US20150299696A1 - SHORT INTERFERING NUCLEIC ACID (siNA) COMPOSITIONS - Google Patents

SHORT INTERFERING NUCLEIC ACID (siNA) COMPOSITIONS Download PDF

Info

Publication number
US20150299696A1
US20150299696A1 US14/398,300 US201314398300A US2015299696A1 US 20150299696 A1 US20150299696 A1 US 20150299696A1 US 201314398300 A US201314398300 A US 201314398300A US 2015299696 A1 US2015299696 A1 US 2015299696A1
Authority
US
United States
Prior art keywords
linker
composition according
strand
nucleotides
inverted abasic
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/398,300
Inventor
Brian Allen Carr
Vasant R. Jadhav
Denise M. Kenski
David M. Tellers
Aarron T. Willingham
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sirna Therapeutics Inc
Merck Sharp and Dohme LLC
Original Assignee
Sirna Therapeutics 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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=49515013&utm_source=***_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20150299696(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Sirna Therapeutics Inc filed Critical Sirna Therapeutics Inc
Priority to US14/398,300 priority Critical patent/US20150299696A1/en
Publication of US20150299696A1 publication Critical patent/US20150299696A1/en
Assigned to MERCK SHARP & DOHME CORP. reassignment MERCK SHARP & DOHME CORP. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CARR, Brian Allen, WILLINGHAM, AARRON T., KENSKI, DENISE M., JADHAV, VASANT R., TELLERS, DAVID M.
Assigned to SIRNA THERAPEUTICS, INC. reassignment SIRNA THERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MERCK SHARP & DOHME CORP.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/111General methods applicable to biologically active non-coding nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3212'-O-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/323Chemical structure of the sugar modified ring structure
    • C12N2310/3231Chemical structure of the sugar modified ring structure having an additional ring, e.g. LNA, ENA
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/34Spatial arrangement of the modifications
    • C12N2310/344Position-specific modifications, e.g. on every purine, at the 3'-end
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/34Spatial arrangement of the modifications
    • C12N2310/346Spatial arrangement of the modifications having a combination of backbone and sugar modifications
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/32Special delivery means, e.g. tissue-specific
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/50Methods for regulating/modulating their activity
    • C12N2320/51Methods for regulating/modulating their activity modulating the chemical stability, e.g. nuclease-resistance

Definitions

  • the present invention relates to compounds, compositions, and methods for the study, diagnosis, and treatment of traits, diseases and conditions that may respond to the modulation of gene expression and/or activity.
  • the present invention is also directed to compounds, compositions, and methods relating to traits, diseases and conditions that may respond to the modulation of expression and/or activity of genes involved in gene expression pathways or other cellular processes that mediate the maintenance or development of such traits, diseases and conditions.
  • the invention relates to chemically modified short interfering nucleic acid (siNA) molecules that may be capable of mediating RNA interference (RNAi) against gene expression, including cocktails of such siNA molecules and formulations of such siNA molecules.
  • siNA molecules are useful, for example, in providing compositions that may prevent, inhibit, or reduce various diseases, traits and conditions that are associated with gene expression or activity in a subject or organism.
  • RNA interference refers to the process of sequence-specific post-transcriptional gene silencing in animals mediated by short interfering RNAs (siRNAs) (Zamore et al., 2000, Cell, 101, 25-33; Bass, 2000, Cell, 101, 235; Fire et al., 1998, Nature, 391, 806; Hamilton et al., 1999, Science, 286, 950-951; Lin et al., 1999, Nature, 402, 128-129; Sharp, 1999, genes & Dev., 13:139-141; and Strauss, 1999, Science, 286, 886).
  • siRNAs short interfering RNAs
  • WO 99/61631 is commonly referred to as post-transcriptional gene silencing or RNA silencing and is also referred to as quelling in fungi.
  • the process of post-transcriptional gene silencing is thought to be an evolutionarily-conserved cellular defense mechanism used to prevent the expression of foreign genes and is commonly shared by diverse flora and phyla (Fire et al., 1999, Trends genet., 15, 358).
  • Such protection from foreign gene expression may have evolved in response to the production of double-stranded RNAs (dsRNAs) derived from viral infection or from the random integration of transposon elements into a host genome via a cellular response that specifically destroys homologous single-stranded RNA or viral genomic RNA.
  • dsRNAs double-stranded RNAs
  • RNAi The therapeutic potential of RNAi lies in the ability to modulate gene expression in a sequence specific manner by harnessing a highly conserved, robust endogenous mechanism of action. This endogenous mechanism of action vastly expands upon the number of available targets for disease modification when compared to existing small molecule and biologic modalities. Nevertheless, a opposed to exogenously supplied small molecule and biologic modalities, the RNA molecules that serve as triggers for RNAi are not well suited for administration due to their inherent instability, especially in biologic systems. This problem has been addressed through innovation, both in terms of chemical modification of RNA triggers (see U.S. Ser. No. 10/444,853, published as US .S Patent Appl. Publ. No. 20040192626) and various delivery approaches (see U.S. Ser.
  • the invention provides a solution to the problem of having a sufficient repertoire of available compounds and compositions for use in RNAi based therapeutics that may be compatible with different delivery modalities and/or routes of administration by providing additional forms of chemically modified short interfering nucleic acid (siNA) molecules.
  • siNA short interfering nucleic acid
  • the present invention provides compounds, compositions, and methods useful for modulating the expression of target genes and for treating diseases and conditions that may respond to such modulation by RNA interference (RNAi).
  • RNAi RNA interference
  • the present invention provides certain chemically modified short interfering nucleic acid (siNA) molecules having any of Formula A, B, C, D, E, F, G, H, I, J, K, L, M, N, O, or P or as otherwise provided in the description and claims herein.
  • siNA short interfering nucleic acid
  • FIG. 1 shows a non-limiting proposed mechanistic representation of target RNA degradation involved in RNAi.
  • Double-stranded RNA dsRNA
  • RdRP RNA-dependent RNA polymerase
  • siNA duplexes RNA-dependent RNA polymerase
  • synthetic or expressed siNA can be introduced directly into a cell by appropriate means.
  • An active siNA complex forms that recognizes a target RNA, resulting in degradation of the target RNA by the RISC endonuclease complex or in the synthesis of additional RNA by RNA-dependent RNA polymerase (RdRP), which can activate DICER and result in additional siNA molecules, thereby amplifying the RNAi response.
  • RdRP RNA-dependent RNA polymerase
  • FIG. 2 shows non-limiting examples of different stabilization chemistries (1-10) that can be used, for example, to stabilize the 5′ and/or 3′-ends of siNA sequences of the invention (e.g., B or a portion thereof with respect to a siNA molecule having any of Formula A, B, C, D, E, F, G, H, I, J, K, L, M, N, O, or P); including (1) [3-3′]-inverted deoxyribose; (2) deoxyribonucleotide; (3) [5′-3′]-3′-deoxyribonucleotide; (4) [5′-3′]-ribonucleotide; (5) [5′-3′]-3′-O-methyl ribonucleotide; (6) 3′-glyceryl; (7) [3′-5′]-3′-deoxyribonucleotide; (8) [3′-3′]-deoxyribonucleotide; (9) [5′-2′]-de
  • FIG. 3 shows non-limiting examples of phosphorylated siNA molecules of the invention, including linear and duplex constructs and asymmetric derivatives thereof.
  • FIG. 4 shows non-limiting examples of chemically modified terminal phosphate groups of the invention. These terminal phosphate groups can be included at the 5′-end of the guide strand of any siNA molecule of the invention (e.g., any of Formula A, B, C, D, E, F, G, H, I, J, K, L, M, N, O, or P).
  • siNA molecule of the invention e.g., any of Formula A, B, C, D, E, F, G, H, I, J, K, L, M, N, O, or P.
  • FIG. 5 shows representative data indicating that 2′-deoxy-2′-fluoro content on the guide strand is important for effective knockdown of ApoB mRNA in vivo.
  • a PVE polymer conjugate delivery vehicle was used to administer the siNAs.
  • mRNA expression in liver tissue was measured at 2 days, 7 days and 14 days post dosing with 3mpk of siNA. Details on modification motifs and siNA sequences are provided in Table 1.
  • FIG. 6 shows representative data indicating that tetraGalNAc (single chemical entity, SCE) siNA conjugates containing alternating 2′-deoxy-2′-fluoro and 2′-O-methyl modification patterns on the guide strands have little impact on ApoB or SSB mRNA expression in vivo.
  • siNA conjugates were administered subcutaneously (30mpk for ApoB and 50mpk for SSB) and mRNA expression in liver tissue was measured 72 hours after dosing. Details on modification motifs and siNA sequences are provided in Table 1.
  • FIG. 7 shows representative data indicating the in vivo activity of subcutaneously delivered tetraGalNAc SCE siNA conjugates in mouse liver 72 hr post dosing.
  • the SSB sequences lacking phosphorothioate linkages at the 5′ end of the guide strand showed little to no activity while the ApoB sequence with 5′-terminal guide strand phosphorothioate linkages showed dose dependent knockdown. Details on modification motifs and siNA sequences are provided in Table 1.
  • FIG. 8 shows representative data indicating the in vivo activity of subcutaneously delivered tetraGalNAc SCE siNA conjugates in mouse liver 24 hr post dosing.
  • the SSB sequences containing phosphorothioate linkages at the 5′ end of the guide strand (pos 1-3) showed dose dependent knockdown. Details on modification motifs and siNA sequences are provided in Table 1.
  • FIG. 9 shows representative data indicating the in vivo activity of subcutaneously delivered tetraGalNAc SCE siNA conjugates in mouse liver 72 hr post dosing. Addition of greater than 3-fold more phosphorothioate content does not appear to improve the activity of the siNA conjugates. Note that all these sequence had three phosphorothioate linkages at 5′ end of the guide strand. Details on modification motifs and siNA sequences are provided in Table 1.
  • FIG. 10 shows representative data indicating the in vivo activity of subcutaneously delivered tetraGalNAc SCE siNA conjugates in mouse liver 72 hr post dosing.
  • the presence of a single phosphorothioate linkage at the first position at the 5′ end of the guide strand is important for in vivo activity.
  • Sequences with phosphorothioate linkages at other positions in the siNA but not at the first position of the 5′ end of the guide strand showed poor activity. Details on modification motifs and siNA sequences are provided in Table 1.
  • FIG. 11 shows representative data indicating that addition of three phosphorothioate modifications to the 5′ end of the guide strand (positions 1-3) significantly improves the knockdown of CTNNB1 mRNA in mice.
  • a 50mpk dose of siNA was administered as a tetraGalNAc SCE conjugate that was injected subcutaneously. Details on modification motifs and siNA sequences are provided in Table 1.
  • FIG. 12 shows representative data indicating the in vivo activity of subcutaneously delivered tetraGalNAc ApoC3 siNA conjugates in mouse liver 72 hr post dosing. Details on modification motifs and siNA sequences are provided in Table 1.
  • FIG. 13 shows representative data indicating the in vivo activity of subcutaneously delivered tetraGalNAc CTNNB1 siNA conjugates in rhesus liver at indicated times. Details on modification motifs and siNA sequences are provided in Table 1.
  • an abasic moiety of the invention is a ribose, deoxyribose, or dideoxyribose sugar.
  • acyclic nucleotide refers to its meaning as is generally accepted in the art.
  • the term generally refers to any nucleotide having an acyclic ribose sugar, for example where any of the ribose carbon/carbon or carbon/oxygen bonds are independently or in combination absent from the nucleotide.
  • alkyl refers to its meaning as is generally accepted in the art.
  • the term generally refers to a saturated or unsaturated hydrocarbon, including straight-chain, branched-chain, alkenyl, alkynyl groups and cyclic groups, but excludes aromatic groups. Notwithstanding the foregoing, alkyl also refers to non-aromatic heterocyclic groups.
  • the alkyl group has 1 to 12 carbons. More preferably, it is a lower alkyl of from 1 to 7 carbons, more preferably 1 to 4 carbons.
  • the alkyl group can be substituted or unsubstituted.
  • the substituted group(s) is preferably, hydroxyl, halogen, cyano, C1-C4 alkoxy, ⁇ O, ⁇ S, NO 2 , SH, NH 2 , or NR 1 R 2 , where R 1 and R 2 independently are H or C1-C4 alkyl.
  • agents that interfere with cell cycle checkpoints refers to compounds that inhibit protein kinases that transduce cell cycle checkpoint signals, thereby sensitizing the cancer cell to DNA damaging agents.
  • RTKs receptor tyrosine kinases
  • androgen receptor modulators refers to compounds that interfere or inhibit the binding of androgens to the receptor, regardless of mechanism.
  • angiogenesis inhibitors refers to compounds that inhibit the formation of new blood vessels, regardless of mechanism.
  • aryl refers to its meaning as is generally accepted in the art.
  • the term generally refers to an aromatic group that has at least one ring having a conjugated pi electron system and includes carbocyclic aryl, heterocyclic aryl and biaryl groups, all of which can be optionally substituted.
  • the preferred substituent(s) of aryl groups are halogen, trihalomethyl, hydroxyl, SH, OH, cyano, C1-C4 alkoxy, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, NH 2 , and NR 1 R 2 groups, where R 1 and R 2 independently are H or C1-C4 alkyl.
  • alkylaryl refers to its meaning as is generally accepted in the art.
  • the term generally refers to an alkyl group (as described above) covalently joined to an aryl group (as described above).
  • Carbocyclic aryl groups are groups wherein the ring atoms on the aromatic ring are all carbon atoms. The carbon atoms are optionally substituted.
  • Heterocyclic aryl groups are groups having from 1 to 3 heteroatoms as ring atoms in the aromatic ring and the remainder of the ring atoms are carbon atoms.
  • Suitable heteroatoms include oxygen, sulfur, and nitrogen, and examples of heterocyclic aryl groups having such heteroatoms include furanyl, thienyl, pyridyl, pyrrolyl, N-lower alkyl pyrrolo, pyrimidyl, pyrazinyl, imidazolyl and the like, all optionally substituted.
  • the alkyl group is a C1-C4 alkyl group.
  • amide refers to its meaning as is generally accepted in the art.
  • the term generally refers to an —C(O)—NH—R, where R is either alkyl, aryl, alkylaryl or hydrogen.
  • antisense region refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term refers to a nucleotide sequence of an siNA molecule having complementarity to a target nucleic acid sequence.
  • the antisense region of an siNA molecule can optionally comprise a nucleic acid sequence having complementarity to a sense region of the siNA molecule. In one embodiment, the antisense region of the siNA molecule is referred to as the antisense strand or guide strand.
  • asymmetric hairpin refers to a linear siNA molecule comprising an antisense region, a loop portion that can comprise nucleotides or non-nucleotides, and a sense region that comprises fewer nucleotides than the antisense region to the extent that the sense region has enough complementary nucleotides to base pair with the antisense region and form a duplex with loop.
  • an asymmetric hairpin siNA molecule of the invention can comprise an antisense region having length sufficient to mediate RNAi in a cell or in vitro system (e.g.
  • the asymmetric hairpin siNA molecule can also comprise a 5′-terminal phosphate group that can be chemically modified.
  • the loop portion of the asymmetric hairpin siNA molecule can comprise nucleotides, non-nucleotides, linker molecules, or conjugate molecules as described herein.
  • biodegradable refers to its meaning as is generally accepted in the art.
  • the term generally refers to degradation in a biological system, for example, enzymatic degradation or chemical degradation.
  • biodegradable linker refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term refers to a linker molecule that is designed to connect one molecule to another molecule, and which is susceptible to degradation in a biological system.
  • the linker can be a nucleic acid or non-nucleic acid based linker.
  • a biodegradable linker can be used to attach a ligand or biologically active molecule to an siNA molecule of the invention. Alternately, a biodegradable linker can be used to connect the sense and antisense strands of an siNA molecule of the invention.
  • the biodegradable linker is designed such that its stability can be modulated for a particular purpose, such as delivery to a particular tissue or cell type.
  • the stability of a nucleic acid-based biodegradable linker molecule can be modulated by using various chemistries, for example combinations of ribonucleotides, deoxyribonucleotides, and chemically modified nucleotides, such as 2′-O-methyl, 2′-fluoro, 2′-amino, 2′-O-amino, 2′-C-allyl, 2′-O-allyl, and other 2′-modified or base modified nucleotides.
  • the biodegradable nucleic acid linker molecule can be a dimer, trimer, tetramer or longer nucleic acid molecule, for example, an oligonucleotide of about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 nucleotides in length, or can comprise a single nucleotide with a phosphorus-based linkage, for example, a phosphoramidate or phosphodiester linkage.
  • the biodegradable nucleic acid linker molecule can also comprise nucleic acid backbone, nucleic acid sugar, or nucleic acid base modifications.
  • biologically active molecule refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term refers to compounds or molecules that are capable of eliciting or modifying a biological response in a system and/or are capable of modulating the pharmacokinetics and/or pharmacodynamics of other biologically active molecules.
  • biologically active molecules include siNA molecules alone or in combination with other molecules including, but not limited to therapeutically active molecules such as antibodies, cholesterol, hormones, antivirals, peptides, proteins, chemotherapeutics, small molecules, vitamins, co-factors, nucleosides, nucleotides, oligonucleotides, enzymatic nucleic acids, antisense nucleic acids, triplex forming oligonucleotides, polyamines, polyamides, polyethylene glycol, other polyethers, 2-5A chimeras, siNA, dsRNA, allozymes, aptamers, decoys and analogs thereof.
  • therapeutically active molecules such as antibodies, cholesterol, hormones, antivirals, peptides, proteins, chemotherapeutics, small molecules, vitamins, co-factors, nucleosides, nucleotides, oligonucleotides, enzymatic nucleic acids, antisense nucleic acids, triplex forming oligon
  • biological system refers to its meaning as is generally accepted in the art.
  • the term generally refers to material, in a purified or unpurified form, from biological sources including, but not limited to, human or animal, wherein the system comprises the components required for RNAi activity.
  • the phrase includes, for example, a cell, tissue, subject, or organism, or extract thereof.
  • the term also includes reconstituted material from a biological source.
  • blunt end refers to its meaning as is generally accepted in the art.
  • the term refers to termini of a double-stranded siNA molecule having no overhanging nucleotides.
  • the two strands of a double-stranded siNA molecule having blunt ends align with each other with matched base-pairs without overhanging nucleotides at the termini.
  • a siNA duplex molecule of the invention can comprise blunt ends at one or both termini of the duplex, such as termini located at the 5′-end of the antisense strand, the 5′-end of the sense strand, or both termini of the duplex.
  • cap also referred to herein as “terminal cap,” as used herein refers to its meaning as is generally accepted in the art.
  • the term refers to a moiety, which can be a chemically modified nucleotide or non-nucleotide that can be incorporated at one or more termini of one or more nucleic acid molecules of the invention.
  • These terminal modifications protect the nucleic acid molecule from exonuclease degradation, and can help in delivery and/or localization within a cell.
  • the cap can be present at the 5′-terminus (5′-cap) or at the 3′-terminal (3′-cap) or can be present on both termini of any nucleic acid molecule of the invention.
  • a cap can be present at the 5′-end, 3-end and/or 5′ and 3′-ends of the sense strand of a nucleic acid molecule of the invention. Additionally, a cap can optionally be present at the 3′-end of the antisense strand of a nucleic acid molecule of the invention.
  • the 5′-cap includes, but is not limited to a polymer; a ligand; locked nucleic acid (LNA); glyceryl; an abasic ribose residue (moiety); inverted deoxy abasic residue (moiety); an inverted nucleotide; 4′,5′-methylene nucleotide; 1-(beta-D-erythrofuranosyl) nucleotide, 4′-thio nucleotide; carbocyclic nucleotide; 1,5-anhydrohexitol nucleotide; L-nucleotides; alpha-nucleotides; modified base nucleotide; phosphorodithioate linkage; threo-pentofuranosyl nucleotide; acyclic 3′,4′-seco nucleotide; acyclic 3,4-dihydroxybutyl nucleotide; acyclic 3,5-d
  • Non-limiting examples of the 3′-cap include, but are not limited to, a polymer; a ligand; locked nucleic acid (LNA); glyceryl; an abasic ribose residue (moiety); inverted deoxy abasic residue (moiety); an inverted nucleotide; 4′,5′-methylene nucleotide; 1-(beta-D-erythrofuranosyl) nucleotide; 4′-thio nucleotide; carbocyclic nucleotide; 5′-amino-alkyl phosphate; 1,3-diamino-2-propyl phosphate; 3-aminopropyl phosphate; 6-aminohexyl phosphate; 1,2-aminododecyl phosphate; hydroxypropyl phosphate; 1,5-anhydrohexitol nucleotide; L-nucleotide; alpha-nucleotide; modified
  • a siNA molecule of the invention having Formula (A) can comprise one or more terminal cap molecules as described above (designated as B) that comprises or includes a covalent attachment to a polymer or ligand via a linker molecule as described herein or as is otherwise known in the art.
  • B terminal cap molecules as described above
  • linker molecule as described herein or as is otherwise known in the art.
  • Non-limiting examples of such linkers are provided in the examples and descriptions herein.
  • the term “cell” as used herein refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term is used in its usual biological sense, and does not refer to an entire multicellular organism, e.g., specifically does not refer to a human being.
  • the cell can be present in an organism, e.g., birds, plants and mammals, such as humans, cows, sheep, apes, monkeys, swine, dogs, and cats.
  • the cell can be prokaryotic (e.g., bacterial cell) or eukaryotic (e.g., mammalian or plant cell).
  • the cell can be of somatic or germ line origin, totipotent or pluripotent, dividing or non-dividing.
  • the cell can also be derived from or can comprise a gamete or embryo, a stem cell, or a fully differentiated cell.
  • chemical modification refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term refers to any modification of the chemical structure of the nucleotides that differs from nucleotides of native siRNA or RNA in general.
  • the term “chemical modification” encompasses the addition, substitution, or modification of native siRNA or RNA at the sugar, base, or internucleotide linkage, as described herein or as is otherwise known in the art.
  • the term “chemical modification” can refer to certain forms of RNA that are naturally occurring in certain biological systems, for example 2′-O-methyl modifications or inosine modifications.
  • nucleic acid molecules of the invention refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the terms generally refer to the formation or existence of hydrogen bond(s) between one nucleic acid sequence and another nucleic acid sequence by either traditional Watson-Crick or other non-traditional types of bonding as described herein. In reference to the nucleic molecules of the present invention, the binding free energy for a nucleic acid molecule with its complementary sequence is sufficient to allow the relevant function of the nucleic acid to proceed, e.g., RNAi activity.
  • Partial complementarity can include various mismatches or non-based paired nucleotides (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more mismatches, non-nucleotide linkers, or non-based paired nucleotides) within the nucleic acid molecule, which can result in bulges, loops, or overhangs that result between the sense strand or sense region and the antisense strand or antisense region of the nucleic acid molecule or between the antisense strand or antisense region of the nucleic acid molecule and a corresponding target nucleic acid molecule.
  • mismatches or non-based paired nucleotides e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more mismatches, non-nucleotide linkers, or non-based paired nucleotides
  • Such partial complementarity can be represented by a % complementarity that is determined by the number of non-base paired nucleotides, i.e., about 50%, 60%, 70%, 80%, 90% etc. depending on the total number of nucleotides involved.
  • Such partial complementarity is permitted to the extent that the nucleic acid molecule (e.g., siNA) maintains its function, for example the ability to mediate sequence specific RNAi.
  • compositions or “formulation” as used herein refer to their generally accepted meaning in the art. These terms generally refer to a composition or formulation, such as in a pharmaceutically acceptable carrier or diluent, in a form suitable for administration, e.g., systemic or local administration, into a cell or subject, including, for example, a human. Suitable forms, in part, depend upon the use or the route of entry, for example oral, transdermal, inhalation, or by injection. Such forms should not prevent the composition or formulation from reaching a target cell (i.e., a cell to which the negatively charged nucleic acid is desirable for delivery). For example, compositions injected into the blood stream should be soluble.
  • compositions include formulations for human and veterinary use.
  • agents suitable for formulation with the nucleic acid molecules of the instant invention include: Lipid Nanoparticles (see for example Semple et al., 2010, Nat Biotechnol ., February; 28(2):172-6.); P-glycoprotein inhibitors (such as Pluronic P85); biodegradable polymers, such as poly (DL-lactide-coglycolide) microspheres for sustained release delivery (Emerich, D F et al, 1999, Cell Transplant, 8, 47-58); and loaded nanoparticles, such as those made of polybutylcyanoacrylate.
  • a “pharmaceutically acceptable composition” or “pharmaceutically acceptable formulation” can refer to a composition or formulation that allows for the effective distribution of the nucleic acid molecules of the instant invention to the physical location most suitable for their desired activity.
  • cytotoxic/cytostatic agents refer to compounds that cause cell death or inhibit cell proliferation primarily by interfering directly with the cell's functioning or inhibit or interfere with cell mytosis, including alkylating agents, tumor necrosis factors, intercalators, hypoxia activatable compounds, microtubule inhibitors/microtubule-stabilizing agents, inhibitors of mitotic kinesins, inhibitors of histone deacetylase, inhibitors of kinases involved in mitotic progression, antimetabolites; biological response modifiers; hormonal/anti-hormonal therapeutic agents, hematopoietic growth factors, monoclonal antibody targeted therapeutic agents, topoisomerase inhibitors, proteasome inhibitors and ubiquitin ligase inhibitors.
  • estrogen receptor modulators refers to compounds that interfere with or inhibit the binding of estrogen to the receptor, regardless of mechanism.
  • gene or “target gene” as used herein refers to their meaning as is generally accepted in the art.
  • the terms generally refer a nucleic acid (e.g., DNA or RNA) sequence that comprises partial length or entire length coding sequences necessary for the production of a polypeptide.
  • the target gene can also include the UTR or non-coding region of the nucleic acid sequence.
  • a gene or target gene can also encode a functional RNA (fRNA) or non-coding RNA (ncRNA), such as small temporal RNA (stRNA), micro RNA (miRNA), small nuclear RNA (snRNA), short interfering RNA (siRNA), small nucleolar RNA (snRNA), ribosomal RNA (rRNA), transfer RNA (tRNA) and precursor RNAs thereof.
  • fRNA functional RNA
  • ncRNA non-coding RNA
  • stRNA small temporal RNA
  • miRNA micro RNA
  • snRNA small nuclear RNA
  • siRNA small nucleolar RNA
  • rRNA ribosomal RNA
  • tRNA transfer RNA
  • siNA molecules targeting fRNA and ncRNA can also be used to manipulate or alter the genotype or phenotype of a subject, organism or cell, by intervening in cellular processes such as genetic imprinting, transcription, translation, or nucleic acid processing (e.g., transamination, methylation etc.).
  • the target gene can be a gene derived from a cell, an endogenous gene, a transgene, or exogenous genes such as genes of a pathogen, for example a virus, which is present in the cell after infection thereof.
  • the cell containing the target gene can be derived from or contained in any organism, for example a plant, animal, protozoan, virus, bacterium, or fungus.
  • Non-limiting examples of plants include monocots, dicots, or gymnosperms.
  • Non-limiting examples of animals include vertebrates or invertebrates.
  • Non-limiting examples of fungi include molds or yeasts.
  • gene targets contemplated herein are also referred to herein generally as “target” sequences (including the target sequences listed by GenBank Accession numbers in U.S. Ser. No. 60/363,124, incorporated by reference herein).
  • HMG-CoA reductase inhibitors refers to inhibitors of 3-hydroxy-3-methylglutaryl-CoA reductase.
  • HMG-CoA reductase inhibitor as used herein includes all pharmaceutically acceptable lactone and open-acid forms (i.e., where the lactone ring is opened to form the free acid) as well as salt and ester forms of compounds that have HMG-CoA reductase inhibitory activity, and therefore the use of such salts, esters, open-acid and lactone forms is included within the scope of this invention.
  • highly conserved sequence region refers to a nucleotide sequence of one or more regions in a target gene that does not vary significantly from one generation to the other or from one biological system to the other.
  • homologous sequence refers to its meaning as is generally accepted in the art.
  • the term generally refers a nucleotide sequence that is shared by one or more polynucleotide sequences, such as genes, gene transcripts and/or non-coding polynucleotides.
  • a homologous sequence can be a nucleotide sequence that is shared by two or more genes encoding related but different proteins, such as different members of a gene family, different protein epitopes, different protein isoforms or completely divergent genes.
  • a homologous sequence can be a nucleotide sequence that is shared by two or more non-coding polynucleotides, such as noncoding DNA or RNA, regulatory sequences, introns, and sites of transcriptional control or regulation. Homologous sequences can also include sequence regions shared by more than one polynucleotide sequence.
  • the term “perfect homology” (or “perfectly homologous”) as used herein refers to complete (100%) homology or “identity” between a reference sequence and a subject nucleic acid sequence.
  • homologous sequences are also contemplated by and within the scope of the instant invention (e.g., at least 95%, 94%, 93%, 92%, 91%, 90%, 89%, 88%, 87%, 86%, 85%, 84%, 83%, 82%, 81%, 80% etc.). Percent homology is the number of matching nucleotides between two sequences divided by the total length being compared, multiplied by 100.
  • RNAi activity refers to an increase in RNAi activity measured in vitro and/or in vivo, where the RNAi activity is a reflection of both the ability of the siNA to mediate RNAi and the stability of the siNAs of the invention.
  • the product of these activities can be increased in vitro and/or in vivo compared to an all RNA siNA or an siNA containing a plurality of ribonucleotides.
  • the activity or stability of the siNA molecule can be decreased (i.e., less than ten-fold), but the overall activity of the siNA molecule is enhanced in vitro and/or in vivo.
  • inhibitor refers to its meaning as is generally accepted in the art.
  • the term generally refers the reduction in the expression of the gene, or level of RNA molecules or equivalent RNA molecules encoding one or more proteins or protein subunits, or activity of one or more proteins or protein subunits, below that observed in the absence of the nucleic acid molecules (e.g., siNA) of the invention.
  • Down-regulation can also be associated with post-transcriptional silencing, such as, RNAi mediated cleavage or by alteration in DNA methylation patterns or DNA chromatin structure.
  • Inhibition, down-regulation or reduction with an siNA molecule can be in reference to an inactive molecule, an attenuated molecule, an siNA molecule with a scrambled sequence, or an siNA molecule with mismatches or alternatively, it can be in reference to the system in the absence of the nucleic acid.
  • inhibitors of cell proliferation and survival signaling pathway refers to pharmaceutical agents that inhibit cell surface receptors and signal transduction cascades downstream of those surface receptors.
  • integrated circuit blockers refers to compounds which selectively antagonize, inhibit or counteract binding of a physiological ligand to the ⁇ ⁇ ⁇ 3 integrin, to compounds which selectively antagonize, inhibit or counteract binding of a physiological ligand to the ⁇ ⁇ ⁇ 5 integrin, to compounds which antagonize, inhibit or counteract binding of a physiological ligand to both the ⁇ ⁇ ⁇ 3 integrin and the ⁇ ⁇ ⁇ 5 integrin, and to compounds which antagonize, inhibit or counteract the activity of the particular integrin(s) expressed on capillary endothelial cells.
  • the term also refers to antagonists of the ⁇ ⁇ ⁇ 6 ⁇ ⁇ ⁇ 8 ⁇ 1 ⁇ 1 ⁇ 2 ⁇ 2 ⁇ 5 ⁇ 1 ⁇ 6 ⁇ 1 and ⁇ 6 ⁇ 4 integrins.
  • the term also refers to antagonists of any combination of ⁇ ⁇ ⁇ 3 , ⁇ ⁇ ⁇ 5 ⁇ ⁇ ⁇ 6 ⁇ ⁇ ⁇ 8 ⁇ 1 ⁇ 1 ⁇ 2 ⁇ 1 ⁇ 5 ⁇ 1 ⁇ 6 ⁇ 1 and ⁇ 6 ⁇ 4 integrins.
  • internucleoside linkage or “internucleoside linker” or “internucleotide linkage” or “internucleotide linker” are used herein interchangeably and refer to any linker or linkage between two nucleoside units, as is known in the art, including, for example, but not limitation, phosphate, analogs of phosphate, phosphonate, guanidinium, hydroxylamine, hydroxylhydrazinyl, amide, carbamate, alkyl, and substituted alkyl linkages.
  • the internucleoside linkages constitute the backbone of a nucleic acid molecule.
  • ligand refers to such compounds and compositions as are generally known in the art. Non-limiting examples of such ligands are described herein including in the documents specifically incorporated by reference herein.
  • a siNA molecule of the invention can be formulated or administered with any covalently linked ligand as described herein or otherwise known in the art.
  • linker refers to its meaning as is generally known in the art. Non-limiting examples of linkers are described herein, for example in Table 8 and including in the documents specifically incorporated by reference herein.
  • lipid nanoparticle refers to lipid-based compositions and formulations as are generally known in the art. Non-limiting examples of such LNPs are described herein including in the documents specifically incorporated by reference herein.
  • a siNA molecule of the invention can be formulated or administered with any LNP as described herein or otherwise known in the art.
  • mammalian or “mammal” as used herein refers to its meaning as is generally accepted in the art.
  • the term generally refers to any warm blooded vertebrate species, such as a human, mouse, rat, dog, cat, hamster, guinea pig, rabbit, livestock, and the like.
  • MDI refers to a unit comprising a can, a secured cap covering the can and a formulation metering valve situated in the cap.
  • MDI systems includes a suitable channeling device. Suitable channeling devices comprise for example, a valve actuator and a cylindrical or cone-like passage through which medicament can be delivered from the filled canister via the metering valve to the nose or mouth of a patient such as a mouthpiece actuator.
  • microRNA or “miRNA” as used herein refers to its meaning as is generally accepted in the art.
  • the term generally refers to an endogenous short RNA molecule found in eukaryotes that is involved in RNA-based gene regulation.
  • a representative set of known endogenous miRNA species is described in the publicly available miRBase sequence database as described in Griffith-Jones et al., Nucleic Acids Research, 2004, 32:D109-D111 and Griffith-Jones et al., Nucleic Acids Research, 2006, 34:D 140-D144, accessible on the World Wide Web at the Wellcome Trust Sanger Institute website.
  • Each mature miRNA is partially complementary to one or more messenger RNA (mRNA) molecules, which are also called “miRNA targets,” thereby regulating the expression of genes associated with the miRNA targets.
  • mRNA messenger RNA
  • modulate refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term refers to when the expression of a gene, or level of one or more RNA molecules (coding or non-coding), or activity of one or more RNA molecules or proteins or protein subunits, is up-regulated or down-regulated, such that expression, level, or activity is greater than or less than that observed in the absence of the molecule that effects modulation.
  • the term “modulate” in some embodiments can refer to inhibition and in other embodiments can refer to potentiation or up-regulation, e.g., of gene expression.
  • modified nucleotide refers to its meaning as is generally accepted in the art.
  • the term generally refers a nucleotide, which contains a modification in the chemical structure of the base, sugar and/or phosphate of the unmodified (or natural) nucleotide as is generally known in the art.
  • modified nucleotides are described herein and in U.S. application Ser. No. 12/064,014.
  • NSAIDs that are selective COX-2 inhibitors refers to NSAIDs, which possess a specificity for inhibiting COX-2 over COX-1 of at least 100 fold as measured by the ratio of IC 50 for COX-2 over IC 50 for COX-1 evaluated by cell or microsomal assays.
  • non-base paired refers to nucleotides that are not base paired between the sense strand or sense region and the antisense strand or antisense region of an double-stranded siNA molecule; and can include for example, but not limitation, mismatches, overhangs, single stranded loops, etc.
  • non-nucleotide refers to any group or compound which can be incorporated into a nucleic acid chain in the place of one or more nucleotide units, such as for example but not limitation abasic moieties or alkyl chains.
  • the group or compound is “abasic” in that it does not contain a commonly recognized nucleotide base, such as adenosine, guanine, cytosine, uracil or thymine and therefore lacks a nucleobase at the 1′-position.
  • nucleotide is used as is generally recognized in the art. Nucleotides generally comprise a nucleobase, a sugar, and an internucleoside linkage, e.g., a phosphate.
  • the base can be a natural bases (standard), modified bases, or a base analog, as are well known in the art. Such bases are generally located at the 1′ position of a nucleotide sugar moiety.
  • nucleotides can be unmodified or modified at the sugar, internucleoside linkage, and/or base moiety, (also referred to interchangeably as nucleotide analogs, modified nucleotides, non-natural nucleotides, non-standard nucleotides and others; see, for example, U.S. application Ser. No. 12/064,014.
  • overhang refers to its meaning as is generally accepted in the art. With reference to exemplary double stranded nucleic acid molecules, the term generally refers to the terminal portion of a nucleotide sequence that is not base paired between the two strands of a double-stranded nucleic acid molecule. Overhangs, when present, are typically at the 3′-end of one or both strands in a siNA duplex.
  • parenteral refers to its meaning as is generally accepted in the art.
  • the term generally refers methods or techniques of administering a molecule, drug, agent, or compound in a manner other than through the digestive tract, and includes epicutaneous, subcutaneous, intravascular (e.g., intravenous), intramuscular, or intrathecal injection or infusion techniques and the like.
  • pathway target refers to any target involved in pathways of gene expression or activity.
  • any given target can have related pathway targets that can include upstream, downstream, or modifier genes in a biologic pathway.
  • pathway target genes can provide additive or synergistic effects in the treatment of diseases, conditions, and traits herein.
  • phosphorothioate refers to an internucleotide phosphate linkage comprising one or more sulfur atoms in place of an oxygen atom. Hence, the term phosphorothioate refers to both phosphorothioate and phosphorodithioate internucleotide linkages.
  • polymer refers to polymeric compounds, compositions and formulations as are generally known in the art. Non-limiting examples of such polymers, including polymeric delivery systems are described herein including in the documents specifically incorporated by reference herein.
  • a siNA molecule of the invention can be formulated or administered with any polymer as described herein or otherwise known in the art.
  • position 1 refers to the position of the first nucleotide at the end of a strand, e.g., antisense strand. All positions referred to herein are the positions of a nucleotide counting from the end of a strand, for example, positions 1-3 from the 5′ end of the antisense strand, refer to the three nucleotides at positions 1, 2, and 3 counting from the 5′ end of the antisense strand.
  • ribonucleotide refers to its meaning as is generally accepted in the art.
  • the term generally refers to a nucleotide with a hydroxyl group at the 2′ position of a (3-D-ribofuranose moiety.
  • RNA refers to its generally accepted meaning in the art.
  • RNA refers to a molecule comprising at least one ribofuranoside moiety.
  • the term can include double-stranded RNA, single-stranded RNA, isolated RNA such as partially purified RNA, essentially pure RNA, synthetic RNA, recombinantly produced RNA, as well as altered RNA that differs from naturally occurring RNA by the addition, deletion, substitution and/or alteration of one or more nucleotides.
  • Such alterations can include addition of non-nucleotide material, such as to the end(s) of the siNA or internally, for example at one or more nucleotides of the RNA.
  • Nucleotides in the RNA molecules of the instant invention can also comprise non-standard nucleotides, such as non-naturally occurring nucleotides or chemically synthesized nucleotides or deoxynucleotides. These altered RNAs can be referred to as analogs or analogs of naturally-occurring RNA.
  • RNA interference refers to the biological process of inhibiting or down regulating gene expression in a cell, as is generally known in the art, and which is mediated by short interfering nucleic acid molecules, see for example Zamore and Haley, 2005, Science, 309, 1519-1524; Vaughn and Martienssen, 2005, Science, 309, 1525-1526; Zamore et al., 2000, Cell, 101, 25-33; Bass, 2001, Nature, 411, 428-429; Elbashir et al., 2001, Nature, 411, 494-498; and Kreutzer et al., International PCT Publication No.
  • RNAi is meant to be equivalent to other terms used to describe sequence specific RNA interference, such as post transcriptional gene silencing, translational inhibition, transcriptional inhibition, or epigenetics.
  • siNA molecules of the invention can be used to epigenetically silence genes at either the post-transcriptional level or the pre-transcriptional level.
  • epigenetic modulation of gene expression by siNA molecules of the invention can result from siNA mediated modification of chromatin structure or methylation patterns to alter gene expression (see, for example, Verdel et al., 2004, Science, 303, 672-676; Pal-Bhadra et al., 2004, Science, 303, 669-672; Allshire, 2002, Science, 297, 1818-1819; Volpe et al., 2002, Science, 297, 1833-1837; Jenuwein, 2002, Science, 297, 2215-2218; and Hall et al., 2002, Science, 297, 2232-2237).
  • modulation of gene expression by siNA molecules of the invention can result from siNA mediated cleavage of RNA (either coding or non-coding RNA) via RISC, or via translational inhibition, as is known in the art or modulation can result from transcriptional inhibition (see for example Janowski et al., 2005, Nature Chemical Biology, 1, 216-222).
  • RNAi inhibitor refers to any molecule that can down regulate, reduce or inhibit RNA interference function or activity in a cell or organism.
  • An RNAi inhibitor can down regulate, reduce or inhibit RNAi (e.g., RNAi mediated cleavage of a target polynucleotide, translational inhibition, or transcriptional silencing) by interaction with or interfering with the function of any component of the RNAi pathway, including protein components such as RISC, or nucleic acid components such as miRNAs or siRNAs.
  • a RNAi inhibitor can be an siNA molecule, an antisense molecule, an aptamer, or a small molecule that interacts with or interferes with the function of RISC, a miRNA, or an siRNA or any other component of the RNAi pathway in a cell or organism.
  • RNAi e.g., RNAi mediated cleavage of a target polynucleotide, translational inhibition, or transcriptional silencing
  • a RNAi inhibitor of the invention can be used to modulate (e.g., up-regulate or down regulate) the expression of a target gene.
  • sense region refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term refers to a nucleotide sequence of an siNA molecule having complementarity to an antisense region of the siNA molecule.
  • the sense region of an siNA molecule can comprise a nucleic acid sequence having homology or sequence identity with a target nucleic acid sequence.
  • the sense region of the siNA molecule is also referred to as the sense strand or passenger strand.
  • short interfering nucleic acid refers to any nucleic acid molecule capable of inhibiting or down regulating gene expression or viral replication by mediating RNA interference (“RNAi”) or gene silencing in a sequence-specific manner. These terms can refer to both individual nucleic acid molecules, a plurality of such nucleic acid molecules, or pools of such nucleic acid molecules.
  • RNAi RNA interference
  • the siNA can be a double-stranded nucleic acid molecule comprising self-complementary sense and antisense strands, wherein the antisense strand comprises a nucleotide sequence that is complementary to a nucleotide sequence in a target nucleic acid molecule or a portion thereof and the sense strand comprises a nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof.
  • the siNA can be a polynucleotide with a duplex, asymmetric duplex, hairpin or asymmetric hairpin secondary structure, having self-complementary sense and antisense regions, wherein the antisense region comprises a nucleotide sequence that is complementary to a nucleotide sequence in a separate target nucleic acid molecule or a portion thereof and the sense region comprises a nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof.
  • the siNA can be a circular single-stranded polynucleotide having two or more loop structures and a stem comprising self-complementary sense and antisense regions, wherein the antisense region comprises nucleotide sequence that is complementary to a nucleotide sequence in a target nucleic acid molecule or a portion thereof and the sense region comprises a nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof, and wherein the circular polynucleotide can be processed either in vivo or in vitro to generate an active siNA molecule capable of mediating RNAi.
  • the siNA can also comprise a single-stranded polynucleotide having a nucleotide sequence complementary to nucleotide sequence in a target nucleic acid molecule or a portion thereof (for example, where such siNA molecule does not require the presence within the siNA molecule of a nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof), wherein the single-stranded polynucleotide can further comprise a terminal phosphate group, such as a 5′-phosphate (see for example, Martinez et al., 2002, Cell, 110, 563-574 and Schwarz et al., 2002, Molecular Cell, 10, 537-568), or 5′,3′-diphosphate.
  • a terminal phosphate group such as a 5′-phosphate (see for example, Martinez et al., 2002, Cell, 110, 563-574 and Schwarz et al., 2002, Molecular Cell, 10, 537-568), or 5′,3′-di
  • subject refers to its meaning as is generally accepted in the art.
  • the term generally refers an organism to which the nucleic acid molecules of the invention can be administered.
  • a subject can be a mammal or mammalian cells, including a human or human cells.
  • the term also refers to an organism, which is a donor or recipient of explanted cells or the cells themselves.
  • systemic administration refers to its meaning as is generally accepted in the art.
  • the term generally refers in vivo systemic absorption or accumulation of drugs in the blood stream followed by distribution throughout the entire body.
  • target refers, to any protein, peptide, or polypeptide, such as encoded by any gene in the GenBank database, including those described herein and/or in U.S. Provisional Patent Application No. 60/363,124, U.S. application Ser. No. 10/923,536 and/or PCT/US03/05028, all of which are incorporated herein by reference for purposes of identifying such targets.
  • target also refers to one or more genes, nucleic acid sequences, or target polynucleotide sequences encoding any target protein, peptide, or polypeptide, such as proteins, peptides, or polypeptides encoded by the genes in the Genebank database or sequences having GenBank Accession Nos.
  • target of interest can include target polynucleotide sequences, such as target DNA or target RNA.
  • target is also meant to include other sequences, such as differing isoforms, mutant target genes, splice variants of target polynucleotides, target polymorphisms, and non-coding (e.g., ncRNA, miRNA, stRNA) or other regulatory polynucleotide sequences as described herein.
  • a double stranded nucleic acid molecule of the invention having complementarity to a target RNA can be used to inhibit or down regulate miRNA or other ncRNA activity.
  • inhibition of miRNA or ncRNA activity can be used to down regulate or inhibit gene expression (e.g., gene targets described herein or otherwise known in the art) that is dependent on miRNA or ncRNA activity.
  • inhibition of miRNA or ncRNA activity by double stranded nucleic acid molecules of the invention e.g.
  • siNA having complementarity to the miRNA or ncRNA can be used to up regulate or promote target gene expression (e.g., gene targets described herein or otherwise known in the art) where the expression of such genes is down regulated, suppressed, or silenced by the miRNA or ncRNA.
  • target gene expression e.g., gene targets described herein or otherwise known in the art
  • Such up-regulation of gene expression can be used to treat diseases and conditions associated with a loss of function or haploinsufficiency as are generally known in the art.
  • target site refers to its meaning as is generally accepted in the art.
  • the term generally refers to a sequence within a target nucleic acid molecule, (e.g., RNA) that is “targeted”, e.g., for cleavage mediated by an siNA construct, which contains sequences within its antisense region that are complementary to the target sequence.
  • terapéuticaally effective amount refers to its meaning as is generally accepted in the art.
  • the term generally refers to the amount of the compound or composition that will elicit the biological or medical response of a cell, tissue, system, animal or human that is be sought by the researcher, veterinarian, medical doctor or other clinician. For example, if a given clinical treatment is considered effective when there is at least a 25% reduction in a measurable parameter associated with a disease or disorder, a therapeutically effective amount of a drug for the treatment of that disease or disorder is that amount necessary to effect at least a 25% reduction in that parameter.
  • universal base refers to its meaning as is generally accepted in the art.
  • the term universal base generally refers to nucleotide base analogs that form base pairs with each of the natural DNA/RNA bases with little or no discrimination between them.
  • Non-limiting examples of universal bases include C-phenyl, C-naphthyl and other aromatic derivatives, inosine, azole carboxamides, and nitroazole derivatives such as 3-nitropyrrole, 4-nitroindole, 5-nitroindole, and 6-nitroindole as known in the art (see for example, Loakes, 2001 , Nucleic Acids Research, 29, 2437-2447).
  • up-regulate refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term refers to an increase in the expression of a gene, or level of RNA molecules or equivalent RNA molecules encoding one or more proteins or protein subunits, or activity of one or more RNAs, proteins or protein subunits, above that observed in the absence of the nucleic acid molecules (e.g., siNA) of the invention. In certain instances, up-regulation or promotion of gene expression with an siNA molecule is above that level observed in the presence of an inactive or attenuated molecule.
  • siNA nucleic acid molecules
  • up-regulation or promotion of gene expression with siNA molecules is above that level observed in the presence of, for example, an siNA molecule with scrambled sequence or with mismatches.
  • up-regulation or promotion of gene expression with a nucleic acid molecule of the instant invention is greater in the presence of the nucleic acid molecule than in its absence.
  • up-regulation or promotion of gene expression is associated with inhibition of RNA mediated gene silencing, such as RNAi mediated cleavage or silencing of a coding or non-coding RNA target that down regulates, inhibits, or silences the expression of the gene of interest to be up-regulated.
  • the down regulation of gene expression can, for example, be induced by a coding RNA or its encoded protein, such as through negative feedback or antagonistic effects.
  • the down regulation of gene expression can, for example, be induced by a non-coding RNA having regulatory control over a gene of interest, for example by silencing expression of the gene via translational inhibition, chromatin structure, methylation, RISC mediated RNA cleavage, or translational inhibition.
  • inhibition or down regulation of targets that down regulate, suppress, or silence a gene of interest can be used to up-regulate expression of the gene of interest toward therapeutic use.
  • vector refers to its meaning as is generally accepted in the art.
  • vector generally refers to any nucleic acid- and/or viral-based expression system or technique used to deliver one or more nucleic acid molecules.
  • the invention features a composition comprising a double-stranded short interfering nucleic acid (siNA) molecule that modulates the expression and/or activity of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure represented by formula (A):
  • siNA short interfering nucleic acid
  • each N is independently a nucleotide wherein one or more Ns may be substituted with a non-nucleotide moiety so long as RNAi activity is maintained;
  • each B 1 , B 2 , and B 3 is independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker, wherein any of B 1 , B 2 , and/or B 3 is
  • the invention features a composition comprising a double-stranded short interfering nucleic acid (siNA) molecule that modulates the expression and/or activity of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure represented by formula (B):
  • siNA short interfering nucleic acid
  • each N is independently a nucleotide wherein one or more Ns may be substituted with a non-nucleotide moiety so long as RNAi activity is maintained;
  • each B 1 , B 2 , and B 3 is independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker, wherein any of B 1 , B 2 , and/or B 3 is
  • the invention features a composition comprising a double-stranded short interfering nucleic acid (siNA) molecule that modulates the expression and/or activity of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure represented by formula (C):
  • siNA short interfering nucleic acid
  • each N is independently a nucleotide wherein one or more Ns may be substituted with a non-nucleotide moiety so long as RNAi activity is maintained;
  • each B 1 , B 2 , and B 3 is independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker, wherein any of B 1 , B 2 , and/or B 3 is
  • the invention features a composition comprising a double-stranded short interfering nucleic acid (siNA) molecule that modulates the expression and/or activity of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure represented by formula (D):
  • siNA short interfering nucleic acid
  • each N is independently a nucleotide wherein one or more Ns may be substituted with a non-nucleotide moiety so long as RNAi activity is maintained;
  • each B 1 , B 2 , and B 3 is independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker, wherein any of B 1 , B 2 , and/or B 3 is
  • the invention features a composition comprising a double-stranded short interfering nucleic acid (siNA) molecule that modulates the expression and/or activity of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure represented by formula (E):
  • siNA short interfering nucleic acid
  • each N is independently a nucleotide wherein one or more Ns may be substituted with a non-nucleotide moiety so long as RNAi activity is maintained;
  • each B 1 , B 2 , and B 3 is independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker, wherein any of B 1 , B 2 , and/or B 3 is
  • the invention features a composition comprising a double-stranded short interfering nucleic acid (siNA) molecule that modulates the expression and/or activity of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure represented by formula (F):
  • siNA short interfering nucleic acid
  • each N is independently a nucleotide wherein one or more Ns may be substituted with a non-nucleotide moiety so long as RNAi activity is maintained;
  • each B 1 , B 2 , and B 3 is independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker, wherein any of B 1 , B 2 , and/or B 3 is
  • the invention features a composition comprising a double-stranded short interfering nucleic acid (siNA) molecule that modulates the expression and/or activity of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure represented by formula (G):
  • siNA short interfering nucleic acid
  • each N is independently a nucleotide wherein one or more Ns may be substituted with a non-nucleotide moiety so long as RNAi activity is maintained;
  • each B 1 , B 2 , and B 3 is independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker, wherein any of B 1 , B 2 , and/or B 3 is
  • the invention features a composition comprising a double-stranded short interfering nucleic acid (siNA) molecule that modulates the expression and/or activity of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure represented by formula (H):
  • siNA short interfering nucleic acid
  • each N is independently a nucleotide wherein one or more Ns may be substituted with a non-nucleotide moiety so long as RNAi activity is maintained;
  • each B 1 , B 2 , and B 3 is independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker, wherein any of B 1 , B 2 , and/or B 3 is
  • the invention features a composition comprising a double-stranded short interfering nucleic acid (siNA) molecule that modulates the expression and/or activity of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure represented by formula (I):
  • siNA short interfering nucleic acid
  • each N is independently a nucleotide wherein one or more Ns may be substituted with a non-nucleotide moiety so long as RNAi activity is maintained;
  • each B 1 , B 2 , and B 3 is independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker, wherein any of B 1 , B 2 , and/or B 3 is
  • the invention features a composition comprising a double-stranded short interfering nucleic acid (siNA) molecule that modulates the expression and/or activity of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure represented by formula (J):
  • siNA short interfering nucleic acid
  • each N is independently a nucleotide wherein one or more Ns may be substituted with a non-nucleotide moiety so long as RNAi activity is maintained;
  • each B 1 , B 2 , and B 3 is independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker, wherein any of B 1 , B 2 , and/or B 3 is
  • each (N) of the passenger strand of a composition according to any of Formula A, B, C, D, E, F, G, H, I or J is independently selected from 2′-O-alkyl, 2′-deoxy-2′-fluoro, 2′-deoxy, and/or LNA modified nucleotide.
  • each (N) of the passenger strand of a composition according to any of Formula A, B, C, D, E, F, G, H, I or J is a 2′-O-alkyl modified nucleotide.
  • each (N) of the passenger strand of a composition according to any of Formula A, B, C, D, E, F, G, H, I or J is a 2′-O-methyl modified nucleotide.
  • x and y of a composition according to any of Formula A, B, C, D, E, F, G, H, I or J are both equal to 2.
  • all N nucleotides of the passenger strand of a composition according to any of Formula A, B, C, D, E, F, G, H, I or J are 2′-deoxy-2′-fluoro nucleotides.
  • 5, 6, 7, 8, 9, or 10 or more pyrimidine N nucleotides of the passenger strand are 2′-O-methyl nucleotides and 5, 6, 7, 8, 9, or 10 or more purine N nucleotides of the passenger strand of a composition according to any of Formula A, B, C, D, E, F, G, H, I or J are 2′-deoxy-2′-fluoro nucleotides.
  • the 5′-terminal N position of a composition according to any of Formula A, B, C, D, E, F, G, H, I or J comprises a terminal phosphate group.
  • all N nucleotides of the guide strand are 2′-deoxy-2′-fluoro nucleotides and all N nucleotides of the guide strand are 2′-O-methyl nucleotides, with no variance being permitted.
  • B1 and B2 of a composition according to any of Formula A, B, C, D, E, F, G, H, I or J each comprise an inverted abasic 2′-deoxyribose moiety and B3 is optionally absent.
  • B1 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B2 comprises an inverted abasic 2′-deoxyribose moiety
  • B3 is optionally absent.
  • B1 comprises one or more galactosamine moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B2 comprises an inverted abasic 2′-deoxyribose moiety
  • B3 is optionally absent.
  • B1 comprises one or more N-acetylgalactosamine moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B2 comprises an inverted abasic 2′-deoxyribose moiety
  • B3 is optionally absent.
  • B1 comprises one or more folate moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B2 comprises an inverted abasic 2′-deoxyribose moiety
  • B3 is optionally absent.
  • B1 comprises one or more cholesterol moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B2 comprises an inverted abasic 2′-deoxyribose moiety
  • B3 is optionally absent.
  • B1 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B2 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B3 is optionally absent.
  • B1 comprises one or more galactosamine moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B2 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B3 is optionally absent.
  • B1 comprises one or more N-acetylgalactosamine moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B2 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B3 is optionally absent.
  • B1 comprises one or more folate moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B2 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B3 is optionally absent.
  • B1 comprises one or more cholesterol moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B2 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B3 is optionally absent.
  • B1 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B2 comprises any of SEQ ID NOS: 54-528 optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B3 is optionally absent.
  • B1 comprises one or more galactosamine moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B2 comprises any of SEQ ID NOS: 54-528 optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B3 is optionally absent.
  • B1 comprises one or more N-acetylgalactosamine moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B2 comprises any of SEQ ID NOS: 54-528 optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B3 is optionally absent.
  • B1 comprises one or more folate moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B2 comprises any of SEQ ID NOS: 54-528 optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B3 is optionally absent.
  • B1 comprises one or more cholesterol moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B2 comprises any of SEQ ID NOS: 54-528 optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B3 is optionally absent.
  • B1 comprises a folate, galactosamine, or cholesterol moiety as described in U.S. Pat. No. 7,491,805.
  • S 1 and S 2 are required and S 3 is optional.
  • S 1 and S 3 are required and S 2 is optional.
  • compositions according to any of Formula A, B, C, D, E, F, G, H, I or J, S 1 , S 2 , and S 3 are required.
  • B 1 comprises:
  • each L is independently a linker and GalNAc is an N-acetyl galactosamine moiety.
  • B1 comprises:
  • B1 comprises:
  • n is an integer from 1 to 20.
  • B 1 and/or B 2 comprises:
  • X is —O—, —S—, —CH 2 — or —NH—; n is 1, 2, 3, or 4; and the bond with “ ” indicates the point of attachment optionally including one or more linkers, which may be the same or different; and further optionally including one or more targeting ligands, solubilizing agents, pharmacokinetics enhancing agents, lipids, and/or masking agents.
  • the linker is a linker shown in Table 8.
  • the invention features a composition comprising a double-stranded short interfering nucleic acid (siNA) molecule that modulates the expression and/or activity of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure represented by formula (K):
  • siNA short interfering nucleic acid
  • B 1 and B 2 are each independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker;
  • every N nucleotide is a 2′-deoxy-2′-fluoro nucleotide and every N nucleotide is a 2′-O-methyl nucleotide;
  • each S is a phosphorothioate or phospho
  • the invention features a composition comprising a double-stranded short interfering nucleic acid (siNA) molecule that modulates the expression and/or activity of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure represented by formula (L):
  • siNA short interfering nucleic acid
  • B 1 and B 2 are each independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker;
  • every N nucleotide is a 2′-deoxy-2′-fluoro nucleotide and every N nucleotide is a 2′-O-methyl nucleotide;
  • each S is a phosphorothioate or phospho
  • the invention features a composition comprising a double-stranded short interfering nucleic acid (siNA) molecule that modulates the expression and/or activity of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure represented by formula (M):
  • siNA short interfering nucleic acid
  • B 1 and B 2 are each independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker;
  • every N nucleotide is a 2′-deoxy-2′-fluoro nucleotide and every N nucleotide is a 2′-O-methyl nucleotide;
  • each S is a phosphorothioate or phospho
  • the invention features a composition comprising a double-stranded short interfering nucleic acid (siNA) molecule that modulates the expression and/or activity of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure represented by formula (N):
  • siNA short interfering nucleic acid
  • B 1 and B 2 are each independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker;
  • every N nucleotide is a 2′-deoxy-2′-fluoro nucleotide and every N nucleotide is a 2′-O-methyl nucleotide;
  • each S is a phosphorothioate or phospho
  • the invention features a composition comprising a double-stranded short interfering nucleic acid (siNA) molecule that modulates the expression and/or activity of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure represented by formula (O):
  • siNA short interfering nucleic acid
  • B 1 and B 2 are each independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker;
  • every N nucleotide is a 2′-deoxy-2′-fluoro nucleotide and every N nucleotide is a 2′-O-methyl nucleotide;
  • each S is a phosphorothioate or phospho
  • the invention features a composition comprising a double-stranded short interfering nucleic acid (siNA) molecule that modulates the expression and/or activity of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure represented by formula (P):
  • siNA short interfering nucleic acid
  • B 1 and B 2 are each independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker;
  • every N nucleotide is a 2′-deoxy-2′-fluoro nucleotide and every N nucleotide is a 2′-O-methyl nucleotide;
  • each S is a phosphorothioate or phospho
  • the 5′-terminal N position of a composition according to any of Formula K, L, M, N, O, or P comprises a terminal phosphate group.
  • B1 and B2 of a composition according to any of Formula K, L, M, N, O, or P each comprise an inverted abasic 2′-deoxyribose moiety.
  • B1 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B2 comprises an inverted abasic 2′-deoxyribose moiety.
  • B1 comprises one or more galactosamine moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B2 comprises an inverted abasic 2′-deoxyribose moiety.
  • B1 comprises one or more N-acetylgalactosamine moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B2 comprises an inverted abasic 2′-deoxyribose moiety.
  • B1 comprises one or more folate moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B2 comprises an inverted abasic 2′-deoxyribose moiety.
  • B1 comprises one or more cholesterol moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B2 comprises an inverted abasic 2′-deoxyribose moiety.
  • B1 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B2 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety.
  • B1 comprises one or more galactosamine moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B2 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety.
  • B1 comprises one or more N-acetylgalactosamine moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B2 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety.
  • B1 comprises one or more folate moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B2 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety.
  • B1 comprises one or more cholesterol moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B2 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety.
  • B1 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B2 comprises any of SEQ ID NOS: 54-528 optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety.
  • B1 comprises one or more galactosamine moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B2 comprises any of SEQ ID NOS: 54-528 optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety.
  • B1 comprises one or more N-acetylgalactosamine moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B2 comprises any of SEQ ID NOS: 54-528 optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety.
  • B1 comprises one or more folate moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B2 comprises any of SEQ ID NOS: 54-528 optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety.
  • B1 comprises one or more cholesterol moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety
  • B2 comprises any of SEQ ID NOS: 54-528 optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety.
