is attached following the protocol of Example 6(A) to furnish the title compound. Deprotection of oligomer from the support, reductive methylation of the oxime linkages, and purification by HPLC as in Examples 6(A) and 6(B) provides the oligomer of choice. Alternatively, appropriately protected tetramers are prepared by solution phase MMI chemistry as per Example 23 of Application PCT/US92/04294. The tetramers are then conjugated sequentially following standard phosphoramidite procedures.
EXAMPLE 7
Synthesis Of Mixed Base MMI Oligomers: T.T, A*T, Cme*T, and
G.T A general procedure for the synthesis of mixed base
MMI linked oligomers has been developed to produce all sixteen pairs of possible dimers (X*Y; X and/or Y = T, dC, dA, dG) . The process consists of two steps on an automated DNA synthesizer ( e . g. , ABI 380 B) and two steps off the synthesizer.
A. Automated Reactions
1. Deprotection of the phthalimido group of 8-12 was carried out with a 3% solution of methyl hydrazine in dichloromethane and methanol (98:2, v/v) utilizing a 630 second step, followed by a dichloromethane wash step (180 seconds) .
2. Amine-aldehyde coupling was carried out with a 0.1 M solution of a bifunctional 3'-C-formyl nucleoside ( e . g. , compounds 13.-27.) in 20 molar equivalent excess dissolved in 25% acetic acid in dichloromethane using a 630 second step, followed by a dichloromethane washing step (130 second) . The excess of bifunctional nucleosides 13.-2J7 was recovered by simple concentration of the solutions.
B. Non-automated Reactions 1. The resulting oxime oligomers (attached to
CPG) were reduced and reductively methylated in manner generally in accordance with step 7 of Example 3.
2. The oligomers were cleaved from the solid support (CPG) by ammonia treatment (55°C, 5 hours) and the resulting solution was concentrated in vacuo . The residue was diluted with 300μl of water and the samples were analyzed by HPLC. A reverse phase C18 column (5 μm, 5.0 X 0.46 cm) and a short (1 cm) guard column were used with a flow-rate of
1 ml/minute. Elution with 0.05 M triethylammonium acetate
(pH 7.0) mixed with acetonitrile (10%→30% for 20 minutes, 30% acetonitrile isocratic for 10 minutes) furnished clean products having the following retention times.
Cme*T = 12.6 minutes
A*T = 15.2 minutes
T*T = 15.5 minutes
G*T = 12.9 minutes T*A = 11.8 minutes
G*A = 07.2 minutes
Each dimer bears a 5'-0-TBDPS group, which was left on the molecule to increase lipophilicity during HPLC analysis.
EXAMPLE 8 Synthesis Of 5' -O-Phthalimido-2' -deoxynucleoside Derivatives. A. 5' -O-Phthalimidothymidine, 3.
The 5' -O-phthalimidothymidine was prepared by the procedure described in the Example 6 of Application Serial No. 08/040,903. B. 2' -Deoxy-5' -O-phtalimidoadenosine, 4.
To a solution of deoxyadenosine (26.9 g, 100 mmol) in DMF (500 ml) were added N-hydroxyphthalimide (21.2 g, 130 mmol) and triphenylphosphine (34.1 g, 130 mmol) . The solution was cooled to 0°C and diisopropyl-azidocarboxylate (29.5 ml, 150 mmol) was then added dropwise. After addition, the solution was warmed to room temperature and stirred for two hours. DMF was removed under reduced pressure and the residual gum was triturated with ether (4 x 300ml) . The precipitate was recrystallized from ethanol in three crops (24.66 g, 62%. m.p. 140-142°C) . - R (DMSO-d6) δ 2.28-2.40 (m, IH, H2 ) , 2.76-2.90 (m, IH, H2" ) . 4.18-4.28 (m, IH, H4') , 4.41 (d, 5.1 Hz, 2H, H5' ,H5") , 4.51-4.61 (m, IH, H3 ' ) , 5.54 (d, 4.0 Hz, IH, OH3' ) , 6.38 (t, 6.9 Hz, IH, HI') , 7.27 (s, 2H, NH2) , 7.78-7.90 (m, 4H, ar-H) , 8.12 (s, IH, H2) , 8.33 (s, IH, H8) . 13C-NMR (DMSO-d6) δ 71.00 (C4') , 77.86 (C2') , 83.66 (C3') , 84.47 (Cl') , 119.14 (C5') , 123.17 and 134.71 (C4", C5", C6", C7") , 128.48 (C3",C8") , 139.48 (C8) , 149.05 (C4) , 152.59 (C2) , 156.03 (C5, C6), 162.98 (C2",C9") . Anal. : C18H16N605+l/2 CH3OH, Calcd. : C, 53.88; H, 4.40; N, 20.38; Found: C, 53.64; H, 4.21; N, 20.22. Mass: [M+H] + 397, [A+2H]+ 136.
C. 2'-Deoxy-06-diphenylcarbamoyl-N2-isobutyryl-5'-O- phthalimidoguanosine, 5.
To a solution of 2'deoxy-06-diphenylcarbamoyl-N2- isobutyryl-guanosine (10.31 g, 19.36 mmoles) in THF (155 ml) and DMF (62 ml) , triphenylphosphine (7.694 g, 29.04 mmoles) and N-hydroxyphthalimide (4.88 g) were added. Diisopropyl azodicarboxylate (6.2 ml) was added to the cooled (0°C) solution over a period of 5 minutes. On complete addition, the solution was warmed to room temperature and stirred for 5 hours. The solvents were evaporated under vacuum and the residue purified by silica gel column chromatography. Elution with dichloromethane/methanol (95:5, v/v) furnished the title compound (9.04 g, 69% yield) . XH-NMR (200 MHz, DMSO- & ) δ 10.64 (l,s,2-NH), 8.59 (1, s, H-8) , 7.76 (4, s,phthaloyl) , 7.5- 7.16 (10, m, N02) , 6.40 (l,t,l'-H), 5.54 (l,d,3'-OH) , 4.64 (l,m, 3'-H), 4.48 (2, m, 5'-H, 5"-H) , 4.21 (1, m, 4'-H), 3.33 (6, d, CH (CH3) 2) , 3.05 (l,m,2'-H), 2.68 (l,m, 2"-H) .
D. N4-Benzoyl-2' -deoxy-5' -O-phthalimidocytosine, 6
TBDP-silylation of N4-benzoyl-2' -deoxy-5' -0- dimethoxytritylcytosine was carried out generally in accordance with Example 7 of Application Serial No.
08/040,903 to furnish a 3'-TBDPSi protected nucleoside. The latter compound was treated with trichloroacetic acid to deprotect the dimethoxytrityl group and furnish N4-benzoyl- 3' -O-tert-butyldiphenylsilyl-2' -deoxycytosine. 1H-NMR (200MHz, CDC13) δ 8.67 (1,broad, H) , 8.16 (1, d, H-5) , 7.9- 7.15 (16,m,TBDP, Benzoyl, 6-H) , 6.27 (l,t, l'-H) , 4.44 (l,m,3'-H), 4.035(l,m,4'-H) , 3.67 (l,dd,5'-H) , 3.245 (l,dd,5"-H) , 2.65 (l,m,2'-H), 2.25 (l,m,2"-H) , 2.06 (1,broad, 5' -OH) , 1.1 (9,s,tBu) . Mitsunobu reaction of N4-benzoyl-3' -O-tert- butyldiphenylsilyl-2' -deoxycytosine with N-hydroxyphthalimide following the procedure described above furnished 81% yield of the desired N4-benzoyl-3' -O-tert-butyldiphenylsilyl-2' - deoxy-5' -O-phthalimidocytosine. MP = 175-177°C. Η- MR (200MHz, CDC13) δ 8.61(2,d, NH,5-H) , 7.91-7.26 (20, m, benzoyl,TBDP, phthal, 6-H) , 6.545 (1, m, l'-H) , 4.70 (l,m, 3'- H) , 4.17 (l,m, 3'H) , 4.035 (l,dd, 5'-H) , 3.67 (l,dd, 5"- H),2.815 (l,m,2'-H) , 1.95 (l,m, 2"-H) , 1.18 (9,s,tBu) .
Treatment of N4-benzoyl-3' -O-tert- butyldiphenylsilyl-2' -deoxy-5' -O-phthalimidocytosine with TBAF in THF furnished the title compound in 85% yield. EXAMPLE 9
Synthesis Of 2' ,3' -Dideoxy-3' -C-formyl-5' -O-phtalimido- nucleoside Derivatives.
A. 3' -Deoxy-3' -C-formyl-5' -O-phtalimidothymidine, 13 5' -O-tert-Butyldiphenylsilyl-3' -C-styrylthymidine was prepared according to the procedure described in Example 82 of Application Serial No. 08/040,903 and deblocked with TBAF to provide 3' -C-styrylthymidine as a white foam in 65% yield. Anal. : C18H21N204 Calcd.: C% 65.63; H% 6.42; N% 8.50; Found: C% 65.23; H% 6.25; N% 8.34.
Mitsunobu reaction of 3' -C-styrylthymidine with N- hydroxyphthalimide gave 97% yield of 5' -O-phtalimido-3' -C- styrylthymidine as a white solid. Anal.: C26H23N306 Calcd. : C% 65.95; H% 4.89; N% 8.87; Found: C% 65.69; H% 5.11; N%'8.59. Oxidative cleavage (Os04/NaI04) of 5' -O-phthalimido-
3' -C-styrylthymidine was carried out as in Example 82 of Application Serial No. 08/040,903 to furnish the title compound 13.
This procedure constitutes an alternate method for the preparation of bifunctional nucleosides such as compounds
13-17.
B. 2' ,3' -Dideoxy-3' -C-formyl-5' -O- phthalimidoadenosine, 14
2' -Deoxyadenosine was conveniently transformed into 5' -0-tert-butyldiphenylsilyl-2' ,3' -dideoxy-3' -C- styryladenosine in 60% yield utilizing the radical reaction described in Example 82 of Application Serial No. 08/040,903. The 5'-0-TBDPS group of the latter compound was deblocked to provide 2', 3' -dideoxy-3' -C-styryladenosine in 91% yield, mp 215-217°C. Anal.: C18H19N502 1/4 H20 Calcd. : C% 63.23; H% 5.74; N% 20.48; Found: C% 63.31; H% 5.68; N% 20.69. 2' , 3' -Dideoxy-3' -C-styryladenosine underwent standard Mitsunobu reaction with N-hydroxyphthalimide to provide 2' ,3' -dideoxy-5' -0-phthalimido-3' -C-styryladenosine in 76% yield with a melting point of 145-147°C. Anal. : C26H22N604-l/4 H20 Calcd. : C% 64.12; H% 4.65; N% 17.25; Found: C% 63.96; H% 4.58; N% 17.51.
Oxidative cleavage (0s04/NaI04) of the styryl group of the latter compound furnished the title compound in 55% yield as a white powder.
C. N4-Benzoyl-2' ,3' -dideoxy-3' -C-formyl-5' -0- phthalimido-5-methylcytosine, 1
5' -O-tert-Butyldiphenylsilyl-3' -C-styrylthymidine was converted into its 4-triazolo derivative generally according to the procedure of Li, et al . , Biochemistry 1987, 26, 1086. The product subsequently was treated with ammonia. Benzoylation of the resulting cytosine derivative provided N4-benzoyl-5' -0-tert-butyldisphenylsilyl-2' , 3 ' -dideoxy-3' -C- styryl-5' -methylcytosine (55% yield for three steps) . Deprotection of the latter product with TBAF and Mitsunobu reaction of the product furnished N4-benzoyl-2' , 3, -dideoxy- 5' -O-phthalimido-3' -C-styryl-5-methyl-cytosine in 45% yield. Melting point = 150-155°C. Anal. : C33H28N406 Calcd. : C% 68.74; H% 4.89; N% 9.71; Found: C% 68.33; H% 4.95; N% 9.57. This product then was treated with 0s04/NaI04 as described in Example 82 of Application Serial No. 08/040,903 to yield title compound JL7 in 57% yield as a white powder. Anal. :
C26H22N407-1.25 H20 Calcd. : C% 59.48; H% 4.70; N% 10.67; Found: C% 59.53; H% 4.83; N% 10.58.
D. 2' ,3' -Dideoxy-3' -C-formyl-N2-isobutyryl-5' -O- phthalimidoguanosine, 15 2' -Deoxyguanosine was converted into the title compound 15. utilizing following reactions: (i) N2- isobutyrylation generally according to R.A. Jones in Oligonucleotide Synthesis, pages 24-27, M.J. Gait, ed. IRL Press, Oxford, 1984; (ii) tert-butyldiphenylsilylation generally according to Nair, Org. Prep. Proc . Int . 1990, 22, 57; (iii) phenoxythiocarbonylation generally according to Robins, J". Am. Chem. Soc. 1983, 105, 4059; (iv) styrylation generally according to Example 82 of Application Serial No. 08/040,903; (v) desilylation by treatment with 1 M TBAF in THF; (vi) Mitsunobu reaction generally according to Example 6 of Application Serial No. 08/040,903; and (vii) oxidative cleavage generally according to Example 82 of Application Serial No. 08/040,903. ~~E NMR (CDC13) δ 1.23 (m, 6, ibu H) , 2.50 (m, 1, 2' H) , 2.60 (m, 1, 2"H), 2.87 (m, 1, CH-ibu) , 3.81 (m, 1, 5"CH2), 4.1-4.4 (m, 3, 3'H, 4'H, 5"CH2), 6.03 (m, 1, l'H) , 7.5-8.2 (m, 4, ArH) , 8.08 (s, 1, C8H) , 9.79 (s, 1, CHO) , 12. 24 (s, 1, NH) . EXAMPLE 10
Synthesis Of 2' ,3' -Dideoxy-5' -O-FMOC-3' -C-formylnucleosides. A. 3' -Deoxy-5' -O-FMOC-3' -C-formylthymidine, 23 3' -Deoxy-3 ' -C-styrylthymidine was protected with FMOC-C1 generally according to Balgobin, et al . , Nucleosides and Nucleotides 1987, 6, 461, to furnish 5' -O-FMOC-3' -deoxy- 3' -C-styrylthymidine in 96% yield as a yellow foam. Anal. : C33H30N2O6-l/2 H20 Calcd. : C% 70.82; H% 5.58; N% 5.00; Found: C% 70.53; H% 5.48; N% 4.98. Oxidative cleavage (Os04/NaI04) of the former compound gave title compound 2_3 in 57% yield. XH NMR (CDC13) δ 1.80 (s, 3, CH3) , 2.22, 2.71 (2m, 2, 2'CH2) , 3.15 (m,l,3' CH) , 4.22 (t, 1, CH FMOC) , 4.45 (s, 2, CH2 FMOC) , 4.55 (m, 2, 5' CH2), 4.62 (m, 1, 4'H) , 6.06 (t, 1, l'H) , 7.3-7.80 (m, 9, Ar H, C6H) , 8.3 (br s, 1, NH) , 9.75 (s, 1, CHO) .
In an analogous manner, 3 '-C-styryl nucleoside derivatives of 2' -deoxycytidine, 2' -deoxyadenosine, and 2'- deoxyguanosine are transformed into 3' -C-formyl-5' -O-FMOC- derivatives of 2' -deoxycytidine 2JL, 2' -deoxyadenosine 2_4, and 2' -deoxyguanosine .25., respectively. EXAMPLE 11
Deprotection Assay During Automated Synthesis Utilizing
Emission Spectra
Equipment and Materials: Luminescence Spectrophotometer: PERKIN ELMER LS50B
Solvent: Spectrophotometer grade MeOH Reagents: (i) 1,2-dihydro-4-hydroxy-2-methyl-l- oxσphthalazine (MW 176.175) ; (ii) N-methyl hydrazine (MW 46.07, d 0.866) ; (iii) N-hydroxyphthalimide (MW 163.13) Synthesis: 1,2-Dihydro-4-hydroxy-2-methyl-l- oxophthalazine
N-Hydroxyphthalimide (10 g, 59.5 mmoles) was dissolved in anhydrous CH2C12 (300 ml) and N-methyl hydrazine (9.7 ml) was added to the stirred solution. The solution turned red and then yellow. When all the starting material had disappeared, the solvent was evaporated and the resulting oil was crystallized from anhydrous ethanol as fine needles. Analysis Protocol A. Neutral conditions The spectrometer was turned on 0.5 hour prior to the measurement . A blank run was performed using spectrophotometer grade methanol. 1,2-Dihydro-4-hydroxy-2- methyl-1-oxophthalazine (0.234 mg) was then dissolved in methanol (100 ml) and the sample analyzed on the spectrophotometer. The excitation wavelength was chosen at 298 nm. and the corresponding emission was observed at 400 nm. The following concentrations (μMol) of 1,2-Dihydro-4- hydroxy-2-methyl-1-oxophthalazine were tested:
Figure imgf000034_0001
A linear plot was obtained using the equation :
Emission = [21 . 0 x 10s] [Concentration (mol/L) ] B. Basic conditions
The procedure of Example 11 (A) was repeated except that the oxophthalazine sample was basified by addition of N- methyl hydrazine (1% volume) . Basification results in the decay of emission intensity by 25 units and moves the excitation wavelength to 485 nm.
C. Acidic conditions.
The procedure of Example 11 (A) was repeated except that the oxophthalazine sample was acidified with glacial acetic acid (0.1 ml) . The intensities of the emission was enhanced considerably and the values are summarized below.
Figure imgf000035_0001
A graphical representation of this data provided a linear plot using the following equation:
Intensity = [459 x 106] [Concentration (mol/L) ] As can be seen, sensitivity of detection was enhanced dramatically under acidic conditions. The foregoing method is very effective for the determination of low concentrations of 1, 2-Dihydro-4-hydroxy-2-methyl-1- oxophthalazine during automated synthesis .
EXAMPLE 12 Solution Phase/Block Synthesis Of Chimeric Oligomers I. General Procedures:
A. Coupling Of An O-Amino Nucleoside To An Aldehydo Nucleoside.
An equimolar mixture of 5' -O-amino nucleoside and 3'- deoxy-3' -C-formyl nucleoside were dissolved in 1:1 toluene/CH2Cl2 containing several drops of acetic acid, mixed at 35 °C on a rotary evaporator at atmospheric pressure for 1 hour, then concentrated to a foam under reduced pressure. This process was repeated until TLC analysis (5% MeOH/CH2Cl2) showed the reaction to be complete (3-6 times) , with the intermediate oxime evident as the only major spot. For MMI dimers in which one of the starting nucleosides bears an amide protecting group on the base (for example i^-benzoyl-S- methylcytosine) , the oxime was stirred overnight with methanolic ammonia, and the resulting solution concentrated to provide the crude product. The so obtained intermediate, base unprotected oxime dimer was dissolved in glacial acetic acid (0.1 M) , placed in an ice bath, and sodium cyanoborohydride (3 x 2 eq) was added over 15 minutes. The mixture was allowed to warm to room temperature over 1 hour, placed in an ice bath, and aqueous formaldehyde (20 eq, 20%) was added in one portion. Sodium cyanoborohydride (3 x 2 eq) was added over 30 minutes, and the mixture was allowed to warm to room temperature over 1 hour. The reaction was terminated by pouring into ice water (5 times volume) with vigorous stirring. The resulting residue was extracted into CH2C12, concentrated, azeotroped (3 times) with toluene, and chromatographed. The column was eluted with 1 to 5% EtOH (95%) /EtOAc, which removed several trace impurities off the column, and the desired product was then obtained by elution with 10% MeOH/CH2Cl2, and concentration of the appropriate fractions.
B. A^-Benzoylation of Cytosine-containing MMI Dimers. The dimer (1 eq) was dissolved in dry DMF, benzoic anhydride (1.1 eq) was added in one portion, and the reaction stirred at room temperature overnight. The solvent was removed, the residue partitioned between 5% aqueous sodium bicarbonate and CH2C12, the organic layer was washed with water, dried over magnesium sulfate, concentrated in vacuo, and chromatographed to afford pure product.
C. Desilylation of MMI Dimers.
The silylated MMI dimer was dissolved in dry THF (0.1 M) and cooled in an ice bath. A solution of tetrabutylammonium fluoride in THF (1 M, 1.5 eq per silyl group) was added dropwise over 5 minutes. The solution was stirred at 0 °C until the reaction was complete as judged by TLC (1-2 hours) , at which point silica (5 g/mmol) was added, and the mixture carefully concentrated. The silica was applied to a column packed in 5% MeOH/CH2Cl2, and eluted with 5% to 10% MeOH/CH2Cl2. The appropriate fractions were combined, concentrated, and the residue azeotroped with EtOAc to provide the silylated product .
D. Dimethoxytritylation of MMI Dimers.
The appropriate dimer (1 eq) and N, N-dimethyl- aminopyridine (0.1 eq) was azeotroped 3 times with dry pyridine, then dissolved in the minimum amount of dry pyridine. Triethylamine (2 eq) and 4,4' -dimethoxytrityl chloride were added to the stirred solution at room temperature, and the mixture diluted with CH2C12 (ca 5 mL/mL pyridine) . Stirring was continued until the reaction was complete (typically overnight) as judged by TLC (10%
MeOH/CH2Cl2 + 0.1% Et3N) , methanol was added to quench the reaction, and the reaction mixture extracted with 5% aqueous sodium bicarbonate. The organic layer was dried (trace magnesium sulfate) , filtered, and concentrated to afford a syrup, which was chromatographed on silica gel (CH2C12 + 0.1% Et3N to 10% MeOH/CH2Cl2 +.0.1% Et3N) . Fractions containing only product were combined and concentrated, and azeotroped with EtOAc to afford the 5' -dimethoxytritylated intermediate as a hard foam. This material was judged to be pure by XH NMR, and used without further purification.
E. Phosphitylation of MMI Dimer.
The appropriate 5' -O-dimethoxytrityl derivative was azeotroped with dry acetonitrile 3 times, then dissolved in dry dichloromethane (0.1 M) at room temperature. Diisopropylammonium tetrazolide (0.5 eq) and 2-cyanoethyl- N,_Z\T,_N' /_N' -tetraisopropylphosphorodiamidite (1.2 eq) was added, and the reaction allowed to stir at room temperature until complete (typically overnight) as judged by TLC (EtOAc + 0.1% Et3N) . The reaction mixture was then directly loaded onto a column packed with 25% EtOAc/hexane + 0.1% Et3N, and eluted with a stepwise gradient to EtOAc + 0.1% Et3N. Fractions containing only the product were pooled and concentrated to yield a hard foam, which was lyophilized from dry 1,4-dioxane to afford the phosphoramidite as a fine white powder.
II. Synthesis of 5' -0-DMT-TTBlMβCBzT-0-phosphoramidite-3' MMI tetramer.
5' -O-TBDPS-3' -C-formyl-T*T oxime dimer.
5' -O-TBDPS-3' -deoxy-3' -C-formyl-thymidine (prepared according to Sanghvi et al . Synthesis 1994, 1163) (0.229 mmole, leq.) and 5' -O-amino-3' -deoxy-3 ' -C-styryl-thymidine (prepared according to the procedure in Example 9 above) (0.229 mmole, leq.) were coupled according to the general procedure described above. The crude oxime dimer was then evaporated and dissolved in (1/1 : v/v) mixture of water and 1,4-dioxane with NaI04 (0.503 mmole, 2.2 eq, ) osmium tetroxide (4% solution in water, 0.229 mmole, O.leq.), 4-methyl-morpholine-N-oxide (0.332 mmole, 1.45 eq.) before being stirred in the dark for 4 hours. After work-up (same as described in the example 9 above) , the residue was chromatographed on silica gel column with 5% MeOH/CH2Cl2 to yield 36% (0.082 mmole) of a white foam. Rf = 0.33 (10% MeOH/CH2Cl2, MS (Electrospray) , M+H = 744.5, -NMR (DMSO-d6) δ 11.30 (ss, 2, NH) , 9.64 (br s, 1, CHO) , 6.10 (m, 2, l'H) , 3.60- 4.52 (m, 8, H3', H4' , H5' , H5' ' ) , 1.76 (s, 3, CH3, thymine) , 1.55 (s, 3, CH3, thymine) .
5'-0-Amino-3'-0-TBDPS-CBz*T oxime dimer. 5' -O-phthalimido-3' -deoxy-3' -C-formyl-5-methyl-i^-benzoyl- cytidine (0.150 mmole, leq.) and 5' -O-amino-3' -O-TBDPS- thymidine (0.15 mmole, leq.) in CH2C12 were coupled following the general procedure described above. The crude oxime dimer was then concentrated under vacuo, neutralized with a 5% aqueous solution of sodium bicarbonate (50 ml) extracted with CH2C12, washed with water and brine (50 ml each) dried over sodium sulfate and finally evaporated to dryness. TLC of the reaction mixture revealed the presence of a single product (Rf = 0.65 10% MeOH/CH2Cl2) . Without further purification, this compound was dissolved in CH2C12 (5 ml) and N-methyl hydrazine (0.225 mmole, 1.5 eq.) was added at room temperature. After 4 hours stirring, the white precipitate was filtered off and the filtrate concentrated and chromatographed on silica gel column. Elution with a mixture of 0.5% to 5% MeOH/CH2Cl2 to yield 51 of a white solid. Rf = 0.55 (10% MeOH/CH2Cl2) , MS (Electr spray), M+H = 850, , M+Na = 872, 'H-NMR (DMSO-d6) , 11.34 (s, 2, NH) , 6.30 (pt, 3, NH2 + l'H) , 6.05 (pt, 1, l'H, thymidine) , 3.57-4.45 (m, 8, H3' , H4' , H5', H5' ' ) , 2.06 (s, 3, CH3, C-5 CH3) , 1.74 (s, 3, CH3, thymine) .
5'-0-TBDPS-3'-0-TBDPS-T*T*CBl*T MMI tetramer.
An equimolar mixture of 5' -O-TBDPS-3' -C-formyl-T*T oxime dimer and 5' -O-amino-3' -0-TBDPS-MeCBz*T oxime dimer (0.153 mmole, 1 eq.) were coupled following the general procedure described above. The MMI tetramer was purified by silica gel column chromatography using 3% MeOH/CH2Cl2 to yield 48% over four synthetic steps (oxime coupling, debenzoylation, reductive methylation and rebenzoylation) of the desired compound. Rf = 0.40 (10% MeOH/CH2Cl2) , MS (Electrospray) , M+H = 1622.9, , M+Na = 1644.3, 'H-NMR (DMSO-d6) δ 11.32 (m, 4, NH) , 6.27 (pt, 1, l'H, 5 methyl cytidine) , 6.05 (m, 3, l'H, thymidine), 2.02 (s, 3, CH3, 5 methyl cytidine), 1.73 (d, 6, CH3, thymine) , 1.51, (s, 3H, CH3, thymine) .
5'-HO-T*T*CBz*T-OH-3' MMI tetramer.
5' -0-TBDPS-T*T*MeCBz*T-0-TBDPS-3' MMI tetramer (0.070 mmole, 1 eq.) was desilylated according the general procedure described above. Deblocked MMI tetramer was purified by silica gel column chromatography. Elution with 5% MeOH/CH2Cl2, pooling and evaporation furnished 80% of the title compound. Rf = 0.56
(15% MeOH/CH2Cl2) , MS (Electrospray) , M+H = 1146.2, M+Na = 1168.
5'-0-DMTr-T*T*CBz*T-OH-3' MMI tetramer.
5'-HO-T*T* eCBz*T-OH-3' MMI tetramer (0.070 mmole, leq.) was dimethoxytritylated according the general procedure described above. Protected MMI tetramer was purified by silica gel column chromatography. Elution with 5% MeOH/CH2Cl2, pooling and evaporation furnished 81% of the title compound. Rf = 0.47
(12% MeOH/CH2Cl2) , MS (Electrospray) , M+H = 1448, M+Na = 1469.7. 5'-0-DMTr-T*T*CBz*T-0-phosphoramidite-3' MMI tetramer.
5'-0-DMTr-T*T*MeCBz*T-OH-3' MMI tetramer (0.045 mmole, 1 eq.) was phosphitylated according the general procedure described above. MMI tetramer was eluted from the silica gel column chromatography with 5% MeOH/CH2Cl2 to yield 85% of the desired compound. Rf = 0.53 (10% MeOH/CH2Cl2) , 31P NMR (CD3CN) δ 148.55, 148.44.
Ill : Synthesis Of 5'-0-DMT- T*T*T*CBz-OH-3' MMI tetramer. 5' -0-amino-T*BzC-0-TBDPS-3' oxime dimer. 5' -O-Phtalimido-3' -deoxy-3' -C-formyl-thymidine (0.450 mmole, 1 eq.) and 5' -O-amino-3' -O-TBDPS-i^-benzoyl-cytidine
(0.45 mmole, 1 eq.) in CH2C12 were coupled following the general procedure described above. The crude product was then concentrated under vacuo, neutralized with a 5% aqueous solution of sodium bicarbonate (50 ml) extracted with CH2C12, washed with water and brine (50 ml each) dried over sodium sulfate and finally evaporated to dryness. TLC revealed the presence of a single product (Rf = 0.65 10% MeOH /CH2Cl2) . Without further purification, this compound was dissolved in CH2C12 (15 mL) and N-methylhydrazine (0.675 mmole, 1.5 eq.) was added at room temperature. After 4 hours of stirring, the white precipitate was filtered off and the filtrate concentrated and chromatographed on silica gel column with a mixture from 0.5% to 5% MeOH/CH2Cl2 to yield 65% of a white solid. Rf = 0.48 (10% MeOH/ CH2C12) , MS (Electrospray) , M+H = 836, M+Na = 858, 'H-NMR (DMSO-d6) δ 11.28 (br s, 2, NH) , 6.22 (pt, 1, NH2 + l'H, 5- methylcytidine) , 6.05 (pt, 1, l'H, thymidine) , 3.57-4.45 (m, 8H, H3', H4', H5', H5" ) , 1.81 (s, 3, CHS, thymine) . 5'-0-TBDPS-T*T*T*CBz-0-TBDPS-3' MMI tetramer. An equimolar mixture of 5' -O-TBDPS-3' -C-formyl-T*T oxime dimer and 5' -0-amino-T*BzC-0-TBDPS-3' oxime dimer (0.343 mmole, 1 eq.) in CH2C12 were coupled following the general procedure described above. The MMI tetramer was purified by silica gel column chromatography using 3% MeOH/CH2Cl2 to yield 56% over four synthetic steps (oxime coupling, debenzoylation, reductive methylation and rebenzoylation) of the title compound. Rf = 0.35 (10% MeOH/CH2Cl2) , MS (Electrospray), M+H = 1607.9, , M+Na = 1630.2, 'H-NMR (DMSO-d6) δ 8.6, 8.9, 9.3 (m, 4, NH) , 6.35 (pt, 1, HI', cytidine) , 6.05 (m, 3, HI', thymidine) , 5.95 (m, 2H, HI', thymidine), 1.88 (d, 6H, CH3, thymidine) , 1.61 (s, 3H, CH3, thymine) . 5'-OH-T*T*T*CBz-OH-3' MMI tetramer.
5'-0-TBDPS-T*T*T*CBz-0-TBDPS-3' MMI tetramer (0.143 mmole, 1 eq.) was desilylated following the general procedure described above. MMI tetramer was purified by silica gel column chromatography using 5% MeOH/CH2Cl2 to yield 80% of the title compound. Rf = 0.34 (15% MeOH/CH2Cl2) , MS (Electrospray) , M+H = 1131.0; M+NH4 = 1167.3.
5'-0-DMTr-T*T*T*CBz-OH-3' .
5' -OH-T*T*T*CBz-OH-3' MMI tetramer (0.113 mmole, leq.) was dimethoxytritylated following the general procedure described above. MMI tetramer was purified by silica gel column chromatography. Elution with 5% MeOH/CH2Cl2, pooling and evaporation furnished 81% yield of the desired compound. Rf = 0.33 (12% MeOH/CH2Cl2) ; MS (Electrospray) M+H = 1435.6; M+Na = 1456.8; 'H NMR (DMS0-d6) δ 9.5 (m, 4, NH) , 6.15 (pt, 1, HI', cytidine) , 6.01 (m, 3, HI', thymidine) .
IV. Synthesis of Oligomers Incorporating MMI Tetramers.
Tetameric MMI containing oligonucleotides correspond to chimeric oligonucleotides where several phosphodiesters and/or phosphorothioates linkages have been replaced by at least three consecutive methylene (methyl) imino (MMI) internucleosidic link.
MMI tetramer (or longer chain) containing chimeric oligonucleotides can be - synthesized using standard phosphoramidite chemistry. In this case the chimeric oligomers were synthesized attached to CPG (Controlled Pore Glass) solid support and the assembling was performed on an automated DNA synthesizer such as Millipore Expedite or Applied Biosystem 380B.
The MMI tetramers synthesized as phosphoramidite elongation units (DMT-X*X*X*X-phosphoramidite, X = T, C, 5MeC, * = 3' -CH2-N(CH3) -0-5' or X*X*X*X MMI ) were prepared according the procedure described above. The initial deoxynucleoside or MMI tetramer determining the 3'-end of the MMI containing chimeric oligonucleotide was loaded on to the CPG solid support via a succinyl linkage following the procedure described in Example 2 above (for example: 5' -0-DMTr-T*T*T*C-0- (succinyl-CPG-NMe2) -3' MMI was loaded onto the CPG providing a loading of 22.67 mmole/g) . The chain elongation of chimeric oligomers was performed in a similar manner as described for the MMI phosphoramidite Dimers. Table 1 : Chimeric Oligomers Containing T*T*T*C and τ*T*C*T MMI Tetramers.
Figure imgf000042_0001
Analytical Data
Figure imgf000042_0002
Column Supelco LC 18 15cm x 4.6mm, 5mm, 5% to 25% CH3CN in 0.05M TEAA, pH 7.00 over 25 minutes, 1ml/min.
EXAMPLE 13
Synthesis of New Monomeric Nucleosides
5' -O-Phthalimido-2' -fluorothymidine.
2' -Fluorothymidine was prepared according to the known procedure (Kawasaki et al . J. Med. Chem. 1993, 36, 83-1) . To this material (1.95 g, 7.5 mmol) and N-hydroxyphthalimide (1.29 g, 7.88 mmol) in dry DMF (60 mL) was added diethyl azodicarboxylate (1.24 mL, 7.88 mol) dropwise at room temperature of 0.5 hour. The mixture was stirred overnight and poured into a rapidly stirred ice cold mixture of ether (150 mL) and water (150 mL) . The aqueous layer was slowly diluted to a total volume of 350 mL with cold water, at whic point solid began forming. The ether was decanted, an additional portion ether was added, stirred, and decanted, and the process repeated. The solution was now diluted to a total volume of 500 mL with water, and allowed to stand on ice for several hours. The solid was collected, and dried t afford 2.11 g (70%) of a white powder: Rf 0.31 (5% MeOH/CH2Cl2) ; Η NMR (CDC13) δ 8.84 (s, IH, NH) , 7.90-7.70 (m, 4H) , 7.26 (s, IH, H-6) , 6.03 (dd, J=3.0, 17.0 Hz, IH, H-l') , 5.11 (ddd, J=1.5, 3.0, 52.8 Hz, IH, H-2') , 4.75 (m, IH) , 4.56 (m, 2H) 4.34 (m, IH) , 3.26 (br s, IH) , 1.94 (s, 3H, CH3) . Anal. Calcd for C18H16N307F«l/4Et20: C, 53.84; H, 4.40; N, 9.91. Found: C, 53.52; H, 4.60; N, 9.96. 5' -O-Amino-2' -fluorothymidine.
5' -O- hthalimido-2' -fluorothymidine (1.2 g, 3 mmol) was dissolved in 30 mL of 10% MeOH/CH2Cl2 with slight warming, then cooled on ice. When the reaction mixture became cloudy, methylhydrazine (0.24 mL, 4.5 mmol) was added dropwise. The clear reaction mixture was stirred for 1.5 hours at 0 °C, then applied directly onto a column packed with 10% MeOH/CH2Cl2. Elution with the same solvent, concentration of the appropriate fractions, and recrystallization of the residue from EtOH provided 0.54 g (65%) of needles: mp 168- 169 °C; Rf 0.32 (10% MeOH/CH2Cl2) ; Η NMR (DMSO-de) δ 11.43
(s, IH, NH) , 7.55 (s, IH, H-6) , 6.23 (s, 2H, ONH2) , 5.89 (dd, J=2.0, 19.5 Hz, IH, H-l') , 5.69 (d, IH, 3'-OH) , 5.09 (ddd, J=2.0, 2.5, 53.3 Hz, IH, H-2') , 4.25-3.65 (m, 4H) , 1.80 (s, 3H, CH3) . Anal. Calcd for C10H14N3O5F: C, 43.64; H, 5.13; N, 15.27. Found: C, 44.00; H, 5.01; N, 15.06. 5' -O-P thalimidouridine.
To a stirred solution of uridine (4.88 g, 20 mmol) , triphenylphosphine (5.76 g, 22 mmol) , and N- hydroxyphthalimide (3.58 g, 22 mmol) was added dropwise diethyl azodicarbohydrate (4.35 g, 25 mmol) over a period of 3 hours at room temperature. The reaction mixture was stirred at room temperature for 12 hours . The solvent was removed under reduced pressure and the residue suspended in CH2C12 (300 mL) . The suspension was poured onto ice water (250 mL) and stirred vigorously to provide pale yellow precipitation. The solids were filtered and washed with water (3 x 50 mL) and ether (3 x 50 mL) . The powder was dried under vacuum to provide 3.03 g (39%) of the title compound. 'H NMR (DMSO-d 6> δ 4.05-4.17 (m, 3, 2', 3', 4' H) , 4.38 (m, 2, 5' CH2) , 5.36 and 5.55 (2d, 2, 2', 3' OH) , 5.62 (d, 1, C5H) , 5.78 (d, 1, l'H), 7.77 (d, 1, C6H) , 7.90 (m, ArH), 11.37 (br s, 1, NH) . 5' -O-Phthalimido-5-methyluridine.
To a stirred mixture of 5-methyluridine (51.64 g, 0.2 mol) , triphenylphosphine (57.64 g, 0.22 mol) and N- hydroxyphthalimide (35.86 g, 0.22 mol) was added dropwise diethyl azodicarboxylate (43.5 g, 0.25 mol) over a period of 4 hours at =5 °C. After complete addition, reaction mixture was warmed to room temperature and stirred for 16 hours. The reaction was =50% (TLC, CH2Cl2:MeOH, 9:1, v/v) complete at this point of time. Therefore, additional quantities of triphenylphosphine (28.8 g, 0.11 mol), N-hydroxyphthalimide (17.9 g, 0.11 mol) and diethyl azodicarboxylate (21.7 g, 0.12 mol) were added in the manner described above. After second addition, the reaction mixture was stirred at room temperature for 16 hours. The reaction was =90% (TLC) complete at this time, therefore, solution was concentrated under vacuum to provide a thick syrupy residue. The residue was poured into a vigorously stirred mixture of ether:ice water (500 mL:1000 ml) to precipitate the product. The precipitate was filtered, washed with water (3 x 100 mL) and ether (3 x 50 ml) , and dried over P205 to provide 56 g (69%) of the title compound. An analytical sample was crystallized from EtOH:CH2Cl2, mp 236-237 °C; Η NMR (DMSO-d6) δ 1.78 (s, 3, CH3) , 4.12 (m, 2', 3', 4' H) , 4.36 (m, 2, 5'CH2) , 5.45 and 5.62 (2d, 2, 3', 4' OH), 7.78 (s, 1, C6H) , 7.90 (m, 4, ArH) , 11.32 (brs, 1, NH) ; Anal. Calcd for C18H17N308• 1/4 H20: C, 53.00; H, 4.32; N, 10.30; Found: C, 53.04; H, 4.28; N, 10.19. 2' -O-Methyl-5' -O-phthalimidouridine.
A stirred mixture of 5' -O-phthalimidouridine (1.94 g, 5 mmol) , dibutyltin oxide (1.48 g, 6 mmol) , tetrabutylammonium iodide (2.02 g, 5.5 mmol) and methyliodine (2.82 g, 20 mmol) in DMF (10 ml) was heated in a sealed flack at 50 °C for 16 hours under argon atmosphere. The reaction mixture was cooled (=10 °C) and to this another addition of methyl iodide (2.82 g, 20 mmol) was made. The flask was sealed and heating continued for 16 hours. The reaction mixture was cooled to room temperature and diluted with CH2C12 (50 ml) . The CH2C12 suspension was transferred onto the top of a prepacked silica gel (CH2C12) column. Elution with CH2C12/CH2C12:MeOH (9:1, v/v) furnished the desired product as homogenous material . Appropriate fractions were pooled and concentrated to provide 0.6 g (29%) of the title compound (contaminated with 20% of the 3'-0-methyl isomer) and 0.61 g (31%) of the unreacted starting material. An analytically pure sample was obtained by fractional crystallization from EtOH:Et20; mp 225-228 °C; Η NMR (DMSO-d6) δ 3.35 (s, 3, OCH3) , 3.83, 4.15, and 4.22 (3 m, 3, 2', 3', 4' H) , 4.40 (m, 2, 5' CH2) , 5.42 (d, 1, 3' OH) , 5.61 (d, 1, C5H) , 5.84 (d, 1, 1' H) , 7.78 (d, 1, C6H) , 7.84 (m, 4, ArH) and 11.4 (br s, 1, NH) ; Anal. Calcd for C18H17N308: C, 53.60; H, 4.24; N, 10.41; Found: C, 53.25; H, 4.34; N, 10.20. 2' -O-Methyl-5' -O-phthalimido-5-methyluridine.
The title compound was synthesized in 61% yield (contaminated with =10% of 3'-0-methyl derivative) starting from 5' -O-phthalimido-5-methyluridine following the foregoing procedure. An analytical sample was obtained by crystallization (EtOH/CH2Cl2) mp 213-214 °C; Η NMR (DMSO-d6) δ 1.71 (s, 3, CH3) , 3.58 (s, 3, OCH3) , 4.42 (m, 2, 5' CH2) , 4.63 (m, 1, 4'H) , 5.36 (m, 1, 2' H) , 5.61 (m, 1, 3' H) , 5.90 (s, 1, 1' H) , 7.50 (s, 1, C6H) , 7.82 (m, 4, ArH) , 11.55 (br s, t, NH) . 3' -O-tert-Butyldiphenylsilyl-2' -O-methyl-5' -O- phthalimido-5-methyluridine.
[Note: The silylation procedure allowed a clean separation of desired 2'-0-methyl derivative from the undesired 3'-0-methyl derivative via silica gel chromatography]
A mixture of 2' -O-methyl-5' -O-phthalimido-5- methyluridine (12.0 g, 28.7 mmol) , imidazole (5.86 g, 86.3 mmol) and tert-butyldiphenylsilylchloride (16.0 g, 57.5 mmol) in DMF (100 mL) was stirred at room temperature for 16 hours under an argon atmosphere. The reaction mixture was concentrated under vacuum to 1/4 of its volume and poured into ice water (500 mL) . The aqueous mixture was extracted with CH2C12 (2 x 250 mL) and washed with water (2 x 100 mL) and dried (MgS04) . The CH2C12 extract was concentrated and the residue was purified by silica gel chromatography. Elution with ether: hexanes (9:1, v/v) furnished the desired product as homogenous material . Appropriate fractions were pooled and concentrated to provide 10.51 g (55.8%) of the title compound. Η NMR (CDC13) δ 1.14 (s, 9, SiMe3) , 1.90 (s, 3,
CH3) , 3.26 (s, 3, OCH3) , 3.38 (m, 1, 4Η) , 4.08 (m, 1, 2' H) , 4.23 (m, 2, 5' CH2) , 4.50 (m, 1, 3' H) , 6.08 (d, 1, 1' H) , 7.34-7.42 (m, 10, Ar H) , 7.60 (s, 1, C6 H) , 7.65-7.85 (m, 4, Ar H) , 8.08 (br s, 1, NH) ; Anal. Calcd for C35H37N308Si : C, 64.10; H, 5.68; N, 6.40; Found: C, 63.96; H, 5.67; N, 6.16. 5' -O-Amino-3' -O-tert-butyldiphenylsilyl-2' -O-methyl-5- methyluridine.
To a stirred solution of 3' -O- ert-butyldiphenylsilyl- 2' -O-methyl-5' -O-phthalimido-5-methyluridine (3.1 g, 4.9 mmol) in CH2C12 (75 mL) was added methylhydrazine (0.39 mL, 7.5 mmol) in one portion at 0 °C. The reaction mixture was allowed to come to room temperature and stirred for 2 hours. The precipitate was filtered and washed with cold (=10 °C) CH2C12 (2 x 25 ml) . The combined filtrates were concentrated and the residue was purified by silica gel column chromatography. Elution with CH2Cl2:MeOH (9:1, v/v) furnished the desired product as homogenous material. Appropriate fractions were pooled and evaporated to yield 7.04 g of the title compound as a white foam. 'H NMR (CDC13) δ 1.11 (s, 9, SiMe3) , 1-82 (s, 3, CH3) , 3.24 (m, 1, 2' H) , 3.38 (s, 3, OCH3) , 3.63 and 3.96 (dd, 2, 5' CH2) , 4.11 and 4.19 (2m, 2, 3', 4' H) , 5.32 (br s, 2, 0NH2) , 5.93 (d, 1, 1' H) , 7.30 (s, 1, C6H) , 7.37-7.50 and 7.63-7.73 (m, 10, Ar H) and 9.22 (br s, 1, NH) ; Anal. Calcd for C27H35N306Si • 1/4 H20: C, 61.16; H, 6.74; H, 7.92; Found: C, 61.03; H, 6.56; N, 7.86.
J^-benzoyl-S' -0-terfc-butyldiphenylsilyl-5-methyl-2' - deoxycytidine.
To i^-benzoyl-δ' - O- ert-butyldimethylsilyl-3 ' - O- tert- butyldiphenylsilyl-5-methyl-2' -deoxycytidine (7.55 g, 10.8 mmol) in dry CH2C12 (110 mL) at 0 °C was added TMS0Tf (6.3 mL, 32.4 mmol) dropwise over 5 minutes. The reaction was stirred 1 hour at 0 °C, and 100 mL ice cold saturated aqueous NaHC03 was added, and the mixture allowed to come to room temperature with vigorous stirring over 1 hour. The organic layer was separated, washed with water, dried over magnesium sulfate, concentrated, dissolved in the minimum amount of CH2C12 and loaded onto a column packed in hexanes . Elution with 0 to 50% EtOAc/hexane provided 2.13 g (28%) of unreacted starting material, and 4.24 g (67%) of the desired product as a foam: Rf 0.42 (30% EtOAc/hexane); Η NMR (CDC13) δ 8.30 (d, 2H) , 7.69-7.36 ( , 14H) , 6.26 (t, J=6.8 Hz, IH, H-l') , 4.46 (m, IH) , 4.00 (m, IH) , 3.68 (m, IH) , 3.27 (m, IH) , 2.45- 1.92 (m, 2H) , 2.04 (s, 3H, CH3) , 1.09 (s, 9H) . Anal. Calcd for C33H37N305Si: C, 67.90; H, 6.39; N, 7.20. Found: C, 67.60; H, 6.34; N, 7.15.
5' -O-Phthalimido-i^-benzoyl-3' -O-tert- butyldiphenylsilyl-5-methyl-2' -deoxycytidine.
To a suspension of i^-benzoyl-S' -O- ert- butyldiphenylsilyl-5-methyl-2' -deoxycytidine (8.1 g, 14 mmol) and N-hydroxyphthalimide (2.6 g, 16 mmol) in dry THF (300 mL) was added diethyl azodicarboxylate (1.24 mL, 7.88 mol) dropwise at room temperature of 0.5 hour. The mixture was stirred overnight, concentrated, and the residue chromatographed (2% Me0H/CH2Cl2) to afford 10.5 g (103%) of slightly impure product, which was suitable for use in further reactions. A sample was recrystallized from ether/hexane (83% recovery) for analysis: J?f 0.47 (30% EtOAc/hexane) ; Η NMR (CDC13) δ 13.32 (s, IH) , 8.33 (d, 2H) , 7.90-7.35 (m, 18H) , 6.56 (dd, J=3.4, 5.4 Hz, IH, H-l') , 4.80 (m, IH) , 4.15-3.55 (m, 3H) , 2.60-1.95 (m, 2H) , 2.16 (s, 3H, CH3) , 1.13 (s, 9H) . Anal. Calcd for C41H40N4O7Si»H2O: C, 65.93; H, 5.67; N, 7.50. Found: C, 65.59; H, 5.56; N, 7.39.
5' -O-Amino-i^-benzoyl-S' -O-terfc-butyldiphenylsilyl-5- methyl-2' -deoxycytidine.
To 5' -0-phthalimido-.rø4-benzoyl-3' - O- tert- butyldiphenylsilyl-5-methyl-2' -deoxycytidine (10.2 g, 14 mmol) in 140 mL of CH2C12 at 0 °C was added methylhydrazine (0.90 mL, 16.8 mmol) dropwise. The solution was stirred at 0 °C until complete as judged by TLC, then filtered into 250 mL water. The filter caked was washed well with CH2C12, and the aqueous layer removed. Toluene (100 mL) was added, and the mixture concentrated. Chromatography (30% EtOAc/hexane) afforded 5.1 g (61%) of a foam: Rf 0.37 (30% EtOAc/hexane) ; 'H NMR (CDC13) δ 13.28 (s, IH) , 8.32 (d, 2H) , 7.75-7.35 (m, 14H) , 6.41 (t, J=6.6 Hz, IH, H-l') , 5.31 (s, 2H, ONH2) , 4.39 (m, IH) , 4.11 (m, IH) , 3.51 (m, IH) , 2.43 (m, IH) , 2.07 (s, 3H, CH3) , 1.89 (m, IH) , 1.09 (s, 9H) . Anal. Calcd for C33H38N405Si#1/4^0: C, 65.70; H, 6.43; N, 9.29. Found: C, 65.66; H, 6.35; N, 9.23.
^-benzoyl-δ' -O-tert-butyldiphenylsilyl-2' ,3' -C-dideoxy- 3' -styryl-5-methylcytidine.
To a solution of 1,2,4-triazole (12.25 g, 177 mmol) in dry acetonitrile (120 mL) at 0 °C is added P0C13 (3.49 mL, 37.4 mmol) dropwise over 30 minutes, followed by Et3N (24.7 mL, 177.3 mmol) over 1 hour. A solution of 5'-0-tert- butyldiphenylsilyl-3 ' -deoxy-3 ' -C-styryl-thymidine (9.31 g, 16.4 mmol) in dry acetonitrile (50 mL) was then added dropwise at 0 °C, and the solution allowed to warm to room temperature over 4 hour, with vigorous stirring.
Triethylamine (25 mL) and water (25 mL) were added and the mixture was concentrated to a small volume, diluted with EtOAc (500 mL) and extracted with 5% aqueous sodium bicarbonate (2 x 200 mL) , water (100 mL) , brine (100 mL) , then dried over magnesium sulfate, filtered and concentrated to afford 10.7 g (106%) of a foam. This material was azeotroped with dry 1,4-dioxane, then dissolved in 160 mL of the same solvent . To this solution was added a mixture of sodium hydride (2.62 g, 65.6 mmol) and benzamide (7.95 g, 65.6 mmol) stirred at room temperature for 0.5 hour via cannula in one portion. The reaction was stirred at room temperature until complete by TLC (1.5 hour) , at which point the reaction was cooled on ice, and quenched with acetic acid (3.8 mL) . The mixture was diluted 1000 mL 1:1 EtOAc/hexane, then extracted with water (2 x 500 mL) , and the organic layers dried over magnesium sulfate, filtered, concentrated, and chromatographed (50% EtOAc/hexane) afforded 8.44 g (77%) of a foam: Rf 0.87 (50% EtOAc/hexane); 'H NMR (CDC13) δ 6.53 (d, J=16 Hz, IH, PhCH=) , 5.98 (dd, J=7.4, 16 Hz, IH, =CH) , 1.74 (s, 3H, CH3) . i^-benzoyl-5' -O-tert-butyldiphenylsilyl-2' ,3' -dideoxy- 3' -C-formyl-5-methylcytidine.
To if-Benzoyl-5' - O- tert-butyldiphenylsilyl-2' ,3' - dideoxy-3' -C-styryl-5-methylcytidine (8.4 g, 12.5 mmol) in 1,4-dioxane (300 mL) was added NaI04 (11.6 g, 54.3 mmol) in warm water (100 mL) . The solution was stirred vigorously, and 3.2 mL aqueous Os04 (4% w/w, 0.5 mmol) was added, and the reaction stirred in the dark for 4 hours. The resulting mixture was partitioned between EtOAc (900 mL) and water (500 mL) , and the resulting emulsion broken by the addition of brine. The organic layer was washed with brine, then with 5% NaS03 (150 mL) , then again with brine, and finally dried over magnesium sulfate, filtered, and concentrated. The residue was chromatographed (0 to 10% MeOH/CH2Cl2) to provide 7.3 g (98%) of a foam: Rf 0.65 (5% MeOH/CH2Cl2) ; Η NMR (CDC13) δ 9.85 (s, IH, CHO) , 6.09 (t, IH, H-l'), 1.83 (s, 3H, CH3) . EXAMPLE 14
Synthesis of Dimeric MMI Nucleosides HO-T*UOHβ-OH
[3' -De(oxyphosphinico) -3' - (methyleneimino) thymidylyl- (3'→5') -2'-0-methyl-5-methyluridine]
A mixture of 5' -0-amino-3' -0- ert-butyldiphenylsilyl-2' - 0-methyl-5-methyluridine (5.25 g, 10 mmol) and 1- [5- ( tert- butyldiphenylsilyl) -2,3-dideoxy-3-C- (formyl) S-D- erythro- pentofuranosyl] thymine (prepared according to Sanghvi et al . Synthesis, 1994, 1163) (4.92 g, 10 mmol) in CH2Cl2:ACOH (50:1 mL) was stirred at room temperature for 5 minutes. The reaction mixture was then coevaporated with toluene (3 x 50 ml) under vacuum. The reaction was complete (by TLC) by the third coevaporation. The residue was dissolved in AcOH (25 ml) and cooled to =15 °C. NaCNBH3 (3 x 250 mg, 12 mmol) was added to the stirred reaction mixture in small portions (fume-hood) . The reaction mixture was stirred for 30 min. at =15 °C and to the cold solution aq. HCHO (30%, 5 ml) was added in one portion. The stirring was continued for 30 minutes and additional amount of NaCNBH3 (3 x 250 mg, 12 mmol) was added in a similar manner. After 2 hours, the reaction mixture was poured into ice-water (250 ml) and extracted with CH2C12 (2 x 250 ml) . The CH2C12 layer was washed with water (2 x 250 ml) and dried MgS04) . The solvent was removed and the residue purified by silica gel column chromatography. Elution with a gradient of CH2Cl2-CH2Cl2:MeOH (95:5, v/v) provided the desired product as homogenous material. Appropriate fractions were pooled and concentrated to furnish 5.08 g (50%) of the 3', 5' -protected MMI dimer. Η NMR (CDC13) δ 1.12 (s, 18, SiMe3) , 1.62 and 1.79 (2s, 6, CH3) , 2.45 (s, 3, N-CH3), 2.65 (m, 2, CH2-N-CH3) , 3.30 (s, 3, 0 CH3) , 5.82 (d, 1, l'H) , 6.17 (t, 1, l'H) , 8.40 (br s, 2, NH) and other protons.
To a stirred solution of 3', 5' -protected MMI dimer (2.2g, 1.88 mmol) in THF (25 ml) was added tetrabutylammonium fluoride (0.52 g, 2 mmol) at room temperature. The stirring was continued for 24 hours. The reaction mixture was loaded on the top of a silica gel column and elution with CH2C12 : MeOH (93:7, v/v) furnished homogenous material. Appropriate fractions were concentrated to furnish 1.0 g (99%) of the title compound as a white foam. 'H NMR at 60 °C (D20) δ 2.26 and 2.29 (2s, 6 CH3) , 2.77 (m, 2, 2' CH2) , 3.10 (s, 3, N-CH3) , 3.88 (s, 3, 0-CH3) , 6.27 (d, 1, l'H), 6.48 (t, 1, 1' H) , 7.90 and 8.09 (2s, 2, C6H) and other protons.
DMT-O-T*U0Mβ-O-Amidite. {3' -De(oxyphosphinico) -3' - [methylene (methylimino) ] - [thymidylyl (5' -O-dimethoxytrityl) ] - (3'-»5' ) -3' -O-β-cyano- ethyldiisopropylaminophosphiryl-2' -O-methyl-5-methyluridine}
The synthesis of the title phosphoramidite was accomplished in two steps from the MMI-2'-0-Me dimer (described above) in 92% overall yield. (General procedures for dimethoxytritylation and phosphitylation are described above) : Η NMR (CD3CN) δ 1.51 and 1.75 92s, 6, CH3) , 5.85 (t, 1, l'H) , 6.10 (pseudo t, 1, l'H) 9.17 (br s, 2, NH) and other protons. 31P (NMR) δ 150.9 and 151.2 ppm. TBDPS-0-T*TF-OH. Coupling reaction of 5' -O-amino-2 ' -fluorothymidine (0.54 g, 2 mmol) and 5' -O- ert-butyldiphenylsilyl-3 ' -deoxy-3 ' -C- formylthymidine (0.98 g, 2 mmol) according to the general procedure provided a fine, powdery solid after pouring into ice water. This material was collected, washed with water, and dried to provide 1.45 g (97%) of product which contained an impurity (ca '10%) by Η NMR. This material was used directly in the desilylation step: Rf 0.44 (10 % Me0H/CH2Cl2) ; Η NMR (CDC13) δ 6.07 (t, J=5.7 Hz, IH, H-l') , 5.75 (d, J=19.1 Hz, IH, H-l'') . TBDPS-0-T*C-0-TBDPS.
Coupling reaction of 5' -0-amino-_N-benzoyl-3 ' -0- tert- butyldiphenylsilyl-5-methyl-2' -deoxycytidine (2.02 g, 3.37 mmol) and 5' -0- ert-butyldiphenylsilyl-3 ' -deoxy-3 ' -C- formylthymidine (1.66 g, 3.37 mmol) according to the general procedure provided 2.8 g (84%) of a hard foam: Rf 0.50 (10 % Me0H/CH2Cl2) ; Η NMR (CDC13) δ 6.33 (t, J=6.0 Hz, IH, H-l') , 6.08 (d, J=6.0 Hz, IH, H-l'') , 2.37 (s, 3H, NCH3) , 1.80 (s, 3H, CH3) , 1.59 (s, 3H, CH3) . Anal. Calcd for C54H68N608Si2*2H20: C, 63.50; H, 7.11; N, 8.23. Found: C, 63.51; H, 6.80; N, 8.42.
TBDPS-0-A*C-0-TBDPS . Coupling reaction of 5' -O-amino-i^-benzoyl-S ' -0- tert- butyldiphenylsilyl-5-methyl-2' -deoxycytidine (1.43 g, 2.39 mmol) and 5' -O- ert-butyldiphenylsilyl-2' , 3' -dideoxy-3 ' -C- formyl adenosine (1.2 g, 2.39 mmol) according to the general procedure provided 1.26 g (53%) of a hard foam: Rf 0.35 (10 % MeOH/CH2Cl2) ; Η NMR (CDC13) δ 8.30 (s, IH) , 8.09 (s, IH) , 6.35-6.22 (m, 2H, H-l', H-l") , 5.87 (s, 2H, NH2) , 2.42 (s, 3H, NCH3) , 1.72 (s, 3H, CH3) . Anal. Calcd for C54H67N906Si2»H20 : C, 64.07; H, 6.87; N, 12.45. Found: C, 64.19; H, 6.91; N, 12.34. TBDPS-0-C*T-0-TBDPS.
Coupling reaction of 5' -0-amino-3' -O- ert-butyldiphenyl- silylthymidine (5.9 g, 12 mmol) and N4-benzoyl-5' -O- tert- butyldiphenylsilyl-2' ,3' -dideoxy-3' -C-formyl-5-methylcytidine (7.15 g, 12 mmol) according to the general procedure provided 7.0 g (60%) of a hard foam. This material was pure by TLC and 'H NMR, and was used -directly in the next step: Rf 0.58 (10% MeOH/CH2Cl2) ; Η NMR (CDC13) δ 6.34 (t, IH, H-l'), 6.12 (t, IH, H-l"), 2.39 (s, 3H, NCH3) , 1.80 (s, 3H, CH3) , 1.60 (s, 3H, CH3) . TBDPS-0-T*CBz-0-TBDPS.