  • B1 comprises a folate, galactosamine, or cholesterol moiety as described in U.S. Pat. No. 7,491,805.
  • B1 comprises:
  • each L is independently a linker and GalNAc is an N-acetyl galactosamine moiety.
  • B1 comprises:
  • B1 comprises:
  • n is an integer from 1 to 20.
  • B 1 and/or B 2 comprises:
  • X is —O—, —S—, —CH 2 — or —NH—; n is 1, 2, 3, or 4; and the bond with “ ” indicates the point of attachment optionally including one or more linkers, which may be the same or different; and further optionally including one or more targeting ligands, solubilizing agents, pharmacokinetics enhancing agents, lipids, and/or masking agents.
  • the linker is a linker shown in Table 8.
  • the siNA molecule is covalently attached to a polymer or ligand via a linker.
  • the siNA molecule is covalently attached to the polymer or ligand via a linker moiety at the 5′-end of the passenger (sense) strand of the siNA molecule.
  • the siNA molecule is covalently attached to the polymer or ligand via a linker moiety at the 3′-end of the passenger (sense) strand of the siNA molecule.
  • the siNA molecule is covalently attached to the polymer or ligand via a linker moiety at the 3′-end of the guide (antisense) strand of the siNA molecule.
  • the linker can be attached to the terminal 3′ and/or 5′ nucleotide position of the passenger or guide strand, or can alternately be attached to a terminal cap moiety such as an abasic moiety or other cap as described herein or otherwise known in the art. Therefore, in totality, a siNA molecule of the invention having Formula (A) can comprise a terminal cap (B) that includes a covalent attachment to a polymer or ligand via a linker molecule as described herein or as is otherwise known in the art. Non-limiting examples of such linkers are provided in the examples herein.
  • one or more terminal cap moieties of a siNA molecule of the invention can comprise a delivery modality.
  • the delivery modality can comprise a ligand or polymer that further includes one or more linker molecules.
  • linker molecules include phosphate ester based linkages, amino based linkers, disulfide based linkers, succinyl based linkers, alkyl or substituted alkyl based linkers, and/or amide based linkers as are generally known in the art.
  • the siNA molecules of the invention are phosphorylated at the 5′ end of the guide or antisense strand.
  • the phosphate group can be a phosphate, a diphosphate or a triphosphate.
  • compositions comprising the double-stranded nucleic acid molecules described herein with optionally a pharmaceutically acceptable carrier or diluent.
  • a composition of the invention further comprises a cryo-protectant.
  • the cryo-protectant is Sucrose, Trehalose, Raffinose, Stachyose, Verbascose, Mannitol, Glucose, Lactose, Maltose, Maltotriose-heptaose, Dextran, Hydroxyethyl Starch, Insulin, Sorbitol, Glycerol, Arginine, Histidine, Lysine, Proline, Dimethylsulfoxide or any combination thereof.
  • the cryo-protectant is Sucrose.
  • the cryo-protectant is Trehalose.
  • the cryo-protectant is a combination of Sucrose and Trehalose.
  • the present invention further provides a compound comprising a double-stranded short interfering nucleic acid (siNA) molecule of the invention covalently attached to a ligand.
  • ligands include steroidal compounds (e.g., cholesterol), galactosamines (e.g., N-acetylgalactosamine), vitamins (e.g., folate), proteins (e.g., monoclonal antibodies), and peptides (e.g., TAT) as are generally known in the art and further provided herein.
  • compositions of the invention can be carried out by known methods, wherein the nucleic acid is introduced into a desired target cell in vitro or in vivo.
  • Non-limiting examples of such carrier systems suitable for use in the present invention include single chemical entity conjugates, nucleic-acid-lipid particles, lipid nanoparticles (LNP), liposomes, lipoplexes, micelles, virosomes, virus like particles (VLP), nucleic acid polymers, and mixtures thereof.
  • LNP lipid nanoparticles
  • VLP virus like particles
  • compositions of the invention can be in the form of an aerosol, dispersion, solution (e.g., an injectable solution), a cream, ointment, tablet, powder, suspension or the like. These compositions may be administered in any suitable way, e.g. orally, sublingually, buccally, parenterally, nasally, or topically. In some embodiments, the compositions are aerosolized and delivered via inhalation.
  • the molecules and compositions of the present invention have utility over a broad range of therapeutic applications. Accordingly another aspect of this invention relates to the use of the compounds and compositions of the invention in treating a subject.
  • the invention thus provides a method for treating a subject, such as a human, suffering from a condition which is associated with the expression of one or more genes, wherein the method comprises administering to the subject an effective amount of a double-stranded short interfering nucleic acid (siNA) molecule of the invention.
  • siNA double-stranded short interfering nucleic acid
  • the condition is one as described herein or is otherwise generally known to one of skill in the art.
  • the present invention provides compositions and methods comprising siNAs having target specificity that may be used to treat diseases and conditions herein or otherwise known in the art that are associated with gene expression.
  • the nucleic acid molecules of the invention comprise at least a 15 nucleotide sequence of the a target sequence, and/or comprises a nucleotide sequence of at least 15 nucleotides complimentary to the target sequence).
  • the siNAs may be provided in several forms.
  • the siNA may be isolated as one or more siNA compounds, or it may be in the form of a transcriptional cassette in a DNA plasmid.
  • the siNA may also be chemically synthesized and may include modifications as described herein or as is otherwise generally known in the art.
  • the siNAs may be administered alone or co-administered with other siNA molecules or with conventional agents that treat a gene related disease or condition as described herein or otherwise known in the art.
  • the siNA molecules of the invention may be used to mediate gene silencing via interaction with RNA transcripts or alternately by interaction with particular gene sequences, wherein such interaction results in modulation of gene silencing either at the transcriptional level or post-transcriptional level such as, for example, but not limited to, RNAi or through cellular processes that modulate the chromatin structure or methylation patterns of the target and prevent transcription of the target gene, with the nucleotide sequence of the target thereby mediating silencing.
  • the target is any GenBank reference sequence as is presently known in the art.
  • the invention provides short interfering nucleic acid (siNA) molecules that may inhibit the expression of the target gene in a cell or mammal.
  • the siNA may be single-stranded or double-stranded. When double-stranded, the siNA comprising a sense and an antisense stand.
  • the antisense strand may be complementary to at least a part of an mRNA formed in the expression of the target gene.
  • the sense strand comprises a region that may be complementary to the antisense strand.
  • the double stranded RNA molecules of the invention may comprise two distinct and separate strands that can be symmetric or asymmetric and are complementary, i.e., two single-stranded RNA molecules, or may comprise one single-stranded molecule in which two complementary portions, e.g., a sense region and an antisense region, are base-paired, and are covalently linked by one or more single-stranded “hairpin” areas (i.e. loops) resulting in, for example, a single-stranded short-hairpin polynucleotide or a circular single-stranded polynucleotide.
  • two complementary portions e.g., a sense region and an antisense region
  • hairpin i.e. loops
  • the linker may be polynucleotide linker or a non-nucleotide linker. In some embodiments, the linker is a non-nucleotide linker.
  • a hairpin or circular siNA molecule of the invention contains one or more loop motifs, wherein at least one of the loop portions of the siNA molecule is biodegradable.
  • a single-stranded hairpin siNA molecule of the invention is designed such that degradation of the loop portion of the siNA molecule in vivo can generate a double-stranded siNA molecule with 3′-terminal overhangs, such as 3′-terminal nucleotide overhangs comprising 1, 2, 3 or 4 nucleotides.
  • a circular siNA molecule of the invention is designed such that degradation of the loop portions of the siNA molecule in vivo can generate a double-stranded siNA molecule with 3′-terminal overhangs, such as 3′-terminal nucleotide overhangs comprising about 2 nucleotides.
  • siNA molecules of the invention may have perfect complementarity between the sense strand or sense region and the antisense strand or antisense region of the siNA molecule.
  • the antisense strand of the siNA molecules of the invention are perfectly complementary to a corresponding target nucleic acid molecule.
  • siNA molecules of the invention have partial complementarity (i.e., less than 100% complementarity) between the sense strand or sense region and the antisense strand or antisense region of the siNA molecule or between the antisense strand or antisense region of the siNA molecule and a corresponding target nucleic acid molecule.
  • the double-stranded nucleic acid molecules of the invention have between about 15 to about 30 (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides in one strand that are complementary to the nucleotides of the other strand.
  • the molecules have between about 15 to about 30 (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides in the sense region that are complementary to the nucleotides of the antisense region. of the double-stranded nucleic acid molecule.
  • the double-stranded nucleic acid molecules of the invention have between about 15 to about 30 (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides in the antisense strand that are complementary to a nucleotide sequence of its corresponding target nucleic acid molecule.
  • the siNA molecule may contain one or more nucleotide deletions, substitutions, mismatches and/or additions; provided, however, that the siNA molecule maintains its activity, for example, to mediate RNAi.
  • the deletion, substitution, mismatch and/or addition may result in a loop or bulge, or alternately a wobble or other alternative (non Watson-Crick) base pair.
  • the double-stranded nucleic acid molecules of the invention have 1 or more (e.g., 1, 2, 3, 4, 5, or 6) nucleotides, in one strand or region that are mismatches or non-base-paired with the other strand or region.
  • the double-stranded nucleic acid molecules of the invention have 1 or more (e.g., 1, 2, 3, 4, 5, or 6) nucleotides in each strand or region that are mismatches or non-base-paired with the other strand or region.
  • the siNA of the invention contains no more than 3 mismatches. If the antisense strand of the siNA contains mismatches to a target sequence, it is preferable that the area of mismatch not be located in the center of the region of complementarity.
  • the siNA molecule may contain one or more nucleotide deletions, substitutions, mismatches and/or additions to a sequence provided herein, however, that the siNA molecule maintains its activity, for example, to mediate RNAi.
  • the deletion, substitution, mismatch and/or addition may result in a loop or bulge, or alternately a wobble or other alternative (non Watson-Crick) base pair.
  • the invention also comprises double-stranded nucleic acid (siNA) molecules as otherwise described hereinabove in which the first strand and second strand are complementary to each other and wherein at least one strand is hybridisable to the polynucleotide sequence of a target sequence under conditions of high stringency, and wherein any of the nucleotides is unmodified or chemically modified.
  • siNA double-stranded nucleic acid
  • Hybridization techniques are well known to the skilled artisan (see for instance, Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989)).
  • Preferred stringent hybridization conditions include overnight incubation at 42° C. in a solution comprising: 50% formamide, 5 ⁇ SSC (150 mM NaCl, 15 mM trisodium citrate), 50 mM sodium phosphate (pH 7.6), 5 ⁇ Denhardt's solution, 10% dextran sulfate, and 20 microgram/ml denatured, sheared salmon sperm DNA; followed by washing the filters in 0.1 ⁇ SSC at about 65° C.
  • the first strand has about 18, 19, 20, 21 or 22 nucleotides that are complementary to the nucleotides of the other strand and at least one strand is hybridisable to target sequence such as a gene in the GenBank database.
  • the first strand has about 19, 20 or 21 nucleotides that are complementary to the nucleotides of the other strand and at least one strand is hybridisable at least one strand is hybridisable to the complement of a target sequence under conditions of high stringency.
  • the siNA molecules of the invention comprise overhangs of about 1 to about 4 (e.g., about 1, 2, 3 or 4) nucleotides.
  • the nucleotides in the overhangs may be the same or different nucleotides.
  • the overhangs occur at the 3′-end at one or both strands of the double-stranded nucleic acid molecule.
  • a double-stranded nucleic acid molecule of the invention may comprise a nucleotide or non-nucleotide overhang at the 3′-end of the antisense strand/region, the 3′-end of the sense strand/region, or both the antisense strand/region and the sense strand/region of the double-stranded nucleic acid molecule.
  • the nucleotides comprising the overhang portion of an siNA molecule of the invention comprise sequences based on the target polynucleotide sequence in which nucleotides comprising the overhang portion of the antisense strand/region of an siNA molecule of the invention can be complementary to nucleotides in the target polynucleotide sequence and/or nucleotides comprising the overhang portion of the sense strand/region of an siNA molecule of the invention can comprise the nucleotides in the target polynucleotide sequence.
  • the overhang comprises a two nucleotide overhang that is complementary to a portion of the target polynucleotide sequence.
  • the overhang comprises a two nucleotide overhang that is not complementary to a portion of the target polynucleotide sequence.
  • the overhang comprises a 3′-UU overhang that is not complementary to a portion of the target polynucleotide sequence.
  • the overhang comprises a UU overhang at the 3′ end of the antisense strand and a TT overhang at the 3′ end of the sense strand.
  • the overhang comprises nucleotides as described in the examples, Tables, and Figures herein.
  • the overhangs are optionally chemically modified at one or more nucleic acid sugar, base, or backbone positions.
  • modified nucleotides in the overhang portion of a double-stranded nucleic acid (siNA) molecule of the invention include: 2′-O-alkyl (e.g., 2′-O-methyl), 2′-deoxy, 2′-deoxy-2′-fluoro, 2′-deoxy-2′-fluoroarabino (FANA), 4′-thio, 2′-O-trifluoromethyl, 2′-O-ethyl-trifluoromethoxy, 2′-O-difluoromethoxy-ethoxy, universal base, acyclic, or 5-C-methyl nucleotides.
  • the overhang nucleotides are each independently, a 2′-O-alkyl nucleotide, a 2′-O-methyl nucleotide, a 2′-dexoy-2-fluoro nucleotide, or a 2′-deoxy ribonucleotide. In some instances the overhang nucleotides are linked by a one or more phosphorothioate linkages.
  • siNA molecules of the invention comprise duplex nucleic acid molecules with blunt ends (i.e., without nucleotide overhangs), where both ends are blunt, or alternatively, where one of the ends is blunt.
  • the siNA molecules of the invention can comprises one blunt end, for example wherein the 5′-end of the antisense strand and the 3′-end of the sense strand do not have any overhanging nucleotides.
  • the siNA molecule comprises one blunt end, for example wherein the 3′-end of the antisense strand and the 5′-end of the sense strand do not have any overhanging nucleotides.
  • siNA molecules of the invention comprise two blunt ends, for example wherein the 3′-end of the antisense strand and the 5′-end of the sense strand as well as the 5′-end of the antisense strand and 3′-end of the sense strand do not have any overhanging nucleotides.
  • the sense strand and/or the antisense strand may further have a cap, such as described herein or as known in the art, at the 3′-end, the 5′-end, or both of the 3′ and 5′-ends of the sense strand and/or antisense strand.
  • the cap may be at either one or both of the terminal nucleotides of the polynucleotide.
  • the cap is at one of both of the ends of the sense strand of a double-stranded siNA molecule.
  • the cap is at the 3′-end of antisense (guide) strand.
  • the caps are at the 3′-end of the sense strand and the 5′-end of the sense strand.
  • terminal caps include an inverted abasic nucleotide, an inverted deoxy abasic nucleotide, an inverted nucleotide moiety, a group shown in the examples and Figures herein, a glyceryl modification, an alkyl or cycloalkyl group, a heterocycle, or any other cap as is generally known in the art.
  • any of the embodiments of the siNA molecules of the invention may have a 5′ phosphate termini.
  • the siNA molecules lack terminal phosphates.
  • RNAi activity and/or stability may be improved compared to corresponding unmodified siNA molecules.
  • Various chemically modified siNA motifs disclosed herein may provide the capacity to maintain RNAi activity that is substantially similar to unmodified or minimally modified active siRNA (see for example Elbashir et al., 2001, EMBO J., 20:6877-6888) while at the same time providing nuclease resistance and pharmacokinetic properties suitable for use in therapeutic applications.
  • siNA molecules having Formula A, B, C, D, E, F, G, H, I, J, K, L, M, N, O, or P have at position 14 from the 5′-end of the antisense strand a ribonucleotide when the nucleotide at that position 14 is a purine.
  • siNA molecules having Formula A, B, C, D, E, F, G, H, I, J, K, L, M, N, O, or P have at position 14 from the 5′-end of the antisense strand a ribonucleotide or a 2′-deoxy-2′-fluoro nucleotide when the nucleotide at that position 14 is a purine.
  • siNA molecules having Formula A, B, C, D, E, F, G, H, I, J, K, L, M, N, O, or P have at position 14 from the 5′-end of the antisense strand is a 2′-deoxy-2′-fluoro nucleotide when the nucleotide at that position 14 is a pyrimidine nucleotide.
  • position 14 from the 5′-end of the antisense strand is a 2′-deoxy-2′-fluoro nucleotide regardless of whether it is a purine or a pyrimidine.
  • siNA molecules having Formula A, B, C, D, E, F, G, H, I, J, K, L, M, N, O, or P comprises (N) nucleotides in the antisense strand (lower strand) that are complementary to nucleotides in a target polynucleotide sequence, which also has complementarity to the N and [N] nucleotides of the antisense (lower) strand.
  • siNA molecules having Formula A, B, C, D, E, F, G, H, I, J, K, L, M, N, O, or P may be applied to any of the embodiments to siNA molecules having Formula A, B, C, D, E, F, G, H, I, J, K, L, M, N, O, or P.
  • the compounds disclosed herein include a transduction moiety.
  • Transduction moieties can include, but are not limited to, cell-penetrating peptide (CPP), peptide transduction domain (PTD), nucleic acid binding proteins, such as RNA binding proteins, or any combination thereof.
  • CPP cell-penetrating peptide
  • PTD peptide transduction domain
  • nucleic acid binding proteins such as RNA binding proteins, or any combination thereof.
  • any B1, B2, and/or B3 of a compound having any of Formula A, B, C, D, E, F, G, H, I, J, K, L, M, N, O, or P comprises a transduction moiety.
  • Nucleic acid binding proteins including double-stranded RNA binding domains, can been used to enhance delivery of oligonucleotides into cells. (See, e.g., Eguchi et al. Nat. Biotech. 27:567-571 (2009)).
  • Exemplary nucleic acid binding domains useful in the embodiments disclosed herein include, but are not limited to, those listed in U.S. Patent Application Publication No. US 2009/0093026.
  • CPPs Cell-penetrating peptides
  • PTDs peptide transfer domains
  • the peptides may be conjugated at either end or both ends by addition of a cysteine or other thiol containing moiety to the C- or N-terminus.
  • peptides When not functionalized on the N-terminus, peptides may be capped by an acetyl group, or may be capped with a lipid, a PEG, or a targeting moiety.
  • the C-terminus of the peptides When the C-terminus of the peptides is unconjugated or unfunctionalized, it may be capped as an amide, or may be capped with a lipid, a PEG, or a targeting moiety.
  • Suitable peptides that can be used in the conjugates discloased herein are listed in Table 2.
  • a PTD of the invention can be defined as one or more cationic peptides that are able to interact with the cell membrane in a manner that enhances macromolecular uptake.
  • these peptides can be configured in a linear sequence or attached via a branched linker as is generally known in the art.
  • the branching mechanism can be built into the peptide by having multiple cysteines or lysines that are specifically used to form linkages. The methods for synthesizing branched amino acid constructs are well established in the art.
  • a transduction moiety can be a cell penetrating peptide (CPP), a cationic polymer, an antibody, a cholesterol or cholesterol derivative, a vitamin compound (e.g., vitamin E, B12, B6, etc.), a tocol, a tocotrienol, a tocopherol, glucose, receptor ligand, antibody or the like capable of cell type specific targeting, to further facilitate the uptake of the anionic biomolecule, such as oligonucleotides and polynucleotides.
  • CPP cell penetrating peptide
  • a cationic polymer an antibody
  • an antibody e.g., a cholesterol or cholesterol derivative
  • a vitamin compound e.g., vitamin E, B12, B6, etc.
  • a tocol e.g., vitamin E, B12, B6, etc.
  • a tocol e.g., a tocotrienol
  • a tocopherol e.g., glucose, receptor ligand,
  • the PTD domain comprises a peptide represented by the following general formula: B1-X1-X2-X3-B2-X4-X5-B3, wherein B1, B2, and B3 are each independently a basic amino acid, the same or different; and X1, X2, X3, X4 and X5 are each independently an alpha-helix enhancing amino acid, the same or different.
  • the PTD domain comprises a polypeptide represented by the following general formula: X-X-R-X-(P/X)-(B/X)-B-(P/X)-X-B-(B/X), wherein X is any alpha helical promoting residue such as alanine; P/X is either proline or X as previously defined; B is a basic amino acid residue, e.g., Arginine (Arg) or lysine (Lys); R is Arginine (Arg) and B/X is either B or X as defined herein.
  • the PTD can be cationic.
  • the PTD can include between 7 and 10 amino acids and have the general formula K-X1-R-X2-X1 wherein X1 is R or K and X2 is any amino acid.
  • An example of such a cationic polypeptide can include the sequence RKKRRQRRR (SEQ ID NO: 528), or functional fragments and variants thereof.
  • a number of protein transduction domains/peptides are known in the art and may facilitate uptake of heterologous molecules linked to the transduction domains (e.g., cargo molecules). Such peptide transduction domains (PTD's) may facilitate uptake through a process referred to as macropinocytosis. Macropinocytosis is a nonselective form of endocytosis.
  • PTDs and CPPs useful in the embodiments disclosed herein include the PTDs and CPPs described in, for example, Langel, Ulo, “Cell Penetrating Peptides, Processes and Applications,” In Langel, Ulo; (Ed.); Handbook of Cell-Penetrating Peptides, 2.sup.nd Ed (2007); Langel, Ulo, (Ed.). “Cell-Penetrating Peptides, Mechanisms and Applications;” In Curr. Pharm. Des.; 2005, 11(28)(2005); Langel, Ulo, “Cell-Penetrating Peptides: Processes and Applications” (2002); Wadia, Jehangir S.; Becker-Hapak, Michelle; Dowdy, Steven F. Protein transport. Cell-Penetrating Peptides (2002), pp. 365-375.
  • Exemplary peptide transduction domains may be derived from the Drosophila homeoprotein Antennapedia transcription protein (AntHD) (Joliot et al., New Biol. 3:1121-34, 1991; Joliot et al., Proc. Natl. Acad. Sci. USA, 88:1864-8, 1991; Le Roux et al., Proc. Natl. Acad. Sci.
  • AntHD Drosophila homeoprotein Antennapedia transcription protein
  • the peptide transduction domain may increase uptake of the biomolecule to which it is fused in a receptor independent fashion, may be capable of transducing a wide range of cell types, and may exhibit minimal or no toxicity (Nagahara et al., Nat. Med. 4:1449-52, 1998).
  • the siNA molecules disclosed herein include PTDs that are cationic in nature. Cationic protein transduction domains track into lipid raft endosomes carrying with them their linked cargo and release their cargo into the cytoplasm by disruption of the endosomal vesicle.
  • the invention provides, in one aspect, methods and compositions that combine the use of PTDs such as TAT and poly-Arg, with a siNA molecule of the invention.
  • protein transduction domains of the embodiments disclosed herein may be any synthetic or naturally-occurring amino acid sequence that can transduce or assist in the transduction of the fusion molecule.
  • transduction may be achieved in accord with the disclosure by use of a nucleic acid construct including phosphodiester and/or phosphorothioate protecting groups and a protein sequence such as an HIV TAT protein or fragment thereof that is linked at the N-terminal or C-terminal end to an oligonucleotide or polynucleotide comprising a phosphodiester and/or phosphorothioate protecting group.
  • the nucleic acid may comprise a phosphodiester and/or phosphorothioate protecting group and may also comprise a nucleic acid binding domain (e.g., a DRBD).
  • the transducing protein domain may be the Antennapedia homeodomain or the HSV VP22 sequence, the N-terminal fragment of a prion protein or suitable transducing fragments thereof such as those generally known in the art.
  • the siNA compositions disclosed herein may include a PTD that has substantial alpha-helicity, for example, to optimize transduction of the biomolecule.
  • the PTD comprises a sequence containing basic amino acid residues that are substantially aligned along at least one face of the peptide.
  • Alpha-helicity can be estimated as is generally known in the art.
  • the PTD is as described in PCT Pub. Nos. WO 08/008,476 and WO 07/095,152, the PTD disclosure of which is hereby expressly incorporated by this reference.
  • Additional transducing domains useful in the embodiments disclosed herein include but are not limited to a TAT fragment that comprises at least amino acids 49 to 56 of TAT up to about the full-length TAT sequence as described in PCT Pub. No. WO 08/008,476.
  • a TAT fragment may include one or more amino acid changes sufficient to increase the alpha-helicity of the fragment.
  • amino acid changes are introduced in the PTDs that add a recognized alpha-helix enhancing amino acid.
  • amino acids are introduced in the PTD's that remove one or more amino acids from the TAT fragment that impede alpha helix formation or stability.
  • the PTD may be a TAT fragment that includes at least one amino acid substitution with an alpha-helix enhancing amino acid.
  • Additional transduction proteins (PTDs) useful in the embodiments disclosed herein include a TAT fragment in which the TAT 49-56 sequence has been modified so that at least two basic amino acids in the sequence are substantially aligned along at least one face of the TAT fragment.
  • Exemplary TAT fragments useful as PTDs in the embodiments disclosed herein may include at least one specified amino acid substitution in at least amino acids 49-56 of TAT which substitution aligns the basic amino acid residues of the 49-56 sequence along at least one face of the segment.
  • the PTD used in the embodiments disclosed herein may be a naturally occurring PTD, such as include the homeodomain of the Drosophila melanogaster protein Antennapedia (Lindsay (2002) Curr. Op. Pharmacol. 2:587-94; Derossi et al. (1994) J. Biol. Chem. 269:10444-50), HSV-1 VP22 (Bennett et al. (2002) Nat. Biotechnol. 20:20), and Buforin II (Park et al. (2000) Proc. Natl. Acad. Sci. U.S.A. 97:8245-50), or the like, or fragments thereof.
  • a naturally occurring PTD such as include the homeodomain of the Drosophila melanogaster protein Antennapedia (Lindsay (2002) Curr. Op. Pharmacol. 2:587-94; Derossi et al. (1994) J. Biol. Chem. 269:10444-50), HSV-1 VP22 (
  • the PTD used in the embodiments disclosed herein may be a recombinant or synthetic PTD designed to mimic and/or enhance the translocating properties of known PTDs, based on consideration of parameters such as electrostatic and hydrophobic properties or secondary structure (Wender et al. (2000) Proc. Natl. Acad. Sci. U.S.A. 97:13003-8; Futaki et al. (2001) J. Biol. Chem. 276:5836-40).
  • An exemplary artificial PTD is transportan (Pooga et al. (1998) FASEB J. 12:67-77; Soomets et al. (2000) Biochim. Biophys. Acta 1467:165-76).
  • Synthetic PTDs such as polylysine, polyarginine, and polyhistidine (which can be positively charged based on the pH of the formulation) e.g., polyarginine (6-15 amino acids) may be useful in the embodiments disclosed herein.
  • PTDs useful in the embodiments disclosed herein include, but are not limited to those provided in Table 2 herein.
  • PTDs useful in the embodiments described herein include PTDs derived from protamine (AAA39985), penetratin (1OMQ_A), TAT (NP — 057853), pVEC, Cationic prion protein domains, P101 (ACT78456), MATa2 (Q6B184), HIV-1 rev (CAA41586), Polyomavirus Vpl (AAP14004), NF-kappaB (NP — 003989), M9 (BAA76626), Vpr (BAH97661), FP_NLS (MPG), Sp-NPS (ACU27162), SN50, Importins and Karyopherins, e.g., Karyopherin alpha (NP — 002255), and Karyopherin beta (NP — 002256), and the like.
  • PTDs useful in the embodiments disclosed herein include those found in International Patent Application Publication No's: WO 09/041,902, WO 05/084158; WO 00/062067, WO 00/034308, and WO 99/55899, each of which is herein incorporated by reference.
  • the transduction moiety may be a chimeric PTD domain comprising sequences derived from at least two different transducing proteins.
  • chimeric transducing proteins useful in the embodiments disclosed herein include a chimera between two different TAT fragments, e.g., one from HIV-1 and the other from HIV-2 or one from a prion protein and one from HIV.
  • TAT fragments e.g., one from HIV-1 and the other from HIV-2 or one from a prion protein and one from HIV.
  • the transduction moiety may be a nucleic acid binding polypeptide, such as an RNA binding protein, or the like optionally linked to a PTD selected from the examples listed above.
  • RNA binding proteins e.g., DRBD
  • histone e.g., histone
  • RDE-4 protein e.g., protamine
  • Exemplary dsRNA binding proteins include but are not limited to: PKR (AAA36409, AAA61926, Q03963), TRBP (P97473, AAA36765), PACT (AAC25672, AAA49947, NP609646), Staufen (AAD17531, AAF98119, AAD17529, P25159), NFAR1 (AF167569), NFAR2 (AF167570, AAF31446, AAC71052, AAA19960, AAA19961, AAG22859), SPNR (AAK20832, AAF59924, A57284), RHA (CAA71668, AAC05725, AAF57297), NREBP (AAK07692, AAF23120, AAF54409, T33856), kanadaptin (AAK29177, AAB88191, AAF55582, NP499172, NP198700, BAB19354), HYL1 (NP563850), hypon
  • CPP and PTD domains described herein include modified peptides such as glycoproteins, the L-optical isomer or the D-optical isomer of amino acids or a combination of both, as well as retro-inverso polypeptides.
  • retro-inverso refers a peptide that comprises an amino-carboxy inversion as well as enantiomeric change in one or more amino acids (i.e., levorotatory (L) to dextrorotary (D)).
  • the CPP and PTD domains described herein encompass D-amino acid modified polypeptides, amino-carboxy inversions of the amino acid sequence, amino-carboxy inversions containing one or more D-amino acids, naturally occurring proteins, recombinantly or synthetically synthesized peptides, non-inverted sequence containing one or more D-amino acids, peptidomimetics, Beta-amino acid analogs, gamma amino acid analogs, and the like.
  • CPP or PTD peptides disclosed herein may also encompass peptide fragments as are generally known in the art.
  • anionic biomolecules disclosed herein may be operably linked to an additional transduction moiety.
  • the transduction moiety can be a synthetic or non-synthetic, linear or branched peptide transduction domain (PTD) as described herein or as is otherwise known in the art.
  • PTD can be a cationic peptide optionally connected via a branching linker installed during automated nucleotide synthesis.
  • the branching linker can be trebler, symmetrical or combinations thereof.
  • transduction moieties disclosed herein can be linked or fused with another transduction moiety as described herein or as is otherwise known in the art, a linker, such as a peptide linker or a nucleotide linker, or can be directly linked to an siNA molecule disclosed herein.
  • a linker such as a peptide linker or a nucleotide linker
  • Non-limiting examples of linkers useful in the embodiments disclosed herein include, but are not limited to GG (SEQ ID NO: 529), GGGGS (SEQ ID NO: 530), GGGGSN (SEQ ID NO: 531), GKSSGSGSESKS (SEQ ID NO: 532), GSTSGSGKSSEGKG (SEQ ID NO: 533), GSTSGKSSEGSGSTKG (SEQ ID NO: 534), GSTSGSGKPGSGEGSTKG (SEQ ID NO: 535), or EGKSSGSGSESKEF (SEQ ID NO: 536)
  • Linking moieties are described, for example, in Huston et al., Proc. Nat'l Acad. Sci.
  • compositions disclosed herein comprise targeting moieties and the like.
  • two or more transduction moieties such as PTDs (e.g., 1-5, 2-4, typically 3) may be linked in series or separated by one or more other domains (e.g., a nucleic acid domain or peptide linkers).
  • Transduction moieties, siNA molecules of the invention, and peptide linkers may be organized in nearly any fashion provided that the construct has the function for which it was intended.
  • Each of several domains e.g., transduction moieties and siNA molecules
  • the domains may be presented in any order.
  • the fusion polypeptides may include tags, e.g., to facilitate identification and/or purification of the fusion polypeptide, such as a 6xHIS tag, a maltose binding protein domain, a GST tag, or the like.
  • tags e.g., to facilitate identification and/or purification of the fusion polypeptide, such as a 6xHIS tag, a maltose binding protein domain, a GST tag, or the like.
  • the siNA compositions described herein include a peptide linker.
  • a peptide linker comprises up to about 20 or 30 amino acids, commonly up to about 10 or 15 amino acids, and still more often from about 1 to 5 amino acids, e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, or more amino acids.
  • the amino acid sequence of the linker is engineered to be flexible so as not to hold the fusion molecule in a single rigid conformation.
  • Peptide linker sequences can be used, e.g., to space the transduction moieties from the siNA molecules of the invention.
  • the peptide linker sequence can be positioned between a transduction moiety, and one or more strands of a siNA molecule, e.g., to provide molecular flexibility.
  • the length of the linker moiety is chosen to optimize the biological activity of the polypeptide comprising a PTD domain fusion construct and can be determined empirically without undue experimentation.
  • the linker moiety should be long enough and flexible enough to allow a nucleic acid binding domain to freely interact with a nucleic acid or vice versa.
  • Exemplary peptide linkers and linker moieties are described in Int. Pub. No. WO/2008/008476, in Huston et al., Proc. Natl. Acad. Sci.
  • siNAs of the invention may be obtained using a number of techniques known to those of skill in the art.
  • the siNA may be chemically synthesized or may be encoded by plasmid (e.g., transcribed as sequences that automatically fold into duplexes with hairpin loops.).
  • siNA may also be generated by cleavage of longer dsRNA (e.g., dsRNA greater than about 25 nucleotides in length) by the E. coli RNase II or Dicer. These enzymes process the dsRNA into biologically active siNA (see, e.g., Yang et al., PNAS USA 99:9942-9947 (2002); Calegari et al.
  • siNA of the invention are chemically synthesized.
  • Oligonucleotides e.g., certain modified oligonucleotides or portions of oligonucleotides lacking ribonucleotides
  • Oligonucleotides are synthesized using protocols known in the art, for example as described in Caruthers et al., 1992 , Methods in Enzymology 211, 3-19, Thompson et al., International PCT Publication No. WO 99/54459, Wincott et al., 1995 , Nucleic Acids Res. 23, 2677-2684, Wincott et al., 1997, Methods Mol.
  • oligonucleotides makes use of common nucleic acid protecting and coupling groups, such as dimethoxytrityl at the 5′-end, and phosphoramidites at the 3′-end.
  • siNA molecules without modifications may be synthesized using procedures as described in Usman et al., 1987 , J. Am. Chem. Soc., 109, 7845; Scaringe et al., 1990 , Nucleic Acids Res., 18, 5433. These syntheses makes use of common nucleic acid protecting and coupling groups, such as dimethoxytrityl at the 5′-end, and phosphoramidites at the 3′-end that can be used for certain siNA molecules of the invention.
  • the siNA molecules of the invention may be synthesized, deprotected, and analyzed according to methods described in U.S. Pat. Nos. 6,995,259, 6,686,463, 6,673,918, 6,649,751, 6,989,442, and U.S. patent application Ser. No. 10/190,359.
  • siNA molecules of the present invention may be synthesized separately and joined together post-synthetically, for example, by ligation (Moore et al., 1992, Science 256, 9923; Draper et al., International PCT Publication No. WO 93/23569; Shabarova et al., 1991 , Nucleic Acids Research 19, 4247; Bellon et al., 1997, Nucleosides & Nucleotides, 16, 951; Bellon et al., 1997, Bioconjugate Chem. 8, 204), or by hybridization following synthesis and/or deprotection.
  • siNA molecules of the invention may also be synthesized using the teachings of Scaringe et al., U.S. Pat. Nos. 5,889,136; 6,008,400; and 6,111,086.
  • the siNA molecules of the invention may be added directly or may be conjugated with a delivery vehicle, or otherwise delivered to target cells or tissues.
  • the invention features conjugates and/or complexes of siNA molecules of the invention.
  • conjugates and/or complexes include ligand based and polymer based delivery modalities that may be used to facilitate delivery of siNA molecules into a biological system, such as a cell.
  • the conjugates and complexes provided by the instant invention may impart therapeutic activity by transferring therapeutic compounds across cellular membranes, altering the pharmacokinetics, and/or modulating the localization of nucleic acid molecules of the invention.
  • Non-limiting, examples of such conjugates are described in U.S. Publication Nos.
  • polyethylene glycol may be covalently attached to siNA compounds of the present invention.
  • the attached PEG may be any molecular weight, preferably from about 100 to about 50,000 daltons (Da).
  • the present invention also provides nucleic acids in kit form.
  • the kit may comprise a container.
  • the kit typically contains a nucleic acid of the invention with instructions for its administration.
  • the nucleic acids may have a targeting moiety or delivery agent attached. Methods of attaching targeting moieties (e.g. antibodies, proteins) or delivery agents (conjugates) are known to those of skill in the art.
  • the kit contains more than one siNA molecule of the invention.
  • the kits may comprise an siNA molecule of the invention with a pharmaceutically acceptable carrier or diluent.
  • the kits may further comprise excipients.
  • nucleic acid molecules and pharmaceutical compositions of the invention may be used to treat diseases, conditions, or phenotypes related to gene expression.
  • diseases, conditions, and phenotypes are described herein and are otherwise known in the art
  • Particular conditions and disease states that may be associated with gene expression modulation include, but are not limited to cancer, proliferative, inflammatory, autoimmune, neurologic, ocular, respiratory, metabolic, dermatological, auditory, liver, kidney, infectious etc. diseases, conditions, or disorders as described herein or otherwise known in the art, and any other diseases, conditions or disorders that may be related to or may respond to the levels of a target (e.g., target DNA, RNA, protein or polynucleotide) in a cell or tissue, alone or in combination with other therapies.
  • a target e.g., target DNA, RNA, protein or polynucleotide
  • Proliferative diseases may include any disease or condition characterized by unregulated cell growth or replication as is known in the art; including leukemias, for example, acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), acute lymphocytic leukemia (ALL), and chronic lymphocytic leukemia, AIDS related cancers such as Kaposi's sarcoma; breast cancers; bone cancers such as Osteosarcoma, Chondrosarcomas, Ewing's sarcoma, Fibrosarcomas, Giant cell tumors, Adamantinomas, and Chordomas; Brain cancers such as Meningiomas, Glioblastomas, Lower-Grade Astrocytomas, Oligodendrocytomas, Pituitary Tumors, Schwannomas, and Metastatic brain cancers; cancers of the head and neck including various lymphomas such as mantle cell lymphoma, non-Hodgkins lymphoma,
  • Inflammatory diseases may include any disease or condition characterized by an inflammatory or allergic process as is known in the art, such as inflammation, acute inflammation, chronic inflammation, respiratory disease, atherosclerosis, psoriasis, dermatitis, restenosis, asthma, allergic rhinitis, atopic dermatitis, septic shock, rheumatoid arthritis, inflammatory bowl disease, inflammatory pelvic disease, pain, ocular inflammatory disease, celiac disease, Leigh Syndrome, Glycerol Kinase Deficiency, Familial eosinophilia (FE), autosomal recessive spastic ataxia, laryngeal inflammatory disease; Tuberculosis, Chronic cholecystitis, Bronchiectasis, Silicosis and other pneumoconioses, and any other inflammatory disease, condition, trait, genotype or phenotype that can respond to the modulation of disease related gene expression in a cell or tissue, alone or in combination
  • Autoimmune diseases may include any disease or condition characterized by autoimmunity as is known in the art, such as multiple sclerosis, diabetes mellitus, lupus, celiac disease, Crohn's disease, ulcerative colitis, Guillain-Barre syndrome, scleroderms, Goodpasture's syndrome, Wegener's granulomatosis, autoimmune epilepsy, Rasmussen's encephalitis, Primary biliary sclerosis, Sclerosing cholangitis, Autoimmune hepatitis, Addison's disease, Hashimoto's thyroiditis, Fibromyalgia, Menier's syndrome; transplantation rejection (e.g., prevention of allograft rejection) pernicious anemia, rheumatoid arthritis, systemic lupus erythematosus, dermatomyositis, Sjogren's syndrome, lupus erythematosus, multiple sclerosis, mya
  • Infectious diseases may include any disease or condition associated with an infectious agent, such as a virus, bacteria, fungus, prion, or parasite.
  • infectious agents such as a virus, bacteria, fungus, prion, or parasite.
  • viral genes that can be targeted using siNA molecules of the invention include Hepatitis C Virus (HCV, for example GenBank Accession Nos: D11168, D50483.1, L38318 and S82227), Hepatitis B Virus (HBV, for example GenBank Accession No. AF100308.1), Human Immunodeficiency Virus type 1 (HIV-1, for example GenBank Accession No. U51188), Human Immunodeficiency Virus type 2 (HIV-2, for example GenBank Accession No.
  • X60667 West Nile Virus (WNV for example GenBank accession No. NC — 001563), cytomegalovirus (CMV for example GenBank Accession No. NC — 001347), respiratory syncytial virus (RSV for example GenBank Accession No. NC — 001781), influenza virus (for example GenBank Accession No. AF037412, rhinovirus (for example, GenBank accession numbers: D00239, X02316, X01087, L24917, M16248, K02121, X01087), papillomavirus (for example GenBank Accession No. NC — 001353), Herpes Simplex Virus (HSV for example GenBank Accession No.
  • siNA molecules designed against conserved regions of various viral genomes will enable efficient inhibition of viral replication in diverse patient populations and may ensure the effectiveness of the siNA molecules against viral quasi species which evolve due to mutations in the non-conserved regions of the viral genome.
  • Non-limiting examples of bacterial infections include Actinomycosis, Anthrax, Aspergillosis, Bacteremia, Bacterial Infections and Mycoses, Bartonella Infections, Botulism, Brucellosis, Burkholderia Infections, Campylobacter Infections, Candidiasis, Cat-Scratch Disease, Chlamydia Infections, Cholera, Clostridium Infections, Coccidioidomycosis, Cross Infection, Cryptococcosis, Dermatomycoses, Dermatomycoses, Diphtheria, Ehrlichiosis, Escherichia coli Infections, Fasciitis, Necrotizing, Fusobacterium Infections, Gas Gangrene, Gram-Negative Bacterial Infections, Gram-Positive Bacterial Infections, Histoplasmosis, Impetigo, Klebsiella Infections, Legionellosis, Lepros
  • Non-limiting examples of fungal infections include Aspergillosis, Blastomycosis, Coccidioidomycosis, Cryptococcosis, Fungal Infections of Fingernails and Toenails, Fungal Sinusitis, Histoplasmosis, Histoplasmosis, Mucormycosis, Nail Fungal Infection, Paracoccidioidomycosis, Sporotrichosis, Valley Fever (Coccidioidomycosis), and Mold Allergy.
  • Neurologic diseases may include any disease or condition affecting the central or peripheral nervous system, including ADHD, AIDS—Neurological Complications, Absence of the Septum Pellucidum, Acquired Epileptiform Aphasia, Acute Disseminated Encephalomyelitis, Adrenoleukodystrophy, Agenesis of the Corpus Callosum, Agnosia, Aicardi Syndrome, Alexander Disease, Alpers' Disease, Alternating Hemiplegia, Alzheimer's Disease, Amyotrophic Lateral Sclerosis, Anencephaly, Aneurysm, Angelman Syndrome, Angiomatosis, Anoxia, Aphasia, Apraxia, Arachnoid Cysts, Arachnoiditis, Arnold-Chiari Malformation, Arteriovenous Malformation, Aspartame, Asperger Syndrome, Ataxia Telangiectasia, Ataxia, Attention Deficit-Hyperactivity Disorder, Autism, Autonomic Dysfunction, Back Pain, Barth Syndrome, Batten Disease, Behcet
  • Respiratory diseases may include any disease or condition affecting the respiratory tract, such as asthma, chronic obstructive pulmonary disease or “COPD”, allergic rhinitis, sinusitis, pulmonary vasoconstriction, inflammation, allergies, impeded respiration, respiratory distress syndrome, cystic fibrosis, pulmonary hypertension, pulmonary vasoconstriction, emphysema, and any other respiratory disease, condition, trait, genotype or phenotype that can respond to the modulation of disease related gene expression in a cell or tissue, alone or in combination with other therapies.
  • COPD chronic obstructive pulmonary disease
  • asthma chronic obstructive pulmonary disease or “COPD”
  • allergic rhinitis sinusitis
  • pulmonary vasoconstriction inflammation
  • allergies impeded respiration
  • respiratory distress syndrome CAD
  • cystic fibrosis pulmonary hypertension
  • pulmonary vasoconstriction emphysema
  • emphysema emphys
  • Ocular diseases may include any disease or condition affecting eye and related structures as is known in the art, such as Cystoid Macular Edema, Asteroid Hyalosis, Pathological Myopia and Posterior Staphyloma, Toxocariasis (Ocular Larva Migrans), Retinal Vein Occlusion, Posterior Vitreous Detachment, Tractional Retinal Tears, Epiretinal Membrane, Diabetic Retinopathy, Lattice Degeneration, Retinal Vein Occlusion, Retinal Artery Occlusion, Macular Degeneration (e.g., age related macular degeneration such as wet AMD or dry AMD), Toxoplasmosis, Choroidal Melanoma, Acquired Retinoschisis, Hollenhorst Plaque, Idiopathic Central Serous Chorioretinopathy, Macular Hole, Presumed Ocular Histoplasmosis Syndrome, Retinal Macroaneursym, Ret
  • Dermatologic diseases may include any disease or condition affecting the skin, dermis, or any substructure therein such as hair, follicle, etc.
  • Dermatological diseases, disorders, conditions, and traits can include psoriasis, ectopic dermatitis, skin cancers such as melanoma and basal cell carcinoma, hair loss, hair removal, alterations in pigmentation, and any other disease, condition, or trait associated with the skin, dermis, or structures therein.
  • Auditory diseases may include any disease or condition affecting the auditory system, including the ear, such as the inner ear, middle ear, outer ear, auditory nerve, and any substructures therein. Auditory diseases, disorders, conditions, and traits can include hearing loss, deafness, tinnitus, Meniere's Disease, vertigo, balance and motion disorders, and any other disease, condition, or trait associated with the ear, or structures therein.
  • Metabolic diseases may include any disease or condition affecting metabolic pathways as in known in the art. Metabolic disease can result in an abnormal metabolic process, either congenital due to inherited enzyme abnormality (inborn errors of metabolism) or acquired due to disease of an endocrine organ or failure of a metabolically important organ such as the liver.
  • metabolic disease includes hyperlipidemia, hypercholesterolemia, cardiovascular disease, atherosclerosis, hypertension, diabetes (e.g., type I and/or type II diabetes), insulin resistance, and/or obesity.
  • Cardiovascular diseases may include any disease or condition affecting the heart and vasculature, including but not limited to, coronary heart disease (CHD), cerebrovascular disease (CVD), aortic stenosis, peripheral vascular disease, atherosclerosis, arteriosclerosis, myocardial infarction (heart attack), cerebrovascular diseases (stroke), transient ischemic attacks (TIA), angina (stable and unstable), atrial fibrillation, arrhythmia, vavular disease, congestive heart failure, hypercholoesterolemia, type I hyperlipoproteinemia, type II hyperlipoproteinemia, type III hyperlipoproteinemia, type IV hyperlipoproteinemia, type V hyperlipoproteinemia, secondary hypertrigliceridemia, and familial lecithin cholesterol acyltransferase deficiency.
  • CHD coronary heart disease
  • CVD cerebrovascular disease
  • aortic stenosis CAD
  • peripheral vascular disease atherosclerosis
  • arteriosclerosis myocardial infarction
  • the siNA molecules of the invention may silence the expression of target genes and thus amenable to the treatment of various diseases and conditions herein or otherwise known in the art. Treatment of a disease may be evaluated by directly measuring the progress of the disease in a subject. It may also be inferred through observing a change or reversal in a condition associated with the disease. Additionally, the siNA molecules of the invention may be used as a prophylaxis. Thus, the use of the nucleic acid molecules and pharmaceutical compositions of the invention may be used to ameliorate, treat, prevent, and/or cure these diseases and others associated with gene expression and/or activity.
  • compositions thereof, and methods of the present disclosure may include those suffering from one or more disease or condition mediated, at least in part, by an aberrant expression level of the target gene or sequence, those at risk of developing a disease caused by or associated with the aberrant levels of a target gene/sequence, or those which may be amenable to treatment by replenishing or increasing the level of RNAi mediated by the corresponding siNA molecule, including a hyperproliferative (e.g., cancer), angiogenic, metabolic, or inflammatory (e.g., arthritis) disease or disorder or condition.
  • a hyperproliferative e.g., cancer
  • angiogenic, metabolic angiogenic
  • inflammatory e.g., arthritis
  • compositions and methods disclosed herein may be useful in the treatment of a wide variety of target viruses, including retrovirus, such as human immunodeficiency virus (HIV), Hepatitis C Virus, Hepatitis B Virus, Coronavirus, as well as respiratory viruses, including human Respiratory Syncytial Virus, human Metapneumovirus, human Parainfluenza virus, Rhinovirus and Influenza virus.
  • retrovirus such as human immunodeficiency virus (HIV), Hepatitis C Virus, Hepatitis B Virus, Coronavirus, as well as respiratory viruses, including human Respiratory Syncytial Virus, human Metapneumovirus, human Parainfluenza virus, Rhinovirus and Influenza virus.
  • compositions and methods of this disclosure may be useful as therapeutic tools to treat or prevent symptoms of, for example, hyperproliferative disorders.
  • hyperproliferative disorders may include neoplasms, carcinomas, sarcomas, tumors, or cancer.
  • More exemplary hyperproliferative disorders may include oral cancer, throat cancer, laryngeal cancer, esophageal cancer, pharyngeal cancer, nasopharyngeal cancer, oropharyngeal cancer, gastrointestinal tract cancer, gastrointestinal stromal tumors (GIST), small intestine cancer, colon cancer, rectal cancer, colorectal cancer, anal cancer, pancreatic cancer, breast cancer, cervical cancer, uterine cancer, vulvar cancer, vaginal cancer, urinary tract cancer, bladder cancer, kidney cancer, adrenocortical cancer, islet cell carcinoma, gallbladder cancer, stomach cancer, prostate cancer, ovarian cancer, endometrial cancer, trophoblastic tumor, testicular cancer, penial cancer, bone cancer, osteosarcoma, liver cancer, extrahepatic bile duct cancer, skin cancer, basal cell carcinoma (BCC), lung cancer, small cell lung cancer, non-small cell lung cancer (NSCLC), brain cancer, melanoma, Kaposi'
  • compositions and methods of this disclosure are useful as therapeutic tools to regulate expression of one or more target gene to treat or prevent symptoms of, for example, inflammatory disorders.
  • inflammatory disorders may include diabetes mellitus, rheumatoid arthritis, pannus growth in inflamed synovial lining, collagen-induced arthritis, spondylarthritis, ankylosing spondylitis, multiple sclerosis, encephalomyelitis, inflammatory bowel disease, Crohn's disease, psoriasis or psoriatic arthritis, myasthenia gravis, systemic lupus erythematosis, graft-versus-host disease, atherosclerosis, and allergies.
  • exemplary disorders that may be treated with the siNA molecules, compositions and methods of the instant disclosure include metabolic disorders, cardiac disease, pulmonary disease, neovascularization, ischemic disorders, age-related macular degeneration, diabetic retinopathy, glomerulonephritis, diabetes, asthma, chronic obstructive pulmonary disease, chronic bronchitis, lymphangiogenesis, and atherosclerosis.
  • siNA molecules of the instant invention may provide useful reagents and methods for a variety of therapeutic, prophylactic, cosmetic, veterinary, diagnostic, target validation, genomic discovery, genetic engineering, and pharmacogenomic applications.
  • the present invention in one aspect, also provides for pharmaceutical compositions of the siNA molecules of the invention, i.e., compositions in a pharmaceutically acceptable carrier or diluent.
  • pharmaceutical compositions include salts, esters, or salts of such esters, of the above compounds, e.g., acid addition salts, for example, salts of hydrochloric, hydrobromic, hydroiodic, acetic acid, and benzene sulfonic acid.
  • Other salts include for example, sodium, potassium, manganese, ammonium, and calcium salts.
  • These formulations or compositions can comprise a pharmaceutically acceptable carrier or diluent as is generally known in the art.
  • the pharmaceutical compositions of the present disclosure are formulated to all the siNA molecule(s) described herein to be bioavailable upon administration to a subject.
  • the invention features a pharmaceutical composition comprising any siNA comprising any formula A, B, C, D, E, F, G, H, I, J, K, L, M, N, O, or P as described herein.
  • the siNA molecules of the invention may preferably be formulated as pharmaceutical compositions prior to administering to a subject, according to techniques known in the art.
  • Pharmaceutical compositions of the present invention may be characterized as being at least sterile and pyrogen-free. Methods for preparing pharmaceutical compositions of the invention are within the skill in the art for example as described in Remington's Pharmaceutical Science, 21 st ed., Mack Publishing Company, Easton, Pa., A.R. Gennaro edit., 2005.
  • Non-limiting examples of various types of formulations for local administration include ointments, lotions, creams, gels, foams, preparations for delivery by transdermal patches, powders, sprays, aerosols, capsules or cartridges for use in an inhaler or insufflator or drops (for example eye or nose drops), solutions/suspensions for nebulization, suppositories, pessaries, retention enemas and chewable or suckable tablets or pellets (for example for the treatment of aphthous ulcers) or liposome or microencapsulation preparations.
  • Ointments, creams and gels may, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agent and/or solvents.
  • suitable thickening and/or gelling agent and/or solvents can thus, for example, include water and/or an oil such as liquid paraffin or a vegetable oil such as arachis oil or castor oil, or a solvent such as polyethylene glycol.
  • Various thickening agents and gelling agents can be used depending on the nature of the base.
  • Non-limiting examples of such agents include soft paraffin, aluminum stearate, cetostearyl alcohol, polyethylene glycols, woolfat, beeswax, carboxypolymethylene and cellulose derivatives, and/or glyceryl monostearate and/or non-ionic emulsifying agents.
  • lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilizing agents, dispersing agents, suspending agents or thickening agents.
  • powders for external application may be formed with the aid of any suitable powder base, for example, talc, lactose or starch.
  • Drops may be formulated with an aqueous or non-aqueous base also comprising one or more dispersing agents, solubilizing agents, suspending agents or preservatives.
  • compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions can contain one or more such sweetening agents, flavoring agents, coloring agents or preservative agents in order to provide pharmaceutically elegant and palatable preparations.
  • Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients that are suitable for the manufacture of tablets.
  • excipients may be, for example, inert diluents; such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia ; and lubricating agents, for example magnesium stearate, stearic acid or talc.
  • the tablets may be uncoated or they can be coated by known techniques. In some cases such coatings may be prepared by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monosterate or glyceryl distearate may be employed.
  • Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin or olive oil.
  • an inert solid diluent for example, calcium carbonate, calcium phosphate or kaolin
  • water or an oil medium for example peanut oil, liquid paraffin or olive oil.
  • Aqueous suspensions contain the active materials in a mixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydropropyl-methylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia ; dispersing or wetting agents may be a naturally-occurring phosphatide, for example, lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate; or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan monoole
  • the aqueous suspensions may also contain one or more preservatives, for example ethyl, or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose or saccharin.
  • preservatives for example ethyl, or n-propyl p-hydroxybenzoate
  • coloring agents for example ethyl, or n-propyl p-hydroxybenzoate
  • flavoring agents for example ethyl, or n-propyl p-hydroxybenzoate
  • sweetening agents such as sucrose or saccharin.
  • Oily suspensions may be formulated by suspending the active ingredients in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin.
  • the oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents and flavoring agents may be added to provide palatable oral preparations. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid
  • compositions of the invention may also be in the form of oil-in-water emulsions.
  • the oily phase may be a vegetable oil or a mineral oil or mixtures of these.
  • Suitable emulsifying agents may be naturally-occurring gums, for example gum acacia or gum tragacanth, naturally-occurring phosphatides, for example soy bean, lecithin, and esters or partial esters derived from fatty acids and hexitol, anhydrides, for example sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monooleate.
  • the emulsions may also contain sweetening and flavoring agents.
  • Syrups and elixirs may be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol, glucose or sucrose. Such formulations may also contain a demulcent, a preservative and flavoring and coloring agents.
  • the pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents that have been mentioned above.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parentally acceptable diluent or solvent, for example as a solution in 1,3-butanediol.
  • Suitable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • the nucleic acid molecules of the invention may also be administered in the form of suppositories, e.g., for rectal administration of the drug.
  • suppositories e.g., for rectal administration of the drug.
  • These compositions may be prepared by mixing the drug with a suitable non-irritating excipient that is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • suitable non-irritating excipient that is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • Such materials include cocoa butter and polyethylene glycols.
  • Nucleic acid molecules of the invention may be administered parenterally in a sterile medium.
  • the drug depending on the vehicle and concentration used, may either be suspended or dissolved in the vehicle.
  • adjuvants such as local anesthetics, preservatives and buffering agents may be dissolved in the vehicle.
  • the siNA compositions, or conjugates, and/or delivery formulations provided herein for use in pulmonary delivery further comprise one or more surfactants.
  • Suitable surfactants or surfactant components for enhancing the uptake of the compositions of the invention include synthetic and natural as well as full and truncated forms of surfactant protein A, surfactant protein B, surfactant protein C, surfactant protein D and surfactant Protein E, di-saturated phosphatidylcholine (other than dipalmitoyl), dipalmitoylphosphatidylcholine, phosphatidylcholine, phosphatidylglycerol, phosphatidylinositol, phosphatidylethanolamine, phosphatidylserine; phosphatidic acid, ubiquinones, lysophosphatidylethanolamine, lysophosphatidylcholine, palmitoyl-lysophosphatidylcholine, dehydroepiandros
  • siNAs and pharmaceutical formulations according to the invention may be administered to a subject alone or used in combination with or include one or more other therapeutic agents, for example, antiviral or anticancer agents.
  • other therapeutic agents for example, antiviral or anticancer agents.
  • combinations of the presently disclosed compounds with other antiviral or anti-cancer or chemotherapeutic agents are within the scope of the invention
  • anti-cancer or chemotherapeutic agents may be found in Cancer Principles and Practice of Oncology by V. T. Devita and S. Hellman (editors), 6 th edition (Feb. 15, 2001), Lippincott Williams & Wilkins Publishers. A person of ordinary skill in the art would be able to discern which combinations of agents would be useful based on the particular characteristics of the drugs and the cancer involved.
  • anti-cancer agents include, but are not limited to, the following: estrogen receptor modulators, androgen receptor modulators, retinoid receptor modulators, cytotoxic/cytostatic agents, antiproliferative agents, prenyl-protein transferase inhibitors, HMG-CoA reductase inhibitors and other angiogenesis inhibitors, inhibitors of cell proliferation and survival signaling, apoptosis inducing agents and agents that interfere with cell cycle checkpoints.
  • the invention provides a combination comprising an siNA molecule of the invention or a pharmaceutically acceptable salt, solvate or physiologically functional derivative thereof together with one or more therapeutic agents as described herein or as is otherwise known in the art.
  • estrogen receptor modulators examples include, but are not limited to, tamoxifen, raloxifene, idoxifene, LY353381, LY117081, toremifene, fulvestrant, 4-[7-(2,2-dimethyl-1-oxopropoxy-4-methyl-2-[4-[2-(1-piperidinyl)ethoxy]phenyl]-2H-1-benzopyran-3-yl]-phenyl-2,2-dimethylpropanoate, 4,4′-dihydroxybenzophenone-2,4-dinitrophenyl-hydrazone, and SH646.
  • Examples of androgen receptor modulators that may be used in combination with the compounds of the invention include, but are not limited to, finasteride and other 5 ⁇ -reductase inhibitors, nilutamide, flutamide, bicalutamide, liarozole, and abiraterone acetate.
  • retinoid receptor modulators examples include, but are not limited to, bexarotene, tretinoin, 13-cis-retinoic acid, 9-cis-retinoic acid, ⁇ -difluoromethylornithine, ILX23-7553, trans-N-(4′-hydroxyphenyl) retinamide, and N-4-carboxyphenyl retinamide.
  • cytotoxic agents examples include, but are not limited to, sertenef, cachectin, ifosfamide, tasonermin, lonidamine, carboplatin, altretamine, prednimustine, dibromodulcitol, ranimustine, fotemustine, nedaplatin, oxaliplatin, temozolomide, heptaplatin, estramustine, improsulfan tosilate, trofosfamide, nimustine, dibrospidium chloride, pumitepa, lobaplatin, satraplatin, profiromycin, cisplatin, irofulven, dexifosfamide, cis-aminedichloro(2-methyl-pyridine)platinum, benzylguanine, glufosfamide, GPX100, (trans, trans, trans)-bis-mu-(hex)platinum, benzylguanine
  • hypoxia activatable compound that may be used in combination with the compounds of the invention is tirapazamine.
  • proteasome inhibitors examples include, but are not limited to, lactacystin and bortezomib.
  • microtubule inhibitors/microtubule-stabilising agents examples include, but are not limited to, paclitaxel, vindesine sulfate, 3′,4′-didehydro-4′-deoxy-8′-norvincaleukoblastine, docetaxol, rhizoxin, dolastatin, mivobulin isethionate, auristatin, cemadotin, RPR109881, BMS184476, vinflunine, cryptophycin, 2,3,4,5,6-pentafluoro-N-(3-fluoro-4-methoxyphenyl)benzene sulfonamide, anhydrovinblastine, N,N-dimethyl-L-valyl-L-valyl-N-methyl-L-valyl-L-prolyl-L-proline-t-butylamide, TDX258, the epothilones (see for example U.
  • topoisomerase inhibitors that may be used in combination with the compounds of the invention include, but are not limited to, are topotecan, hycaptamine, irinotecan, rubitecan, 6-ethoxypropionyl-3′,4′-O-exo-benzylidene-chartreusin, 9-methoxy-N,N-dimethyl-5-nitropyrazolo[3,4,5-kl]acridine-2-(6H) propanamine, 1-amino-9-ethyl-5-fluoro-2,3-dihydro-9-hydroxy-4-methyl-1H,12H-benzo[de]pyrano[3′,4′:b,7]-indolizino[1,2b]quinoline-10,13(9H,15H)dione, lurtotecan, 7-[2-(N-isopropylamino)ethyl]-(20S)camptothecin, BNP1350, BNPI1100
  • inhibitors of mitotic kinesins include, but are not limited to, inhibitors described in PCT Publications WO 01/30768, WO 01/98278, WO 03/050,064, WO 03/050,122, WO 03/049,527, WO 03/049,679, WO 03/049,678, WO04/039774, WO03/079973, WO03/099211, WO03/105855, WO03/106417, WO04/037171, WO04/058148, WO04/058700, WO04/126699, WO05/018638, WO05/019206, WO05/019205, WO05/018547, WO05/017190, US2005/0176776.