TBDPS-0-T* eC-0-TBDPS (2.70 g, 2.75 mmol) was N- benzoylated according to the general procedure, then chromatographed (30% to 50% EtOAc/hexanes) to yield 1.80 g (60%) of a hard foam: Rf 0.54 (50% EtOAc/hexanes) ; Η NMR (CDC13) δ 6.35 (t, J=7.0 Hz, IH, H-l') , 6.09 (t, J=5.9 Hz, IH, H-l"), 2.40 (s, 3H, NCH3) , 1.97 (s, 3H, CH3) , 1.60 (s, 3H, CH3) . Anal. Calcd for C61H72N6O9Si2»0.5 H20: C, 66.70; H, 6.70; N, 7.65. Found: C, 66.65; H, 6.55; N, 7.55. TBDPS-0-CBz*T-0-TBDPS. TBDPS-0-MeC*T-0-TBDPS (7.0 g, 7.11 mmol) was benzoylated according to the general procedure, then chromatographed (2% MeOH/CH2Cl2) to yield 7.7 g (100%) of a hard foam: Rf 0.25 (2% MeOH/CH2Cl2) ; Η NMR (CDC13) δ 6.35 (dd, J=1.7, 6.0 Hz, IH, H-l') , 6.12 (t, J=6.0 Hz, IH, H-l") , 2.42 (s, 3H, NCH3) , 1.81 (s, 6H, 2 CH3 's) . Anal. Calcd for C61H72N609Si2 : C, 67.25; H, 6.66; N, 7.71. Found: C, 66.92; H, 6.51; N, 7.43. HO-T*TF-OH.
TBDPS-0-T*TF-OH (1.30 g, 1.70 mmol) was desilylated according to the general procedure to afford 0.65 g (67%) of a hard foam: Rf 0.29 (10% MeOH/CH2Cl2) ; XH NMR (DMSO-dff) δ 11.44 (s, IH) , 11.26 (s, IH) , 7.82 (s, IH) , (7.52 s, IH) , 6.03 (t, J= 5.3 Hz, IH, H-l') , 5.88 (dd J=2.0 , 18.9 Hz, IH, H-l") , 5.71 (d, IH, 3"-OH) , 5.08 (t, IH, 5'-OH) , 5.12 (dm, J=74.4 Hz, IH, H-2") , 4.2-3.4 (m, 9H) , 2.72 (m, IH) , 2.59 (S, 3H, NCH3) , 2.15 (m, 2H) , 1.80 (s, 3H, CH3) , 1.77 (s, 3H, CH3) . Anal. Calcd for C22H30N5O9F*H2O: C, 48.44; H, 5.91; N, 12.84. Found: C, 48.80; H, 5.92; N, 12.53. HO-T*CBz-OH.
TBDPS-0-T*MeCBz-0-TBDPS (1.70 g, 1.56 mmol) was desilylated according to the general procedure to afford 0.85 g (89%) of a hard foam: Rf 0.44 (10% MeOH/CH2Cl2) ; Η NMR (DMSO-d6) δ 13.02 (s, IH) , 11.26 (s, IH) , 8.19 (m, 2H) , (7.84 s, 2H) , 7.62-7.40 (m, 3H) , 6.15 (t, J= 6.4 Hz, IH, H-l') , 6.03 (t J=5.4 Hz, IH, H-l") , 5.38 (d, IH, 3-OH) , 5.11 (t, IH, 5-OH) , 4.38 (t, J=5.0 Hz, IH) , 4.3-3.5 (m, 8H) , 2.80-2.45 (m, 2H) , 2.61 (s, 3H, NCH3) , 2.17 (m, 3H) , 2.05 (s, 3H, CH3) , 1.77 (s, 3H, CH3) . Anal. Calcd for C29H36N6O9»0.5 H20: C, 56.03; H, 6.00; N, 13.52. Found: C, 56.18; H, 5.97; N, 13.30.
HO-CBl*T-OH.
TBDPS-0-MeCBz*T-0-TBDPS (6.97 g, 6.4 mmol) was desilylated according to the general procedure to afford 3.14 g (80%) of a hard foam: Rf 0.50 (10% MeOH/CH2Cl2) ; Η NMR (DMSO-de) δ 13.12 (s, IH) , 11.32 (s, IH) , (8.31 s, IH) , 8.22 (m, 2H) , 7.70-7.40 (m, 4H) , 6.16 (t, J= 6.9 Hz, IH, H-l') , 6.03 (t J=4.6 Hz, IH, H-l"), 5.32 (d, IH, 3-OH) , 5.25 (t, IH, 5-OH) , 4.30-3.60 (m, 9H) , 2.80-2.45 (m, 2H) , -2.62 (s, 3H, NCH3) , 2.35-1.90 (m, 3H) , 2.04 (s, 3H, CH3) , 1.80 (s, 3H, CH3) . Anal. Calcd for C29H36N609: C, 56.86; H, 5.92; N, 13.72. Found: C, 56.48; H, 6.23; N, 13.35. DMT-0-T*TF-OH.
HO-T*TF-OH (0.65 g, 1.23 mmol) was tritylated according to the general procedure to afford 0.72 g (73%) of the dimethoxytrityl ether derivative: Rf 0.39 (10% MeOH/CH2Cl2 + 0.1% Et3N) ;'H NMR (CDCl3) δ 6.08 (t, J=5.4 Hz, IH, H-l') , 5.68 (d, J=19.5 Hz, IH, H-l") , 5.07 (dd J=54.0, 4.2 Hz, IH, H- 2" ) . DMT-0-T*CBz-OH.
HO-T*MeCBz-OH (0.80 g, 1.31 mmol) was tritylated according to the general procedure to afford 0.95 g (79%) of the dimethoxytrityl ether derivative: Rf 0.45 (10% MeOH/CH2Cl2 + 0.1% Et3N) ; 'H NMR (CDC13) δ 6.21 (t, J=6.3 Hz, IH, H-l') , 6.09 (t, J=5.5 Hz, IH, H-l") . DMT-0-CBl*T-OH.
HO-MeCBz*T-OH (1.22 g, 2.0 mmol) was tritylated according to the general procedure. The reaction was incomplete. The unreacted starting was separated from the starting material via chromatography and the unreacted starting material tritylated again according to the general procedure. The products obtained were combined to afford 1.25.g (68%) of the dimethoxytrityl ether derivative: Rf 0.45 (10% MeOH/CH2Cl2 + 0.1% Et3N) ; 'H NMR (CDCl3) δ 6.20 (t, J=6.3 Hz, IH, H-l') , 6.11 (t, J=5.2 Hz, IH, H-l") . DMT-0-T*T-0-TBDPS.
The MMI dimer Tr-0-T*T-0-TBDPS was prepared from Tr-O-T- I and CH2=NO-T-0-TBDPS via our radical coupling reaction we described in F. Debart et. al . Tetrahedron Lett . 1992, 33 , 2645) , followed by our reductive methylation of the intermediate hydroxylamine that we described in J.-J. Vasseur et. al . J. Am . Chem. Soc . 1992, 114 , 4006. This material (6.78 g, 6.8 mmol) was dissolved in MeOH (150 mL) , 0.5 N HCl in ether (2 mL) was added, and the reaction stirred at room temperature for 2 hours. Triethylamine (1.5 mL) was added, and the solution was concentrated, then chromatographed (5% MeOH/CH2Cl2) to afford 4.25 g (83%) of the 5' -unprotected dimer (Rf 0.30, 5% MeOH/CH2Cl2) as a hard foam. This material was then dimethoxytritylated according to the general procedure to provide 5.3 g (89%) of a foam: Rf 0.56 (1% MeOH/CH2Cl2) ; Η NMR (CDC13) δ 6.31 (t, IH, H-l') , 6.10 (t, IH, H-l") , 2.35 (s, 3H, NCH3) , 1.73 (s, 3H, CH3) , 1.48 (s, 3H, CH3) .
DMT-0-T*T-OH.
DMT-0-T*T-0-TBDPS (2.7 g, 2.6 mmol) was desilylated according to the general procedure, except that the crude material was chromatographed using a stepwise gradient of 0 to 4% MeOH/CH2Cl2 + 0.1% Et3N. Concentration of the appropriate fractions provided 1.78 g (84%) of the dimethoxytrityl ether derivative: Rf 0.44 (5 % MeOH/CH2Cl2 + 0.1% Et3N) ; 'H NMR (CDCl3) δ 6.22 (t, J=6.4 Hz, IH, H-l') 6.08, (t, J=5.4 Hz, IH, H-l") .
DMT-0-CBz*CBz-0-TBDPS.
To a solution of 1, 2,4-triazole (11 g, 159 mmol) in dry acetonitrile (115 mL) at 0 °C is added P0C13 (3.4 mL, 37 mmol) dropwise over 30 minutes, followed by Et3N (26 mL, 185 mmol) over 1 hour. A solution of DMT-0-T*T-0-TBDPS (2.6 g,
2.47 mmol) in dry acetonitrile (10 mL) was then added dropwise at 0 °C, and the solution allowed to warm to room temperature over 3.5 hours, with vigorous stirring. The mixture was poured into ice water (200 mL) and extracted with EtOAc (2 x 200 mL) . The combined organic layers were washed with saturated aqueous NaHC03 (2 x 100 mL) , water (100 mL) , and brine (100 mL) , then dried over magnesium sulfate, filtered and concentrated to afford 2.56 g (90%) of a yellow foam. This material was azeotroped with dry 1,4-dioxane, dissolved in 25 mL of the same solvent. To this solution was added a mixture of sodium hydride (0.35 g, 8.87 mmol) and benzamide (1.08 g, 8.87 mmol) stirred at room temperature for 0.5 hour via cannula in one portion. The reaction was stirred at room temperature until complete by TLC, at which point the reaction was cooled on ice, and quenched with acetic acid
(0.5 mL, 8.87 mmol) . The mixture was diluted with water (200 mL) and CH2C12 (200 mL) , and the organic layers washed with water (2x200 mL) , brine, and then dried over magnesium sulfate, filtered and concentrated. Chromatography (CH2C12 + 0.1% Et3N) afforded 2.06 g (74%) of a foam: Rf 0.95 (CH2C12 + 0.1% Et3N) . DMT-0-CBz*CBz-0H.
DMT-0-MeCBz*MeCBz-OTBDPS (1.36 g, 1.1 mmol) was desilylated according to the general procedure, except that the column was packed with 10% hexanes/CH2Cl2 + 0.1% Et3N, then eluted with the same solvent, follow by a stepwise gradient of 0 to 5% MeOH/CH2Cl2 + 0.1% Et3N. Concentration of the appropriate fractions provided 0.69 g (63%) of the dimethoxytrityl ether derivative: Rf 0.63 (1% MeOH/CH2Cl2 + 0.1% Et3N) ; Η NMR (CDC13) δ 6.25-6.05 (m, 2H, H-l', H-l") . DMT-0-T*TF-0-Amidite. DMT-0-T*TF-0H (0.72 g, 0.9 mmol) was phosphitylated according to the general procedure to afford 0.79 g (85%) of the phosphoramidite derivative: Rf 0.86 (EtOAc + 0.1% Et3N) ; 'H NMR (CD3CN) δ 6.09 (t, J=5.6 Hz, IH, H-l') 5.81 (d, J=18.8 Hz, IH, H-l") ; 31P NMR (CD3CN) δ 151.66 (d, J=9.0 Hz), 151.35 (d, J=7.9 Hz) .
DMT-0-T*CBz-0-Amidite.
DMT-0-T*MeCBz-OH (0.94 g, 1.0 mmol) was phosphitylated according to the general procedure to afford 0.71 g (62%) of the phosphoramidite derivative: Rf 0.83 (EtOAc + 0.1% Et3N) ; Η NMR (CD3CN) δ 6.20-6.05 (m, 2H, H-l', H-l") ; 31P NMR (CD3CN) δ 149.65,' 149.43.
DMT-0-CBz*T-0-Amidite.
DMT-0-MeCBz*T-OH (1.17 g, 1.28 mmol) was phosphitylated according to the general procedure to afford 1.10 g (77%) of the phosphoramidite: Rf 0.55 (80% EtOAc/hexanes + 0.1% Et3N) ; 'H NMR (CD3CN) δ 6.20-6.05 (m, 2H, H-l', H-l") ; 31P NMR (CD3CN) δ 149.63, 149.24.
DMT-0-CBz*CBz-0-Amidite .
DMT-0-MeCBz*MeCBz-OH (0.30 g, 0.3 mmol) was phosphitylated according to the general procedure to afford 0.31 g (81%) of the phosphoramidite: Rf 0.81 (80% EtOAc/hexanes + 0.1% Et3N) ; 'H NMR ( CD3CN) δ 6 . 20 - 6 . 05 (m, 2H, H- l ' , H- l " ) ; 31P NMR ( CD3CN) δ 149 . 70 , 149 . 45 . DMT-0-T*T-0-Amidite .
DMT-0-T*T-OH (1.78 g, 2.19 mmol) was phosphitylated according to the general procedure, except that the organic layer was extracted with 5% aqueous sodium bicarbonate, dried, filtered, and concentrated prior to chromatography (1% MeOH/CH2Cl2 + 0.1% Et3N) . This resulted in contamination with an impurity, and additional chromatography (1:1 CH2Cl2/EtOAc + 0.1% Et3N to 2% MeOH in 1:1 CH2Cl2/EtOAc + 0.1% Et3N) was necessary to completely purify the product. Fractions containing only pure product were pooled, concentrated, and lyophilized from 1,4-dioxane to afford 1.28 g (58%) of the phosphoramidite derivative: Rf 0.91 (5% MeOH/CH2Cl2 + 0.1% Et3N) ; *H NMR (CD3CN) δ 6.20-6.05 (m, 2H, H-l', H-l") ; 31P NMR (CD3CN) δ 149.59, 149.23. EXAMPLE 15
Incorporation Of MMI Dimers And Assembly of Chimeric Oligonucleotides Methylene (methyl) imino (MMI) linked chimeric oligonucleotides correspond to oligonucleotides where one or several phosphodiesters and/or phosphorothioates linkages have been replaced by MMI internucleosidic linkage. Thus, creating an alternating motif of MMI→P=0→MMI or MMI→P=S→MMI linked chimeric oligomers.
The chimeric oligonucleotides containing MMI dimers were synthesized utilizing standard phosphoramidite chemistry. In this case the chimeric oligomers were synthesized attached to CPG (Controlled Pore Glass) solid support and the assembling was performed on an automated DNA synthesizer such as
Millipore Expedite or Applied Biosystem 380B. MMI dimers synthesized as phosphoramidite elongation units (DMT-X*X- phosphoramidite, X = A, T, G, C, 5MeC, * = 3' -CH2-N(CH3) -0-5' or X*X MMI) were obtained according the procedure described above in the Example 14. The initial deoxynucleoside or MMI dimer determining the 3'-end of the MMI containing chimeric oligonucleotide was loaded on to the CPG solid support via a succinyl linkage following the procedure described in Example 2 above. For example: 5' -0-DMTr-T*C-0- (succinyl-CPG-NMe2) -3' was loaded onto the CPG providing a loading of 24.20 mmole/g and 5'-0-DMTr-MeCBz*MeCBz-0- (succinyl-CPG-NMe2) -3' was loaded onto the CPG providing a loading of 22.32 mmole/g. Protocol For The Incorporation Of the MMI Dimers Into Oligomers Step 1 - Detritylation:
1 mmole of 5' -0-DMT-O- (succinyl-CPG-NMe2) -3 ' MMI dimer was packed into a small column and connected in line to a Millipore Expedite DNA synthesizer. TCA (3%) solution was passed through the column according the standard protocol used for standard amidite chemistry. Step 2 - Coupling (Of MMI Phosphoramidite Dimers) : MMI phosphoramidite dimers were diluted in anhydrous acetonitrile up to a concentration of 0.0673 Mol/1. Using the standard coupling step 0.217 ml of a mixture of MMI dimer solution and lH-tetrazole (l/l, v/v) was passed through the column with an extended coupling wait step of 300 seconds. Step 3 - Oxidation or Sulfurization of the phosphite triester linkage:
This step was carried out using the standard oxidizing protocol required for the commercial deoxyribonucleoside phosphoramidites. Step 4 - Capping:
This step was carried out using the standard capping protocol required for commercial deoxyribonucleoside phosphoramidites. Step 5- Isolation And Purification: At the end of the automated synthesis, MMI containing oligonucleotides were concomitantly deprotected and cleaved from the solid support by treatment with concentrated aqueous ammonia solution (30%) at 55 °C for 10 hours. The ammonia solution was then evaporated and the full length oligonucleotides were separated from the failure sequences on reverse phase HPLC (column: RCM Waters 8 X 10 Bondapack HC18HA, flow rate 2 ml/minutes, gradient 5% to 25% CH,CN in TEAA 0.05 N pH 7.00 over 25 minutes) .
Table 2: Chimeric Oligomers Containing MMI Linkages (Denoted by *)
Figure imgf000059_0001
Analytical Data For Oligomers In Table 2
Figure imgf000060_0001
Column Supelco LC 18 15cm x 4.6mm, 5mm, 5% to 25 CH,CN in 0.05M TEAA, pH 7.00 over 25 minutes, 1ml/minute.
Table 3 : Chimeric Oligomers Containing T*2,.0MβT MMI dimers
Figure imgf000060_0002
Analytical Data For Oligomers In Table 3
Figure imgf000061_0001
Column Supelco LC 18 15cm x 4.6mm, 5mm, 5% to 25 % CH3CN in 0.05M TEAA, pH 7.00 over 25 minutes, lml/minute.
Table 4 : Chimeric Oligomers Containing T*2,.PT MMI dimers
Figure imgf000061_0002
Analytical Data For Oligomers In Table 4
Figure imgf000061_0003
Column Supelco LC 18 15cm x 4.6mm, 5mm, 5% to 25' CH3CN m 0.05M TEAA, pH 7.00 over 25 minutes, lml/minute. EXAMPLE 16
Post-Oligo modifications (Regiospecific Reductive Alkylation Of
Imino Linkage After Chimeric Oligomer Synthesis)
General Procedure The key feature of this procedure is to utilize the oximes dimers (prepared according to our published procedure in J". Am . Chem . Soc . 1992, 114 , 4006) as their phosphoramidites and incorporate into oligomers as described above. For illustrative purposes, we describe two of such oxime dimers (DMT-0-T*T- phosphoramidite * = 3'-CH=N-0-5' or abbreviated as T*T oxime and DMT-0-T*GiBu phosphoramidite * = 3'-CH=N-0-5' or abbreviated as T*G oxime) as phosphoramidites and incorporated into various oligomers (see Tables 5-8 for examples) .
The automated assembly of chimeric oligomers containing oxime linked dimers was performed on a Millipore Expedite DNA synthesizer using previously described methodology (see Example 15 for details) . After deprotection and purification of the oxime containing chimeric oligonucleotides, all of the oximes linkages were selectively reduced and alkylated according the protocol described below.
The following procedure was carried out in a small flask (2 mL) :
1) - Oxime containing oligonucleotide (1 μmole) was dissolved in 0.23 mL water 2) - Diluted with MeOH 0.5 mL
3) - Bromocresol Green solution was added in water 15 ml
4) - NaBH3CN (40 mg) was added
5) - HCl/MeOH 2N solution (0.1 ml) was added
6) - Mixture was vortexed for 5 minutes 7) - Repeat steps 5 and 6 once
8) - Add the desired aldehyde (R-CHO) 100 eq/secondary imino linkage
9) - Vortex for 30 minutes
10) - Add NaBH3CN 40mg 11) - Add 0.1 mL HCl/MeOH 2N solution
12) - Vortex for 2 minutes
13) - Repeat steps 11 and 12 six times 14) - Neutralization with triethylammonium acetate 2N solution in water 0.3 mL
The solution was evaporated and the residue loaded on a Sephadex G-25 desalting column. Elution with water, pooling of appropriate fractions and evaporation furnished the desired oligonucleotides. Purification by reverse phase HPLC (column: RCM Waters 8 X 10 B ndapack HC18HA, flow rate 2 mL/minutes, gradient: 5% → 25% CH3CN in TEAA 0.05 N pH 7.00 over 25 minutes) gave analytically pure chimeric oligomers.
Table 5. Chimeric Oligomers Prepared Via Post-Oligo Modification Method Containing MMI Linkages
Figure imgf000063_0001
Column Supelco LC 18 15cm x 4.6mm, 5mm, 5% to 25% CH3CN in 0.05M TEAA, pH 7.00 over 25 minutes, lml/minute. Table 6. Chimeric Oligomers Prepared Via Post-Oligo Modification Method Containing MEI [methylene(ethylimino) ] Linkages
Figure imgf000064_0001
Column Supelco LC 18 15cm x 4.6mm, 5mm, 5% to 25% CH3CN in 0.05M TEAA, pH 7.00 over 25 minutes, lml/minute.
Table 7. Chimeric Oligomers Prepared Via Post-Oligo Modification Method Containing MPI [methylene(propylimino) ] Linkages
Figure imgf000064_0002
Column Supelco LC 18 15cm x 4.6mm, 5mm, 5% to 25% CH3CN in 0.05M TEAA, pH 7.00 over 25 minutes, lml/minute. Table 8. Chimeric Oligomers Prepared Via Post-Oligo Modification Method Containing McPI [methylene(eyelopropylimino) ] Linkages
ID No . Sequence
11045 G P0Cp0GPST*TP0T*TP0T*TP0T*TP0T*TP0 GP0CP0G- 3 ' McPI 12 Phosphodiesters + 5 McPI Linkages
Figure imgf000065_0001
Column Supelco LC 18 15cm x 4.6mm, 5mm, 5% to 25% CH3CN in 0.05M TEAA, pH 7.00 pver 25 minutes, lml/minute.
EXAMPLE 17
Preparation of Dimeric MMI Nucleosides Incoporating
Methylene(aralkylimido) Linkages
5' - O - ert-Butyldiphenylsilyl-3 ' -de (oxyphosphinico) -3 ' - [methylene (l-butyl-4- (1-pyrenyl) imino) ] thymidylyl- (3'-* 5') -3'- O-tert-butyldiphenylsilylthymidine.
To an oven-dried 200 mL recovery flask was added 3.51g (3.05 mmols) of 5 ' - O- ert-butyldiphenylsilyl-3 ' - de (oxyphosphinico) -3' -methyleneiminothymidylyl- (3'→ 5' ) -3 ' - O- tert-butylsilylthymidine and l-pyrenebutan-4-al (1.05 g, 3.85 mmols) were added as solids in one portion. The mixture was taken up in 20 mL of anhydrous CH2C12 and 80 mL of anhydrous toluene was added along with 1.0 mL of glacial AcOH yielding a pale golden solution. The mixture was evaporated on a rotary evaporator to a thick syrup. The syrup was then redissolved in CH2C12, rediluted with toluene and acetic acid, then evaporated three additional times at which point TLC analysis on silica gel (19:1 CH2Cl2/MeOH) showed the complete consumption of dimeric starting material . The flask containing the mixture was then equipped with a magnetic stir bar, a rubber septum, and an argon inlet and dissolved in 7.5 mL CH2C12 and diluted with 7.5 mL MeOH. Bromocresol green indicator solution in MeOH was added dropwise to give a more notable yellow color to the solution and a catalytic amount of AcOH was also added at this time. The mixture was then equilibrated in an ice water bath. At ice bath temperature, NaCNBH3 (0.850 g, 13.52 mmol) was added in four approximately equal portions over a period of 4 h. Over this time, HOAc was added dropwise upon the appearance of blue-green color to restore the original gold-yellow tint and maintain the pH of the mixture. After the final addition, the mixture was allowed to stir 2 additional hours at ice bath temperature. The mixture was then partitioned between water and CH2C12. The aqueous phase was extracted 3 additional times with CH2C12 and the combined organic phase was washed successively with saturated NaHC03, water, and brine and then dried over anh. Na2S04. The solution was decanted from the solids, evaporated and chromatographed on silica using 98:2 CH2Cl2/MeOH to yield 3.241 g of the title compound as a ivory foam.
Further transformations of this compound ( e . g. bis desilylation, 5' -dimethoxytritylation, and 3' -O-β-cyanoethyl- (N,N-diisopropylamino)phosphydylation) were carried out as described for the methyl (methyleneimino) analogs above. The compounds were characterized by 4ϊ and 13C NMR at either 200 or 400 MHZ. The final phosphoramidite product showed the expected two diastereotopic resonances in the 31P spectrum at 148.21 ppm and 147.11 ppm, respectively. 5' -O-tert-Butyldiphenylsilyl-3 ' -de (oxyphosphinico) -3 ' - [methylene- (l-propyl-3-l-pyrenylimino) ] thymidylyl- (3'-»5' ) -3' - O-ter -butyldiphenylsilylthymidine.
To an oven-dried 50 mL round bottom flask equipped with a septum, positive pressure argon line and magnetic stirbar was added 1.08 g (1.12 mmols) of 5' - 0- ert-butyldiphenylsilyl-3' - de (oxyphos-phinico) -3' -methyleneiminothymidylyl- (3'→5' ) -3' -0- tert-butylsilylthymidine as a solid. The dimer was taken up in 3 mL of anhydrous CH2C12 and 3 mL of anhydrous MeOH was added yielding a pale golden solution. 5 drops of bromocresol green was added followed by glacial AcOH dropwise until the solution retained a yellow coloration beyond the initial color of the solution. l-pyrenepropan-3-al (0.5783 g, 2.239 mmols) was added as a solid in one portion with stirring. The mixture was then allowed to stir at ambient temperature lh under argon after which time it was equilibrated in an ice-water bath. At ice bath temperature, NaCNBH3 (0.282 g, 4.48 mmol) was added in four approximately equal portions over a period of 4 h. Over this time, HOAc was added dropwise upon the appearance of blue- green color to maintain the pH of the mixture. After the final addition, the mixture was allowed to stir 2 additional hours at ice bath temperature. The mixture was then partitioned between water and CH2C12. The aqueous phase was extracted 3 additional times with CH2Cl2 and the combined organic phase was washed successively with saturated NaHC03, water, and brine and then dried over anh. Na2S04. The solution was decanted from the solids, evaporated and chromatographed on silica using 98:2 CH2Cl2/MeOH to yield 0.961 g of the title compound as a white foam.
Further transformations of this compound ( e . g. bis desilylation, 5' -dimethoxytritylation, and 3' -O-β-cyanoethyl- (N,N-diisopropylamino)phosphydylation) were carried out as described for the methyl (methyleneimino) analogs above. The compounds in this sequence were characterized by ~~E and 13C NMR at either 200 or 400 MHZ. The final phosphoramidite product showed the expected two diastereotopic resonances in the 31P spectrum at 149.59 ppm and 149.03 ppm, respectively.
EXAMPLE 18 Incorporation of Dimeric MMI Nucleosides Having Methylene- (aralkylimido) Linkages in Oligomers
The propypyreneyl T-T dimer of Example 17 was incorporated into a oligomer of the sequence Gp0GpoAp0T*Tp0Gp0Gp0Tp0C in a lOμmol synthesis using the standard protocols above. The oligomer was characterized by NMR and mass spectra. MS (Electrospray) , M+H = 3704.44. The oligomer was further shown by CGE to be full length material free of short mer sequences.
EVALUATION
PROCEDURE 1 - Nuclease Resistance A. Evaluation of the resistance of oligonucleotide- mimicking macromolecules to serum and cytoplasmic nucleases.
Oligonucleotide-mimicking macromolecules of the invention can be assessed for their resistance to serum nucleases by incubation of the oligonucleotide-mimicking macromolecules in media containing various concentrations of fetal calf serum or adult human serum. Labelled oligonucleotide-mimicking macromolecules are incubated for various times, treated with protease K and then analyzed by gel electrophoresis on 20% polyacrylamine-urea denaturing gels and subsequent autoradiography. Autoradiograms are quantitated by laser densitometry. Based upon the location of the modified linkage and the known length of the oligonucleotide-mimicking macromolecules it is possible to determine the effect on nuclease degradation by the particular modification. For the cytoplasmic nucleases, an HL 60 cell line can be used. A post-mitochondrial supernatant is prepared by differential centrifugation and the labelled macromolecules are incubated in this supernatant for various times. Following the incubation, macromolecules are assessed for degradation as outlined above for serum nucleolytic degradation. Autoradiography results are quantitated for evaluation of the macromolecules of the invention. It is expected that the macromolecules will be completely resistant to serum and cytoplasmic nucleases.
B. Evaluation of the resistance of oligonucleotide- mimicking macromolecules to specific endo- and exo-nucle- ases. Evaluation of the resistance of natural oligonucleotides and oligonucleotide-mimicking macromolecules of the invention to specific nucleases (i.e., endonucleases, 3',5'-exo-, and 5' , 3' -exonucleases) can be done to determine the exact effect of the macromolecule linkage on degradation. The oligonucleotide-mimicking macromolecules are incubated in defined reaction buffers specific for various selected nucleases. Following treatment of the products with protease K, urea is added and analysis on 20% polyacrylamide gels containing urea is done. Gel products are visualized by staining with Stains All reagent (Sigma Chemical Co.) . Laser densitometry is used to quantitate the extent of degradation. The effects of the macromolecules linkage are determined for specific nucleases and compared with the results obtained from the serum and cytoplasmic systems. As with the serum and cytoplasmic nucleases, it is expected that the oligonucleotide- mimicking macromolecules of the invention will be completely resistant to endo- and exo-nucleases.
PROCEDURE 2 - 5-Lipoxygenase Analysis and Assays A. Therapeutics
For therapeutic use, an animal suspected' of having a disease characterized by excessive or abnormal supply of 5-lipoxygenase is treated by administering the macromolecule of the invention. Persons of ordinary skill can easily determine optimum dosages, dosing methodologies and repetition rates. Such treatment is generally continued until either a cure is effected or a diminution in the diseased state is achieved. Long term treatment is likely for some diseases. B. Research Reagents
The oligonucleotide-mimicking macromolecules of this invention will also be useful as research reagents when used to cleave or otherwise modulate 5-lipoxygenase mRNA in crude cell lysates or in partially purified or wholly purified RNA preparations. This application of the invention is accomplished, for example, by lysing cells by standard methods, optimally extracting the RNA and then treating it with a composition at concentrations ranging, for instance, from about 100 to about 500 ng per 10 Mg of total RNA in a buffer consisting, for, example, of 50 mm phosphate, pH ranging from about 4-10 at a temperature from about 30° to about 50° C. The cleaved 5-lipoxygenase RNA can be analyzed by agarose gel electrophoresis and hybridization with radiolabeled DNA probes or by other standard methods. C. Diagnostics
The oligonucleotide-mimicking macromolecules of the invention will also be useful in diagnostic applications, particularly for the determination of the expression of specific mRNA species in various tissues or the expression of abnormal or mutant RNA species. In this example, while the macromolecules target a abnormal mRNA by being designed complementary to the abnormal sequence, they would not hybridize to normal mRNA.
Tissue samples can be homogenized, and RNA extracted by standard methods. The crude homogenate or extract can be treated for example to effect cleavage of the target RNA. The product can then be hybridized to a solid support which contains a bound oligonucleotide complementary to a region on the 5' side of the cleavage site. Both the normal and abnormal 5' region of the mRNA would bind to the solid support. The 3' region of the abnormal RNA, which is cleaved, would not be bound to the support and therefore would be separated from the normal mRNA.
Targeted mRNA species for modulation relates to 5- lipoxygenase; however, persons of ordinary skill in the art will appreciate that the present invention is not so limited and it is generally applicable. The inhibition or modulation of production of the enzyme 5-lipoxygenase is expected to have significant therapeutic benefits in the treatment of disease. In order to assess the effectiveness of the compositions, an assay or series of assays is required. D. In Vitro Assays
The cellular assays for 5-lipoxygenase preferably use the human promyelocytic leukemia cell line HL-60. These cells can be induced to differentiate into either a monocyte like cell or neutrophil like cell by various known agents. Treatment of the cells with 1.3% dimethyl sulfoxide, DMSO, is known to promote differentiation of the cells into neutrophils. It has now been found that basal HL-60 cells do not synthesize detectable levels of 5-lipoxygenase protein or secrete leukotrienes (a downstream product of 5-lipoxygenase) . Differentiation of the cells with DMSO causes an appearance of 5-lipoxygenase protein and leukotriene biosynthesis 48 hours after addition of DMSO. Thus induction of 5-lipoxygenase protein synthesis can be utilized as a test system for analysis of oligonucleotide- mimicking macromolecules which interfere with 5-lipoxygenase synthesis in these cells. A second test system for oligonucleotide-mimicking macromolecules makes use of the fact that 5-lipoxygenase is a "suicide" enzyme in that it inactivates itself upon reacting with substrate. Treatment of differentiated HL-60 or other cells expressing 5 lipoxygenase, with 10 μM A23187, a calcium ionophore, promotes translocation of 5-lipoxygenase from the cytosol to the membrane with subsequent activation of the enzyme. Following activation and several rounds of catalysis, the enzyme becomes catalytically inactive. Thus, treatment of the cells with calcium ionophore inactivates endogenous 5- lipoxygenase. It takes the cells approximately 24 hours to recover from A23187 treatment as measured by their ability to synthesize leukotriene B4. Macromolecules directed against 5- lipoxygenase can be tested for activity in two HL-60 model systems using the following quantitative assays. The assays are described from the most direct measurement of inhibition of 5-lipoxygenase protein synthesis in intact cells to more downstream events such as measurement of 5-lipoxygenase activity in intact cells. A direct effect which oligonucleotide-mimicking macromolecules can exert on intact cells and which can be easily be quantitated is specific inhibition of 5-lipoxygenase protein synthesis. To perform this technique, cells can be labelled with 35S-methionine (50 μCi/mL) for 2 hours at 37° C to label newly synthesized protein. Cells are extracted to solubilize total cellular proteins and 5-lipoxygenase is immunoprecipitated with 5-lipoxygenase antibody followed by elution from protein A Sepharose beads. The immunoprecipitated proteins are resolved by SDS-polyacrylamide gel electrophoresis and exposed for autoradiography. The amount of immunopre¬ cipitated 5-lipoxygenase is quantitated by scanning densitometry.
A predicted result from these experiments would be as follows. The amount of 5-lipoxygenase protein immuno- precipitated from control cells would be normalized to 100%. Treatment of the cells with 1 μM, 10 μM, and 30 μM of the macromolecules of the invention for 48 hours would reduce immunoprecipitated 5-lipoxygenase by 5%, 25% and 75% of control, respectively.
Measurement of 5-lipoxygenase enzyme activity in cellular homogenates could also be used to quantitate the amount of enzyme present which is capable of synthesizing leukotrienes. A radiometric assay has now been developed for quantitating 5- lipoxygenase enzyme activity in cell homogenates using reverse phase HPLC. Cells' are broken by sonication in a buffer containing protease inhibitors and EDTA. The cell homogenate is centrifuged at 10,000 x g for 30 min and the supernatants analyzed for 5-lipoxygenase activity. Cytosolic proteins are incubated with 10 μM 14C-arachidonic acid, 2mM ATP, 50 μM free calcium, 100 μg/ml phosphatidylcholine, and 50 mM bis-Tris buffer, pH 7.0, for 5 min at 37° C. The reactions are quenched by the addition of an equal volume of acetone and the fatty acids extracted with ethyl acetate. The substrate and reaction products are separated by reverse phase HPLC on a Novapak C18 column (Waters Inc., Millford, MA) . Radioactive peaks are detected by a Beckman model 171 radiochromatography detector. The amount of arachidonic acid converted into di-HETE's and mono-HETE's is used as a measure of 5-lipoxygenase activity.
A predicted result for treatment of DMSO differentiated HL-60 cells for 72 hours with effective the macromolecules of the invention at 1 μM, 10 μM, and 30 μM would be as follows.' Control cells oxidize 200 pmol arachidonic acid/ 5 min/ 106 cells. Cells treated with 1 μM, 10 μM, and 30 μM of an effective oligonucleotide-mimicking macromolecule would oxidize 195 pmol, 140 pmol, and 60 pmol of arachidonic acid/ 5 min/ 10s cells respectively. A quantitative competitive enzyme linked immunosorbant assay (ELISA) for the measurement of total 5-lipoxygenase protein in cells has been developed. Human 5-lipoxygenase expressed in E. coli and purified by extraction, Q-Sepharose, hydroxyapatite, and reverse phase HPLC is used as a standard and as the primary antigen to coat microtiter plates. 25 ng of purified 5-lipoxygenase is bound to the microtiter plates overnight at 4°C. The wells are blocked for 90 min with 5% goat serum diluted in 20 mM Tris-HCL buffer, pH 7.4, in the presence of 150 mM NaCl (TBS) . Cell extracts (0.2% Triton X- 100, 12,000 x g for 30 min.) or purified 5-lipoxygenase were incubated with a 1:4000 dilution of 5-lipoxygenase polyclonal antibody in a total volume of 100 μL in the microtiter wells for 90 min. The antibodies are prepared by immunizing rabbits with purified human recombinant 5-lipoxygenase. The wells are washed with TBS containing 0.05% tween 20 (TBST) , then incubated with 100 μL of a 1:1000 dilution of peroxidase conjugated goat anti-rabbit IgG (Cappel Laboratories, Malvern, PA) for 60 min at 25°C. The wells are washed with TBST and the amount of peroxidase labelled second antibody determined by development with tetramethylbenzidine.
Predicted results from such an assay using a 30 mer oligonucleotide-mimicking macromolecule at 1 μM, 10 μM, and 30 μM would be 30 ng, 18 ng and 5 ng of 5-lipoxygenase per 106 cells, respectively with untreated cells containing about 34 ng 5-1ipoxygenase.
A net effect of inhibition of 5-lipoxygenase biosynthesis is a diminution in the quantities of leukotrienes released from stimulated cells. DMSO-differentiated HL-60 cells release leukotriene B4 upon stimulation with the calcium ionophore A23187. Leukotriene B4 released into the cell medium can be quantitated by radioimmunoassay using commercially available diagnostic kits (New England Nuclear, Boston, MA) . Leukotriene B4 production can be detected in HL-60 cells 48 hours following addition of DMSO to differentiate the cells into a neutrophil- like cell. Cells (2 x 105 cells/mL) will be treated with increasing concentrations of the macromolecule for 48-72 hours in the presence of 1.3% DMSO. The cells are washed and resuspended at a concentration of 2 x 106 cell/mL in Dulbecco's phosphate buffered saline containing 1% delipidated bovine serum albumin. Cells are stimulated with 10 μM calcium ionophore A23187 for 15 min and the quantity of LTB4 produced from 5 x 105 cell determined by radioimmunoassay as described by the manufacturer. Using this assay the following results would likely be obtained with an oligonucleotide-mimicking macromolecule directed to the 5-LO mRNA. Cells will be treated for 72 hours with either 1 μM, 10 μM or 30 μM of the macromolecule in the presence of 1.3% DMSO. The quantity of LTB4 produced from 5 x 105 cells would be expected to be about 75 pg, 50 pg, and 35 pg, respectively with untreated differentiated cells producing 75 pg LTB4.
E. In Vivo Assay Inhibition of the production of 5-lipoxygenase in the mouse can be demonstrated in accordance with the following protocol. Topical application of arachidonic acid results in the rapid production of leukotriene B4, leukotriene C4 and prostaglandin E2 in the skin followed by edema and cellular infiltration. Certain inhibitors of 5-lipoxygenase have been known to exhibit activity in this assay. For the assay, 2 mg of arachidonic acid is applied to a mouse ear with the contralateral ear serving as a control. The polymorphonuclear cell infiltrate is assayed by myeloperoxidase activity in homogenates taken from a biopsy 1 hour following the administration of arachidonic acid. The edematous response is quantitated by measurement of ear thickness and wet weight of a punch biopsy. Measurement of leukotriene B4 produced in biopsy specimens is performed as a direct measurement of 5- lipoxygenase activity in the tissue. Oligonucleotide-mimicking macromolecules will be applied topically to both ears 12 to 24 hours prior to administration of arachidonic acid to allow optimal activity of the compounds. Both ears are pretreated for 24 hours with either 0.1 μmol, 0.3 μmol, or 1.0 μmol of the macromolecule prior to challenge with arachidonic acid. Values are expressed as the mean for three animals per concentration. Inhibition of polymorphonuclear cell infiltration for 0.1 μmol, 0.3 μmol, and 1 μmol is expected to be about 10%, 75% and 92% of control activity, respectively. Inhibition of edema is expected to be about 3%, 58% and 90%, respectively while inhibition of leukotriene B4 production would be expected to be about 15%, 79% and 99%, respectively. PROCEDURE 3 - Solid Support Biochemistry Reagents
Oligomers of the present invention also can advantageously be used as solid-phase biochemistry reagents ( see, e . g. , "Solid-Phase Biochemistry - Analytical and Synthetic Aspects" , W. H. Scouten, ed. , John Wiley & Sons, New York, 1983) , notably in solid-phase biosystems, especially bioassays or solid-phase techniques which concerns diagnostic detection/quantitation or affinity purification of complementary nucleic acids ( see, e. g. , "Affinity Chromatography - A Practical Approach" , P. D. G. Dean, W. S. Johnson and F. A. Middle, eds., IRL Press Ltd., Oxford 1986; "Nucleic Acid Hybridization - A Practical Approach" , B. D. Harnes and S. J. Higgins, IRL Press Ltd., Oxford 1987) . Present day methods for performing such bioassays or purification techniques almost exclusively utilize "normal" oligonucleotides either physically adsorbed or bound through a substantially permanent covalent anchoring linkage to beaded solid supports such as cellulose, glass beads, including those with controlled porosity (Mizutani, et al . , J. Chromatogr. , 1986, 356, 202), "Sephadex", "Sepharose", agarose, polyacrylamide, porous particulate alumina, hydroxyalkyl methacrylate gels, diol-bonded silica, porous ceramics, or contiguous materials such as filter discs of nylon and nitrocellulose. In one example employing the chemical synthesis of MMI containing oligo-dT on cellulose beads, affinity isolation of poly A tail containing mRNA is effected (Gilham in "Methods in Enzymology , " L. Grossmann and K. Moldave, eds., vol. 21, part D, page 191, Academic Press, New York and London, 1971) .
All the above-mentioned methods are applicable, however, it is especially desirable to utilize covalent linkage over the physical adsorption of the molecules in question, since the latter approach has the disadvantage that some of the immobilized molecules can be washed out (desorbed) during the hybridization or affinity process. Further, certain types of studies concerning the use of the oligomers of the invention in solid-phase biochemistry can be approached, facilitated, or greatly accelerated by use of the recently-reported "light- directed, spatially addressable, parallel chemical synthesis" technology (Fodor, et al . , Science, 1991, 251 , 161 ) , a technique that combines solid-phase chemistry and photolithography to produce thousands of highly diverse, but identifiable, permanently immobilized compounds in a substantially simultaneous way.
PROCEDURE 4 - Investigation of mutant β-amyloid precursor protein (βAPP)
Point mutations in the gene encoding β-amyloid have been suggested in familial Alzheimer's disease (FAD) . To analysis samples of individual biological cellular material, MMI oligomers are used to probe for such point mutation expressed proteins. A MMI containing oligomer of the selected sequence coding for the protein of interest is synthesized as above. Upon completion of the oligomer sequence and prior to removal of the oligomer from the solid support, a fluorescein label is added to the oligomer using fluorescein phosphoramidite (Cat. No. 10-1963-95, Glen Research Corp., Sterling VA) . This amidite is attached in the normal manner as the last phosphate residue added. Alternatively, the oligomers can be labeled with other reporter molecules (acridine or psoralen, both also available from Glen Research or rhodamine) before or after oligomer synthesis. Labeled oligomers are contacted with tissue or cell samples suspected of abnormal βAPP expression under conditions in which specific hybridization can occur, and the sample is washed to remove unbound oligomers. Label remaining in the sample indicates bound oligonucleotide and is quantitated using a fluorimeter, fluorescence microscope or other routine means. Tissue or cell samples suspected of expressing a point mutation in the βAPP gene are incubated with a fluorescein- labeled oligomer which is targeted to the mutant codon 717, codon 670 or codon 671 of βAPP mRNA. An identical sample of cells or tissues is incubated with a second labeled oligomer which is targeted to the same region of normal βAPP mRNA, under conditions in which specific hybridization can occur, and the sample is washed to remove unbound oligomer. Label remaining in the sample indicates bound oligomer and can be quantitated using a fluorimeter or other routine means. The presence of mutant βAPP is indicated if the first sample binds labeled oligomer and the second sample does not bind fluorescent label. Double labeling can also be used with the oligomers to specifically detect expression of mutant βAPP. A single tissue sample is incubated with a rhodamine-labeled oligomer which is targeted to codon 717, codon 670 or codon 671 of mutant βAPP mRNA and a fluorescein-labeled oligomer which is targeted to the translation initiation site of mutant βAPP mRNA, under conditions in which specific hybridization can occur. The sample is washed to remove unbound oligomer and labels are detected by and fluori etry with appropriate filters. The presence of mutant βAPP is indicated if the sample does not bind rhodamine-labeled oligomer but does retain the fluorescein label.
PROCEDURE 5 - Detection of mutant H-ras gene expression
Point mutations in the H-ras gene have been implicated in numerous aberrations of the Ras pathway. MMI oligomers are labeled after synthesis with fluorescein or other fluorescent tag as illustrated above. Alternatively, the oligomers are labeled with other reporter molecules before or after oligomer synthesis. Labeled oligomers are contacted with tissue or cell samples suspected of abnormal ras expression under conditions in which specific hybridization can occur, and the sample is washed to remove unbound oligomer. Label remaining in the sample indicates bound oligomer and is quantitated using a fluorimeter, fluorescence microscope or other routine means. Tissue or cell samples suspected of expressing a point mutation in the H-ras gene are incubated with a fluorescein- labeled oligomer which is targeted to the mutant codon 12, codon 13 or codon 61 of H-ras mRNA. An identical sample of cells or tissues is incubated with a second labeled oligomer which is targeted to the same region of normal H-ras mRNA, under conditions in which specific hybridization can occur, and the sample is washed to remove unbound oligomer. Label remaining in the sample indicates bound oligomer and can be quantitated using a fluorimeter or other routine means . The presence of mutant H-ras is indicated if the first sample binds labeled oligomer and the second sample does not bind fluorescent label.
Double labeling can also be used with oligomers to specifically detect expression of mutant ras. A single tissue sample is incubated with a rhodamine-labeled oligomer which is targeted to codon 12, codon 13 or codon 61 of mutant H-ras mRNA and a fluorescein-labeled oligomer which is targeted to the translation initiation site of ras mRNA, under conditions in which specific hybridization can occur. The sample is washed to remove unbound oligomer and labels are detected by and fluorimetry with appropriate filters. The presence of mutant ras is indicated if the sample does not bind rhodamine-labeled oligomer but does retain the fluorescein label .
PROCEDURE 6 - Effect of MMI-containing antisense oligonucleotides on PKC-α mRNA levels: A549 cells were treated with compounds ID No.s 9495 and 9496 as well as a phosphorothioate oligonucleotide standard of the same sequence as compound ID No. 9495 above, at doses from 100 to 400 nM for four hours in the presence of the cationic lipids DOTMA/DOPE, washed and allowed to recover for an additional 20 hours. Total RNA was extracted and 20μg of each was resolved on 1.2% gels and transferred to nylon membranes. These blots were probed with a 32P radiolabeled PKC-α cDNA probe and then stripped and reprobed with a radiolabeled G3PDH probe to confirm equal RNA loading. PKC-α transcripts were examined and quantified with a Phosphorlmager (Molecular Dynamics, Sunnyvale CA) . The results indicate that oligonucleotides of seqences 9495 and 9496 gave approximately 75% reduction of PKC- a mRNA levels, with IC50s of approximately 80 nM and 120 nM, respectively. The phosphorothioate oligonucleotide standard, known to be active against PKC-α, had an IC50 in this assay of approximately 175 nM. Thus the MMI compounds exhibited greater activity than the test standard. The high specific binding of the test compounds to the PKC-α sequence can also be used to distinguish PKC-α mRNA from other mRNA of other PKC isozymes such as the β, T and η isozymes.
PROCEDURE 7 - Northern blot analysis of ras expression in vivo
Cells were treated with oligomers in Opti-MEM reduced- serum medium containing 2.5 μl DOTMA. Oligomer was then added to the desired concentration. After 4 hours of treatment, the medium was replaced with medium without oligonucleotide. Cells were harvested 48 hours after oligomer treatment and RNA was isolated using a standard CsCl purification method. Kingston, R.E., in Current Protocols in Molecular Biology, (F.M. Ausubel, R. Brent, R.E. Kingston, D.D. Moore, J.A. Smith, J.G. Seidman and K. Strahl, eds.), John Wiley and Sons, NY. Northern hybridization: 10 μg of each RNA was electrophoresed on a 1.2% agarose/formaldehyde gel and transferred overnight to GeneBind 45 nylon membrane (Pharmacia LKB, Piscataway, NJ) using standard methods. Kingston, R.E., in Current Protocols in Molecular Biology, (F.M. Ausubel, R. Brent, R.E. Kingston, D.D. Moore, J.A. Smith, J.G. Seidman and K. Strahl, eds.), John Wiley and Sons, NY. RNA was UV- crosslinked to the membrane. Double-stranded 32P-labeled probes were synthesized using the Prime a Gene labeling kit (Promega, Madison WI) . The ras probe was a Sall-Nhel fragment of a cDNA clone of the activated (mutant) H-ras mRNA having a GGC-to-GTC mutation at codon-12. The control probe was G3PDH. Blots were prehybridized for 15 minutes at 68 °C with the QuickHyb hybridization solution (Stratagene, La Jolla, CA) . The heat- denatured radioactive probe (2.5 x 10s counts/2 ml hybridization solution) mixed with 100 μl of 10 mg/ml salmon sperm DNA was added and the membrane was hybridized for 1 hour at 68 °C. The blots were washed twice for 15 minutes at room temperature in 2x SSC/0.1% SDS and once for 30 minutes at 60 °C with 0.1XSSC/0.1%SDS. Blots were autoradiographed and the intensity of signal was quantitated using an ImageQuant Phosphorlmager (Molecular Dynamics, Sunnyvale, CA) . Northern blots were first hybridized with the ras probe, then stripped by boiling for 15 minutes in 0.Ix SSC/0.1%SDS and rehybridized with the control G3PDH probe to check for correct sample loading. The MMI-containing oligomers, compound ID No.s 9502, 9503 and 9504, identified in previous Examples above, had IC50s of approximately 100 nM, 90 nM and 120 nM, respectively. All were more active in this assay than a known-active phosphorothioate oligonucleotide of the same sequence that was used as the test standard. This standard phosphorothioate oligonucleotide had C50 of approximately 150 nM.
Those skilled in the art will appreciate that numerous changes and modifications may be made to the preferred embodiments of the invention and that such changes and modifications may be made without departing from the spirit of the invention. It is therefore intended that the appended claims cover all such equivalent variations as fall within the true spirit and scope of the invention.

Claims

WHAT IS CLAIMED IS:
1. A process for forming covalent linkages, comprising the steps of:
(a) providing a support-bound synthon having structure:
Figure imgf000081_0001
and
(b) contacting said support-bound synthon with a solution-phase synthon having structure:
Figure imgf000081_0002
said contacting being for a time and under reaction conditions effective to form a covalent linkage having structure CH=N-RA-CH2, CH2-CH=N-RA, CH2-RA-N=CH, or RA-N=CH- CH2; wherein:
Z-L and Y2 are selected such that (i) Z1 is C(0)H and Y2 is CH2RANH2; or
(ii) Z is CH2RANH2 and Y2 is C(0)H; or (iii) Zx is CH2C(0)H and Y2 is RANH2; or (iv) Zi is RANH2 and Y2 is CH2C(0)H; each RA is, independently, 0 or NR2;
Y1 is OH, ORHP, CH2OH, or CH2ORHP where RHP is a hydroxyl protecting group;
(P) is a solid support; each Lc is, independently, a covalent linkage having structure CH=N-RA-CH2, CH2-CH=N-RA, CH2-RA-N=CH, RA-N=CH-CH2, 0- P(0)20-CH2, or O-P(S) (0)0-CH2; n is 0-200; each R2 is, independently, H; alkyl or substituted alkyl having 1 to about 10 carbon atoms; alkenyl or substituted alkenyl having 2 to about 10 carbon atoms; alkynyl or substituted alkynyl having 2 to about 10 carbon atoms; alkaryl, substituted alkaryl, aralkyl, or substituted aralkyl having 7 to about 14 carbon atoms; alicyclic; heterocyclic; a reporter molecule; an RNA cleaving group; a group for improving the pharmacokinetic properties of an oligonucleotide; or a group for improving the pharmacodynamic properties of an oligonucleotide; each Bx is, independently, a nucleosidic base; each Q is, independently, 0, S, CH2, CHF or CF2; and each X is, independently, H; OH; alkyl or substituted alkyl having 1 to about 10 carbon atoms; alkaryl, substituted alkaryl, aralkyl, or substituted aralkyl having 7 to about 14 carbon atoms; F; Cl; Br; CN; CF3; OCF3; OCN; O-alkyl; S-alkyl; N-alkyl; O-alkenyl; S-alkenyl; N-alkenyl; SOCH3; S02CH3; ON02; N02; N3; NH2; heterocycloalkyl; heterocycloalkaryl; aminoalkylamino; polyalkylamino or substituted silyl; an RNA cleaving group; a group for improving the pharmacokinetic properties of an oligonucleotide; or a group for improving the pharmacodynamic properties of an oligonucleotide.
2. The process of claim 1 wherein RA is 0.
3. The process of claim 1 wherein RA is NH or NCH3.
4. The process of claim 1 wherein Zx is C(0)H and Y2 is CH2RANH2. The process of claim 1 wherein Zx is CH2RANH2 and Y2 is
C(0)H.
6. The process of claim 1 wherein Zτ is CH2C(0)H and Y2 is RANH2.
7. The process of claim 1 wherein is RANH2 and Y2 is CH2C(0)H.
8. The process of claim 1 wherein said support bound synthon is prepared by contacting a synthon having structure:
Figure imgf000083_0001
wherein Y2 is CH2RA- (phthalimido) or RA- (phthalimido) with hydrazine, methylhydrazine, or a combination thereof.
9. The process of claim 1 further comprising reducing said covalent linkage to form a reduced linkage having structure O^-NR^A-CHa, CH^CHS-NR^R^ CH^RA-NR^O^, or RA-NRX- CH2-CH2 wherein each R± is, independently, H; alkyl or substituted alkyl having 1 to about 10 carbon atoms; alkenyl or substituted alkenyl having 2 to about 10 carbon atoms; alkynyl or substituted alkynyl having 2 to about 10 carbon atoms; alkaryl, substituted alkaryl, aralkyl, or substituted aralkyl having 7 to about 14 carbon atoms; alicyclic; heterocyclic; a reporter molecule; an RNA cleaving group; a group for improving the pharmacokinetic properties of an oligonucleotide; or a group for improving the pharmacodynamic properties of an oligonucleotide.
10. The process of claim 9 wherein Zx is C(0)H and Y2 is CH2RANH2.
11. The process of claim 9 wherein RA is 0.
12. The process of claim 9 wherein Z is CH2C(0)H and Y2 is RANH2.
13. The process of claim 9 further comprising alkylating a free amine group in said reduced linkage.
14. The process of claim 13 further comprising cleaving the product of said claim from said support.
15. The process of claim 1 further comprising cleaving the product of said claim from said support.
16. The process of claim 14 further comprising removing said hydroxyl protecting group from the product of said claim.
17. The process of claim 16 further comprising contacting said product in the presence of acid with a 2'- cyanoethylphosphpramidite nucleotide or a 2' -terminal cyanoethylphosphoramidite oligonucleotide to form a phosphite- linked structure.
18. The process of claim 17 further comprising oxidizing said phosphite linkage structure and hydrolyzing said cyanoethyl group.
19. The process of claim 1 further comprising removing said hydroxyl protecting group from the product of said claim.
20. A compound having structure:
Figure imgf000085_0001
wherein:
Zj. is C(0)H, CH2C(0)H, CH2RANH2, or RANH2; RA is O or NR2;
Yx is OH, ORHP, CH2OH, or CH2ORHP where RHP is a hydroxyl protecting group;
R2 is H; alkyl or substituted alkyl having 1 to about 10 carbon atoms; alkenyl or substituted alkenyl having 2 to about 10 carbon atoms; alkynyl or substituted alkynyl having 2 to about 10 carbon atoms; alkaryl, substituted alkaryl, aralkyl, or substituted aralkyl having 7 to about 14 carbon atoms; alicyclic; heterocyclic; a reporter molecule; an RNA cleaving group; a group for improving the pharmacokinetic properties of an oligonucleotide; or a group for improving the pharmacody¬ namic properties of an oligonucleotide;
Bx is a nucleosidic base;
Q is 0, S, CH2, CHF or CF2; and
Xx is H; OH; alkyl or substituted alkyl having 1 to about 10 carbon atoms; alkaryl, substituted alkaryl, aralkyl, or substituted aralkyl having 7 to about 14 carbon atoms; F; Cl; Br; CN; CF3; 0CF3; OCN; O-alkyl; S-alkyl; N-alkyl; O-alkenyl; S-alkenyl; N-alkenyl; S0CH3; S02CH3; ON02; N02; N3; NH2; heterocycloalkyl; heterocycloalkaryl; aminoalkylamino; polyalkylamino or substituted silyl; an RNA cleaving group; a group for improving the pharmacokinetic properties of an oligonucleotide; or a group for improving the pharmacodynamic properties of an oligonucleotide.
21. The compound of claim 20 wherein RA is O.
22. The compound of claim 20 wherein RA is NH.
23. The compound of claim 20 wherein Z is C(0)H or
CH2C(0)H.
24 . The compound of claim 20 wherein Zx is CH2RANH2 or
RA H2 ,
25. The compound of claim 20 wherein RHP is phthalimido, fluorenylmethoxycarbonyl, t-butyldimethylsilyl, or t- butyldiphenylsilyl.
26. A compound having structure:
Figure imgf000086_0001
wherein : Y2 is C (0) H , CH2C (0) H, CH2RANH2 , or RANH2 ;
Z2 is OH, 0RHP , CH20H, or CH20RHP ; n is 0 - 200 ;
(P) is a solid support; each Lc is, independently, a covalent linkage having structure CH=N-RA-CH2, CH2-CH=N-RA, CH2-RA-N=CH, RA-N=CH-CH2, CH2- NR^RA-CHS, CHs-CHz-NRi-RA, CH^RA-NR^O^, RA-NR-L-CT^-CH^ 0-P(0)20- CH2, or O-P(S) (0)0-CH2; each RA is, independently, O or NR2; each Rx and R2 is, independently, H; alkyl or substituted alkyl having 1 to about 10 carbon atoms; alkenyl or substituted alkenyl having 2 to about 10 carbon atoms; alkynyl or substituted alkynyl having 2 to about 10 carbon atoms; alkaryl, substituted alkaryl, aralkyl, or substituted aralkyl having 7 to about 14 carbon atoms; alicyclic; heterocyclic; a reporter molecule; an RNA cleaving group; a group for improving the pharmacokinetic properties of an oligonucleotide; or a group for improving the pharmacodynamic properties of an oligonucleotide; each Bx is, independently, a nucleosidic base; each Q is, independently, 0, S, CH2, CHF or CF2; and each X is, independently, H; OH; alkyl or substituted alkyl having 1 to about 10 carbon atoms; alkaryl, substituted alkaryl, aralkyl, or substituted aralkyl having 7 to about 14 carbon atoms; F; Cl; Br; CN; CF3; OCF3; OCN; O-alkyl; S-alkyl; N-alkyl; O-alkenyl; S-alkenyl; N-alkenyl; SOCH3; S02CH3; ON02; N02; N3; NH2; heterocycloalkyl; heterocycloalkaryl; aminoalkylamino; polyalkylamino or substituted silyl; an RNA cleaving group; a group for improving the pharmacokinetic properties of an oligonucleotide; or a group for improving the pharmacodynamic properties of an oligonucleotide.
27. The compound of claim 26 wherein RA is 0.
28. The compound of claim 26 wherein Y2 is CH2RANH2 or RANH2.
29. The compound of claim 26 wherein Lc is CH=N-RA-CH2, CH2-CH=N-RA, CH2-RA-N=CH, or RA-N=CH-CH2.
30. The compound of claim 26 wherein Lc is CH^NR-L-RA-CH;,,
Figure imgf000087_0001
31. The compound of claim 30 wherein R is H.
32. The compound of claim 30 wherein Rx is CH3.
PCT/US1994/014883 1993-12-28 1994-12-28 Backbone modified oligonucleotide analogs and solid phase synthesis thereof WO1995018136A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP95906115A EP0737201A4 (en) 1993-12-28 1994-12-28 Backbone modified oligonucleotide analogs and solid phase synthesis thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/174,379 1993-12-28
US08/174,379 US5541307A (en) 1990-07-27 1993-12-28 Backbone modified oligonucleotide analogs and solid phase synthesis thereof

Publications (1)

Publication Number Publication Date
WO1995018136A1 true WO1995018136A1 (en) 1995-07-06

Family

ID=22635952

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1994/014883 WO1995018136A1 (en) 1993-12-28 1994-12-28 Backbone modified oligonucleotide analogs and solid phase synthesis thereof

Country Status (3)

Country Link
US (1) US5541307A (en)
EP (1) EP0737201A4 (en)
WO (1) WO1995018136A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0763052A1 (en) * 1994-05-31 1997-03-19 Isis Pharmaceuticals, Inc. ANTISENSE OLIGONUCLEOTIDE MODULATION OF raf GENE EXPRESSION
EP0813539A1 (en) * 1995-03-06 1997-12-29 Isis Pharmaceuticals, Inc. Improved process for the synthesis of 2'-o-substituted pyrimidines and oligomeric compounds therefrom
WO1999013105A1 (en) * 1997-09-05 1999-03-18 Mikael Kubista Method for the preparation of a probe for nucleic acid hybridization
DE19819735A1 (en) * 1998-05-02 1999-11-04 Novartis Ag Device and method for producing an arrangement of chain molecules on a carrier material
JP2000300299A (en) * 1999-03-26 2000-10-31 Vysis Inc Solid-phase nucleic acid labeling by amino group transfer
US6776986B1 (en) 1996-06-06 2004-08-17 Novartis Ag Inhibition of HIV-1 replication by antisense RNA expression