  • inhibitors of mitotic kinesins include, but are not limited to inhibitors of KSP, inhibitors of MKLP1, inhibitors of CENP-E, inhibitors of MCAK, inhibitors of Kif14, inhibitors of Mphosphl and inhibitors of Rab6-KIFL.
  • histone deacetylase inhibitors examples include, but are not limited to, TSA, oxamflatin, PXD101, MG98, valproic acid and scriptaid. Further reference to other histone deacetylase inhibitors may be found in the following manuscript; Miller, T. A. et al. J. Med. Chem. 46(24):5097-5116 (2003).
  • Inhibitors of kinases involved in mitotic progression include, but are not limited to, inhibitors of aurora kinase, inhibitors of Polo-like kinases (PLK) (in particular inhibitors of PLK-1), inhibitors of bub-1 and inhibitors of bub-R1.
  • PLK Polo-like kinases
  • Antiproliferative agents that may be used in combination with the compounds of the invention include, but are not limited to, antisense RNA and DNA oligonucleotides such as G3139, ODN698, RVASKRAS, GEM231, and INX3001, and antimetabolites such as enocitabine, carmofur, tegafur, pentostatin, doxifluridine, trimetrexate, fludarabine, capecitabine, galocitabine, cytarabine ocfosfate, fosteabine sodium hydrate, raltitrexed, paltitrexid, emitefur, tiazofurin, decitabine, nolatrexed, pemetrexed, nelzarabine, 2′-deoxy-2′-methylidenecytidine, 2′-fluoromethylene-2′-deoxycytidine, N-[5-(2,3-dihydro-benzofuryl)sulfonyl
  • monoclonal antibody targeted therapeutic agents examples include those therapeutic agents which have cytotoxic agents or radioisotopes attached to a cancer cell specific or target cell specific monoclonal antibody, such as, for example, Bexxar.
  • HMG-CoA reductase inhibitors examples include, but are not limited to, lovastatin (MEVACOR®; see U.S. Pat. Nos. 4,231,938, 4,294,926 and 4,319,039), simvastatin (ZOCOR®; see U.S. Pat. Nos. 4,444,784, 4,820,850 and 4,916,239), pravastatin (PRAVACHOL®; see U.S. Pat. Nos. 4,346,227, 4,537,859, 4,410,629, 5,030,447 and 5,180,589), fluvastatin (LESCOL®; see U.S. Pat. Nos.
  • prenyl-protein transferase inhibitors examples include, but are not limited to, may be found in the following publications and patents: WO 96/30343, WO 97/18813, WO 97/21701, WO 97/23478, WO 97/38665, WO 98/28980, WO 98/29119, WO 95/32987, U.S. Pat. No. 5,420,245, U.S. Pat. No. 5,523,430, U.S. Pat. No. 5,532,359, U.S. Pat. No. 5,510,510, U.S. Pat. No. 5,589,485, U.S. Pat. No. 5,602,098, European Patent Publ.
  • angiogenesis inhibitors examples include, but are not limited to, tyrosine kinase inhibitors, such as inhibitors of the tyrosine kinase receptors Flt-1 (VEGFR1) and Flk-1/KDR (VEGFR2), inhibitors of epidermal-derived, fibroblast-derived, or platelet derived growth factors, MMP (matrix metalloprotease) inhibitors, integrin blockers, interferon- ⁇ , interleukin-12, pentosan polysulfate, cyclooxygenase inhibitors, including nonsteroidal anti-inflammatories (NSAIDs) like aspirin and ibuprofen as well as selective cyclooxy-genase-2 inhibitors like celecoxib and rofecoxib ( PNAS , Vol.
  • NSAIDs nonsteroidal anti-inflammatories
  • steroidal anti-inflammatories such as corticosteroids, mineralocorticoids, dexamethasone, prednisone, prednisolone, methylpred, betamethasone), carboxyamidotriazole, combretastatin A-4, squalamine, 6-O-chloroacetyl-carbonyl)-fumagillol, thalidomide, angiostatin, troponin-1, angiotensin II antagonists (see Fernandez et al., J. Lab. Clin. Med.
  • agents that modulate or inhibit angiogenesis may also be used in combination with the compounds of the instant invention and include agents that modulate or inhibit the coagulation and fibrinolysis systems (see review in Clin. Chem. La. Med. 38:679-692 (2000)).
  • agents that modulate or inhibit the coagulation and fibrinolysis pathways include, but are not limited to, heparin (see Thromb. Haemost. 80:10-23 (1998)), low molecular weight heparins and carboxypeptidase U inhibitors (also known as inhibitors of active thrombin activatable fibrinolysis inhibitor [TAFIa]) (see Thrombosis Res. 0.101:329-354 (2001)).
  • TAFIa inhibitors have been described in PCT Publication WO 03/013,526 and U.S. Ser. No. 60/349,925 (filed Jan. 18, 2002).
  • Agents that interfere with cell cycle checkpoints include, but are not limited to, inhibitors of ATR, ATM, the Chk1 and Chk2 kinases and cdk and cdc kinase inhibitors and are specifically exemplified by 7-hydroxystaurosporin, flavopiridol, CYC202 (Cyclacel) and BMS-387032.
  • RTKs receptor tyrosine kinases
  • agents that interfere with receptor tyrosine kinases (RTKs) include, but are not limited to, inhibitors of c-Kit, Eph, PDGF, Flt3 and HBV.
  • Further agents include inhibitors of RTKs as described by Bume-Jensen and Hunter, Nature, 411:355-365, 2001.
  • Inhibitors of cell proliferation and survival signaling pathway that may be used in combination with the compounds of the invention include, but are not limited to, inhibitors of EGFR (for example gefitinib and erlotinib), inhibitors of ERB-2 (for example trastuzumab), inhibitors of IGFR, inhibitors of cytokine receptors, inhibitors of HBV, inhibitors of PI3K (for example LY294002), serine/threonine kinases (including but not limited to inhibitors of Akt such as described in WO 02/083064, WO 02/083139, WO 02/083140, US 2004-0116432, WO 02/083138, US 2004-0102360, WO 03/086404, WO 03/086279, WO 03/086394, WO 03/084473, WO 03/086403, WO 2004/041162, WO 2004/096131, WO 2004/096129, WO 2004/096135, WO 2004/096130,
  • Apoptosis inducing agents that may be used in combination with the compounds of the invention include, but are not limited to, activators of TNF receptor family members (including the TRAIL receptors).
  • NSAIDs that are selective COX-2 inhibitors that may be used in combination with the compounds of the invention include, but are not limited to, those NSAIDs disclosed in U.S. Pat. No. 5,474,995, U.S. Pat. No. 5,861,419, U.S. Pat. No. 6,001,843, U.S. Pat. No. 6,020,343, U.S. Pat. No. 5,409,944, U.S. Pat. No. 5,436,265, U.S. Pat. No. 5,536,752, U.S. Pat. No. 5,550,142, U.S. Pat. No. 5,604,260, U.S. Pat. No. 5,698,584, U.S. Pat. No.
  • Inhibitors of COX-2 that are particularly useful in combination with the compounds of the invention include: 3-phenyl-4-(4-(methylsulfonyl)phenyl)-2-(5H)-furanone; and 5-chloro-3-(4-methylsulfonyl)-phenyl-2-(2-methyl-5-pyridinyl)pyridine; or a pharmaceutically acceptable salt thereof.
  • Angiogenesis inhibitors that may be used in combination with the compounds of the invention include, but are not limited to, endostatin, ukrain, ranpirnase, IM862, 5-methoxy-4-[2-methyl-3-(3-methyl-2-butenyl)oxiranyl]-1-oxaspiro[2,5]oct-6-yl(chloroacetyl)carbamate, acetyldinanaline, 5-amino-1-[[3,5-dichloro-4-(4-chlorobenzoyl)-phenyl]methyl]-1H-1,2,3-triazole-4-carboxamide, CM101, squalamine, combretastatin, RPI4610, NX31838, sulfated mannopentaose phosphate, 7,7-(carbonyl-bis[imino-N-methyl-4,2-pyrrolocarbonylimino[N-methyl-4,2-pyrrole]-carbonylimino
  • Tyrosine kinase inhibitors that may be used in combination with the compounds of the invention include, but are not limited to, N-(trifluoromethylphenyl)-5-methylisoxazol-4-carboxamide, 3-[(2,4-dimethylpyrrol-5-yl)methylidenyl)indolin-2-one, 17-(allylamino)-17-demethoxygeldanamycin, 4-(3-chloro-4-fluorophenylamino)-7-methoxy-6-[3-(4-morpholinyl)propoxyl]quinazoline, N-(3-ethynylphenyl)-6,7-bis(2-methoxyethoxy)-4-quinazolinamine, BIBX1382, 2,3,9,10,11,12-hexahydro-10-(hydroxymethyl)-10-hydroxy-9-methyl-9,12-epoxy-1H-diindolo[1,2,3-fg:3′,2′,1′-
  • Combinations with compounds other than anti-cancer compounds are also encompassed in the instant compositions and methods.
  • combinations of the instantly claimed compounds with PPAR- ⁇ (i.e., PPAR-gamma) agonists and PPAR- ⁇ (i.e., PPAR-delta) agonists are useful in the treatment of certain malignancies.
  • PPAR- ⁇ and PPAR- ⁇ are the nuclear peroxisome proliferator-activated receptors y and ⁇ .
  • the expression of PPAR- ⁇ on endothelial cells and its involvement in angiogenesis has been reported in the literature (see J. Cardiovasc. Pharmacol. 31:909-913 (1998); J. Biol. Chem. 274:9116-9121 (1999); Invest.
  • Examples of PPAR- ⁇ agonists and PPAR- ⁇ / ⁇ agonists that may be used in combination with the compounds of the invention include, but are not limited to, thiazolidinediones (such as DRF2725, CS-011, troglitazone, rosiglitazone, and pioglitazone), fenofibrate, gemfibrozil, clofibrate, GW2570, SB219994, AR-H039242, JTT-501, MCC-555, GW2331, GW409544, NN2344, KRP297, NP0110, DRF4158, NN622, G1262570, PNU182716, DRF552926, 2-[(5,7-dipropyl-3-trifluoromethyl-1,2-benzisoxazol-6-yl)oxy]-2-methylpropionic acid (disclosed in U.S.
  • thiazolidinediones such as DRF2725, CS-011, tro
  • Another embodiment of the instant invention is the use of the presently disclosed compounds in combination with gene therapy for the treatment of cancer.
  • Gene therapy may be used to deliver any tumor suppressing gene. Examples of such genes include, but are not limited to, p53, which may be delivered via recombinant virus-mediated gene transfer (see U.S. Pat. No.
  • a uPA/uPAR antagonist (“Adenovirus-Mediated Delivery of a uPA/uPAR Antagonist Suppresses Angiogenesis-Dependent Tumor Growth and Dissemination in Mice,” Gene Therapy , August 5(8):1105-13 (1998)), and interferon gamma (J Immunol 164:217-222 (2000)).
  • the compounds of the instant invention may also be administered in combination with an inhibitor of inherent multidrug resistance (MDR), in particular MDR associated with high levels of expression of transporter proteins.
  • MDR inhibitors include inhibitors of p-glycoprotein (P-gp), such as LY335979, XR9576, OC144-093, R101922, VX853 and PSC833 (valspodar).
  • a compound of the present invention may be employed in conjunction with anti-emetic agents to treat nausea or emesis, including acute, delayed, late-phase, and anticipatory emesis, which may result from the use of a compound of the present invention, alone or with radiation therapy.
  • a compound of the present invention may be used in conjunction with other anti-emetic agents, especially neurokinin-1 receptor antagonists, 5HT3 receptor antagonists, such as ondansetron, granisetron, tropisetron, and zatisetron, GABAB receptor agonists, such as baclofen, a corticosteroid such as Decadron (dexamethasone), Kenalog, Aristocort, Nasalide, Preferid, Benecorten or others such as disclosed in U.S. Pat. Nos.
  • neurokinin-1 receptor antagonists especially 5HT3 receptor antagonists, such as ondansetron, granisetron, tropisetron, and zatisetron, GABAB receptor agonists, such as baclofen, a corticosteroid such as Decadron (dexamethasone), Kenalog, Aristocort, Nasalide, Preferid, Benecorten or others such as disclosed in U.S. Pat. Nos.
  • an antidopaminergic such as the phenothiazines (for example prochlorperazine, fluphenazine, thioridazine and mesoridazine), metoclopramide or dronabinol.
  • an anti-emesis agent selected from a neurokinin-1 receptor antagonist, a 5HT3 receptor antagonist and a corticosteroid is administered as an adjuvant for the treatment or prevention of emesis that may result upon administration of the instant compounds.
  • Neurokinin-1 receptor antagonists of use in conjunction with the compounds of the present invention are fully described, for example, in U.S. Pat. Nos. 5,162,339, 5,232,929, 5,242,930, 5,373,003, 5,387,595, 5,459,270, 5,494,926, 5,496,833, 5,637,699, 5,719,147; European Patent Publication Nos.
  • the neurokinin-1 receptor antagonist for use in conjunction with the compounds of the present invention is selected from: 2-(R)-(1-(R)-(3,5-bis(trifluoromethyl)-phenyl)ethoxy)-3-(S)-(4-fluorophenyl)-4-(3-(5-oxo-1H,4H-1,2,4-triazolo)methyl)morpholine, or a pharmaceutically acceptable salt thereof, which is described in U.S. Pat. No. 5,719,147.
  • a compound of the instant invention may also be administered with an agent useful in the treatment of anemia.
  • an anemia treatment agent is, for example, a continuous eythropoiesis receptor activator (such as epoetin alfa).
  • a compound of the instant invention may also be administered with an agent useful in the treatment of neutropenia.
  • a neutropenia treatment agent is, for example, a hematopoietic growth factor which regulates the production and function of neutrophils such as a human granulocyte colony stimulating factor, (G-CSF).
  • G-CSF human granulocyte colony stimulating factor
  • Examples of a G-CSF include filgrastim and PEG-filgrastim.
  • a compound of the instant invention may also be administered with an immunologic-enhancing drug, such as levamisole, isoprinosine and Zadaxin.
  • an immunologic-enhancing drug such as levamisole, isoprinosine and Zadaxin.
  • a compound of the instant invention may also be useful for treating or preventing liver disease or cancer in combination with other siNA therapeutics.
  • the compounds of the instant invention may also be administered in combination with ⁇ -secretase inhibitors and/or inhibitors of NOTCH signaling.
  • Such inhibitors include compounds described in WO 01/90084, WO 02/30912, WO 01/70677, WO 03/013506, WO 02/36555, WO 03/093252, WO 03/093264, WO 03/093251, WO 03/093253, WO 2004/039800, WO 2004/039370, WO 2005/030731, WO 2005/014553, U.S. Ser. No.
  • a compound of the instant invention may also be useful for treating or preventing cancer in combination with PARP inhibitors.
  • a compound of the instant invention may also be useful for treating cancer in combination with the following therapeutic agents: abarelix (Plenaxis Depot®); aldesleukin (Prokine®); Aldesleukin (Proleukin®); Alemtuzumabb (Campath®); alitretinoin (Panretin®); allopurinol (Zyloprim®); altretamine (Hexalen®); amifostine (Ethyol®); anastrozole (Arimidex®); arsenic trioxide (Trisenox®); asparaginase (Elspar®); azacitidine (Vidaza®); bendamustine hydrochloride (Treanda®); bevacuzimab (Avastin®); bexarotene capsules (Targretin®); bexarotene gel (Targretin®); bleomycin (Blenoxane®); bortezomib (Velcade®); brefeldin
  • the invention also provides a combination comprising an siNA molecule of the invention targeting one gene together with another inhibitor targeting a second target gene.
  • compositions comprising a combination as defined above together with a pharmaceutically acceptable diluent or carrier represent a further aspect of the invention.
  • an siNA molecule is administered simultaneously or sequentially in a coordinated treatment protocol with one or more secondary or adjunctive therapeutic agents described herein or known in the art.
  • the coordinate administration may be done in either order, and there may be a time period while only one or both (or all) active therapeutic agents, individually or collectively, exert their biological activities.
  • a distinguishing aspect of all such coordinate treatment methods is that the siNA molecule(s) present in a composition elicits some favorable clinical response, which may or may not be in conjunction with a secondary clinical response provided by the secondary therapeutic agent.
  • the coordinate administration of an siNA molecule with a secondary therapeutic agent as contemplated herein may yield an enhanced (e.g., synergistic) therapeutic response beyond the therapeutic response elicited by either or both the purified siNA molecule and the secondary therapeutic agent alone.
  • the individual compounds of such combinations may be administered either sequentially or simultaneously in separate or combined pharmaceutical formulations.
  • the individual compounds will be administered simultaneously in a combined pharmaceutical formulation.
  • the described molecules could be used in combination with one or more known compounds, treatments, or procedures to prevent or treat diseases, disorders, conditions, and traits described herein in a subject or organism as are known in the art, such as other gene inhibitors.
  • RNAi research indicates the need for methods that can modulate gene expression for therapeutic use.
  • one aspect of the invention comprises a method of treating a subject including, but not limited to, a human suffering from a disease or a condition which is mediated by the action of target gene expression, which method comprises administering to said subject an effective amount of a double-stranded siNA molecule of the invention.
  • the siNA molecules comprises sequence having at least a 15 nucleotides complementary to a target nucleic acid.
  • the siNA molecule comprises any molecule herein having formula A, B, C, D, E, F, G, H, I, J, K, L, M, N, O or P.
  • the disease or condition may be cancer, a proliferative, inflammatory, autoimmune, neurologic, ocular, respiratory, metabolic, dermatological, auditory, liver, kidney, or infectious disease as described herein or otherwise known in the art.
  • the molecules and compositions of the instant invention may be useful in a method for treating cancer, proliferative, inflammatory, autoimmune, neurologic, ocular, respiratory, metabolic, dermatological, auditory, liver, kidney, or infectious diseases.
  • the administration of the siNA molecule may be via local administration or systemic administration.
  • the invention features contacting the subject or organism with an siNA molecule of the invention via local administration to relevant tissues or cells, such as lung cells and tissues, such as via pulmonary delivery.
  • the invention features contacting the subject or organism with an siNA molecule of the invention via systemic administration (such as via intravenous or subcutaneous administration of siNA) to relevant tissues or cells in a subject or organism.
  • siNA molecules of the invention may also used as reagents in ex vivo applications.
  • siNA reagents may be introduced into tissue or cells that are transplanted into a subject for therapeutic effect.
  • the cells and/or tissue may be derived from an organism or subject that later receives the explant, or may be derived from another organism or subject prior to transplantation.
  • the siNA molecules may be used to modulate the expression of one or more genes in the cells or tissue, such that the cells or tissue obtain a desired phenotype or are able to perform a function when transplanted in vivo.
  • certain target cells from a patient are extracted.
  • These extracted cells are contacted with siNAs targeting a specific nucleotide sequence within the cells under conditions suitable for uptake of the siNAs by these cells (e.g., using delivery reagents such as cationic lipids, liposomes and the like or using techniques such as electroporation to facilitate the delivery of siNAs into cells).
  • delivery reagents such as cationic lipids, liposomes and the like or using techniques such as electroporation to facilitate the delivery of siNAs into cells.
  • the cells are then reintroduced back into the same patient or other patients.
  • a pharmaceutically effective dose of the siNA molecules or pharmaceutical compositions of the invention is administered to the subject.
  • a pharmaceutically effective dose is that dose required to prevent, inhibit the occurrence, or treat (alleviate a symptom to some extent, preferably all of the symptoms) a disease state.
  • One skilled in the art may readily determine a therapeutically effective dose of the siNA of the invention to be administered to a given subject, by taking into account factors, such as the size and weight of the subject, the extent of the disease progression or penetration, the age, health, and sex of the subject, the route of administration, and whether the administration is regional or systemic.
  • an amount between 0.1 ⁇ g/kg and 140 mg/kg body weight/day of active ingredients is administered dependent upon potency of the siNA of the disclosure.
  • the amount of active ingredient that may be combined with the carrier materials to produce a single dosage form varies depending upon the host treated and the particular mode of administration. Optimal dosing schedules may be calculated from measurements of drug accumulation in the body of the patient.
  • the siNA molecules of the invention may be administered in a single dose or in multiple doses.
  • siNA molecules of the instant invention may be administered once monthly, once weekly, once daily (QD), or divided into multiple monthly, weekly, or daily doses, such as, for example, but not limitation, twice daily (BID), three times daily (TID), once every two weeks.
  • BID twice daily
  • TID three times daily
  • Persons of ordinary skill in the art may easily estimate repetition rates for dosing based on measured residence times and concentrations of the drug in bodily fluids or tissues.
  • the administration may be continuous, i.e., every day, or intermittently.
  • intermittent administration of a compound of the instant invention may be administration one to six days per week or it may mean administration in cycles (e.g. daily administration for two to eight consecutive weeks, then a rest period with no administration for up to one week) or it may mean administration on alternate days.
  • compositions or formulations may be administered in a variety of ways.
  • administration methods of the invention include oral, buccal, sublingual, parenteral (i.e., intraarticularly, intravenously, intraperitoneally, subcutaneously, or intramuscularly), local rectal administration or other local administration.
  • the composition of the invention may be administered by insufflation and inhalation. Administration may be accomplished via single or divided doses.
  • the pharmaceutical compositions are administered intravenously or intraperitoneally by a bolus injection (see, e.g., U.S. Pat. No. 5,286,634).
  • siNA molecule with or without a vehicle may be locally delivered by direct injection or by use of an infusion pump.
  • Direct injection of the siNA molecules of this disclosure may take place using standard needle and syringe methodologies, or by needle free technologies, such as those described in Conroy et al, (1999 , Clin. Cancer Res. 5:2330) and PCT Publication No. WO 99/31262.
  • lipid nucleic acid particles may be administered by direct injection at the site of disease or by injection at a site distal from the site of disease (see, e.g., Culver, HUMAN GENE THERAPY, MaryAnn Liebert, Inc., Publishers, New York. pp. 70-71(1994)).
  • the siNA molecules of the invention and formulations or compositions thereof are administered to a cell, subject, or organism as is described herein and as is generally known in the art.
  • the siNA may be administered to the subject systemically as described herein or otherwise known in the art, either alone as a monotherapy or in combination with additional therapies described herein or as are known in the art.
  • Systemic administration may include, for example, pulmonary (inhalation, nebulization etc.) intravenous, subcutaneous, intramuscular, catheterization, nasopharyngeal, transdermal, or oral/gastrointestinal administration as is generally known in the art.
  • the siNA may be administered to the subject locally or to local tissues as described herein or otherwise known in the art, either alone as a monotherapy or in combination with additional therapies as are known in the art.
  • Local administration may include, for example, inhalation, nebulization, catheterization, implantation, direct injection, dermal/transdermal application, patches, stenting, ear/eye drops, or portal vein administration to relevant tissues, or any other local administration technique, method or procedure, as is generally known in the art.
  • the siNA molecules of the invention and formulations or compositions thereof are administered to the liver as is generally known in the art (see for example Wen et al., 2004, World J Gastroenterol., 10, 244-9; Murao et al., 2002, Pharm Res., 19, 1808-14; Liu et al., 2003 , gene Ther., 10, 180-7; Hong et al., 2003, J Pharm Pharmacol., 54, 51-8; Herrmann et al., 2004 , Arch Virol., 149, 1611-7; and Matsuno et al., 2003 , gene Ther., 10, 1559-66).
  • the invention features the use of methods to deliver the siNA molecules of the instant invention to hematopoietic cells, including monocytes and lymphocytes. These methods are described in detail by Hartmann et al., 1998, J. Phamacol. Exp. Ther., 285(2), 920-928; Kronenwett et al., 1998, Blood, 91(3), 852-862; Filion and Phillips, 1997, Biochim. Biophys. Acta., 1329(2), 345-356; Ma and Wei, 1996, Leuk. Res., 20(11/12), 925-930; and Bongartz et al., 1994 , Nucleic Acids Research, 22(22), 4681-8.
  • the siNA molecules of the invention and formulations or compositions thereof are administered directly or topically (e.g., locally) to the dermis or follicles as is generally known in the art (see for example Brand, 2001, Curr. Opin. Mol. Ther., 3, 244-8; Regnier et al., 1998, J. Drug Target, 5, 275-89; Kanikkannan, 2002, BioDrugs, 16, 339-47; Wraight et al., 2001, Pharmacol. Ther., 90, 89-104; and Preat and Dujardin, 2001, STP PharmaSciences, 11, 57-68).
  • the siNA molecules of the invention and formulations or compositions thereof are administered directly or topically using a hydroalcoholic gel formulation comprising an alcohol (e.g., ethanol or isopropanol), water, and optionally including additional agents such isopropyl myristate and carbomer 980.
  • a hydroalcoholic gel formulation comprising an alcohol (e.g., ethanol or isopropanol), water, and optionally including additional agents such isopropyl myristate and carbomer 980.
  • the siNA are formulated to be administered topically to the nasal cavity.
  • Topical preparations may be administered by one or more applications per day to the affected area; over skin areas occlusive dressings may advantageously be used. Continuous or prolonged delivery may be achieved by an adhesive reservoir system.
  • an siNA molecule of the invention is administered iontophoretically, for example to a particular organ or compartment (e.g., the eye, back of the eye, heart, liver, kidney, bladder, prostate, tumor, CNS etc.).
  • a particular organ or compartment e.g., the eye, back of the eye, heart, liver, kidney, bladder, prostate, tumor, CNS etc.
  • iontophoretic delivery are described in, for example, WO 03/043689 and WO 03/030989, which are incorporated by reference in their entireties herein.
  • the siNA molecules of the invention and formulations or compositions thereof are administered to the lung as is described herein and as is generally known in the art.
  • the siNA molecules of the invention and formulations or compositions thereof are administered to lung tissues and cells as is described in U.S. Patent Publication Nos. 2006/0062758; 2006/0014289; and 2004/0077540.
  • compositions of the present invention may be made into aerosol formulations (i.e., they may be “nebulized”) to be administered via inhalation (e.g., intranasally or intratracheally) (see, Brigham et al., Am. J. Sci., 298:278 (1989)). Aerosol formulations may be placed into pressurized acceptable propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like.
  • the siNA molecules of the invention and formulations thereof are administered via pulmonary delivery, such as by inhalation of an aerosol or spray dried formulation administered by an inhalation device or nebulizer, providing rapid local uptake of the nucleic acid molecules into relevant pulmonary tissues.
  • Solid particulate compositions containing respirable dry particles of micronized nucleic acid compositions may be prepared by grinding dried or lyophilized nucleic acid compositions, and then passing the micronized composition through, for example, a 400 mesh screen to break up or separate out large agglomerates.
  • a solid particulate composition comprising the siNA compositions of the invention may optionally contain a dispersant which serves to facilitate the formation of an aerosol as well as other therapeutic compounds.
  • a suitable dispersant is lactose, which may be blended with the nucleic acid compound in any suitable ratio, such as a 1 to 1 ratio by weight.
  • Spray compositions comprising siNA molecules or compositions of the invention may, for example, be formulated as aqueous solutions or suspensions or as aerosols delivered from pressurized packs, such as a metered dose inhaler, with the use of a suitable liquefied propellant.
  • aerosol compositions of the invention suitable for inhalation may be either a suspension or a solution and generally contain an siNA molecule comprising formula (A), and a suitable propellant such as a fluorocarbon or hydrogen-containing chlorofluorocarbon or mixtures thereof, particularly hydrofluoroalkanes, especially 1,1,1,2-tetrafluoroethane, 1,1,1,2,3,3,3-heptafluoro-n-propane or a mixture thereof.
  • the aerosol composition may optionally contain additional formulation excipients well known in the art such as surfactants.
  • additional formulation excipients well known in the art such as surfactants.
  • Non-limiting examples include oleic acid, lecithin or an oligolactic acid or derivative such as those described in WO94/21229 and WO98/34596 and co-solvents for example ethanol.
  • a pharmaceutical aerosol formulation of the invention comprising a compound of the invention and a fluorocarbon or hydrogen-containing chlorofluorocarbon or mixtures thereof as propellant, optionally in combination with a surfactant and/or a co-solvent.
  • the aerosol formulations of the invention may be buffered by the addition of suitable buffering agents.
  • Aerosol formulations may include optional additives including preservatives if the formulation is not prepared sterile.
  • Non-limiting examples include, methyl hydroxybenzoate, anti-oxidants, flavorings, volatile oils, buffering agents and emulsifiers and other formulation surfactants.
  • fluorocarbon or perfluorocarbon carriers are used to reduce degradation and provide safer biocompatible non-liquid particulate suspension compositions of the invention (e.g., siNA and/or LNP formulations thereof).
  • a device comprising a nebulizer delivers a composition of the invention (e.g., siNA and/or LNP formulations thereof) comprising fluorochemicals that are bacteriostatic thereby decreasing the potential for microbial growth in compatible devices.
  • Capsules and cartridges comprising the composition of the invention for use in an inhaler or insufflator, of for example gelatin, may be formulated containing a powder mix for inhalation of a compound of the invention and a suitable powder base such as lactose or starch.
  • each capsule or cartridge contains an siNA molecule comprising formula (A), and one or more excipients.
  • the compound of the invention may be presented without excipients such as lactose
  • the aerosol compositions of the present invention may be administered into the respiratory system as a formulation including particles of respirable size, e.g. particles of a size sufficiently small to pass through the nose, mouth and larynx upon inhalation and through the bronchi and alveoli of the lungs.
  • respirable particles range from about 0.5 to 10 microns in size.
  • the particulate range may be from 1 to 5 microns.
  • the particulate range may be from 2 to 3 microns.
  • Particles of non-respirable size which are included in the aerosol tend to deposit in the throat and be swallowed, and the quantity of non-respirable particles in the aerosol is thus minimized.
  • a particle size in the range of 10-500 um is preferred to ensure retention in the nasal cavity.
  • an siNA composition of the invention is administered topically to the nose for example, for the treatment of rhinitis, via pressurized aerosol formulations, aqueous formulations administered to the nose by pressurized pump or by nebulization.
  • Suitable formulations contain water as the diluent or carrier for this purpose.
  • the aqueous formulations for administration of the composition of the invention to the lung or nose may be provided with conventional excipients such as buffering agents, tonicity modifying agents and the like.
  • siNA molecules of the invention may be formulated and delivered as particles and/or aerosols as discussed above and dispensed from various aerosolization devices known by those of skill in the art.
  • Aerosols of liquid or non-liquid particles comprising an siNA molecule or formulation of the invention may be produced by any suitable means, such as with a device comprising a nebulizer (see for example U.S. Pat. No. 4,501,729) such as ultrasonic or air jet nebulizers.
  • a nebulizer see for example U.S. Pat. No. 4,501,729
  • ultrasonic or air jet nebulizers such as ultrasonic or air jet nebulizers.
  • Solid particle aerosols comprising an siNA molecule or formulation of the invention and surfactant may be produced with any solid particulate aerosol generator.
  • One type of solid particle aerosol generator used with the siNA molecules of the invention is an insufflator.
  • a second type of illustrative aerosol generator comprises a metered dose inhaler (“MDI”). MDIs containing siNA molecules or formulations taught herein may be prepared by methods of the art (for example, see Byron, above and WO96/32099).
  • the siNA molecules may also be formulated as a fluid formulation for delivery from a fluid dispenser, such as those described and illustrated in WO05/044354.
  • nebulizer devices are used in applications for conscious, spontaneously breathing subjects, and for controlled ventilated subjects of all ages.
  • the nebulizer devices may be used for targeted topical and systemic drug delivery to the lung.
  • a device comprising a nebulizer is used to deliver an siNA molecule or formulation of the invention locally to lung or pulmonary tissues.
  • a device comprising a nebulizer is used to deliver a an siNA molecule or formulation of the invention systemically.
  • the siNA molecules of the invention may also be used for diagnostic applications, research applications, and/or manufacture of medicants.
  • the invention features a method for diagnosing a disease, trait, or condition in a subject comprising administering to the subject a composition of the invention under conditions suitable for the diagnosis of the disease, trait, or condition in the subject.
  • siNA molecules of the invention may be used to down regulate or inhibit the expression of proteins arising from haplotype polymorphisms that are associated with a trait, disease or condition in a subject or organism. Analysis of genes, or protein or RNA levels may be used to identify subjects with such polymorphisms or those subjects who are at risk of developing traits, conditions, or diseases described herein. These subjects may be amenable to treatment, for example, treatment with siNA molecules of the invention and any other composition useful in treating diseases related to target gene expression. As such, analysis of protein or RNA levels may be used to determine treatment type and the course of therapy in treating a subject. Monitoring of protein or RNA levels may be used to predict treatment outcome and to determine the efficacy of compounds and compositions that modulate the level and/or activity of certain peptides and/or proteins associated with a trait, disorder, condition, or disease.
  • the invention comprises use of a double-stranded nucleic acid according to the invention for use in the manufacture of a medicament.
  • the medicament is for use in treating a disease or a condition that is mediated by the action of one or more target genes.
  • the medicant is for use in treating any disease or condition herein or otherwise known in the art.
  • the medicament is for use in the treatment of cancer.
  • the siNA molecules of the invention may be for use in a method for treating any disease or condition contemplated herein.
  • the siNA molecules of the invention may be used in a method for treating cancer.
  • siRNAs with drug-like properties are dependent on the incorporation of chemical modifications to improve duration of RNA knockdown and potency while minimizing non-specific (off-target) effects and stimulation of innate immunity.
  • the growing diversity of siRNA delivery vehicles has expanded beyond traditional lipid encapsulated nano-particles to include dynamic polymer conjugates and conjugation with various targeting ligands including antibodies, sugars, and cholesterol.
  • Many of these delivery strategies expose the siRNA cargo to serum and/or cellular nucleases which can compromise the structural integrity and in vivo efficacy of both unmodified siRNAs and modified siRNAs which have not been optimized for nuclease stability.
  • siRNA stability is needed for development of therapeutic siRNAs with cross-delivery platform compatibility and potentially enhanced duration due to improvement of stability attributes along with pharmacokinetic and pharmacodynamic profiles of the siRNA molecule.
  • modified siNA molecules disclosed herein may improve serum stability while maintaining robust potency.
  • the modification criteria of the present invention may be applied to any siRNA sequence.
  • LNP lipid nanoparticle
  • PC polymer conjugates
  • delivery targeting ligands e.g., galactosamine, folate, cholesterol, etc.
  • siNA molecules having Formula A-P originated with experiments aimed that exploring the role of placing 2′-O-methyl chemical modifications at specific positions within the siNA guide strand.
  • the rationale was to identify specific positions of the guide strand that were tolerant or sensitive to chemical modifications and to use this information to map interactions between the guide strand and the RNA binding pocket within the Ago2 protein.
  • This position-based approach was also a distinct and complementary to the approach of assigning modifications based on the sequence content of the siNA (e.g. purine or pyrimidine).
  • siNA motif having alternating 2′-O-methyl modifications at odd positions on the guide strand e.g. positions 1, 3, 5, etc
  • ribonucleotides at even positions e.g. positions 2, 4, 6, etc.
  • siNA molecules with such alternating motifs were then designed to target ApoB and were complexed with a PVE-based polymer conjugate delivery vehicle for characterization in vivo.
  • the siNA complexes were administered to mice by intravenous injection at a concentration of 3 mg per kg (mpk). A significant improvement in ApoB mRNA reduction was conferred by the inclusion of 2′-deoxy-2′-fluoro modifications within the guide strand as shown in FIG. 5 .
  • FIG. 5 shows that the same guide strand motif was not effective when delivered to mice using a tetra-GalNAc conjugate (4GalNAc) single chemical entity (SCE) delivery platform.
  • FIG. 6 shows that for both ApoB and SSB targets, the SCE delivered guide strands having alternating 2′-O-methyl and 2′-deoxy-2′-fluoro modifications did not significantly reduce mRNA expression levels in mice.
  • an ApoB targeted tetra-GalNAc conjugate (4GalNAc) single chemical entity (SCE) siNA having 2′-O-methyl pyrimidines and 2′-deoxy-2′-fluoro purines with phosphorothioate modified ribonucleotides at positions 1-3 of the guide strand when administered to mice via SC delivery, did demonstrate dose dependent activity in mice ( FIG. 7 ).
  • the key difference between the sequences tested is the presence of three phosphorothioate linkages at the 5′ end of guide strand in the active motif.
  • the SSB siNA construct was modified accordingly. As shown in FIG. 8 , the SSB siNA construct having phosphorothioates at the 5′-end of the guide strand showed activity as a tetra-GalNAc conjugate.
  • siNA modification motifs were tested in different biological media including serum, S9, and lysosomal lysate fractions. This data was compared to in vivo metabolism data observed in mouse and rhesus. The lysosomal lysate fractions are enriched for late endosome/lysosome vesicles and serve as a source of lysosomal enzymes. The benefit of improved stability in lysosomal lysates at pH 5.5 and in mouse or rhesus liver was observed with the inclusion of 5′-terminal guide strand phosphorothioate linkages (Table 6). These results demonstrate that the siNA single chemical entity conjugates require more stability in during exposure to the endocytosis pathway compared to polymer conjugate delivery systems.
  • siNA molecules having phosphorothioates at the 5′-end of the guide strand are still susceptible to degradation in lysosomal lysates as well in mouse or rhesus liver (Table 6).
  • the poor in vivo activity of the siNA molecules having 2′-modifications at the 5′-end of the guide strand can be rescued by the inclusion of phosphorothioate modifications at the 5′ end of the guide strands at position 1 and optionally through position 3.
  • siNAs targeting CTNNB1 were synthesized and conjugated to the same SCE delivery vehicle used in FIG. 6 .
  • the two individual, complementary strands of the siNA were synthesized separately using solid phase synthesis, then purified separately by reversed phase solid phase extraction (SPE).
  • SPE reversed phase solid phase extraction
  • the complementary strands were annealed to form the double strand (duplex) and delivered in the desired concentration and buffer of choice.
  • the single strand oligonucleotides were synthesized using phosphoramidite chemistry on an automated solid-phase synthesizer, using procedures as are generally known in the art (see for example U.S. application Ser. No. 12/064,014).
  • a synthesis column was packed with solid support derivatized with the first nucleoside residue (natural or chemically modified). Synthesis was initiated by detritylation of the acid labile 5′-O-dimethoxytrityl group to release the 5′-hydroxyl.
  • a suitably protected phosphoramidite and a suitable activator in acetonitrile were delivered simultaneously to the synthesis column resulting in coupling of the amidite to the 5′-hydroxyl.
  • the column was then washed with a solvent, such as acetonitrile.
  • An oxidizing solution such as an iodine solution was pumped through the column to oxidize the phosphite triester linkage P(III) to its phosphotriester P(V) analog.
  • Unreacted 5′-hydroxyl groups were capped using reagents such as acetic anhydride in the presence of 2,6-lutidine and N-methylimidazole.
  • the elongation cycle was resumed with the detritylation step for the next phosphoramidite incorporation. This process was repeated until the desired sequence was synthesized.
  • the synthesis concluded with the final 5′-terminus protecting group (trityl or 5′-O-dimethoxytrityl).
  • the solid-support and associated oligonucleotide were dried under argon pressure or vacuum.
  • Aqueous base was added and the mixture was heated to effect cleavage of the succinyl linkage, removal of the cyanoethyl phosphate protecting group, and deprotection of the exocyclic amine protection.
  • the trityl-on solution of each crude single strand was purified using chromatographic purification, such as SPE RPC purification.
  • the hydrophobic nature of the trityl group permits stronger retention of the desired full-length oligo than the non-tritylated truncated failure sequences.
  • the failure sequences were selectively washed from the resin with a suitable solvent, such as low percent acetonitrile. Retained oligonucleotides were then detritylated on-column with trifluoroacetic acid to remove the acid-labile trityl group. Residual acid was washed from the column, a salt exchange was performed, and a final desalting of the material commenced.
  • the full-length oligo was recovered in a purified form with an aqueous-organic solvent. The final product was then analyzed for purity (HPLC), identity (Maldi-TOF MS), and yield (UV A 260 ). The oligos were dried via lyophilization or vacuum condensation.
  • the dried oligos were dissolved in appropriate buffers followed by mixing equal molar amounts (calculated using the theoretical extinction coefficient) of the sense and antisense oligonucleotide strands. The solution was then analyzed for purity of duplex by chromatographic methods and desired final concentration. If the analysis indicated an excess of either strand, then the additional non-excess strand was titrated until duplexing was complete. When analysis indicated that the target product purity has been achieved the material was delivered and ready for use.
  • the material is transferred to the tangential flow filtration (TFF) system for concentration and desalting, as opposed to doing this prior to the annealing step.
  • TMF tangential flow filtration
  • Ultrafiltration The annealed product solution is concentrated using a TFF system containing an appropriate molecular weight cut-off membrane. Following concentration, the product solution is desalted via diafiltration using Milli-Q water until the conductivity of the filtrate is that of water.
  • Lyophilization The concentrated solution is transferred to a bottle, flash frozen and attached to a lyophilizer. The product is then freeze-dried to a powder. The bottle is removed from the lyophilizer and is now ready for use.
  • the resulting solution was stirred for 16 h at room temperature, quenched with acetic acid (0.75 mL, 4.0 equiv), treated with MP-TMT resin (Part No: 801472, from Biotage) (9 g), aged at room temperature for 16 h, filtered, and concentrated the filtrate to a foam solid.
  • the solid was then dissolved in CH 2 Cl 2 (140 mL), and washed with AcOH/NaCl solution (140 mL).
  • the AcOH/NaCl solution was prepared with 1 mL AcOH and 100 mL 20% NaCl solution.
  • the bottom organic layer was concentrated, and purified on a SiO 2 column (220 g), eluting with CH 2 Cl 2 /MeOH.
  • HEK293 cells stably transfected with luciferase vector that contains target sites for siNA in 3′UTR of renilla luciferase were generated. These cells were seeded on 96-well tissue culture plates (Corning: #3903) at a density of 7.5e3 cells per well in DMEM 10% serum media. Cellular plates were then incubated at 37° C./5% CO2 for 24 hr. After incubation, plates were treated with test compounds co-transfected with transfection reagent Lipofectamine 2000 (invitrogen: #11668-019) in Opti-MEM (Gibco: #31985) in accordance to manufacturers protocol. The treatment concentrations ranged from 10 nM to 0.03 pM.
  • Treated plates were then incubated for 24 hr at 37° C./5% CO2. Following treatment incubation, cells were lysed and processed in accordance to Dual-GloTM Luciferase Assay (Promega: E2920) and read on a TECAN safire2 plate reader.
  • Dual-GloTM Luciferase Assay Promega: E2920
  • HepG2 cells (ATCC: HB-8065) were seeded on collagen coated plates (BioCoat: 356649) at a density of 7.5e3 cells per well in DMEM 10% serum media. Cellular plates were then incubated at 37° C./5% CO2 for 24 hr. After incubation, plates were treated with test compounds co-transfected with transfection reagent Lipofectamine 2000 (invitrogen: 11668-019) in Opti-MEM (Gibco: 31985) in accordance to invitrogen protocol. The treatment concentrations ranged from 10 nM to 0.03 pM. Treated plates were then incubated for 24 hr at 37° C./5% CO2.
  • Cryogenically preserved male rhesus primary hepatocyte cells were obtained from Celsis. Frozen cells were defrosted for 2 minutes in a 37° C. water bath and resuspended using thawing/plating media (Gibco: CM3000) at a ratio of 1-3 mL frozen cells per 50 mL of T/ P media , in order to dilute out DMSO. Cells were spun down at 80 ⁇ g for 5 minutes to pellet cells. Pelleted cell were then diluted in T/ P media for seeding of cells to center 60 wells of a 96 well collagen coated plate (Biocoat: 354649) at 4.0e4 cells per well. Seeded cells were incubated for 18 hr at 37° C./5% CO2.
  • CD1 female mice were dosed by subcutaneous injection in 200 ul volume. Animals were observed for behavioral or physiological changes. Animals were sacrificed 72 hrs post dose by CO2 asphyxiation followed by ex-sanguination via cardiac puncture. The liver samples were as 3 mm punches from the medial lobe and put into RNAlater tubes for isolation of total RNA. The mRNA knockdown analysis was conducted by Taqman analysis using standard procedures.
  • Test siNAs were incubated in either 100% mouse serum or 0.4 mg/mL rat lysosomal lysate at 20 ug/mL.
  • the 96-well Clarity OTX cartridge plates (Phenomenex, Torrance, Calif.) were used with a 96-well plate vacuum manifold following the manufacturer's instructions.
  • the eluants were next lyophilized over night then reconstituted in 1 mM EDTA for LC/MS analysis.
  • Data acquisition and processing for the LC/MS system wereperformed using Excalibur software.
  • a Hypersil Gold C18 reversed phase column (2.1 ⁇ 50 mm) was used. The column temperature was maintained at 70 C.
  • the mobile phases used were as follows: A, 1.7 mM triethylamine (TEA) and 100 mM 1,1,1,3,3,3-hexafluoro-2-propanol (HFIP) in HPLC water; and B, methanol.
  • the sample injection volume was 10 uL, and the flow rate was maintained at 0.4 mL/min.
  • the mass spectrometer was a linear ion trap coupled to an Orbitrap allowing high-resolution mass measurements (LTQ-Orbitrap, Thermo Fisher Scientific, San Jose, Calif.), fitted with an ESI source, and operated in negative ion mode.
  • Zero charge state siNA and metabolite masses were obtained by deconvolution of charge states using Thermo Extract software. Metabolites were identified by comparison of experimental masses to theoretical nuclease metabolite masses.
  • liver tissue was homogenized in 1 mL of Tris-EDTA buffer, pH 7.2 using a tissue homogenizer with a stainless steel ball at 4 C, for 2 min.
  • siNA and their metabolites were extracted from liver lysates using a combination liquid-liquid and solid phase extraction.
  • a 500 mL volume of 5% ammonium hydroxide solution was added to each sample mixture, vortexed for and centrifuged for 10 min at 14 000 rpm.
  • the aqueous phase was transferred to a fresh tube and an equal volume of a buffer (17.2 mM TEA and 200 mM HFIP, pH8.5) was added to the aqueous phase and the sample tubes were vortexed.
  • the sample solutions were then extracted on an Oasis HLB SPE cartridge (10 mg, Waters, Mass., USA).
  • the SPE cartridges were first conditioned with 1 mL of acetonitrile followed by two aliquots of 1 mL of buffer (8.6 mM TEA and 100 mM HFIP, pH 8.5).
  • the sample solutions were then loaded onto the SPE cartridges.
  • the cartridges were washed with two aliquots of 0.5 mL buffer (8.6 mM TEA and 100 mM HFIP, pH8.5) followed by 0.5 mL of 100 mM triethylammonium bicarbonate (TEAB).
  • TEAB triethylammonium bicarbonate
  • Mobile phase ‘A’ consisted of 1.7 mMTEA and 100 mMHFIP (pH 7.5) in water, and mobile phase ‘B’ consisted of methanol.
  • the effluent from the HPLC column was introduced into the ion source of the LTQ-Orbitrap mass spectrometer operated in negative mode.
  • Zero charge state siNA and metabolite masses were obtained by deconvolution of charge states using Thermo Extract software. Metabolites were identified by comparison of experimental masses to theoretical nuclease metabolite masses.
  • R H, Boc, Cbz, Ac, PEG, lipid, targeting ligand, linker(s) and/or peptide(s).

Abstract

The present invention relates to compounds, compositions, and methods for the study, diagnosis, and treatment of traits, diseases and conditions that respond to the modulation of gene expression and/or activity, and/or modulate a gene expression pathway. Specifically, the invention relates to double-stranded nucleic acid molecules including small nucleic acid molecules, such as short interfering nucleic acid (siNA) molecules that are capable of mediating or that mediate RNA interference (RNAi) against target gene expression.

Description

    FIELD OF THE INVENTION
  • The present invention relates to compounds, compositions, and methods for the study, diagnosis, and treatment of traits, diseases and conditions that may respond to the modulation of gene expression and/or activity. The present invention is also directed to compounds, compositions, and methods relating to traits, diseases and conditions that may respond to the modulation of expression and/or activity of genes involved in gene expression pathways or other cellular processes that mediate the maintenance or development of such traits, diseases and conditions. Specifically, the invention relates to chemically modified short interfering nucleic acid (siNA) molecules that may be capable of mediating RNA interference (RNAi) against gene expression, including cocktails of such siNA molecules and formulations of such siNA molecules. Such siNA molecules are useful, for example, in providing compositions that may prevent, inhibit, or reduce various diseases, traits and conditions that are associated with gene expression or activity in a subject or organism.
  • BACKGROUND OF THE INVENTION
  • The following is a discussion of relevant art pertaining to RNAi. The discussion is provided only for understanding of the invention that follows. The summary is not an admission that any of the work described below is prior art to the claimed invention.
  • RNA interference refers to the process of sequence-specific post-transcriptional gene silencing in animals mediated by short interfering RNAs (siRNAs) (Zamore et al., 2000, Cell, 101, 25-33; Bass, 2000, Cell, 101, 235; Fire et al., 1998, Nature, 391, 806; Hamilton et al., 1999, Science, 286, 950-951; Lin et al., 1999, Nature, 402, 128-129; Sharp, 1999, genes & Dev., 13:139-141; and Strauss, 1999, Science, 286, 886). The corresponding process in plants (Heifetz et al., International PCT Publication No. WO 99/61631) is commonly referred to as post-transcriptional gene silencing or RNA silencing and is also referred to as quelling in fungi. The process of post-transcriptional gene silencing is thought to be an evolutionarily-conserved cellular defense mechanism used to prevent the expression of foreign genes and is commonly shared by diverse flora and phyla (Fire et al., 1999, Trends genet., 15, 358). Such protection from foreign gene expression may have evolved in response to the production of double-stranded RNAs (dsRNAs) derived from viral infection or from the random integration of transposon elements into a host genome via a cellular response that specifically destroys homologous single-stranded RNA or viral genomic RNA.
  • The therapeutic potential of RNAi lies in the ability to modulate gene expression in a sequence specific manner by harnessing a highly conserved, robust endogenous mechanism of action. This endogenous mechanism of action vastly expands upon the number of available targets for disease modification when compared to existing small molecule and biologic modalities. Nevertheless, a opposed to exogenously supplied small molecule and biologic modalities, the RNA molecules that serve as triggers for RNAi are not well suited for administration due to their inherent instability, especially in biologic systems. This problem has been addressed through innovation, both in terms of chemical modification of RNA triggers (see U.S. Ser. No. 10/444,853, published as US .S Patent Appl. Publ. No. 20040192626) and various delivery approaches (see U.S. Ser. No. 11/586,102, published as US US-.S. Patent Appl. Publ. No. 20080020058)), which have provided compounds and compositions that may be available for clinical development. Nevertheless there remains a need for additional RNA triggers that are available to expand the repertoire of available compounds and compositions for use in RNAi based therapeutics, and especially compounds and compositions that are compatible with different delivery systems and/or routes of administration.
  • SUMMARY OF THE INVENTION
  • The invention provides a solution to the problem of having a sufficient repertoire of available compounds and compositions for use in RNAi based therapeutics that may be compatible with different delivery modalities and/or routes of administration by providing additional forms of chemically modified short interfering nucleic acid (siNA) molecules.
  • The present invention provides compounds, compositions, and methods useful for modulating the expression of target genes and for treating diseases and conditions that may respond to such modulation by RNA interference (RNAi). Specially, the present invention provides certain chemically modified short interfering nucleic acid (siNA) molecules having any of Formula A, B, C, D, E, F, G, H, I, J, K, L, M, N, O, or P or as otherwise provided in the description and claims herein. These siNA molecules may be used, inter alia, as RNAi based therapeutic compounds and compositions.
  • These and other aspects of the invention will be apparent upon reference to the following detailed description and attached figures. Moreover, it is contemplated that any method or composition described herein can be implemented with respect to any other method or composition described herein and that different embodiments may be combined.
  • Additionally, patents, patent applications, and other documents are cited throughout the specification to describe and more specifically set forth various aspects of this invention. Each of these references cited herein is hereby incorporated by reference in its entirety, including the drawings.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 shows a non-limiting proposed mechanistic representation of target RNA degradation involved in RNAi. Double-stranded RNA (dsRNA), which is generated by RNA-dependent RNA polymerase (RdRP) from foreign single-stranded RNA, for example viral, transposon, or other exogenous RNA, activates the DICER enzyme that in turn generates siNA duplexes. Alternately, synthetic or expressed siNA can be introduced directly into a cell by appropriate means. An active siNA complex forms that recognizes a target RNA, resulting in degradation of the target RNA by the RISC endonuclease complex or in the synthesis of additional RNA by RNA-dependent RNA polymerase (RdRP), which can activate DICER and result in additional siNA molecules, thereby amplifying the RNAi response.
  • FIG. 2 shows non-limiting examples of different stabilization chemistries (1-10) that can be used, for example, to stabilize the 5′ and/or 3′-ends of siNA sequences of the invention (e.g., B or a portion thereof with respect to a siNA molecule having any of Formula A, B, C, D, E, F, G, H, I, J, K, L, M, N, O, or P); including (1) [3-3′]-inverted deoxyribose; (2) deoxyribonucleotide; (3) [5′-3′]-3′-deoxyribonucleotide; (4) [5′-3′]-ribonucleotide; (5) [5′-3′]-3′-O-methyl ribonucleotide; (6) 3′-glyceryl; (7) [3′-5′]-3′-deoxyribonucleotide; (8) [3′-3′]-deoxyribonucleotide; (9) [5′-2′]-deoxyribonucleotide; and (10) [5-3′]-dideoxyribonucleotide (when X═O). In addition to modified and unmodified backbone chemistries indicated in the figure, these chemistries can be combined with different sugar and base nucleotide modifications as described herein.
  • FIG. 3 shows non-limiting examples of phosphorylated siNA molecules of the invention, including linear and duplex constructs and asymmetric derivatives thereof.
  • FIG. 4 shows non-limiting examples of chemically modified terminal phosphate groups of the invention. These terminal phosphate groups can be included at the 5′-end of the guide strand of any siNA molecule of the invention (e.g., any of Formula A, B, C, D, E, F, G, H, I, J, K, L, M, N, O, or P).
  • FIG. 5 shows representative data indicating that 2′-deoxy-2′-fluoro content on the guide strand is important for effective knockdown of ApoB mRNA in vivo. A PVE polymer conjugate delivery vehicle was used to administer the siNAs. mRNA expression in liver tissue was measured at 2 days, 7 days and 14 days post dosing with 3mpk of siNA. Details on modification motifs and siNA sequences are provided in Table 1.
  • FIG. 6 shows representative data indicating that tetraGalNAc (single chemical entity, SCE) siNA conjugates containing alternating 2′-deoxy-2′-fluoro and 2′-O-methyl modification patterns on the guide strands have little impact on ApoB or SSB mRNA expression in vivo. siNA conjugates were administered subcutaneously (30mpk for ApoB and 50mpk for SSB) and mRNA expression in liver tissue was measured 72 hours after dosing. Details on modification motifs and siNA sequences are provided in Table 1.
  • FIG. 7 shows representative data indicating the in vivo activity of subcutaneously delivered tetraGalNAc SCE siNA conjugates in mouse liver 72 hr post dosing. The SSB sequences lacking phosphorothioate linkages at the 5′ end of the guide strand showed little to no activity while the ApoB sequence with 5′-terminal guide strand phosphorothioate linkages showed dose dependent knockdown. Details on modification motifs and siNA sequences are provided in Table 1.
  • FIG. 8 shows representative data indicating the in vivo activity of subcutaneously delivered tetraGalNAc SCE siNA conjugates in mouse liver 24 hr post dosing. The SSB sequences containing phosphorothioate linkages at the 5′ end of the guide strand (pos 1-3) showed dose dependent knockdown. Details on modification motifs and siNA sequences are provided in Table 1.
  • FIG. 9 shows representative data indicating the in vivo activity of subcutaneously delivered tetraGalNAc SCE siNA conjugates in mouse liver 72 hr post dosing. Addition of greater than 3-fold more phosphorothioate content does not appear to improve the activity of the siNA conjugates. Note that all these sequence had three phosphorothioate linkages at 5′ end of the guide strand. Details on modification motifs and siNA sequences are provided in Table 1.
  • FIG. 10 shows representative data indicating the in vivo activity of subcutaneously delivered tetraGalNAc SCE siNA conjugates in mouse liver 72 hr post dosing. The presence of a single phosphorothioate linkage at the first position at the 5′ end of the guide strand is important for in vivo activity. Sequences with phosphorothioate linkages at other positions in the siNA but not at the first position of the 5′ end of the guide strand showed poor activity. Details on modification motifs and siNA sequences are provided in Table 1.
  • FIG. 11 shows representative data indicating that addition of three phosphorothioate modifications to the 5′ end of the guide strand (positions 1-3) significantly improves the knockdown of CTNNB1 mRNA in mice. A 50mpk dose of siNA was administered as a tetraGalNAc SCE conjugate that was injected subcutaneously. Details on modification motifs and siNA sequences are provided in Table 1.
  • FIG. 12 shows representative data indicating the in vivo activity of subcutaneously delivered tetraGalNAc ApoC3 siNA conjugates in mouse liver 72 hr post dosing. Details on modification motifs and siNA sequences are provided in Table 1.
  • FIG. 13 shows representative data indicating the in vivo activity of subcutaneously delivered tetraGalNAc CTNNB1 siNA conjugates in rhesus liver at indicated times. Details on modification motifs and siNA sequences are provided in Table 1.
  • DETAILED DESCRIPTION OF THE INVENTION Terms and Definitions
  • The following terminology and definitions apply as used in the present application.
  • As used in this specification and the appended claims, the singular forms “a,” “an,” and “the” include plural referents unless the content clearly dictates otherwise. Thus, for example, reference to “a cell” includes a combination of two or more cells, and the like.
  • Any concentration range, percentage range, ratio range or integer range is to be understood to include the value of any integer within the recited range, and when appropriate, fractions thereof (such as on tenth and one hundredth of an integer), unless otherwise indicated.
  • “About” or “approximately,” as used herein, in reference to a number are generally taken to include numbers that fall within a range of 5% in either direction (greater than or less than) of the number unless otherwise stated or otherwise evident from the context (except where such number would exceed 100% of a possible value). Where ranges are stated, the endpoints are included within the range unless otherwise stated or otherwise evident from the context.
  • The term “abasic” as used herein refers to its meaning as is generally accepted in the art. The term generally refers to sugar moieties lacking a nucleobase or having a hydrogen atom (H) or other non-nucleobase chemical groups in place of a nucleobase at the 1′ position of the sugar moiety, see for example Adamic et al., U.S. Pat. No. 5,998,203. In one embodiment, an abasic moiety of the invention is a ribose, deoxyribose, or dideoxyribose sugar.
  • The term “acyclic nucleotide” as used herein refers to its meaning as is generally accepted in the art. The term generally refers to any nucleotide having an acyclic ribose sugar, for example where any of the ribose carbon/carbon or carbon/oxygen bonds are independently or in combination absent from the nucleotide.
  • The term “alkyl” as used herein refers to its meaning as is generally accepted in the art. The term generally refers to a saturated or unsaturated hydrocarbon, including straight-chain, branched-chain, alkenyl, alkynyl groups and cyclic groups, but excludes aromatic groups. Notwithstanding the foregoing, alkyl also refers to non-aromatic heterocyclic groups. Preferably, the alkyl group has 1 to 12 carbons. More preferably, it is a lower alkyl of from 1 to 7 carbons, more preferably 1 to 4 carbons. The alkyl group can be substituted or unsubstituted. When substituted, the substituted group(s) is preferably, hydroxyl, halogen, cyano, C1-C4 alkoxy, ═O, ═S, NO2, SH, NH2, or NR1R2, where R1 and R2 independently are H or C1-C4 alkyl.
  • The phrase “agents that interfere with cell cycle checkpoints” refers to compounds that inhibit protein kinases that transduce cell cycle checkpoint signals, thereby sensitizing the cancer cell to DNA damaging agents.
  • The phrase “agents that interfere with receptor tyrosine kinases (RTKs)” refers to compounds that inhibit RTKs and therefore inhibit mechanisms involved in oncogenesis and tumor progression.
  • The phrase “androgen receptor modulators” refers to compounds that interfere or inhibit the binding of androgens to the receptor, regardless of mechanism.
  • The phrase “angiogenesis inhibitors” refers to compounds that inhibit the formation of new blood vessels, regardless of mechanism.
  • The term “aryl” as used herein refers to its meaning as is generally accepted in the art. The term generally refers to an aromatic group that has at least one ring having a conjugated pi electron system and includes carbocyclic aryl, heterocyclic aryl and biaryl groups, all of which can be optionally substituted. The preferred substituent(s) of aryl groups are halogen, trihalomethyl, hydroxyl, SH, OH, cyano, C1-C4 alkoxy, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, NH2, and NR1R2 groups, where R1 and R2 independently are H or C1-C4 alkyl.
  • The term “alkylaryl” as used herein refers to its meaning as is generally accepted in the art. The term generally refers to an alkyl group (as described above) covalently joined to an aryl group (as described above). Carbocyclic aryl groups are groups wherein the ring atoms on the aromatic ring are all carbon atoms. The carbon atoms are optionally substituted. Heterocyclic aryl groups are groups having from 1 to 3 heteroatoms as ring atoms in the aromatic ring and the remainder of the ring atoms are carbon atoms. Suitable heteroatoms include oxygen, sulfur, and nitrogen, and examples of heterocyclic aryl groups having such heteroatoms include furanyl, thienyl, pyridyl, pyrrolyl, N-lower alkyl pyrrolo, pyrimidyl, pyrazinyl, imidazolyl and the like, all optionally substituted. Preferably, the alkyl group is a C1-C4 alkyl group.