Families Citing this family (844)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6753423B1 (en) 1990-01-11 2004-06-22 Isis Pharmaceuticals, Inc. Compositions and methods for enhanced biostability and altered biodistribution of oligonucleotides in mammals
US20040142899A1 (en) * 1990-01-11 2004-07-22 Isis Pharmaceuticals, Inc. Compositions and methods for enhanced biostability and altered biodistribution of oligonucleotides in mammals
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
US5386023A (en) * 1990-07-27 1995-01-31 Isis Pharmaceuticals Backbone modified oligonucleotide analogs and preparation thereof through reductive coupling
US5623070A (en) 1990-07-27 1997-04-22 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5792844A (en) * 1990-07-27 1998-08-11 Isis Pharmaceuticals, Inc. Oligonucleoside linkages containing adjacent nitrogen atoms
YU187991A (en) * 1990-12-11 1994-09-09 Hoechst Aktiengesellschaft 3- (2) -AMINO-ALI THIOL-MODIFIED, FLUORESCENT-DYED NUCLEOSIDES, NUCLEOTIDS AND OLIGONUCLEOTIDES, PROCESS FOR THEIR OBTAINING AND THEIR USE
US5965722A (en) * 1991-05-21 1999-10-12 Isis Pharmaceuticals, Inc. Antisense inhibition of ras gene with chimeric and alternating oligonucleotides
US6335434B1 (en) 1998-06-16 2002-01-01 Isis Pharmaceuticals, Inc., Nucleosidic and non-nucleosidic folate conjugates
US8153602B1 (en) 1991-11-19 2012-04-10 Isis Pharmaceuticals, Inc. Composition and methods for the pulmonary delivery of nucleic acids
US6537973B1 (en) 1992-03-16 2003-03-25 Isis Pharmaceuticals, Inc. Oligonucleotide inhibition of protein kinase C
US5985558A (en) 1997-04-14 1999-11-16 Isis Pharmaceuticals Inc. Antisense oligonucleotide compositions and methods for the inibition of c-Jun and c-Fos
EP0728139B1 (en) 1993-09-03 2003-08-13 Isis Pharmaceuticals, Inc. Amine-derivatized nucleosides and oligonucleosides
US6420549B1 (en) 1995-06-06 2002-07-16 Isis Pharmaceuticals, Inc. Oligonucleotide analogs having modified dimers
US5854033A (en) 1995-11-21 1998-12-29 Yale University Rolling circle replication reporter systems
US20040171028A1 (en) * 1996-06-06 2004-09-02 Baker Brenda F. Phosphorous-linked oligomeric compounds and their use in gene modulation
US5898031A (en) 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
US9096636B2 (en) 1996-06-06 2015-08-04 Isis Pharmaceuticals, Inc. Chimeric oligomeric compounds and their use in gene modulation
US20040266706A1 (en) * 2002-11-05 2004-12-30 Muthiah Manoharan Cross-linked oligomeric compounds and their use in gene modulation
US20030044941A1 (en) 1996-06-06 2003-03-06 Crooke Stanley T. Human RNase III and compositions and uses thereof
US7812149B2 (en) 1996-06-06 2010-10-12 Isis Pharmaceuticals, Inc. 2′-Fluoro substituted oligomeric compounds and compositions for use in gene modulations
US20040161844A1 (en) * 1996-06-06 2004-08-19 Baker Brenda F. Sugar and backbone-surrogate-containing oligomeric compounds and compositions for use in gene modulation
US6111085A (en) * 1996-09-13 2000-08-29 Isis Pharmaceuticals, Inc. Carbamate-derivatized nucleosides and oligonucleosides
US6127533A (en) * 1997-02-14 2000-10-03 Isis Pharmaceuticals, Inc. 2'-O-aminooxy-modified oligonucleotides
US6576752B1 (en) 1997-02-14 2003-06-10 Isis Pharmaceuticals, Inc. Aminooxy functionalized oligomers
US6172209B1 (en) 1997-02-14 2001-01-09 Isis Pharmaceuticals Inc. Aminooxy-modified oligonucleotides and methods for making same
US6251591B1 (en) 1997-02-27 2001-06-26 Lorne Park Research, Inc. Quantitative method for detecting nucleotide concentration
US6046004A (en) * 1997-02-27 2000-04-04 Lorne Park Research, Inc. Solution hybridization of nucleic acids with antisense probes having modified backbones
EP1012331B1 (en) 1997-07-01 2006-03-29 Isis Pharmaceuticals, Inc. Compositions and methods for the delivery of oligonucleotides via the alimentary canal
US6383808B1 (en) 2000-09-11 2002-05-07 Isis Pharmaceuticals, Inc. Antisense inhibition of clusterin expression
US6809193B2 (en) 1997-08-13 2004-10-26 Isis Pharmaceuticals, Inc. Antisense oligonucleotide compositions and methods for the modulation of JNK proteins
US20070149472A1 (en) * 1997-08-13 2007-06-28 Mckay Robert Antisense oligonucleotide compositions and methods for the modulation of jnk proteins
US5877309A (en) 1997-08-13 1999-03-02 Isis Pharmaceuticals, Inc. Antisense oligonucleotides against JNK
US6133246A (en) * 1997-08-13 2000-10-17 Isis Pharmaceuticals Inc. Antisense oligonucleotide compositions and methods for the modulation of JNK proteins
US6518017B1 (en) * 1997-10-02 2003-02-11 Oasis Biosciences Incorporated Combinatorial antisense library
US20030165888A1 (en) * 2001-07-18 2003-09-04 Brown Bob D. Oligonucleotide probes and primers comprising universal bases for diagnostic purposes
WO2000018885A1 (en) 1998-09-29 2000-04-06 Gamida Cell Ltd. Methods of controlling proliferation and differentiation of stem and progenitor cells
US20040186071A1 (en) 1998-04-13 2004-09-23 Bennett C. Frank Antisense modulation of CD40 expression
AU753270B2 (en) 1998-05-21 2002-10-10 Isis Pharmaceuticals, Inc. Compositions and methods for topical delivery of oligonucleotides
EP1080103A4 (en) 1998-05-21 2003-07-02 Isis Pharmaceuticals Inc Compositions and methods for non-parenteral delivery of oligonucleotides
US6255050B1 (en) 1998-05-22 2001-07-03 Lorne Park Research, Inc. Dynamic hybridization system
US6300319B1 (en) 1998-06-16 2001-10-09 Isis Pharmaceuticals, Inc. Targeted oligonucleotide conjugates
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
US6043352A (en) 1998-08-07 2000-03-28 Isis Pharmaceuticals, Inc. 2'-O-Dimethylaminoethyloxyethyl-modified oligonucleotides
US6673912B1 (en) 1998-08-07 2004-01-06 Isis Pharmaceuticals, Inc. 2′-O-aminoethyloxyethyl-modified oligonucleotides
US20040009938A1 (en) * 1998-08-07 2004-01-15 Muthiah Manoharan Methods of enhancing renal uptake of oligonucleotides
US6225293B1 (en) 1998-09-02 2001-05-01 Isis Pharmaceuticals, Inc. Methods and compounds for tracking the biodistribution of macromolecule-carrier combinations
US6077709A (en) 1998-09-29 2000-06-20 Isis Pharmaceuticals Inc. Antisense modulation of Survivin expression
US6667176B1 (en) 2000-01-11 2003-12-23 Geron Corporation cDNA libraries reflecting gene expression during growth and differentiation of human pluripotent stem cells
EP1129064B1 (en) 1998-11-12 2008-01-09 Invitrogen Corporation Transfection reagents
US6492111B1 (en) * 1998-11-25 2002-12-10 Isis Pharmaceuticals, Inc. In situ binary synthesis of biologically effective molecules
US6087112A (en) * 1998-12-30 2000-07-11 Oligos Etc. Inc. Arrays with modified oligonucleotide and polynucleotide compositions
US20030180789A1 (en) * 1998-12-30 2003-09-25 Dale Roderic M.K. Arrays with modified oligonucleotide and polynucleotide compositions
US6300320B1 (en) 1999-01-05 2001-10-09 Isis Pharmaceuticals, Inc. Modulation of c-jun using inhibitors of protein kinase C
US6403779B1 (en) 1999-01-08 2002-06-11 Isis Pharmaceuticals, Inc. Regioselective synthesis of 2′-O-modified nucleosides
US6127124A (en) * 1999-01-20 2000-10-03 Isis Pharmaceuticals, Inc. Fluorescence based nuclease assay
US6207819B1 (en) * 1999-02-12 2001-03-27 Isis Pharmaceuticals, Inc. Compounds, processes and intermediates for synthesis of mixed backbone oligomeric compounds
KR20020013519A (en) * 1999-04-08 2002-02-20 추후제출 Antisense Oligonucleotides Comprising Universal and/or Degenerate Bases
US7098192B2 (en) 1999-04-08 2006-08-29 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of STAT3 expression
US7534605B2 (en) * 1999-06-08 2009-05-19 Yissum Research Development Company Of The Hebrew University Of Jerusalem CD44 polypeptides, polynucleotides encoding same, antibodies directed thereagainst and method of using same for diagnosing and treating inflammatory diseases
US6656730B1 (en) 1999-06-15 2003-12-02 Isis Pharmaceuticals, Inc. Oligonucleotides conjugated to protein-binding drugs
US6593466B1 (en) 1999-07-07 2003-07-15 Isis Pharmaceuticals, Inc. Guanidinium functionalized nucleotides and precursors thereof
US6147200A (en) * 1999-08-19 2000-11-14 Isis Pharmaceuticals, Inc. 2'-O-acetamido modified monomers and oligomers
US6277982B1 (en) 1999-08-20 2001-08-21 Isis Pharmaceuticals, Inc. Alkylation of alcohols, amines, thiols and their derivatives by cyclic sulfate intermediates
US7332275B2 (en) 1999-10-13 2008-02-19 Sequenom, Inc. Methods for detecting methylated nucleotides
US20020055479A1 (en) 2000-01-18 2002-05-09 Cowsert Lex M. Antisense modulation of PTP1B expression
US6261840B1 (en) 2000-01-18 2001-07-17 Isis Pharmaceuticals, Inc. Antisense modulation of PTP1B expression
US6911535B2 (en) * 2000-03-22 2005-06-28 Solvlink Biosciences Biomolecule/polymer conjugates
DE60140864D1 (en) * 2000-04-13 2010-02-04 Thomas N Wight THERAPEUTIC SUMMARIES AND METHOD FOR MODULATING V3, AN ISOFORM FROM VERSICAN
US6680172B1 (en) 2000-05-16 2004-01-20 Regents Of The University Of Michigan Treatments and markers for cancers of the central nervous system
US6656700B2 (en) 2000-05-26 2003-12-02 Amersham Plc Isoforms of human pregnancy-associated protein-E
US6686188B2 (en) * 2000-05-26 2004-02-03 Amersham Plc Polynucleotide encoding a human myosin-like polypeptide expressed predominantly in heart and muscle
US6958214B2 (en) 2000-07-10 2005-10-25 Sequenom, Inc. Polymorphic kinase anchor proteins and nucleic acids encoding the same
AU2001276691A1 (en) * 2000-08-03 2002-02-18 Matsushita Electric Industrial Co., Ltd. Brushless motor and method of manufacturing the brushless motor
US8568766B2 (en) 2000-08-24 2013-10-29 Gattadahalli M. Anantharamaiah Peptides and peptide mimetics to treat pathologies associated with eye disease
US20020123474A1 (en) * 2000-10-04 2002-09-05 Shannon Mark E. Human GTP-Rho binding protein2
EP2336166A1 (en) 2000-10-12 2011-06-22 University Of Rochester Compositions that inhibit proliferation of cancer cells
AU2002232387A1 (en) * 2000-10-27 2002-05-06 Invitrogen Corporation Method for introducing antisense oligonucleotides into eucaryotic cells
US7767802B2 (en) 2001-01-09 2010-08-03 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of anti-apoptotic genes
US6573051B2 (en) * 2001-03-09 2003-06-03 Molecular Staging, Inc. Open circle probes with intramolecular stem structures
ES2328796T3 (en) 2001-03-14 2009-11-18 Myriad Genetics, Inc. TSG101-GAG INTERACTION AND USE OF THE SAME.
US7803915B2 (en) 2001-06-20 2010-09-28 Genentech, Inc. Antibody compositions for the diagnosis and treatment of tumor
CA2633171C (en) 2001-06-20 2012-11-20 Genentech, Inc. Antibodies against tumor-associated antigenic target (tat) polypeptides
AU2002315393A1 (en) 2001-06-21 2003-01-08 Isis Pharmaceuticals, Inc. Antisense modulation of superoxide dismutase 1, soluble expression
US6964950B2 (en) 2001-07-25 2005-11-15 Isis Pharmaceuticals, Inc. Antisense modulation of C-reactive protein expression
US7425545B2 (en) 2001-07-25 2008-09-16 Isis Pharmaceuticals, Inc. Modulation of C-reactive protein expression
US20030096772A1 (en) 2001-07-30 2003-05-22 Crooke Rosanne M. Antisense modulation of acyl CoA cholesterol acyltransferase-2 expression
US7407943B2 (en) 2001-08-01 2008-08-05 Isis Pharmaceuticals, Inc. Antisense modulation of apolipoprotein B expression
US20040096880A1 (en) * 2001-08-07 2004-05-20 Kmiec Eric B. Compositions and methods for the treatment of diseases exhibiting protein misassembly and aggregation
US7227014B2 (en) 2001-08-07 2007-06-05 Isis Pharmaceuticals, Inc. Antisense modulation of apolipoprotein (a) expression
US20030109476A1 (en) * 2001-08-07 2003-06-12 Kmiec Eric B. Compositions and methods for the prevention and treatment of Huntington's disease
DE60238143D1 (en) 2001-09-18 2010-12-09 Genentech Inc COMPOSITIONS AND METHODS FOR THE DIAGNOSIS OF TUMORS
NZ577565A (en) 2001-10-09 2010-10-29 Isis Pharmaceuticals Inc Antisense modulation of insulin-like growth factor binding protein 5 expressions
US6750019B2 (en) 2001-10-09 2004-06-15 Isis Pharmaceuticals, Inc. Antisense modulation of insulin-like growth factor binding protein 5 expression
US20030170678A1 (en) * 2001-10-25 2003-09-11 Neurogenetics, Inc. Genetic markers for Alzheimer's disease and methods using the same
US20030224380A1 (en) * 2001-10-25 2003-12-04 The General Hospital Corporation Genes and polymorphisms on chromosome 10 associated with Alzheimer's disease and other neurodegenerative diseases
AU2002364945A1 (en) * 2001-10-25 2003-07-09 Neurogenetics, Inc. Genes and polymorphisms on chromosome 10 associated with alzheimer's disease and other neurodegenerative diseases
US6965025B2 (en) 2001-12-10 2005-11-15 Isis Pharmaceuticals, Inc. Antisense modulation of connective tissue growth factor expression
WO2003057160A2 (en) 2002-01-02 2003-07-17 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
IL152904A0 (en) 2002-01-24 2003-06-24 Gamida Cell Ltd Utilization of retinoid and vitamin d receptor antagonists for expansion of renewable stem cell populations
WO2003062404A1 (en) * 2002-01-25 2003-07-31 Gamida-Cell Ltd. Methods of expanding stem and progenitor cells and expanded cell populations obtained thereby
US7553619B2 (en) 2002-02-08 2009-06-30 Qiagen Gmbh Detection method using dissociated rolling circle amplification
US20030191075A1 (en) * 2002-02-22 2003-10-09 Cook Phillip Dan Method of using modified oligonucleotides for hepatic delivery
US20030180712A1 (en) 2002-03-20 2003-09-25 Biostratum Ab Inhibition of the beta3 subunit of L-type Ca2+ channels
US7169916B2 (en) * 2002-04-01 2007-01-30 Isis Pharmaceuticals, Inc. Chloral-free DCA in oligonucleotide synthesis
MXPA04010092A (en) 2002-04-16 2004-12-13 Genentech Inc Compositions and methods for the diagnosis and treatment of tumor.
EP1501863A4 (en) 2002-05-03 2007-01-24 Sequenom Inc Kinase anchor protein muteins, peptides thereof, and related methods
US7199107B2 (en) 2002-05-23 2007-04-03 Isis Pharmaceuticals, Inc. Antisense modulation of kinesin-like 1 expression
WO2003106617A2 (en) * 2002-06-12 2003-12-24 Tel Aviv Medical Center Research Development Fund Oligonucleotides antibodies and kits including same for treating prostate cancer and determining predisposition thereto
AU2003276131A1 (en) * 2002-06-18 2003-12-31 Epigenesis Pharmaceuticals, Inc. A dry powder oligonucleotide formulation, preparation and its uses
US20050287648A1 (en) 2002-08-05 2005-12-29 University Of Rochester Protein Transducing Domain/Deaminase Chimeric Proteins, Related Compounds, and Uses Thereof
CN1694959B (en) 2002-09-13 2013-09-18 雷普利瑟公司 Non-sequence complementary antiviral oligonucleotides
EP1546170A4 (en) * 2002-09-20 2007-08-29 Univ Yale Riboswitches, methods for their use, and compositions for use with riboswitches
EP1549767A4 (en) 2002-09-26 2006-06-07 Amgen Inc Modulation of forkhead box o1a expression
US9150606B2 (en) 2002-11-05 2015-10-06 Isis Pharmaceuticals, Inc. Compositions comprising alternating 2'-modified nucleosides for 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
CA2504720C (en) 2002-11-05 2013-12-24 Isis Pharmaceuticals, Inc. Chimeric oligomeric compounds and their use in gene modulation
US9150605B2 (en) * 2002-11-05 2015-10-06 Isis Pharmaceuticals, Inc. Compositions comprising alternating 2′-modified nucleosides for use in gene modulation
ES2420914T3 (en) 2002-11-13 2013-08-27 Genzyme Corporation Antisense modulation of apolipoprotein B expression
EP2336318B1 (en) 2002-11-13 2013-04-24 Genzyme Corporation Antisense modulation of apolipoprotein b expression
US20060009378A1 (en) 2002-11-14 2006-01-12 Itshak Golan Novel galectin sequences and compositions and methods utilizing same for treating or diagnosing arthritis and other chronic inflammatory diseases
US7754450B2 (en) * 2002-11-15 2010-07-13 Morphotek, Inc. Methods of generating high-production of antibodies from hybridomas created by in vitro immunization
JP4915980B2 (en) 2002-11-15 2012-04-11 エムユーエスシー ファウンデーション フォー リサーチ デベロップメント Complement receptor 2 targeted complement regulator
EP1624753B1 (en) 2002-11-21 2012-01-25 The University of Utah Research Foundation Purinergic modulation of smell
US7144999B2 (en) 2002-11-23 2006-12-05 Isis Pharmaceuticals, Inc. Modulation of hypoxia-inducible factor 1 alpha expression
US20040121338A1 (en) * 2002-12-19 2004-06-24 Alsmadi Osama A. Real-time detection of rolling circle amplification products
US9487823B2 (en) 2002-12-20 2016-11-08 Qiagen Gmbh Nucleic acid amplification
WO2004058987A2 (en) 2002-12-20 2004-07-15 Qiagen Gmbh Nucleic acid amplification
US6977153B2 (en) * 2002-12-31 2005-12-20 Qiagen Gmbh Rolling circle amplification of RNA
ES2400033T3 (en) 2003-02-11 2013-04-05 Antisense Therapeutics Ltd Modulation of insulin-like growth factor I receptor expression
US7002006B2 (en) * 2003-02-12 2006-02-21 Isis Pharmaceuticals, Inc. Protection of nucleosides
CA2517074A1 (en) 2003-02-27 2004-09-10 Yeda Research And Development Co., Ltd. Nucleic acid molecules, polypeptides, antibodies and compositions containing same useful for treating and detecting influenza virus infection
US7803781B2 (en) 2003-02-28 2010-09-28 Isis Pharmaceuticals, Inc. Modulation of growth hormone receptor expression and insulin-like growth factor expression
US20040185559A1 (en) 2003-03-21 2004-09-23 Isis Pharmaceuticals Inc. Modulation of diacylglycerol acyltransferase 1 expression
US8043834B2 (en) 2003-03-31 2011-10-25 Qiagen Gmbh Universal reagents for rolling circle amplification and methods of use
US7598227B2 (en) 2003-04-16 2009-10-06 Isis Pharmaceuticals Inc. Modulation of apolipoprotein C-III expression
US7399853B2 (en) 2003-04-28 2008-07-15 Isis Pharmaceuticals Modulation of glucagon receptor expression
JP2006525027A (en) 2003-05-01 2006-11-09 ジェン−プロウブ インコーポレイテッド Oligonucleotides containing molecular switches
CA2540692C (en) * 2003-06-02 2013-05-28 Isis Pharmaceuticals, Inc. Oligonucleotide synthesis with alternative solvents
BRPI0410886A (en) 2003-06-03 2006-07-04 Isis Pharmaceuticals Inc double stranded compound, pharmaceutical composition, pharmaceutically acceptable salt, methods of modifying human survivin-encoding nucleic acid, inhibiting suvivin expression in cells or tissues, and treating a condition associated with suvivin expression or overexpression, and single stranded RNA oligonucleotide
EP1633770B1 (en) 2003-06-13 2015-04-29 Alnylam Europe AG Double-stranded ribonucleic acid with increased effectiveness in an organism
CA2533701A1 (en) 2003-07-31 2005-02-17 Isis Pharmaceuticals, Inc. Oligomeric compounds and compositions for use in modulation of small non-coding rnas
US7825235B2 (en) 2003-08-18 2010-11-02 Isis Pharmaceuticals, Inc. Modulation of diacylglycerol acyltransferase 2 expression
US20050053981A1 (en) 2003-09-09 2005-03-10 Swayze Eric E. Gapped oligomeric compounds having linked bicyclic sugar moieties at the termini
US20070123480A1 (en) * 2003-09-11 2007-05-31 Replicor Inc. Oligonucleotides targeting prion diseases
US7425544B2 (en) 2003-09-18 2008-09-16 Eli Lilly And Company Modulation of eIF4E expression
WO2005027962A1 (en) 2003-09-18 2005-03-31 Isis Pharmaceuticals, Inc. 4’-thionucleosides and oligomeric compounds
US7125945B2 (en) * 2003-09-19 2006-10-24 Varian, Inc. Functionalized polymer for oligonucleotide purification
US20050191653A1 (en) 2003-11-03 2005-09-01 Freier Susan M. Modulation of SGLT2 expression
EP1689432B1 (en) 2003-11-17 2009-12-30 Genentech, Inc. Compositions and methods for the treatment of tumor of hematopoietic origin
WO2005071080A2 (en) 2004-01-20 2005-08-04 Isis Pharmaceuticals, Inc. Modulation of glucocorticoid receptor expression
US7468431B2 (en) 2004-01-22 2008-12-23 Isis Pharmaceuticals, Inc. Modulation of eIF4E-BP2 expression
US8778900B2 (en) * 2004-01-22 2014-07-15 Isis Pharmaceuticals, Inc. Modulation of eIF4E-BP1 expression
US7842459B2 (en) 2004-01-27 2010-11-30 Compugen Ltd. Nucleotide and amino acid sequences, and assays and methods of use thereof for diagnosis
US8569474B2 (en) 2004-03-09 2013-10-29 Isis Pharmaceuticals, Inc. Double stranded constructs comprising one or more short strands hybridized to a longer strand
US8790919B2 (en) 2004-03-15 2014-07-29 Isis Pharmaceuticals, Inc. Compositions and methods for optimizing cleavage of RNA by RNase H
US20050244869A1 (en) 2004-04-05 2005-11-03 Brown-Driver Vickie L Modulation of transthyretin expression
US20050267300A1 (en) 2004-04-05 2005-12-01 Muthiah Manoharan Processes and reagents for oligonucleotide synthesis and purification
US20050260755A1 (en) * 2004-04-06 2005-11-24 Isis Pharmaceuticals, Inc. Sequential delivery of oligomeric compounds
AU2005323437B2 (en) 2004-04-30 2011-10-06 Alnylam Pharmaceuticals, Inc. Oligonucleotides comprising a C5-modified pyrimidine
EP3225633B1 (en) 2004-05-21 2020-03-25 The UAB Research Foundation Variable lymphocyte receptors, related polypeptides and nucleic acids, and uses thereof
US8394947B2 (en) 2004-06-03 2013-03-12 Isis Pharmaceuticals, Inc. Positionally modified siRNA constructs
US7884086B2 (en) 2004-09-08 2011-02-08 Isis Pharmaceuticals, Inc. Conjugates for use in hepatocyte free uptake assays
WO2006030442A2 (en) 2004-09-16 2006-03-23 Gamida-Cell Ltd. Methods of ex vivo progenitor and stem cell expansion by co-culture with mesenchymal cells
WO2006032144A1 (en) * 2004-09-23 2006-03-30 Arc Pharmaceuticals, Inc. Pharmaceutical compositions and methods relating to inhibiting fibrous adhesions or inflammatory disease using fucans from various echinoderm sources
US20060105348A1 (en) * 2004-11-15 2006-05-18 Lee Jun E Compositions and methods for the detection and discrimination of nucleic acids
US20060275792A1 (en) * 2004-11-15 2006-12-07 Lee Jun E Enhancement of nucleic acid amplification using double-stranded DNA binding proteins
WO2006050999A2 (en) * 2004-11-15 2006-05-18 Obe Therapy Biotechnology S.A.S Methods of reducing body fat
US20080260637A1 (en) 2004-11-17 2008-10-23 Dalia Dickman Methods of Detecting Prostate Cancer
WO2006086777A2 (en) 2005-02-11 2006-08-17 Memorial Sloan Kettering Cancer Center Methods and compositions for detecting a drug resistant egfr mutant
CN101175769A (en) 2005-03-10 2008-05-07 健泰科生物技术公司 Methods and compositions for modulating vascular integrity
US7476733B2 (en) 2005-03-25 2009-01-13 The United States Of America As Represented By The Department Of Health And Human Services Development of a real-time PCR assay for detection of pneumococcal DNA and diagnosis of pneumococccal disease
US8309303B2 (en) 2005-04-01 2012-11-13 Qiagen Gmbh Reverse transcription and amplification of RNA with simultaneous degradation of DNA
EP1891141B1 (en) 2005-05-31 2016-11-16 Ecole Polytechnique Fédérale de Lausanne (EPFL) Triblock copolymers for cytoplasmic delivery of gene-based drugs
US20090209621A1 (en) 2005-06-03 2009-08-20 The Johns Hopkins University Compositions and methods for decreasing microrna expression for the treatment of neoplasia
WO2006138145A1 (en) 2005-06-14 2006-12-28 Northwestern University Nucleic acid functionalized nanoparticles for therapeutic applications
CA2614531C (en) 2005-07-07 2015-06-16 Avraham Hochberg Nucleic acid agents for downregulating h19, and methods of using same
US8067571B2 (en) 2005-07-13 2011-11-29 Avi Biopharma, Inc. Antibacterial antisense oligonucleotide and method
US7776532B2 (en) 2005-08-11 2010-08-17 Synthetic Genomics, Inc. Method for in vitro recombination
JP2009504168A (en) 2005-08-17 2009-02-05 メディクシス エス.エー. Composition and method for confirming CK19 expression
WO2007027775A2 (en) 2005-08-29 2007-03-08 Isis Pharmaceuticals, Inc. Methods for use in modulating mir-122a
EP1762627A1 (en) 2005-09-09 2007-03-14 Qiagen GmbH Method for the activation of a nucleic acid for performing a polymerase reaction
IL172297A (en) 2005-10-03 2016-03-31 Compugen Ltd Soluble vegfr-1 variants for diagnosis of preeclamsia
EP2392645A1 (en) 2005-10-14 2011-12-07 MUSC Foundation For Research Development Targeting PAX2 for the induction of DEFB1-mediated tumor immunity and cancer therapy
US8080534B2 (en) 2005-10-14 2011-12-20 Phigenix, Inc Targeting PAX2 for the treatment of breast cancer
JP5111385B2 (en) * 2005-10-28 2013-01-09 アルナイラム ファーマシューティカルズ, インコーポレイテッド Composition and method for suppressing expression of huntingtin gene
US7545323B2 (en) * 2005-10-31 2009-06-09 The Boeing Company Phased array antenna systems and methods
AU2006311730B2 (en) 2005-11-09 2010-12-02 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of Factor V Leiden mutant gene
JP2009516710A (en) 2005-11-21 2009-04-23 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 Modulating the expression of eIF4E-BP2
US8846393B2 (en) 2005-11-29 2014-09-30 Gamida-Cell Ltd. Methods of improving stem cell homing and engraftment
JP2009524411A (en) * 2005-12-21 2009-07-02 イェール ユニバーシティー Methods and compositions related to the regulation of riboswitches
JP5713377B2 (en) 2005-12-28 2015-05-07 ザ スクリプス リサーチ インスティテュート Natural antisense and non-coding RNA transcripts as drug targets
US20100184021A1 (en) 2006-01-16 2010-07-22 Compugen Ltd. Novel nucleotide and amino acid sequences, and methods of use thereof for diagnosis
US7569686B1 (en) 2006-01-27 2009-08-04 Isis Pharmaceuticals, Inc. Compounds and methods for synthesis of bicyclic nucleic acid analogs
EP2388328A1 (en) 2006-01-27 2011-11-23 Isis Pharmaceuticals, Inc. Oligomeric compounds and compositions for the use in modulation of micrornas
ES2516815T3 (en) 2006-01-27 2014-10-31 Isis Pharmaceuticals, Inc. Analogs of bicyclic nucleic acids modified at position 6
EP2527354A1 (en) 2006-03-31 2012-11-28 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of Eg5 gene
MX2008014005A (en) 2006-05-03 2009-01-27 Baltic Technology Dev Ltd Antisense agents combining strongly bound base - modified oligonucleotide and artificial nuclease.
US8586554B2 (en) 2006-05-05 2013-11-19 Isis Pharmaceuticals, Inc. Compounds and methods for modulating expression of PTP1B
US7666854B2 (en) * 2006-05-11 2010-02-23 Isis Pharmaceuticals, Inc. Bis-modified bicyclic nucleic acid analogs
DK2066684T3 (en) * 2006-05-11 2012-10-22 Isis Pharmaceuticals Inc 5'-Modified Bicyclic Nucleic Acid Analogs
EP3872179A1 (en) 2006-05-11 2021-09-01 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the pcsk9 gene
CA2652770A1 (en) 2006-05-19 2007-11-29 Alnylam Pharmaceuticals, Inc. Rnai modulation of aha and therapeutic uses thereof
WO2007137220A2 (en) 2006-05-22 2007-11-29 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of ikk-b gene
US20090209625A1 (en) * 2006-05-23 2009-08-20 Sanjay Bhanot Modulation of chrebp expression
US9506056B2 (en) 2006-06-08 2016-11-29 Northwestern University Nucleic acid functionalized nanoparticles for therapeutic applications
US8198253B2 (en) 2006-07-19 2012-06-12 Isis Pharmaceuticals, Inc. Compositions and their uses directed to HBXIP
US20100137440A1 (en) * 2006-09-11 2010-06-03 Yale University Lysine riboswitches, structure-based compound design with lysine riboswitches, and methods and compositions for use of and with lysine riboswitches
AU2007299629C1 (en) 2006-09-21 2012-05-10 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the HAMP gene
KR101129509B1 (en) 2006-10-03 2012-04-13 알닐람 파마슈티칼스 인코포레이티드 Lipid containing formulations
EP2081596A4 (en) 2006-10-06 2010-07-21 Univ Utah Res Found Method of detecting ocular diseases and pathologic conditions and treatment of same
US8999317B2 (en) 2006-11-01 2015-04-07 University Of Rochester Methods and compositions related to the structure and function of APOBEC3G
KR20090089462A (en) 2006-12-11 2009-08-21 유니버시티 오브 유타 리써치 파운데이션 Compositions and methods for treating pathologic angiogenesis and vascular permeability
US20100129358A1 (en) 2006-12-22 2010-05-27 University Of Utah Research Foundation Method of detecting ocular diseases and pathologic conditions and treatment of same
ATE548454T1 (en) * 2007-01-16 2012-03-15 Yissum Res Dev Co NUCLEIC ACID ACTIVE SUBSTANCES FOR STOPPING H19 FOR THE TREATMENT OF RHEUMATOID ARTHRITIS
US20100196403A1 (en) * 2007-01-29 2010-08-05 Jacob Hochman Antibody conjugates for circumventing multi-drug resistance
KR101488800B1 (en) 2007-02-09 2015-02-04 노오쓰웨스턴 유니버시티 Particles for detecting intracellular targets
JP2010520749A (en) * 2007-02-27 2010-06-17 ノースウェスタン ユニバーシティ Binding molecules to nanoparticles
CN101801185A (en) 2007-03-22 2010-08-11 耶鲁大学 Methods and compositions related to riboswitches that control alternative splicing
PE20090064A1 (en) 2007-03-26 2009-03-02 Novartis Ag DOUBLE-CHAIN RIBONUCLEIC ACID TO INHIBIT THE EXPRESSION OF THE HUMAN E6AP GENE AND THE PHARMACEUTICAL COMPOSITION THAT INCLUDES IT
AU2008232891B2 (en) 2007-03-29 2012-01-12 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of a gene from the Ebola
US20110091377A1 (en) 2007-05-11 2011-04-21 The Johns Hopkins University Biomarkers for melanoma
CA2687684A1 (en) * 2007-05-29 2008-12-11 Yale University Methods and compositions related to riboswitches that control alternative splicing and rna processing
SG174103A1 (en) * 2007-05-29 2011-09-29 Univ Yale Riboswitches and methods and compositions for use of and with riboswitches
AU2008260277C1 (en) 2007-05-30 2014-04-17 Isis Pharmaceuticals, Inc. N-substituted-aminomethylene bridged bicyclic nucleic acid analogs
US7807372B2 (en) * 2007-06-04 2010-10-05 Northwestern University Screening sequence selectivity of oligonucleotide-binding molecules using nanoparticle based colorimetric assay
DK2173760T4 (en) 2007-06-08 2016-02-08 Isis Pharmaceuticals Inc Carbocyclic bicyclic nukleinsyreanaloge
EP2176280B2 (en) * 2007-07-05 2015-06-24 Isis Pharmaceuticals, Inc. 6-disubstituted bicyclic nucleic acid analogs
CN103215269B (en) 2007-07-05 2015-01-21 诺瓦提斯公司 Dsrna for treating viral infection
US8088904B2 (en) 2007-08-15 2012-01-03 Isis Pharmaceuticals, Inc. Tetrahydropyran nucleic acid analogs
JP2010538005A (en) 2007-08-28 2010-12-09 ユーエービー リサーチ ファウンデーション Synthetic apolipoprotein E mimetic polypeptides and methods of use
JP2010537638A (en) 2007-08-28 2010-12-09 ユーエービー リサーチ ファウンデーション Synthetic apolipoprotein E mimetic polypeptides and methods of use
PL2769729T3 (en) 2007-09-04 2019-09-30 Compugen Ltd. Polypeptides and polynucleotides, and uses thereof as a drug target for producing drugs and biologics
WO2009039466A1 (en) 2007-09-20 2009-03-26 Vanderbilt University Free solution measurement of molecular interactions by backscattering interferometry
WO2009039442A1 (en) * 2007-09-21 2009-03-26 California Institute Of Technology Nfia in glial fate determination, glioma therapy and astrocytoma treatment
EP2205741A2 (en) 2007-10-02 2010-07-14 Amgen Inc. Increasing erythropoietin using nucleic acids hybridizable to micro-rna and precursors thereof
US8097712B2 (en) 2007-11-07 2012-01-17 Beelogics Inc. Compositions for conferring tolerance to viral disease in social insects, and the use thereof
EP2222851B1 (en) 2007-11-20 2017-06-28 Ionis Pharmaceuticals, Inc. Modulation of cd40 expression
WO2009067647A1 (en) * 2007-11-21 2009-05-28 Isis Pharmaceuticals, Inc. Carbocyclic alpha-l-bicyclic nucleic acid analogs
JP5530933B2 (en) 2007-12-10 2014-06-25 アルナイラム ファーマシューティカルズ, インコーポレイテッド Compositions and methods for inhibiting factor VII gene expression
WO2009086558A1 (en) 2008-01-02 2009-07-09 Tekmira Pharmaceuticals Corporation Improved compositions and methods for the delivery of nucleic acids
US8530640B2 (en) * 2008-02-07 2013-09-10 Isis Pharmaceuticals, Inc. Bicyclic cyclohexitol nucleic acid analogs
EA019531B1 (en) 2008-03-05 2014-04-30 Элнилэм Фармасьютикалз, Инк. COMPOSITIONS AND METHODS FOR INHIBITING EXPRESSION OF Eg5 AND VEGF GENES
WO2009117589A1 (en) 2008-03-21 2009-09-24 Isis Pharmaceuticals, Inc. Oligomeric compounds comprising tricyclic nucleosides and methods for their use
WO2009124238A1 (en) * 2008-04-04 2009-10-08 Isis Pharmaceuticals, Inc. Oligomeric compounds comprising neutrally linked terminal bicyclic nucleosides
AU2009234266B2 (en) 2008-04-11 2015-08-06 Tekmira Pharmaceuticals Corporation Site-specific delivery of nucleic acids by combining targeting ligands with endosomolytic components
JP5685182B2 (en) 2008-04-18 2015-03-18 バクスター・インターナショナル・インコーポレイテッドBaxter International Incorp0Rated Microsphere-based composition for preventing and / or reversing primary autoimmune diabetes
EP2280995A2 (en) * 2008-04-29 2011-02-09 Wyeth LLC Methods for treating inflammation
US8082730B2 (en) * 2008-05-20 2011-12-27 Caterpillar Inc. Engine system having particulate reduction device and method
CA2728467A1 (en) * 2008-07-15 2010-01-21 F. Hoffmann-La Roche Ag Compositions and methods for inhibiting expression of tgf-beta receptor genes
AU2009275387B2 (en) 2008-08-25 2010-07-08 Excaliard Pharmaceuticals, Inc. Antisense oligonucleotides directed against connective tissue growth factor and uses thereof
EP2331690B1 (en) 2008-09-02 2016-01-13 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expression of mutant egfr gene
EP2342616A2 (en) 2008-09-23 2011-07-13 Alnylam Pharmaceuticals Inc. Chemical modifications of monomers and oligonucleotides with cycloaddition
WO2010036696A1 (en) 2008-09-24 2010-04-01 Isis Pharmaceuticals, Inc. Cyclohexenyl nucleic acid analogs
US8501805B2 (en) * 2008-09-24 2013-08-06 Isis Pharmaceuticals, Inc. Substituted alpha-L-bicyclic nucleosides
EP2334793B1 (en) 2008-09-25 2016-04-06 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of serum amyloid a gene
EP2743265B1 (en) 2008-10-09 2017-03-15 Arbutus Biopharma Corporation Improved amino lipids and methods for the delivery of nucleic acids
PL2379084T3 (en) 2008-10-15 2018-04-30 Ionis Pharmaceuticals, Inc. Modulation of factor 11 expression
IL302142B1 (en) 2008-10-20 2024-03-01 Alnylam Pharmaceuticals Inc Compositions and Methods for Inhabiting Expression of TRANSTHYRETIN
EP2447274B1 (en) 2008-10-24 2017-10-04 Ionis Pharmaceuticals, Inc. Oligomeric compounds and methods
US20120059045A1 (en) 2008-10-24 2012-03-08 Isis Pharmaceuticals, Inc. Methods of using oligomeric compounds comprising 2'-substituted nucleosides
MX2011004909A (en) * 2008-11-17 2011-05-30 Hoffmann La Roche Compositions and methods for inhibiting expression of factor vii genes.
DK2365803T3 (en) 2008-11-24 2018-01-22 Univ Northwestern POLYVALENT RNA NANOPARTICLE COMPOSITIONS
WO2010061393A1 (en) 2008-11-30 2010-06-03 Compugen Ltd. He4 variant nucleotide and amino acid sequences, and methods of use thereof
SG171914A1 (en) 2008-12-02 2011-07-28 Chiralgen Ltd Method for the synthesis of phosphorus atom modified nucleic acids
EP2370580B1 (en) 2008-12-04 2019-09-11 CuRNA, Inc. Treatment of sirtuin 1 (sirt1) related diseases by inhibition of natural antisense transcript to sirtuin 1
CN102317458B (en) 2008-12-04 2018-01-02 库尔纳公司 Pass through treatment of the suppression of erythropoietin(EPO) (EPO) natural antisense transcript to EPO relevant diseases
WO2010065787A2 (en) 2008-12-04 2010-06-10 Curna, Inc. Treatment of tumor suppressor gene related diseases by inhibition of natural antisense transcript to the gene
EP2633854B1 (en) 2008-12-05 2015-09-16 Yeda Research And Development Co. Ltd. miRNA-9 or miRNA-9* for use in treating ALS
AU2009324534B2 (en) 2008-12-10 2015-07-30 Alnylam Pharmaceuticals, Inc. GNAQ targeted dsRNA compositions and methods for inhibiting expression
WO2010080554A1 (en) 2008-12-17 2010-07-15 Avi Biopharma, Inc. Antisense compositions and methods for modulating contact hypersensitivity or contact dermatitis
US20100233270A1 (en) 2009-01-08 2010-09-16 Northwestern University Delivery of Oligonucleotide-Functionalized Nanoparticles
WO2010088537A2 (en) 2009-01-29 2010-08-05 Alnylam Pharmaceuticals, Inc. Improved lipid formulation
AU2010211133A1 (en) 2009-02-03 2011-07-21 F. Hoffmann-La Roche Ag Compositions and methods for inhibiting expression of PTP1B genes
WO2010090762A1 (en) 2009-02-04 2010-08-12 Rxi Pharmaceuticals Corporation Rna duplexes with single stranded phosphorothioate nucleotide regions for additional functionality
US8536320B2 (en) 2009-02-06 2013-09-17 Isis Pharmaceuticals, Inc. Tetrahydropyran nucleic acid analogs
JP5766126B2 (en) 2009-02-12 2015-08-19 クルナ・インコーポレーテッド Treatment of brain-derived neurotrophic factor (BDNF) -related diseases by suppression of natural antisense transcripts against BDNF
CA2752239C (en) 2009-02-12 2021-03-30 Opko Curna, Llc Treatment of glial cell derived neurotrophic factor (gdnf) related diseases by inhibition of natural antisense transcript to gdnf
US20120041051A1 (en) 2009-02-26 2012-02-16 Kevin Fitzgerald Compositions And Methods For Inhibiting Expression Of MIG-12 Gene
WO2010101951A1 (en) 2009-03-02 2010-09-10 Alnylam Pharmaceuticals, Inc. Nucleic acid chemical modifications
EP2403946A4 (en) 2009-03-04 2012-11-14 Treatment of sirtuin 1 (sirt1) related diseases by inhibition of natural antisense transcript to sirt 1
US20100267806A1 (en) 2009-03-12 2010-10-21 David Bumcrot LIPID FORMULATED COMPOSITIONS AND METHODS FOR INHIBITING EXPRESSION OF Eg5 AND VEGF GENES
CN102482677B (en) 2009-03-16 2017-10-17 库尔纳公司 Nuclear factor (red blood cell derives 2) sample 2 (NRF2) relevant disease is treated by suppressing NRF2 natural antisense transcript
CN102549159B (en) 2009-03-17 2016-08-10 库尔纳公司 By suppressing to treat the disease that DLK1 is correlated with for the natural antisense transcript of δ sample 1 congener (DLK1)
CA2757694C (en) 2009-04-15 2020-10-06 Northwestern University Delivery of oligonucleotide-functionalized nanoparticles
EP3248618A1 (en) 2009-04-22 2017-11-29 Massachusetts Institute Of Technology Innate immune suppression enables repeated delivery of long rna molecules
DK2424987T3 (en) 2009-05-01 2018-02-26 Curna Inc TREATMENT OF HEMOGLOBIN (HBF / HBG) RELATED DISEASES BY INHIBITION OF NATURAL ANTISENSE TRANSCRIPTION TO HBF / HBG
AU2010245933B2 (en) 2009-05-05 2016-06-16 Arbutus Biopharma Corporation Methods of delivering oligonucleotides to immune cells
NZ596186A (en) 2009-05-05 2014-03-28 Alnylam Pharmaceuticals Inc Lipid compositions
ES2609655T3 (en) 2009-05-06 2017-04-21 Curna, Inc. Treatment of diseases related to tristetraproline (TTP) by inhibition of natural antisense transcript for TTP
WO2010129799A2 (en) 2009-05-06 2010-11-11 Curna, Inc. Treatment of lipid transport and metabolism gene related diseases by inhibition of natural antisense transcript to a lipid transport and metabolism gene
WO2010132665A1 (en) 2009-05-15 2010-11-18 Yale University Gemm riboswitches, structure-based compound design with gemm riboswitches, and methods and compositions for use of and with gemm riboswitches
BRPI1012769A2 (en) * 2009-05-15 2018-01-30 Hoffmann La Roche compositions and methods for inhibiting glucocorticoid receptor (gcr) gene expression
EP2432881B1 (en) 2009-05-18 2017-11-15 CuRNA, Inc. Treatment of reprogramming factor related diseases by inhibition of natural antisense transcript to a reprogramming factor
WO2010135695A2 (en) 2009-05-22 2010-11-25 Curna, Inc. TREATMENT OF TRANSCRIPTION FACTOR E3 (TFE3) and INSULIN RECEPTOR SUBSTRATE 2 (IRS2) RELATED DISEASES BY INHIBITION OF NATURAL ANTISENSE TRANSCRIPT TO TFE3
ES2618576T3 (en) 2009-05-28 2017-06-21 Curna, Inc. Treatment of diseases related to an antiviral gene by inhibiting a natural antisense transcript to an antiviral gene
HUE038796T2 (en) 2009-06-10 2018-11-28 Arbutus Biopharma Corp Improved lipid formulation
JP6128846B2 (en) 2009-06-16 2017-05-17 クルナ・インコーポレーテッド Treatment of PON1 gene-related diseases by suppression of natural antisense transcripts against paraoxonase (PON1)
WO2010148050A2 (en) 2009-06-16 2010-12-23 Curna, Inc. Treatment of collagen gene related diseases by inhibition of natural antisense transcript to a collagen gene
WO2010151671A2 (en) 2009-06-24 2010-12-29 Curna, Inc. Treatment of tumor necrosis factor receptor 2 (tnfr2) related diseases by inhibition of natural antisense transcript to tnfr2
CA2765815A1 (en) 2009-06-26 2010-12-29 Opko Curna, Llc Treatment of down syndrome gene related diseases by inhibition of natural antisense transcript to a down syndrome gene
KR101885383B1 (en) 2009-07-06 2018-08-03 웨이브 라이프 사이언시스 리미티드 Novel nucleic acid prodrugs and methods of use thereof
US8927513B2 (en) 2009-07-07 2015-01-06 Alnylam Pharmaceuticals, Inc. 5′ phosphate mimics
US9512164B2 (en) 2009-07-07 2016-12-06 Alnylam Pharmaceuticals, Inc. Oligonucleotide end caps
CA2769665A1 (en) 2009-08-05 2011-02-10 Opko Curna, Llc Treatment of insulin gene (ins) related diseases by inhibition of natural antisense transcript to an insulin gene (ins)
EP2462153B1 (en) 2009-08-06 2015-07-29 Isis Pharmaceuticals, Inc. Bicyclic cyclohexose nucleic acid analogs
EP2464336A4 (en) 2009-08-14 2013-11-20 Alnylam Pharmaceuticals Inc Lipid formulated compositions and methods for inhibiting expression of a gene from the ebola virus
WO2011022420A1 (en) 2009-08-17 2011-02-24 Yale University Methylation biomarkers and methods of use
KR101892760B1 (en) 2009-08-25 2018-08-28 큐알엔에이, 인크. Treatment of 'iq motif containing gtpase activating protein' (iqgap) related diseases by inhibition of natural antisense transcript to iqgap
AU2010289400B2 (en) 2009-09-02 2014-10-23 Curis, Inc. Mutant smoothened and methods of using the same
US8962584B2 (en) 2009-10-14 2015-02-24 Yissum Research Development Company Of The Hebrew University Of Jerusalem, Ltd. Compositions for controlling Varroa mites in bees
CN105368836A (en) 2009-10-14 2016-03-02 耶路撒冷希伯来大学伊森姆研究发展公司 Compositions for controlling varroa mites in bees
CA2778442A1 (en) 2009-10-22 2011-04-28 Genentech, Inc. Methods and compositions for modulating hepsin activation of macrophage-stimulating protein
JP6147502B2 (en) 2009-10-27 2017-06-14 スウィフト バイオサイエンシーズ, インコーポレイテッド Polynucleotide primers and probes
US9376690B2 (en) 2009-10-30 2016-06-28 Northwestern University Templated nanoconjugates
KR20120102674A (en) 2009-11-03 2012-09-18 유니버시티 오브 버지니아 페이턴트 파운데이션 Versatile, visible method for detecting polymeric analytes
AR078921A1 (en) * 2009-11-09 2011-12-14 Hoffmann La Roche COMPOSITIONS AND METHODS TO INHIBIT THE EXPRESSION OF GENES OF THE QUINESINAS SUPERFAMILY, KIF10
WO2011058555A1 (en) 2009-11-12 2011-05-19 Yeda Research And Development Co. Ltd. A method of editing dna in a cell and constructs capable of same
CN102712928B (en) 2009-11-13 2017-08-04 萨雷普塔治疗公司 Antisense antiviral compound and the method for treating influenza infection
CA2781469A1 (en) 2009-11-23 2011-05-26 Swift Biosciences, Inc. Devices to extend single stranded target molecules
CN102741294A (en) 2009-11-30 2012-10-17 霍夫曼-拉罗奇有限公司 Antibodies for treating and diagnosing tumors expressing SLC34A2 (TAT211 = SEQID 2)
NO2513310T3 (en) 2009-12-16 2018-03-31
US20110152349A1 (en) 2009-12-18 2011-06-23 Anke Geick Compositions and methods for inhibiting expression of il-18 genes
CN102869776B (en) 2009-12-23 2017-06-23 库尔纳公司 HGF relevant diseases are treated by suppressing the natural antisense transcript of HGF (HGF)
US9068183B2 (en) 2009-12-23 2015-06-30 Curna, Inc. Treatment of uncoupling protein 2 (UCP2) related diseases by inhibition of natural antisense transcript to UCP2
CA2785173A1 (en) 2009-12-29 2011-07-28 Curna, Inc. Treatment of nuclear respiratory factor 1 (nrf1) related diseases by inhibition of natural antisense transcript to nrf1
KR101853508B1 (en) 2009-12-29 2018-06-20 큐알엔에이, 인크. TREATMENT OF TUMOR PROTEIN 63 (p63) RELATED DISEASES BY INHIBITION OF NATURAL ANTISENSE TRANSCRIPT TO p63
US8946181B2 (en) 2010-01-04 2015-02-03 Curna, Inc. Treatment of interferon regulatory factor 8 (IRF8) related diseases by inhibition of natural antisense transcript to IRF8
RU2612161C2 (en) 2010-01-06 2017-03-02 Курна, Инк. Treatment of pancreatic developmental gene related diseases by inhibition of natural antisense transcript to pancreatic developmental gene
WO2011085102A1 (en) 2010-01-11 2011-07-14 Isis Pharmaceuticals, Inc. Base modified bicyclic nucleosides and oligomeric compounds prepared therefrom
WO2011085347A2 (en) 2010-01-11 2011-07-14 Opko Curna, Llc Treatment of sex hormone binding globulin (shbg) related diseases by inhibition of natural antisense transcript to shbg
WO2011088076A2 (en) 2010-01-12 2011-07-21 Yale University Structured rna motifs and compounds and methods for their use
CN102782135A (en) 2010-01-25 2012-11-14 库尔纳公司 Treatment of RNase H1 related diseases by inhibition of natural antisense transcript to RNase H1
WO2011100131A2 (en) 2010-01-28 2011-08-18 Alnylam Pharmacuticals, Inc. Monomers and oligonucleotides comprising cycloaddition adduct(s)
WO2011094580A2 (en) 2010-01-28 2011-08-04 Alnylam Pharmaceuticals, Inc. Chelated copper for use in the preparation of conjugated oligonucleotides
CA2824843A1 (en) 2010-02-04 2011-08-11 Ico Therapeutics Inc. Dosing regimens for treating and preventing ocular disorders using c-raf antisense
TW201129365A (en) 2010-02-05 2011-09-01 Hoffmann La Roche Compositions and methods for inhibiting expression of IKK2 genes
CA2790506A1 (en) 2010-02-22 2011-08-25 Curna, Inc. Treatment of pyrroline-5-carboxylate reductase 1 (pycr1) related diseases by inhibition of natural antisense transcript to pycr1
PE20130214A1 (en) 2010-02-23 2013-03-11 Genentech Inc COMPOSITIONS AND METHODS FOR THE DIAGNOSIS AND TREATMENT OF TUMORS
WO2011105901A2 (en) 2010-02-23 2011-09-01 Academisch Ziekenhuis Bij De Universiteit Van Amsterdam Antagonists of complement component 9 (c9) and uses thereof
WO2011105902A2 (en) 2010-02-23 2011-09-01 Academisch Ziekenhuis Bij De Universiteit Van Amsterdam Antagonists of complement component 8-beta (c8-beta) and uses thereof
WO2011105900A2 (en) 2010-02-23 2011-09-01 Academisch Ziekenhuis Bij De Universiteit Van Amsterdam Antagonists of complement component 8-alpha (c8-alpha) and uses thereof
MY163887A (en) 2010-03-08 2017-11-15 Monsanto Technology Llc Polynucleotide molecules for gene regulations in plants
WO2011112516A1 (en) 2010-03-08 2011-09-15 Ico Therapeutics Inc. Treating and preventing hepatitis c virus infection using c-raf kinase antisense oligonucleotides
WO2011113054A2 (en) 2010-03-12 2011-09-15 Aurasense Llc Crosslinked polynucleotide structure
ES2743600T3 (en) 2010-03-12 2020-02-20 Brigham & Womens Hospital Inc Methods of treatment of vascular inflammatory disorders
EP2545173A2 (en) 2010-03-12 2013-01-16 Sarepta Therapeutics, Inc. Antisense modulation of nuclear hormone receptors
WO2011115818A1 (en) 2010-03-17 2011-09-22 Isis Pharmaceuticals, Inc. 5'-substituted bicyclic nucleosides and oligomeric compounds prepared therefrom
WO2011120046A2 (en) 2010-03-26 2011-09-29 Swift Biosciences, Inc. Methods and compositions for isolating polynucleotides
AU2011235276B2 (en) 2010-03-29 2015-09-03 Alnylam Pharmaceuticals, Inc. SiRNA therapy for transthyretin (TTR) related ocular amyloidosis
WO2011123621A2 (en) 2010-04-01 2011-10-06 Alnylam Pharmaceuticals Inc. 2' and 5' modified monomers and oligonucleotides
TWI644675B (en) 2010-04-09 2018-12-21 可娜公司 Treatment of fibroblast growth factor 21 (fgf21) related diseases by inhibition of natural antisense transcript to fgf21
US20110269194A1 (en) 2010-04-20 2011-11-03 Swift Biosciences, Inc. Materials and methods for nucleic acid fractionation by solid phase entrapment and enzyme-mediated detachment
US20130260460A1 (en) 2010-04-22 2013-10-03 Isis Pharmaceuticals Inc Conformationally restricted dinucleotide monomers and oligonucleotides
WO2011133876A2 (en) 2010-04-22 2011-10-27 Alnylam Pharmaceuticals, Inc. Oligonucleotides comprising acyclic and abasic nucleosides and analogs
WO2011133871A2 (en) 2010-04-22 2011-10-27 Alnylam Pharmaceuticals, Inc. 5'-end derivatives
US9127033B2 (en) 2010-04-28 2015-09-08 Isis Pharmaceuticals, Inc. 5′ modified nucleosides and oligomeric compounds prepared therefrom
EP3173419A1 (en) 2010-04-28 2017-05-31 Ionis Pharmaceuticals, Inc. Modified nucleosides, analogs thereof and oligomeric compounds prepared therefrom
MX343559B (en) 2010-04-29 2016-11-10 Ionis Pharmaceuticals Inc Modulation of transthyretin expression.
MX342239B (en) 2010-05-03 2016-09-21 Genentech Inc * Compositions and methods for the diagnosis and treatment of tumor.
CN107988228B (en) 2010-05-03 2022-01-25 库尔纳公司 Treatment of Sirtuin (SIRT) related diseases by inhibition of natural antisense transcript to Sirtuin (SIRT)
EP2569431B1 (en) 2010-05-13 2015-09-23 Sarepta Therapeutics, Inc. Methods for identifying compounds which modulate interleukins 17 and 23 signaling activity
TWI531370B (en) 2010-05-14 2016-05-01 可娜公司 Treatment of par4 related diseases by inhibition of natural antisense transcript to par4
US20130203045A1 (en) 2010-05-26 2013-08-08 University Of Virginia Patent Foundation Method for detecting nucleic acids based on aggregate formation
DK2576783T3 (en) 2010-05-26 2018-03-12 Curna Inc TREATMENT OF ATONAL HOMOLOGY 1- (ATOH1) RELATED DISEASES BY INHIBITION OF NATURAL ANTISENCE TRANSCRIPTS AT ATOH1
KR101981705B1 (en) 2010-05-28 2019-05-24 사렙타 쎄러퓨틱스, 인코퍼레이티드 Oligonucleotide analogues having modified intersubunit linkages and/or terminal groups
EP2576579B1 (en) 2010-06-02 2018-08-08 Alnylam Pharmaceuticals, Inc. Compositions and methods directed to treating liver fibrosis
WO2011156278A1 (en) 2010-06-07 2011-12-15 Isis Pharmaceuticals, Inc. Bicyclic nucleosides and oligomeric compounds prepared therefrom
WO2011156202A1 (en) 2010-06-08 2011-12-15 Isis Pharmaceuticals, Inc. Substituted 2 '-amino and 2 '-thio-bicyclic nucleosides and oligomeric compounds prepared therefrom
US9638632B2 (en) 2010-06-11 2017-05-02 Vanderbilt University Multiplexed interferometric detection system and method
WO2011163466A1 (en) 2010-06-23 2011-12-29 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Regulation of skin pigmentation by neuregulin-1 (nrg-1)
CA2805024A1 (en) 2010-06-30 2012-01-05 Compugen Ltd. Polypeptides and uses thereof as a drug for treatment of multiples sclerosis, rheumatoid arthritis and other autoimmune disorders
US20130143955A1 (en) 2010-08-09 2013-06-06 Yale University Cyclic di-GMP-II Riboswitches, Motifs, and Compounds, and Methods for Their Use
WO2012031243A2 (en) 2010-09-03 2012-03-08 Avi Biopharma, Inc. dsRNA MOLECULES COMPRISING OLIGONUCLEOTIDE ANALOGS HAVING MODIFIED INTERSUBUNIT LINKAGES AND/OR TERMINAL GROUPS
US20130210901A1 (en) 2010-09-20 2013-08-15 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Method of treating neurodegenerative diseases
JP5868324B2 (en) 2010-09-24 2016-02-24 株式会社Wave Life Sciences Japan Asymmetric auxiliary group
WO2012047968A2 (en) 2010-10-05 2012-04-12 Genentech, Inc. Mutant smoothened and methods of using the same
RU2624048C2 (en) 2010-10-06 2017-06-30 Курна, Инк. Treatment of diseases associated with the sialidase 4 (neu4) gene, by gene neu4 natural antisense transcript inhibition
CN103517990A (en) 2010-10-07 2014-01-15 通用医疗公司 Biomarkers of cancer
US8951983B2 (en) 2010-10-17 2015-02-10 Yeda Research And Development Co. Ltd. Methods and compositions for the treatment of insulin-associated medical conditions
EP3434772A3 (en) 2010-10-18 2019-03-20 Arrowhead Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of rrm2 genes
JP6049623B2 (en) 2010-10-22 2016-12-21 カッパーアールエヌエー,インコーポレイテッド Treatment of IDUA-related diseases by inhibition of natural antisense transcripts to α-L-iduronidase (IDUA)
KR101913232B1 (en) 2010-10-27 2018-10-30 큐알엔에이, 인크. Treatment of interferon-related developmental regulator 1(ifrd1) related diseases by inhibition of natural antisense transcript to ifrd1
CN110123830A (en) 2010-11-09 2019-08-16 阿尔尼拉姆医药品有限公司 Composition and method for inhibiting the lipid of the expression of Eg5 and VEGF gene to prepare
DK2638163T3 (en) 2010-11-12 2017-07-24 Massachusetts Gen Hospital POLYCOMB-ASSOCIATED NON-CODING RNAs
CA3077910A1 (en) 2010-11-17 2012-05-24 Ionis Pharmaceuticals, Inc. Modulation of alpha synuclein expression
JP6071893B2 (en) 2010-11-23 2017-02-01 カッパーアールエヌエー,インコーポレイテッド Treatment of NANOG-related diseases by inhibition of natural antisense transcripts to NANOG
US9150926B2 (en) 2010-12-06 2015-10-06 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Diagnosis and treatment of adrenocortical tumors using human microRNA-483
US9127275B2 (en) 2010-12-10 2015-09-08 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of klf-1 and bcl11a genes
US9193973B2 (en) 2010-12-10 2015-11-24 Alynylam Pharmaceuticals, Inc. Compositions and methods for increasing erythropoietin (EPO) production
US9045749B2 (en) 2011-01-14 2015-06-02 The General Hospital Corporation Methods targeting miR-128 for regulating cholesterol/lipid metabolism
MX365647B (en) 2011-02-02 2019-06-10 Excaliard Pharmaceuticals Inc Method of treating keloids or hypertrophic scars using antisense compounds targeting connective tissue growth factor (ctgf).
EP2670404B1 (en) 2011-02-02 2018-08-29 The Trustees of Princeton University Sirtuin modulators as virus production modulators
US9562853B2 (en) 2011-02-22 2017-02-07 Vanderbilt University Nonaqueous backscattering interferometric methods
KR102365961B1 (en) 2011-03-29 2022-02-23 알닐람 파마슈티칼스 인코포레이티드 Compositions and methods for inhibiting expression of tmprss6 gene
EP2694660B1 (en) 2011-04-03 2018-08-08 The General Hospital Corporation Efficient protein expression in vivo using modified rna (mod-rna)
WO2012140627A1 (en) 2011-04-15 2012-10-18 Compugen Ltd. Polypeptides and polynucleotides, and uses thereof for treatment of immune related disorders and cancer
WO2012149154A1 (en) 2011-04-26 2012-11-01 Swift Biosciences, Inc. Polynucleotide primers and probes
WO2012151268A1 (en) 2011-05-02 2012-11-08 University Of Virginia Patent Foundation Method and system for high throughput optical and label free detection of analytes
WO2012151289A2 (en) 2011-05-02 2012-11-08 University Of Virginia Patent Foundation Method and system to detect aggregate formation on a substrate
CN103620036B (en) 2011-06-09 2016-12-21 库尔纳公司 FXN relevant disease is treated by the natural antisense transcript of suppression Frataxin (FXN)
WO2012170347A1 (en) 2011-06-09 2012-12-13 Isis Pharmaceuticals, Inc. Bicyclic nucleosides and oligomeric compounds prepared therefrom
EP2723351B1 (en) 2011-06-21 2018-02-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibition of expression of protein c (proc) genes
CA3191066A1 (en) 2011-06-21 2012-12-27 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibition of expression of apolipoprotein c-iii (apoc3) genes
AU2012272970A1 (en) 2011-06-21 2014-02-06 Alnylam Pharmaceuticals, Inc. Angiopoietin-like 3 (ANGPTL3) iRNA compositions and methods of use thereof
EP2723390B1 (en) 2011-06-23 2017-12-27 Alnylam Pharmaceuticals, Inc. Serpina1 sirnas: compositions of matter and methods of treatment
US20140227293A1 (en) 2011-06-30 2014-08-14 Trustees Of Boston University Method for controlling tumor growth, angiogenesis and metastasis using immunoglobulin containing and proline rich receptor-1 (igpr-1)
WO2013001517A1 (en) 2011-06-30 2013-01-03 Compugen Ltd. Polypeptides and uses thereof for treatment of autoimmune disorders and infection
CN103635576B (en) 2011-06-30 2016-09-21 箭头研究公司 For suppressing compositions and the method for the gene expression of hepatitis B virus
DK2734208T3 (en) 2011-07-19 2017-06-19 Wave Life Sciences Ltd PROCEDURES FOR SYNTHESIS OF FUNCTIONALIZED NUCLEIC ACIDS
JP2014526887A (en) 2011-08-01 2014-10-09 アルナイラム ファーマシューティカルズ, インコーポレイテッド How to improve the success rate of hematopoietic stem cell transplantation
CA2844012C (en) 2011-08-04 2021-03-16 Yeda Research And Development Co. Ltd. Micro-rnas and compositions comprising same for the treatment and diagnosis of serotonin-, adrenalin-, noradrenalin-, glutamate-, and corticotropin-releasing hormone- associated medical conditions
US9840715B1 (en) 2011-09-13 2017-12-12 Monsanto Technology Llc Methods and compositions for delaying senescence and improving disease tolerance and yield in plants
BR112014005958A2 (en) 2011-09-13 2020-10-13 Monsanto Technology Llc agricultural chemical methods and compositions for plant control, method of reducing expression of an accase gene in a plant, microbial expression cassette, method for making a polynucleotide, method of identifying polynucleotides useful in modulating expression of the accase gene and herbicidal composition
WO2013040057A1 (en) 2011-09-13 2013-03-21 Monsanto Technology Llc Methods and compositions for weed control
US10806146B2 (en) 2011-09-13 2020-10-20 Monsanto Technology Llc Methods and compositions for weed control
CA2848685A1 (en) 2011-09-13 2013-03-21 Monsanto Technology Llc Methods and compositions for weed control comprising topical application of a glutamine synthetase polynucleotide
UY34328A (en) 2011-09-13 2013-04-30 Monsanto Technology Llc ? COMPOSITIONS AND METHODS TO CONTROL MALEZAS UNDERSTANDING A POLINUCLEOTIDE AND TRANSFER AGENT, AND THAT MODULATE PROTOPORPHYRINOGEN IX OXIDASE.
US10760086B2 (en) 2011-09-13 2020-09-01 Monsanto Technology Llc Methods and compositions for weed control
US10829828B2 (en) 2011-09-13 2020-11-10 Monsanto Technology Llc Methods and compositions for weed control
WO2013040499A1 (en) 2011-09-14 2013-03-21 Northwestern University Nanoconjugates able to cross the blood-brain barrier
DK2756080T3 (en) 2011-09-14 2019-05-20 Translate Bio Ma Inc MULTIMERIC OILONCLEOTID CONNECTIONS
US9920326B1 (en) 2011-09-14 2018-03-20 Monsanto Technology Llc Methods and compositions for increasing invertase activity in plants
US9580713B2 (en) 2011-09-17 2017-02-28 Yale University Fluoride-responsive riboswitches, fluoride transporters, and methods of use
EP2766482B1 (en) 2011-10-11 2016-12-07 The Brigham and Women's Hospital, Inc. Micrornas in neurodegenerative disorders
RS57645B1 (en) 2011-10-14 2018-11-30 Hoffmann La Roche Anti-htra1 antibodies and methods of use
EP2771464B1 (en) 2011-10-27 2018-03-21 Yeda Research and Development Co. Ltd. Methods of treating cancer
WO2013074834A1 (en) 2011-11-18 2013-05-23 Sarepta Therapeutics, Inc. Functionally-modified oligonucleotides and subunits thereof
WO2013090457A2 (en) 2011-12-12 2013-06-20 Oncoimmunin Inc. In vivo delivery of oligonucleotides
WO2013106358A1 (en) 2012-01-10 2013-07-18 Hussain M Mahmood Method of treating hyperlipidemia and atherosclerosis with mir-30c
SG2014008304A (en) 2012-02-01 2014-06-27 Compugen Ltd C10rf32 antibodies, and uses thereof for treatment of cancer
WO2013121426A1 (en) 2012-02-13 2013-08-22 Gamida-Cell Ltd. Culturing of mesenchymal stem cells
WO2013124816A2 (en) 2012-02-22 2013-08-29 Brainstem Biotec Ltd. Generation of neural stem cells and motor neurons
EP3401393B1 (en) 2012-02-22 2020-02-19 Exostem Biotec Ltd Micrornas for the generation of astrocytes
JP6509723B2 (en) 2012-03-13 2019-05-08 スウィフト バイオサイエンシーズ, インコーポレイテッド Methods and compositions for size-controlled homopolymeric tailing of substrate polynucleotides by nucleic acid polymerase
ES2694592T3 (en) 2012-03-15 2018-12-21 Curna, Inc. Treatment of diseases related to brain-derived neurotrophic factor (BDNF) by inhibition of the natural antisense transcript of BDNF
EP2639238A1 (en) 2012-03-15 2013-09-18 Universität Bern Tricyclic nucleosides and oligomeric compounds prepared therefrom
US9221864B2 (en) 2012-04-09 2015-12-29 Isis Pharmaceuticals, Inc. Tricyclic nucleic acid analogs
WO2013154799A1 (en) 2012-04-09 2013-10-17 Isis Pharmaceuticals, Inc. Tricyclic nucleosides and oligomeric compounds prepared therefrom
US9133461B2 (en) 2012-04-10 2015-09-15 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the ALAS1 gene
CN107083385A (en) 2012-04-20 2017-08-22 艾珀特玛治疗公司 The miRNA conditioning agents of heat production
US9127274B2 (en) 2012-04-26 2015-09-08 Alnylam Pharmaceuticals, Inc. Serpinc1 iRNA compositions and methods of use thereof
EP3597741A1 (en) 2012-04-27 2020-01-22 Duke University Genetic correction of mutated genes
US9273949B2 (en) 2012-05-11 2016-03-01 Vanderbilt University Backscattering interferometric methods
US10059941B2 (en) 2012-05-16 2018-08-28 Translate Bio Ma, Inc. Compositions and methods for modulating SMN gene family expression
CA2873809A1 (en) 2012-05-16 2013-11-21 Rana Therapeutics, Inc. Compositions and methods for modulating gene expression
MX360866B (en) 2012-05-24 2018-11-09 A B Seeds Ltd Compositions and methods for silencing gene expression.
WO2013184209A1 (en) 2012-06-04 2013-12-12 Ludwig Institute For Cancer Research Ltd. Mif for use in methods of treating subjects with a neurodegenerative disorder
KR101850319B1 (en) 2012-07-13 2018-04-20 웨이브 라이프 사이언시스 리미티드 Asymmetric auxiliary group
US20140038182A1 (en) 2012-07-17 2014-02-06 Dna Logix, Inc. Cooperative primers, probes, and applications thereof
US9175266B2 (en) 2012-07-23 2015-11-03 Gamida Cell Ltd. Enhancement of natural killer (NK) cell proliferation and activity
US9567569B2 (en) 2012-07-23 2017-02-14 Gamida Cell Ltd. Methods of culturing and expanding mesenchymal stem cells
WO2014022852A1 (en) 2012-08-03 2014-02-06 Aptamir Therapeutics, Inc. Cell-specific delivery of mirna modulators for the treatment of obesity and related disorders
US9403865B2 (en) 2012-08-15 2016-08-02 Ionis Pharmaceuticals, Inc. Method of preparing oligomeric compounds using modified capping protocols
AU2013306006B2 (en) 2012-08-20 2017-07-06 The Regents Of The University Of California Polynucleotides having bioreversible groups
US9029335B2 (en) 2012-10-16 2015-05-12 Isis Pharmaceuticals, Inc. Substituted 2′-thio-bicyclic nucleosides and oligomeric compounds prepared therefrom
US10077451B2 (en) 2012-10-18 2018-09-18 Monsanto Technology Llc Methods and compositions for plant pest control
AU2013337277B2 (en) 2012-11-05 2018-03-08 Foundation Medicine, Inc. Novel NTRK1 fusion molecules and uses thereof
WO2014106838A2 (en) 2013-01-01 2014-07-10 A.B. Seeds Ltd. Methods of introducing dsrna to plant seeds for modulating gene expression
US10683505B2 (en) 2013-01-01 2020-06-16 Monsanto Technology Llc Methods of introducing dsRNA to plant seeds for modulating gene expression
EP3939614A1 (en) 2013-01-18 2022-01-19 Foundation Medicine, Inc. Methods of treating cholangiocarcinoma
US10000767B2 (en) 2013-01-28 2018-06-19 Monsanto Technology Llc Methods and compositions for plant pest control
WO2014120861A2 (en) 2013-01-31 2014-08-07 Isis Pharmaceuticals, Inc. Method of preparing oligomeric compounds using modified coupling protocols
US20150366890A1 (en) 2013-02-25 2015-12-24 Trustees Of Boston University Compositions and methods for treating fungal infections
AU2014249015B2 (en) 2013-03-13 2020-04-16 Monsanto Technology Llc Methods and compositions for weed control
US10612019B2 (en) 2013-03-13 2020-04-07 Monsanto Technology Llc Methods and compositions for weed control
US20140283211A1 (en) 2013-03-14 2014-09-18 Monsanto Technology Llc Methods and Compositions for Plant Pest Control
PT2970974T (en) 2013-03-14 2017-11-29 Alnylam Pharmaceuticals Inc Complement component c5 irna compositions and methods of use thereof
US10568328B2 (en) 2013-03-15 2020-02-25 Monsanto Technology Llc Methods and compositions for weed control
EP4286517A3 (en) 2013-04-04 2024-03-13 President and Fellows of Harvard College Therapeutic uses of genome editing with crispr/cas systems
RU2686080C2 (en) 2013-05-01 2019-04-24 Ионис Фармасьютикалз, Инк. Compositions and methods
SG10201804472YA (en) 2013-05-22 2018-07-30 Alnylam Pharmaceuticals Inc SERPINA1 iRNA COMPOSITIONS AND METHODS OF USE THEREOF
SG11201510565TA (en) 2013-05-22 2016-01-28 Alnylam Pharmaceuticals Inc Tmprss6 irna compositions and methods of use thereof
EP3004396B1 (en) 2013-06-06 2019-10-16 The General Hospital Corporation Compositions for the treatment of cancer
ES2862125T3 (en) 2013-06-13 2021-10-07 Antisense Therapeutics Ltd Combination therapy for acromegaly
BR112016000555B1 (en) 2013-07-19 2022-12-27 Monsanto Technology Llc METHOD FOR CONTROLLING AN INFESTATION OF THE LEPTINOTARSA SPECIES IN A PLANT, INSECTICIDAL COMPOSITION AND CONSTRUCTION OF RECOMBINANT DNA
US9850496B2 (en) 2013-07-19 2017-12-26 Monsanto Technology Llc Compositions and methods for controlling Leptinotarsa
CN105579582A (en) 2013-07-25 2016-05-11 埃克西奎雷股份有限公司 Spherical nucleic acid-based constructs as immunostimulatory agents for prophylactic and therapeutic use
HUE048710T2 (en) 2013-08-08 2020-08-28 Scripps Research Inst A method for the site-specific enzymatic labelling of nucleic acids in vitro by incorporation of unnatural nucleotides
US10077444B2 (en) 2013-10-02 2018-09-18 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the LECT2 gene
US10519446B2 (en) 2013-10-04 2019-12-31 Novartis Ag Organic compounds to treat hepatitis B virus
WO2015051366A2 (en) 2013-10-04 2015-04-09 Novartis Ag Novel formats for organic compounds for use in rna interference
LT3052628T (en) 2013-10-04 2020-09-10 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the alas1 gene
KR102298475B1 (en) 2013-10-04 2021-09-06 노파르티스 아게 3' END CAPS FOR RNAi AGENTS FOR USE IN RNA INTERFERENCE
EP3055426B1 (en) 2013-10-09 2019-06-19 The United States of America as represented by The Secretary Department of Health and Human Services Detection of hepatitis delta virus (hdv) for the diagnosis and treatment of sjögren's syndrome and lymphoma
US11162096B2 (en) 2013-10-14 2021-11-02 Ionis Pharmaceuticals, Inc Methods for modulating expression of C9ORF72 antisense transcript
WO2015061246A1 (en) 2013-10-21 2015-04-30 Isis Pharmaceuticals, Inc. Method for solution phase detritylation of oligomeric compounds
EP3060680B1 (en) 2013-10-21 2019-02-27 The General Hospital Corporation Methods relating to circulating tumor cell clusters and the treatment of cancer
WO2015066708A1 (en) 2013-11-04 2015-05-07 Northwestern University Quantification and spatio-temporal tracking of a target using a spherical nucleic acid (sna)
CA2929533C (en) 2013-11-04 2023-06-06 Monsanto Technology Llc Compositions and methods for controlling arthropod parasite and pest infestations
US10182988B2 (en) 2013-12-03 2019-01-22 Northwestern University Liposomal particles, methods of making same and uses thereof
CA2844640A1 (en) 2013-12-06 2015-06-06 The University Of British Columbia Method for treatment of castration-resistant prostate cancer
US10385388B2 (en) 2013-12-06 2019-08-20 Swift Biosciences, Inc. Cleavable competitor polynucleotides
UA119253C2 (en) 2013-12-10 2019-05-27 Біолоджикс, Інк. Compositions and methods for virus control in varroa mite and bees
EP3798306A1 (en) 2013-12-12 2021-03-31 Alnylam Pharmaceuticals, Inc. Complement component irna compositions and methods of use thereof
US10900083B2 (en) 2013-12-20 2021-01-26 The General Hospital Corporation Methods and assays relating to circulating tumor cells
MX368629B (en) 2014-01-15 2019-10-08 Monsanto Technology Llc Methods and compositions for weed control using epsps polynucleotides.
SG10201912897UA (en) 2014-01-16 2020-02-27 Wave Life Sciences Ltd Chiral design
JP6736467B2 (en) 2014-02-04 2020-08-05 ジェネンテック, インコーポレイテッド Smoothing mutant and method of using the same
WO2015118537A2 (en) 2014-02-05 2015-08-13 Yeda Research And Development Co. Ltd. Micro-rnas and compositions comprising same for the treatment and diagnosis of serotonin-, adrenalin-, noradrenalin-, glutamate-, and corticotropin-releasing hormone- associated medical conditions
KR20230152154A (en) 2014-02-11 2023-11-02 알닐람 파마슈티칼스 인코포레이티드 KETOHEXOKINASE (KHK) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
US10036019B2 (en) 2014-03-17 2018-07-31 Ionis Pharmaceuticals, Inc. Bicyclic carbocyclic nucleosides and oligomeric compounds prepared therefrom
SG11201607761UA (en) 2014-03-19 2016-10-28 Ionis Pharmaceuticals Inc Compositions for modulating ataxin 2 expression
WO2015143245A1 (en) 2014-03-19 2015-09-24 Isis Pharmaceuticals, Inc. Methods for modulating ataxin 2 expression
PT3757214T (en) 2014-04-01 2022-08-26 Biogen Ma Inc Compositions for modulating sod-1 expression
BR112016022711A2 (en) 2014-04-01 2017-10-31 Monsanto Technology Llc compositions and methods for insect pest control
EP3943607A1 (en) 2014-04-09 2022-01-26 The Scripps Research Institute Import of unnatural or modified nucleoside triphosphates into cells via nucleic acid triphosphate transporters
WO2015164693A1 (en) 2014-04-24 2015-10-29 Isis Pharmaceuticals, Inc. Oligomeric compounds comprising alpha-beta-constrained nucleic acid
PE20170010A1 (en) 2014-05-01 2017-03-04 Ionis Pharmaceuticals Inc COMPOSITIONS AND METHODS TO MODULATE THE EXPRESSION OF THE COMPLEMENT FACTOR B
EP3137115B1 (en) 2014-05-01 2020-10-14 Ionis Pharmaceuticals, Inc. Method for synthesis of reactive conjugate clusters
TW201607559A (en) 2014-05-12 2016-03-01 阿尼拉製藥公司 Methods and compositions for treating a SERPINC1-associated disorder
SG10202104570TA (en) 2014-05-22 2021-06-29 Alnylam Pharmaceuticals Inc Angiotensinogen (agt) irna compositions and methods of use thereof
AU2015270925A1 (en) 2014-06-02 2016-12-22 Children's Medical Center Corporation Methods and compositions for immunomodulation
EP3155100B1 (en) 2014-06-10 2021-12-22 Erasmus University Medical Center Rotterdam Antisense oligonucleotides useful in treatment of pompe disease
CA2953347A1 (en) 2014-06-23 2015-12-30 Monsanto Technology Llc Compositions and methods for regulating gene expression via rna interference
WO2015200697A1 (en) 2014-06-25 2015-12-30 The General Hospital Corporation Targeting human satellite ii (hsatii)
EP3161138A4 (en) 2014-06-25 2017-12-06 Monsanto Technology LLC Methods and compositions for delivering nucleic acids to plant cells and regulating gene expression
EP3169310A1 (en) 2014-07-15 2017-05-24 Life Technologies Corporation Compositions with lipid aggregates and methods for efficient delivery of molecules to cells
ES2799404T3 (en) 2014-07-15 2020-12-17 Yissum Research And Development Company Of The Hebrew Univ Of Jerusalem Ltd Isolated CD44 polypeptides and uses thereof
US9951327B1 (en) 2014-07-17 2018-04-24 Integrated Dna Technologies, Inc. Efficient and rapid method for assembling and cloning double-stranded DNA fragments
RU2021123470A (en) 2014-07-29 2021-09-06 Монсанто Текнолоджи Ллс COMPOSITIONS AND METHODS FOR COMBATING PESTS
JP7081923B2 (en) 2014-07-31 2022-06-07 ユーエイビー リサーチ ファンデーション Higher effectiveness for removing apoE mimetic peptides and plasma cholesterol
CA2958431A1 (en) 2014-08-19 2016-02-25 Northwestern University Protein/oligonucleotide core-shell nanoparticle therapeutics
WO2016030899A1 (en) 2014-08-28 2016-03-03 Yeda Research And Development Co. Ltd. Methods of treating amyotrophic lateral scleroses
EP4043567A1 (en) 2014-08-29 2022-08-17 Children's Medical Center Corporation Methods and compositions for the treatment of cancer
WO2016033424A1 (en) 2014-08-29 2016-03-03 Genzyme Corporation Methods for the prevention and treatment of major adverse cardiovascular events using compounds that modulate apolipoprotein b
ES2928500T3 (en) 2014-08-29 2022-11-18 Alnylam Pharmaceuticals Inc Patisiran for use in the treatment of transthyretin-mediated amyloidosis
EP3191591A1 (en) 2014-09-12 2017-07-19 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting complement component c5 and methods of use thereof
KR102473092B1 (en) 2014-09-15 2022-12-01 칠드런'즈 메디컬 센터 코포레이션 Methods and compositions to increase somatic cell nuclear transfer (scnt) efficiency by removing histone h3-lysine trimethylation
EP3663403A1 (en) 2014-09-26 2020-06-10 University of Massachusetts Rna-modulating agents
JOP20200115A1 (en) 2014-10-10 2017-06-16 Alnylam Pharmaceuticals Inc Compositions And Methods For Inhibition Of HAO1 (Hydroxyacid Oxidase 1 (Glycolate Oxidase)) Gene Expression
WO2016061487A1 (en) 2014-10-17 2016-04-21 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting aminolevulinic acid synthase-1 (alas1) and uses thereof
EP3904519A1 (en) 2014-10-30 2021-11-03 Genzyme Corporation Polynucleotide agents targeting serpinc1 (at3) and methods of use thereof
JOP20200092A1 (en) 2014-11-10 2017-06-16 Alnylam Pharmaceuticals Inc HEPATITIS B VIRUS (HBV) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2016081444A1 (en) 2014-11-17 2016-05-26 Alnylam Pharmaceuticals, Inc. Apolipoprotein c3 (apoc3) irna compositions and methods of use thereof
CA2968531A1 (en) 2014-11-21 2016-05-26 Northwestern University The sequence-specific cellular uptake of spherical nucleic acid nanoparticle conjugates
JP6997623B2 (en) 2014-12-12 2022-02-04 エム. ウルフ、トッド Compositions and Methods for Editing Intracellular Nucleic Acids Utilizing Oligonucleotides
EP3234141A4 (en) 2014-12-18 2018-06-20 Alnylam Pharmaceuticals, Inc. Reversir tm compounds
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
WO2016115490A1 (en) 2015-01-16 2016-07-21 Ionis Pharmaceuticals, Inc. Compounds and methods for modulation of dux4
PL3256589T3 (en) 2015-01-22 2022-02-21 Monsanto Technology Llc Compositions and methods for controlling leptinotarsa
WO2016118812A1 (en) 2015-01-23 2016-07-28 Vanderbilt University A robust interferometer and methods of using same
EP3256487A4 (en) 2015-02-09 2018-07-18 Duke University Compositions and methods for epigenome editing
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
EP3262173A2 (en) 2015-02-23 2018-01-03 Crispr Therapeutics AG Materials and methods for treatment of human genetic diseases including hemoglobinopathies
WO2016137923A1 (en) 2015-02-23 2016-09-01 Ionis Pharmaceuticals, Inc. Method for solution phase detritylation of oligomeric compounds
US11129844B2 (en) 2015-03-03 2021-09-28 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating MECP2 expression
EP3268475B1 (en) 2015-03-11 2020-10-21 Yissum Research and Development Company of the Hebrew University of Jerusalem Ltd. Decoy oligonucleotides for the treatment of diseases
EP4218771A1 (en) 2015-03-27 2023-08-02 Yeda Research and Development Co. Ltd Methods of treating motor neuron diseases
KR20180020125A (en) 2015-03-27 2018-02-27 프레지던트 앤드 펠로우즈 오브 하바드 칼리지 Modified T cells and methods for their manufacture and use
US10961532B2 (en) 2015-04-07 2021-03-30 The General Hospital Corporation Methods for reactivating genes on the inactive X chromosome
US10745702B2 (en) 2015-04-08 2020-08-18 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the LECT2 gene
WO2016167780A1 (en) 2015-04-16 2016-10-20 Ionis Pharmaceuticals, Inc. Compositions for modulating expression of c9orf72 antisense transcript
MX2017014215A (en) 2015-05-04 2018-03-28 Monsanto Technology Llc Compositions and methods for controlling arthropod parasite and pest infestations.
US20180161300A1 (en) 2015-05-11 2018-06-14 Yeda Research And Development Co., Ltd. Citrin inhibitors for the treatment of cancer
CN107750125A (en) 2015-06-02 2018-03-02 孟山都技术有限公司 For by the composition and method in delivery of polynucleotides to plant
WO2016196782A1 (en) 2015-06-03 2016-12-08 Monsanto Technology Llc Methods and compositions for introducing nucleic acids into plants
EP3302489A4 (en) 2015-06-04 2019-02-06 Sarepta Therapeutics, Inc. Methods and compounds for treatment of lymphocyte-related diseases and conditions
EP3307316A1 (en) 2015-06-12 2018-04-18 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions and methods of use thereof
WO2016205323A1 (en) 2015-06-18 2016-12-22 Alnylam Pharmaceuticals, Inc. Polynucleotde agents targeting hydroxyacid oxidase (glycolate oxidase, hao1) and methods of use thereof
WO2016209862A1 (en) 2015-06-23 2016-12-29 Alnylam Pharmaceuticals, Inc. Glucokinase (gck) irna compositions and methods of use thereof
JP2018524588A (en) 2015-06-26 2018-08-30 ベス・イスラエル・ディーコネス・メディカル・センター,インコーポレイテッド Cancer therapy targeting tetraspanin 33 (Tspan33) in bone marrow-derived suppressor cells