  • The term “amide” as used herein refers to its meaning as is generally accepted in the art. The term generally refers to an —C(O)—NH—R, where R is either alkyl, aryl, alkylaryl or hydrogen.
  • The phrase “antisense region” as used herein refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term refers to a nucleotide sequence of an siNA molecule having complementarity to a target nucleic acid sequence. In addition, the antisense region of an siNA molecule can optionally comprise a nucleic acid sequence having complementarity to a sense region of the siNA molecule. In one embodiment, the antisense region of the siNA molecule is referred to as the antisense strand or guide strand.
  • The phrase “asymmetric hairpin” refers to a linear siNA molecule comprising an antisense region, a loop portion that can comprise nucleotides or non-nucleotides, and a sense region that comprises fewer nucleotides than the antisense region to the extent that the sense region has enough complementary nucleotides to base pair with the antisense region and form a duplex with loop. For example, an asymmetric hairpin siNA molecule of the invention can comprise an antisense region having length sufficient to mediate RNAi in a cell or in vitro system (e.g. about 15 to about 30, or about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides) and a loop region comprising about 4 to about 12 (e.g., about 4, 5, 6, 7, 8, 9, 10, 11, or 12) nucleotides, and a sense region having about 3 to about 25 (e.g., about 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25) nucleotides that are complementary to the antisense region. The asymmetric hairpin siNA molecule can also comprise a 5′-terminal phosphate group that can be chemically modified. The loop portion of the asymmetric hairpin siNA molecule can comprise nucleotides, non-nucleotides, linker molecules, or conjugate molecules as described herein.
  • The term “biodegradable” as used herein refers to its meaning as is generally accepted in the art. The term generally refers to degradation in a biological system, for example, enzymatic degradation or chemical degradation.
  • The term “biodegradable linker” as used herein refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term refers to a linker molecule that is designed to connect one molecule to another molecule, and which is susceptible to degradation in a biological system. The linker can be a nucleic acid or non-nucleic acid based linker. For example, a biodegradable linker can be used to attach a ligand or biologically active molecule to an siNA molecule of the invention. Alternately, a biodegradable linker can be used to connect the sense and antisense strands of an siNA molecule of the invention. The biodegradable linker is designed such that its stability can be modulated for a particular purpose, such as delivery to a particular tissue or cell type. The stability of a nucleic acid-based biodegradable linker molecule can be modulated by using various chemistries, for example combinations of ribonucleotides, deoxyribonucleotides, and chemically modified nucleotides, such as 2′-O-methyl, 2′-fluoro, 2′-amino, 2′-O-amino, 2′-C-allyl, 2′-O-allyl, and other 2′-modified or base modified nucleotides. The biodegradable nucleic acid linker molecule can be a dimer, trimer, tetramer or longer nucleic acid molecule, for example, an oligonucleotide of about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 nucleotides in length, or can comprise a single nucleotide with a phosphorus-based linkage, for example, a phosphoramidate or phosphodiester linkage. The biodegradable nucleic acid linker molecule can also comprise nucleic acid backbone, nucleic acid sugar, or nucleic acid base modifications.
  • The phrase “biologically active molecule” as used herein refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term refers to compounds or molecules that are capable of eliciting or modifying a biological response in a system and/or are capable of modulating the pharmacokinetics and/or pharmacodynamics of other biologically active molecules. Examples of biologically active molecules, include siNA molecules alone or in combination with other molecules including, but not limited to therapeutically active molecules such as antibodies, cholesterol, hormones, antivirals, peptides, proteins, chemotherapeutics, small molecules, vitamins, co-factors, nucleosides, nucleotides, oligonucleotides, enzymatic nucleic acids, antisense nucleic acids, triplex forming oligonucleotides, polyamines, polyamides, polyethylene glycol, other polyethers, 2-5A chimeras, siNA, dsRNA, allozymes, aptamers, decoys and analogs thereof.
  • The phrase “biological system” as used herein refers to its meaning as is generally accepted in the art. The term generally refers to material, in a purified or unpurified form, from biological sources including, but not limited to, human or animal, wherein the system comprises the components required for RNAi activity. Thus, the phrase includes, for example, a cell, tissue, subject, or organism, or extract thereof. The term also includes reconstituted material from a biological source.
  • The phrase “blunt end” as used herein refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term refers to termini of a double-stranded siNA molecule having no overhanging nucleotides. For example, the two strands of a double-stranded siNA molecule having blunt ends align with each other with matched base-pairs without overhanging nucleotides at the termini. A siNA duplex molecule of the invention can comprise blunt ends at one or both termini of the duplex, such as termini located at the 5′-end of the antisense strand, the 5′-end of the sense strand, or both termini of the duplex.
  • The term “cap” also referred to herein as “terminal cap,” as used herein refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term refers to a moiety, which can be a chemically modified nucleotide or non-nucleotide that can be incorporated at one or more termini of one or more nucleic acid molecules of the invention. These terminal modifications protect the nucleic acid molecule from exonuclease degradation, and can help in delivery and/or localization within a cell. The cap can be present at the 5′-terminus (5′-cap) or at the 3′-terminal (3′-cap) or can be present on both termini of any nucleic acid molecule of the invention. A cap can be present at the 5′-end, 3-end and/or 5′ and 3′-ends of the sense strand of a nucleic acid molecule of the invention. Additionally, a cap can optionally be present at the 3′-end of the antisense strand of a nucleic acid molecule of the invention. In non-limiting examples, the 5′-cap includes, but is not limited to a polymer; a ligand; locked nucleic acid (LNA); glyceryl; an abasic ribose residue (moiety); inverted deoxy abasic residue (moiety); an inverted nucleotide; 4′,5′-methylene nucleotide; 1-(beta-D-erythrofuranosyl) nucleotide, 4′-thio nucleotide; carbocyclic nucleotide; 1,5-anhydrohexitol nucleotide; L-nucleotides; alpha-nucleotides; modified base nucleotide; phosphorodithioate linkage; threo-pentofuranosyl nucleotide; acyclic 3′,4′-seco nucleotide; acyclic 3,4-dihydroxybutyl nucleotide; acyclic 3,5-dihydroxypentyl nucleotide; 3′-3′-inverted nucleotide moiety; 3′-3′-inverted abasic moiety; 3′-2′-inverted nucleotide moiety; 3′-2′-inverted abasic moiety; 1,4-butanediol phosphate; 3′-phosphoramidate; hexylphosphate; aminohexyl phosphate; 3′-phosphate; 3′-phosphorothioate; phosphorodithioate; or bridging or non-bridging methylphosphonate moiety. Non-limiting examples of the 3′-cap include, but are not limited to, a polymer; a ligand; locked nucleic acid (LNA); glyceryl; an abasic ribose residue (moiety); inverted deoxy abasic residue (moiety); an inverted nucleotide; 4′,5′-methylene nucleotide; 1-(beta-D-erythrofuranosyl) nucleotide; 4′-thio nucleotide; carbocyclic nucleotide; 5′-amino-alkyl phosphate; 1,3-diamino-2-propyl phosphate; 3-aminopropyl phosphate; 6-aminohexyl phosphate; 1,2-aminododecyl phosphate; hydroxypropyl phosphate; 1,5-anhydrohexitol nucleotide; L-nucleotide; alpha-nucleotide; modified base nucleotide; phosphorodithioate; threo-pentofuranosyl nucleotide; acyclic 3′,4′-seco nucleotide; 3,4-dihydroxybutyl nucleotide; 3,5-dihydroxypentyl nucleotide, 5′-5′-inverted nucleotide moiety; 5′-5′-inverted abasic moiety; 5′-phosphoramidate; 5′-phosphorothioate; 1,4-butanediol phosphate; 5′-amino; bridging and/or non-bridging 5′-phosphoramidate; phosphorothioate and/or phosphorodithioate; bridging or non bridging methylphosphonate; and 5′-mercapto moieties (for more details see Beaucage and Iyer, 1993, Tetrahedron 49, 1925; incorporated by reference herein). In certain embodiments, a siNA molecule of the invention having Formula (A) can comprise one or more terminal cap molecules as described above (designated as B) that comprises or includes a covalent attachment to a polymer or ligand via a linker molecule as described herein or as is otherwise known in the art. Non-limiting examples of such linkers are provided in the examples and descriptions herein.
  • The term “cell” as used herein refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term is used in its usual biological sense, and does not refer to an entire multicellular organism, e.g., specifically does not refer to a human being. The cell can be present in an organism, e.g., birds, plants and mammals, such as humans, cows, sheep, apes, monkeys, swine, dogs, and cats. The cell can be prokaryotic (e.g., bacterial cell) or eukaryotic (e.g., mammalian or plant cell). The cell can be of somatic or germ line origin, totipotent or pluripotent, dividing or non-dividing. The cell can also be derived from or can comprise a gamete or embryo, a stem cell, or a fully differentiated cell.
  • The phrase “chemical modification” as used herein refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term refers to any modification of the chemical structure of the nucleotides that differs from nucleotides of native siRNA or RNA in general. The term “chemical modification” encompasses the addition, substitution, or modification of native siRNA or RNA at the sugar, base, or internucleotide linkage, as described herein or as is otherwise known in the art. In certain embodiments, the term “chemical modification” can refer to certain forms of RNA that are naturally occurring in certain biological systems, for example 2′-O-methyl modifications or inosine modifications.
  • The term “complementarity” or “complementary” as used herein refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the terms generally refer to the formation or existence of hydrogen bond(s) between one nucleic acid sequence and another nucleic acid sequence by either traditional Watson-Crick or other non-traditional types of bonding as described herein. In reference to the nucleic molecules of the present invention, the binding free energy for a nucleic acid molecule with its complementary sequence is sufficient to allow the relevant function of the nucleic acid to proceed, e.g., RNAi activity. Determination of binding free energies for nucleic acid molecules is well known in the art (see, e.g., Turner et al., 1987, CSH Symp. Quant. Biol. LII pp. 123-133; Frier et al., 1986, Proc. Nat. Acad. Sci. USA 83:9373-9377; Turner et al., 1987, J. Am. Chem. Soc. 109:3783-3785). Perfect complementary means that all the contiguous residues of a nucleic acid sequence will hydrogen bond with the same number of contiguous residues in a second nucleic acid sequence. Partial complementarity can include various mismatches or non-based paired nucleotides (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more mismatches, non-nucleotide linkers, or non-based paired nucleotides) within the nucleic acid molecule, which can result in bulges, loops, or overhangs that result between the sense strand or sense region and the antisense strand or antisense region of the nucleic acid molecule or between the antisense strand or antisense region of the nucleic acid molecule and a corresponding target nucleic acid molecule. Such partial complementarity can be represented by a % complementarity that is determined by the number of non-base paired nucleotides, i.e., about 50%, 60%, 70%, 80%, 90% etc. depending on the total number of nucleotides involved. Such partial complementarity is permitted to the extent that the nucleic acid molecule (e.g., siNA) maintains its function, for example the ability to mediate sequence specific RNAi.
  • The terms “composition” or “formulation” as used herein refer to their generally accepted meaning in the art. These terms generally refer to a composition or formulation, such as in a pharmaceutically acceptable carrier or diluent, in a form suitable for administration, e.g., systemic or local administration, into a cell or subject, including, for example, a human. Suitable forms, in part, depend upon the use or the route of entry, for example oral, transdermal, inhalation, or by injection. Such forms should not prevent the composition or formulation from reaching a target cell (i.e., a cell to which the negatively charged nucleic acid is desirable for delivery). For example, compositions injected into the blood stream should be soluble. Other factors are known in the art, and include considerations such as toxicity and forms that prevent the composition or formulation from exerting its effect. As used herein, pharmaceutical formulations include formulations for human and veterinary use. Non-limiting examples of agents suitable for formulation with the nucleic acid molecules of the instant invention include: Lipid Nanoparticles (see for example Semple et al., 2010, Nat Biotechnol., February; 28(2):172-6.); P-glycoprotein inhibitors (such as Pluronic P85); biodegradable polymers, such as poly (DL-lactide-coglycolide) microspheres for sustained release delivery (Emerich, D F et al, 1999, Cell Transplant, 8, 47-58); and loaded nanoparticles, such as those made of polybutylcyanoacrylate. Other non-limiting examples of delivery strategies for the nucleic acid molecules of the instant invention include material described in Boado et al., 1998, J. Pharm. Sci., 87, 1308-1315; Tyler et al., 1999, FEBS Lett., 421, 280-284; Pardridge et al., 1995, PNAS USA., 92, 5592-5596; Boado, 1995, Adv. Drug Delivery Rev., 15, 73-107; Aldrian-Herrada et al., 1998, Nucleic Acids Res., 26, 4910-4916; and Tyler et al., 1999, PNAS USA., 96, 7053-7058. A “pharmaceutically acceptable composition” or “pharmaceutically acceptable formulation” can refer to a composition or formulation that allows for the effective distribution of the nucleic acid molecules of the instant invention to the physical location most suitable for their desired activity.
  • The phrase “cytotoxic/cytostatic agents” refer to compounds that cause cell death or inhibit cell proliferation primarily by interfering directly with the cell's functioning or inhibit or interfere with cell mytosis, including alkylating agents, tumor necrosis factors, intercalators, hypoxia activatable compounds, microtubule inhibitors/microtubule-stabilizing agents, inhibitors of mitotic kinesins, inhibitors of histone deacetylase, inhibitors of kinases involved in mitotic progression, antimetabolites; biological response modifiers; hormonal/anti-hormonal therapeutic agents, hematopoietic growth factors, monoclonal antibody targeted therapeutic agents, topoisomerase inhibitors, proteasome inhibitors and ubiquitin ligase inhibitors.
  • The phrase “estrogen receptor modulators” refers to compounds that interfere with or inhibit the binding of estrogen to the receptor, regardless of mechanism.
  • The term “gene” or “target gene” as used herein refers to their meaning as is generally accepted in the art. The terms generally refer a nucleic acid (e.g., DNA or RNA) sequence that comprises partial length or entire length coding sequences necessary for the production of a polypeptide. The target gene can also include the UTR or non-coding region of the nucleic acid sequence. A gene or target gene can also encode a functional RNA (fRNA) or non-coding RNA (ncRNA), such as small temporal RNA (stRNA), micro RNA (miRNA), small nuclear RNA (snRNA), short interfering RNA (siRNA), small nucleolar RNA (snRNA), ribosomal RNA (rRNA), transfer RNA (tRNA) and precursor RNAs thereof. Such non-coding RNAs can serve as target nucleic acid molecules for siNA mediated RNA interference in modulating the activity of fRNA or ncRNA involved in functional or regulatory cellular processes. Aberrant fRNA or ncRNA activity leading to disease can therefore be modulated by siNA molecules of the invention. siNA molecules targeting fRNA and ncRNA can also be used to manipulate or alter the genotype or phenotype of a subject, organism or cell, by intervening in cellular processes such as genetic imprinting, transcription, translation, or nucleic acid processing (e.g., transamination, methylation etc.). The target gene can be a gene derived from a cell, an endogenous gene, a transgene, or exogenous genes such as genes of a pathogen, for example a virus, which is present in the cell after infection thereof. The cell containing the target gene can be derived from or contained in any organism, for example a plant, animal, protozoan, virus, bacterium, or fungus. Non-limiting examples of plants include monocots, dicots, or gymnosperms. Non-limiting examples of animals include vertebrates or invertebrates. Non-limiting examples of fungi include molds or yeasts. For a review, see for example Snyder and Gerstein, 2003, Science, 300, 258-260. In certain embodiments, gene targets contemplated herein are also referred to herein generally as “target” sequences (including the target sequences listed by GenBank Accession numbers in U.S. Ser. No. 60/363,124, incorporated by reference herein).
  • The phrase “HMG-CoA reductase inhibitors” refers to inhibitors of 3-hydroxy-3-methylglutaryl-CoA reductase. The term HMG-CoA reductase inhibitor as used herein includes all pharmaceutically acceptable lactone and open-acid forms (i.e., where the lactone ring is opened to form the free acid) as well as salt and ester forms of compounds that have HMG-CoA reductase inhibitory activity, and therefore the use of such salts, esters, open-acid and lactone forms is included within the scope of this invention.
  • The phrase “highly conserved sequence region” refers to a nucleotide sequence of one or more regions in a target gene that does not vary significantly from one generation to the other or from one biological system to the other.
  • The phrase “homologous sequence” as used herein refers to its meaning as is generally accepted in the art. The term generally refers a nucleotide sequence that is shared by one or more polynucleotide sequences, such as genes, gene transcripts and/or non-coding polynucleotides. For example, a homologous sequence can be a nucleotide sequence that is shared by two or more genes encoding related but different proteins, such as different members of a gene family, different protein epitopes, different protein isoforms or completely divergent genes. A homologous sequence can be a nucleotide sequence that is shared by two or more non-coding polynucleotides, such as noncoding DNA or RNA, regulatory sequences, introns, and sites of transcriptional control or regulation. Homologous sequences can also include sequence regions shared by more than one polynucleotide sequence. The term “perfect homology” (or “perfectly homologous”) as used herein refers to complete (100%) homology or “identity” between a reference sequence and a subject nucleic acid sequence. Homology does not need to be perfect identity (100%), however, as partially homologous sequences are also contemplated by and within the scope of the instant invention (e.g., at least 95%, 94%, 93%, 92%, 91%, 90%, 89%, 88%, 87%, 86%, 85%, 84%, 83%, 82%, 81%, 80% etc.). Percent homology is the number of matching nucleotides between two sequences divided by the total length being compared, multiplied by 100.
  • The phrase “improved RNAi activity” refers to an increase in RNAi activity measured in vitro and/or in vivo, where the RNAi activity is a reflection of both the ability of the siNA to mediate RNAi and the stability of the siNAs of the invention. In this invention, the product of these activities can be increased in vitro and/or in vivo compared to an all RNA siNA or an siNA containing a plurality of ribonucleotides. In some cases, the activity or stability of the siNA molecule can be decreased (i.e., less than ten-fold), but the overall activity of the siNA molecule is enhanced in vitro and/or in vivo.
  • The term “including” (and any form thereof, such as “includes” and “include”), “comprising” (and any form thereof, such as “has” or “have”) or “containing” (and any form thereof such as “contains” or “contain”) are inclusive and open-ended and do not exclude additional, unrecited elements or method steps.
  • The terms “inhibit,” “down-regulate,” or “reduce” as used herein refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term generally refers the reduction in the expression of the gene, or level of RNA molecules or equivalent RNA molecules encoding one or more proteins or protein subunits, or activity of one or more proteins or protein subunits, below that observed in the absence of the nucleic acid molecules (e.g., siNA) of the invention. Down-regulation can also be associated with post-transcriptional silencing, such as, RNAi mediated cleavage or by alteration in DNA methylation patterns or DNA chromatin structure. Inhibition, down-regulation or reduction with an siNA molecule can be in reference to an inactive molecule, an attenuated molecule, an siNA molecule with a scrambled sequence, or an siNA molecule with mismatches or alternatively, it can be in reference to the system in the absence of the nucleic acid.
  • The phrase “inhibitors of cell proliferation and survival signaling pathway” refers to pharmaceutical agents that inhibit cell surface receptors and signal transduction cascades downstream of those surface receptors.
  • The term “integrin blockers” refers to compounds which selectively antagonize, inhibit or counteract binding of a physiological ligand to the αωβ3 integrin, to compounds which selectively antagonize, inhibit or counteract binding of a physiological ligand to the αωβ5 integrin, to compounds which antagonize, inhibit or counteract binding of a physiological ligand to both the αωβ3 integrin and the αωβ5 integrin, and to compounds which antagonize, inhibit or counteract the activity of the particular integrin(s) expressed on capillary endothelial cells. The term also refers to antagonists of the αωβ6 αωβ8 α1β1 α2β2 α5β1 α6β1 and α6β4 integrins. The term also refers to antagonists of any combination of αωβ3, αωβ5 αωβ6 αωβ8 α1β1 α2β1 α5β1 α6β1 and α6β4 integrins.
  • The terms “intermittent” or “intermittently” as used herein refers to its meaning as is generally accepted in the art. The term generally refers to periodic stopping and starting at either regular or irregular intervals.
  • The terms “internucleoside linkage” or “internucleoside linker” or “internucleotide linkage” or “internucleotide linker” are used herein interchangeably and refer to any linker or linkage between two nucleoside units, as is known in the art, including, for example, but not limitation, phosphate, analogs of phosphate, phosphonate, guanidinium, hydroxylamine, hydroxylhydrazinyl, amide, carbamate, alkyl, and substituted alkyl linkages. The internucleoside linkages constitute the backbone of a nucleic acid molecule.
  • The term “ligand” refers to such compounds and compositions as are generally known in the art. Non-limiting examples of such ligands are described herein including in the documents specifically incorporated by reference herein. A siNA molecule of the invention can be formulated or administered with any covalently linked ligand as described herein or otherwise known in the art.
  • The term “linker” as used herein refers to its meaning as is generally known in the art. Non-limiting examples of linkers are described herein, for example in Table 8 and including in the documents specifically incorporated by reference herein.
  • The term “lipid nanoparticle” or “LNP” refers to lipid-based compositions and formulations as are generally known in the art. Non-limiting examples of such LNPs are described herein including in the documents specifically incorporated by reference herein. A siNA molecule of the invention can be formulated or administered with any LNP as described herein or otherwise known in the art.
  • The terms “mammalian” or “mammal” as used herein refers to its meaning as is generally accepted in the art. The term generally refers to any warm blooded vertebrate species, such as a human, mouse, rat, dog, cat, hamster, guinea pig, rabbit, livestock, and the like.
  • The phrase “metered dose inhaler” or MDI refers to a unit comprising a can, a secured cap covering the can and a formulation metering valve situated in the cap. MDI systems includes a suitable channeling device. Suitable channeling devices comprise for example, a valve actuator and a cylindrical or cone-like passage through which medicament can be delivered from the filled canister via the metering valve to the nose or mouth of a patient such as a mouthpiece actuator.
  • The term “microRNA” or “miRNA” as used herein refers to its meaning as is generally accepted in the art. The term generally refers to an endogenous short RNA molecule found in eukaryotes that is involved in RNA-based gene regulation. A representative set of known endogenous miRNA species is described in the publicly available miRBase sequence database as described in Griffith-Jones et al., Nucleic Acids Research, 2004, 32:D109-D111 and Griffith-Jones et al., Nucleic Acids Research, 2006, 34:D 140-D144, accessible on the World Wide Web at the Wellcome Trust Sanger Institute website. Each mature miRNA is partially complementary to one or more messenger RNA (mRNA) molecules, which are also called “miRNA targets,” thereby regulating the expression of genes associated with the miRNA targets.
  • The term “modulate” as used herein refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term refers to when the expression of a gene, or level of one or more RNA molecules (coding or non-coding), or activity of one or more RNA molecules or proteins or protein subunits, is up-regulated or down-regulated, such that expression, level, or activity is greater than or less than that observed in the absence of the molecule that effects modulation. For example, the term “modulate” in some embodiments can refer to inhibition and in other embodiments can refer to potentiation or up-regulation, e.g., of gene expression.
  • The phrase “modified nucleotide” as used herein refers to its meaning as is generally accepted in the art. The term generally refers a nucleotide, which contains a modification in the chemical structure of the base, sugar and/or phosphate of the unmodified (or natural) nucleotide as is generally known in the art. Non-limiting examples of modified nucleotides are described herein and in U.S. application Ser. No. 12/064,014.
  • The phrase “NSAIDs that are selective COX-2 inhibitors” for purposes herein, refers to NSAIDs, which possess a specificity for inhibiting COX-2 over COX-1 of at least 100 fold as measured by the ratio of IC50 for COX-2 over IC50 for COX-1 evaluated by cell or microsomal assays.
  • The phrase “non-base paired” refers to nucleotides that are not base paired between the sense strand or sense region and the antisense strand or antisense region of an double-stranded siNA molecule; and can include for example, but not limitation, mismatches, overhangs, single stranded loops, etc.
  • The term “non-nucleotide” refers to any group or compound which can be incorporated into a nucleic acid chain in the place of one or more nucleotide units, such as for example but not limitation abasic moieties or alkyl chains. The group or compound is “abasic” in that it does not contain a commonly recognized nucleotide base, such as adenosine, guanine, cytosine, uracil or thymine and therefore lacks a nucleobase at the 1′-position.
  • The term “nucleotide” is used as is generally recognized in the art. Nucleotides generally comprise a nucleobase, a sugar, and an internucleoside linkage, e.g., a phosphate. The base can be a natural bases (standard), modified bases, or a base analog, as are well known in the art. Such bases are generally located at the 1′ position of a nucleotide sugar moiety. Additionally, the nucleotides can be unmodified or modified at the sugar, internucleoside linkage, and/or base moiety, (also referred to interchangeably as nucleotide analogs, modified nucleotides, non-natural nucleotides, non-standard nucleotides and others; see, for example, U.S. application Ser. No. 12/064,014.
  • The term “overhang” as used herein refers to its meaning as is generally accepted in the art. With reference to exemplary double stranded nucleic acid molecules, the term generally refers to the terminal portion of a nucleotide sequence that is not base paired between the two strands of a double-stranded nucleic acid molecule. Overhangs, when present, are typically at the 3′-end of one or both strands in a siNA duplex.
  • The term “parenteral” as used herein refers to its meaning as is generally accepted in the art. The term generally refers methods or techniques of administering a molecule, drug, agent, or compound in a manner other than through the digestive tract, and includes epicutaneous, subcutaneous, intravascular (e.g., intravenous), intramuscular, or intrathecal injection or infusion techniques and the like.
  • The phrase “pathway target” refers to any target involved in pathways of gene expression or activity. For example, any given target can have related pathway targets that can include upstream, downstream, or modifier genes in a biologic pathway. These pathway target genes can provide additive or synergistic effects in the treatment of diseases, conditions, and traits herein.
  • The term “phosphorothioate” refers to an internucleotide phosphate linkage comprising one or more sulfur atoms in place of an oxygen atom. Hence, the term phosphorothioate refers to both phosphorothioate and phosphorodithioate internucleotide linkages.
  • The term “polymer” refers to polymeric compounds, compositions and formulations as are generally known in the art. Non-limiting examples of such polymers, including polymeric delivery systems are described herein including in the documents specifically incorporated by reference herein. A siNA molecule of the invention can be formulated or administered with any polymer as described herein or otherwise known in the art.
  • The term “position 1” refers to the position of the first nucleotide at the end of a strand, e.g., antisense strand. All positions referred to herein are the positions of a nucleotide counting from the end of a strand, for example, positions 1-3 from the 5′ end of the antisense strand, refer to the three nucleotides at positions 1, 2, and 3 counting from the 5′ end of the antisense strand.
  • The term “ribonucleotide” as used herein refers to its meaning as is generally accepted in the art. The term generally refers to a nucleotide with a hydroxyl group at the 2′ position of a (3-D-ribofuranose moiety.
  • The term “RNA” as used herein refers to its generally accepted meaning in the art. Generally, the term RNA refers to a molecule comprising at least one ribofuranoside moiety. The term can include double-stranded RNA, single-stranded RNA, isolated RNA such as partially purified RNA, essentially pure RNA, synthetic RNA, recombinantly produced RNA, as well as altered RNA that differs from naturally occurring RNA by the addition, deletion, substitution and/or alteration of one or more nucleotides. Such alterations can include addition of non-nucleotide material, such as to the end(s) of the siNA or internally, for example at one or more nucleotides of the RNA. Nucleotides in the RNA molecules of the instant invention can also comprise non-standard nucleotides, such as non-naturally occurring nucleotides or chemically synthesized nucleotides or deoxynucleotides. These altered RNAs can be referred to as analogs or analogs of naturally-occurring RNA.
  • The phrase “RNA interference” or term “RNAi” refer to the biological process of inhibiting or down regulating gene expression in a cell, as is generally known in the art, and which is mediated by short interfering nucleic acid molecules, see for example Zamore and Haley, 2005, Science, 309, 1519-1524; Vaughn and Martienssen, 2005, Science, 309, 1525-1526; Zamore et al., 2000, Cell, 101, 25-33; Bass, 2001, Nature, 411, 428-429; Elbashir et al., 2001, Nature, 411, 494-498; and Kreutzer et al., International PCT Publication No. WO 00/44895; Zernicka-Goetz et al., International PCT Publication No. WO 01/36646; Fire, International PCT Publication No. WO 99/32619; Plaetinck et al., International PCT Publication No. WO 00/01846; Mello and Fire, International PCT Publication No. WO 01/29058; Deschamps-Depaillette, International PCT Publication No. WO 99/07409; and Li et al., International PCT Publication No. WO 00/44914; Allshire, 2002, Science, 297, 1818-1819; Volpe et al., 2002, Science, 297, 1833-1837; Jenuwein, 2002, Science, 297, 2215-2218; and Hall et al., 2002, Science, 297, 2232-2237; Hutvagner and Zamore, 2002, Science, 297, 2056-60; McManus et al., 2002, RNA, 8, 842-850; Reinhart et al., 2002, Gene & Dev., 16, 1616-1626; and Reinhart & Bartel, 2002, Science, 297, 1831). Additionally, the term RNAi is meant to be equivalent to other terms used to describe sequence specific RNA interference, such as post transcriptional gene silencing, translational inhibition, transcriptional inhibition, or epigenetics. For example, siNA molecules of the invention can be used to epigenetically silence genes at either the post-transcriptional level or the pre-transcriptional level. In a non-limiting example, epigenetic modulation of gene expression by siNA molecules of the invention can result from siNA mediated modification of chromatin structure or methylation patterns to alter gene expression (see, for example, Verdel et al., 2004, Science, 303, 672-676; Pal-Bhadra et al., 2004, Science, 303, 669-672; Allshire, 2002, Science, 297, 1818-1819; Volpe et al., 2002, Science, 297, 1833-1837; Jenuwein, 2002, Science, 297, 2215-2218; and Hall et al., 2002, Science, 297, 2232-2237). In another non-limiting example, modulation of gene expression by siNA molecules of the invention can result from siNA mediated cleavage of RNA (either coding or non-coding RNA) via RISC, or via translational inhibition, as is known in the art or modulation can result from transcriptional inhibition (see for example Janowski et al., 2005, Nature Chemical Biology, 1, 216-222).
  • The phrase “RNAi inhibitor” refers to any molecule that can down regulate, reduce or inhibit RNA interference function or activity in a cell or organism. An RNAi inhibitor can down regulate, reduce or inhibit RNAi (e.g., RNAi mediated cleavage of a target polynucleotide, translational inhibition, or transcriptional silencing) by interaction with or interfering with the function of any component of the RNAi pathway, including protein components such as RISC, or nucleic acid components such as miRNAs or siRNAs. A RNAi inhibitor can be an siNA molecule, an antisense molecule, an aptamer, or a small molecule that interacts with or interferes with the function of RISC, a miRNA, or an siRNA or any other component of the RNAi pathway in a cell or organism. By inhibiting RNAi (e.g., RNAi mediated cleavage of a target polynucleotide, translational inhibition, or transcriptional silencing), a RNAi inhibitor of the invention can be used to modulate (e.g., up-regulate or down regulate) the expression of a target gene.
  • The phrase “sense region” as used herein refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term refers to a nucleotide sequence of an siNA molecule having complementarity to an antisense region of the siNA molecule. In addition, the sense region of an siNA molecule can comprise a nucleic acid sequence having homology or sequence identity with a target nucleic acid sequence. In one embodiment, the sense region of the siNA molecule is also referred to as the sense strand or passenger strand.
  • The phrases “short interfering nucleic acid”, “siNA”, “short interfering RNA”, “siRNA”, “short interfering nucleic acid molecule”, “short interfering oligonucleotide molecule”, or “chemically modified short interfering nucleic acid molecule” refer to any nucleic acid molecule capable of inhibiting or down regulating gene expression or viral replication by mediating RNA interference (“RNAi”) or gene silencing in a sequence-specific manner. These terms can refer to both individual nucleic acid molecules, a plurality of such nucleic acid molecules, or pools of such nucleic acid molecules. The siNA can be a double-stranded nucleic acid molecule comprising self-complementary sense and antisense strands, wherein the antisense strand comprises a nucleotide sequence that is complementary to a nucleotide sequence in a target nucleic acid molecule or a portion thereof and the sense strand comprises a nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof. The siNA can be a polynucleotide with a duplex, asymmetric duplex, hairpin or asymmetric hairpin secondary structure, having self-complementary sense and antisense regions, wherein the antisense region comprises a nucleotide sequence that is complementary to a nucleotide sequence in a separate target nucleic acid molecule or a portion thereof and the sense region comprises a nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof. The siNA can be a circular single-stranded polynucleotide having two or more loop structures and a stem comprising self-complementary sense and antisense regions, wherein the antisense region comprises nucleotide sequence that is complementary to a nucleotide sequence in a target nucleic acid molecule or a portion thereof and the sense region comprises a nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof, and wherein the circular polynucleotide can be processed either in vivo or in vitro to generate an active siNA molecule capable of mediating RNAi. The siNA can also comprise a single-stranded polynucleotide having a nucleotide sequence complementary to nucleotide sequence in a target nucleic acid molecule or a portion thereof (for example, where such siNA molecule does not require the presence within the siNA molecule of a nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof), wherein the single-stranded polynucleotide can further comprise a terminal phosphate group, such as a 5′-phosphate (see for example, Martinez et al., 2002, Cell, 110, 563-574 and Schwarz et al., 2002, Molecular Cell, 10, 537-568), or 5′,3′-diphosphate.
  • The term “subject” as used herein refers to its meaning as is generally accepted in the art. The term generally refers an organism to which the nucleic acid molecules of the invention can be administered. A subject can be a mammal or mammalian cells, including a human or human cells. The term also refers to an organism, which is a donor or recipient of explanted cells or the cells themselves.
  • The phrase “systemic administration” as used herein refers to its meaning as is generally accepted in the art. The term generally refers in vivo systemic absorption or accumulation of drugs in the blood stream followed by distribution throughout the entire body.
  • The term “target” as used herein refers, to any protein, peptide, or polypeptide, such as encoded by any gene in the GenBank database, including those described herein and/or in U.S. Provisional Patent Application No. 60/363,124, U.S. application Ser. No. 10/923,536 and/or PCT/US03/05028, all of which are incorporated herein by reference for purposes of identifying such targets. The term “target” also refers to one or more genes, nucleic acid sequences, or target polynucleotide sequences encoding any target protein, peptide, or polypeptide, such as proteins, peptides, or polypeptides encoded by the genes in the Genebank database or sequences having GenBank Accession Nos. shown herein and/or in U.S. Provisional Patent Application No. 60/363,124, U.S. application Ser. No. 10/923,536 and/or PCT/US03/05028, all of which are incorporated herein by reference for purposes of identify such targets. The target of interest can include target polynucleotide sequences, such as target DNA or target RNA. The term “target” is also meant to include other sequences, such as differing isoforms, mutant target genes, splice variants of target polynucleotides, target polymorphisms, and non-coding (e.g., ncRNA, miRNA, stRNA) or other regulatory polynucleotide sequences as described herein. Therefore, in various embodiments of the invention, a double stranded nucleic acid molecule of the invention (e.g., siNA) having complementarity to a target RNA can be used to inhibit or down regulate miRNA or other ncRNA activity. In one embodiment, inhibition of miRNA or ncRNA activity can be used to down regulate or inhibit gene expression (e.g., gene targets described herein or otherwise known in the art) that is dependent on miRNA or ncRNA activity. In another embodiment, inhibition of miRNA or ncRNA activity by double stranded nucleic acid molecules of the invention (e.g. siNA) having complementarity to the miRNA or ncRNA can be used to up regulate or promote target gene expression (e.g., gene targets described herein or otherwise known in the art) where the expression of such genes is down regulated, suppressed, or silenced by the miRNA or ncRNA. Such up-regulation of gene expression can be used to treat diseases and conditions associated with a loss of function or haploinsufficiency as are generally known in the art.
  • The phrase “target site” as used herein refers to its meaning as is generally accepted in the art. The term generally refers to a sequence within a target nucleic acid molecule, (e.g., RNA) that is “targeted”, e.g., for cleavage mediated by an siNA construct, which contains sequences within its antisense region that are complementary to the target sequence.
  • The phrase “therapeutically effective amount” as used herein refers to its meaning as is generally accepted in the art. The term generally refers to the amount of the compound or composition that will elicit the biological or medical response of a cell, tissue, system, animal or human that is be sought by the researcher, veterinarian, medical doctor or other clinician. For example, if a given clinical treatment is considered effective when there is at least a 25% reduction in a measurable parameter associated with a disease or disorder, a therapeutically effective amount of a drug for the treatment of that disease or disorder is that amount necessary to effect at least a 25% reduction in that parameter.
  • The phrase “universal base” as used herein refers to its meaning as is generally accepted in the art. The term universal base generally refers to nucleotide base analogs that form base pairs with each of the natural DNA/RNA bases with little or no discrimination between them. Non-limiting examples of universal bases include C-phenyl, C-naphthyl and other aromatic derivatives, inosine, azole carboxamides, and nitroazole derivatives such as 3-nitropyrrole, 4-nitroindole, 5-nitroindole, and 6-nitroindole as known in the art (see for example, Loakes, 2001, Nucleic Acids Research, 29, 2437-2447).
  • The term “up-regulate” as used herein refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term refers to an increase in the expression of a gene, or level of RNA molecules or equivalent RNA molecules encoding one or more proteins or protein subunits, or activity of one or more RNAs, proteins or protein subunits, above that observed in the absence of the nucleic acid molecules (e.g., siNA) of the invention. In certain instances, up-regulation or promotion of gene expression with an siNA molecule is above that level observed in the presence of an inactive or attenuated molecule. In other instances, up-regulation or promotion of gene expression with siNA molecules is above that level observed in the presence of, for example, an siNA molecule with scrambled sequence or with mismatches. In still other instances, up-regulation or promotion of gene expression with a nucleic acid molecule of the instant invention is greater in the presence of the nucleic acid molecule than in its absence. In some instances, up-regulation or promotion of gene expression is associated with inhibition of RNA mediated gene silencing, such as RNAi mediated cleavage or silencing of a coding or non-coding RNA target that down regulates, inhibits, or silences the expression of the gene of interest to be up-regulated. The down regulation of gene expression can, for example, be induced by a coding RNA or its encoded protein, such as through negative feedback or antagonistic effects. The down regulation of gene expression can, for example, be induced by a non-coding RNA having regulatory control over a gene of interest, for example by silencing expression of the gene via translational inhibition, chromatin structure, methylation, RISC mediated RNA cleavage, or translational inhibition. As such, inhibition or down regulation of targets that down regulate, suppress, or silence a gene of interest can be used to up-regulate expression of the gene of interest toward therapeutic use.
  • The term “vector” as used herein refers to its meaning as is generally accepted in the art. The term vector generally refers to any nucleic acid- and/or viral-based expression system or technique used to deliver one or more nucleic acid molecules.
  • siNA Molecules of the Invention
  • In one embodiment, the invention features a composition comprising a double-stranded short interfering nucleic acid (siNA) molecule that modulates the expression and/or activity of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure represented by formula (A):
  • Figure US20150299696A1-20151022-C00001
  • wherein,
    (a) the upper strand is the passenger strand and the lower strand is the guide strand of the double-stranded nucleic acid molecule; the guide strand is complementary to the target sequence and the passenger strand is complementary to the guide strand, wherein one or more mismatches between the guide strand and passenger strand and/or between the guide strand and the target sequence are tolerated so long as RNAi activity is maintained;
    (b) each N is independently a nucleotide wherein one or more Ns may be substituted with a non-nucleotide moiety so long as RNAi activity is maintained;
    (c) each B1, B2, and B3 is independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker, wherein any of B1, B2, and/or B3 is optionally absent;
    (d) x is an integer from 0 to 4, provided that when x is 1, 2, 3, or 4, one or more of the (N)x nucleotides can be complementary to nucleotides in target sequence, and one or more phosphorothioate internucleotide linkage(s) “s” can be present in the (N)x region when x is 1, 2, 3, or 4;
    (e) y is an integer from 0 to 4, provided that when y is 1, 2, 3, or 4, one or more of the (N)y nucleotides can be complementary to nucleotides in target sequence, and one or more phosphorothioate internucleotide linkage(s) “s” can be present in the (N)y region when y is 1, 2, 3, or 4;
    (f) N nucleotides of the guide strand are 2′-deoxy-2′-fluoro nucleotides and N nucleotides of the guide strand are 2′-O-methyl nucleotides, with an optional variance of 1 or 2 N or N nucleotides being tolerated provided that as RNAi activity is maintained;
    (g) S=a phosphorothioate or phosphorodithioate internucleotide linkage wherein S1 is required and S2 and S3 are optional; and
    (h) N nucleotides of the passenger strand are independently selected from ribonucleotide, 2′-O-alkyl nucleotide, 2′-deoxy-2′-fluoro nucleotide, 2′-deoxy nucleotide, and LNA.
  • In one embodiment, the invention features a composition comprising a double-stranded short interfering nucleic acid (siNA) molecule that modulates the expression and/or activity of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure represented by formula (B):
  • Figure US20150299696A1-20151022-C00002
  • wherein,
    (a) the upper strand is the passenger strand and the lower strand is the guide strand of the double-stranded nucleic acid molecule; the guide strand is complementary to the target sequence and the passenger strand is complementary to the guide strand, wherein one or more mismatches between the guide strand and passenger strand and/or between the guide strand and the target sequence are tolerated so long as RNAi activity is maintained;
    (b) each N is independently a nucleotide wherein one or more Ns may be substituted with a non-nucleotide moiety so long as RNAi activity is maintained;
    (c) each B1, B2, and B3 is independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker, wherein any of B1, B2, and/or B3 is optionally absent;
    (d) x is an integer from 0 to 4, provided that when x is 1, 2, 3, or 4, one or more of the (N)x nucleotides can be complementary to nucleotides in target sequence, and one or more phosphorothioate internucleotide linkage(s) “s” can be present in the (N)x region when x is 1, 2, 3, or 4;
    (e) y is an integer from 0 to 4, provided that when y is 1, 2, 3, or 4, one or more of the (N)y nucleotides can be complementary to nucleotides in target sequence, and one or more phosphorothioate internucleotide linkage(s) “s” can be present in the (N)y region when y is 1, 2, 3, or 4;
    (f) N nucleotides of the guide strand are 2′-deoxy-2′-fluoro nucleotides and N nucleotides of the guide strand are 2′-O-methyl nucleotides, with an optional variance of 1 or 2 N or N nucleotides being tolerated provided that as RNAi activity is maintained;
    (g) S=a phosphorothioate or phosphorodithioate internucleotide linkage wherein S1 is required and S2 and S3 are optional; and
    (h) N nucleotides of the passenger strand are independently selected from ribonucleotide, 2′-O-alkyl nucleotide, 2′-deoxy-2′-fluoro nucleotide, 2′-deoxy nucleotide, and LNA.
  • In one embodiment, the invention features a composition comprising a double-stranded short interfering nucleic acid (siNA) molecule that modulates the expression and/or activity of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure represented by formula (C):
  • Figure US20150299696A1-20151022-C00003
  • wherein,
    (a) the upper strand is the passenger strand and the lower strand is the guide strand of the double-stranded nucleic acid molecule; the guide strand is complementary to the target sequence and the passenger strand is complementary to the guide strand, wherein one or more mismatches between the guide strand and passenger strand and/or between the guide strand and the target sequence are tolerated so long as RNAi activity is maintained;
    (b) each N is independently a nucleotide wherein one or more Ns may be substituted with a non-nucleotide moiety so long as RNAi activity is maintained;
    (c) each B1, B2, and B3 is independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker, wherein any of B1, B2, and/or B3 is optionally absent;
    (d) x is an integer from 0 to 4, provided that when x is 1, 2, 3, or 4, one or more of the (N)x nucleotides can be complementary to nucleotides in target sequence, and one or more phosphorothioate internucleotide linkage(s) “s” can be present in the (N)x region when x is 1, 2, 3, or 4;
    (e) y is an integer from 0 to 4, provided that when y is 1, 2, 3, or 4, one or more of the (N)y nucleotides can be complementary to nucleotides in target sequence, and one or more phosphorothioate internucleotide linkage(s) “s” can be present in the (N)y region when y is 1, 2, 3, or 4;
    (f) N nucleotides of the guide strand are 2′-deoxy-2′-fluoro nucleotides and N nucleotides of the guide strand are 2′-O-methyl nucleotides, with an optional variance of 1 or 2 N or N nucleotides being tolerated provided that as RNAi activity is maintained;
    (g) S=a phosphorothioate or phosphorodithioate internucleotide linkage wherein S1 is required and S2 and S3 are optional; and
    (h) N nucleotides of the passenger strand are independently selected from ribonucleotide, 2′-O-alkyl nucleotide, 2′-deoxy-2′-fluoro nucleotide, 2′-deoxy nucleotide, and LNA.
    In one embodiment, the invention features a composition comprising a double-stranded short interfering nucleic acid (siNA) molecule that modulates the expression and/or activity of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure represented by formula (D):
  • Figure US20150299696A1-20151022-C00004
  • wherein,
    (a) the upper strand is the passenger strand and the lower strand is the guide strand of the double-stranded nucleic acid molecule; the guide strand is complementary to the target sequence and the passenger strand is complementary to the guide strand, wherein one or more mismatches between the guide strand and passenger strand and/or between the guide strand and the target sequence are tolerated so long as RNAi activity is maintained;
    (b) each N is independently a nucleotide wherein one or more Ns may be substituted with a non-nucleotide moiety so long as RNAi activity is maintained;
    (c) each B1, B2, and B3 is independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker, wherein any of B1, B2, and/or B3 is optionally absent;
    (d) x is an integer from 0 to 4, provided that when x is 1, 2, 3, or 4, one or more of the (N)x nucleotides can be complementary to nucleotides in target sequence, and one or more phosphorothioate internucleotide linkage(s) “s” can be present in the (N)x region when x is 1, 2, 3, or 4;
    (e) y is an integer from 0 to 4, provided that when y is 1, 2, 3, or 4, one or more of the (N)y nucleotides can be complementary to nucleotides in target sequence, and one or more phosphorothioate internucleotide linkage(s) “s” can be present in the (N)y region when y is 1, 2, 3, or 4;
    (f) N nucleotides of the guide strand are 2′-deoxy-2′-fluoro nucleotides and N nucleotides of the guide strand are 2′-O-methyl nucleotides, with an optional variance of 1 or 2 N or N nucleotides being tolerated provided that as RNAi activity is maintained;
    (g) S=a phosphorothioate or phosphorodithioate internucleotide linkage wherein S1 is required and S2 and S3 are optional; and
    (h) N nucleotides of the passenger strand are independently selected from ribonucleotide, 2′-O-alkyl nucleotide, 2′-deoxy-2′-fluoro nucleotide, 2′-deoxy nucleotide, and LNA.
  • In one embodiment, the invention features a composition comprising a double-stranded short interfering nucleic acid (siNA) molecule that modulates the expression and/or activity of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure represented by formula (E):
  • Figure US20150299696A1-20151022-C00005
  • wherein,
    (a) the upper strand is the passenger strand and the lower strand is the guide strand of the double-stranded nucleic acid molecule; the guide strand is complementary to the target sequence and the passenger strand is complementary to the guide strand, wherein one or more mismatches between the guide strand and passenger strand and/or between the guide strand and the target sequence are tolerated so long as RNAi activity is maintained;
    (b) each N is independently a nucleotide wherein one or more Ns may be substituted with a non-nucleotide moiety so long as RNAi activity is maintained;
    (c) each B1, B2, and B3 is independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker, wherein any of B1, B2, and/or B3 is optionally absent;
    (d) x is an integer from 0 to 4, provided that when x is 1, 2, 3, or 4, one or more of the (N)x nucleotides can be complementary to nucleotides in target sequence, and one or more phosphorothioate internucleotide linkage(s) “s” can be present in the (N)x region when x is 1, 2, 3, or 4;
    (e) y is an integer from 0 to 4, provided that when y is 1, 2, 3, or 4, one or more of the (N)y nucleotides can be complementary to nucleotides in target sequence, and one or more phosphorothioate internucleotide linkage(s) “s” can be present in the (N)y region when y is 1, 2, 3, or 4;
    (f) N nucleotides of the guide strand are 2′-deoxy-2′-fluoro nucleotides and N nucleotides of the guide strand are 2′-O-methyl nucleotides, with an optional variance of 1 or 2 N or N nucleotides being tolerated provided that as RNAi activity is maintained;
    (g) S=a phosphorothioate or phosphorodithioate internucleotide linkage wherein S1 is required and S2 and S3 are optional; and
    (h) N nucleotides of the passenger strand are independently selected from ribonucleotide, 2′-O-alkyl nucleotide, 2′-deoxy-2′-fluoro nucleotide, 2′-deoxy nucleotide, and LNA.
  • In one embodiment, the invention features a composition comprising a double-stranded short interfering nucleic acid (siNA) molecule that modulates the expression and/or activity of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure represented by formula (F):
  • Figure US20150299696A1-20151022-C00006
  • wherein,
    (a) the upper strand is the passenger strand and the lower strand is the guide strand of the double-stranded nucleic acid molecule; the guide strand is complementary to the target sequence and the passenger strand is complementary to the guide strand, wherein one or more mismatches between the guide strand and passenger strand and/or between the guide strand and the target sequence are tolerated so long as RNAi activity is maintained;
    (b) each N is independently a nucleotide wherein one or more Ns may be substituted with a non-nucleotide moiety so long as RNAi activity is maintained;
    (c) each B1, B2, and B3 is independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker, wherein any of B1, B2, and/or B3 is optionally absent;
    (d) x is an integer from 0 to 4, provided that when x is 1, 2, 3, or 4, one or more of the (N)x nucleotides can be complementary to nucleotides in target sequence, and one or more phosphorothioate internucleotide linkage(s) “s” can be present in the (N)x region when x is 1, 2, 3, or 4;
    (e) y is an integer from 0 to 4, provided that when y is 1, 2, 3, or 4, one or more of the (N)y nucleotides can be complementary to nucleotides in target sequence, and one or more phosphorothioate internucleotide linkage(s) “s” can be present in the (N)Y region when y is 1, 2, 3, or 4;
    (f) N nucleotides of the guide strand are 2′-deoxy-2′-fluoro nucleotides and N nucleotides of the guide strand are 2′-O-methyl nucleotides, with an optional variance of 1 or 2 N or N nucleotides being tolerated provided that as RNAi activity is maintained;
    (g) S=a phosphorothioate or phosphorodithioate internucleotide linkage wherein S1 is required and S2 and S3 are optional; and
    (h) N nucleotides of the passenger strand are independently selected from ribonucleotide, 2′-O-alkyl nucleotide, 2′-deoxy-2′-fluoro nucleotide, 2′-deoxy nucleotide, and LNA.
  • In one embodiment, the invention features a composition comprising a double-stranded short interfering nucleic acid (siNA) molecule that modulates the expression and/or activity of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure represented by formula (G):
  • Figure US20150299696A1-20151022-C00007
  • wherein,
    (a) the upper strand is the passenger strand and the lower strand is the guide strand of the double-stranded nucleic acid molecule; the guide strand is complementary to the target sequence and the passenger strand is complementary to the guide strand, wherein one or more mismatches between the guide strand and passenger strand and/or between the guide strand and the target sequence are tolerated so long as RNAi activity is maintained;
    (b) each N is independently a nucleotide wherein one or more Ns may be substituted with a non-nucleotide moiety so long as RNAi activity is maintained;
    (c) each B1, B2, and B3 is independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker, wherein any of B1, B2, and/or B3 is optionally absent;
    (d) x is an integer from 0 to 4, provided that when x is 1, 2, 3, or 4, one or more of the (N)x nucleotides can be complementary to nucleotides in target sequence, and one or more phosphorothioate internucleotide linkage(s) “s” can be present in the (N)x region when x is 1, 2, 3, or 4;
    (e) y is an integer from 0 to 4, provided that when y is 1, 2, 3, or 4, one or more of the (N)y nucleotides can be complementary to nucleotides in target sequence, and one or more phosphorothioate internucleotide linkage(s) “s” can be present in the (N)y region when y is 1, 2, 3, or 4;
    (f) N nucleotides of the guide strand are 2′-deoxy-2′-fluoro nucleotides and N nucleotides of the guide strand are 2′-O-methyl nucleotides, with an optional variance of 1 or 2 N or N nucleotides being tolerated provided that as RNAi activity is maintained;
    (g) S=a phosphorothioate or phosphorodithioate internucleotide linkage wherein S1 is required and S2 and S3 are optional; and
    (h) N nucleotides of the passenger strand are independently selected from ribonucleotide, 2′-O-alkyl nucleotide, 2′-deoxy-2′-fluoro nucleotide, 2′-deoxy nucleotide, and LNA.
  • In one embodiment, the invention features a composition comprising a double-stranded short interfering nucleic acid (siNA) molecule that modulates the expression and/or activity of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure represented by formula (H):
  • Figure US20150299696A1-20151022-C00008
  • wherein,
    (a) the upper strand is the passenger strand and the lower strand is the guide strand of the double-stranded nucleic acid molecule; the guide strand is complementary to the target sequence and the passenger strand is complementary to the guide strand, wherein one or more mismatches between the guide strand and passenger strand and/or between the guide strand and the target sequence are tolerated so long as RNAi activity is maintained;
    (b) each N is independently a nucleotide wherein one or more Ns may be substituted with a non-nucleotide moiety so long as RNAi activity is maintained;
    (c) each B1, B2, and B3 is independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker, wherein any of B1, B2, and/or B3 is optionally absent;
    (d) x is an integer from 0 to 4, provided that when x is 1, 2, 3, or 4, one or more of the (N)x nucleotides can be complementary to nucleotides in target sequence, and one or more phosphorothioate internucleotide linkage(s) “s” can be present in the (N)x region when x is 1, 2, 3, or 4;
    (e) y is an integer from 0 to 4, provided that when y is 1, 2, 3, or 4, one or more of the (N)y nucleotides can be complementary to nucleotides in target sequence, and one or more phosphorothioate internucleotide linkage(s) “s” can be present in the (N)y region when y is 1, 2, 3, or 4;
    (f) N nucleotides of the guide strand are 2′-deoxy-2′-fluoro nucleotides and N nucleotides of the guide strand are 2′-O-methyl nucleotides, with an optional variance of 1 or 2 N or N nucleotides being tolerated provided that as RNAi activity is maintained;
    (g) S=a phosphorothioate or phosphorodithioate internucleotide linkage wherein S1 is required and S2 and S3 are optional; and
    (h) N nucleotides of the passenger strand are independently selected from ribonucleotide, 2′-O-alkyl nucleotide, 2′-deoxy-2′-fluoro nucleotide, 2′-deoxy nucleotide, and LNA.
  • In one embodiment, the invention features a composition comprising a double-stranded short interfering nucleic acid (siNA) molecule that modulates the expression and/or activity of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure represented by formula (I):
  • Figure US20150299696A1-20151022-C00009
  • wherein,
    (a) the upper strand is the passenger strand and the lower strand is the guide strand of the double-stranded nucleic acid molecule; the guide strand is complementary to the target sequence and the passenger strand is complementary to the guide strand, wherein one or more mismatches between the guide strand and passenger strand and/or between the guide strand and the target sequence are tolerated so long as RNAi activity is maintained;
    (b) each N is independently a nucleotide wherein one or more Ns may be substituted with a non-nucleotide moiety so long as RNAi activity is maintained;
    (c) each B1, B2, and B3 is independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker, wherein any of B1, B2, and/or B3 is optionally absent;
    (d) x is an integer from 0 to 4, provided that when x is 1, 2, 3, or 4, one or more of the (N)x nucleotides can be complementary to nucleotides in target sequence, and one or more phosphorothioate internucleotide linkage(s) “s” can be present in the (N)x region when x is 1, 2, 3, or 4;
    (e) y is an integer from 0 to 4, provided that when y is 1, 2, 3, or 4, one or more of the (N)y nucleotides can be complementary to nucleotides in target sequence, and one or more phosphorothioate internucleotide linkage(s) “s” can be present in the (N)y region when y is 1, 2, 3, or 4;
    (f) N nucleotides of the guide strand are 2′-deoxy-2′-fluoro nucleotides and N nucleotides of the guide strand are 2′-O-methyl nucleotides, with an optional variance of 1 or 2 N or N nucleotides being tolerated provided that as RNAi activity is maintained;
    (g) S=a phosphorothioate or phosphorodithioate internucleotide linkage wherein S1 is required and S2 and S3 are optional; and
    (h) N nucleotides of the passenger strand are independently selected from ribonucleotide, 2′-O-alkyl nucleotide, 2′-deoxy-2′-fluoro nucleotide, 2′-deoxy nucleotide, and LNA.
  • In one embodiment, the invention features a composition comprising a double-stranded short interfering nucleic acid (siNA) molecule that modulates the expression and/or activity of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure represented by formula (J):
  • Figure US20150299696A1-20151022-C00010
  • wherein,
    (a) the upper strand is the passenger strand and the lower strand is the guide strand of the double-stranded nucleic acid molecule; the guide strand is complementary to the target sequence and the passenger strand is complementary to the guide strand, wherein one or more mismatches between the guide strand and passenger strand and/or between the guide strand and the target sequence are tolerated so long as RNAi activity is maintained;
    (b) each N is independently a nucleotide wherein one or more Ns may be substituted with a non-nucleotide moiety so long as RNAi activity is maintained;
    (c) each B1, B2, and B3 is independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker, wherein any of B1, B2, and/or B3 is optionally absent;
    (d) x is an integer from 0 to 4, provided that when x is 1, 2, 3, or 4, one or more of the (N)x nucleotides can be complementary to nucleotides in target sequence, and one or more phosphorothioate internucleotide linkage(s) “s” can be present in the (N)x region when x is 1, 2, 3, or 4;
    (e) y is an integer from 0 to 4, provided that when y is 1, 2, 3, or 4, one or more of the (N)y nucleotides can be complementary to nucleotides in target sequence, and one or more phosphorothioate internucleotide linkage(s) “s” can be present in the (N)Y region when y is 1, 2, 3, or 4;
    (f) N nucleotides of the guide strand are 2′-deoxy-2′-fluoro nucleotides and N nucleotides of the guide strand are 2′-O-methyl nucleotides, with an optional variance of 1 or 2 N or N nucleotides being tolerated provided that as RNAi activity is maintained;
    (g) S=a phosphorothioate or phosphorodithioate internucleotide linkage wherein S1 is required and S2 and S3 are optional; and
    (h) N nucleotides of the passenger strand are independently selected from ribonucleotide, 2′-O-alkyl nucleotide, 2′-deoxy-2′-fluoro nucleotide, 2′-deoxy nucleotide, and LNA.
  • In some embodiments, each (N) of the passenger strand of a composition according to any of Formula A, B, C, D, E, F, G, H, I or J is independently selected from 2′-O-alkyl, 2′-deoxy-2′-fluoro, 2′-deoxy, and/or LNA modified nucleotide.
  • In some embodiments, each (N) of the passenger strand of a composition according to any of Formula A, B, C, D, E, F, G, H, I or J is a 2′-O-alkyl modified nucleotide.
  • In some embodiments, each (N) of the passenger strand of a composition according to any of Formula A, B, C, D, E, F, G, H, I or J is a 2′-O-methyl modified nucleotide.
  • In some embodiments, x and y of a composition according to any of Formula A, B, C, D, E, F, G, H, I or J are both equal to 2.
  • In some embodiments, all N nucleotides of the passenger strand of a composition according to any of Formula A, B, C, D, E, F, G, H, I or J are 2′-deoxy-2′-fluoro nucleotides.
  • In some embodiments, 5, 6, 7, 8, 9, or 10 or more pyrimidine N nucleotides of the passenger strand are 2′-O-methyl nucleotides and 5, 6, 7, 8, 9, or 10 or more purine N nucleotides of the passenger strand of a composition according to any of Formula A, B, C, D, E, F, G, H, I or J are 2′-deoxy-2′-fluoro nucleotides.
  • In some embodiments, the 5′-terminal N position of a composition according to any of Formula A, B, C, D, E, F, G, H, I or J comprises a terminal phosphate group.
  • In some embodiments, with respect to a composition according to any of Formula A, B, C, D, E, F, G, H, I or J, all N nucleotides of the guide strand are 2′-deoxy-2′-fluoro nucleotides and all N nucleotides of the guide strand are 2′-O-methyl nucleotides, with no variance being permitted.
  • In some embodiments, B1 and B2 of a composition according to any of Formula A, B, C, D, E, F, G, H, I or J each comprise an inverted abasic 2′-deoxyribose moiety and B3 is optionally absent.
  • In some embodiments, with respect to a composition according to any of Formula A, B, C, D, E, F, G, H, I or J, B1 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, B2 comprises an inverted abasic 2′-deoxyribose moiety, and B3 is optionally absent.
  • In some embodiments, with respect to a composition according to any of Formula A, B, C, D, E, F, G, H, I or J, B1 comprises one or more galactosamine moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, B2 comprises an inverted abasic 2′-deoxyribose moiety, and B3 is optionally absent.
  • In some embodiments, with respect to a composition according to any of Formula A, B, C, D, E, F, G, H, I or J, B1 comprises one or more N-acetylgalactosamine moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, B2 comprises an inverted abasic 2′-deoxyribose moiety, and B3 is optionally absent.
  • In some embodiments, with respect to a composition according to any of Formula A, B, C, D, E, F, G, H, I or J, B1 comprises one or more folate moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, B2 comprises an inverted abasic 2′-deoxyribose moiety, and B3 is optionally absent.
  • In some embodiments, with respect to a composition according to any of Formula A, B, C, D, E, F, G, H, I or J, B1 comprises one or more cholesterol moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, B2 comprises an inverted abasic 2′-deoxyribose moiety, and B3 is optionally absent.
  • In some embodiments, with respect to a composition according to any of Formula A, B, C, D, E, F, G, H, I or J, B1 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, B2 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B3 is optionally absent.