CA2991045A1 (en) 2015-06-29 2017-01-05 Caris Science, Inc. Therapeutic oligonucleotides binding c1q
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
CA3205381A1 (en) 2015-07-17 2017-01-26 Alnylam Pharmaceuticals, Inc. Multi-targeted single entity conjugates
US10941176B2 (en) 2015-07-28 2021-03-09 Caris Science, Inc. Therapeutic oligonucleotides
WO2017021963A1 (en) 2015-08-03 2017-02-09 Biokine Therapeutics Ltd. Cxcr4 binding agents for treatment of diseases
WO2017021961A1 (en) 2015-08-04 2017-02-09 Yeda Research And Development Co. Ltd. Methods of screening for riboswitches and attenuators
US10130651B2 (en) 2015-08-07 2018-11-20 Arrowhead Pharmaceuticals, Inc. RNAi Therapy for Hepatitis B Virus Infection
SG10202007937SA (en) 2015-09-02 2020-09-29 Alnylam Pharmaceuticals Inc PROGRAMMED CELL DEATH 1 LIGAND 1 (PD-L1) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
US20190048340A1 (en) 2015-09-24 2019-02-14 Crispr Therapeutics Ag Novel family of rna-programmable endonucleases and their uses in genome editing and other applications
US10533175B2 (en) 2015-09-25 2020-01-14 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating Ataxin 3 expression
US11369692B2 (en) 2015-10-28 2022-06-28 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of Duchenne Muscular Dystrophy
PE20181009A1 (en) 2015-10-30 2018-06-26 Genentech Inc ANTI-HtrA1 ANTIBODIES AND METHODS OF USE OF THEM
ES2938883T3 (en) 2015-11-05 2023-04-17 Los Angeles Childrens Hospital Oligo antisense for use in the treatment of acute myeloid leukemia
BR112018008971A2 (en) 2015-11-06 2018-11-27 Crispr Therapeutics Ag Materials and Methods for Treatment of Type 1a Glycogen Storage Disease
WO2017081686A1 (en) 2015-11-10 2017-05-18 B. G. Negev Technologies And Applications Ltd., At Ben-Gurion University Means and methods for reducing tumorigenicity of cancer stem cells
US11001622B2 (en) 2015-11-19 2021-05-11 The Brigham And Women's Hospital, Inc. Method of treating autoimmune disease with lymphocyte antigen CD5-like (CD5L) protein
AU2016359629B2 (en) 2015-11-23 2023-03-09 Ranjan BATRA Tracking and manipulating cellular RNA via nuclear delivery of CRISPR/Cas9
EP3967758A1 (en) 2015-12-01 2022-03-16 CRISPR Therapeutics AG Materials and methods for treatment of alpha-1 antitrypsin deficiency
US11058709B1 (en) 2015-12-04 2021-07-13 Ionis Pharmaceuticals, Inc. Methods of treating breast cancer
US10993995B2 (en) 2015-12-07 2021-05-04 Erasmus University Medical Center Rotterdam Enzymatic replacement therapy and antisense therapy for pompe disease
US11761007B2 (en) 2015-12-18 2023-09-19 The Scripps Research Institute Production of unnatural nucleotides using a CRISPR/Cas9 system
EP3394260B1 (en) 2015-12-23 2021-02-17 CRISPR Therapeutics AG Materials and methods for treatment of amyotrophic lateral sclerosis and/or frontal temporal lobular degeneration
US10907160B2 (en) 2016-01-05 2021-02-02 Ionis Pharmaceuticals, Inc. Methods for reducing LRRK2 expression
US10627396B2 (en) 2016-01-29 2020-04-21 Vanderbilt University Free-solution response function interferometry
US20190038771A1 (en) 2016-02-02 2019-02-07 Crispr Therapeutics Ag Materials and methods for treatment of severe combined immunodeficiency (scid) or omenn syndrome
WO2017136558A1 (en) 2016-02-04 2017-08-10 Curis, Inc. Mutant smoothened and methods of using the same
WO2017141109A1 (en) 2016-02-18 2017-08-24 Crispr Therapeutics Ag Materials and methods for treatment of severe combined immunodeficiency (scid) or omenn syndrome
CA3054284A1 (en) 2016-02-25 2017-08-31 The Brigham And Women's Hospital, Inc. Treatment methods for fibrosis targeting smoc2
WO2017161168A1 (en) 2016-03-16 2017-09-21 Ionis Pharmaceuticals, Inc. Modulation of dyrk1b expression
WO2017161172A1 (en) 2016-03-16 2017-09-21 Ionis Pharmaceuticals, Inc. Methods of modulating keap1
WO2017158422A1 (en) 2016-03-16 2017-09-21 Crispr Therapeutics Ag Materials and methods for treatment of hereditary haemochromatosis
CA3018066A1 (en) 2016-03-18 2017-09-21 Caris Science, Inc. Oligonucleotide probes and uses thereof
CA3021467A1 (en) 2016-04-18 2017-10-26 Crispr Therapeutics Ag Materials and methods for treatment of hemoglobinopathies
MA45295A (en) 2016-04-19 2019-02-27 Alnylam Pharmaceuticals Inc HIGH DENSITY LIPOPROTEIN BINDING PROTEIN (HDLBP / VIGILINE) RNA COMPOSITION AND METHODS FOR USING THEM
WO2017191503A1 (en) 2016-05-05 2017-11-09 Crispr Therapeutics Ag Materials and methods for treatment of hemoglobinopathies
AU2017271579B2 (en) 2016-05-25 2023-10-19 Caris Science, Inc. Oligonucleotide probes and uses thereof
US20190256845A1 (en) 2016-06-10 2019-08-22 Alnylam Pharmaceuticals, Inc. COMPLEMENT COMPONENT C5 iRNA COMPOSITIONS AND METHODS OF USE THEREOF FOR TREATING PAROXYSMAL NOCTURNAL HEMOGLOBINURIA (PNH)
EP3471781A4 (en) 2016-06-17 2020-05-06 Ionis Pharmaceuticals, Inc. Modulation of gys1 expression
HUE060123T2 (en) 2016-06-24 2023-01-28 Scripps Research Inst Novel nucleoside triphosphate transporter and uses thereof
US11427838B2 (en) 2016-06-29 2022-08-30 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of myotonic dystrophy type 1 (DM1) and other related disorders
EP3478313B1 (en) 2016-06-29 2022-05-04 CRISPR Therapeutics AG Materials and methods for treatment of amyotrophic lateral sclerosis (als) and other related disorders
US11564997B2 (en) 2016-06-29 2023-01-31 Crispr Therapeutics Ag Materials and methods for treatment of friedreich ataxia and other related disorders
US11801313B2 (en) 2016-07-06 2023-10-31 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of pain related disorders
CN109843914B (en) 2016-07-06 2024-03-15 沃泰克斯药物股份有限公司 Materials and methods for treating pain-related disorders
WO2018007871A1 (en) 2016-07-08 2018-01-11 Crispr Therapeutics Ag Materials and methods for treatment of transthyretin amyloidosis
CA3030701A1 (en) 2016-07-11 2018-01-18 Translate Bio Ma, Inc. Nucleic acid conjugates and uses thereof
CN109844103A (en) 2016-07-21 2019-06-04 美克斯细胞有限公司 Method and composition for modifier group DNA
WO2018020323A2 (en) 2016-07-25 2018-02-01 Crispr Therapeutics Ag Materials and methods for treatment of fatty acid disorders
JOP20170161A1 (en) 2016-08-04 2019-01-30 Arrowhead Pharmaceuticals Inc RNAi Agents for Hepatitis B Virus Infection
NL2017295B1 (en) 2016-08-05 2018-02-14 Univ Erasmus Med Ct Rotterdam Antisense oligomeric compound for Pompe disease
NL2017294B1 (en) 2016-08-05 2018-02-14 Univ Erasmus Med Ct Rotterdam Natural cryptic exon removal by pairs of antisense oligonucleotides.
WO2018039629A2 (en) 2016-08-25 2018-03-01 Northwestern University Micellar spherical nucleic acids from thermoresponsive, traceless templates
HUE059718T2 (en) 2016-09-02 2022-12-28 Dicerna Pharmaceuticals Inc 4'-phosphate analogs and oligonucleotides comprising the same
CN109661233A (en) 2016-10-06 2019-04-19 Ionis 制药公司 The method that oligomeric compound is conjugated
US11459568B2 (en) 2016-10-31 2022-10-04 University Of Massachusetts Targeting microRNA-101-3p in cancer therapy
JOP20190104A1 (en) 2016-11-10 2019-05-07 Ionis Pharmaceuticals Inc Compounds and methods for reducing atxn3 expression
TWI788312B (en) 2016-11-23 2023-01-01 美商阿尼拉製藥公司 SERPINA1 iRNA COMPOSITIONS AND METHODS OF USE THEREOF
EP3330276A1 (en) 2016-11-30 2018-06-06 Universität Bern Novel bicyclic nucleosides and oligomers prepared therefrom
EP3548620A4 (en) 2016-12-02 2020-07-22 Cold Spring Harbor Laboratory Modulation of lnc05 expression
US11753460B2 (en) 2016-12-13 2023-09-12 Seattle Children's Hospital Methods of exogenous drug activation of chemical-induced signaling complexes expressed in engineered cells in vitro and in vivo
AU2017376950B2 (en) 2016-12-16 2024-02-22 Alnylam Pharmaceuticals, Inc. Methods for treating or preventing TTR-associated diseases using transthyretin (TTR) iRNA compositions
US11197928B2 (en) 2017-01-13 2021-12-14 Board Of Regents, The University Of Texas System Sustained production of high affinity antigen specific antibody by high dose BAFF receptor-targeting mAb-siRNA conjugate
SG11201905508VA (en) 2017-01-23 2019-08-27 Regeneron Pharma Hsd17b13 variants and uses thereof
WO2018154462A2 (en) 2017-02-22 2018-08-30 Crispr Therapeutics Ag Materials and methods for treatment of spinocerebellar ataxia type 2 (sca2) and other spinocerebellar ataxia type 2 protein (atxn2) gene related conditions or disorders
JP2020508056A (en) 2017-02-22 2020-03-19 クリスパー・セラピューティクス・アクチェンゲゼルシャフトCRISPR Therapeutics AG Compositions and methods for gene editing
US11559588B2 (en) 2017-02-22 2023-01-24 Crispr Therapeutics Ag Materials and methods for treatment of Spinocerebellar Ataxia Type 1 (SCA1) and other Spinocerebellar Ataxia Type 1 Protein (ATXN1) gene related conditions or disorders
WO2018154459A1 (en) 2017-02-22 2018-08-30 Crispr Therapeutics Ag Materials and methods for treatment of primary hyperoxaluria type 1 (ph1) and other alanine-glyoxylate aminotransferase (agxt) gene related conditions or disorders
WO2018154418A1 (en) 2017-02-22 2018-08-30 Crispr Therapeutics Ag Materials and methods for treatment of early onset parkinson's disease (park1) and other synuclein, alpha (snca) gene related conditions or disorders
WO2018165564A1 (en) 2017-03-09 2018-09-13 Ionis Pharmaceuticals, Inc. Morpholino modified oligomeric compounds
WO2018183969A1 (en) 2017-03-30 2018-10-04 California Institute Of Technology Barcoded rapid assay platform for efficient analysis of candidate molecules and methods of making and using the platform
TWI801377B (en) 2017-04-18 2023-05-11 美商阿尼拉製藥公司 Methods for the treatment of subjects having a hepatitis b virus (hbv) infection
WO2018193428A1 (en) 2017-04-20 2018-10-25 Synthena Ag Modified oligomeric compounds comprising tricyclo-dna nucleosides and uses thereof
US11767520B2 (en) 2017-04-20 2023-09-26 Atyr Pharma, Inc. Compositions and methods for treating lung inflammation
JP2020517613A (en) 2017-04-20 2020-06-18 シンセナ アーゲー Modified oligomeric compounds containing tricyclo DNA nucleosides and uses thereof
WO2018195486A1 (en) 2017-04-21 2018-10-25 The Broad Institute, Inc. Targeted delivery to beta cells
JP7398279B2 (en) 2017-05-10 2023-12-14 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Targeted editing of cellular RNA by CRISPR/CAS9 nuclear delivery
KR20200005596A (en) 2017-05-12 2020-01-15 크리스퍼 테라퓨틱스 아게 Materials and methods for engineering cells, and their use in immuno-oncology
AU2018300069A1 (en) 2017-07-11 2020-02-27 Synthorx, Inc. Incorporation of unnatural nucleotides and methods thereof
JP7277432B2 (en) 2017-07-13 2023-05-19 アルナイラム ファーマシューティカルズ, インコーポレイテッド Lactate dehydrogenase A (LDHA) iRNA compositions and methods of use thereof
KR20200028997A (en) 2017-07-13 2020-03-17 노오쓰웨스턴 유니버시티 General and direct method of preparing oligonucleotide-functionalized metal-organic framework nanoparticles
US20200181220A1 (en) 2017-08-03 2020-06-11 Synthorx, Inc. Cytokine conjugates for the treatment of proliferative and infectious diseases
US11197884B2 (en) 2017-08-18 2021-12-14 Ionis Pharmaceuticals, Inc. Modulation of the notch signaling pathway for treatment of respiratory disorders
WO2019051173A1 (en) 2017-09-08 2019-03-14 Ionis Pharmaceuticals, Inc. Modulators of smad7 expression
KR20200089656A (en) 2017-09-19 2020-07-27 알닐람 파마슈티칼스 인코포레이티드 Compositions and methods for treating transthyretin (TTR) mediated amyloidosis
CA3079172A1 (en) 2017-10-17 2019-04-25 Crispr Therapeutics Ag Compositions and methods for gene editing for hemophilia a
US20210180091A1 (en) 2017-10-26 2021-06-17 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of hemoglobinopathies
AU2018360697A1 (en) 2017-11-01 2020-05-14 Alnylam Pharmaceuticals, Inc. Complement component C3 iRNA compositions and methods of use thereof
TWI809004B (en) 2017-11-09 2023-07-21 美商Ionis製藥公司 Compounds and methods for reducing snca expression
WO2019092505A1 (en) 2017-11-09 2019-05-16 Casebia Therapeutics Llp Self-inactivating (sin) crispr/cas or crispr/cpf1 systems and uses thereof
US20200385719A1 (en) 2017-11-16 2020-12-10 Alnylam Pharmaceuticals, Inc. Kisspeptin 1 (kiss1) irna compositions and methods of use thereof
EP3714054A1 (en) 2017-11-20 2020-09-30 Alnylam Pharmaceuticals, Inc. Serum amyloid p component (apcs) irna compositions and methods of use thereof
US20190153440A1 (en) 2017-11-21 2019-05-23 Casebia Therapeutics Llp Materials and methods for treatment of autosomal dominant retinitis pigmentosa
CA3084955A1 (en) 2017-12-05 2019-06-13 Vertex Pharmaceuticals Incorporated Crispr-cas9 modified cd34+ human hematopoietic stem and progenitor cells and uses thereof
EP3724332A1 (en) 2017-12-14 2020-10-21 CRISPR Therapeutics AG Novel rna-programmable endonuclease systems and their use in genome editing and other applications
BR112020012088A2 (en) 2017-12-18 2020-11-17 Alnylam Pharmaceuticals, Inc. compositions of the high mobility group box-1 (hmgb1) and methods of using them
CA3084633A1 (en) 2017-12-21 2019-06-27 Crispr Therapeutics Ag Materials and methods for treatment of usher syndrome type 2a and/or non-syndromic autosomal recessive retinitis pigmentosa (arrp)
JP7402163B2 (en) 2017-12-21 2023-12-20 クリスパー セラピューティクス アーゲー Materials and methods for the treatment of Usher syndrome type 2A
WO2019126641A2 (en) 2017-12-21 2019-06-27 Ionis Pharmaceuticals, Inc. Modulation of frataxin expression
CA3088180A1 (en) 2018-01-12 2019-07-18 Crispr Therapeutics Ag Compositions and methods for gene editing by targeting transferrin
MX2020007369A (en) 2018-01-15 2020-10-28 Ionis Pharmaceuticals Inc Modulators of dnm2 expression.
JP2021510714A (en) 2018-01-19 2021-04-30 シンセナ アーゲー Tricyclo-DNA nucleoside precursor and the process for preparing it
WO2019147743A1 (en) 2018-01-26 2019-08-01 Massachusetts Institute Of Technology Structure-guided chemical modification of guide rna and its applications
EP3749768A1 (en) 2018-02-05 2020-12-16 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of hemoglobinopathies
US11566236B2 (en) 2018-02-05 2023-01-31 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of hemoglobinopathies
EP3749368A1 (en) 2018-02-08 2020-12-16 Yeda Research and Development Co. Ltd Methods of identifying and using agents for treating diseases associated with intestinal barrier dysfunction
US20210130824A1 (en) 2018-02-16 2021-05-06 Crispr Therapeutics Ag Compositions and methods for gene editing by targeting fibrinogen-alpha
CA3091857A1 (en) 2018-02-26 2019-08-29 Synthorx, Inc. Il-15 conjugates and uses thereof
US11732260B2 (en) 2018-03-02 2023-08-22 Ionis Pharmaceuticals, Inc. Compounds and methods for the modulation of amyloid-β precursor protein
TW202000199A (en) 2018-03-02 2020-01-01 美商Ionis製藥公司 Modulators of IRF4 expression
CN112105625A (en) 2018-03-07 2020-12-18 赛诺菲 Nucleotide precursors, nucleotide analogs, and oligomeric compounds containing the same
WO2019183150A1 (en) 2018-03-19 2019-09-26 Casebia Therapeutics Limited Liability Partnership Novel rna-programmable endonuclease systems and uses thereof
US11661601B2 (en) 2018-03-22 2023-05-30 Ionis Pharmaceuticals, Inc. Methods for modulating FMR1 expression
EP4051799A2 (en) 2018-03-30 2022-09-07 Rheinische Friedrich-Wilhelms-Universität Bonn Aptamers for targeted activaton of t cell-mediated immunity
AU2019247490A1 (en) 2018-04-06 2020-10-22 Children's Medical Center Corporation Compositions and methods for somatic cell reprogramming and modulating imprinting
EP3775204A4 (en) 2018-04-11 2021-12-29 Ionis Pharmaceuticals, Inc. Modulators of ezh2 expression
WO2019204668A1 (en) 2018-04-18 2019-10-24 Casebia Therapeutics Limited Liability Partnership Compositions and methods for knockdown of apo(a) by gene editing for treatment of cardiovascular disease
UY38225A (en) 2018-05-09 2019-11-29 Ionis Pharmaceuticals Inc COMPOUNDS AND METHODS TO REDUCE THE EXPRESSION OF ATXN3
MX2020011913A (en) 2018-05-09 2021-01-29 Ionis Pharmaceuticals Inc Compounds and methods for reducing fxi expression.
TW202016304A (en) 2018-05-14 2020-05-01 美商阿尼拉製藥公司 Angiotensinogen (agt) irna compositions and methods of use thereof
WO2019241648A1 (en) 2018-06-14 2019-12-19 Ionis Pharmaceuticals, Inc. Compounds and methods for increasing stmn2 expression
US11332746B1 (en) 2018-06-27 2022-05-17 Ionis Pharmaceuticals, Inc. Compounds and methods for reducing LRRK2 expression
JP2021529518A (en) 2018-06-28 2021-11-04 クリスパー セラピューティクス アーゲー Compositions and Methods for Genome Editing by Insertion of Donor polynucleotide
JOP20210018A1 (en) 2018-07-25 2021-01-21 Ionis Pharmaceuticals Inc Compounds and methods for reducing atxn2 expression
EA202190528A1 (en) 2018-08-13 2021-04-23 Элнилэм Фармасьютикалз, Инк. COMPOSITIONS OF HEPATITIS B VIRUS (HBV) dsRNA AGENTS AND METHODS OF THEIR APPLICATION
US20210348162A1 (en) 2018-08-16 2021-11-11 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the lect2 gene
US20210292766A1 (en) 2018-08-29 2021-09-23 University Of Massachusetts Inhibition of Protein Kinases to Treat Friedreich Ataxia
US20220056220A1 (en) 2018-09-14 2022-02-24 Northwestern University Programming protein polymerization with dna
WO2020060986A1 (en) 2018-09-18 2020-03-26 Alnylam Pharmaceuticals, Inc. Ketohexokinase (khk) irna compositions and methods of use thereof
US20210348159A1 (en) 2018-10-17 2021-11-11 Crispr Therapeutics Ag Compositions and methods for delivering transgenes
US10913951B2 (en) 2018-10-31 2021-02-09 University of Pittsburgh—of the Commonwealth System of Higher Education Silencing of HNF4A-P2 isoforms with siRNA to improve hepatocyte function in liver failure
TW202028222A (en) 2018-11-14 2020-08-01 美商Ionis製藥公司 Modulators of foxp3 expression
EP3880821A4 (en) 2018-11-15 2023-01-25 Ionis Pharmaceuticals, Inc. Modulators of irf5 expression
IL263184A (en) 2018-11-21 2020-05-31 Yarden Yosef Method of treating cancer and compositions for same
WO2020118259A1 (en) 2018-12-06 2020-06-11 Northwestern University Protein crystal engineering through dna hybridization interactions
CN113631709A (en) 2018-12-20 2021-11-09 普拉克西斯精密药物股份有限公司 Compositions and methods for treating KCNT 1-related disorders
HRP20231338T1 (en) 2018-12-20 2024-02-16 Vir Biotechnology, Inc. Combination hbv therapy
MX2021008628A (en) 2019-01-16 2021-11-17 Genzyme Corp Serpinc1 irna compositions and methods of use thereof.
JP2022519532A (en) 2019-01-31 2022-03-24 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 Modulator of YAP1 expression
MX2021009259A (en) 2019-02-06 2021-08-24 Synthorx Inc Il-2 conjugates and methods of use thereof.
SG11202108357PA (en) 2019-02-15 2021-08-30 Crispr Therapeutics Ag Gene editing for hemophilia a with improved factor viii expression
WO2020171889A1 (en) 2019-02-19 2020-08-27 University Of Rochester Blocking lipid accumulation or inflammation in thyroid eye disease
EP3927378A1 (en) 2019-02-21 2021-12-29 Yissum Research Development Company of the Hebrew University of Jerusalem Ltd. Method for reduction drug-induced nephrotoxicity
MX2021010152A (en) 2019-02-27 2021-09-14 Ionis Pharmaceuticals Inc Modulators of malat1 expression.
MA55297A (en) 2019-03-12 2022-01-19 Bayer Healthcare Llc NOVEL HIGH-FIDELITY PROGRAMMABLE RNA ENDONUCLEASE SYSTEMS AND THEIR USES
JP2022527108A (en) 2019-03-29 2022-05-30 ディセルナ ファーマシューティカルズ インコーポレイテッド Compositions and Methods for the Treatment of KRAS-Related Diseases or Disorders
JP2022527105A (en) 2019-03-29 2022-05-30 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 Compounds and Methods for Modulating UBE3A-ATS
CA3136676A1 (en) 2019-05-03 2020-11-12 Dicerna Pharmaceuticals, Inc. Double-stranded nucleic acid inhibitor molecules with shortened sense strands
US20210047649A1 (en) 2019-05-08 2021-02-18 Vertex Pharmaceuticals Incorporated Crispr/cas all-in-two vector systems for treatment of dmd
US20220339256A1 (en) 2019-05-13 2022-10-27 Vir Biotechnology, Inc. Compositions and methods for treating hepatitis b virus (hbv) infection
JP2022538784A (en) 2019-06-14 2022-09-06 ザ スクリプス リサーチ インスティテュート Reagents and methods for replication, transcription and translation in semi-synthetic organisms
US20210008161A1 (en) 2019-06-17 2021-01-14 Crispr Therapeutics Ag Methods and compositions for improved homology directed repair
WO2020255008A1 (en) 2019-06-18 2020-12-24 Janssen Sciences Ireland Unlimited Company COMBINATION OF HEPATITIS B VIRUS (HBV) VACCINES AND HBV-TARGETING RNAi
JP2022536945A (en) 2019-06-18 2022-08-22 ヤンセン・サイエンシズ・アイルランド・アンリミテッド・カンパニー Combination of hepatitis B virus (HBV) vaccine and RNAi targeting HBV
EP3956450A4 (en) 2019-07-26 2022-11-16 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating gfap
WO2021022109A1 (en) 2019-08-01 2021-02-04 Alnylam Pharmaceuticals, Inc. SERPIN FAMILY F MEMBER 2 (SERPINF2) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2021022108A2 (en) 2019-08-01 2021-02-04 Alnylam Pharmaceuticals, Inc. CARBOXYPEPTIDASE B2 (CPB2) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
EP4013870A1 (en) 2019-08-13 2022-06-22 Alnylam Pharmaceuticals, Inc. Small ribosomal protein subunit 25 (rps25) irna agent compositions and methods of use thereof
WO2021030706A1 (en) 2019-08-15 2021-02-18 Synthorx, Inc. Immuno oncology combination therapies with il-2 conjugates
WO2021030778A1 (en) 2019-08-15 2021-02-18 Ionis Pharmaceuticals, Inc. Linkage modified oligomeric compounds and uses thereof
CA3148135A1 (en) 2019-08-23 2021-03-04 Carolina E. CAFFARO Il-15 conjugates and uses thereof
MX2022002689A (en) 2019-09-03 2022-04-07 Alnylam Pharmaceuticals Inc Compositions and methods for inhibiting expression of the lect2 gene.
KR20220061158A (en) 2019-09-10 2022-05-12 신톡스, 인크. IL-2 conjugates and methods of use for treating autoimmune diseases
EP4038189A1 (en) 2019-10-04 2022-08-10 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing ugt1a1 gene expression
EP4045652A1 (en) 2019-10-18 2022-08-24 Alnylam Pharmaceuticals, Inc. Solute carrier family member irna compositions and methods of use thereof
EP4048793A1 (en) 2019-10-22 2022-08-31 Alnylam Pharmaceuticals, Inc. Complement component c3 irna compositions and methods of use thereof
AR120341A1 (en) 2019-11-01 2022-02-09 Alnylam Pharmaceuticals Inc COMPOSITIONS OF RNAi AGENTS AGAINST HUNTINGTINE (HTT) AND THEIR METHODS OF USE
US20230040920A1 (en) 2019-11-01 2023-02-09 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing dnajb1-prkaca fusion gene expression
MX2022005251A (en) 2019-11-04 2022-06-08 Synthorx Inc Interleukin 10 conjugates and uses thereof.
CN114981431A (en) 2019-11-13 2022-08-30 阿尔尼拉姆医药品有限公司 Methods and compositions for treating Angiotensinogen (AGT) -related disorders
EP4061945A1 (en) 2019-11-22 2022-09-28 Alnylam Pharmaceuticals, Inc. Ataxin3 (atxn3) rnai agent compositions and methods of use thereof
CN115335521A (en) 2019-11-27 2022-11-11 克里斯珀医疗股份公司 Method for synthesizing RNA molecules
EP4073251A1 (en) 2019-12-13 2022-10-19 Alnylam Pharmaceuticals, Inc. Human chromosome 9 open reading frame 72 (c9orf72) irna agent compositions and methods of use thereof
WO2021126734A1 (en) 2019-12-16 2021-06-24 Alnylam Pharmaceuticals, Inc. Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
EP4077674A1 (en) 2019-12-18 2022-10-26 Alia Therapeutics S.R.L. Compositions and methods for treating retinitis pigmentosa
US20230123981A1 (en) 2020-01-15 2023-04-20 Dicerna Pharmaceuticals, Inc. 4'-o-methylene phosphonate nucleic acids and analogues thereof
US20230057461A1 (en) 2020-01-27 2023-02-23 The U.S.A., As Represented By The Secretary, Department Of Health And Human Services Rab13 and net1 antisense oligonucleotides to treat metastatic cancer
WO2021154941A1 (en) 2020-01-31 2021-08-05 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions for use in the treatment of amyotrophic lateral sclerosis (als)
CA3170377A1 (en) 2020-02-10 2021-08-19 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing vegf-a expression
CN115397989A (en) 2020-02-18 2022-11-25 阿尔尼拉姆医药品有限公司 Apolipoprotein C3 (APOC 3) iRNA compositions and methods of use thereof
TW202140787A (en) 2020-02-28 2021-11-01 美商Ionis製藥公司 Compounds and methods for modulating smn2
EP4114947A1 (en) 2020-03-05 2023-01-11 Alnylam Pharmaceuticals, Inc. Complement component c3 irna compositions and methods of use thereof for treating or preventing complement component c3-associated diseases
WO2021178736A1 (en) 2020-03-06 2021-09-10 Alnylam Pharmaceuticals, Inc. KETOHEXOKINASE (KHK) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2021188611A1 (en) 2020-03-18 2021-09-23 Alnylam Pharmaceuticals, Inc. Compositions and methods for treating subjects having a heterozygous alanine-glyoxylate aminotransferase gene (agxt) variant
CN116209759A (en) 2020-03-26 2023-06-02 阿尔尼拉姆医药品有限公司 Coronavirus iRNA compositions and methods of use thereof
WO2021202443A2 (en) 2020-03-30 2021-10-07 Alnylam Pharmaceucticals, Inc. Compositions and methods for silencing dnajc15 gene expression
BR112022020145A2 (en) 2020-04-06 2023-01-03 Alnylam Pharmaceuticals Inc COMPOSITIONS AND METHODS FOR SILENCING THE MYOC EXPRESSION
WO2021206917A1 (en) 2020-04-07 2021-10-14 Alnylam Pharmaceuticals, Inc. ANGIOTENSIN-CONVERTING ENZYME 2 (ACE2) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
EP4133077A1 (en) 2020-04-07 2023-02-15 Alnylam Pharmaceuticals, Inc. Transmembrane serine protease 2 (tmprss2) irna compositions and methods of use thereof
WO2021207189A1 (en) 2020-04-07 2021-10-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing scn9a expression
KR20230018377A (en) 2020-04-27 2023-02-07 알닐람 파마슈티칼스 인코포레이티드 Apolipoprotein E (APOE) IRNA preparation composition and method of use thereof
KR20230017789A (en) 2020-04-30 2023-02-06 알닐람 파마슈티칼스 인코포레이티드 Complement Factor B (CFB) iRNA Compositions and Methods of Use Thereof
WO2021222768A2 (en) 2020-05-01 2021-11-04 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating atxn1
EP4150078A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of argininosuccinate lyase (asl)
WO2021231673A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of leucine rich repeat kinase 2 (lrrk2)
EP4150090A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of otoferlin (otof)
EP4150077A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of transmembrane channel-like protein 1 (tmc1)
WO2021231679A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of gap junction protein beta 2 (gjb2)
CA3162416C (en) 2020-05-15 2023-07-04 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of argininosuccinate synthetase (ass1)
EP4150076A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of methyl-cpg binding protein 2 (mecp2)
EP4150089A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of retinoschisin 1 (rs1)
EP4153746A1 (en) 2020-05-21 2023-03-29 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting marc1 gene expression
CA3179051A1 (en) 2020-05-22 2021-11-25 Chandra Vargeese Double stranded oligonucleotide compositions and methods relating thereto
AR122534A1 (en) 2020-06-03 2022-09-21 Triplet Therapeutics Inc METHODS FOR THE TREATMENT OF NUCLEOTIDE REPEAT EXPANSION DISORDERS ASSOCIATED WITH MSH3 ACTIVITY
EP4161552A1 (en) 2020-06-05 2023-04-12 The Broad Institute, Inc. Compositions and methods for treating neoplasia
EP4162050A1 (en) 2020-06-09 2023-04-12 Alnylam Pharmaceuticals, Inc. Rnai compositions and methods of use thereof for delivery by inhalation
CN116209760A (en) 2020-06-18 2023-06-02 阿尔尼拉姆医药品有限公司 Xanthine Dehydrogenase (XDH) iRNA compositions and methods of use thereof
BR112022026316A2 (en) 2020-06-24 2023-03-07 Vir Biotechnology Inc ENGINEERED HEPATITIS B VIRUS NEUTRALIZING ANTIBODIES AND THEIR USES
TW202216203A (en) 2020-06-25 2022-05-01 美商欣爍克斯公司 Immuno oncology combination therapy with il-2 conjugates and anti-egfr antibodies
CA3185749A1 (en) 2020-06-29 2022-01-06 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating plp1
IL300283A (en) 2020-08-04 2023-04-01 Dicerna Pharmaceuticals Inc Systemic delivery of oligonucleotides
EP4217489A1 (en) 2020-09-24 2023-08-02 Alnylam Pharmaceuticals, Inc. Dipeptidyl peptidase 4 (dpp4) irna compositions and methods of use thereof
US20220290136A1 (en) 2020-09-30 2022-09-15 Crispr Therapeutics Ag Materials and methods for treatment of amyotrophic lateral sclerosis
TW202229552A (en) 2020-10-05 2022-08-01 美商艾拉倫製藥股份有限公司 G protein-coupled receptor 75 (gpr75) irna compositions and methods of use thereof
EP3978608A1 (en) 2020-10-05 2022-04-06 SQY Therapeutics Oligomeric compound for dystrophin rescue in dmd patients throughout skipping of exon-51
MX2023004029A (en) 2020-10-09 2023-04-27 Synthorx Inc Immuno oncology combination therapy with il-2 conjugates and pembrolizumab.
MX2023004032A (en) 2020-10-09 2023-04-27 Synthorx Inc Immuno oncology therapies with il-2 conjugates.
EP4228637A1 (en) 2020-10-15 2023-08-23 Yeda Research and Development Co. Ltd Method of treating myeloid malignancies
CN116368146A (en) 2020-10-20 2023-06-30 赛诺菲 Novel ligands for asialoglycoprotein receptors
EP4232581A1 (en) 2020-10-21 2023-08-30 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating primary hyperoxaluria
EP4232582A1 (en) 2020-10-23 2023-08-30 Alnylam Pharmaceuticals, Inc. Mucin 5b (muc5b) irna compositions and methods of use thereof
JP2023549500A (en) 2020-11-13 2023-11-27 アルナイラム ファーマシューティカルズ, インコーポレイテッド Coagulation factor V (F5) iRNA composition and method of use thereof
EP4136092A4 (en) 2020-11-18 2023-10-11 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating angiotensinogen expression
TW202237150A (en) 2020-12-01 2022-10-01 美商艾拉倫製藥股份有限公司 Methods and compositions for inhibition of hao1 (hydroxyacid oxidase 1 (glycolate oxidase)) gene expression
EP4259795A1 (en) 2020-12-08 2023-10-18 Alnylam Pharmaceuticals, Inc. Coagulation factor x (f10) irna compositions and methods of use thereof
CA3206285A1 (en) 2020-12-23 2022-06-30 Flagship Pioneering, Inc. Compositions of modified trems and uses thereof
WO2022150260A1 (en) 2021-01-05 2022-07-14 Alnylam Pharmaceuticals, Inc. COMPLEMENT COMPONENT 9 (C9) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2022174000A2 (en) 2021-02-12 2022-08-18 Alnylam Pharmaceuticals, Inc. Superoxide dismutase 1 (sod1) irna compositions and methods of use thereof for treating or preventing superoxide dismutase 1- (sod1-) associated neurodegenerative diseases
TW202245843A (en) 2021-02-12 2022-12-01 美商欣爍克斯公司 Skin cancer combination therapy with il-2 conjugates and cemiplimab
EP4291243A1 (en) 2021-02-12 2023-12-20 Synthorx, Inc. Lung cancer combination therapy with il-2 conjugates and an anti-pd-1 antibody or antigen-binding fragment thereof
JP2024509783A (en) 2021-02-25 2024-03-05 アルナイラム ファーマシューティカルズ, インコーポレイテッド Prion protein (PRNP) IRNA compositions and methods of use thereof
WO2022182574A1 (en) 2021-02-26 2022-09-01 Alnylam Pharmaceuticals, Inc. KETOHEXOKINASE (KHK) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2022187435A1 (en) 2021-03-04 2022-09-09 Alnylam Pharmaceuticals, Inc. Angiopoietin-like 3 (angptl3) irna compositions and methods of use thereof
EP4304640A1 (en) 2021-03-12 2024-01-17 Northwestern University Antiviral vaccines using spherical nucleic acids
WO2022192519A1 (en) 2021-03-12 2022-09-15 Alnylam Pharmaceuticals, Inc. Glycogen synthase kinase 3 alpha (gsk3a) irna compositions and methods of use thereof
CA3214499A1 (en) 2021-03-29 2022-10-06 Alnylam Pharmaceuticals, Inc. Huntingtin (htt) irna agent compositions and methods of use thereof
WO2022212153A1 (en) 2021-04-01 2022-10-06 Alnylam Pharmaceuticals, Inc. Proline dehydrogenase 2 (prodh2) irna compositions and methods of use thereof
EP4330392A1 (en) 2021-04-26 2024-03-06 Alnylam Pharmaceuticals, Inc. Transmembrane protease, serine 6 (tmprss6) irna compositions and methods of use thereof
WO2022232343A1 (en) 2021-04-29 2022-11-03 Alnylam Pharmaceuticals, Inc. Signal transducer and activator of transcription factor 6 (stat6) irna compositions and methods of use thereof
WO2022235537A1 (en) 2021-05-03 2022-11-10 Alnylam Pharmaceuticals, Inc. Compositions and methods for treating transthyretin (ttr) mediated amyloidosis
EP4341401A1 (en) 2021-05-18 2024-03-27 Alnylam Pharmaceuticals, Inc. Sodium-glucose cotransporter-2 (sglt2) irna compositions and methods of use thereof
WO2022246023A1 (en) 2021-05-20 2022-11-24 Korro Bio, Inc. Methods and compositions for adar-mediated editing
WO2022256283A2 (en) 2021-06-01 2022-12-08 Korro Bio, Inc. Methods for restoring protein function using adar
WO2022256395A1 (en) 2021-06-02 2022-12-08 Alnylam Pharmaceuticals, Inc. Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
TW202313679A (en) 2021-06-03 2023-04-01 美商欣爍克斯公司 Head and neck cancer combination therapy comprising an il-2 conjugate and a pd-1 antagonist
BR112023025224A2 (en) 2021-06-04 2024-02-27 Alnylam Pharmaceuticals Inc HUMAN CHROMOSOME 9 (C9ORF72) OPEN READING BOARD 72 IRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
WO2022260939A2 (en) 2021-06-08 2022-12-15 Alnylam Pharmaceuticals, Inc. Compositions and methods for treating or preventing stargardt's disease and/or retinal binding protein 4 (rbp4)-associated disorders
EP4101928A1 (en) 2021-06-11 2022-12-14 Bayer AG Type v rna programmable endonuclease systems
BR112023023768A2 (en) 2021-06-11 2024-02-27 Bayer Ag TYPE V RNA PROGRAMMABLE ENDONUCLEASE SYSTEMS
CA3223192A1 (en) 2021-06-18 2022-12-22 Ionis Pharmaceuticals, Inc. Compounds and methods for reducing ifnar1 expression
WO2023278410A1 (en) 2021-06-29 2023-01-05 Korro Bio, Inc. Methods and compositions for adar-mediated editing
US20230194709A9 (en) 2021-06-29 2023-06-22 Seagate Technology Llc Range information detection using coherent pulse sets with selected waveform characteristics
AU2022303164A1 (en) 2021-06-30 2024-01-18 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating an angiotensinogen- (agt-) associated disorder
WO2023285431A1 (en) 2021-07-12 2023-01-19 Alia Therapeutics Srl Compositions and methods for allele specific treatment of retinitis pigmentosa
WO2023003805A1 (en) 2021-07-19 2023-01-26 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating subjects having or at risk of developing a non-primary hyperoxaluria disease or disorder
IL309905A (en) 2021-07-23 2024-03-01 Alnylam Pharmaceuticals Inc Beta-catenin (ctnnb1) irna compositions and methods of use thereof
WO2023009687A1 (en) 2021-07-29 2023-02-02 Alnylam Pharmaceuticals, Inc. 3-hydroxy-3-methylglutaryl-coa reductase (hmgcr) irna compositions and methods of use thereof
CN117795074A (en) 2021-08-03 2024-03-29 阿尔尼拉姆医药品有限公司 Transthyretin (TTR) iRNA compositions and methods of use thereof
TW202337474A (en) 2021-08-04 2023-10-01 美商艾拉倫製藥股份有限公司 Irna compositions and methods for silencing angiotensinogen (agt)
CA3228916A1 (en) 2021-08-11 2023-02-16 King's College London Compositions and methods for improved treatment of disorders affecting the central nervous system
AR126771A1 (en) 2021-08-13 2023-11-15 Alnylam Pharmaceuticals Inc RNAi COMPOSITIONS AGAINST FACTOR XII (F12) AND THEIR METHODS OF USE
EP4144841A1 (en) 2021-09-07 2023-03-08 Bayer AG Novel small rna programmable endonuclease systems with impoved pam specificity and uses thereof
WO2023044370A2 (en) 2021-09-17 2023-03-23 Alnylam Pharmaceuticals, Inc. Irna compositions and methods for silencing complement component 3 (c3)
WO2023044094A1 (en) 2021-09-20 2023-03-23 Alnylam Pharmaceuticals, Inc. Inhibin subunit beta e (inhbe) modulator compositions and methods of use thereof
WO2023069603A1 (en) 2021-10-22 2023-04-27 Korro Bio, Inc. Methods and compositions for disrupting nrf2-keap1 protein interaction by adar mediated rna editing
WO2023076451A1 (en) 2021-10-29 2023-05-04 Alnylam Pharmaceuticals, Inc. Complement factor b (cfb) irna compositions and methods of use thereof
TW202334418A (en) 2021-10-29 2023-09-01 美商艾拉倫製藥股份有限公司 Huntingtin (htt) irna agent compositions and methods of use thereof
WO2023122573A1 (en) 2021-12-20 2023-06-29 Synthorx, Inc. Head and neck cancer combination therapy comprising an il-2 conjugate and pembrolizumab
WO2023118349A1 (en) 2021-12-21 2023-06-29 Alia Therapeutics Srl Type ii cas proteins and applications thereof
WO2023118068A1 (en) 2021-12-23 2023-06-29 Bayer Aktiengesellschaft Novel small type v rna programmable endonuclease systems
WO2023122750A1 (en) 2021-12-23 2023-06-29 Synthorx, Inc. Cancer combination therapy with il-2 conjugates and cetuximab
WO2023141314A2 (en) 2022-01-24 2023-07-27 Alnylam Pharmaceuticals, Inc. Heparin sulfate biosynthesis pathway enzyme irna agent compositions and methods of use thereof
WO2023177866A1 (en) 2022-03-18 2023-09-21 Dicerna Pharmaceuticals, Inc. Decarboxylative acetoxylation using mn(ii) or mn(iii) reagent for synthesis of 4'-acetoxy- nucleoside and use thereof for synthesis of corresponding 4'-(dimethoxyphosphoryl)methoxy- nucleotide
WO2023194359A1 (en) 2022-04-04 2023-10-12 Alia Therapeutics Srl Compositions and methods for treatment of usher syndrome type 2a
WO2023233290A1 (en) 2022-05-31 2023-12-07 Janssen Sciences Ireland Unlimited Company Rnai agents targeting pd-l1
WO2023237587A1 (en) 2022-06-10 2023-12-14 Bayer Aktiengesellschaft Novel small type v rna programmable endonuclease systems
EP4311579A1 (en) 2022-07-29 2024-01-31 Association Française contre les Myopathies B cell-specific mab-sirna conjugates improve myasthenia
WO2024035952A1 (en) 2022-08-12 2024-02-15 Remix Therapeutics Inc. Methods and compositions for modulating splicing at alternative splice sites
WO2024039776A2 (en) 2022-08-18 2024-02-22 Alnylam Pharmaceuticals, Inc. Universal non-targeting sirna compositions and methods of use thereof
WO2024059165A1 (en) 2022-09-15 2024-03-21 Alnylam Pharmaceuticals, Inc. 17b-hydroxysteroid dehydrogenase type 13 (hsd17b13) irna compositions and methods of use thereof
WO2024056880A2 (en) 2022-09-16 2024-03-21 Alia Therapeutics Srl Enqp type ii cas proteins and applications thereof