  • In some embodiments, with respect to a composition according to any of Formula A, B, C, D, E, F, G, H, I or J, B1 comprises one or more galactosamine moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, B2 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B3 is optionally absent.
  • In some embodiments, with respect to a composition according to any of Formula A, B, C, D, E, F, G, H, I or J, B1 comprises one or more N-acetylgalactosamine moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, B2 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B3 is optionally absent.
  • In some embodiments, with respect to a composition according to any of Formula A, B, C, D, E, F, G, H, I or J, B1 comprises one or more folate moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, B2 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B3 is optionally absent.
  • In some embodiments, with respect to a composition according to any of Formula A, B, C, D, E, F, G, H, I or J, B1 comprises one or more cholesterol moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, B2 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B3 is optionally absent.
  • In some embodiments, with respect to a composition according to any of Formula A, B, C, D, E, F, G, H, I or J, B1 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, B2 comprises any of SEQ ID NOS: 54-528 optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B3 is optionally absent.
  • In some embodiments, with respect to a composition according to any of Formula A, B, C, D, E, F, G, H, I or J, B1 comprises one or more galactosamine moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, B2 comprises any of SEQ ID NOS: 54-528 optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B3 is optionally absent.
  • In some embodiments, with respect to a composition according to any of Formula A, B, C, D, E, F, G, H, I or J, B1 comprises one or more N-acetylgalactosamine moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, B2 comprises any of SEQ ID NOS: 54-528 optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B3 is optionally absent.
  • In some embodiments, with respect to a composition according to any of Formula A, B, C, D, E, F, G, H, I or J, B1 comprises one or more folate moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, B2 comprises any of SEQ ID NOS: 54-528 optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B3 is optionally absent.
  • In some embodiments, with respect to a composition according to any of Formula A, B, C, D, E, F, G, H, I or J, B1 comprises one or more cholesterol moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, B2 comprises any of SEQ ID NOS: 54-528 optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B3 is optionally absent.
  • In some embodiments, with respect to a composition according to any of Formula A, B, C, D, E, F, G, H, I or J, B1 comprises a folate, galactosamine, or cholesterol moiety as described in U.S. Pat. No. 7,491,805.
  • In some embodiments, with respect to a composition according to any of Formula A, B, C, D, E, F, G, H, I or J, S1 and S2 are required and S3 is optional.
  • In some embodiments, with respect to a composition according to any of Formula A, B, C, D or E, S1 and S3 are required and S2 is optional.
  • In some embodiments, with respect to a composition according to any of Formula A, B, C, D, E, F, G, H, I or J, S1, S2, and S3 are required.
  • In some embodiments, with respect to a composition according to any of Formula A, B, C, D, E, F, G, H, I or J, B1 comprises:
  • Figure US20150299696A1-20151022-C00011
  • wherein each L is independently a linker and GalNAc is an N-acetyl galactosamine moiety.
  • In some embodiments, with respect to a composition according to any of Formula A, B, C, D, E, F, G, H, I or J, B1 comprises:
  • Figure US20150299696A1-20151022-C00012
  • In some embodiments, with respect to a composition according to any of Formula A, B, C, D, E, F, G, H, I or J, B1 comprises:
  • Figure US20150299696A1-20151022-C00013
  • wherein n is an integer from 1 to 20.
    In some embodiments, with respect to a composition according to any of Formula A, B, C, D, E, F, G, H, I or J, B1 and/or B2 comprises:
  • Figure US20150299696A1-20151022-C00014
  • wherein X is —O—, —S—, —CH2— or —NH—; n is 1, 2, 3, or 4; and the bond with “
    Figure US20150299696A1-20151022-P00001
    ” indicates the point of attachment optionally including one or more linkers, which may be the same or different; and further optionally including one or more targeting ligands, solubilizing agents, pharmacokinetics enhancing agents, lipids, and/or masking agents. In certain embodiments, the linker is a linker shown in Table 8.
  • In one embodiment, the invention features a composition comprising a double-stranded short interfering nucleic acid (siNA) molecule that modulates the expression and/or activity of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure represented by formula (K):
  • Figure US20150299696A1-20151022-C00015
  • wherein,
    (a) the upper strand is the passenger strand and the lower strand is the guide strand of the double-stranded nucleic acid molecule; the guide strand is complementary to the target sequence and the passenger strand is complementary to the guide strand, wherein one or more mismatches between the guide strand and passenger strand and/or between the guide strand and the target sequence are tolerated so long as RNAi activity is maintained;
    (b) B1 and B2, are each independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker;
    (c) every N nucleotide is a 2′-deoxy-2′-fluoro nucleotide and every N nucleotide is a 2′-O-methyl nucleotide;
    (d) each S is a phosphorothioate or phosphorodithioate internucleotide linkage.
    In one embodiment, the invention features a composition comprising a double-stranded short interfering nucleic acid (siNA) molecule that modulates the expression and/or activity of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure represented by formula (L):
  • Figure US20150299696A1-20151022-C00016
  • wherein,
    (a) the upper strand is the passenger strand and the lower strand is the guide strand of the double-stranded nucleic acid molecule; the guide strand is complementary to the target sequence and the passenger strand is complementary to the guide strand, wherein one or more mismatches between the guide strand and passenger strand and/or between the guide strand and the target sequence are tolerated so long as RNAi activity is maintained;
    (b) B1 and B2, are each independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker;
    (c) every N nucleotide is a 2′-deoxy-2′-fluoro nucleotide and every N nucleotide is a 2′-O-methyl nucleotide;
    (d) each S is a phosphorothioate or phosphorodithioate internucleotide linkage.
  • In one embodiment, the invention features a composition comprising a double-stranded short interfering nucleic acid (siNA) molecule that modulates the expression and/or activity of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure represented by formula (M):
  • Figure US20150299696A1-20151022-C00017
  • wherein,
    (a) the upper strand is the passenger strand and the lower strand is the guide strand of the double-stranded nucleic acid molecule; the guide strand is complementary to the target sequence and the passenger strand is complementary to the guide strand, wherein one or more mismatches between the guide strand and passenger strand and/or between the guide strand and the target sequence are tolerated so long as RNAi activity is maintained;
    (b) B1 and B2, are each independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker;
    (c) every N nucleotide is a 2′-deoxy-2′-fluoro nucleotide and every N nucleotide is a 2′-O-methyl nucleotide;
    (d) each S is a phosphorothioate or phosphorodithioate internucleotide linkage.
  • In one embodiment, the invention features a composition comprising a double-stranded short interfering nucleic acid (siNA) molecule that modulates the expression and/or activity of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure represented by formula (N):
  • Figure US20150299696A1-20151022-C00018
  • wherein,
    (a) the upper strand is the passenger strand and the lower strand is the guide strand of the double-stranded nucleic acid molecule; the guide strand is complementary to the target sequence and the passenger strand is complementary to the guide strand, wherein one or more mismatches between the guide strand and passenger strand and/or between the guide strand and the target sequence are tolerated so long as RNAi activity is maintained;
    (b) B1 and B2, are each independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker;
    (c) every N nucleotide is a 2′-deoxy-2′-fluoro nucleotide and every N nucleotide is a 2′-O-methyl nucleotide;
    (d) each S is a phosphorothioate or phosphorodithioate internucleotide linkage.
    In one embodiment, the invention features a composition comprising a double-stranded short interfering nucleic acid (siNA) molecule that modulates the expression and/or activity of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure represented by formula (O):
  • Figure US20150299696A1-20151022-C00019
  • wherein,
    (a) the upper strand is the passenger strand and the lower strand is the guide strand of the double-stranded nucleic acid molecule; the guide strand is complementary to the target sequence and the passenger strand is complementary to the guide strand, wherein one or more mismatches between the guide strand and passenger strand and/or between the guide strand and the target sequence are tolerated so long as RNAi activity is maintained;
    (b) B1 and B2, are each independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker;
    (c) every N nucleotide is a 2′-deoxy-2′-fluoro nucleotide and every N nucleotide is a 2′-O-methyl nucleotide;
    (d) each S is a phosphorothioate or phosphorodithioate internucleotide linkage.
  • In one embodiment, the invention features a composition comprising a double-stranded short interfering nucleic acid (siNA) molecule that modulates the expression and/or activity of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure represented by formula (P):
  • Figure US20150299696A1-20151022-C00020
  • wherein,
    (a) the upper strand is the passenger strand and the lower strand is the guide strand of the double-stranded nucleic acid molecule; the guide strand is complementary to the target sequence and the passenger strand is complementary to the guide strand, wherein one or more mismatches between the guide strand and passenger strand and/or between the guide strand and the target sequence are tolerated so long as RNAi activity is maintained;
    (b) B1 and B2, are each independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker;
    (c) every N nucleotide is a 2′-deoxy-2′-fluoro nucleotide and every N nucleotide is a 2′-O-methyl nucleotide;
    (d) each S is a phosphorothioate or phosphorodithioate internucleotide linkage.
  • In some embodiments, the 5′-terminal N position of a composition according to any of Formula K, L, M, N, O, or P comprises a terminal phosphate group.
  • In some embodiments, B1 and B2 of a composition according to any of Formula K, L, M, N, O, or P each comprise an inverted abasic 2′-deoxyribose moiety.
  • In some embodiments, with respect to a composition according to any of Formula K, L, M, N, O, or P, B1 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B2 comprises an inverted abasic 2′-deoxyribose moiety.
  • In some embodiments, with respect to a composition according to any of Formula K, L, M, N, O, or P, B1 comprises one or more galactosamine moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B2 comprises an inverted abasic 2′-deoxyribose moiety.
  • In some embodiments, with respect to a composition according to any of Formula K, L, M, N, O, or P, B1 comprises one or more N-acetylgalactosamine moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B2 comprises an inverted abasic 2′-deoxyribose moiety.
  • In some embodiments, with respect to a composition according to any of Formula K, L, M, N, O, or P, B1 comprises one or more folate moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B2 comprises an inverted abasic 2′-deoxyribose moiety.
  • In some embodiments, with respect to a composition according to any of Formula K, L, M, N, O, or P, B1 comprises one or more cholesterol moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B2 comprises an inverted abasic 2′-deoxyribose moiety.
  • In some embodiments, with respect to a composition according to any of Formula K, L, M, N, O, or P, B1 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B2 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety.
  • In some embodiments, with respect to a composition according to any of Formula K, L, M, N, O, or P, B1 comprises one or more galactosamine moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B2 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety.
  • In some embodiments, with respect to a composition according to any of Formula K, L, M, N, O, or P, B1 comprises one or more N-acetylgalactosamine moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B2 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety.
  • In some embodiments, with respect to a composition according to any of Formula K, L, M, N, O, or P, B1 comprises one or more folate moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B2 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety.
  • In some embodiments, with respect to a composition according to any of Formula K, L, M, N, O, or P, B1 comprises one or more cholesterol moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B2 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety.
  • In some embodiments, with respect to a composition according to any of Formula K, L, M, N, O, or P, B1 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B2 comprises any of SEQ ID NOS: 54-528 optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety.
  • In some embodiments, with respect to a composition according to any of Formula K, L, M, N, O, or P, B1 comprises one or more galactosamine moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B2 comprises any of SEQ ID NOS: 54-528 optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety.
  • In some embodiments, with respect to a composition according to any of Formula K, L, M, N, O, or P, B1 comprises one or more N-acetylgalactosamine moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B2 comprises any of SEQ ID NOS: 54-528 optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety.
  • In some embodiments, with respect to a composition according to any of Formula K, L, M, N, O, or P, B1 comprises one or more folate moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B2 comprises any of SEQ ID NOS: 54-528 optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety.
  • In some embodiments, with respect to a composition according to any of Formula F, G, or H, B1 comprises one or more cholesterol moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B2 comprises any of SEQ ID NOS: 54-528 optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety.
  • In some embodiments, with respect to a composition according to any of Formula K, L, M, N, O, or P, B1 comprises a folate, galactosamine, or cholesterol moiety as described in U.S. Pat. No. 7,491,805.
  • In some embodiments, with respect to a composition according to any of Formula K, L, M, N, O, or P, B1 comprises:
  • Figure US20150299696A1-20151022-C00021
  • wherein each L is independently a linker and GalNAc is an N-acetyl galactosamine moiety.
  • In some embodiments, with respect to a composition according to any of Formula K, L, M, N, O, or P, B1 comprises:
  • Figure US20150299696A1-20151022-C00022
  • In some embodiments, with respect to a composition according to any of Formula K, L, M, N, O, or P, B1 comprises:
  • Figure US20150299696A1-20151022-C00023
  • wherein n is an integer from 1 to 20.
  • In some embodiments, with respect to a composition according to any of Formula K, L, M, N, O, or P, B1 and/or B2 comprises:
  • Figure US20150299696A1-20151022-C00024
  • wherein X is —O—, —S—, —CH2— or —NH—; n is 1, 2, 3, or 4; and the bond with “
    Figure US20150299696A1-20151022-P00001
    ” indicates the point of attachment optionally including one or more linkers, which may be the same or different; and further optionally including one or more targeting ligands, solubilizing agents, pharmacokinetics enhancing agents, lipids, and/or masking agents. In certain embodiments, the linker is a linker shown in Table 8.
  • With respect to any siNA having Formula A, B, C, D, E, F, G, H, I, J, K, L, M, N, O, or P described herein, in certain embodiments, the siNA molecule is covalently attached to a polymer or ligand via a linker. In certain embodiments, the siNA molecule is covalently attached to the polymer or ligand via a linker moiety at the 5′-end of the passenger (sense) strand of the siNA molecule. In other embodiments, the siNA molecule is covalently attached to the polymer or ligand via a linker moiety at the 3′-end of the passenger (sense) strand of the siNA molecule. In other embodiments, the siNA molecule is covalently attached to the polymer or ligand via a linker moiety at the 3′-end of the guide (antisense) strand of the siNA molecule. In any of the above embodiments, the linker can be attached to the terminal 3′ and/or 5′ nucleotide position of the passenger or guide strand, or can alternately be attached to a terminal cap moiety such as an abasic moiety or other cap as described herein or otherwise known in the art. Therefore, in totality, a siNA molecule of the invention having Formula (A) can comprise a terminal cap (B) that includes a covalent attachment to a polymer or ligand via a linker molecule as described herein or as is otherwise known in the art. Non-limiting examples of such linkers are provided in the examples herein.
  • In certain embodiments, one or more terminal cap moieties of a siNA molecule of the invention (i.e. any B of any compound having Formula A, B, C, D, E, F, G, H, I, J, K, L, M, N, O, or P herein) can comprise a delivery modality. The delivery modality can comprise a ligand or polymer that further includes one or more linker molecules. Non-limiting examples of such linker molecules include phosphate ester based linkages, amino based linkers, disulfide based linkers, succinyl based linkers, alkyl or substituted alkyl based linkers, and/or amide based linkers as are generally known in the art.
  • In some embodiments, the siNA molecules of the invention are phosphorylated at the 5′ end of the guide or antisense strand. The phosphate group can be a phosphate, a diphosphate or a triphosphate.
  • The present invention further provides compositions comprising the double-stranded nucleic acid molecules described herein with optionally a pharmaceutically acceptable carrier or diluent.
  • In some embodiments, a composition of the invention further comprises a cryo-protectant. In some embodiments, the cryo-protectant is Sucrose, Trehalose, Raffinose, Stachyose, Verbascose, Mannitol, Glucose, Lactose, Maltose, Maltotriose-heptaose, Dextran, Hydroxyethyl Starch, Insulin, Sorbitol, Glycerol, Arginine, Histidine, Lysine, Proline, Dimethylsulfoxide or any combination thereof. In some embodiments, the cryo-protectant is Sucrose. In some embodiments, the cryo-protectant is Trehalose. In some embodiments, the cryo-protectant is a combination of Sucrose and Trehalose.
  • The present invention further provides a compound comprising a double-stranded short interfering nucleic acid (siNA) molecule of the invention covalently attached to a ligand. Non limiting examples of ligands include steroidal compounds (e.g., cholesterol), galactosamines (e.g., N-acetylgalactosamine), vitamins (e.g., folate), proteins (e.g., monoclonal antibodies), and peptides (e.g., TAT) as are generally known in the art and further provided herein.
  • The administration of the compositions of the invention can be carried out by known methods, wherein the nucleic acid is introduced into a desired target cell in vitro or in vivo.
  • Commonly used techniques for introduction of the nucleic acid molecules of the invention into cells, tissues, and organisms include the use of various carrier systems, reagents and vectors. Non-limiting examples of such carrier systems suitable for use in the present invention include single chemical entity conjugates, nucleic-acid-lipid particles, lipid nanoparticles (LNP), liposomes, lipoplexes, micelles, virosomes, virus like particles (VLP), nucleic acid polymers, and mixtures thereof.
  • The compositions of the invention can be in the form of an aerosol, dispersion, solution (e.g., an injectable solution), a cream, ointment, tablet, powder, suspension or the like. These compositions may be administered in any suitable way, e.g. orally, sublingually, buccally, parenterally, nasally, or topically. In some embodiments, the compositions are aerosolized and delivered via inhalation.
  • The molecules and compositions of the present invention have utility over a broad range of therapeutic applications. Accordingly another aspect of this invention relates to the use of the compounds and compositions of the invention in treating a subject. The invention thus provides a method for treating a subject, such as a human, suffering from a condition which is associated with the expression of one or more genes, wherein the method comprises administering to the subject an effective amount of a double-stranded short interfering nucleic acid (siNA) molecule of the invention. Thus, the siNA molecules of the invention treat the disease or condition. In some embodiments, the condition is one as described herein or is otherwise generally known to one of skill in the art.
  • The present invention provides compositions and methods comprising siNAs having target specificity that may be used to treat diseases and conditions herein or otherwise known in the art that are associated with gene expression. In particular aspects and embodiments of the invention, the nucleic acid molecules of the invention comprise at least a 15 nucleotide sequence of the a target sequence, and/or comprises a nucleotide sequence of at least 15 nucleotides complimentary to the target sequence). The siNAs may be provided in several forms. For example, the siNA may be isolated as one or more siNA compounds, or it may be in the form of a transcriptional cassette in a DNA plasmid. The siNA may also be chemically synthesized and may include modifications as described herein or as is otherwise generally known in the art. The siNAs may be administered alone or co-administered with other siNA molecules or with conventional agents that treat a gene related disease or condition as described herein or otherwise known in the art.
  • The siNA molecules of the invention may be used to mediate gene silencing via interaction with RNA transcripts or alternately by interaction with particular gene sequences, wherein such interaction results in modulation of gene silencing either at the transcriptional level or post-transcriptional level such as, for example, but not limited to, RNAi or through cellular processes that modulate the chromatin structure or methylation patterns of the target and prevent transcription of the target gene, with the nucleotide sequence of the target thereby mediating silencing. More specifically, the target is any GenBank reference sequence as is presently known in the art.
  • In one aspect, the invention provides short interfering nucleic acid (siNA) molecules that may inhibit the expression of the target gene in a cell or mammal. The siNA may be single-stranded or double-stranded. When double-stranded, the siNA comprising a sense and an antisense stand. The antisense strand may be complementary to at least a part of an mRNA formed in the expression of the target gene. The sense strand comprises a region that may be complementary to the antisense strand.
  • The double stranded RNA molecules of the invention may comprise two distinct and separate strands that can be symmetric or asymmetric and are complementary, i.e., two single-stranded RNA molecules, or may comprise one single-stranded molecule in which two complementary portions, e.g., a sense region and an antisense region, are base-paired, and are covalently linked by one or more single-stranded “hairpin” areas (i.e. loops) resulting in, for example, a single-stranded short-hairpin polynucleotide or a circular single-stranded polynucleotide.
  • The linker may be polynucleotide linker or a non-nucleotide linker. In some embodiments, the linker is a non-nucleotide linker. In some embodiments, a hairpin or circular siNA molecule of the invention contains one or more loop motifs, wherein at least one of the loop portions of the siNA molecule is biodegradable. For example, a single-stranded hairpin siNA molecule of the invention is designed such that degradation of the loop portion of the siNA molecule in vivo can generate a double-stranded siNA molecule with 3′-terminal overhangs, such as 3′-terminal nucleotide overhangs comprising 1, 2, 3 or 4 nucleotides. Or alternatively, a circular siNA molecule of the invention is designed such that degradation of the loop portions of the siNA molecule in vivo can generate a double-stranded siNA molecule with 3′-terminal overhangs, such as 3′-terminal nucleotide overhangs comprising about 2 nucleotides.
  • In some embodiments, siNA molecules of the invention may have perfect complementarity between the sense strand or sense region and the antisense strand or antisense region of the siNA molecule. In other or the same embodiments, the antisense strand of the siNA molecules of the invention are perfectly complementary to a corresponding target nucleic acid molecule.
  • In yet other embodiments, siNA molecules of the invention have partial complementarity (i.e., less than 100% complementarity) between the sense strand or sense region and the antisense strand or antisense region of the siNA molecule or between the antisense strand or antisense region of the siNA molecule and a corresponding target nucleic acid molecule. Thus, in some embodiments, the double-stranded nucleic acid molecules of the invention, have between about 15 to about 30 (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides in one strand that are complementary to the nucleotides of the other strand. In other embodiments, the molecules have between about 15 to about 30 (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides in the sense region that are complementary to the nucleotides of the antisense region. of the double-stranded nucleic acid molecule. In certain embodiments, the double-stranded nucleic acid molecules of the invention have between about 15 to about 30 (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides in the antisense strand that are complementary to a nucleotide sequence of its corresponding target nucleic acid molecule.
  • In other embodiments, the siNA molecule may contain one or more nucleotide deletions, substitutions, mismatches and/or additions; provided, however, that the siNA molecule maintains its activity, for example, to mediate RNAi. In a non-limiting example, the deletion, substitution, mismatch and/or addition may result in a loop or bulge, or alternately a wobble or other alternative (non Watson-Crick) base pair. Thus, in some embodiments, for example, the double-stranded nucleic acid molecules of the invention, have 1 or more (e.g., 1, 2, 3, 4, 5, or 6) nucleotides, in one strand or region that are mismatches or non-base-paired with the other strand or region. In other embodiments, the double-stranded nucleic acid molecules of the invention, have 1 or more (e.g., 1, 2, 3, 4, 5, or 6) nucleotides in each strand or region that are mismatches or non-base-paired with the other strand or region. In a preferred embodiment, the siNA of the invention contains no more than 3 mismatches. If the antisense strand of the siNA contains mismatches to a target sequence, it is preferable that the area of mismatch not be located in the center of the region of complementarity.
  • In other embodiments, the siNA molecule may contain one or more nucleotide deletions, substitutions, mismatches and/or additions to a sequence provided herein, however, that the siNA molecule maintains its activity, for example, to mediate RNAi. In a non-limiting example, the deletion, substitution, mismatch and/or addition may result in a loop or bulge, or alternately a wobble or other alternative (non Watson-Crick) base pair.
  • The invention also comprises double-stranded nucleic acid (siNA) molecules as otherwise described hereinabove in which the first strand and second strand are complementary to each other and wherein at least one strand is hybridisable to the polynucleotide sequence of a target sequence under conditions of high stringency, and wherein any of the nucleotides is unmodified or chemically modified.
  • Hybridization techniques are well known to the skilled artisan (see for instance, Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989)). Preferred stringent hybridization conditions include overnight incubation at 42° C. in a solution comprising: 50% formamide, 5×SSC (150 mM NaCl, 15 mM trisodium citrate), 50 mM sodium phosphate (pH 7.6), 5×Denhardt's solution, 10% dextran sulfate, and 20 microgram/ml denatured, sheared salmon sperm DNA; followed by washing the filters in 0.1×SSC at about 65° C.
  • In some embodiments, the first strand has about 18, 19, 20, 21 or 22 nucleotides that are complementary to the nucleotides of the other strand and at least one strand is hybridisable to target sequence such as a gene in the GenBank database. In a more preferred embodiment, the first strand has about 19, 20 or 21 nucleotides that are complementary to the nucleotides of the other strand and at least one strand is hybridisable at least one strand is hybridisable to the complement of a target sequence under conditions of high stringency.
  • In certain embodiments, the siNA molecules of the invention comprise overhangs of about 1 to about 4 (e.g., about 1, 2, 3 or 4) nucleotides. The nucleotides in the overhangs may be the same or different nucleotides. In some embodiments, the overhangs occur at the 3′-end at one or both strands of the double-stranded nucleic acid molecule. For example, a double-stranded nucleic acid molecule of the invention may comprise a nucleotide or non-nucleotide overhang at the 3′-end of the antisense strand/region, the 3′-end of the sense strand/region, or both the antisense strand/region and the sense strand/region of the double-stranded nucleic acid molecule.
  • In some embodiments, the nucleotides comprising the overhang portion of an siNA molecule of the invention comprise sequences based on the target polynucleotide sequence in which nucleotides comprising the overhang portion of the antisense strand/region of an siNA molecule of the invention can be complementary to nucleotides in the target polynucleotide sequence and/or nucleotides comprising the overhang portion of the sense strand/region of an siNA molecule of the invention can comprise the nucleotides in the target polynucleotide sequence. Thus, in some embodiments, the overhang comprises a two nucleotide overhang that is complementary to a portion of the target polynucleotide sequence. In other embodiments, however, the overhang comprises a two nucleotide overhang that is not complementary to a portion of the target polynucleotide sequence. In certain embodiments, the overhang comprises a 3′-UU overhang that is not complementary to a portion of the target polynucleotide sequence. In other embodiments, the overhang comprises a UU overhang at the 3′ end of the antisense strand and a TT overhang at the 3′ end of the sense strand. In other embodiments, the overhang comprises nucleotides as described in the examples, Tables, and Figures herein.
  • In any of the embodiments of the siNA molecules described herein having 3′-terminal nucleotide overhangs, the overhangs are optionally chemically modified at one or more nucleic acid sugar, base, or backbone positions. Representative, but not limiting examples of modified nucleotides in the overhang portion of a double-stranded nucleic acid (siNA) molecule of the invention include: 2′-O-alkyl (e.g., 2′-O-methyl), 2′-deoxy, 2′-deoxy-2′-fluoro, 2′-deoxy-2′-fluoroarabino (FANA), 4′-thio, 2′-O-trifluoromethyl, 2′-O-ethyl-trifluoromethoxy, 2′-O-difluoromethoxy-ethoxy, universal base, acyclic, or 5-C-methyl nucleotides. In more preferred embodiments, the overhang nucleotides are each independently, a 2′-O-alkyl nucleotide, a 2′-O-methyl nucleotide, a 2′-dexoy-2-fluoro nucleotide, or a 2′-deoxy ribonucleotide. In some instances the overhang nucleotides are linked by a one or more phosphorothioate linkages.
  • In yet other embodiments, siNA molecules of the invention comprise duplex nucleic acid molecules with blunt ends (i.e., without nucleotide overhangs), where both ends are blunt, or alternatively, where one of the ends is blunt. In some embodiments, the siNA molecules of the invention can comprises one blunt end, for example wherein the 5′-end of the antisense strand and the 3′-end of the sense strand do not have any overhanging nucleotides. In another example, the siNA molecule comprises one blunt end, for example wherein the 3′-end of the antisense strand and the 5′-end of the sense strand do not have any overhanging nucleotides. In other embodiments, siNA molecules of the invention comprise two blunt ends, for example wherein the 3′-end of the antisense strand and the 5′-end of the sense strand as well as the 5′-end of the antisense strand and 3′-end of the sense strand do not have any overhanging nucleotides.
  • In any of the embodiments or aspects of the siNA molecules of the invention, the sense strand and/or the antisense strand may further have a cap, such as described herein or as known in the art, at the 3′-end, the 5′-end, or both of the 3′ and 5′-ends of the sense strand and/or antisense strand. Or as in the case of a hairpin siNA molecule, the cap may be at either one or both of the terminal nucleotides of the polynucleotide. In some embodiments, the cap is at one of both of the ends of the sense strand of a double-stranded siNA molecule. In other embodiments, the cap is at the 3′-end of antisense (guide) strand. In preferred embodiments, the caps are at the 3′-end of the sense strand and the 5′-end of the sense strand.
  • Representative, but non-limiting examples of such terminal caps include an inverted abasic nucleotide, an inverted deoxy abasic nucleotide, an inverted nucleotide moiety, a group shown in the examples and Figures herein, a glyceryl modification, an alkyl or cycloalkyl group, a heterocycle, or any other cap as is generally known in the art.
  • Any of the embodiments of the siNA molecules of the invention may have a 5′ phosphate termini. In some embodiments, the siNA molecules lack terminal phosphates.
  • Applicants describe herein chemically modified siNA molecules that may have improved RNAi activity and/or stability compared to corresponding unmodified siNA molecules. Various chemically modified siNA motifs disclosed herein may provide the capacity to maintain RNAi activity that is substantially similar to unmodified or minimally modified active siRNA (see for example Elbashir et al., 2001, EMBO J., 20:6877-6888) while at the same time providing nuclease resistance and pharmacokinetic properties suitable for use in therapeutic applications.
  • Any of the above described modifications, or combinations thereof, including those in the references cited, can be applied to any of these embodiments.
  • In certain embodiments, siNA molecules having Formula A, B, C, D, E, F, G, H, I, J, K, L, M, N, O, or P have at position 14 from the 5′-end of the antisense strand a ribonucleotide when the nucleotide at that position 14 is a purine.
  • In certain embodiments, siNA molecules having Formula A, B, C, D, E, F, G, H, I, J, K, L, M, N, O, or P have at position 14 from the 5′-end of the antisense strand a ribonucleotide or a 2′-deoxy-2′-fluoro nucleotide when the nucleotide at that position 14 is a purine.
  • In certain embodiments, siNA molecules having Formula A, B, C, D, E, F, G, H, I, J, K, L, M, N, O, or P have at position 14 from the 5′-end of the antisense strand is a 2′-deoxy-2′-fluoro nucleotide when the nucleotide at that position 14 is a pyrimidine nucleotide. In particularly preferred embodiments, position 14 from the 5′-end of the antisense strand is a 2′-deoxy-2′-fluoro nucleotide regardless of whether it is a purine or a pyrimidine.
  • In some embodiments, siNA molecules having Formula A, B, C, D, E, F, G, H, I, J, K, L, M, N, O, or P comprises (N) nucleotides in the antisense strand (lower strand) that are complementary to nucleotides in a target polynucleotide sequence, which also has complementarity to the N and [N] nucleotides of the antisense (lower) strand.
  • Any of the above described modifications, or combinations thereof, discussed above as applicable to siNAs of the invention, including those in the references cited, may be applied to any of the embodiments to siNA molecules having Formula A, B, C, D, E, F, G, H, I, J, K, L, M, N, O, or P.
  • In some embodiments, the compounds disclosed herein (e.g., having Formula A, B, C, D, E, F, G, H, I, J, K, L, M, N, O, or P) include a transduction moiety. Transduction moieties can include, but are not limited to, cell-penetrating peptide (CPP), peptide transduction domain (PTD), nucleic acid binding proteins, such as RNA binding proteins, or any combination thereof. In certain embodiments, any B1, B2, and/or B3 of a compound having any of Formula A, B, C, D, E, F, G, H, I, J, K, L, M, N, O, or P comprises a transduction moiety.
  • Nucleic acid binding proteins, including double-stranded RNA binding domains, can been used to enhance delivery of oligonucleotides into cells. (See, e.g., Eguchi et al. Nat. Biotech. 27:567-571 (2009)). Exemplary nucleic acid binding domains useful in the embodiments disclosed herein include, but are not limited to, those listed in U.S. Patent Application Publication No. US 2009/0093026.
  • Cell-penetrating peptides (CPPs) or peptide transfer domains (PTDs) can be been used successfully to induce intracellular uptake of DNA, antisense oligonucleotides (Astriab-Fisher et al., Pharm. Res. 19:744-54, 2002), small molecules (Polyakov et al. Biocong. Chem. 11:762-71, 2000), and iron particles (Dodd et al. J. Immunol. Methods 256:89-105, 2001; Wunderbaldinger et al., Bioconjug. Chem. 13:264-8, 2002; Lewin et al., Nat. Biotechnol. 18:410-4, 2000; Josephson et al., Bioconjug. Chem. 10:186-91, 1999), suggesting that the size of the cargo is not a limiting factor.
  • The peptides may be conjugated at either end or both ends by addition of a cysteine or other thiol containing moiety to the C- or N-terminus. When not functionalized on the N-terminus, peptides may be capped by an acetyl group, or may be capped with a lipid, a PEG, or a targeting moiety. When the C-terminus of the peptides is unconjugated or unfunctionalized, it may be capped as an amide, or may be capped with a lipid, a PEG, or a targeting moiety. Suitable peptides that can be used in the conjugates discloased herein are listed in Table 2.
  • In some embodiments, a PTD of the invention can be defined as one or more cationic peptides that are able to interact with the cell membrane in a manner that enhances macromolecular uptake. In some embodiments, these peptides can be configured in a linear sequence or attached via a branched linker as is generally known in the art. Alternatively the branching mechanism can be built into the peptide by having multiple cysteines or lysines that are specifically used to form linkages. The methods for synthesizing branched amino acid constructs are well established in the art. In some embodiments, a transduction moiety can be a cell penetrating peptide (CPP), a cationic polymer, an antibody, a cholesterol or cholesterol derivative, a vitamin compound (e.g., vitamin E, B12, B6, etc.), a tocol, a tocotrienol, a tocopherol, glucose, receptor ligand, antibody or the like capable of cell type specific targeting, to further facilitate the uptake of the anionic biomolecule, such as oligonucleotides and polynucleotides.
  • In some embodiments, the PTD domain comprises a peptide represented by the following general formula: B1-X1-X2-X3-B2-X4-X5-B3, wherein B1, B2, and B3 are each independently a basic amino acid, the same or different; and X1, X2, X3, X4 and X5 are each independently an alpha-helix enhancing amino acid, the same or different.
  • In some embodiments, the PTD domain comprises a polypeptide represented by the following general formula: X-X-R-X-(P/X)-(B/X)-B-(P/X)-X-B-(B/X), wherein X is any alpha helical promoting residue such as alanine; P/X is either proline or X as previously defined; B is a basic amino acid residue, e.g., Arginine (Arg) or lysine (Lys); R is Arginine (Arg) and B/X is either B or X as defined herein.
  • In some embodiments, the PTD can be cationic. For example, in some embodiments, the PTD can include between 7 and 10 amino acids and have the general formula K-X1-R-X2-X1 wherein X1 is R or K and X2 is any amino acid. An example of such a cationic polypeptide can include the sequence RKKRRQRRR (SEQ ID NO: 528), or functional fragments and variants thereof.
  • A number of protein transduction domains/peptides are known in the art and may facilitate uptake of heterologous molecules linked to the transduction domains (e.g., cargo molecules). Such peptide transduction domains (PTD's) may facilitate uptake through a process referred to as macropinocytosis. Macropinocytosis is a nonselective form of endocytosis.
  • PTDs and CPPs useful in the embodiments disclosed herein include the PTDs and CPPs described in, for example, Langel, Ulo, “Cell Penetrating Peptides, Processes and Applications,” In Langel, Ulo; (Ed.); Handbook of Cell-Penetrating Peptides, 2.sup.nd Ed (2007); Langel, Ulo, (Ed.). “Cell-Penetrating Peptides, Mechanisms and Applications;” In Curr. Pharm. Des.; 2005, 11(28)(2005); Langel, Ulo, “Cell-Penetrating Peptides: Processes and Applications” (2002); Wadia, Jehangir S.; Becker-Hapak, Michelle; Dowdy, Steven F. Protein transport. Cell-Penetrating Peptides (2002), pp. 365-375.
  • Exemplary peptide transduction domains (PTD's) may be derived from the Drosophila homeoprotein Antennapedia transcription protein (AntHD) (Joliot et al., New Biol. 3:1121-34, 1991; Joliot et al., Proc. Natl. Acad. Sci. USA, 88:1864-8, 1991; Le Roux et al., Proc. Natl. Acad. Sci. USA, 90:9120-4, 1993), the herpes simplex virus structural protein VP22 (Elliott and O'Hare, Cell 88:223-33, 1997), the HIV-1 transcriptional activator TAT protein (Green and Loewenstein, Cell 55:1179-1188, 1988; Frankel and Pabo, Cell 55:1189-1193, 1988), and more recently the cationic N-terminal domain of prion proteins. Other exemplary peptide transduction domains are described in International Patent Application Publication No. WO 08/008,476. The peptide transduction domain may increase uptake of the biomolecule to which it is fused in a receptor independent fashion, may be capable of transducing a wide range of cell types, and may exhibit minimal or no toxicity (Nagahara et al., Nat. Med. 4:1449-52, 1998).
  • In some embodiments, the siNA molecules disclosed herein include PTDs that are cationic in nature. Cationic protein transduction domains track into lipid raft endosomes carrying with them their linked cargo and release their cargo into the cytoplasm by disruption of the endosomal vesicle. The invention provides, in one aspect, methods and compositions that combine the use of PTDs such as TAT and poly-Arg, with a siNA molecule of the invention.
  • In general, protein transduction domains of the embodiments disclosed herein may be any synthetic or naturally-occurring amino acid sequence that can transduce or assist in the transduction of the fusion molecule. For example, transduction may be achieved in accord with the disclosure by use of a nucleic acid construct including phosphodiester and/or phosphorothioate protecting groups and a protein sequence such as an HIV TAT protein or fragment thereof that is linked at the N-terminal or C-terminal end to an oligonucleotide or polynucleotide comprising a phosphodiester and/or phosphorothioate protecting group. In some aspects, the nucleic acid may comprise a phosphodiester and/or phosphorothioate protecting group and may also comprise a nucleic acid binding domain (e.g., a DRBD). The transducing protein domain, for example, may be the Antennapedia homeodomain or the HSV VP22 sequence, the N-terminal fragment of a prion protein or suitable transducing fragments thereof such as those generally known in the art.
  • In some embodiments, the siNA compositions disclosed herein may include a PTD that has substantial alpha-helicity, for example, to optimize transduction of the biomolecule. In another embodiment, the PTD comprises a sequence containing basic amino acid residues that are substantially aligned along at least one face of the peptide. Alpha-helicity can be estimated as is generally known in the art.
  • In some embodiments, with respect to certain siNA compositions described herein, the PTD is as described in PCT Pub. Nos. WO 08/008,476 and WO 07/095,152, the PTD disclosure of which is hereby expressly incorporated by this reference. Additional transducing domains useful in the embodiments disclosed herein include but are not limited to a TAT fragment that comprises at least amino acids 49 to 56 of TAT up to about the full-length TAT sequence as described in PCT Pub. No. WO 08/008,476. In some embodiments, a TAT fragment may include one or more amino acid changes sufficient to increase the alpha-helicity of the fragment. In some embodiments, amino acid changes are introduced in the PTDs that add a recognized alpha-helix enhancing amino acid. In some embodiments, amino acids are introduced in the PTD's that remove one or more amino acids from the TAT fragment that impede alpha helix formation or stability. In some embodiments, for example, the PTD may be a TAT fragment that includes at least one amino acid substitution with an alpha-helix enhancing amino acid.
  • Additional transduction proteins (PTDs) useful in the embodiments disclosed herein include a TAT fragment in which the TAT 49-56 sequence has been modified so that at least two basic amino acids in the sequence are substantially aligned along at least one face of the TAT fragment. Exemplary TAT fragments useful as PTDs in the embodiments disclosed herein may include at least one specified amino acid substitution in at least amino acids 49-56 of TAT which substitution aligns the basic amino acid residues of the 49-56 sequence along at least one face of the segment.
  • In some embodiments, the PTD used in the embodiments disclosed herein may be a naturally occurring PTD, such as include the homeodomain of the Drosophila melanogaster protein Antennapedia (Lindsay (2002) Curr. Op. Pharmacol. 2:587-94; Derossi et al. (1994) J. Biol. Chem. 269:10444-50), HSV-1 VP22 (Bennett et al. (2002) Nat. Biotechnol. 20:20), and Buforin II (Park et al. (2000) Proc. Natl. Acad. Sci. U.S.A. 97:8245-50), or the like, or fragments thereof.
  • In some embodiments, the PTD used in the embodiments disclosed herein may be a recombinant or synthetic PTD designed to mimic and/or enhance the translocating properties of known PTDs, based on consideration of parameters such as electrostatic and hydrophobic properties or secondary structure (Wender et al. (2000) Proc. Natl. Acad. Sci. U.S.A. 97:13003-8; Futaki et al. (2001) J. Biol. Chem. 276:5836-40). An exemplary artificial PTD is transportan (Pooga et al. (1998) FASEB J. 12:67-77; Soomets et al. (2000) Biochim. Biophys. Acta 1467:165-76). Synthetic PTDs such as polylysine, polyarginine, and polyhistidine (which can be positively charged based on the pH of the formulation) e.g., polyarginine (6-15 amino acids) may be useful in the embodiments disclosed herein.
  • Other PTDs useful in the embodiments disclosed herein include, but are not limited to those provided in Table 2 herein.
  • Yet other PTDs useful in the embodiments described herein include PTDs derived from protamine (AAA39985), penetratin (1OMQ_A), TAT (NP057853), pVEC, Cationic prion protein domains, P101 (ACT78456), MATa2 (Q6B184), HIV-1 rev (CAA41586), Polyomavirus Vpl (AAP14004), NF-kappaB (NP003989), M9 (BAA76626), Vpr (BAH97661), FP_NLS (MPG), Sp-NPS (ACU27162), SN50, Importins and Karyopherins, e.g., Karyopherin alpha (NP002255), and Karyopherin beta (NP002256), and the like. Other PTDs useful in the embodiments disclosed herein include those found in International Patent Application Publication No's: WO 09/041,902, WO 05/084158; WO 00/062067, WO 00/034308, and WO 99/55899, each of which is herein incorporated by reference.
  • In some embodiments, the transduction moiety may be a chimeric PTD domain comprising sequences derived from at least two different transducing proteins. For example, chimeric transducing proteins useful in the embodiments disclosed herein include a chimera between two different TAT fragments, e.g., one from HIV-1 and the other from HIV-2 or one from a prion protein and one from HIV. S. Deshayes, M. C. Morris, G. Divita and F. Heitz Cell-penetrating peptides: tools for intracellular delivery of therapeutics 2005, V62, N 16, p 1839.
  • In some embodiments, the transduction moiety may be a nucleic acid binding polypeptide, such as an RNA binding protein, or the like optionally linked to a PTD selected from the examples listed above. Exemplary RNA binding proteins (e.g., DRBD) include histone, RDE-4 protein, or protamine. Exemplary dsRNA binding proteins (with Accession numbers listed in parenthesis) include but are not limited to: PKR (AAA36409, AAA61926, Q03963), TRBP (P97473, AAA36765), PACT (AAC25672, AAA49947, NP609646), Staufen (AAD17531, AAF98119, AAD17529, P25159), NFAR1 (AF167569), NFAR2 (AF167570, AAF31446, AAC71052, AAA19960, AAA19961, AAG22859), SPNR (AAK20832, AAF59924, A57284), RHA (CAA71668, AAC05725, AAF57297), NREBP (AAK07692, AAF23120, AAF54409, T33856), kanadaptin (AAK29177, AAB88191, AAF55582, NP499172, NP198700, BAB19354), HYL1 (NP563850), hyponastic leaves (CAC05659, BAB00641), human rhinovirus polyprotein (ACT09659), ADAR1 (AAB97118, P55266, AAK16102, AAB51687, AF051275), ADAR2P78563, P51400, AAK17102, AAF63702), ADAR3 (AAF78094, AAB41862, AAF76894), TENR (XP059592, CAA59168), RNaseIII (AAF80558, AAF59169, Z81070Q02555/S55784, P05797), and Dicer (BAA78691, AF408401, AAF56056, 544849, AAF03534, Q9884), RDE-4 (AY071926), F1120399 (NP060273, BAB26260), CG1434 (AAF48360, EAA12065, CAA21662), CG13139 (XP059208, XP143416, XP110450, AAF52926, EEA14824), DGCRK6 (BAB83032, XP110167) CG1800 (AAF57175, EAA08039), F1120036 (AAH22270, XP134159), MRP-L45 (BAB14234, XP129893), CG2109 (AAF52025), CG12493 (NP647927), CG10630 (AAF50777), CG17686 (AAD50502), T22A3.5 (CAB03384) and Accession number EAA14308. Nucleic acid binding polypeptides can comprise any of the full length polypeptides of the foregoing accession numbers, as well as fragments or variants thereof, including as modified polypeptides comprising from 1-14 amino acid substitutions.
  • The skilled artisan will readily appreciate that the CPP and PTD domains described herein include modified peptides such as glycoproteins, the L-optical isomer or the D-optical isomer of amino acids or a combination of both, as well as retro-inverso polypeptides. As used herein, the term “retro-inverso” refers a peptide that comprises an amino-carboxy inversion as well as enantiomeric change in one or more amino acids (i.e., levorotatory (L) to dextrorotary (D)). The CPP and PTD domains described herein encompass D-amino acid modified polypeptides, amino-carboxy inversions of the amino acid sequence, amino-carboxy inversions containing one or more D-amino acids, naturally occurring proteins, recombinantly or synthetically synthesized peptides, non-inverted sequence containing one or more D-amino acids, peptidomimetics, Beta-amino acid analogs, gamma amino acid analogs, and the like.
  • The CPP or PTD peptides disclosed herein may also encompass peptide fragments as are generally known in the art.
  • In some embodiments, in addition to including one or more protecting groups disclosed herein, anionic biomolecules disclosed herein may be operably linked to an additional transduction moiety. In some embodiments, the transduction moiety can be a synthetic or non-synthetic, linear or branched peptide transduction domain (PTD) as described herein or as is otherwise known in the art. The PTD can be a cationic peptide optionally connected via a branching linker installed during automated nucleotide synthesis. These linkers have been established and are described by Horn et al., 1989: Chang et al., 1991; Foldesi et al, 1991, M. S. Shchepinov, I. A. Udalova, A. J. Bridgman, and E. M. Southern, Nucleic Acids Res, 1997, 25, 4447-4454, T. Horn, C. A. Chang, and M. S. Urdea, Nucleic Acids Res, 1997, 25, 4842-4849, M. S. Shchepinov, K. U. Mir, J. K. Elder, M. D. Frank-Kamenetskii, and E. M. Southern, Nucleic Acids Res, 1999, 27, 3035-41 The branching linker can be trebler, symmetrical or combinations thereof. The transduction moieties disclosed herein can be linked or fused with another transduction moiety as described herein or as is otherwise known in the art, a linker, such as a peptide linker or a nucleotide linker, or can be directly linked to an siNA molecule disclosed herein. Non-limiting examples of linkers useful in the embodiments disclosed herein include, but are not limited to GG (SEQ ID NO: 529), GGGGS (SEQ ID NO: 530), GGGGSN (SEQ ID NO: 531), GKSSGSGSESKS (SEQ ID NO: 532), GSTSGSGKSSEGKG (SEQ ID NO: 533), GSTSGSGKSSEGSGSTKG (SEQ ID NO: 534), GSTSGSGKPGSGEGSTKG (SEQ ID NO: 535), or EGKSSGSGSESKEF (SEQ ID NO: 536) Linking moieties are described, for example, in Huston et al., Proc. Nat'l Acad. Sci. 85:5879, 1988; Whitlow et al., Protein Engineering 6:989, 1993; and Newton et al., Biochemistry 35:545, 1996. Other suitable peptide linkers are those described in U.S. Pat. Nos. 4,751,180 and 4,935,233, which are hereby incorporated by reference.
  • In some embodiments, the compositions disclosed herein comprise targeting moieties and the like.
  • For example, in some embodiments, two or more transduction moieties, such as PTDs (e.g., 1-5, 2-4, typically 3) may be linked in series or separated by one or more other domains (e.g., a nucleic acid domain or peptide linkers). Transduction moieties, siNA molecules of the invention, and peptide linkers, may be organized in nearly any fashion provided that the construct has the function for which it was intended. Each of several domains (e.g., transduction moieties and siNA molecules) may be directly linked or can be separated by a linker peptide. The domains may be presented in any order. Additionally, the fusion polypeptides may include tags, e.g., to facilitate identification and/or purification of the fusion polypeptide, such as a 6xHIS tag, a maltose binding protein domain, a GST tag, or the like.
  • In some embodiments, the siNA compositions described herein include a peptide linker. For example, in some embodiments, a peptide linker comprises up to about 20 or 30 amino acids, commonly up to about 10 or 15 amino acids, and still more often from about 1 to 5 amino acids, e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, or more amino acids. In some embodiments, the amino acid sequence of the linker is engineered to be flexible so as not to hold the fusion molecule in a single rigid conformation. Peptide linker sequences can be used, e.g., to space the transduction moieties from the siNA molecules of the invention. For example, the peptide linker sequence can be positioned between a transduction moiety, and one or more strands of a siNA molecule, e.g., to provide molecular flexibility. The length of the linker moiety is chosen to optimize the biological activity of the polypeptide comprising a PTD domain fusion construct and can be determined empirically without undue experimentation. The linker moiety should be long enough and flexible enough to allow a nucleic acid binding domain to freely interact with a nucleic acid or vice versa. Exemplary peptide linkers and linker moieties are described in Int. Pub. No. WO/2008/008476, in Huston et al., Proc. Natl. Acad. Sci. 85:5879, 1988; Whitlow et al., Protein Engineering 6:989, 1993; and Newton et al., Biochemistry 35:545, 1996. Other suitable peptide linkers are those described in U.S. Pat. Nos. 4,751,180 and 4,935,233, which are incorporated herein by reference.
  • Generation/Synthesis of siNA Molecules
  • The siNAs of the invention may be obtained using a number of techniques known to those of skill in the art. For example the siNA may be chemically synthesized or may be encoded by plasmid (e.g., transcribed as sequences that automatically fold into duplexes with hairpin loops.). siNA may also be generated by cleavage of longer dsRNA (e.g., dsRNA greater than about 25 nucleotides in length) by the E. coli RNase II or Dicer. These enzymes process the dsRNA into biologically active siNA (see, e.g., Yang et al., PNAS USA 99:9942-9947 (2002); Calegari et al. PNAS USA 99:14236 (2002) Byron et al. Ambion Tech Notes; 10 (1):4-6 (2009); Kawaski et al., Nucleic Acids Res., 31:981-987 (2003), Knight and Bass, Science, 293:2269-2271 (2001) and Roberston et al., J. Biol. Chem 243:82(1969).
  • Preferably, siNA of the invention are chemically synthesized. Oligonucleotides (e.g., certain modified oligonucleotides or portions of oligonucleotides lacking ribonucleotides) are synthesized using protocols known in the art, for example as described in Caruthers et al., 1992, Methods in Enzymology 211, 3-19, Thompson et al., International PCT Publication No. WO 99/54459, Wincott et al., 1995, Nucleic Acids Res. 23, 2677-2684, Wincott et al., 1997, Methods Mol. Bio., 74, 59, Brennan et al., 1998, Biotechnol Bioeng., 61, 33-45, and Brennan, U.S. Pat. No. 6,001,311. The synthesis of oligonucleotides makes use of common nucleic acid protecting and coupling groups, such as dimethoxytrityl at the 5′-end, and phosphoramidites at the 3′-end.
  • siNA molecules without modifications may be synthesized using procedures as described in Usman et al., 1987, J. Am. Chem. Soc., 109, 7845; Scaringe et al., 1990, Nucleic Acids Res., 18, 5433. These syntheses makes use of common nucleic acid protecting and coupling groups, such as dimethoxytrityl at the 5′-end, and phosphoramidites at the 3′-end that can be used for certain siNA molecules of the invention.
  • In certain embodiments, the siNA molecules of the invention may be synthesized, deprotected, and analyzed according to methods described in U.S. Pat. Nos. 6,995,259, 6,686,463, 6,673,918, 6,649,751, 6,989,442, and U.S. patent application Ser. No. 10/190,359.
  • Alternatively, the siNA molecules of the present invention may be synthesized separately and joined together post-synthetically, for example, by ligation (Moore et al., 1992, Science 256, 9923; Draper et al., International PCT Publication No. WO 93/23569; Shabarova et al., 1991, Nucleic Acids Research 19, 4247; Bellon et al., 1997, Nucleosides & Nucleotides, 16, 951; Bellon et al., 1997, Bioconjugate Chem. 8, 204), or by hybridization following synthesis and/or deprotection.
  • Various siNA molecules of the invention may also be synthesized using the teachings of Scaringe et al., U.S. Pat. Nos. 5,889,136; 6,008,400; and 6,111,086.
  • Carrier/Delivery Systems
  • The siNA molecules of the invention may be added directly or may be conjugated with a delivery vehicle, or otherwise delivered to target cells or tissues. In certain embodiments, the invention features conjugates and/or complexes of siNA molecules of the invention. Such conjugates and/or complexes include ligand based and polymer based delivery modalities that may be used to facilitate delivery of siNA molecules into a biological system, such as a cell. The conjugates and complexes provided by the instant invention may impart therapeutic activity by transferring therapeutic compounds across cellular membranes, altering the pharmacokinetics, and/or modulating the localization of nucleic acid molecules of the invention. Non-limiting, examples of such conjugates are described in U.S. Publication Nos. US2008/0152661 A1 and US 2004/0162260 A1 (e.g., CDM-LBA, CDM-Pip-LBA, CDM-PEG, CDM-NAG, etc.) and U.S. patent application Ser. Nos. 10/427,160 10/201,394, 61/322,422, 61/378,609, and 61/315,223; and U.S. Pat. Nos. 6,528,631; 6,335,434; 6,235,886; 6,153,737; 5,214,136; and 5,138,045.
  • In various embodiments, polyethylene glycol (PEG) may be covalently attached to siNA compounds of the present invention. The attached PEG may be any molecular weight, preferably from about 100 to about 50,000 daltons (Da).
  • Kits
  • The present invention also provides nucleic acids in kit form. The kit may comprise a container. The kit typically contains a nucleic acid of the invention with instructions for its administration. In certain instances, the nucleic acids may have a targeting moiety or delivery agent attached. Methods of attaching targeting moieties (e.g. antibodies, proteins) or delivery agents (conjugates) are known to those of skill in the art. In certain instances, the kit contains more than one siNA molecule of the invention. The kits may comprise an siNA molecule of the invention with a pharmaceutically acceptable carrier or diluent. The kits may further comprise excipients.
  • Therapeutic Uses/Pharmaceutical Compositions
  • The nucleic acid molecules and pharmaceutical compositions of the invention may be used to treat diseases, conditions, or phenotypes related to gene expression. Non-limiting examples of such diseases, conditions, and phenotypes are described herein and are otherwise known in the art
  • Indications
  • Particular conditions and disease states that may be associated with gene expression modulation include, but are not limited to cancer, proliferative, inflammatory, autoimmune, neurologic, ocular, respiratory, metabolic, dermatological, auditory, liver, kidney, infectious etc. diseases, conditions, or disorders as described herein or otherwise known in the art, and any other diseases, conditions or disorders that may be related to or may respond to the levels of a target (e.g., target DNA, RNA, protein or polynucleotide) in a cell or tissue, alone or in combination with other therapies.
  • Proliferative diseases (cancer) may include any disease or condition characterized by unregulated cell growth or replication as is known in the art; including leukemias, for example, acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), acute lymphocytic leukemia (ALL), and chronic lymphocytic leukemia, AIDS related cancers such as Kaposi's sarcoma; breast cancers; bone cancers such as Osteosarcoma, Chondrosarcomas, Ewing's sarcoma, Fibrosarcomas, Giant cell tumors, Adamantinomas, and Chordomas; Brain cancers such as Meningiomas, Glioblastomas, Lower-Grade Astrocytomas, Oligodendrocytomas, Pituitary Tumors, Schwannomas, and Metastatic brain cancers; cancers of the head and neck including various lymphomas such as mantle cell lymphoma, non-Hodgkins lymphoma, adenoma, squamous cell carcinoma, laryngeal carcinoma, gallbladder and bile duct cancers, cancers of the retina such as retinoblastoma, cancers of the esophagus, gastric cancers, multiple myeloma, ovarian cancer, uterine cancer, thyroid cancer, testicular cancer, endometrial cancer, melanoma, colorectal cancer, lung cancer, bladder cancer, prostate cancer, lung cancer (including non-small cell lung carcinoma), pancreatic cancer, sarcomas, Wilms' tumor, cervical cancer, head and neck cancer, skin cancers, nasopharyngeal carcinoma, liposarcoma, epithelial carcinoma, renal cell carcinoma, gallbladder adeno carcinoma, parotid adenocarcinoma, endometrial sarcoma, multidrug resistant cancers; and proliferative diseases and conditions, such as neovascularization associated with tumor angiogenesis, macular degeneration (e.g., wet/dry AMD), corneal neovascularization, diabetic retinopathy, neovascular glaucoma, myopic degeneration and other proliferative diseases and conditions such as restenosis and polycystic kidney disease, and any other cancer or proliferative disease, condition, trait, genotype or phenotype that can respond to the modulation of disease related gene expression in a cell or tissue, alone or in combination with other therapies.
  • Inflammatory diseases may include any disease or condition characterized by an inflammatory or allergic process as is known in the art, such as inflammation, acute inflammation, chronic inflammation, respiratory disease, atherosclerosis, psoriasis, dermatitis, restenosis, asthma, allergic rhinitis, atopic dermatitis, septic shock, rheumatoid arthritis, inflammatory bowl disease, inflammatory pelvic disease, pain, ocular inflammatory disease, celiac disease, Leigh Syndrome, Glycerol Kinase Deficiency, Familial eosinophilia (FE), autosomal recessive spastic ataxia, laryngeal inflammatory disease; Tuberculosis, Chronic cholecystitis, Bronchiectasis, Silicosis and other pneumoconioses, and any other inflammatory disease, condition, trait, genotype or phenotype that can respond to the modulation of disease related gene expression in a cell or tissue, alone or in combination with other therapies.
  • Autoimmune diseases may include any disease or condition characterized by autoimmunity as is known in the art, such as multiple sclerosis, diabetes mellitus, lupus, celiac disease, Crohn's disease, ulcerative colitis, Guillain-Barre syndrome, scleroderms, Goodpasture's syndrome, Wegener's granulomatosis, autoimmune epilepsy, Rasmussen's encephalitis, Primary biliary sclerosis, Sclerosing cholangitis, Autoimmune hepatitis, Addison's disease, Hashimoto's thyroiditis, Fibromyalgia, Menier's syndrome; transplantation rejection (e.g., prevention of allograft rejection) pernicious anemia, rheumatoid arthritis, systemic lupus erythematosus, dermatomyositis, Sjogren's syndrome, lupus erythematosus, multiple sclerosis, myasthenia gravis, Reiter's syndrome, Grave's disease, and any other autoimmune disease, condition, trait, genotype or phenotype that can respond to the modulation of disease related gene expression in a cell or tissue, alone or in combination with other therapies.
  • Infectious diseases may include any disease or condition associated with an infectious agent, such as a virus, bacteria, fungus, prion, or parasite. Non-limiting examples of various viral genes that can be targeted using siNA molecules of the invention include Hepatitis C Virus (HCV, for example GenBank Accession Nos: D11168, D50483.1, L38318 and S82227), Hepatitis B Virus (HBV, for example GenBank Accession No. AF100308.1), Human Immunodeficiency Virus type 1 (HIV-1, for example GenBank Accession No. U51188), Human Immunodeficiency Virus type 2 (HIV-2, for example GenBank Accession No. X60667), West Nile Virus (WNV for example GenBank accession No. NC001563), cytomegalovirus (CMV for example GenBank Accession No. NC001347), respiratory syncytial virus (RSV for example GenBank Accession No. NC001781), influenza virus (for example GenBank Accession No. AF037412, rhinovirus (for example, GenBank accession numbers: D00239, X02316, X01087, L24917, M16248, K02121, X01087), papillomavirus (for example GenBank Accession No. NC001353), Herpes Simplex Virus (HSV for example GenBank Accession No. NC001345), and other viruses such as HTLV (for example GenBank Accession No. AJ430458). Due to the high sequence variability of many viral genomes, selection of siNA molecules for broad therapeutic applications would likely involve the conserved regions of the viral genome. Nonlimiting examples of conserved regions of the viral genomes include but are not limited to 5′-Non Coding Regions (NCR), 3′-Non Coding Regions (NCR) and/or internal ribosome entry sites (IRES). siNA molecules designed against conserved regions of various viral genomes will enable efficient inhibition of viral replication in diverse patient populations and may ensure the effectiveness of the siNA molecules against viral quasi species which evolve due to mutations in the non-conserved regions of the viral genome. Non-limiting examples of bacterial infections include Actinomycosis, Anthrax, Aspergillosis, Bacteremia, Bacterial Infections and Mycoses, Bartonella Infections, Botulism, Brucellosis, Burkholderia Infections, Campylobacter Infections, Candidiasis, Cat-Scratch Disease, Chlamydia Infections, Cholera, Clostridium Infections, Coccidioidomycosis, Cross Infection, Cryptococcosis, Dermatomycoses, Dermatomycoses, Diphtheria, Ehrlichiosis, Escherichia coli Infections, Fasciitis, Necrotizing, Fusobacterium Infections, Gas Gangrene, Gram-Negative Bacterial Infections, Gram-Positive Bacterial Infections, Histoplasmosis, Impetigo, Klebsiella Infections, Legionellosis, Leprosy, Leptospirosis, Listeria Infections, Lyme Disease, Maduromycosis, Melioidosis, Mycobacterium Infections, Mycoplasma Infections, Mycoses, Nocardia Infections, Onychomycosis, Ornithosis, Plague, Pneumococcal Infections, Pseudomonas Infections, Q Fever, Rat-Bite Fever, Relapsing Fever, Rheumatic Fever, Rickettsia Infections, Rocky Mountain Spotted Fever, Salmonella Infections, Scarlet Fever, Scrub Typhus, Sepsis, Sexually Transmitted Diseases—Bacterial, Bacterial Skin Diseases, Staphylococcal Infections, Streptococcal Infections, Tetanus, Tick-Borne Diseases, Tuberculosis, Tularemia, Typhoid Fever, Typhus, Epidemic Louse-Borne, Vibrio Infections, Yaws, Yersinia Infections, Zoonoses, and Zygomycosis. Non-limiting examples of fungal infections include Aspergillosis, Blastomycosis, Coccidioidomycosis, Cryptococcosis, Fungal Infections of Fingernails and Toenails, Fungal Sinusitis, Histoplasmosis, Histoplasmosis, Mucormycosis, Nail Fungal Infection, Paracoccidioidomycosis, Sporotrichosis, Valley Fever (Coccidioidomycosis), and Mold Allergy.