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3687808A (en) * 1969-08-14 1972-08-29 Univ Leland Stanford Junior Synthetic polynucleotides
US4707440A (en) * 1984-01-30 1987-11-17 Enzo Biochem, Inc. Nucleic acid hybridization assay and detectable molecules useful in such assay
US5386023A (en) * 1990-07-27 1995-01-31 Isis Pharmaceuticals Backbone modified oligonucleotide analogs and preparation thereof through reductive coupling
US5223618A (en) * 1990-08-13 1993-06-29 Isis Pharmaceuticals, Inc. 4'-desmethyl nucleoside analog compounds
US5138045A (en) * 1990-07-27 1992-08-11 Isis Pharmaceuticals Polyamine conjugated oligonucleotides
US5378825A (en) * 1990-07-27 1995-01-03 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogs

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
BIOCONJUGATE CHEMISTRY, issued May 1990, JOHN GOODCHILD, "Conjugates of Oligonucleotides and Modified Oligonucleotides: A Review of their Synthesis and Properties", pages 165-186. *
J. ORG. CHEM., Vol. 52, No. 19, issued 1987, EUGENE P. STIRCHAK et al., "Uncharged Stereoregular Nucleic Acid Analogues. 1. Synthesis of a Cytosin-Containing Oligomer with Carbamate Internucleoside Linkages", page s4202-4206. *
See also references of EP0737201A4 *
TETRAHEDRON LETTERS, Volume 28, Number 7, issued 1987, JAMES M. COULL et al., "Synthesis and Characterization of a Carbamate-Linked Oligonucleoside", pages 745-748. *