  • Neurologic diseases may include any disease or condition affecting the central or peripheral nervous system, including ADHD, AIDS—Neurological Complications, Absence of the Septum Pellucidum, Acquired Epileptiform Aphasia, Acute Disseminated Encephalomyelitis, Adrenoleukodystrophy, Agenesis of the Corpus Callosum, Agnosia, Aicardi Syndrome, Alexander Disease, Alpers' Disease, Alternating Hemiplegia, Alzheimer's Disease, Amyotrophic Lateral Sclerosis, Anencephaly, Aneurysm, Angelman Syndrome, Angiomatosis, Anoxia, Aphasia, Apraxia, Arachnoid Cysts, Arachnoiditis, Arnold-Chiari Malformation, Arteriovenous Malformation, Aspartame, Asperger Syndrome, Ataxia Telangiectasia, Ataxia, Attention Deficit-Hyperactivity Disorder, Autism, Autonomic Dysfunction, Back Pain, Barth Syndrome, Batten Disease, Behcet's Disease, Bell's Palsy, Benign Essential Blepharospasm, Benign Focal Amyotrophy, Benign Intracranial Hypertension, Bernhardt-Roth Syndrome, Binswanger's Disease, Blepharospasm, Bloch-Sulzberger Syndrome, Brachial Plexus Birth Injuries, Brachial Plexus Injuries, Bradbury-Eggleston Syndrome, Brain Aneurysm, Brain Injury, Brain and Spinal Tumors, Brown-Sequard Syndrome, Bulbospinal Muscular Atrophy, Canavan Disease, Carpal Tunnel Syndrome, Causalgia, Cavernomas, Cavernous Angioma, Cavernous Malformation, Central Cervical Cord Syndrome, Central Cord Syndrome, Central Pain Syndrome, Cephalic Disorders, Cerebellar Degeneration, Cerebellar Hypoplasia, Cerebral Aneurysm, Cerebral Arteriosclerosis, Cerebral Atrophy, Cerebral Beriberi, Cerebral Gigantism, Cerebral Hypoxia, Cerebral Palsy, Cerebro-Oculo-Facio-Skeletal Syndrome, Charcot-Marie-Tooth Disorder, Chiari Malformation, Chorea, Choreoacanthocytosis, Chronic Inflammatory Demyelinating Polyneuropathy (CIDP), Chronic Orthostatic Intolerance, Chronic Pain, Cockayne Syndrome Type II, Coffin Lowry Syndrome, Coma, including Persistent Vegetative State, Complex Regional Pain Syndrome, Congenital Facial Diplegia, Congenital Myasthenia, Congenital Myopathy, Congenital Vascular Cavernous Malformations, Corticobasal Degeneration, Cranial Arteritis, Craniosynostosis, Creutzfeldt-Jakob Disease, Cumulative Trauma Disorders, Cushing's Syndrome, Cytomegalic Inclusion Body Disease (CIBD), Cytomegalovirus Infection, Dancing Eyes-Dancing Feet Syndrome, Dandy-Walker Syndrome, Dawson Disease, De Morsier's Syndrome, Dejerine-Klumpke Palsy, Dementia—Multi-Infarct, Dementia—Subcortical, Dementia With Lewy Bodies, Dermatomyositis, Developmental Dyspraxia, Devic's Syndrome, Diabetic Neuropathy, Diffuse Sclerosis, Dravet's Syndrome, Dysautonomia, Dysgraphia, Dyslexia, Dysphagia, Dyspraxia, Dystonias, Early Infantile Epileptic Encephalopathy, Empty Sella Syndrome, Encephalitis Lethargica, Encephalitis and Meningitis, Encephaloceles, Encephalopathy, Encephalotrigeminal Angiomatosis, Epilepsy, Erb's Palsy, Erb-Duchenne and Dejerine-Klumpke Palsies, Fabry's Disease, Fahr's Syndrome, Fainting, Familial Dysautonomia, Familial Hemangioma, Familial Idiopathic Basal Ganglia Calcification, Familial Spastic Paralysis, Febrile Seizures (e.g., GEFS and GEFS plus), Fisher Syndrome, Floppy Infant Syndrome, Friedreich's Ataxia, Gaucher's Disease, Gerstmann's Syndrome, Gerstmann-Straussler-Scheinker Disease, Giant Cell Arteritis, Giant Cell Inclusion Disease, Globoid Cell Leukodystrophy, Glossopharyngeal Neuralgia, Guillain-Barre Syndrome, HTLV-1 Associated Myelopathy, Hallervorden-Spatz Disease, Head Injury, Headache, Hemicrania Continua, Hemifacial Spasm, Hemiplegia Alterans, Hereditary Neuropathies, Hereditary Spastic Paraplegia, Heredopathia Atactica Polyneuritiformis, Herpes Zoster Oticus, Herpes Zoster, Hirayama Syndrome, Holoprosencephaly, Huntington's Disease, Hydranencephaly, Hydrocephalus—Normal Pressure, Hydrocephalus, Hydromyelia, Hypercortisolism, Hypersomnia, Hypertonia, Hypotonia, Hypoxia, Immune-Mediated Encephalomyelitis, Inclusion Body Myositis, Incontinentia Pigmenti, Infantile Hypotonia, Infantile Phytanic Acid Storage Disease, Infantile Refsum Disease, Infantile Spasms, Inflammatory Myopathy, Intestinal Lipodystrophy, Intracranial Cysts, Intracranial Hypertension, Isaac's Syndrome, Joubert Syndrome, Kearns-Sayre Syndrome, Kennedy's Disease, Kinsbourne syndrome, Kleine-Levin syndrome, Klippel Feil Syndrome, Klippel-Trenaunay Syndrome (KTS), Kliiver-Bucy Syndrome, Korsakoffs Amnesic Syndrome, Krabbe Disease, Kugelberg-Welander Disease, Kuru, Lambert-Eaton Myasthenic Syndrome, Landau-Kleffner Syndrome, Lateral Femoral Cutaneous Nerve Entrapment, Lateral Medullary Syndrome, Learning Disabilities, Leigh's Disease, Lennox-Gastaut Syndrome, Lesch-Nyhan Syndrome, Leukodystrophy, Levine-Critchley Syndrome, Lewy Body Dementia, Lissencephaly, Locked-In Syndrome, Lou Gehrig's Disease, Lupus—Neurological Sequelae, Lyme Disease—Neurological Complications, Machado-Joseph Disease, Macrencephaly, Megalencephaly, Melkersson-Rosenthal Syndrome, Meningitis, Menkes Disease, Meralgia Paresthetica, Metachromatic Leukodystrophy, Microcephaly, Migraine, Miller Fisher Syndrome, Mini-Strokes, Mitochondrial Myopathies, Mobius Syndrome, Monomelic Amyotrophy, Motor Neuron Diseases, Moyamoya Disease, Mucolipidoses, Mucopolysaccharidoses, Multi-Infarct Dementia, Multifocal Motor Neuropathy, Multiple Sclerosis, Multiple System Atrophy with Orthostatic Hypotension, Multiple System Atrophy, Muscular Dystrophy, Myasthenia—Congenital, Myasthenia Gravis, Myelinoclastic Diffuse Sclerosis, Myoclonic Encephalopathy of Infants, Myoclonus, Myopathy—Congenital, Myopathy—Thyrotoxic, Myopathy, Myotonia Congenita, Myotonia, Narcolepsy, Neuroacanthocytosis, Neurodegeneration with Brain Iron Accumulation, Neurofibromatosis, Neuroleptic Malignant Syndrome, Neurological Complications of AIDS, Neurological Manifestations of Pompe Disease, Neuromyelitis Optica, Neuromyotonia, Neuronal Ceroid Lipofuscinosis, Neuronal Migration Disorders, Neuropathy—Hereditary, Neurosarcoidosis, Neurotoxicity, Nevus Cavernosus, Niemann-Pick Disease, O'Sullivan-McLeod Syndrome, Occipital Neuralgia, Occult Spinal Dysraphism Sequence, Ohtahara Syndrome, Olivopontocerebellar Atrophy, Opsoclonus Myoclonus, Orthostatic Hypotension, Overuse Syndrome, Pain—Chronic, Paraneoplastic Syndromes, Paresthesia, Parkinson's Disease, Parmyotonia Congenita, Paroxysmal Choreoathetosis, Paroxysmal Hemicrania, Parry-Romberg, Pelizaeus-Merzbacher Disease, Pena Shokeir II Syndrome, Perineural Cysts, Periodic Paralyses, Peripheral Neuropathy, Periventricular Leukomalacia, Persistent Vegetative State, Pervasive Developmental Disorders, Phytanic Acid Storage Disease, Pick's Disease, Piriformis Syndrome, Pituitary Tumors, Polymyositis, Pompe Disease, Porencephaly, Post-Polio Syndrome, Postherpetic Neuralgia, Postinfectious Encephalomyelitis, Postural Hypotension, Postural Orthostatic Tachycardia Syndrome, Postural Tachycardia Syndrome, Primary Lateral Sclerosis, Prion Diseases, Progressive Hemifacial Atrophy, Progressive Locomotor Ataxia, Progressive Multifocal Leukoencephalopathy, Progressive Sclerosing Poliodystrophy, Progressive Supranuclear Palsy, Pseudotumor Cerebri, Pyridoxine Dependent and Pyridoxine Responsive Siezure Disorders, Ramsay Hunt Syndrome Type I, Ramsay Hunt Syndrome Type II, Rasmussen's Encephalitis and other autoimmune epilepsies, Reflex Sympathetic Dystrophy Syndrome, Refsum Disease—Infantile, Refsum Disease, Repetitive Motion Disorders, Repetitive Stress Injuries, Restless Legs Syndrome, Retrovirus-Associated Myelopathy, Rett Syndrome, Reye's Syndrome, Riley-Day Syndrome, SUNCT Headache, Sacral Nerve Root Cysts, Saint Vitus Dance, Salivary Gland Disease, Sandhoff Disease, Schilder's Disease, Schizencephaly, Seizure Disorders, Septo-Optic Dysplasia, Severe Myoclonic Epilepsy of Infancy (SMEI), Shaken Baby Syndrome, Shingles, Shy-Drager Syndrome, Sjogren's Syndrome, Sleep Apnea, Sleeping Sickness, Soto's Syndrome, Spasticity, Spina Bifida, Spinal Cord Infarction, Spinal Cord Injury, Spinal Cord Tumors, Spinal Muscular Atrophy, Spinocerebellar Atrophy, Steele-Richardson-Olszewski Syndrome, Stiff-Person Syndrome, Striatonigral Degeneration, Stroke, Sturge-Weber Syndrome, Subacute Sclerosing Panencephalitis, Subcortical Arteriosclerotic Encephalopathy, Swallowing Disorders, Sydenham Chorea, Syncope, Syphilitic Spinal Sclerosis, Syringohydromyelia, Syringomyelia, Systemic Lupus Erythematosus, Tabes Dorsalis, Tardive Dyskinesia, Tarlov Cysts, Tay-Sachs Disease, Temporal Arteritis, Tethered Spinal Cord Syndrome, Thomsen Disease, Thoracic Outlet Syndrome, Thyrotoxic Myopathy, Tic Douloureux, Todd's Paralysis, Tourette Syndrome, Transient Ischemic Attack, Transmissible Spongiform Encephalopathies, Transverse Myelitis, Traumatic Brain Injury, Tremor, Trigeminal Neuralgia, Tropical Spastic Paraparesis, Tuberous Sclerosis, Vascular Erectile Tumor, Vasculitis including Temporal Arteritis, Von Economo's Disease, Von Hippel-Lindau disease (VHL), Von Recklinghausen's Disease, Wallenberg's Syndrome, Werdnig-Hoffman Disease, Wernicke-Korsakoff Syndrome, West Syndrome, Whipple's Disease, Williams Syndrome, Wilson's Disease, X-Linked Spinal and Bulbar Muscular Atrophy, and Zellweger Syndrome.
  • Respiratory diseases may include any disease or condition affecting the respiratory tract, such as asthma, chronic obstructive pulmonary disease or “COPD”, allergic rhinitis, sinusitis, pulmonary vasoconstriction, inflammation, allergies, impeded respiration, respiratory distress syndrome, cystic fibrosis, pulmonary hypertension, pulmonary vasoconstriction, emphysema, and any other respiratory disease, condition, trait, genotype or phenotype that can respond to the modulation of disease related gene expression in a cell or tissue, alone or in combination with other therapies.
  • Ocular diseases may include any disease or condition affecting eye and related structures as is known in the art, such as Cystoid Macular Edema, Asteroid Hyalosis, Pathological Myopia and Posterior Staphyloma, Toxocariasis (Ocular Larva Migrans), Retinal Vein Occlusion, Posterior Vitreous Detachment, Tractional Retinal Tears, Epiretinal Membrane, Diabetic Retinopathy, Lattice Degeneration, Retinal Vein Occlusion, Retinal Artery Occlusion, Macular Degeneration (e.g., age related macular degeneration such as wet AMD or dry AMD), Toxoplasmosis, Choroidal Melanoma, Acquired Retinoschisis, Hollenhorst Plaque, Idiopathic Central Serous Chorioretinopathy, Macular Hole, Presumed Ocular Histoplasmosis Syndrome, Retinal Macroaneursym, Retinitis Pigmentosa, Retinal Detachment, Hypertensive Retinopathy, Retinal Pigment Epithelium (RPE) Detachment, Papillophlebitis, Ocular Ischemic Syndrome, Coats' Disease, Leber's Miliary Aneurysm, Conjunctival Neoplasms, Allergic Conjunctivitis, Vernal Conjunctivitis, Acute Bacterial Conjunctivitis, Allergic Conjunctivitis &Vernal Keratoconjunctivitis, Viral Conjunctivitis, Bacterial Conjunctivitis, Chlamydial & Gonococcal Conjunctivitis, Conjunctival Laceration, Episcleritis, Scleritis, Pingueculitis, Pterygium, Superior Limbic Keratoconjunctivitis (SLK of Theodore), Toxic Conjunctivitis, Conjunctivitis with Pseudomembrane, Giant Papillary Conjunctivitis, Terrien's Marginal Degeneration, Acanthamoeba Keratitis, Fungal Keratitis, Filamentary Keratitis, Bacterial Keratitis, Keratitis Sicca/Dry Eye Syndrome, Bacterial Keratitis, Herpes Simplex Keratitis, Sterile Corneal Infiltrates, Phlyctenulosis, Corneal Abrasion & Recurrent Corneal Erosion, Corneal Foreign Body, Chemical Burs, Epithelial Basement Membrane Dystrophy (EBMD), Thygeson's Superficial Punctate Keratopathy, Corneal Laceration, Salzmann's Nodular Degeneration, Fuchs' Endothelial Dystrophy, Crystalline Lens Subluxation, Ciliary-Block Glaucoma, Primary Open-Angle Glaucoma, Pigment Dispersion Syndrome and Pigmentary Glaucoma, Pseudoexfoliation Syndrom and Pseudoexfoliative Glaucoma, Anterior Uveitis, Primary Open Angle Glaucoma, Uveitic Glaucoma & Glaucomatocyclitic Crisis, Pigment Dispersion Syndrome & Pigmentary Glaucoma, Acute Angle Closure Glaucoma, Anterior Uveitis, Hyphema, Angle Recession Glaucoma, Lens Induced Glaucoma, Pseudoexfoliation Syndrome and Pseudoexfoliative Glaucoma, Axenfeld-Rieger Syndrome, Neovascular Glaucoma, Pars Planitis, Choroidal Rupture, Duane's Refraction Syndrome, Toxic/Nutritional Optic Neuropathy, Aberrant Regeneration of Cranial Nerve III, Intracranial Mass Lesions, Carotid-Cavernous Sinus Fistula, Anterior Ischemic Optic Neuropathy, Optic Disc Edema & Papilledema, Cranial Nerve III Palsy, Cranial Nerve IV Palsy, Cranial Nerve VI Palsy, Cranial Nerve VII (Facial Nerve) Palsy, Horner's Syndrome, Internuclear Ophthalmoplegia, Optic Nerve Head Hypoplasia, Optic Pit, Tonic Pupil, Optic Nerve Head Drusen, Demyelinating Optic Neuropathy (Optic Neuritis, Retrobulbar Optic Neuritis), Amaurosis Fugax and Transient Ischemic Attack, Pseudotumor Cerebri, Pituitary Adenoma, Molluscum Contagiosum, Canaliculitis, Verruca and Papilloma, Pediculosis and Pthiriasis, Blepharitis, Hordeolum, Preseptal Cellulitis, Chalazion, Basal Cell Carcinoma, Herpes Zoster Ophthalmicus, Pediculosis & Phthiriasis, Blow-out Fracture, Chronic Epiphora, Dacryocystitis, Herpes Simplex Blepharitis, Orbital Cellulitis, Senile Entropion, and Squamous Cell Carcinoma.
  • Dermatologic diseases may include any disease or condition affecting the skin, dermis, or any substructure therein such as hair, follicle, etc. Dermatological diseases, disorders, conditions, and traits can include psoriasis, ectopic dermatitis, skin cancers such as melanoma and basal cell carcinoma, hair loss, hair removal, alterations in pigmentation, and any other disease, condition, or trait associated with the skin, dermis, or structures therein.
  • Auditory diseases may include any disease or condition affecting the auditory system, including the ear, such as the inner ear, middle ear, outer ear, auditory nerve, and any substructures therein. Auditory diseases, disorders, conditions, and traits can include hearing loss, deafness, tinnitus, Meniere's Disease, vertigo, balance and motion disorders, and any other disease, condition, or trait associated with the ear, or structures therein.
  • Metabolic diseases may include any disease or condition affecting metabolic pathways as in known in the art. Metabolic disease can result in an abnormal metabolic process, either congenital due to inherited enzyme abnormality (inborn errors of metabolism) or acquired due to disease of an endocrine organ or failure of a metabolically important organ such as the liver. In one embodiment, metabolic disease includes hyperlipidemia, hypercholesterolemia, cardiovascular disease, atherosclerosis, hypertension, diabetes (e.g., type I and/or type II diabetes), insulin resistance, and/or obesity.
  • Cardiovascular diseases may include any disease or condition affecting the heart and vasculature, including but not limited to, coronary heart disease (CHD), cerebrovascular disease (CVD), aortic stenosis, peripheral vascular disease, atherosclerosis, arteriosclerosis, myocardial infarction (heart attack), cerebrovascular diseases (stroke), transient ischemic attacks (TIA), angina (stable and unstable), atrial fibrillation, arrhythmia, vavular disease, congestive heart failure, hypercholoesterolemia, type I hyperlipoproteinemia, type II hyperlipoproteinemia, type III hyperlipoproteinemia, type IV hyperlipoproteinemia, type V hyperlipoproteinemia, secondary hypertrigliceridemia, and familial lecithin cholesterol acyltransferase deficiency.
  • It is understood that the siNA molecules of the invention may silence the expression of target genes and thus amenable to the treatment of various diseases and conditions herein or otherwise known in the art. Treatment of a disease may be evaluated by directly measuring the progress of the disease in a subject. It may also be inferred through observing a change or reversal in a condition associated with the disease. Additionally, the siNA molecules of the invention may be used as a prophylaxis. Thus, the use of the nucleic acid molecules and pharmaceutical compositions of the invention may be used to ameliorate, treat, prevent, and/or cure these diseases and others associated with gene expression and/or activity.
  • Subjects (e.g., mammalian, human) that may be amendable for treatment using the siNA molecules of the invention (optionally further substituted or modified or conjugated), compositions thereof, and methods of the present disclosure may include those suffering from one or more disease or condition mediated, at least in part, by an aberrant expression level of the target gene or sequence, those at risk of developing a disease caused by or associated with the aberrant levels of a target gene/sequence, or those which may be amenable to treatment by replenishing or increasing the level of RNAi mediated by the corresponding siNA molecule, including a hyperproliferative (e.g., cancer), angiogenic, metabolic, or inflammatory (e.g., arthritis) disease or disorder or condition.
  • Compositions and methods disclosed herein may be useful in the treatment of a wide variety of target viruses, including retrovirus, such as human immunodeficiency virus (HIV), Hepatitis C Virus, Hepatitis B Virus, Coronavirus, as well as respiratory viruses, including human Respiratory Syncytial Virus, human Metapneumovirus, human Parainfluenza virus, Rhinovirus and Influenza virus.
  • In other examples, the compositions and methods of this disclosure may be useful as therapeutic tools to treat or prevent symptoms of, for example, hyperproliferative disorders. Exemplary hyperproliferative disorders may include neoplasms, carcinomas, sarcomas, tumors, or cancer. More exemplary hyperproliferative disorders may include oral cancer, throat cancer, laryngeal cancer, esophageal cancer, pharyngeal cancer, nasopharyngeal cancer, oropharyngeal cancer, gastrointestinal tract cancer, gastrointestinal stromal tumors (GIST), small intestine cancer, colon cancer, rectal cancer, colorectal cancer, anal cancer, pancreatic cancer, breast cancer, cervical cancer, uterine cancer, vulvar cancer, vaginal cancer, urinary tract cancer, bladder cancer, kidney cancer, adrenocortical cancer, islet cell carcinoma, gallbladder cancer, stomach cancer, prostate cancer, ovarian cancer, endometrial cancer, trophoblastic tumor, testicular cancer, penial cancer, bone cancer, osteosarcoma, liver cancer, extrahepatic bile duct cancer, skin cancer, basal cell carcinoma (BCC), lung cancer, small cell lung cancer, non-small cell lung cancer (NSCLC), brain cancer, melanoma, Kaposi's sarcoma, eye cancer, head and neck cancer, squamous cell carcinoma of head and neck, tymoma, thymic carcinoma, thyroid cancer, parathyroid cancer, Hippel-Lindau syndrome, leukemia, acute myeloid leukemia, chronic myelogenous leukemia, acute lymphoblastic leukemia, hairy cell leukemia, lymphoma, non-Hodgkin's lymphoma, Burkitt's lymphoma, T-cell lymphoma, multiple myeloma, malignant pleural mesothelioma, Barrett's adenocarcinoma, Wilm's tumor, or the like. In other examples, the compositions and methods of this disclosure are useful as therapeutic tools to regulate expression of one or more target gene to treat or prevent symptoms of, for example, inflammatory disorders. Exemplary inflammatory disorders may include diabetes mellitus, rheumatoid arthritis, pannus growth in inflamed synovial lining, collagen-induced arthritis, spondylarthritis, ankylosing spondylitis, multiple sclerosis, encephalomyelitis, inflammatory bowel disease, Crohn's disease, psoriasis or psoriatic arthritis, myasthenia gravis, systemic lupus erythematosis, graft-versus-host disease, atherosclerosis, and allergies.
  • Other exemplary disorders that may be treated with the siNA molecules, compositions and methods of the instant disclosure include metabolic disorders, cardiac disease, pulmonary disease, neovascularization, ischemic disorders, age-related macular degeneration, diabetic retinopathy, glomerulonephritis, diabetes, asthma, chronic obstructive pulmonary disease, chronic bronchitis, lymphangiogenesis, and atherosclerosis.
  • Pharmaceutical Compositions
  • The siNA molecules of the instant invention may provide useful reagents and methods for a variety of therapeutic, prophylactic, cosmetic, veterinary, diagnostic, target validation, genomic discovery, genetic engineering, and pharmacogenomic applications.
  • Formulations
  • Thus, the present invention, in one aspect, also provides for pharmaceutical compositions of the siNA molecules of the invention, i.e., compositions in a pharmaceutically acceptable carrier or diluent. These pharmaceutical compositions include salts, esters, or salts of such esters, of the above compounds, e.g., acid addition salts, for example, salts of hydrochloric, hydrobromic, hydroiodic, acetic acid, and benzene sulfonic acid. Other salts include for example, sodium, potassium, manganese, ammonium, and calcium salts. These formulations or compositions can comprise a pharmaceutically acceptable carrier or diluent as is generally known in the art. The pharmaceutical compositions of the present disclosure are formulated to all the siNA molecule(s) described herein to be bioavailable upon administration to a subject.
  • In one embodiment, the invention features a pharmaceutical composition comprising any siNA comprising any formula A, B, C, D, E, F, G, H, I, J, K, L, M, N, O, or P as described herein.
  • The siNA molecules of the invention may preferably be formulated as pharmaceutical compositions prior to administering to a subject, according to techniques known in the art. Pharmaceutical compositions of the present invention may be characterized as being at least sterile and pyrogen-free. Methods for preparing pharmaceutical compositions of the invention are within the skill in the art for example as described in Remington's Pharmaceutical Science, 21st ed., Mack Publishing Company, Easton, Pa., A.R. Gennaro edit., 2005.
  • Non-limiting examples of various types of formulations for local administration include ointments, lotions, creams, gels, foams, preparations for delivery by transdermal patches, powders, sprays, aerosols, capsules or cartridges for use in an inhaler or insufflator or drops (for example eye or nose drops), solutions/suspensions for nebulization, suppositories, pessaries, retention enemas and chewable or suckable tablets or pellets (for example for the treatment of aphthous ulcers) or liposome or microencapsulation preparations.
  • Ointments, creams and gels, may, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agent and/or solvents. Non limiting examples of such bases can thus, for example, include water and/or an oil such as liquid paraffin or a vegetable oil such as arachis oil or castor oil, or a solvent such as polyethylene glycol. Various thickening agents and gelling agents can be used depending on the nature of the base. Non-limiting examples of such agents include soft paraffin, aluminum stearate, cetostearyl alcohol, polyethylene glycols, woolfat, beeswax, carboxypolymethylene and cellulose derivatives, and/or glyceryl monostearate and/or non-ionic emulsifying agents.
  • In one embodiment lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilizing agents, dispersing agents, suspending agents or thickening agents.
  • In one embodiment powders for external application may be formed with the aid of any suitable powder base, for example, talc, lactose or starch. Drops may be formulated with an aqueous or non-aqueous base also comprising one or more dispersing agents, solubilizing agents, suspending agents or preservatives.
  • Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions can contain one or more such sweetening agents, flavoring agents, coloring agents or preservative agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients that are suitable for the manufacture of tablets. These excipients may be, for example, inert diluents; such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia; and lubricating agents, for example magnesium stearate, stearic acid or talc. The tablets may be uncoated or they can be coated by known techniques. In some cases such coatings may be prepared by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monosterate or glyceryl distearate may be employed.
  • Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin or olive oil.
  • Aqueous suspensions contain the active materials in a mixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydropropyl-methylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents may be a naturally-occurring phosphatide, for example, lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate; or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan monooleate. The aqueous suspensions may also contain one or more preservatives, for example ethyl, or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose or saccharin.
  • Oily suspensions may be formulated by suspending the active ingredients in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents and flavoring agents may be added to provide palatable oral preparations. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid
  • Pharmaceutical compositions of the invention may also be in the form of oil-in-water emulsions. The oily phase may be a vegetable oil or a mineral oil or mixtures of these. Suitable emulsifying agents may be naturally-occurring gums, for example gum acacia or gum tragacanth, naturally-occurring phosphatides, for example soy bean, lecithin, and esters or partial esters derived from fatty acids and hexitol, anhydrides, for example sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monooleate. The emulsions may also contain sweetening and flavoring agents.
  • Syrups and elixirs may be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol, glucose or sucrose. Such formulations may also contain a demulcent, a preservative and flavoring and coloring agents. The pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents that have been mentioned above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parentally acceptable diluent or solvent, for example as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables.
  • The nucleic acid molecules of the invention may also be administered in the form of suppositories, e.g., for rectal administration of the drug. These compositions may be prepared by mixing the drug with a suitable non-irritating excipient that is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials include cocoa butter and polyethylene glycols.
  • Nucleic acid molecules of the invention may be administered parenterally in a sterile medium. The drug, depending on the vehicle and concentration used, may either be suspended or dissolved in the vehicle. Advantageously, adjuvants such as local anesthetics, preservatives and buffering agents may be dissolved in the vehicle.
  • In other embodiments, the siNA compositions, or conjugates, and/or delivery formulations provided herein for use in pulmonary delivery further comprise one or more surfactants. Suitable surfactants or surfactant components for enhancing the uptake of the compositions of the invention include synthetic and natural as well as full and truncated forms of surfactant protein A, surfactant protein B, surfactant protein C, surfactant protein D and surfactant Protein E, di-saturated phosphatidylcholine (other than dipalmitoyl), dipalmitoylphosphatidylcholine, phosphatidylcholine, phosphatidylglycerol, phosphatidylinositol, phosphatidylethanolamine, phosphatidylserine; phosphatidic acid, ubiquinones, lysophosphatidylethanolamine, lysophosphatidylcholine, palmitoyl-lysophosphatidylcholine, dehydroepiandrosterone, dolichols, sulfatidic acid, glycerol-3-phosphate, dihydroxyacetone phosphate, glycerol, glycero-3-phosphocholine, dihydroxyacetone, palmitate, cytidine diphosphate (CDP) diacylglycerol, CDP choline, choline, choline phosphate; as well as natural and artificial lamellar bodies which are the natural carrier vehicles for the components of surfactant, omega-3 fatty acids, polyenic acid, polyenoic acid, lecithin, palmitinic acid, non-ionic block copolymers of ethylene or propylene oxides, polyoxypropylene, monomeric and polymeric, polyoxyethylene, monomeric and polymeric, poly (vinyl amine) with dextran and/or alkanoyl side chains, Brij 35, Triton X-100 and synthetic surfactants ALEC, Exosurf, Survan and Atovaquone, among others. These surfactants may be used either as single or part of a multiple component surfactant in a formulation, or as covalently bound additions to the 5′ and/or 3′ ends of the nucleic acid component of a pharmaceutical composition herein.
  • b. Combinations
  • The siNAs and pharmaceutical formulations according to the invention may be administered to a subject alone or used in combination with or include one or more other therapeutic agents, for example, antiviral or anticancer agents. Thus, combinations of the presently disclosed compounds with other antiviral or anti-cancer or chemotherapeutic agents are within the scope of the invention
  • Examples of anti-cancer or chemotherapeutic agents may be found in Cancer Principles and Practice of Oncology by V. T. Devita and S. Hellman (editors), 6th edition (Feb. 15, 2001), Lippincott Williams & Wilkins Publishers. A person of ordinary skill in the art would be able to discern which combinations of agents would be useful based on the particular characteristics of the drugs and the cancer involved. Such anti-cancer agents include, but are not limited to, the following: estrogen receptor modulators, androgen receptor modulators, retinoid receptor modulators, cytotoxic/cytostatic agents, antiproliferative agents, prenyl-protein transferase inhibitors, HMG-CoA reductase inhibitors and other angiogenesis inhibitors, inhibitors of cell proliferation and survival signaling, apoptosis inducing agents and agents that interfere with cell cycle checkpoints.
  • In a further embodiment, therefore, the invention provides a combination comprising an siNA molecule of the invention or a pharmaceutically acceptable salt, solvate or physiologically functional derivative thereof together with one or more therapeutic agents as described herein or as is otherwise known in the art.
  • Examples of estrogen receptor modulators that may be used in combination with the compounds of the invention include, but are not limited to, tamoxifen, raloxifene, idoxifene, LY353381, LY117081, toremifene, fulvestrant, 4-[7-(2,2-dimethyl-1-oxopropoxy-4-methyl-2-[4-[2-(1-piperidinyl)ethoxy]phenyl]-2H-1-benzopyran-3-yl]-phenyl-2,2-dimethylpropanoate, 4,4′-dihydroxybenzophenone-2,4-dinitrophenyl-hydrazone, and SH646.
  • Examples of androgen receptor modulators that may be used in combination with the compounds of the invention include, but are not limited to, finasteride and other 5α-reductase inhibitors, nilutamide, flutamide, bicalutamide, liarozole, and abiraterone acetate.
  • Examples of such retinoid receptor modulators that may be used in combination with the compounds of the invention include, but are not limited to, bexarotene, tretinoin, 13-cis-retinoic acid, 9-cis-retinoic acid, α-difluoromethylornithine, ILX23-7553, trans-N-(4′-hydroxyphenyl) retinamide, and N-4-carboxyphenyl retinamide.
  • Examples of cytotoxic agents that may be used in combination with the compounds of the invention include, but are not limited to, sertenef, cachectin, ifosfamide, tasonermin, lonidamine, carboplatin, altretamine, prednimustine, dibromodulcitol, ranimustine, fotemustine, nedaplatin, oxaliplatin, temozolomide, heptaplatin, estramustine, improsulfan tosilate, trofosfamide, nimustine, dibrospidium chloride, pumitepa, lobaplatin, satraplatin, profiromycin, cisplatin, irofulven, dexifosfamide, cis-aminedichloro(2-methyl-pyridine)platinum, benzylguanine, glufosfamide, GPX100, (trans, trans, trans)-bis-mu-(hexane-1,6-diamine)-mu-[diamine-platinum(II)]bis[diamine(chloro)platinum (II)]tetrachloride, diarizidinylspermine, arsenic trioxide, 1-(11-dodecylamino-10-hydroxyundecyl)-3,7-dimethylxanthine, zorubicin, idarubicin, daunorubicin, bisantrene, mitoxantrone, pirarubicin, pinafide, valrubicin, amrubicin, antineoplaston, 3′-deamino-3′-morpholino-13-deoxo-10-hydroxycarminomycin, annamycin, galarubicin, elinafide, MEN10755, and 4-demethoxy-3-deamino-3-aziridinyl-4-methylsulphonyl-daunorubicin (see WO 00/50032).
  • An example of a hypoxia activatable compound that may be used in combination with the compounds of the invention is tirapazamine.
  • Examples of proteasome inhibitors that may be used in combination with the compounds of the invention include, but are not limited to, lactacystin and bortezomib.
  • Examples of microtubule inhibitors/microtubule-stabilising agents that may be used in combination with the compounds of the invention include, but are not limited to, paclitaxel, vindesine sulfate, 3′,4′-didehydro-4′-deoxy-8′-norvincaleukoblastine, docetaxol, rhizoxin, dolastatin, mivobulin isethionate, auristatin, cemadotin, RPR109881, BMS184476, vinflunine, cryptophycin, 2,3,4,5,6-pentafluoro-N-(3-fluoro-4-methoxyphenyl)benzene sulfonamide, anhydrovinblastine, N,N-dimethyl-L-valyl-L-valyl-N-methyl-L-valyl-L-prolyl-L-proline-t-butylamide, TDX258, the epothilones (see for example U.S. Pat. Nos. 6,284,781 and 6,288,237) and BMS188797.
  • Some examples of topoisomerase inhibitors that may be used in combination with the compounds of the invention include, but are not limited to, are topotecan, hycaptamine, irinotecan, rubitecan, 6-ethoxypropionyl-3′,4′-O-exo-benzylidene-chartreusin, 9-methoxy-N,N-dimethyl-5-nitropyrazolo[3,4,5-kl]acridine-2-(6H) propanamine, 1-amino-9-ethyl-5-fluoro-2,3-dihydro-9-hydroxy-4-methyl-1H,12H-benzo[de]pyrano[3′,4′:b,7]-indolizino[1,2b]quinoline-10,13(9H,15H)dione, lurtotecan, 7-[2-(N-isopropylamino)ethyl]-(20S)camptothecin, BNP1350, BNPI1100, BN80915, BN80942, etoposide phosphate, teniposide, sobuzoxane, 2′-dimethylamino-2′-deoxy-etoposide, GL331, N-[2-(dimethylamino)ethyl]-9-hydroxy-5,6-dimethyl-6H-pyrido[4,3-b]carbazole-1-carboxamide, asulacrine, (5a,5aB,8aa,9b)-9-[2-[N-[2-(dimethylamino)ethyl]-N-methylamino]ethyl]-5-[4-hydroxy-3,5-dimethoxyphenyl]-5,5a,6,8,8a,9-hexohydrofuro(3′,4′:6,7)naphtho(2,3-d)-1,3-dioxol-6-one, 2,3-(methylenedioxy)-5-methyl-7-hydroxy-8-methoxybenzo[c]-phenanthridinium, 6,9-bis[(2-aminoethyl)amino]benzo[g]isoquinoline-5,10-dione, 5-(3-aminopropylamino)-7,10-dihydroxy-2-(2-hydroxyethylaminomethyl)-6H-pyrazolo[4,5,1-de]acridin-6-one, N-[1-[2(diethylamino)ethylamino]-7-methoxy-9-oxo-9H-thioxanthen-4-ylmethyl]formamide, N-(2-(dimethylamino)ethyl)acridine-4-carboxamide, 6-[[2-(dimethylamino)ethyl]amino]-3-hydroxy-7H-indeno[2,1-c]quinolin-7-one, and dimesna.
  • Examples of inhibitors of mitotic kinesins, and in particular the human mitotic kinesin KSP, that may be used in combination with the compounds of the invention include, but are not limited to, inhibitors described in PCT Publications WO 01/30768, WO 01/98278, WO 03/050,064, WO 03/050,122, WO 03/049,527, WO 03/049,679, WO 03/049,678, WO04/039774, WO03/079973, WO03/099211, WO03/105855, WO03/106417, WO04/037171, WO04/058148, WO04/058700, WO04/126699, WO05/018638, WO05/019206, WO05/019205, WO05/018547, WO05/017190, US2005/0176776. In an embodiment inhibitors of mitotic kinesins include, but are not limited to inhibitors of KSP, inhibitors of MKLP1, inhibitors of CENP-E, inhibitors of MCAK, inhibitors of Kif14, inhibitors of Mphosphl and inhibitors of Rab6-KIFL.
  • Examples of “histone deacetylase inhibitors” that may be used in combination with the compounds of the invention include, but are not limited to, TSA, oxamflatin, PXD101, MG98, valproic acid and scriptaid. Further reference to other histone deacetylase inhibitors may be found in the following manuscript; Miller, T. A. et al. J. Med. Chem. 46(24):5097-5116 (2003).
  • Inhibitors of kinases involved in mitotic progression that may be used in combination with the compounds of the invention include, but are not limited to, inhibitors of aurora kinase, inhibitors of Polo-like kinases (PLK) (in particular inhibitors of PLK-1), inhibitors of bub-1 and inhibitors of bub-R1.
  • Antiproliferative agents that may be used in combination with the compounds of the invention include, but are not limited to, antisense RNA and DNA oligonucleotides such as G3139, ODN698, RVASKRAS, GEM231, and INX3001, and antimetabolites such as enocitabine, carmofur, tegafur, pentostatin, doxifluridine, trimetrexate, fludarabine, capecitabine, galocitabine, cytarabine ocfosfate, fosteabine sodium hydrate, raltitrexed, paltitrexid, emitefur, tiazofurin, decitabine, nolatrexed, pemetrexed, nelzarabine, 2′-deoxy-2′-methylidenecytidine, 2′-fluoromethylene-2′-deoxycytidine, N-[5-(2,3-dihydro-benzofuryl)sulfonyl]-N′-(3,4-dichlorophenyl)urea, N6-[4-deoxy-4-[N2-[2(E),4(E)-tetradecadienoyl]glycylamino]-L-glycero-B-L-manno-heptopyranosyl]adenine, aplidine, ecteinascidin, troxacitabine, 4-[2-amino-4-oxo-4,6,7,8-tetrahydro-3H-pyrimidino[5,4-b][1,4]thiazin-6-yl-(S)-ethyl]-2,5-thienoyl-L-glutamic acid, aminopterin, 5-flurouracil, alanosine, 11-acetyl-8-(carbamoyloxymethyl)-4-formyl-6-methoxy-14-oxa-1,11-diazatetracyclo(7.4.1.0.0)-tetradeca-2,4,6-trien-9-yl acetic acid ester, swainsonine, lometrexol, dexrazoxane, methioninase, 2′-cyano-2′-deoxy-N4-palmitoyl-1-B-D-arabin furanosyl cytosine and 3-aminopyridine-2-carboxaldehyde thiosemicarbazone. Examples of monoclonal antibody targeted therapeutic agents that may be used in combination with the compounds of the invention include those therapeutic agents which have cytotoxic agents or radioisotopes attached to a cancer cell specific or target cell specific monoclonal antibody, such as, for example, Bexxar.
  • Examples of HMG-CoA reductase inhibitors that may be used that may be used in combination with the compounds of the invention include, but are not limited to, lovastatin (MEVACOR®; see U.S. Pat. Nos. 4,231,938, 4,294,926 and 4,319,039), simvastatin (ZOCOR®; see U.S. Pat. Nos. 4,444,784, 4,820,850 and 4,916,239), pravastatin (PRAVACHOL®; see U.S. Pat. Nos. 4,346,227, 4,537,859, 4,410,629, 5,030,447 and 5,180,589), fluvastatin (LESCOL®; see U.S. Pat. Nos. 5,354,772, 4,911,165, 4,929,437, 5,189,164, 5,118,853, 5,290,946 and 5,356,896) and atorvastatin (LIPITOR®; see U.S. Pat. Nos. 5,273,995, 4,681,893, 5,489,691 and 5,342,952). The structural formulas of these and additional HMG-CoA reductase inhibitors that may be used in the instant methods are described at page 87 of M. Yalpani, “Cholesterol Lowering Drugs”, Chemistry & Industry, pp. 85-89 (5 Feb. 1996) and U.S. Pat. Nos. 4,782,084 and 4,885,314.
  • Examples of prenyl-protein transferase inhibitors that may be used in combination with the compounds of the invention include, but are not limited to, may be found in the following publications and patents: WO 96/30343, WO 97/18813, WO 97/21701, WO 97/23478, WO 97/38665, WO 98/28980, WO 98/29119, WO 95/32987, U.S. Pat. No. 5,420,245, U.S. Pat. No. 5,523,430, U.S. Pat. No. 5,532,359, U.S. Pat. No. 5,510,510, U.S. Pat. No. 5,589,485, U.S. Pat. No. 5,602,098, European Patent Publ. 0 618 221, European Patent Publ. 0 675 112, European Patent Publ. 0 604 181, European Patent Publ. 0 696 593, WO 94/19357, WO 95/08542, WO 95/11917, WO 95/12612, WO 95/12572, WO 95/10514, U.S. Pat. No. 5,661,152, WO 95/10515, WO 95/10516, WO 95/24612, WO 95/34535, WO 95/25086, WO 96/05529, WO 96/06138, WO 96/06193, WO 96/16443, WO 96/21701, WO 96/21456, WO 96/22278, WO 96/24611, WO 96/24612, WO 96/05168, WO 96/05169, WO 96/00736, U.S. Pat. No. 5,571,792, WO 96/17861, WO 96/33159, WO 96/34850, WO 96/34851, WO 96/30017, WO 96/30018, WO 96/30362, WO 96/30363, WO 96/31111, WO 96/31477, WO 96/31478, WO 96/31501, WO 97/00252, WO 97/03047, WO 97/03050, WO 97/04785, WO 97/02920, WO 97/17070, WO 97/23478, WO 97/26246, WO 97/30053, WO 97/44350, WO 98/02436, and U.S. Pat. No. 5,532,359. For an example of the role of a prenyl-protein transferase inhibitor on angiogenesis see European J. of Cancer, Vol. 35, No. 9, pp. 1394-1401 (1999).
  • Examples of angiogenesis inhibitors that may be used in combination with the compounds of the invention include, but are not limited to, tyrosine kinase inhibitors, such as inhibitors of the tyrosine kinase receptors Flt-1 (VEGFR1) and Flk-1/KDR (VEGFR2), inhibitors of epidermal-derived, fibroblast-derived, or platelet derived growth factors, MMP (matrix metalloprotease) inhibitors, integrin blockers, interferon-α, interleukin-12, pentosan polysulfate, cyclooxygenase inhibitors, including nonsteroidal anti-inflammatories (NSAIDs) like aspirin and ibuprofen as well as selective cyclooxy-genase-2 inhibitors like celecoxib and rofecoxib (PNAS, Vol. 89, p. 7384 (1992); JNCI, Vol. 69, p. 475 (1982); Arch. Opthalmol., Vol. 108, p. 573 (1990); Anat. Rec., Vol. 238, p. 68 (1994); FEBS Letters, Vol. 372, p. 83 (1995); Clin, Orthop. Vol. 313, p. 76 (1995); J. Mol. Endocrinol., Vol. 16, p. 107 (1996); Jpn. J. Pharmacol., Vol. 75, p. 105 (1997); Cancer Res., Vol. 57, p. 1625 (1997); Cell, Vol. 93, p. 705 (1998); Intl. J. Mol. Med., Vol. 2, p. 715 (1998); J. Biol. Chem., Vol. 274, p. 9116 (1999)), steroidal anti-inflammatories (such as corticosteroids, mineralocorticoids, dexamethasone, prednisone, prednisolone, methylpred, betamethasone), carboxyamidotriazole, combretastatin A-4, squalamine, 6-O-chloroacetyl-carbonyl)-fumagillol, thalidomide, angiostatin, troponin-1, angiotensin II antagonists (see Fernandez et al., J. Lab. Clin. Med. 105:141-145 (1985)), and antibodies to VEGF (see, Nature Biotechnology, Vol. 17, pp. 963-968 (October 1999); Kim et al., Nature, 362, 841-844 (1993); WO 00/44777; and WO 00/61186).
  • Other therapeutic agents that modulate or inhibit angiogenesis may also be used in combination with the compounds of the instant invention and include agents that modulate or inhibit the coagulation and fibrinolysis systems (see review in Clin. Chem. La. Med. 38:679-692 (2000)). Examples of such agents that modulate or inhibit the coagulation and fibrinolysis pathways that may be used in combination with the compounds of the invention include, but are not limited to, heparin (see Thromb. Haemost. 80:10-23 (1998)), low molecular weight heparins and carboxypeptidase U inhibitors (also known as inhibitors of active thrombin activatable fibrinolysis inhibitor [TAFIa]) (see Thrombosis Res. 0.101:329-354 (2001)). TAFIa inhibitors have been described in PCT Publication WO 03/013,526 and U.S. Ser. No. 60/349,925 (filed Jan. 18, 2002).
  • Agents that interfere with cell cycle checkpoints that may be used in combination with the compounds of the invention include, but are not limited to, inhibitors of ATR, ATM, the Chk1 and Chk2 kinases and cdk and cdc kinase inhibitors and are specifically exemplified by 7-hydroxystaurosporin, flavopiridol, CYC202 (Cyclacel) and BMS-387032.
  • Agents that interfere with receptor tyrosine kinases (RTKs) that may be used in combination with the compounds of the invention include, but are not limited to, inhibitors of c-Kit, Eph, PDGF, Flt3 and HBV. Further agents include inhibitors of RTKs as described by Bume-Jensen and Hunter, Nature, 411:355-365, 2001.
  • Inhibitors of cell proliferation and survival signaling pathway that may be used in combination with the compounds of the invention include, but are not limited to, inhibitors of EGFR (for example gefitinib and erlotinib), inhibitors of ERB-2 (for example trastuzumab), inhibitors of IGFR, inhibitors of cytokine receptors, inhibitors of HBV, inhibitors of PI3K (for example LY294002), serine/threonine kinases (including but not limited to inhibitors of Akt such as described in WO 02/083064, WO 02/083139, WO 02/083140, US 2004-0116432, WO 02/083138, US 2004-0102360, WO 03/086404, WO 03/086279, WO 03/086394, WO 03/084473, WO 03/086403, WO 2004/041162, WO 2004/096131, WO 2004/096129, WO 2004/096135, WO 2004/096130, WO 2005/100356, WO 2005/100344), inhibitors of Raf kinase (for example BAY-43-9006), inhibitors of MEK (for example CI-1040 and PD-098059) and inhibitors of mTOR (for example Wyeth CCI-779). Such agents include small molecule inhibitor compounds and antibody antagonists.
  • Apoptosis inducing agents that may be used in combination with the compounds of the invention include, but are not limited to, activators of TNF receptor family members (including the TRAIL receptors).
  • NSAIDs that are selective COX-2 inhibitors that may be used in combination with the compounds of the invention include, but are not limited to, those NSAIDs disclosed in U.S. Pat. No. 5,474,995, U.S. Pat. No. 5,861,419, U.S. Pat. No. 6,001,843, U.S. Pat. No. 6,020,343, U.S. Pat. No. 5,409,944, U.S. Pat. No. 5,436,265, U.S. Pat. No. 5,536,752, U.S. Pat. No. 5,550,142, U.S. Pat. No. 5,604,260, U.S. Pat. No. 5,698,584, U.S. Pat. No. 5,710,140, WO 94/15932, U.S. Pat. No. 5,344,991, U.S. Pat. No. 5,134,142, U.S. Pat. No. 5,380,738, U.S. Pat. No. 5,393,790, U.S. Pat. No. 5,466,823, U.S. Pat. No. 5,633,272, and U.S. Pat. No. 5,932,598, all of which are hereby incorporated by reference.
  • Inhibitors of COX-2 that are particularly useful in combination with the compounds of the invention include: 3-phenyl-4-(4-(methylsulfonyl)phenyl)-2-(5H)-furanone; and 5-chloro-3-(4-methylsulfonyl)-phenyl-2-(2-methyl-5-pyridinyl)pyridine; or a pharmaceutically acceptable salt thereof.
  • Compounds that have been described as specific inhibitors of COX-2 and are therefore useful in the present invention include, but are not limited to: parecoxib, CELEBREX® and BEXTRA® or a pharmaceutically acceptable salt thereof.
  • Angiogenesis inhibitors that may be used in combination with the compounds of the invention include, but are not limited to, endostatin, ukrain, ranpirnase, IM862, 5-methoxy-4-[2-methyl-3-(3-methyl-2-butenyl)oxiranyl]-1-oxaspiro[2,5]oct-6-yl(chloroacetyl)carbamate, acetyldinanaline, 5-amino-1-[[3,5-dichloro-4-(4-chlorobenzoyl)-phenyl]methyl]-1H-1,2,3-triazole-4-carboxamide, CM101, squalamine, combretastatin, RPI4610, NX31838, sulfated mannopentaose phosphate, 7,7-(carbonyl-bis[imino-N-methyl-4,2-pyrrolocarbonylimino[N-methyl-4,2-pyrrole]-carbonylimino]-bis-(1,3-naphthalene disulfonate), and 3-[(2,4-dimethylpyrrol-5-yl)methylene]-2-indolinone (SU5416).
  • Tyrosine kinase inhibitors that may be used in combination with the compounds of the invention include, but are not limited to, N-(trifluoromethylphenyl)-5-methylisoxazol-4-carboxamide, 3-[(2,4-dimethylpyrrol-5-yl)methylidenyl)indolin-2-one, 17-(allylamino)-17-demethoxygeldanamycin, 4-(3-chloro-4-fluorophenylamino)-7-methoxy-6-[3-(4-morpholinyl)propoxyl]quinazoline, N-(3-ethynylphenyl)-6,7-bis(2-methoxyethoxy)-4-quinazolinamine, BIBX1382, 2,3,9,10,11,12-hexahydro-10-(hydroxymethyl)-10-hydroxy-9-methyl-9,12-epoxy-1H-diindolo[1,2,3-fg:3′,2′,1′-kl]pyrrolo[3,4-i][1,6]benzodiazocin-1-one, SH268, genistein, imatinib (STI571), CEP2563, 4-(3-chlorophenylamino)-5,6-dimethyl-7H-pyrrolo[2,3-d]pyrimidinemethane sulfonate, 4-(3-bromo-4-hydroxyphenyl)amino-6,7-dimethoxyquinazoline, 4-(4′-hydroxyphenyl)amino-6,7-dimethoxyquinazoline, SU6668, STI571A, N-4-chlorophenyl-4-(4-pyridylmethyl)-1-phthalazinamine, and EMD121974.
  • Combinations with compounds other than anti-cancer compounds are also encompassed in the instant compositions and methods. For example, combinations of the instantly claimed compounds with PPAR-γ (i.e., PPAR-gamma) agonists and PPAR-δ (i.e., PPAR-delta) agonists are useful in the treatment of certain malignancies. PPAR-γ and PPAR-δ are the nuclear peroxisome proliferator-activated receptors y and δ. The expression of PPAR-γ on endothelial cells and its involvement in angiogenesis has been reported in the literature (see J. Cardiovasc. Pharmacol. 31:909-913 (1998); J. Biol. Chem. 274:9116-9121 (1999); Invest. Ophthalmol Vis. Sci. 41:2309-2317 (2000)). More recently, PPAR-γ agonists have been shown to inhibit the angiogenic response to VEGF in vitro; both troglitazone and rosiglitazone maleate inhibit the development of retinal neovascularization in mice. (Arch. Ophthamol. 119:709-717 (2001)). Examples of PPAR-γ agonists and PPAR-γ/α agonists that may be used in combination with the compounds of the invention include, but are not limited to, thiazolidinediones (such as DRF2725, CS-011, troglitazone, rosiglitazone, and pioglitazone), fenofibrate, gemfibrozil, clofibrate, GW2570, SB219994, AR-H039242, JTT-501, MCC-555, GW2331, GW409544, NN2344, KRP297, NP0110, DRF4158, NN622, G1262570, PNU182716, DRF552926, 2-[(5,7-dipropyl-3-trifluoromethyl-1,2-benzisoxazol-6-yl)oxy]-2-methylpropionic acid (disclosed in U.S. Ser. No. 09/782,856), and 2(R)-7-(3-(2-chloro-4-(4-fluorophenoxy) phenoxy)propoxy)-2-ethylchromane-2-carboxylic acid (disclosed in U.S. Ser. No. 60/235,708 and 60/244,697).
  • Another embodiment of the instant invention is the use of the presently disclosed compounds in combination with gene therapy for the treatment of cancer. For an overview of genetic strategies to treating cancer see Hall et al. (Am J Hum Genet 61:785-789 (1997)) and Kufe et al. (Cancer Medicine, 5th Ed, pp 876-889, BC Decker, Hamilton, 2000). Gene therapy may be used to deliver any tumor suppressing gene. Examples of such genes include, but are not limited to, p53, which may be delivered via recombinant virus-mediated gene transfer (see U.S. Pat. No. 6,069,134, for example), a uPA/uPAR antagonist (“Adenovirus-Mediated Delivery of a uPA/uPAR Antagonist Suppresses Angiogenesis-Dependent Tumor Growth and Dissemination in Mice,” Gene Therapy, August 5(8):1105-13 (1998)), and interferon gamma (J Immunol 164:217-222 (2000)).
  • The compounds of the instant invention may also be administered in combination with an inhibitor of inherent multidrug resistance (MDR), in particular MDR associated with high levels of expression of transporter proteins. Such MDR inhibitors include inhibitors of p-glycoprotein (P-gp), such as LY335979, XR9576, OC144-093, R101922, VX853 and PSC833 (valspodar).
  • A compound of the present invention may be employed in conjunction with anti-emetic agents to treat nausea or emesis, including acute, delayed, late-phase, and anticipatory emesis, which may result from the use of a compound of the present invention, alone or with radiation therapy. For the prevention or treatment of emesis, a compound of the present invention may be used in conjunction with other anti-emetic agents, especially neurokinin-1 receptor antagonists, 5HT3 receptor antagonists, such as ondansetron, granisetron, tropisetron, and zatisetron, GABAB receptor agonists, such as baclofen, a corticosteroid such as Decadron (dexamethasone), Kenalog, Aristocort, Nasalide, Preferid, Benecorten or others such as disclosed in U.S. Pat. Nos. 2,789,118, 2,990,401, 3,048,581, 3,126,375, 3,929,768, 3,996,359, 3,928,326 and 3,749,712, an antidopaminergic, such as the phenothiazines (for example prochlorperazine, fluphenazine, thioridazine and mesoridazine), metoclopramide or dronabinol. In an embodiment, an anti-emesis agent selected from a neurokinin-1 receptor antagonist, a 5HT3 receptor antagonist and a corticosteroid is administered as an adjuvant for the treatment or prevention of emesis that may result upon administration of the instant compounds.
  • Neurokinin-1 receptor antagonists of use in conjunction with the compounds of the present invention are fully described, for example, in U.S. Pat. Nos. 5,162,339, 5,232,929, 5,242,930, 5,373,003, 5,387,595, 5,459,270, 5,494,926, 5,496,833, 5,637,699, 5,719,147; European Patent Publication Nos. EP 0 360 390, 0 394 989, 0 428 434, 0 429 366, 0 430 771, 0 436 334, 0 443 132, 0 482 539, 0 498 069, 0 499 313, 0 512 901, 0 512 902, 0 514 273, 0 514 274, 0 514 275, 0 514 276, 0 515 681, 0 517 589, 0 520 555, 0 522 808, 0 528 495, 0 532 456, 0 533 280, 0 536 817, 0 545 478, 0 558 156, 0 577 394, 0 585 913, 0 590 152, 0 599 538, 0 610 793, 0 634 402, 0 686 629, 0 693 489, 0 694 535, 0 699 655, 0 699 674, 0 707 006, 0 708 101, 0 709 375, 0 709 376, 0 714 891, 0 723 959, 0 733 632 and 0 776 893; PCT International Patent Publication Nos. WO 90/05525, 90/05729, 91/09844, 91/18899, 92/01688, 92/06079, 92/12151, 92/15585, 92/17449, 92/20661, 92/20676, 92/21677, 92/22569, 93/00330, 93/00331, 93/01159, 93/01165, 93/01169, 93/01170, 93/06099, 93/09116, 93/10073, 93/14084, 93/14113, 93/18023, 93/19064, 93/21155, 93/21181, 93/23380, 93/24465, 94/00440, 94/01402, 94/02461, 94/02595, 94/03429, 94/03445, 94/04494, 94/04496, 94/05625, 94/07843, 94/08997, 94/10165, 94/10167, 94/10168, 94/10170, 94/11368, 94/13639, 94/13663, 94/14767, 94/15903, 94/19320, 94/19323, 94/20500, 94/26735, 94/26740, 94/29309, 95/02595, 95/04040, 95/04042, 95/06645, 95/07886, 95/07908, 95/08549, 95/11880, 95/14017, 95/15311, 95/16679, 95/17382, 95/18124, 95/18129, 95/19344, 95/20575, 95/21819, 95/22525, 95/23798, 95/26338, 95/28418, 95/30674, 95/30687, 95/33744, 96/05181, 96/05193, 96/05203, 96/06094, 96/07649, 96/10562, 96/16939, 96/18643, 96/20197, 96/21661, 96/29304, 96/29317, 96/29326, 96/29328, 96/31214, 96/32385, 96/37489, 97/01553, 97/01554, 97/03066, 97/08144, 97/14671, 97/17362, 97/18206, 97/19084, 97/19942 and 97/21702; and in British Patent Publication Nos. 2 266 529, 2 268 931, 2 269 170, 2 269 590, 2 271 774, 2 292 144, 2 293 168, 2 293 169, and 2 302 689. The preparation of such compounds is fully described in the aforementioned patents and publications, which are incorporated herein by reference.
  • In an embodiment, the neurokinin-1 receptor antagonist for use in conjunction with the compounds of the present invention is selected from: 2-(R)-(1-(R)-(3,5-bis(trifluoromethyl)-phenyl)ethoxy)-3-(S)-(4-fluorophenyl)-4-(3-(5-oxo-1H,4H-1,2,4-triazolo)methyl)morpholine, or a pharmaceutically acceptable salt thereof, which is described in U.S. Pat. No. 5,719,147.
  • A compound of the instant invention may also be administered with an agent useful in the treatment of anemia. Such an anemia treatment agent is, for example, a continuous eythropoiesis receptor activator (such as epoetin alfa).
  • A compound of the instant invention may also be administered with an agent useful in the treatment of neutropenia. Such a neutropenia treatment agent is, for example, a hematopoietic growth factor which regulates the production and function of neutrophils such as a human granulocyte colony stimulating factor, (G-CSF). Examples of a G-CSF include filgrastim and PEG-filgrastim.
  • A compound of the instant invention may also be administered with an immunologic-enhancing drug, such as levamisole, isoprinosine and Zadaxin.
  • A compound of the instant invention may also be useful for treating or preventing liver disease or cancer in combination with other siNA therapeutics.
  • The compounds of the instant invention may also be administered in combination with γ-secretase inhibitors and/or inhibitors of NOTCH signaling. Such inhibitors include compounds described in WO 01/90084, WO 02/30912, WO 01/70677, WO 03/013506, WO 02/36555, WO 03/093252, WO 03/093264, WO 03/093251, WO 03/093253, WO 2004/039800, WO 2004/039370, WO 2005/030731, WO 2005/014553, U.S. Ser. No. 10/957,251, WO 2004/089911, WO 02/081435, WO 02/081433, WO 03/018543, WO 2004/031137, WO 2004/031139, WO 2004/031138, WO 2004/101538, WO 2004/101539 and WO 02/47671 (including LY-450139).
  • A compound of the instant invention may also be useful for treating or preventing cancer in combination with PARP inhibitors.