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0763052A1 (en) * 1994-05-31 1997-03-19 Isis Pharmaceuticals, Inc. ANTISENSE OLIGONUCLEOTIDE MODULATION OF raf GENE EXPRESSION
EP0763052A4 (en) * 1994-05-31 1998-10-14 Isis Pharmaceuticals Inc ANTISENSE OLIGONUCLEOTIDE MODULATION OF raf GENE EXPRESSION
EP0813539A1 (en) * 1995-03-06 1997-12-29 Isis Pharmaceuticals, Inc. Improved process for the synthesis of 2'-o-substituted pyrimidines and oligomeric compounds therefrom
EP0813539A4 (en) * 1995-03-06 2002-01-23 Isis Pharmaceuticals Inc Improved process for the synthesis of 2'-o-substituted pyrimidines and oligomeric compounds therefrom
US6776986B1 (en) 1996-06-06 2004-08-17 Novartis Ag Inhibition of HIV-1 replication by antisense RNA expression
US6461871B1 (en) 1997-05-09 2002-10-08 Lightup Technologies Ab Method for the preparation of a probe for nucleic acid hybridization
WO1999013105A1 (en) * 1997-09-05 1999-03-18 Mikael Kubista Method for the preparation of a probe for nucleic acid hybridization
GB2344823A (en) * 1997-09-05 2000-06-21 Kubista Mikael Method for the preparation of a probe for nucleic acid hybridization
GB2344823B (en) * 1997-09-05 2002-09-04 Kubista Mikael Method for the preparation of a probe for nucleic acid hybridization
DE19819735A1 (en) * 1998-05-02 1999-11-04 Novartis Ag Device and method for producing an arrangement of chain molecules on a carrier material
JP2000300299A (en) * 1999-03-26 2000-10-31 Vysis Inc Solid-phase nucleic acid labeling by amino group transfer
EP1046649B1 (en) * 1999-03-26 2006-06-28 Abbott Molecular Inc. Solid phase nucleic acid labeling by transamination

Also Published As

Publication number Publication date
US5541307A (en) 1996-07-30
EP0737201A4 (en) 1998-12-30
EP0737201A1 (en) 1996-10-16

Similar Documents

Publication Publication Date Title
WO1995018136A1 (en) Backbone modified oligonucleotide analogs and solid phase synthesis thereof
US5378825A (en) Backbone modified oligonucleotide analogs
US5618704A (en) Backbone-modified oligonucleotide analogs and preparation thereof through radical coupling
US6900301B2 (en) Backbone modified oligonucleotide analogues
US5602240A (en) Backbone modified oligonucleotide analogs
US5808023A (en) Modified oligonucleotides
US6214551B1 (en) Oligonucleoside linkages containing adjacent nitrogen atoms
US5677437A (en) Heteroatomic oligonucleoside linkages
US5489677A (en) Oligonucleoside linkages containing adjacent oxygen and nitrogen atoms
US5792844A (en) Oligonucleoside linkages containing adjacent nitrogen atoms
US6001841A (en) Cyclobutyl oligonucleotide surrogates
IE83315B1 (en) Backbone modified oligonucleotide analogs
US20020183502A1 (en) Backbone-modified oligonucleotide analogs and methods for using same
EP0649429B1 (en) Heteroatomic oligonucleoside linkages
WO1994022886A1 (en) Heteroatomic oligonucleoside linkages
WO1994022894A1 (en) Backbone modified oligonucleotide analogs and preparation thereof through radical coupling
WO1994022893A1 (en) Oligonucleoside linkages containing adjacent nitrogen atoms

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): CA JP US

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 1995906115

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1995906115

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: CA

WWR Wipo information: refused in national office

Ref document number: 1995906115

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1995906115

Country of ref document: EP