  • A compound of the instant invention may also be useful for treating cancer in combination with the following therapeutic agents: abarelix (Plenaxis Depot®); aldesleukin (Prokine®); Aldesleukin (Proleukin®); Alemtuzumabb (Campath®); alitretinoin (Panretin®); allopurinol (Zyloprim®); altretamine (Hexalen®); amifostine (Ethyol®); anastrozole (Arimidex®); arsenic trioxide (Trisenox®); asparaginase (Elspar®); azacitidine (Vidaza®); bendamustine hydrochloride (Treanda®); bevacuzimab (Avastin®); bexarotene capsules (Targretin®); bexarotene gel (Targretin®); bleomycin (Blenoxane®); bortezomib (Velcade®); brefeldin A; busulfan intravenous (Busulfex®); busulfan oral (Myleran®); calusterone (Methosarb®); capecitabine (Xeloda®); carboplatin (Paraplatin®); carmustine (BCNU®, BiCNU®); carmustine (Gliadel®); carmustine with Polifeprosan 20 Implant (Gliadel Wafer®); celecoxib (Celebrex®); cetuximab (Erbitux®); chlorambucil (Leukeran®); cisplatin (Platinol®); cladribine (Leustatin®, 2-CdA®; clofarabine (Clolar®); cyclophosphamide (Cytoxan®, Neosar®); cyclophosphamide (Cytoxan Injection®); cyclophosphamide (Cytoxan Tablet®); cytarabine (Cytosar-U®); cytarabine liposomal (DepoCyt®); dacarbazine (DTIC-Dome®); dactinomycin, actinomycin D (Cosmegen®); dalteparin sodium injection (Fragmin®); Darbepoetin alfa (Aranesp®); dasatinib (Sprycel®); daunorubicin liposomal (DanuoXome®); daunorubicin, daunomycin (Daunorubicin®); daunorubicin, daunomycin (Cerubidine®); degarelix (Firmagon®); Denileukin diftitox (Ontak®); dexrazoxane (Zinecard®); dexrazoxane hydrochloride (Totect®); didemnin B; 17-DMAG; docetaxel (Taxotere®); doxorubicin (Adriamycin PFS®); doxorubicin (Adriamycin®, Rubex®); doxorubicin (Adriamycin PFS Injection®); doxorubicin liposomal (Doxil®); dromostanolone propionate (Dromostanolone®); dromostanolone propionate (Masterone Injection®); eculizumab injection (Soliris®); Elliott's B Solution (Elliott's B Solution®); eltrombopag (Promacta®); epirubicin (Ellence®); Epoetin alfa (Epogen®); erlotinib (Tarceva®); estramustine (Emcyt®); ethinyl estradiol; etoposide phosphate (Etopophos®); etoposide, VP-16 (Vepesid®); everolimus tablets (Afinitor®); exemestane (Aromasin®); ferumoxytol (Feraheme Injection®); Filgrastim (Neupogen®); floxuridine (intraarterial) (FUDR®); fludarabine (Fludara®); fluorouracil, 5-FU (Adrucil®); fulvestrant (Faslodex®); gefitinib (Iressa®); geldanamycin; gemcitabine (Gemzar®); gemtuzumab ozogamicin (Mylotarg®); goserelin acetate (Zoladex Implant®); goserelin acetate (Zoladex®); histrelin acetate (Histrelin Implant®); hydroxyurea (Hydrea®); Ibritumomab Tiuxetan (Zevalin®); idarubicin (Idamycin®); ifosfamide (IFEX®); imatinib mesylate (Gleevec®); interferon alfa 2a (Roferon A®); Interferon alfa-2b (Intron A®); iobenguane I 123 injection (AdreView®); irinotecan (Camptosar®); ixabepilone (Ixempra®); lapatinib tablets (Tykerb®); lenalidomide (Revlimid®); letrozole (Femara®); leucovorin (Wellcovorin®, Leucovorin®); Leuprolide Acetate (Eligard®); levamisole (Ergamisol®); lomustine, CCNU (CeeBU®); meclorethamine, nitrogen mustard (Mustargen®); megestrol acetate (Megace®); melphalan, L-PAM (Alkeran®); mercaptopurine, 6-MP (Purinethol®); mesna (Mesnex®); mesna (Mesnex Tabs®); methotrexate (Methotrexate®); methoxsalen (Uvadex®); 8-methoxypsoralen; mitomycin C (Mutamycin®); mitotane (Lysodren®); mitoxantrone (Novantrone®); mitramycin; nandrolone phenpropionate (Durabolin-50®); nelarabine (Arranon®); nilotinib (Tasigna®); Nofetumomab (Verluma®); ofatumumab (Arzerra®); Oprelvekin (Neumega®); oxaliplatin (Eloxatin®); paclitaxel (Paxene®); paclitaxel (Taxol®); paclitaxel protein-bound particles (Abraxane®); palifermin (Kepivance®); pamidronate (Aredia®); panitumumab (Vectibix®); pazopanib tablets (Votrienttm®); pegademase (Adagen (Pegademase Bovine)®); pegaspargase (Oncaspar®); Pegfilgrastim (Neulasta®); pemetrexed disodium (Alimta®); pentostatin (Nipent®); pipobroman (Vercyte®); plerixafor (Mozobil®); plicamycin, mithramycin (Mithracin®); porfimer sodium (Photofrin®); pralatrexate injection (Folotyn®); procarbazine (Matulane®); quinacrine (Atabrine®); rapamycin; Rasburicase (Elitek®); raloxifene hydrochloride (Evista®); Rituximab (Rituxan®); romidepsin (Istodax®); romiplostim (Nplate®); sargramostim (Leukine®); Sargramostim (Prokine®); sorafenib (Nexavar®); streptozocin (Zanosar®); sunitinib maleate (Sutent®); talc (Sclerosol®); tamoxifen (Nolvadex®); temozolomide (Temodar®); temsirolimus (Torisel®); teniposide, VM-26 (Vumon®); testolactone (Teslac®); thioguanine, 6-TG (Thioguanine®); thiopurine; thiotepa (Thioplex®); topotecan (Hycamtin®); toremifene (Fareston®); Tositumomab (Bexxar®); Tositumomab/I-131 tositumomab (Bexxar®); trans-retinoic acid; Trastuzumab (Herceptin®); tretinoin, ATRA (Vesanoid®); triethylenemelamine; Uracil Mustard (Uracil Mustard Capsules®); valrubicin (Valstar®); vinblastine (Velban®); vincristine (Oncovin®); vinorelbine (Navelbine®); vorinostat (Zolinza®); wortmannin; and zoledronate (Zometa®).
  • The invention also provides a combination comprising an siNA molecule of the invention targeting one gene together with another inhibitor targeting a second target gene.
  • The combinations referred to above may conveniently be presented for use in the form of a pharmaceutical formulation and thus pharmaceutical compositions comprising a combination as defined above together with a pharmaceutically acceptable diluent or carrier represent a further aspect of the invention.
  • To practice the coordinate administration methods of this disclosure, an siNA molecule is administered simultaneously or sequentially in a coordinated treatment protocol with one or more secondary or adjunctive therapeutic agents described herein or known in the art. The coordinate administration may be done in either order, and there may be a time period while only one or both (or all) active therapeutic agents, individually or collectively, exert their biological activities. A distinguishing aspect of all such coordinate treatment methods is that the siNA molecule(s) present in a composition elicits some favorable clinical response, which may or may not be in conjunction with a secondary clinical response provided by the secondary therapeutic agent. For example, the coordinate administration of an siNA molecule with a secondary therapeutic agent as contemplated herein may yield an enhanced (e.g., synergistic) therapeutic response beyond the therapeutic response elicited by either or both the purified siNA molecule and the secondary therapeutic agent alone.
  • The individual compounds of such combinations may be administered either sequentially or simultaneously in separate or combined pharmaceutical formulations. In one embodiment, the individual compounds will be administered simultaneously in a combined pharmaceutical formulation.
  • Thus, the described molecules could be used in combination with one or more known compounds, treatments, or procedures to prevent or treat diseases, disorders, conditions, and traits described herein in a subject or organism as are known in the art, such as other gene inhibitors.
  • Therapeutic Applications
  • The present body of knowledge in RNAi research indicates the need for methods that can modulate gene expression for therapeutic use.
  • Thus, one aspect of the invention comprises a method of treating a subject including, but not limited to, a human suffering from a disease or a condition which is mediated by the action of target gene expression, which method comprises administering to said subject an effective amount of a double-stranded siNA molecule of the invention. In one embodiment of this aspect, the siNA molecules comprises sequence having at least a 15 nucleotides complementary to a target nucleic acid. In other embodiments, the siNA molecule comprises any molecule herein having formula A, B, C, D, E, F, G, H, I, J, K, L, M, N, O or P.
  • In some embodiments of this aspect, the disease or condition may be cancer, a proliferative, inflammatory, autoimmune, neurologic, ocular, respiratory, metabolic, dermatological, auditory, liver, kidney, or infectious disease as described herein or otherwise known in the art. Thus, in certain embodiments the molecules and compositions of the instant invention may be useful in a method for treating cancer, proliferative, inflammatory, autoimmune, neurologic, ocular, respiratory, metabolic, dermatological, auditory, liver, kidney, or infectious diseases.
  • In certain embodiments, the administration of the siNA molecule may be via local administration or systemic administration. In other embodiments, the invention features contacting the subject or organism with an siNA molecule of the invention via local administration to relevant tissues or cells, such as lung cells and tissues, such as via pulmonary delivery. In yet other embodiments, the invention features contacting the subject or organism with an siNA molecule of the invention via systemic administration (such as via intravenous or subcutaneous administration of siNA) to relevant tissues or cells in a subject or organism.
  • siNA molecules of the invention may also used as reagents in ex vivo applications. For example, siNA reagents may be introduced into tissue or cells that are transplanted into a subject for therapeutic effect. The cells and/or tissue may be derived from an organism or subject that later receives the explant, or may be derived from another organism or subject prior to transplantation. The siNA molecules may be used to modulate the expression of one or more genes in the cells or tissue, such that the cells or tissue obtain a desired phenotype or are able to perform a function when transplanted in vivo. In one embodiment, certain target cells from a patient are extracted. These extracted cells are contacted with siNAs targeting a specific nucleotide sequence within the cells under conditions suitable for uptake of the siNAs by these cells (e.g., using delivery reagents such as cationic lipids, liposomes and the like or using techniques such as electroporation to facilitate the delivery of siNAs into cells). The cells are then reintroduced back into the same patient or other patients.
  • For therapeutic applications, a pharmaceutically effective dose of the siNA molecules or pharmaceutical compositions of the invention is administered to the subject. A pharmaceutically effective dose is that dose required to prevent, inhibit the occurrence, or treat (alleviate a symptom to some extent, preferably all of the symptoms) a disease state. One skilled in the art may readily determine a therapeutically effective dose of the siNA of the invention to be administered to a given subject, by taking into account factors, such as the size and weight of the subject, the extent of the disease progression or penetration, the age, health, and sex of the subject, the route of administration, and whether the administration is regional or systemic. Generally, an amount between 0.1 μg/kg and 140 mg/kg body weight/day of active ingredients is administered dependent upon potency of the siNA of the disclosure. The amount of active ingredient that may be combined with the carrier materials to produce a single dosage form varies depending upon the host treated and the particular mode of administration. Optimal dosing schedules may be calculated from measurements of drug accumulation in the body of the patient. The siNA molecules of the invention may be administered in a single dose or in multiple doses.
  • siNA molecules of the instant invention may be administered once monthly, once weekly, once daily (QD), or divided into multiple monthly, weekly, or daily doses, such as, for example, but not limitation, twice daily (BID), three times daily (TID), once every two weeks. Persons of ordinary skill in the art may easily estimate repetition rates for dosing based on measured residence times and concentrations of the drug in bodily fluids or tissues.
  • In addition, the administration may be continuous, i.e., every day, or intermittently. For example, intermittent administration of a compound of the instant invention may be administration one to six days per week or it may mean administration in cycles (e.g. daily administration for two to eight consecutive weeks, then a rest period with no administration for up to one week) or it may mean administration on alternate days.
  • Administration
  • Compositions or formulations may be administered in a variety of ways. Non-limiting examples of administration methods of the invention include oral, buccal, sublingual, parenteral (i.e., intraarticularly, intravenously, intraperitoneally, subcutaneously, or intramuscularly), local rectal administration or other local administration. In one embodiment, the composition of the invention may be administered by insufflation and inhalation. Administration may be accomplished via single or divided doses. In some embodiments, the pharmaceutical compositions are administered intravenously or intraperitoneally by a bolus injection (see, e.g., U.S. Pat. No. 5,286,634).
  • An siNA molecule with or without a vehicle may be locally delivered by direct injection or by use of an infusion pump. Direct injection of the siNA molecules of this disclosure, whether subcutaneous, intramuscular, or intradermal, may take place using standard needle and syringe methodologies, or by needle free technologies, such as those described in Conroy et al, (1999, Clin. Cancer Res. 5:2330) and PCT Publication No. WO 99/31262. For example, but not limitation, lipid nucleic acid particles may be administered by direct injection at the site of disease or by injection at a site distal from the site of disease (see, e.g., Culver, HUMAN GENE THERAPY, MaryAnn Liebert, Inc., Publishers, New York. pp. 70-71(1994)). In one embodiment, the siNA molecules of the invention and formulations or compositions thereof are administered to a cell, subject, or organism as is described herein and as is generally known in the art.
  • In Vivo Administration
  • In any of the methods of treatment of the invention, the siNA may be administered to the subject systemically as described herein or otherwise known in the art, either alone as a monotherapy or in combination with additional therapies described herein or as are known in the art. Systemic administration may include, for example, pulmonary (inhalation, nebulization etc.) intravenous, subcutaneous, intramuscular, catheterization, nasopharyngeal, transdermal, or oral/gastrointestinal administration as is generally known in the art.
  • In any of the methods of treatment or prevention of the invention, the siNA may be administered to the subject locally or to local tissues as described herein or otherwise known in the art, either alone as a monotherapy or in combination with additional therapies as are known in the art. Local administration may include, for example, inhalation, nebulization, catheterization, implantation, direct injection, dermal/transdermal application, patches, stenting, ear/eye drops, or portal vein administration to relevant tissues, or any other local administration technique, method or procedure, as is generally known in the art.
  • In one embodiment, the siNA molecules of the invention and formulations or compositions thereof are administered to the liver as is generally known in the art (see for example Wen et al., 2004, World J Gastroenterol., 10, 244-9; Murao et al., 2002, Pharm Res., 19, 1808-14; Liu et al., 2003, gene Ther., 10, 180-7; Hong et al., 2003, J Pharm Pharmacol., 54, 51-8; Herrmann et al., 2004, Arch Virol., 149, 1611-7; and Matsuno et al., 2003, gene Ther., 10, 1559-66).
  • In one embodiment, the invention features the use of methods to deliver the siNA molecules of the instant invention to hematopoietic cells, including monocytes and lymphocytes. These methods are described in detail by Hartmann et al., 1998, J. Phamacol. Exp. Ther., 285(2), 920-928; Kronenwett et al., 1998, Blood, 91(3), 852-862; Filion and Phillips, 1997, Biochim. Biophys. Acta., 1329(2), 345-356; Ma and Wei, 1996, Leuk. Res., 20(11/12), 925-930; and Bongartz et al., 1994, Nucleic Acids Research, 22(22), 4681-8.
  • In one embodiment, the siNA molecules of the invention and formulations or compositions thereof are administered directly or topically (e.g., locally) to the dermis or follicles as is generally known in the art (see for example Brand, 2001, Curr. Opin. Mol. Ther., 3, 244-8; Regnier et al., 1998, J. Drug Target, 5, 275-89; Kanikkannan, 2002, BioDrugs, 16, 339-47; Wraight et al., 2001, Pharmacol. Ther., 90, 89-104; and Preat and Dujardin, 2001, STP PharmaSciences, 11, 57-68). In one embodiment, the siNA molecules of the invention and formulations or compositions thereof are administered directly or topically using a hydroalcoholic gel formulation comprising an alcohol (e.g., ethanol or isopropanol), water, and optionally including additional agents such isopropyl myristate and carbomer 980. In other embodiments, the siNA are formulated to be administered topically to the nasal cavity. Topical preparations may be administered by one or more applications per day to the affected area; over skin areas occlusive dressings may advantageously be used. Continuous or prolonged delivery may be achieved by an adhesive reservoir system.
  • In one embodiment, an siNA molecule of the invention is administered iontophoretically, for example to a particular organ or compartment (e.g., the eye, back of the eye, heart, liver, kidney, bladder, prostate, tumor, CNS etc.). Non-limiting examples of iontophoretic delivery are described in, for example, WO 03/043689 and WO 03/030989, which are incorporated by reference in their entireties herein.
  • In one embodiment, the siNA molecules of the invention and formulations or compositions thereof are administered to the lung as is described herein and as is generally known in the art. In another embodiment, the siNA molecules of the invention and formulations or compositions thereof are administered to lung tissues and cells as is described in U.S. Patent Publication Nos. 2006/0062758; 2006/0014289; and 2004/0077540.
  • Aerosols and Delivery Devices
  • a. Aerosol Formulations
  • The compositions of the present invention, either alone or in combination with other suitable components, may be made into aerosol formulations (i.e., they may be “nebulized”) to be administered via inhalation (e.g., intranasally or intratracheally) (see, Brigham et al., Am. J. Sci., 298:278 (1989)). Aerosol formulations may be placed into pressurized acceptable propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like.
  • In one embodiment, the siNA molecules of the invention and formulations thereof are administered via pulmonary delivery, such as by inhalation of an aerosol or spray dried formulation administered by an inhalation device or nebulizer, providing rapid local uptake of the nucleic acid molecules into relevant pulmonary tissues. Solid particulate compositions containing respirable dry particles of micronized nucleic acid compositions may be prepared by grinding dried or lyophilized nucleic acid compositions, and then passing the micronized composition through, for example, a 400 mesh screen to break up or separate out large agglomerates. A solid particulate composition comprising the siNA compositions of the invention may optionally contain a dispersant which serves to facilitate the formation of an aerosol as well as other therapeutic compounds. A suitable dispersant is lactose, which may be blended with the nucleic acid compound in any suitable ratio, such as a 1 to 1 ratio by weight.
  • Spray compositions comprising siNA molecules or compositions of the invention may, for example, be formulated as aqueous solutions or suspensions or as aerosols delivered from pressurized packs, such as a metered dose inhaler, with the use of a suitable liquefied propellant. In one embodiment, aerosol compositions of the invention suitable for inhalation may be either a suspension or a solution and generally contain an siNA molecule comprising formula (A), and a suitable propellant such as a fluorocarbon or hydrogen-containing chlorofluorocarbon or mixtures thereof, particularly hydrofluoroalkanes, especially 1,1,1,2-tetrafluoroethane, 1,1,1,2,3,3,3-heptafluoro-n-propane or a mixture thereof. The aerosol composition may optionally contain additional formulation excipients well known in the art such as surfactants. Non-limiting examples include oleic acid, lecithin or an oligolactic acid or derivative such as those described in WO94/21229 and WO98/34596 and co-solvents for example ethanol. In one embodiment a pharmaceutical aerosol formulation of the invention comprising a compound of the invention and a fluorocarbon or hydrogen-containing chlorofluorocarbon or mixtures thereof as propellant, optionally in combination with a surfactant and/or a co-solvent.
  • The aerosol formulations of the invention may be buffered by the addition of suitable buffering agents.
  • Aerosol formulations may include optional additives including preservatives if the formulation is not prepared sterile. Non-limiting examples include, methyl hydroxybenzoate, anti-oxidants, flavorings, volatile oils, buffering agents and emulsifiers and other formulation surfactants. In one embodiment, fluorocarbon or perfluorocarbon carriers are used to reduce degradation and provide safer biocompatible non-liquid particulate suspension compositions of the invention (e.g., siNA and/or LNP formulations thereof). In another embodiment, a device comprising a nebulizer delivers a composition of the invention (e.g., siNA and/or LNP formulations thereof) comprising fluorochemicals that are bacteriostatic thereby decreasing the potential for microbial growth in compatible devices.
  • Capsules and cartridges comprising the composition of the invention for use in an inhaler or insufflator, of for example gelatin, may be formulated containing a powder mix for inhalation of a compound of the invention and a suitable powder base such as lactose or starch. In one embodiment, each capsule or cartridge contains an siNA molecule comprising formula (A), and one or more excipients. In another embodiment, the compound of the invention may be presented without excipients such as lactose
  • The aerosol compositions of the present invention may be administered into the respiratory system as a formulation including particles of respirable size, e.g. particles of a size sufficiently small to pass through the nose, mouth and larynx upon inhalation and through the bronchi and alveoli of the lungs. In general, respirable particles range from about 0.5 to 10 microns in size. In one embodiment, the particulate range may be from 1 to 5 microns. In another embodiment, the particulate range may be from 2 to 3 microns. Particles of non-respirable size which are included in the aerosol tend to deposit in the throat and be swallowed, and the quantity of non-respirable particles in the aerosol is thus minimized. For nasal administration, a particle size in the range of 10-500 um is preferred to ensure retention in the nasal cavity.
  • In some embodiments, an siNA composition of the invention is administered topically to the nose for example, for the treatment of rhinitis, via pressurized aerosol formulations, aqueous formulations administered to the nose by pressurized pump or by nebulization. Suitable formulations contain water as the diluent or carrier for this purpose. In certain embodiments, the aqueous formulations for administration of the composition of the invention to the lung or nose may be provided with conventional excipients such as buffering agents, tonicity modifying agents and the like.
  • b. Devices
  • The siNA molecules of the invention may be formulated and delivered as particles and/or aerosols as discussed above and dispensed from various aerosolization devices known by those of skill in the art.
  • Aerosols of liquid or non-liquid particles comprising an siNA molecule or formulation of the invention may be produced by any suitable means, such as with a device comprising a nebulizer (see for example U.S. Pat. No. 4,501,729) such as ultrasonic or air jet nebulizers.
  • Solid particle aerosols comprising an siNA molecule or formulation of the invention and surfactant may be produced with any solid particulate aerosol generator. One type of solid particle aerosol generator used with the siNA molecules of the invention is an insufflator. A second type of illustrative aerosol generator comprises a metered dose inhaler (“MDI”). MDIs containing siNA molecules or formulations taught herein may be prepared by methods of the art (for example, see Byron, above and WO96/32099).
  • The siNA molecules may also be formulated as a fluid formulation for delivery from a fluid dispenser, such as those described and illustrated in WO05/044354.
  • In certain embodiments of the invention, nebulizer devices are used in applications for conscious, spontaneously breathing subjects, and for controlled ventilated subjects of all ages. The nebulizer devices may be used for targeted topical and systemic drug delivery to the lung. In one embodiment, a device comprising a nebulizer is used to deliver an siNA molecule or formulation of the invention locally to lung or pulmonary tissues. In another embodiment, a device comprising a nebulizer is used to deliver a an siNA molecule or formulation of the invention systemically.
  • Other Applications/Uses of siNA Molecules of the Invention
  • The siNA molecules of the invention may also be used for diagnostic applications, research applications, and/or manufacture of medicants.
  • In one aspect, the invention features a method for diagnosing a disease, trait, or condition in a subject comprising administering to the subject a composition of the invention under conditions suitable for the diagnosis of the disease, trait, or condition in the subject.
  • In one embodiment, siNA molecules of the invention may be used to down regulate or inhibit the expression of proteins arising from haplotype polymorphisms that are associated with a trait, disease or condition in a subject or organism. Analysis of genes, or protein or RNA levels may be used to identify subjects with such polymorphisms or those subjects who are at risk of developing traits, conditions, or diseases described herein. These subjects may be amenable to treatment, for example, treatment with siNA molecules of the invention and any other composition useful in treating diseases related to target gene expression. As such, analysis of protein or RNA levels may be used to determine treatment type and the course of therapy in treating a subject. Monitoring of protein or RNA levels may be used to predict treatment outcome and to determine the efficacy of compounds and compositions that modulate the level and/or activity of certain peptides and/or proteins associated with a trait, disorder, condition, or disease.
  • In another embodiment, the invention comprises use of a double-stranded nucleic acid according to the invention for use in the manufacture of a medicament. In an embodiment, the medicament is for use in treating a disease or a condition that is mediated by the action of one or more target genes. In one embodiment, the medicant is for use in treating any disease or condition herein or otherwise known in the art. In some embodiments, the medicament is for use in the treatment of cancer.
  • In certain embodiments, the siNA molecules of the invention may be for use in a method for treating any disease or condition contemplated herein.
  • In certain embodiments, the siNA molecules of the invention may be used in a method for treating cancer.
  • EXAMPLES
  • The invention will now be illustrated with the following non-limiting examples. Those of skill in the art will readily recognize a variety of non-critical parameters which can be changed or modified to yield essentially the same results.
  • Example 1 Identification of Highly Potent Stabilized siNA Molecules with Prolonged Duration
  • The development of therapeutic siRNAs with drug-like properties is dependent on the incorporation of chemical modifications to improve duration of RNA knockdown and potency while minimizing non-specific (off-target) effects and stimulation of innate immunity. The growing diversity of siRNA delivery vehicles has expanded beyond traditional lipid encapsulated nano-particles to include dynamic polymer conjugates and conjugation with various targeting ligands including antibodies, sugars, and cholesterol. Many of these delivery strategies expose the siRNA cargo to serum and/or cellular nucleases which can compromise the structural integrity and in vivo efficacy of both unmodified siRNAs and modified siRNAs which have not been optimized for nuclease stability. Additionally, these alternative delivery strategies employ different targeting ligands and mechanisms of endosomal escape, thereby exposing the siRNA cargo to different cellular micro-environments with varied pH and nuclease content. Therefore optimization of siRNA stability is needed for development of therapeutic siRNAs with cross-delivery platform compatibility and potentially enhanced duration due to improvement of stability attributes along with pharmacokinetic and pharmacodynamic profiles of the siRNA molecule.
  • The modified siNA molecules disclosed herein may improve serum stability while maintaining robust potency. The modification criteria of the present invention may be applied to any siRNA sequence.
  • Traditional lipid nanoparticle (LNP) delivery vehicles encapsulate the siRNA and thus limit exposure to serum nucleases upon intravenous dosing of siRNA-LNP complexes in animals. The adoption of alternative delivery platforms such as polymer conjugates (PC) or direct attachment of delivery targeting ligands (e.g., galactosamine, folate, cholesterol, etc.) exposes the siRNA molecule to serum nucleases and potentially hostile intracellular environments. Therefore the development of a stabilized siRNAs with strategies that may be applied to siRNA irrespective of sequence to improve nuclease/chemical stability while retaining requisite RNA knockdown and potency may be useful in conjunction with heterogeneous delivery vehicle platforms.
  • Short Interfering Nucleic Acid Molecules of Formula a, B, C, D, E, F, G, H, I, J, K, L, M, N, O and P
  • The discovery and development of siNA molecules having Formula A-P originated with experiments aimed that exploring the role of placing 2′-O-methyl chemical modifications at specific positions within the siNA guide strand. The rationale was to identify specific positions of the guide strand that were tolerant or sensitive to chemical modifications and to use this information to map interactions between the guide strand and the RNA binding pocket within the Ago2 protein. This position-based approach was also a distinct and complementary to the approach of assigning modifications based on the sequence content of the siNA (e.g. purine or pyrimidine).
  • As a starting point, a siNA motif having alternating 2′-O-methyl modifications at odd positions on the guide strand ( e.g. positions 1, 3, 5, etc) and ribonucleotides at even positions ( e.g. positions 2, 4, 6, etc.) was altered in such a manner that the unmodified ribose residues at the even positions were replaced with 2′-deoxy-2′-fluoro modifications. siNA molecules with such alternating motifs were then designed to target ApoB and were complexed with a PVE-based polymer conjugate delivery vehicle for characterization in vivo. The siNA complexes were administered to mice by intravenous injection at a concentration of 3 mg per kg (mpk). A significant improvement in ApoB mRNA reduction was conferred by the inclusion of 2′-deoxy-2′-fluoro modifications within the guide strand as shown in FIG. 5.
  • Despite the promising results seen with the R-008357859-000V guide strand having alternating 2′-O-methyl and 2′-deoxy-2′-fluoro modifications in the context of polymer conjugate delivery (FIG. 5), the same guide strand motif was not effective when delivered to mice using a tetra-GalNAc conjugate (4GalNAc) single chemical entity (SCE) delivery platform. FIG. 6 shows that for both ApoB and SSB targets, the SCE delivered guide strands having alternating 2′-O-methyl and 2′-deoxy-2′-fluoro modifications did not significantly reduce mRNA expression levels in mice. Nevertheless, an ApoB targeted tetra-GalNAc conjugate (4GalNAc) single chemical entity (SCE) siNA having 2′-O-methyl pyrimidines and 2′-deoxy-2′-fluoro purines with phosphorothioate modified ribonucleotides at positions 1-3 of the guide strand, when administered to mice via SC delivery, did demonstrate dose dependent activity in mice (FIG. 7). However, when a SSB targeted sequence having 2′-O-methyl pyrimidines and 2′-deoxy-2′-fluoro purines with 2′-deoxy, 2′-deoxy-2′fluoro, and 2′-O-methyl respectively at positions 1, 2, and 3 of the guide strand was tested as a tetra-GalNAc conjugate, no knockdown was observed (FIG. 7). This result is surprising since both of these siNAs were shown to be very stable in serum and in S9 fractions (Table 6) and were functional when delivered as a polymer conjugate.
  • The key difference between the sequences tested is the presence of three phosphorothioate linkages at the 5′ end of guide strand in the active motif. To determine if the presence of the three 5′-terminal phosphorothioates could rescue activity of SSB-GalNAc conjugate, the SSB siNA construct was modified accordingly. As shown in FIG. 8, the SSB siNA construct having phosphorothioates at the 5′-end of the guide strand showed activity as a tetra-GalNAc conjugate. This was a surprising and unexpected result based on the serum/S9 stability and robust activity of the SSB sequence having 2′-O-methyl pyrimidines and 2′-deoxy-2′-fluoro purines with 2′-deoxy, 2′-deoxy-2′fluoro, and 2′-O-methyl at positions 1, 2, and 3 of the guide strand when administered as a polymer conjugate (data not shown).
  • Next, increasing the phosphorothioate content was explored to determine if the siNA SCE activity could be increased further since the phosphorothioate linkages could be playing role in improving pharmacokinetics, for example by binding to serum proteins. As shown as in FIG. 9, increasing the phosphorothioate linkages on either the guide or passenger strand did not improve activity. Note that all these constructs retained the three 5′-terminal guide strand phosphorothioate linkages. To elucidate this further, the specific placement of phosphorothioate linkages was investigated in both the guide and passenger strands. The data clearly showed that the placement of phosphorothioate linkages at the 5′-end of the guide strand, specifically between first and second nucleotide, is important for activity (FIG. 10). Because the phosphorothioate linkages at other positions did not impact the RNAi activity, it is the placement of phosphorothioate linkages at the 5′-end of the guide strand, and not their general presence within the siNA sequence, that was found to be important for in vivo activity with single chemical entity delivery modalities.
  • To investigate the role of phosphorothioate linkages, the stability of various siNA modification motifs were tested in different biological media including serum, S9, and lysosomal lysate fractions. This data was compared to in vivo metabolism data observed in mouse and rhesus. The lysosomal lysate fractions are enriched for late endosome/lysosome vesicles and serve as a source of lysosomal enzymes. The benefit of improved stability in lysosomal lysates at pH 5.5 and in mouse or rhesus liver was observed with the inclusion of 5′-terminal guide strand phosphorothioate linkages (Table 6). These results demonstrate that the siNA single chemical entity conjugates require more stability in during exposure to the endocytosis pathway compared to polymer conjugate delivery systems.
  • The presence of phosphorothioates or 2′ modifications alone may not be sufficient to provide adequate stability for siNA conjugate. For example, siNA molecules having phosphorothioates at the 5′-end of the guide strand are still susceptible to degradation in lysosomal lysates as well in mouse or rhesus liver (Table 6). The poor in vivo activity of the siNA molecules having 2′-modifications at the 5′-end of the guide strand can be rescued by the inclusion of phosphorothioate modifications at the 5′ end of the guide strands at position 1 and optionally through position 3. In one example, siNAs targeting CTNNB1 were synthesized and conjugated to the same SCE delivery vehicle used in FIG. 6. In vivo knockdown of CTNNB1 mRNA demonstrates that the inclusion of phosphorothioates in conjunction with 2′-sugar modifications at the 5′-end of the guide strand may result in significantly improved efficacy (FIG. 11). Therefore, the presence of 2′-sugar modifications in combination with phosphorothioate modification of the 5′-end of the guide strand appear to provide optimized stability and activity (Table 6 and 7). Application of this principle has resulted in alternate 2′-O-methyl and 2′-deoxy-2′-fluoro modified siNA molecules that show robust activity as GalNAc conjugates. As shown in FIG. 12, these molecules demonstrate >90% knockdown of ApoC3 mRNA in vivo. As shown in Table 6, application of this principle demonstrates robust stability in lysosomal lysates as well as in vivo in mice. These results hold true in vivo in non-human primes (rhesus) as well (FIG. 13).
  • Materials and Methods
  • siNA Synthesis
  • For each oligonucleotide of a target sequence, the two individual, complementary strands of the siNA were synthesized separately using solid phase synthesis, then purified separately by reversed phase solid phase extraction (SPE). The complementary strands were annealed to form the double strand (duplex) and delivered in the desired concentration and buffer of choice.
  • Briefly, the single strand oligonucleotides were synthesized using phosphoramidite chemistry on an automated solid-phase synthesizer, using procedures as are generally known in the art (see for example U.S. application Ser. No. 12/064,014). A synthesis column was packed with solid support derivatized with the first nucleoside residue (natural or chemically modified). Synthesis was initiated by detritylation of the acid labile 5′-O-dimethoxytrityl group to release the 5′-hydroxyl. A suitably protected phosphoramidite and a suitable activator in acetonitrile were delivered simultaneously to the synthesis column resulting in coupling of the amidite to the 5′-hydroxyl. The column was then washed with a solvent, such as acetonitrile. An oxidizing solution, such as an iodine solution was pumped through the column to oxidize the phosphite triester linkage P(III) to its phosphotriester P(V) analog. Unreacted 5′-hydroxyl groups were capped using reagents such as acetic anhydride in the presence of 2,6-lutidine and N-methylimidazole. The elongation cycle was resumed with the detritylation step for the next phosphoramidite incorporation. This process was repeated until the desired sequence was synthesized. The synthesis concluded with the final 5′-terminus protecting group (trityl or 5′-O-dimethoxytrityl).
  • Upon completion of the synthesis, the solid-support and associated oligonucleotide were dried under argon pressure or vacuum. Aqueous base was added and the mixture was heated to effect cleavage of the succinyl linkage, removal of the cyanoethyl phosphate protecting group, and deprotection of the exocyclic amine protection.
  • The following process was performed on single strands that do not contain ribonucleotides. After treating the solid support with the aqueous base, the mixture was filtered to separate the solid support from the deprotected crude synthesis material. The solid support was then rinsed with water, which is combined with the filtrate. The resultant basic solution allows for retention of the 5′-O-dimethoxytrityl group to remain on the 5′ terminal position (trityl-on).
  • For single strands that contain ribonucleotides, the following process was performed. After treating the solid support with the aqueous base, the mixture was filtered to separate the solid support from the deprotected crude synthesis material. The solid support was then rinsed with dimethylsulfoxide (DMSO), which was combined with the filtrate. Fluoride reagent, such as triethylamine trihydrofluoride, was added to the mixture, and the solution was heated. The reaction was quenched with suitable buffer to provide a solution of crude single strand with the 5′-O-dimethoxytrityl group on the final 5′ terminal position.
  • The trityl-on solution of each crude single strand was purified using chromatographic purification, such as SPE RPC purification. The hydrophobic nature of the trityl group permits stronger retention of the desired full-length oligo than the non-tritylated truncated failure sequences. The failure sequences were selectively washed from the resin with a suitable solvent, such as low percent acetonitrile. Retained oligonucleotides were then detritylated on-column with trifluoroacetic acid to remove the acid-labile trityl group. Residual acid was washed from the column, a salt exchange was performed, and a final desalting of the material commenced. The full-length oligo was recovered in a purified form with an aqueous-organic solvent. The final product was then analyzed for purity (HPLC), identity (Maldi-TOF MS), and yield (UV A260). The oligos were dried via lyophilization or vacuum condensation.
  • Annealing:
  • Based on the analysis of the product, the dried oligos were dissolved in appropriate buffers followed by mixing equal molar amounts (calculated using the theoretical extinction coefficient) of the sense and antisense oligonucleotide strands. The solution was then analyzed for purity of duplex by chromatographic methods and desired final concentration. If the analysis indicated an excess of either strand, then the additional non-excess strand was titrated until duplexing was complete. When analysis indicated that the target product purity has been achieved the material was delivered and ready for use.
  • Further Synthesis Steps for Commercial Preparations
  • Once analysis indicates that the target product purity has been achieved after the annealing step, the material is transferred to the tangential flow filtration (TFF) system for concentration and desalting, as opposed to doing this prior to the annealing step.
  • Ultrafiltration: The annealed product solution is concentrated using a TFF system containing an appropriate molecular weight cut-off membrane. Following concentration, the product solution is desalted via diafiltration using Milli-Q water until the conductivity of the filtrate is that of water.
  • Lyophilization: The concentrated solution is transferred to a bottle, flash frozen and attached to a lyophilizer. The product is then freeze-dried to a powder. The bottle is removed from the lyophilizer and is now ready for use.
  • Synthesis of siNA Single Chemical Entities (SCE)
    TetraGalNAc Synthesis and Preparation of TetraGalNAc-siNA conjugates
  • Synthesis of (2S)-2,6-bis[bis(prop-2-yn-1-yl)amino]hexanoic acid (1)
  • Into a 2000-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen was placed a solution of (2S)-2,6-diaminohexanoic acid (50 g, 342.03 mmol, 1.00 equiv) in acetonitrile (1000 mL) and heated to 50° C. To this was added potassium hydroxide (22.6 g, 0.4025 mol, 1.00 equiv, 85%). The resulting solution was stirred for 30 min. Then 3-bromoprop-1-yne (29.5 mL, 1.00 equiv) was added. The resulting solution was stirred for 1 hour at 50° C. additional potassium hydroxide (22.6 g, 0.4025 mol, 1.00 equiv) was added to the solution and stirred for 30 min at 50° C. To this was added 3-bromoprop-1-yne (29.5 mL, 1.00 equiv). The resulting solution was stirred for 1 hour. To this was added potassium hydroxide (22.6 g, 0.4025 mol, 1.00 equiv) again. The resulting solution was stirred for 30 min at 50° C., followed by addition of more 3-bromoprop-1-yne (29.5 mL, 1.00 equiv). The resulting solution was stirred for 1 hour. To this was added potassium hydroxide (22.6 g, 0.4025 mol, 1.00 equiv). The resulting solution was stirred for 30 min. To this was added 3-bromoprop-1-yne (29.5 mL, 1.00 equiv). The resulting solution was stirred for 3 hours. The reaction mixture was cooled to 25° C. with a water/ice bath. The solid was filtered out. The filtrate was adjusted to pH 4 with HCl (6M). The solid was filtered out. The filtrate was concentrated under vacuum. The residue was applied onto a silica gel column and eluted with dichloromethane/methanol (100:1-25:1). This resulted in (2S)-2, 6-bis[bis(prop-2-yn-1-yl)amino]hexanoic acid (1) as a light yellow oil.
  • MS(ES, m/z): 297.2, [M−H]− 1HNMR(CDCl3, 500 MHz, ppm): 3.62 (d, J=2.0 Hz, 4H), 3.52-3.49 (m, 1H), 3.50 (d, J=2.4 Hz, 4H), 2.62 (t, J=7.1 Hz, 2H), 2.30 (t, J=2.4 Hz, 2H), 2.27 (t, J=2.4 Hz, 2H), 1.88-1.79 (m, 2H), 1.60-1.53 (m, 2H), 1.52-1.43 (m, 2H).
  • Synthesis of 2-(2-hydroxyethoxy)ethyl 4-methylbenzenesulfonate (3)
  • Into a 2000-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen was placed a solution of 2-(2-hydroxyethoxy)ethan-1-ol (2, 42.4 g, 399.55 mmol, 1.00 equiv) in dichloromethane (1000 mL) and triethylamine (27.9 g, 275.72 mmol, 0.25 equiv). To the above was added p-toluenesulfonyl chloride (19.1 g, 100.18 mmol, 0.50 equiv). After stirred for 1 h at 25° C., the resulting mixture was washed with 1×500 mL of aq. potassium hydrosulfate (1M) and 1×500 mL of aq. sodium bicarbonate (5%) respectively. The organic layer was dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was applied onto a silica gel column and eluted with dichloromethane/methanol (100:1). This resulted in 2-(2-hydroxyethoxy)ethyl 4-methylbenzenesulfonate (3) as a colorless oil.
  • Synthesis of 2-(2-azidoethoxy)ethan-1-ol (4)
  • Into a 500-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen was placed a solution of 2-(2-[[(4-2-(2-hydroxyethoxy)ethyl 4-methylbenzenesulfonate (3, 50 g, 192.08 mmol, 1.00 equiv) in N,N-dimethylformamide (250 mL). This was followed by the addition of sodium azide (18.79 g, 289.03 mmol, 1.50 equiv) at 25° C. The resulting solution was stirred for 5 h at 100° C. in an oil bath. The reaction mixture was cooled and filtered. The filtrate was concentrated under vacuum. The residual solution was diluted with 1000 mL of dichloromethane and washed with 1×500 mL of water. The organic layer was dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was applied onto a silica gel column and eluted with dichloromethane/methanol (80:1). This resulted in 2-(2-azidoethoxy)ethan-1-ol (4) as a colorless oil.
  • 1HNMR (CDCl3, 400 MHz, ppm): 3.42-3.45 (t, J=4.8 Hz, 2H), 3.63-3.65 (t, J=4.8 Hz, 2H), 3.71-3.74 (t, J=4.8 Hz, 2H), 3.71-3.79 (m, 2H).
  • Synthesis of (3R,4R,5R,6R)-3-acetamido-6-(acetoxymethyl)tetrahydro-2H-pyran-2,4,5-triyl triacetate (6)
  • Into a 2000-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen was placed a solution of (3R,4R,5R,6R)-3-amino-6-(hydroxymethyl)tetrahydro-2H-pyran-2,4,5-triol hydrochloride (5, 120 g, 556.50 mmol, 1.00 equiv) in pyridine (1200 mL). This was followed by the addition of acetic anhydride (341.6 g, 3.35 mol, 6.00 equiv) dropwise with stirring at 0° C. The resulting solution was stirred overnight at 25° C. The reaction was then quenched by the addition of 8000 mL of water/ice. The solid was collected by filtration. This resulted in (3R,4R,5R,6R)-3-acetamido-6-(acetoxymethyl)tetrahydro-2H-pyran-2,4,5-triyl triacetate (6) as a white solid.
  • Synthesis of (3aR,5R,6R,7R,7aR)-5-(acetoxymethyl)-2-methyl-5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]oxazole-6,7-diyl diacetate (7)
  • Into a 2000-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen was placed a solution of (3R,4R,5R,6R)-3-acetamido-6-(acetoxymethyl)tetrahydro-2H-pyran-2,4,5-triyl triacetate (6, 30 g, 77.05 mmol, 1.00 equiv) in dichloromethane (1500 mL), then added iron (III) chloride (30 g, 184.95 mmol, 2.40 equiv). The resulting mixture was stirred for 2 h at 25° C. The reaction was then quenched by the addition of 1000 mL of water/ice. The organic layer was washed with 1×1000 mL of sodium aq. bicarbonate and 1×1000 mL of water, dried over anhydrous sodium sulfate and concentrated under vacuum. This resulted in (3aR,5R,6R,7R,7aR)-5-(acetoxymethyl)-2-methyl-5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]oxazole-6,7-diyl diacetate (7, 20 g, 79%) as yellow oil.
  • 1HNMR(CDCl3, 300 MHz, ppm): 2.03 (s, 9H), 2.12 (s, 3H), 3.97-4.27 (m, 4H), 4.90-4.93 (m, J=3.3 Hz, 1H), 5.45-5.47 (t, J=3.0 Hz, 1H), 5.98-6.00 (d, J=6.6 Hz, 1H).
  • Synthesis of (2R,3R,4R,5R,6R)-5-acetamido-2-(acetoxymethyl)-6-[2-(2-azidoethoxy)ethoxy]tetrahydro-2H-pyran-3,4-diyl diacetate (8)
  • Into a 500-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen was placed a solution of (3aR,5R,6R,7R,7aR)-5-(acetoxymethyl)-2-methyl-5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]oxazole-6,7-diyl diacetate (7, 40 g, 121.47 mmol, 1.00 equiv) in 1,2-dichloroethane (200 mL), 2-(2-azidoethoxy)ethan-1-ol (4, 23.89 g, 182.18 mmol, 1.50 equiv). To the above several 4A zeolite was added. The resulting mixture was stirred for 1 h at 25° C. Then trimethylsilyl trifluoromethanesulfonate (10.8 mL, 0.50 equiv) was added. After stirred overnight at 25° C., the reaction mixture was diluted with 500 mL of dichloromethane and washed with 1×500 mL of water, 1×500 mL of aq. sodium bicarbonate and 1×500 mL of water. The organic layer was dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was applied onto a silica gel column and eluted with dichloromethane/methanol (100:1). This resulted in (2R,3R,4R,5R,6R)-5-acetamido-2-(acetoxymethyl)-6-[2-(2-azidoethoxy)ethoxy]tetrahydro-2H-pyran-3,4-diyl diacetate (8) as a colorless oil.
  • MS(m/z): 461.1, [M+H]′
  • 1HNMR(CDCl3, 500 MHz, ppm) 5.78 (d, J=8.90 Hz, 1H), 5.36 (d, J=2.9 Hz, 1H), 5.22 (dd, J=11.2, 3.6 Hz, 1H), 4.77 (d, J=8.3 Hz, 1H), 4.19-4.12 (m, 2H), 4.11-4.05 (m, 1H), 3.98-3.92 (m, 2H), 3.82-3.78 (m, 1H), 3.71-3.63 (m, 4H), 3.49-3.38 (m, 2H), 2.16 (s, 3H), 2.05 (s, 3H), 2.01 (s, 3H), 1.97 (s, 3H).
  • Synthesis of (S)-2,6-bis(bis((1-(2-(2-(((2R,3R,4R,5R,6R)-3-acetamido-4,5-diacetoxy-6-(acetoxymethyl)tetrahydro-2H-pyran-2-yl)oxy)ethoxy)ethyl)-1H-1,2,3-triazol-4-yl)methyl)amino)hexanoic acid (9, tetraGalNAc acetate)
  • Into a 250-mL round bottom flask purged and maintained with an inert atmosphere of nitrogen was charged (2S)-2,6-bis[bis(prop-2-yn-1-yl)amino]hexanoic acid (1, 1.0 g, 1.0 equiv), (2R,3R,4R,5R,6R)-5-acetamido-2-(acetoxymethyl)-6-[2-(2-azidoethoxy)ethoxy]tetrahydro-2H-pyran-3,4-diyl diacetate (8, 9.26 g, 6.0 equiv), anhydrous THF 50 mL, CuBr.SMe2 (0.138 g, 0.20 equiv), and anhydrous DBU (1.5 ml, 3.0 equiv) in respective order. The resulting solution was stirred for 16 h at room temperature, quenched with acetic acid (0.75 mL, 4.0 equiv), treated with MP-TMT resin (Part No: 801472, from Biotage) (9 g), aged at room temperature for 16 h, filtered, and concentrated the filtrate to a foam solid. The solid was then dissolved in CH2Cl2 (140 mL), and washed with AcOH/NaCl solution (140 mL). The AcOH/NaCl solution was prepared with 1 mL AcOH and 100 mL 20% NaCl solution. The bottom organic layer was concentrated, and purified on a SiO2 column (220 g), eluting with CH2Cl2/MeOH. This resulted in (S)-2,6-bis(bis((1-(2-(2-(((2R,3R,4R,5R,6R)-3-acetamido-4,5-diacetoxy-6-(acetoxymethyl)tetrahydro-2H-pyran-2-yl)oxy)ethoxy)ethyl)-1H-1,2,3-triazol-4-yl)methyl)amino)hexanoic acid (9) as a white solid.
  • MS(m/z): 2139.5, [M+H]+
  • Synthesis of (S)-2,6-bis(bis((1-(2-(2-(((2R,3R,4R,5R,6R)-3-acetamido-4,5-dihydroxy-6-(hydroxymethyl)tetrahydro-2H-pyran-2-yl)oxy)ethoxy)ethyl)-1H-1,2,3-triazol-4-yl)methyl)amino)hexanoic acid (10, tetraGalNAc)
  • Into a 250-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen was charged (S)-2,6-bis(bis((1-(2-(2-(((2R,3R,4R,5R,6R)-3-acetamido-4,5-diacetoxy-6-(acetoxymethyl)tetrahydro-2H-pyran-2-yl)oxy)ethoxy)ethyl)-1H-1,2,3-triazol-4-yl)methyl)amino)hexanoic acid (9, 6.9 g, 1.0 equiv), Na2CO3 (6.83 g, 20 eq), water (56 mL), and MeOH (32 mL) in respective order. The reaction was aged at room temperature for 16 h, concentrated to residue, redissolved in water (50 mL), and purified on Combiflash C18 gold reverse column (415 g), eluting with water/MeCN. After concentration under vacuum, the product was dissolved in minimum amount of water, and lyophilized to obtain (S)-2,6-bis(bis((1-(2-(2-(((2R,3R,4R,5R,6R)-3-acetamido-4,5-dihydroxy-6-(hydroxymethyl)tetrahydro-2H-pyran-2-yl)oxy)ethoxy)ethyl)-1H-1,2,3-triazol-4-yl)methyl)amino)hexanoic acid (10) as a white solid.
  • MS(m/z): 1657 [M+Na]+
  • 1HNMR(D2O, 500 MHz, ppm): 8.05 (s, 2H), 7.91 (s, 2H), 4.62 (t, J=5.0 Hz, 4H), 4.57 (t, J=5.0 Hz, 4H), 4.45-4.41 (d, J=8.6 Hz, 4H), 3.99-3.82 (m, 28H), 3.80-3.61 (m, 28H), 3.14 (t, J=7.1 Hz, 1H), 2.52 (broad s, 2H), 1.99 (s, 6H), 1.98 (s, 6H), 1.73 (m, 2H), 1.60 (m, 2H), 1.29 (m, 2H).
  • Synthesis of tetraGalNAc-siNA conjugate (12) via tetraGalNAc acetate 9
  • To a solution of tetraGalNAc acetate (9, 58.7 mg, 0.027 mmol) in acetonitrile (1.5 ml) was added DIPEA (2.2 mg, 0.055 mmol) and HATU (10.44 mg, 0.027 mmol). The mixture was stirred at room temperature for 30 min, transferred into a solution siNA (11, 0.014 mmol) in water (1.5 ml) and acetonitrile (1.5 ml) via a syringe pump over 20 min, and stirred for 30 min before it was concentrated under vacuum down to 1.5 mL. Sodium carbonate (218 mg, 2.059 mmol) was then added, followed by MeOH (0.50 ml). The resulting solution was stirred at room temperature for 16 h, concentrated, purified via dialysis, and lyophilized to yield compound 12. The same approach can be used to generate compound 13.
  • RNA Silencing Activity of Compounds Transfected with Lipofectamine in Luciferase Constructs:
  • HEK293 cells stably transfected with luciferase vector that contains target sites for siNA in 3′UTR of renilla luciferase were generated. These cells were seeded on 96-well tissue culture plates (Corning: #3903) at a density of 7.5e3 cells per well in DMEM 10% serum media. Cellular plates were then incubated at 37° C./5% CO2 for 24 hr. After incubation, plates were treated with test compounds co-transfected with transfection reagent Lipofectamine 2000 (invitrogen: #11668-019) in Opti-MEM (Gibco: #31985) in accordance to manufacturers protocol. The treatment concentrations ranged from 10 nM to 0.03 pM. Treated plates were then incubated for 24 hr at 37° C./5% CO2. Following treatment incubation, cells were lysed and processed in accordance to Dual-Glo™ Luciferase Assay (Promega: E2920) and read on a TECAN safire2 plate reader.
  • RNA Silencing Activity of Compounds Transfected with Lipofectamine in HepG2 Cells:
  • HepG2 cells (ATCC: HB-8065) were seeded on collagen coated plates (BioCoat: 356649) at a density of 7.5e3 cells per well in DMEM 10% serum media. Cellular plates were then incubated at 37° C./5% CO2 for 24 hr. After incubation, plates were treated with test compounds co-transfected with transfection reagent Lipofectamine 2000 (invitrogen: 11668-019) in Opti-MEM (Gibco: 31985) in accordance to invitrogen protocol. The treatment concentrations ranged from 10 nM to 0.03 pM. Treated plates were then incubated for 24 hr at 37° C./5% CO2. Following treatment incubation, cells were lysed with PLA Buffer (AB: 4448542) in accordance to supplied protocol. Resulting cell lysate was reverse transcribed to cDNA using High Capacity cDNA Kit (AB: 4368813) and run through qPCR using Life Technology 7900.
  • RNA Silencing Activity of Compounds in Cryopreserved Primary Rhesus Hepatocytes:
  • Cryogenically preserved male rhesus primary hepatocyte cells were obtained from Celsis. Frozen cells were defrosted for 2 minutes in a 37° C. water bath and resuspended using thawing/plating media (Gibco: CM3000) at a ratio of 1-3 mL frozen cells per 50 mL of T/P media, in order to dilute out DMSO. Cells were spun down at 80×g for 5 minutes to pellet cells. Pelleted cell were then diluted in T/P media for seeding of cells to center 60 wells of a 96 well collagen coated plate (Biocoat: 354649) at 4.0e4 cells per well. Seeded cells were incubated for 18 hr at 37° C./5% CO2. After incubation, cells were washed once with Maintenance media (Gibco: CM4000) and aspirated dry. Immediately following wash, cells were treated with SCE diluted in maintenance media at a concentration range of 2 uM to 5 pM and allowed to incubate for 48 hr at 37° C./5% CO2. Following treatment incubation, cells were lysed with PLA Buffer (AB: 4448542) in accordance to provided protocol. Resulting cell lysate was reverse transcribed to cDNA using High Capacity cDNA Kit (AB: 4368813) and run through qPCR using Life Technology 7900.
  • In Vivo Evaluation of RNAi Activity:
  • CD1 female mice were dosed by subcutaneous injection in 200 ul volume. Animals were observed for behavioral or physiological changes. Animals were sacrificed 72 hrs post dose by CO2 asphyxiation followed by ex-sanguination via cardiac puncture. The liver samples were as 3 mm punches from the medial lobe and put into RNAlater tubes for isolation of total RNA. The mRNA knockdown analysis was conducted by Taqman analysis using standard procedures.
  • In Vitro siNA Metabolic Stability Assay
  • Test siNAs were incubated in either 100% mouse serum or 0.4 mg/mL rat lysosomal lysate at 20 ug/mL. To extract oligonucleotides from biomatrices the 96-well Clarity OTX cartridge plates (Phenomenex, Torrance, Calif.) were used with a 96-well plate vacuum manifold following the manufacturer's instructions. The eluants were next lyophilized over night then reconstituted in 1 mM EDTA for LC/MS analysis. Data acquisition and processing for the LC/MS system wereperformed using Excalibur software. A Hypersil Gold C18 reversed phase column (2.1×50 mm) was used. The column temperature was maintained at 70 C. The mobile phases used were as follows: A, 1.7 mM triethylamine (TEA) and 100 mM 1,1,1,3,3,3-hexafluoro-2-propanol (HFIP) in HPLC water; and B, methanol. The sample injection volume was 10 uL, and the flow rate was maintained at 0.4 mL/min. The mass spectrometer was a linear ion trap coupled to an Orbitrap allowing high-resolution mass measurements (LTQ-Orbitrap, Thermo Fisher Scientific, San Jose, Calif.), fitted with an ESI source, and operated in negative ion mode. Zero charge state siNA and metabolite masses were obtained by deconvolution of charge states using Thermo Extract software. Metabolites were identified by comparison of experimental masses to theoretical nuclease metabolite masses.
  • In Vivo siNA Liver Metabolite Identification
  • Approximately 150 mg to 300 mg liver tissue was homogenized in 1 mL of Tris-EDTA buffer, pH 7.2 using a tissue homogenizer with a stainless steel ball at 4 C, for 2 min. siNA and their metabolites were extracted from liver lysates using a combination liquid-liquid and solid phase extraction. A 500 mL volume of 5% ammonium hydroxide solution was added to each sample mixture, vortexed for and centrifuged for 10 min at 14 000 rpm. The aqueous phase was transferred to a fresh tube and an equal volume of a buffer (17.2 mM TEA and 200 mM HFIP, pH8.5) was added to the aqueous phase and the sample tubes were vortexed. The sample solutions were then extracted on an Oasis HLB SPE cartridge (10 mg, Waters, Mass., USA). The SPE cartridges were first conditioned with 1 mL of acetonitrile followed by two aliquots of 1 mL of buffer (8.6 mM TEA and 100 mM HFIP, pH 8.5). The sample solutions were then loaded onto the SPE cartridges. The cartridges were washed with two aliquots of 0.5 mL buffer (8.6 mM TEA and 100 mM HFIP, pH8.5) followed by 0.5 mL of 100 mM triethylammonium bicarbonate (TEAB). Samples were eluted with two aliquots of 0.5 mL 100 mM TEA in 60:40 (v/v) acetonitrile/water solution. The eluants were next lyophilized over night then reconstituted in 1 mM EDTA for LC/MS analysis. All LC/MS analyses were performed using a Thermo LTQ-Orbitrap mass spectrometer (Thermo Scientific, Inc.) Chromatographic separation of siNAs and their metabolites was conducted using a Thermo Hypersil Gold C18 column (2.1×50 mm) at a flow rate of 0.2 mL/min. The column was heated to 70 C to denature the siNA duplex. Mobile phase ‘A’ consisted of 1.7 mMTEA and 100 mMHFIP (pH 7.5) in water, and mobile phase ‘B’ consisted of methanol. The effluent from the HPLC column was introduced into the ion source of the LTQ-Orbitrap mass spectrometer operated in negative mode. Zero charge state siNA and metabolite masses were obtained by deconvolution of charge states using Thermo Extract software. Metabolites were identified by comparison of experimental masses to theoretical nuclease metabolite masses.
  • Figure US20150299696A1-20151022-C00025
    Figure US20150299696A1-20151022-C00026
  • Figure US20150299696A1-20151022-C00027
  • Figure US20150299696A1-20151022-C00028
  • One skilled in the art would readily appreciate that the present invention is well adapted to carry out the objects and obtain the ends and advantages mentioned, as well as those inherent therein. The methods and compositions described herein, as presently representative of preferred embodiments, are exemplary and are not intended as limitations on the scope of the invention. Changes therein and other uses will occur to those skilled in the art, which are encompassed within the spirit of the invention, are defined by the scope of the claims.
  • TABLE 1
    Duplex Target Site SEQ ID SEQ ID
    ID R Number (human) NO: 1 Target Sequence Modified Sequence NO: 2
    129302- R-008491391- 1797 1 CUGUUGGAUUGAUUCGAAA UuUcGaAuCaAuCcAaCaGUsU 7
    DC 000N
    129302- R-008491391- 1797 1 CUGUUGGAUUGAUUCGAAA GalNAcB CUgUUggaUUgaUUCgaaaUsU B 8
    DC 000N
    129303- R-008491393- 1797 1 CUGUUGGAUUGAUUCGAAA GalNAcB CUgUUggaUUgaUUCgaaaUsU B 8
    DC 000F
    129303- R-008491393- 1797 1 CUGUUGGAUUGAUUCGAAA UsusUscGaAuCaAuCcAaCaGUsU 9
    DC 000F
    129304- R-008491389- 1797 1 CUGUUGGAUUGAUUCGAAA GalNAcB CUgUUggaUUgaUUCgaaaUsU B 8
    DC 000R
    129304- R-008491389- 1797 1 CUGUUGGAUUGAUUCGAAA uuuCgaaUCaaUCcaaCagUsU 10
    DC 000R
    129305- R-008491392- 1797 1 CUGUUGGAUUGAUUCGAAA GalNAcB CUgUUggaUUgaUUCgaaaUsU B 8
    DC 000X
    129305- R-008491392- 1797 1 CUGUUGGAUUGAUUCGAAA usususCgaaUCaaUCcaaCagUsU 11
    DC 000X
    132037- R-008499711- 1797 1 CUGUUGGAUUGAUUCGAAA GalNAcB CUgUUggaUUgaUUCgaaaUsU B 8
    DC 000V
    132037- R-008499711- 1797 1 CUGUUGGAUUGAUUCGAAA pUsusUscGaAuCaAuCcAaCaGUsU 12
    DC 000V
    132038- R-008499712- 1797 1 CUGUUGGAUUGAUUCGAAA GalNAcB CUgUUggaUUgaUUCgaaaUsU B 8
    DC 000D
    132038- R-008499712- 1797 1 CUGUUGGAUUGAUUCGAAA pususUscGaAuCaAuCcAaCaGUsU 13
    DC 000D
    131992- R-008498416- 1797 1 CUGUUGGAUUGAUUCGAAA GalNAcB CUgUUggaUUgaUUCgaaaUsU B 8
    DC 000P
    131992- R-008498416- 1797 1 CUGUUGGAUUGAUUCGAAA p UsusUscGaAuCaAuCcAaCaGUsU 14
    DC 000P
    131996- R-008498418- 1797 1 CUGUUGGAUUGAUUCGAAA GalNAcB CUgUUggaUUgaUUCgaaaUsU B 8
    DC 000G
    131996- R-008498418- 1797 1 CUGUUGGAUUGAUUCGAAA p ususUscGaAuCaAuCcAaCaGUsU 15
    DC 000G
    132039- R-008499709- 1797 1 CUGUUGGAUUGAUUCGAAA GalNAcB CUgUUggaUUgaUUCgaaaUsU B 8
    DC 000X
    132039- R-008499709- 1797 1 CUGUUGGAUUGAUUCGAAA UsusUscGaAuCaAuCcAaCaGUsU 16
    DC 000X
    132040- R-008499710- 1797 1 CUGUUGGAUUGAUUCGAAA GalNAcB CUgUUggaUUgaUUCgaaaUsU B 8
    DC 000L
    132040- R-008499710- 1797 1 CUGUUGGAUUGAUUCGAAA ususUscGaAuCaAuCcAaCaGUsU 17
    DC 000L
    132041- R-008499713- 1797 1 CUGUUGGAUUGAUUCGAAA GalNAcB CUgUUggaUUgaUUCgaaaUsU B 8
    DC 000M
    132041- R-008499713- 1797 1 CUGUUGGAUUGAUUCGAAA ususUscGaAuCaAuCcAaCaGUsU 18
    DC 000M
    142795- R-008543009- 273 2 GGAGCAAGUUUACUGACAA p UsusGsuCaGuAaAcUuGcUcCUsU 19
    DC 000U
    142795- R-008543009- 273 2 GGAGCAAGUUUACUGACAA GalNAcB gGaGcAaGuUuAcUgAcAaUsU B 20
    DC 000U
    142804- R-008543018- 442 3 AAGUAUGUUCUCAUGUCUU pasAsgsAcAuGaGaAcauAcUuUsU 21
    DC 000C
    142804- R-008543018- 442 3 AAGUAUGUUCUCAUGUCUU GalNAcB AaGuAuGuUcUcAuGuCuUUsU B 22
    DC 000C
    126735- R-008417879- 9514 4 CUUUAACAAUUCCUGAAAU AsUsUsUCaggaaUUguUaaagUsU 23
    DC 000B
    126735- R-008417879- 9514 4 CUUUAACAAUUCCUGAAAU GalNAcB CUUUaaCaaUUCCUgaaaU TsT B 24
    DC 000B
    129230- R-008480267- 291 5 ACAACAGACUUUAAUGUAA TuACaUUaaagUCugUUgUUsU 25
    DC 000V
    129230- R-008480267- 291 5 ACAACAGACUUUAAUGUAA GalNAcB aCaaCagaCUUUaaUgUaaTsT B 26
    DC 000V
    129282- R-008484697- 291 5 ACAACAGACUUUAAUGUAA GalNAcB aCaaCagaCUUUaaUgUaaTsT B 26
    DC 000P
    129282- R-008484697- 291 5 ACAACAGACUUUAAUGUAA UsUsAsCaUUaaagUCugUUgUUsU 27
    DC 000P
    129283- R-008491315- 291 5 ACAACAGACUUUAAUGUAA TsusAsCaUUaaagUCugUUgUUsU 28
    DC 000J
    129283- R-008491315- 291 5 ACAACAGACUUUAAUGUAA GalNAcaCaaCagaCUUUaaUgUaaTsT B 29
    DC 000J
    133061- R-008502422- 1797 6 UUUCGAAUCAAUCCAACAG GalNAcB CUgUUggaUUgaUUCgaaaUsU B 8
    DC 000F
    133061- R-008502422- 1797 6 UUUCGAAUCAAUCCAACAG UsuUcGaAuCaAuCcAaCaGUsU 30
    DC 000F
    133064- R-008502424- 1797 6 UUUCGAAUCAAUCCAACAG GalNAcB CUgUUggaUUgaUUCgaaaUsU B 8
    DC 000Y
    133064- R-008502424- 1797 6 UUUCGAAUCAAUCCAACAG UusUcGaAuCaAuCcAaCaGUsU 31
    DC 000Y
    133064- R-008502424- 1797 54 UUUCGAAUCAAUCCAACAG GalNAcB CUgUUggaUUgaUUCgaaaUsU B 8
    DC 000Y
    133064- R-008502424- 1797 54 UUUCGAAUCAAUCCAACAG UusUcGaAuCaAuCcAaCaGUsU 31
    DC 000Y
    61657- R-008291260- 9514 4 CUUUAACAAUUCCUGAAAU LB CUUUAACAAUUCCUGAAAU TsT B 32
    DC 000L
    61657- R-008291260- 9514 4 CUUUAACAAUUCCUGAAAU AUUUCAGGAAUUGUUAAAGUsU 33
    DC 000L
    81858- R-008357859- 9514 4 CUUUAACAAUUCCUGAAAU LB CUUUAACAAUUCCUGAAAU TsT B 32
    DC 000V
    81858- R-008357859- 9514 4 CUUUAACAAUUCCUGAAAU AuUuCaGgAaUuGuUaAaGUsU 34
    DC 000V
    115748- R-008461704- 9514 4 CUUUAACAAUUCCUGAAAU AuUuCaGgAaUuGuUaAaGUsU 34
    DC 000E
    115748- R-008461704- 9514 4 CUUUAACAAUUCCUGAAAU LB CUUUaaCaaUUCCUgaaaU TsT B 35
    DC 000E
    115750- R-008462079- 291 5 ACAACAGACUUUAAUGUAA LB aCaaCagaCUUUaaUgUaaTsT B 36
    DC 000W
    115750- R-008462079- 291 5 ACAACAGACUUUAAUGUAA UuAcAuUaAaGuCuGuUgUUsU 37
    DC 000W
    110573- R-008428355- 1797 1 CUGUUGGAUUGAUUCGAAA UuUcGaAuCaAuCcAaCaGUsU 7
    DC 000S
    110573- R-008428355- 1797 1 CUGUUGGAUUGAUUCGAAA LB CUgUUggaUUgaUUCgaaaUsU B 38
    DC 000S
    128797- R-008490480- 1797 1 CUGUUGGAUUGAUUCGAAA UsusUscGaAuCaAuCcAaCaGUsU 9
    DC 000C
    128797- R-008490480- 1797 1 CUGUUGGAUUGAUUCGAAA LB CUgUUggaUUgaUUCgaaaUsU B 38
    DC 000C
    57407- R-008277564- 9514 4 CUUUAACAAUUCCUGAAAU AsUsUsUCaggaaUUguUaaagUsU 23
    DC 000P
    57407- R-008277564- 9514 4 CUUUAACAAUUCCUGAAAU LB CUUUaaCaaUUCCUgaaaU TsT B 35
    DC 000P
    129288- R-008491383- 9514 4 CUUUAACAAUUCCUGAAAU AsUsUsUsCaggaaUUguUaasasgsUsU 39
    DC 000N
    129288- R-008491383- 9514 4 CUUUAACAAUUCCUGAAAU GalNAcB 40
    DC 000N CsUsUsUsaaCaaUUCCUgaasasUsTsT B
    129289- R-008491382- 9514 4 CUUUAACAAUUCCUGAAAU AsUsUsUCaggaaUUguUaaagUsU 23
    DC 000E
    129289- R-008491382- 9514 4 CUUUAACAAUUCCUGAAAU GalNAcB 40
    DC 000E CsUsUsUsaaCaaUUCCUgaasasUsTsT B
    129290- R-008491377- 9514 4 CUUUAACAAUUCCUGAAAU AsUsUsUsCaggaaUUguUaaagUsU 41
    DC 000F
    129290- R-008491377- 9514 4 CUUUAACAAUUCCUGAAAU GalNAcB CUUUaaCaaUUCCUgaasasUsTsT B 42
    DC 000F
    129291- R-008491378- 9514 4 CUUUAACAAUUCCUGAAAU GalNAcB CUUUaaCaaUUCCUgaaaU TsT B 24
    DC 000P
    129291- R-008491378- 9514 4 CUUUAACAAUUCCUGAAAU AsUsUsUsCaggaaUUguUaasasgsUsU 39
    DC 000P
    129292- R-008491380- 9514 4 CUUUAACAAUUCCUGAAAU AsUsUsUCaggaaUUguUaaagUsU 23
    DC 000M
    129292- R-008491380- 9514 4 UUUAACAAUUCCUGAAAU GalNAcB 43
    DC 000M CsUsUsUsaaCaaUUCCUgaaaU TsT B
    133063- R-008502423- 1797 6 UUUCGAAUCAAUCCAACAG GalNAcB CUgUUggaUUgaUUCgaaaUsU B 8
    DC 000P
    133063- R-008502423- 1797 6 UUUCGAAUCAAUCCAACAG UuUscGaAuCaAuCcAaCaGUsU 44
    DC 000P
    133057- R-008502426- 1797 6 UUUCGAAUCAAUCCAACAG GalNAcB CUgUUggaUUgaUUCgaaaUsU  B 8
    DC 000R
    133057- R-008502426- 1797 6 UUUCGAAUCAAUCCAACAG UsuUscGaAuCaAuCcAaCaGUsU 45
    DC 000R
    133058- R-008502427- 1797 6 UUUCGAAUCAAUCCAACAG GalNAcB CUgUUggaUUgaUUCgaaaUsU B 8
    DC 000Z
    133058- R-008502427- 1797 6 UUUCGAAUCAAUCCAACAG UusUscGaAuCaAuCcAaCaGUsU 46
    DC 000Z
    133066- R-008502428- 1797 6 UUUCGAAUCAAUCCAACAG GalNAcB CUgUUggaUUgaUUCgaaaUsU B 8
    DC 000H
    133066- R-008502428- 1797 6 UUUCGAAUCAAUCCAACAG UsusUcGaAuCaAuCcAaCaGUsU 47
    DC 000H
    133055- R-008502429- 1797 6 UUUCGAAUCAAUCCAACAG GalNAcB CUgUUggaUUgaUUCgaaaUsU B 8
    DC 000S
    133055- R-008502429- 1797 6 UUUCGAAUCAAUCCAACAG UsuUcGaAuCasAuCcAaCaGsUsU 48
    DC 000S
    133056- R-008502430- 1797 6 UUUCGAAUCAAUCCAACAG GalNAcB CUgUUggaUUgaUUCgaaaUsU B 8
    DC 000F
    133056- R-008502430- 1797 6 UUUCGAAUCAAUCCAACAG UuUcGaAuCaAuCcAaCsasGsUsU 49
    DC 000F
    133062- R-008502431- 1797 6 UUUCGAAUCAAUCCAACAG GalNAcB CUgUUggaUUgaUUCgaaaUsU B 8
    DC 000P
    133062- R-008502431- 1797 6 UUUCGAAUCAAUCCAACAG UuUcGaAuCsasAsuCcAaCaGUsU 50
    DC 000P
    133065- R-008502432- 1797 6 UUUCGAAUCAAUCCAACAG GalNAcB CUgUUggaUUgaUUCgaaaUsU B 8
    DC 000Y
    133065- R-008502432- 1797 6 UUUCGAAUCAAUCCAACAG UsusUscsGaAuCaAuCcAaCaGUsU 51
    DC 000Y
    129278- R-008489620- 9514 4 CUUUAACAAUUCCUGAAAU GalNAcB CUUUaaCaaUUCCUgaaaU TsT B 24
    DC 000E
    129278- R-008489620- 9514 4 CUUUAACAAUUCCUGAAAU AuUuCaGgAaUuGuUaAaGUsU 34
    DC 000E
    129287- R-008489628- 291 5 ACAACAGACUUUAAUGUAA GalNAcB aCaaCagaCUUUaaUgUaaTsT B 26
    DC 000Z
    129287- R-008489628- 291 5 ACAACAGACUUUAAUGUAA UuAcAuUaAaGuCuGuUgUUsU 37
    DC 000Z
    104995- R-008415011- 9514 4 CUUUAACAAUUCCUGAAAU LB CUUUaaCaaUUCCUgaaaUUsU B 52
    DC 000K
    104995- R-008415011- 9514 4 CUUUAACAAUUCCUGAAAU auT UCaggaaUUguUaaagUsU 53
    DC 000K
    wherein:
    A, U, C, and G = Adenosine, Uridine, Cytidine and Guanosine ribonucleotides respectively
    a, u, c and g = 2′-deoxy-2′-fluoro (2′-F) modified Adenosine, Uridine, Cytidine and Guanosine respectively
    A, U, C and G = 2′-O-methyl (2′-OMe) modified Adenosine, Uridine, Cytidine and Guanosine respectively
    A, U, C, and G = 2′-deoxy (2′-H) modified Adenosine, Uridine, Cytidine and Guanosine respectively
    B = inverted abasic
    T = thymidine
    s = phosphorothioate linkage
    u = 2′-benzyl Uridine
    p = terminal phosphate
    LB = animohexyl phosphate linker - inverted abasic cap ---
    GalNAcB = tetraGalNAc-animohexyl phosphate linker - inverted abasic cap ---
  • TABLE 2
    Sequence SEQ ID
    CGLFEAIEEFIENLWELLIDGWYGYGRKKRRQRR 54
    CGLFEAIEGFIENGWEGMIDGWYGYGHKKHHQHH 55
    C-bAla-LFEAIEGFIENGWEGMIDGWYGYGRKKRRQRR 56
    CGLFEAIEGFIENGLKGLIDWWYGYGRKKRRQRR 57
    CGLFEAIEGFIEWGWEGMIDGWYGYGRKKRRQRR 58
    CRRQRRKKRGYGYWGDIMGEWGNEIFGEIAEFLG 59
    CGLFEAIEGFIENGWEGMIDGWYGYGRKKRRQR 60
    CYGRKKRRQRRGLFEAIEGFIENGWEGMIDGWYG 61
    CIFGAIAGFIKNILKGLIDG 62
    CIFGAIAGFIRNIW 63
    CGLFHALLHLLHSLWHGLLHAWYGYGHKKHHQHR 64
    CGLFEAIEGLIENGWEGMIDGWYGYGRKKRRQRR 65
    CGLFELIEGFIENGWEGMIDGWYGYGRKKRRQRR 66
    CGLFEAIEGFIENGWEGLIDGWYGYGOOOOOQRR (O = ornithine) 67
    CGLFGAIEGFIENGWEGLIDGWYGYGRKKRRQRR 68
    CGLFEAIEGFLENGWEGMIDGWYGYGRKKRRQRR 69
    CGLFEAIEGFIENGLEGMIDGWYGYGRKKRRQRR 70
    CGLFGAIEGFIENGWEGMIDGWYGYGRKKRRQRR 71
    CGLFEAIEGFIENGWEG-Nle-IDGWYGYGRKKRRQRR 72
    CGIFGAIAGFIKNIWKGLIDW 73
    CYGRKKRRQRRGLFEAIEGFIENGWKGLIDAWYG 74
    CGLLEALEGLLESLWEGLLEAWYGYGRKKRRQRR 75
    CGLFEAIEGFIENGWEGMIDNWYGYGRKKRRQRR 76
    CIFGAIAGFIKNIWEGLIEAWYGLHLLHHLLHHLHHLLHHLLHL 77
    CIFGAIAGFIKNIWEGLIDAF 78
    CIFGAIAGFIKNIWEGLI 79
    CGLFEAIEGFIENGWEGMIDGWYGYGRKKRRQRRK(stearyl) 80
    CGLFEAIAGFIEGGWPGLINGWYGYGRKKRRQRRLHLLHHLLHHLHHLLHHLLHLLHHLLHHL 81
    CGLFEAIEGFIENGWEGMIDGWYGGGGLHLLHHLLHHLHHLLHHLLHLLHHLLHHL 82
    CGLFEAIEGFIENGWEGMIDGWYGLHLLHHLLHHLHHLLHHLLHL 83
    CGLFEALLELLESLWELLLEAYGRKKRRQRR 84
    CGLFEAIEGFIENGWEGMIDGWYGYGRKKRRQRR 85
    CGLFEAIEGFIENGWEGMADGWYGYGRKKRRQRR 86
    CGIFGAIAGFIKNIWEGLIDWWYGYGRKKRRQRR 87
    CGFLPAIAGILSQLFEGLIDGWYGYGRKKRRQRR 88
    CFFGAIWGFIKSIL 89
    CIFGAIAGFIKNIWKGLIDWWYG 90
    CGLFEAIEGFIWNGWEGMIDGWYGYGRKKRRQRR 91
    CGLFEAIAEFIENGWEGMIDGWYGYGRKKRRQRR 92
    CYGRKKRRQRRGLFEAIEGFIENGWKGLIDWWYG 93
    CGLFEAIEGFIEEGWEGMIDGWYGYGRKKRRQRR 94
    CGLFEAIEGFIENAWEGMIDGWYGYGRKKRRQRR 95
    CGLFEAIEGFIENGWEGMIDLWYGYGRKKRRQRR 96
    CRLLRLLLRLWRRLLRLLR 97
    CGGFEAIEGFIENGWEGMIDGWYGYGRKKRRQRR 98
    CGLFEKIEGFIENGWEGMIDGWYGYGRKKRRQRR 99
    CGLFEAIEGFIENGWENMIDGWYGYGRKKRRQRR 100
    CIFGAIAGFIKNILKGL 101
    CIFGAIAGFIKNILKGLIDGWYG 102
    CGLFEAIEGFIENGWEGMIDGWYG-(PEG)3-YGRKKRRQRR 103
    CGLFEALLELLESLWELLLEAYGRKKRRQRRLHLLHHLLHHLHHLLHHLLHL 104
    CYGRKKRRQRRWEAALAEALAEALAEHLAEALAEALEALAA 105
    CIFGAIAGFIKNIWEGLIDGWYGKLALKLALKALKAALKLA 106
    CFFGAIWEFIRSILEGLIDGWYGYGRKKRRQRR 107
    CGLFHALLHLLHSLWHLLLHAWYGYGRKKRRQRR 108
    CGLFHALLHLLHSLWHLLLHAWYGYGHKKHHQHR 109
    CGLFGALLELLESLWKGLLEWYGRKKRRQRR 110
    CRRQRRKKRGYGYWGDILGEWGNEIFGEIAEFLG 111
    CGLFEALEGFLENGWEGLLDGWYGYGROORRQRR (O = ornithine) 112
    CGLFGEIEELIENGLKNLIDWWYGYGRKKRRQRR 113
    CRRQRRKKRGYGYWWDILGKWGNEIFGEIAEFLG all (D) aminos 114
    CGIFGAIAGFIKNIL 115
    CGIFGAIAGLLKNIFK 116
    CIFGAIAGFIKNIWKGLIDW 117
    CIFGAIAGFIKNIWK 118
    CGLFEEIEGFIENGWEGLIDWWYGYGHKKHHQHR 119
    CGLFGEIEELIENGLKNLIDWWYGYGHKKHHQHR 120
    CGLFEEIEEFIENGWEGLIDWWYGYGHKKHHQHR 121
    stearyl-WEAALAEALAEALAEHLAEALAEALEALAAYGRKKRRQRRC 122
    CGLFEAIEGFIENGWKGLIDGWYGGLFEAIEGFIENGWKGLIDWWYG 123
    CGFFHAFFHFFHSFWHGFFEA 124
    CGNFGEIEELIEEGLENLIDWWNG 125
    CFFGAIWEFIRNILEGF 126
    CFFGAIWEFIHSIL 127
    CGLFHALLHLLHSLWHGLLEA 128
    CIFGAIAGFIKNIWEGL 129
    CIFGAIAGLLKNIFEGLIDGWYGYGRKKRRQRR 130
    CGFIGAIANLLSKIFEGLIDGWYGYGRKKRRQRR 131
    CGLFEAIEELIENLWKGLIDAWYGYGRKKRRQRR 132
    CGIFGAIAGLLKNIFKGLIDA 133
    CGIFGAIAGLLKNIFKGLIDW 134
    CGIFEAIAGLLKNIFK 135
    CGIFEEIAGLLKNIFK 136
    CGLFEAIAGFIEGGWPGLINGWYGYGRKKRRQRRLHLLHHLLHHLHHLLHHLLHL 137
    CGLFEAIEGFIENGWKGMIDWWYGYGRKKRRQRRK(stearyl) 138
    CGLFGEIEEFIENGWKGLIDWWYG 139
    CIFGAIAGFIKNIWLHLLHHLLHHLHHLLHHLLHL 140
    CGIFGAIEGFIENGWKGLIDAWYGYRKKRRQRR 141
    CELFGAIEGFIENGWKGLIDWWYGYGRKKRRQRR 142
    CIFGIDDLIIGLLFVAIVEAGIGGYLLGSYGRKKRRQRR 143
    GLFGALAEALAEALAEHLAEALAEALEALAAGGSC 144
    CGFIGAIANLLSKIFEGLIDGWYGYGRKKRRQRR all (D) 145
    CFFGAIWEFIRSILKGLI 146
    CFFGAIWEFIRSILK 147
    CFFGAIWEFIRSILE 148
    CIFGAIAGFIKNIWE 149
    CIFGAIAGFIKNIWKGLIDA 150
    CFFEAIEEFIKNILK 151
    CIFGAIAGLLRNIF 152
    CGIFGAIAGLLKNIW 153
    CLFGAIWEFIKSIL 154
    CFWGAIWEFIKSIL 155
    CFGGAIWEFIKSIL 156
    CFAGAIWEFIKSIL 157
    CGLFEAIEGFIENGWEGM(SO2)IDGWYGYGRKKRRQRR 158
    CGLFEAIEGFIENGWEGMIDWWYGYGRKKRRQRR 159
    CFFGAIWEFIKSIG 160
    CFFGAIWEFIKSIA 161
    CFFGAIWEFIKSIN 162
    CFFGAIWEFIKSIW 163
    CFFGAIWEFIKSILEGLIDWWYGYGHKKHHQHR 164
    Ac-CLHLLHHLLHHLHHLLHHLLHLLHHLLHHL-NH2 165
    Ac-LHLLHHLLHHLHHLLHHLLHLLHHLLHHLGGGRKKRRQRRRPPQC-NH2 166
    CRKKRRQRRRPPQGGGLHLLHHLLHHLHHLLHHLLHLLHHLLHHL 167
    CLHLLHHLLHHLHHLLHHLLHLLHHLLHHLGGGRKKRRQRRRPPQ 168
    CGLFHAIAHFIHGGWHGLIHGWYGYGRKKRRQRR 169
    CGLFKAIAKFIKGGWKGLIKGWYGYGRKKRRQRR 170
    CGLFEAIAGFIENGWEGMIDGWYGYGRKKRRQRR 171
    CWEAALAEALAEALAEHLAEALAEALEALAAYGRKKRRQRR 172
    CGLFEAIEGFIENGWEGMIDGWYGRKKRRQRRRPPQ 173
    GLFEAIEGFIENGWEGMIDGWYGYGRKKRRQRRC 174
    Ac-LHLLHHLLHHLHHLLHHLLHLLHHLLHHLRKKRRQRRRPPQ-NH2 175
    Ac-LHLLHHLLHHLHHLLHHLLHLLHHLLHHLGPGRKKRRQRRRPPQ-NH2 176
    Ac-LIRLWSHLIHIWFQNRRLKWKKK-NH2 177
    Ac-RKKRRQRRRPPQQQQQQ-NH2 178
    Ac-GLFEAIEGFIENGWEGMIDGWYGYGRKKRRQRR-NH2 179
    Ac-LHLLHHLLHHLHHLLHHLLHLLHHLLHHLGGGRRRRRRRRR-NH2 180
    Ac-LHLLHHLLHHLHHLLHHLLHLLHHLLHHL-(Peg)12-RKKRRQRRRPPQ-NH2 181
    Ac-GLFGAIAGFIENGWEGMIDGWYGLIRLWSHLIWFQNRRLKWLLL-NH2 182
    Ac-HHHHHRKKRRQRRRPPQGGGLHLLHHLLHHLHHLLHHLLHLLHHLLHHL-NH2 183
    Ac-LHLLHHLLHHLHHLLHHLLHLLHHLLHHL-(Peg)2-RKKRRQRRRPPQ-NH2 184
    Ac-LHLLHHLLHHLHHLLHHLLLLHHLLHHLGGGRQIKIWFQNRRMKWKKGG-NH2 185
    Ac-KLLKLLLKLWLKLLKLLLKLLGGGRKKRRQRRRPPQ-NH2 186
    Ac-LHHLLHHLLHLLHHLLHHLHHLLHHLLHLC-NH2 all (D) 187
    Ac-LHLLHHLLHHLHHLLHHLLHLLHHLLHHL-PEG6-RKKRRQRRRPPQC-NH2 188
    Ac-GLFEAIEGFIENGWEGMIDGWYGYGRKKRRQRRC-NH2 189
    CGLFEAIEGFIENGWEGMIDGWYGYGRKKRRQRR all (D) 190
    CGLFEAIEGFIENGWEGMIDGWYGYGRRRRRRRRR-NH2 191
    YGRKKRRQRRGLFEAIEGFIENGWEGMIDGWYGC-NH2 192
    CGVFVLGFLGFLATAGSYGRKKRRQRR-NH2 193
    CGLFKAIAKFIKGGWKGLIKGWYG-NH2 194
    CGLFEAIEGFIENGWEGMIDGWYGYGRKKR 195
    CGLFEAIEGFIENGWEGMIDGWYGYGRKKRRQRRYGRKKRRQRR 196
    CGLFEAIEGFIENGWEGMIDGWYGYGRKKRRQRRYGRKKRRQRR 197
    CGLFEAIKGFIENGWEGMIDGWYGYGRKKRRQRR 198
    CGLFEAIHGFIENGWEGMIDGWYGYGRKKRRQRR 199
    CGLFEAIRGFIENGWEGMIDGWYGYGRKKRRQRR 200
    CGLFEAIDGFIENGWEGMIDGWYGYGRKKRRQRR 201
    CRLFEAIEGFIENGWEGMIDGWYGYGRKKRRQRR 202
    CGGGLFEAIEGFIENGWEGMIDGWYGYGRKKRRQRR 203
    CGLFEAIEGFIENGWEGMIDGWYGGGGYGRKKRRQRR 204
    CGLFEAIEGFIENGWEGMIDGWYG-(PEG)11-YGRKKRRQRR 205
    CFLGFLLGVGSAIASGIAVSKVLHL 206
    CGVFVLGFLGFLATAGSAMGARSLTLSAYGRKKRRQRR 207
    Ac-GLWRALWRLLRSLWRLLWRA-mercaptoethylamide 208
    C-Nle-LFEAIEGFIENGWEGMIDGWYGYGRKKRRQRR 209
    CELFEAIEGFIENGWEGMIDGWYGYGRKKRRQRR 210
    CGFFGAIAGFLEGGWEGMIAGWHGYGRKKRRQRR 211
    CFLGFLLGVGSAIASGIAVSKVLHLYGRKKRRQRR 212
    GLFEAIEGFIENGWEGLAEALAEALEALAAGGSC 213
    CGLFEAIEGFIENGWEGMIDGWYGLHLLHHLLHHLHHLLHHLLHLLHHLLHHL 214
    CGLFEAIEGFIENGWEGMIDGWYGYGRKKRRQRRLHLLHHLLHHLHHLLHHLLHLLHHLLHHL 215
    CGLFGAIAGFIEGGWTGMIDGWYGYGRKKRRQRR 216
    CGLFGAIAGFIEGGWQGMVDGWYGYGRKKRRQRR 217
    CGLFGAIAGFIENGWQGLIDGWYGYGRKKRRQRR 218
    CGLFGAIAGFIENGWEGLVDGWYGYGRKKRRQRR 219
    CGLFGAIAGFIEGGWSGMIDGWYGYGRKKRRQRR 220
    CGLFGAIAGFIEGGWPGLVAGWYGYGRKKRRQRR 221
    CGLFGAIAGFIENGWEGMVDGWYGYGRKKRRQRR 222
    CGLFGAIAGFIEGGWPGLINGWYGYGRKKRRQRR 223
    CGLFGAIAGFIENGWEGLIDGWYGYGRKKRRQRR 224
    CGLFGAIAGFIENGWEGMIDGWYGYGRKKRRQRR 225
    CGLFGAIAGFIENGWEGMIDGWYGSSKKKK 226
    CGLFGAIAGFIENGWEGLIDGWYGYGRKKRRQRR 227
    CGLFEAIEGFIENGWEGLIDGWYGYGRKKRRQRR 228
    CGLFGAIAGFIENGWEGLIEGWYGGGRKKRRQRR 229
    CGLFEAIEGFIENGWEGMIDGWYGGGRKKRRQRR 230
    CGLFEAIAGFIENGWEGLIDGWYGYGRKKRRQRR 231
    CGLFEAIAEFIENGWEGLIEGWYGGRKKRRQRR 232
    CGLFEAIEGFIENGWEGMIDGWYGRKKRRQRRR 233
    CKLLKLLLKLWLKLLKLLLKLL 234
    CKLLKLLLKLWLKLLKLLLKLLYGRKKRRQRR 235
    GLFEAIEGFIENGWEGMIDGWYGC 236
    CVLFEAIEGFIENGWEGMIDGWYGYGRKKRRQRR 237
    CSLFEAIEGFIENGWEGMIDGWYGYGRKKRRQRR 238
    CGLFEAIEGFIENGWEGMIDGWYGYGRKKRRQ 239
    CGLFEAIEGFIENGWEGMIDGWYGYGRKKRR 240
    CGLFEAIEGFIENGWEGMIDGWYGYGKKKKKQKK 241
    CGLFEAIEGFIENGWEGMIDGWYGGLFEAIEGFIENGWEGMIDGWYG 242
    CGLFEAIEGFIENGWEGMIDGWYGYGRKKRRQRRGLFEAIEGFIENGWEGMIDGWYGYGRKKRRQRR 243
    RRQRRKKRGYGYWGDIMGEWGNEIFGEIAEFLGC 244
    CRRQRRKKRGYGYWGDIMGEWGNEIFGEIAEFLG 245
    GLFEAIEGFIENGWEGMIDGWYGYGRK-K(D)-RRQRR 246
    GLFEAIEGFIENGWEGMIDGWYGYGRKK-R(D)-RQRR 247
    GL-F(D)-EAIEGFIENGWEGMIDGWYGYGRKKRRQRR 248
    GLF-E(D)-AIEGFIENGWEGMIDGWYGYGRKKRRQRR 249
    CGLFEAIEGFIENGWEGMIDGWYG 250
    CYGRKKRRQRR 251
    YGRKKRRQRRC 252
    RRQRRKKRGYGYWGDIMGEWGNEIFGEIAEFLGC all(D) 253
    CRRQRRKKRGYGYWGDIMGEWGNEIFGEIAEFLG all(D) 254
    CGLFEAIEGFIENGWEGMIDGAYGYGRKKRRQRR 255
    CGLFEALLELLESLWELLLEAWYGYGRKKRRQRR 256
    CGLFEAIEGFNENGWEGMIDGWYGYGRKKRRQRR 257
    CGLFEAIEGFIENEWEGMIDGWYGYGRKKRRQRR 258
    K(stearoyl)GLFEAIEGFIENGWEGMIDGWYGYGRKKRRQRRC 259
    CGLFEAIK(stearoyl)GFIENGWEGMIDGWYGYGRKKRRQRR 260
    CGLFEAIKGFIENGWEGMIDGWYGYGRK(stearoyl)KRRQRR 261
    CGLFEAIEGFIENPWEGMIDGWYGYGRKKRRQRR 262
    (stearyl)GLFEAIEGFIENPWEGMIDGWYGYGRKKRRQRRC 263
    CGLFGAIAGFIEGGWPGLINGWYGYGRKKRRQRRLHLLHHLLHHLHHLLHHLLHLLHHLLHHL 264
    CGLFGAIAGFIEGGWPGLINGWYGYGRKKRRQRRLHLLHHLLHHLHHLLHHLLHL 265
    CGLFEAIAGFIEGGWPGLINGWYGYGRKKRRQRR 266
    CGLEEAIEGFIENGWEGMIDGWYGYGRKKRRQRR 267
    CGLFNAIEGFIENGWEGMIDGWYGYGRKKRRQRR 268
    CGLFAAIEGFIENGWEGMIDGWYGYGRKKRRQRR 269
    CGLFEAIENFIENGWEGMIDGWYGYGRKKRRQRR 270
    CGLFEAIEKFIENGWEGMIDGWYGYGRKKRRQRR 271
    CGLFEAIEGFAENGWEGMIDGWYGYGRKKRRQRR 272
    CGLFEAIEGFIENWWEGMIDGWYGYGRKKRRQRR 273
    CGLFEAIEGFIENNWEGMIDGWYGYGRKKRRQRR 274
    CGLFEAIEGFIENGEEGMIDGWYGYGRKKRRQRR 275
    CGLFEAIEGFIENGWAGMIDGWYGYGRKKRRQRR 276
    CGLFEAIEGFIENGWNGMIDGWYGYGRKKRRQRR 277
    CGLFEAIEGFIENGWGGMIDGWYGYGRKKRRQRR 278
    CGLFEAIEGFIENGWEGMIDAWYGYGRKKRRQRR 279
    CGLFEAIEGFIENGWLGMIDGWYGYGRKKRRQRR 280
    CGLFEAIEGFIENGWKGMIDGWYGYGRKKRRQRR 281
    CGLFEAIEGFIENGWEGMIDKWYGYGRKKRRQRR 282
    CGLFEAIEGFIENGWEGMIDEWYGYGRKKRRQRR 283
    CGLFEAIEGFIENGWEGMIDGLYGYGRKKRRQRR 284
    CGLFEAIEGFIENGWEGMIDGNYGYGRKKRRQRR 285
    CGLFEAIEGFIENGWEGMIDGKYGYGRKKRRQRR 286
    CGLFEAIEGFIENGWEGMIDGEYGYGRKKRRQRR 287
    CGLFEALEELLEGGWEGLIEAWYGYGRKKRRQRR 288
    CELFGAIWEFIEGGWEGLIEAWYGYGRKKRRQRR 289
    CGLFEALEEFIEGGWEGLLEAWYGYGRKKRRQRR 290
    CGLFEALEEFIENGWEGLLEAWYGYGRKKRRQRR 291
    CGLFEAIEGFIESGWEGLIDGWYGYGRKKRRQRR 292
    CGLFEAIEEFIEGGWEGLIEAWYGYGRKKRRQRR 293
    CGLFEAIEGFIENGWEGLIDAWYGYGRKKRRQRR 294
    CGLFEAIEGFILNGWEGMIDGWYGYGRKKRRQRR 295
    CGLFEAIEGFIKNGWEGMIDGWYGYGRKKRRQRR 296
    CGLFEAIEGFIGNGWEGMIDGWYGYGRKKRRQRR 297
    CGLFEAIEGFIELGWEGMIDGWYGYGRKKRRQRR 298
    CGLFEAIEGFIEKGWEGMIDGWYGYGRKKRRQRR 299
    CGLFEAIAEFIEGGWEGLIEGWYGYGRKKRRQRR 300
    CRGWEVLKYWWNLLQY 301
    CRGWEVLKYWWNLLQYYGRKKRRQRR 302
    CGLFGAIAGFIENGWEGMIDGWYGFRYGRKKRRQRR 303
    Ac-CGLFEAIEGFIENGWEGMIDGWYGYGRKKRRQRR-CO2H 304
    CGLLEALEGLLENGWEGLLEAWYGYGRKKRRQRR 305
    CLRHLLRHLLRHLRHLLRHLRHLLRHLLRH 306
    CGIFEAIEGFIENGWEGIIDGWYGYGROORRQRR (O = ornithine) 307
    CGIGAVLKVLTTGLPALISWIKRKRQQ 308
    CGIGAVLKVLTTGLPALISWIHHHHQQ 309
    CGAFEAIEGFIENGWEGMIDGWYGYGRKKRRQRR 310
    Ac-LHLLHHLLHHLHHLLHHLLHLLHHLLHHLRRRRR 311
    CGLFGAIWGFIENWWKGLIDWWYGYGRKKRRQRR 312
    CGLFGAIEGFIENGWKGLIDAWYGYGRKKRRQRR 313
    CGLFEAIAGFIENGWKGLIDWWYGYGRKKRRQRR 314
    GLFEAIEGFIENGWKGLIDWWYGYGRKKRRQRRC 315
    YGRKKRRQRRGLFEAIEGFIENGWKGLIDAWYGC 316
    YGRKKRRQRRGLFEAIEGFIENGWKGLIDWWYGC 317
    CGLFHAIHGFIENGWHGLIDWWYGYGRKKRRQRR 318
    CGLFEAIEGFIENGWKGLIDWWYGYGRKKRRQRRK(stearyl) 319
    CGLFKALLKLLKSLWKLLLKAWYGYGHKKHHQHR 320
    CGLFKALLKLLKSLWKGLLKAWYGYGHKKHHQHR 321
    CGLAKALLKLLKSLWKGLIEAWYGYGRKKRRQRR 322
    CGIFGAIAGFIKNIW 323
    CIFGAIAGFIKNIWEGLIDGWYGYGRKKRRQRR 324
    CGIFGAIAGFIKNIWEGLIDGYGRKKRRQRR 325
    CGIFGAIAGFIKNIWKGLIDAWYGYGRKKRRQRR 326
    CIFGAIAGFIKNIWKGLIDWWYGYGRKKRRQRR 327
    CLFGAIAGFIKNIW 328
    CGL(R5)EAIEGF(S8)ENGWEGMIDGWYGYGRKKRRQRR 329
    CGLFEA(S5)EGF(S5)ENGWEGMIDGWYGYGRKKRRQRR 330
    CGLFEAIEGFIENGWEGAIDGWYGYGRKKRRQRR 331
    CGLFEAIEGFIENGWEGEIDGWYGYGRKKRRQRR 332
    CGIFGAIAGFIKNGWEGMVDWYGYGRKKRRQRR 333
    CGLFEAIAGFIENGWEGMIDGWYGFYGRKKRRQRR 334
    CGIFGAIAGFIKNGWEGMIDGWYGYGRKKRRQRR 335
    CIFGAIAGFIKNIW 336
    CIFGAIAGFIKNIWYGRKKRRQRR 337
    CGIFGAIAGFIKNIWEGMIDGWYGYGRKKRRQRR 338
    CGLFEAIEGFIENGWEGLIEAYGRKKRRQRR 339
    CGLFEALLGFIENGWEGLIDGYGRKKRRQRR 340
    CGLFGAIEGFIENGWEGLIDGWYGYGRKKRRQRRR 341
    CELFGAIEGFIENGWEGMIDGWYGYGRKKRRQRRR 342
    CGLFEAIEGFIENGWEGMIDGWYGYGHKKHHQHR 343
    CGLFGAIEGFIEGGWPGLINGWYGYGRKKRRQRRR 344
    CGLFKALLKLLKSLWKLLLKAYGRKKRRQRR 345
    CGLFKALLKLLKSLWKLLLKAWYGYGRKKRRQRR 346
    CGLFRALLRLLRSLWRLLLRAYGRKKRRQRR 347
    CGLFEAILGFIENGWEGLIDGWYGYGRKKRRQRR 348
    CGLFEAIWEFIENGWEGLIDGWYGYGRKKRRQRR 349
    CGLFEAIEGFIENGWEGMIDGWYGGGGLHLLHHLLHHLHHLLHHLLHL 350
    CGPVEDAITAAIGRVADTVGTYGRKKRRQRR 351
    CMDGTLFPGDDDLAIPATEFFSTKA 352
    CGLFEALEEFIEGGWEGLLEAWYGYGRKKRRQRR 353
    CGLFEALEEFIENGWEGLLEAWYGYGRKKRRQRR 354
    CELFGAIWEFIEGGWEGLIEAYGRKKRRQRR 355
    CGLFEAIEGFIEEGWEGMIDGWYGYGRKKRRQRR 356
    CGLFEAIAEFIENGWEGMIDGWYGYGRKKRRQRR 357
    CGLFEAIAEFIEGLWEGLIEGWYGYGRKKRRQRR 358
    CGLLEALEGLLESLWEGLLEAWYGYGRKKRRQRR 359
    CGLFEAIEGFIENGWEGMIDIWYGYGRKKRRQRR 360
    CGLFEAIEGFIENGWRGMIDGWYGYGRKKRRQRR 361
    CGLFEAIEGFIENGWDGMIDGWYGYGRKKRRQRR 362
    CGLFEAIEGFIENHWEGMIDGWYGYGRKKRRQRR 363
    CGLFEAIEGFIENWWKGLIDWWYGYGRKKRRQRR 364
    GLFEAIEGFIENGWKGLIDAWYGYGRKKRRQRRC 365
    CGLFEAIEGFIENGWKGMIDAWYGYGRKKRRQRR 366
    CGLFEAIEGFIENGWKGMIDWWYGYGRKKRRQRR 367
    CGLAEAIEGFIENGLKGLIDWWYGYGRKKRRQRR 368
    RRQRRKKRGYGYWGDILGEWGNEIFGEIAEFLGC all(D) 369
    CRRQRRKKRGYGYWGDILGEWGNEIFGEIAEFLG all(D) 370
    CGLFEAIEGFIENGWKGLIDWWYGYGRKKRRQRR 371
    CGFFEAIEGFIENGLKGLIDAWYGYGRKKRRQRR 372
    CGLFEAIEGFIENGLKGLIDAWYGYGRKKRRQRR 373
    CELFGAIEGFIENGWKGLIDAWYGYGRKKRRQRR 374
    CGLFKAIKGFIKNGWKGLIKAWYGYGRKKRRQRR 375
    CGLAEALLELLESLWKGLIEAYGRKKRRQRR 376
    CGIFGAIEGFIENGWKGLIDAWYGYGRKKRRQRR 377
    CGIAGAIAGFIKNIWEGLIDWWYGYGRKKRRQRR 378
    CGIAGAIAGFIKNIWKGLIDAWYGYGRKKRRQRR 379
    CGIFGAIAGFIKNIWEGLIDGWYGKKKKKKKKK 380
    CG(R5)FEAIEG(S8)IENGWEGMIDGWYGYGRKKRRQRR 381
    CGLFEAIEGF(R5)ENGWEG(S8)IDGWYGYGRKKRRQRR 382
    GLFEAIEGFIENGWEGMIDGWYGCYGRKKRRQRR 383
    GLFEAIEGFIENGWEGMIDGWYGGCGYGRKKRRQRR 384
    GLLEALEGLLENGWEGLLDGWYGYGRKKRRQRR 385
    CFFGAIWEFIRNIL 386
    CIFGAIAGFIRSIL 387
    CGLFEEIEEFIENGWEGLIDWWYGYGRKKRRQRR 388
    CGFFGAIWEFIKSIL 389
    GFFGAIWEFIKSILC 390
    CGLFEALEGFIENGWEGLLDGWYGYGROORRQRR (O = ornithine) 391
    CGLFEALLELLENGWELLLEAWYGYGRKKRRQRR 392
    CGLFEALLELLENGWELLLDGWYGYGRKKRRQRR 393
    CALFEAIEAFIENGWEAMIDAWYGYGRKKRRQRR 394
    CGLFGAIWGFIENGWEGLIDGWYGYGRKKRRQRR 395
    CGLFEAIEELIENLWKGLIDWWYGYGRKKRRQRR 396
    CGLFEEIEGFIENGWKGLIDWWYGYGRKKRRQRR 397
    CGLFEEIEGFIENGWKGLIDWWYGYGHKKHHQHR 398
    CFFGAIWEFIKNILKGLIDGWYG 399
    CGIFGAIAGFIRSIL 400
    CGLFEEIEGFIENGWEGMIDGWYGYGRKKRRQRR 401
    CGLFEAIEGFIENGWEGMIDGWNGYGRKKRRQRR 402
    AGYLLGKINLKALAALAKKILHHHHHHKKKKKKC 403
    Bis CGLFEAIEGFIENGWEGMIDWWYGYGRKKRRQRR 404
    CGLFEAIEGFIENGWEGMIDGWYG-(PEG)6-YGRKKRRQRR 405
    CGIFGAIWNGIKSLFEGLIDGWYGYGRKKRRQRR 406
    CGIFGAIEGFIENGWEGLIDWWYGYGRKKRRQRR 407
    CIFGAIAGFIKNIWEGLIDWWYGYGRKKRRQRR 408
    CGLFEAIEGFIENGWKGLIDGWYGGLFEAIEGFIENGWKGLIDWWYG 409
    CWEAALAEALAEALAEHLAEALAEALEALAAYGRKKRRQRRK(stearyl) 410
    CGLFEAIEGFIENGWKGLIDWWYGYGRKKRRQRR 411
    CGLFEELEELLEEGWEGLLEAYGRKKRRQRR 412
    CGNFEEIEEFIEEGLRNFIDWWYGYGHKKHHQHR 413
    CFFGAIWEFIRNILEGLIDWWYGYGRKKRRQRR 414
    CFFGAIWEFIKNILLHLLHHLLHHLHHLLHHLLHL 415
    CGLFEAIEGFIENGWEGMIDGWYGYGRKKRRQRR all(D) 416
    CGFFHAFFHFFHSFWHGFFEA 417
    CGLFHALLHLLHSLWHGLLHWWYGYGHKKHHQHR 418
    CGLFGALLELLESLWEGLLEWYGRKKRRQRR 419
    CGLFGALLELLESLWEGLLEWYGHKKHHQHR 420
    CGLFHALLHLLHSLWKGLLEWWYGF 421
    CIFGAIAGFIRSILEGF 422
    CGIFGAIAGFIKNIWKGLIDA 423
    CFFEAIEEFIKNIWK 424
    CGLFEAIEGFIENGWKGLIDWLAEALAEALEALAA 425
    GCGIFGAIAEFIKNIW 426
    CIFGAIAEFIKNIWKGLIDW 427
    CFFGAIWEFIKSILELLLEAYGHKKHHQHRR 428
    CWFGAIWEFIKSIL 429
    CAFGAIWEFIKSIL 430
    CFLGAIWEFIKSIL 431
    CFFGAIWEFIKSIK 432
    CGFIGAIANLLSKIFEGLIDGWYGYGRKKRRQRR all(D) 433
    CFFGAIWEFIKSIL 434
    CIFGAIAGFIKNIWLHLLHHLLHHLHHLLHHLLHL all(D) 435
    CFFGAIAEFIKNIW 436
    CIFEAIWGFIKNIW 437
    stearyl-AGYLLGKINLKALAALAKKILHHHHHHKKKKKKC 438
    CIFEAIAGFIKNIWKGLIDWWYGYGRKKRRQRR 439
    CGLFEAIEGFIENGWKGLIDWWYGGRPRESGKKRKRKRLKP 440
    C(b-Ala)GFGEIEEFIENGLKNLIDWWYGYGHKKHHQHR 441
    C(b-Ala)GFEFIEEFIENGLKNLIDWWYGYGRKKRRQRR 442
    C(b-Ala)GFEFIEEFIENGLKNLIDWWYGYGHKKHHQHR 443
    CGGIEEIAGLLSKILKGLIDWWYGYGHKKHHQHR 444
    CGFIGAIANLLSKIFEGLIDWWYGYGRKKRRQRR 445
    CGFIGAIAELLEKIFEGLIDWWYGYGRKKRRQRR 446
    CGFIGAIAELLEKIFEGLIDWWYGYGHKKHHQHR 447
    CFFGAIWEFIRNILEGLIDWWYGYGHKKHHQHR 448
    CFFGAIWEFIKSILLHLLHHLLHHLHHLLHHLLHL 449
    CFFGAIWEFIRSILLHLLHHLLHHLHHLLHHLLHL 450
    CGFFGAIWEFIRSILEGFIDWWYGYGYGHKKHHQHR 451
    CGLFEAIWEFIKSILEGLLEAYGHKKHHQHR 452
    CGLFEAIWEFIKSILEGLLEAWYGYGHKKHHQHR 453
    CGIFGAIAGFIKNIWKYGRKKRRQRR 454
    CGLFEALLELLESLWELLLEAWYGYGHKKHHQHR 455
    CIFGAIAGFIRNIWKGLIDGWYG 456
    CGIFGAIAGFIRNIWKGLIDGWYG 457
    CFFGAIWEFIKNILKLHLLHHLLHHLHHLLHHLLHL 458
    CFFGAIWEFIRNILLHLLHHLLHHLHHLLHHLLHL 459
    CFFGKIWEFIKSIL 460
    CYGRKKRRQRRGLFEALLELLESLWELLLEA 461
    462
    CWWGAIEGFIKSIL 463
    CFFGAIWEWIKSIL 464
    CFFGAIWEFWKSIL 465
    CFFGAIWEFIKFIL 466
    CFFGAIWEFIKKIL 467
    CFFGAIWEFIKGIL 468
    CFFGAIWEFIKLIL 469
    CFFGAIWEFIKWIL 470
    CFFGAIWEFIKSFL 471
    CFFGAIWEFIKSKL 472
    CFFGFIWEFIKSIL 473
    CIFGAIAGFIKNILKGLIDAF 474
    CFFGKIWELWEWIL 475
    CFFGAIWEFAKSIL 476
    CFFGAIWEFIKSAL 477
    CFFGAIWEFIKSWL 478
    CFFGAIWEFIKSILK 479
    CFFGAIWEFIKSILE 480
    CFFKAIWEFIKSIL 481
    CFFNAIWEFIKSIL 482
    CFFGGIWEFIKSIL 483
    CFFGNIWEFIKSIL 484
    CFFGALWEFIKSIL 485
    CFFGAAWEFIKSIL 486
    CGLFHALLHLLHSLWHGLLDG 487
    CGLFHALLHLLHSLWHGLLEW 488
    CGLFHALLHLLHSLWHLLLEA 489
    CGLFHALLHLLHSLWKLLLEW 490
    CKFGAIWEFIKSIL 491
    CFKGAIWEFIKSIL 492
    CFFGAIWKFIKSIL 493
    CFFGAIWAFIKSIL 494
    CFFGAIWLFIKSIL 495
    CFFGAIWFFIKSIL 496
    CFFGAIWNFIKSIL 497
    CFFGAIWELIKSIL 498
    CFFGAIWEAIKSIL 499
    CGLFEAIEGFIENGWEGLAEALAEALEALAAYGRKKRRQRR 500
    CIFGAIAGFIKNIWEGMIDGWYGYGRKKRRQRR 501
    CIFGAIAGFIKNIWEGLIDAWYGYGRKKRRQRR 502
    CIFGAIAGFIKNIWKGLIDAWYGYGRKKRRQRR 503
    CIFGAIAGFIKNIWIFGAIAGFIKNIWWYGYGRKKRRQRR 504
    CGLFGAIAGFIENGWEGLIEGWYG 505
    CGLFEAIEGFIENGWEGLIDGWYGYGOOOOOQRR (O = ornithine) 506
    CGLFEAIEGFIENGWKGLIDWWYGYGRKKRRQRR 507
    CGLFEAIEGFIENGWEGLIDGWYGYGRKKRRQRRK(stearyl) 508
    CYGHKKHHQHRGLFEAIEGFIENGWKGLIDWWYG 509
    CYGHKKHHQHRGLFEAIEEFIENGWEGLIDGWYG 510
    CGLFEAIEGFIENGWKGLIDGWYGYGRKKRRQRRK(stearyl) 511
    CGLFEAIEGFIENGWHGMIDGWYGYGRKKRRQRR 512
    IFGIDDLIIGLLFVAIVEAGIGGYLLGSYGRKKRRQRRC 513
    CGFFGEIAELIEEGLKGLIDWWNG 514
    CGLFGEIEELIEEGLENLIDWWNG 515
    CFFGAIWEFIHSIL all (D) 516
    CFFGAIWEFIHNIL 517
    CFFGAIWEFIHSIFK 518
    CGIFEAIAGLLKWIFK 519
    CGIFELIAGLLKNIFK 520
    CGIFEAIAGLLKSILKK(stearyl) 521
    CGIFGAIAGLLKSILKK(stearyl) 522
    CIFGAIAGFIKNILKGL all (D) 523
    CIFGAIAGFIKNILKGLIDGWWYG 524
    CIFGAIAGFIKNIWHGLI 525
    CIFGAIAGFIKNILKGLK(stearyl) 526
    GLGKLINKIFGAIAGFIC all (D) 527
  • TABLE 3
    motif pyrimidine purine 5′ end 3′ end
    PS02
    2′OMe 2′F iB dTs; dT; iB
    PS03
    2′OMe 2′OH iB dTs; dT; iB
    PS05
    2′OMe 2′F iB omeUs; omeU; iB
  • TABLE 4
    posi-
    pyrim- tion
    motif idine purine 5′ end 3′ end 14
    GS10 2′OMe 2′F rXs; rXs; rXs & YA = mUs; mU 2′F
    2′OMe
    GS10ffd
    2′OMe 2′F fX; fX; dX mUs; mU 2′ F
    GS13
    1, 3 . . . 17, 19 = mUs; mU
    2′ OMe
    GS13b
    1, 3 . . . 17, 19 = mUs; mU 2′F
    2′OMe & 2, 4 . . . 16,
    18 = 2′F
    GS13b- s 1, 3 . . . 17, 19 = mUs; mU 2′F
    2′OMe & 2, 4 . . . 16,
    18 = 2′F (position
    1-3 thioates)
  • TABLE 5
    Start
    Target Seq Motif Duplex R- ORS
    Name Loc Name number # Passenger strand Guide strand
    ApoB 9514 Sci13 R-008291260- 9122 6amiL; iB; omeC; omeU; omeU; omeU; omeA; rU; omeU; rU; omeC; rA;
    (PS03GS13) 000L rA; rA; omeC; rA; rA; omeU; omeU; omeC; omeG; rG; omeA; rA; omeU; rU;
    omeC; omeU; rG; rA; rA; rA; omeU; omeG; rU; omeU; rA; omeA; rA;
    dTs; dT; iBSup omeG; omeUs; omeUSup
    ApoB 9514 PS03GS13b R-008357859- 15792 6amiL; iB; omeC; omeU; omeU; omeU; omeA; fluU; omeU; fluU; omeC; fluA;
    000V rA; rA; omeC; rA; rA; omeU; omeU; omeC; omeG; fluG; omeA; fluA; omeU; fluU;
    omeC; omeU; rG; rA; rA; rA; omeU; omeG; fluU; omeU; fluA; omeA; fluA;
    dTs; dT; iBSup omeG; omeUs; omeUSup
    ApoB 9514 PS02GS13b R-008461704- 24314 6amiL; iB; omeC; omeU; omeU; omeU; omeA; fluU; omeU; fluU; omeC; fluA;
    000E fluA; fluA; omeC; fluA; fluA; omeU; omeU; omeG; fluG; omeA; fluA; omeU; fluU;
    omeC; omeC; omeU; fluG; fluA; fluA; omeG; fluU; omeU; fluA; omeA; fluA;
    fluA; omeU; dTs; dT; iBSup omeG; omeUs; omeUSup
    SSB 291 PS02GS13b R-008462079- 24314 6amiL; iB; fluA; omeC; fluA; fluA; omeC; omeU; fluU; omeA; fluC; omeA; fluU;
    000W fluA; fluG; fluA; omeC; omeU; omeU; omeU; fluA; omeA; fluA; omeG; fluU;
    omeU; fluA; fluA; omeU; fluG; omeU; omeC; fluU; omeG; fluU; omeU; fluG;
    fluA; fluA; dTs; dT; iBSup omeU; omeUs; omeUSup
    CTNNB1 1797 PS05GS13b R-008428355- 23491 6amiL; iB; omeC; omeU; fluG; omeU; omeU; omeU; fluU; omeU; fluC; omeG; fluA;
    000S fluG; fluG; fluA; omeU; omeU; fluG; omeA; fluU; omeC; fluA; omeA; fluU;
    fluA; omeU; omeU; omeC; fluG; fluA; omeC; fluC; omeA; fluA; omeC; fluA;
    fluA; fluA; omeUs; omeU; iBSup omeG; omeUs; omeUSup
    CTNNB1 1797 PS05GS13b-s R-008490480- 28071 6amiL; iB; omeC; omeU; fluG; omeU; omeU; omeUs; fluUs; omeUs; fluC; omeG;
    000C fluG; fluG; fluA; omeU; omeU; fluG; fluA; omeA; fluU; omeC; fluA; omeA;
    fluA; omeU; omeU; omeC; fluG; fluA; fluU; omeC; fluC; omeA; fluA; omeC;
    fluA; fluA; omeUs; omeU; iBSup fluA; omeG; omeUs; omeUSup
    ApoB 9514 Sci10 R-008277564- 6922 6amiL; iB; omeC; omeU; omeU; omeU; rAs; rUs; rUs; omeU; omeC; fluA;
    (PS02GS10) 000P fluA; fluA; omeC; fluA; fluA; omeU; omeU; fluG; fluG; fluA; fluA; omeU; omeU;
    omeC; omeC; omeU; fluG; fluA; fluA; fluG; fluU; omeU; fluA; fluA; fluA;
    fluA; omeU; dTs; dT; iBSup fluG; omeUs; omeUSup
    ApoB 9514 Sci10ffd R-008415011- 21732 6amiL; iB; omeC; omeU; omeU; omeU; fluA; fluU; dT; omeU; omeC; fluA;
    (PS05GS10ffd) 000K fluA; fluA; omeC; fluA; fluA; omeU; omeU; fluG; fluG; fluA; fluA; omeU; omeU;
    omeC; omeC; omeU; fluG; fluA; fluA; fluG; fluU; omeU; fluA; fluA; fluA;
    fluA; omeU; omeUs; omeU; iBSup fluG; omeUs; omeUSup
  • TABLE 6
    % of Parent
    % of Parent % of Parent Rat % of Parent % of Parent % of Parent % of Parent % of Parent
    Mouse Monkey Lysosomal in vivo in vivo in vivo in vivo in vivo
    Serum S9@pH 5.5 Lysate mouse liver mouse liver rhesus liver rhesus liver rhesus liver
    2 hr 4 hr 4 hr 4 hr post 24 hr post 6 hr post 24 hr post 48 hr post
    R# incubation incubation incubation dose dose dose dose dose
    R-008417879- PS 97 93 121 0 nd 0 nd
    000B
    GS 97 102 35 51 nd 3 nd
    R-008480267- PS 99 48 nd 0 nd nd nd nd
    000V
    GS 94 7 nd 0 nd nd nd nd
    R-008491315- PS nd nd nd 0 nd nd nd nd
    000J
    GS nd nd nd 13 nd nd nd nd
    R-008484697- PS 100 nd nd nd nd nd nd nd
    000P
    GS
    0 nd nd nd nd nd nd nd
    R-008491391- PS 98 95 120 50 nd 31 nd 0
    000N
    GS 99 109 0 0 nd 0 nd 0
    R-008491393- PS 100 99 143 39 nd 25 nd 0
    000F
    GS
    100 106 123 95 nd 84 nd 87
    R-008502422- PS nd nd 100 0 nd nd nd nd
    000F
    GS nd nd 100 100 nd nd nd nd
    R-008502424- PS nd nd 100 0 nd nd nd nd
    000Y
    GS nd nd 2 10 nd nd nd nd
    PS—Paasenger
    GS—Guide
    nd—not determined
  • TABLE 7
    % KD IC50 w/ IP in rhesus
    in LF2K in pri.
    Gene SC Dose mouse HEK-Luc hepatocytes
    Entry Target R # mpk liver [pM] [nM]
    1 CTNNB1 R-008491391-000N 30 33 8.7 0.81
    2 CTNNB1 R-008491393-000F 30 76 52 1.01
    3 CTNNB1 R-008491389-000R 30 17 16.1 0.37
    4 CTNNB1 R-008491392-000X 30 76 7.5 0.97
    5 CTNNB1 R-008499711-000V 5 27 3.6 1.12
    6 CTNNB1 R-008499712-000D 5 39 4 1.64
    7 CTNNB1 R-008498416-000P 5 45 6.7 2.36
    8 CTNNB1 R-008498418-000G 5 39 5.6 3.78
    9 CTNNB1 R-008499709-000X 5 38 4.3 39.48
    10 CTNNB1 R-008499710-000L 5 37 6.8 5.18
    11 CTNNB1 R-008499713-000M 5 40 28.9 3.20
    12 ApoC3 R-008543009-000U 15 97 nd nd
    13 ApoC3 R-008543018-000C 15 92 nd nd
    15 ApoB R-008417879-000B 5 43 85.3 1.5
    16 SSB R-008480267-000V 50 12 29.1 4.23
    17 SSB R-008484697-000P 15 59 23.4 nd
  • TABLE 8
    Figure US20150299696A1-20151022-C00029
    Figure US20150299696A1-20151022-C00030
    Figure US20150299696A1-20151022-C00031
    Figure US20150299696A1-20151022-C00032
    Figure US20150299696A1-20151022-C00033
    Figure US20150299696A1-20151022-C00034
    Figure US20150299696A1-20151022-C00035
    Figure US20150299696A1-20151022-C00036
    Figure US20150299696A1-20151022-C00037
    R = H, Boc, Cbz, Ac, PEG, lipid, targeting ligand, linker(s) and/or peptide(s).
    “nucleotide” can be substituted with non-nucleotide moiety such as abasic or linkers as are generally known in the art.
    n = 0 to 750.

Claims (42)

1. A composition comprising:
a double-stranded short interfering nucleic acid (siNA) molecule that modulates expression of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure selected from the structures represented by
Figure US20150299696A1-20151022-C00038
wherein,
(a) the upper strand is the passenger strand and the lower strand is the guide strand of the double-stranded nucleic acid molecule; the guide strand is complementary to the target sequence and the passenger strand is complementary to the guide strand, wherein one or more mismatches between the guide strand and passenger strand and/or between the guide strand and the target sequence are tolerated so long as RNAi activity is maintained;
(b) each N is independently a nucleotide wherein one or more Ns may be substituted with a non-nucleotide moiety so long as RNAi activity is maintained;
(c) each B1, B2, and B3 is independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker, wherein any of B1, B2, and/or B3 is optionally absent;
(d) x is an integer from 0 to 4, provided that when x is 1, 2, 3, or 4, one or more of the (N)x nucleotides can be complementary to nucleotides in target sequence, and one or more phosphorothioate internucleotide linkage(s) “s” can be present in the (N)x region when x is 1, 2, 3, or 4;
(e) y is an integer from 0 to 4, provided that when y is 1, 2, 3, or 4, one or more of the (N)y nucleotides can be complementary to nucleotides in target sequence, and one or more phosphorothioate internucleotide linkage(s) “s” can be present in the (N)y region when y is 1, 2, 3, or 4;
(f) N nucleotides of the guide strand are 2′-deoxy-2′-fluoro nucleotides and N nucleotides of the guide strand are 2′-O-methyl nucleotides, with an optional variance of 1 or 2 N or N nucleotides being tolerated provided that as RNAi activity is maintained;
(g) S=a phosphorothioate or phosphorodithioate internucleotide linkage wherein S1 is required and S2 and S3 are optional; and
(h) N nucleotides of the passenger strand are independently selected from ribonucleotide, 2′-O-alkyl nucleotide, 2′-deoxy-2′-fluoro nucleotide, 2′-deoxy nucleotide, and LNA.
2-10. (canceled)
11. The composition according to claim 1, wherein each (N) of the passenger strand is independently selected from 2′-O-alkyl, 2′-deoxy-2′-fluoro, 2′-deoxy, and/or LNA modified nucleotide.
12. The composition according to claim 1, wherein each (N) of the passenger strand is a 2′-O-alkyl modified nucleotide.
13. The composition according to claim 1, wherein each (N) of the passenger strand is a 2′-O-methyl modified nucleotide.
14. The composition according to claim 1, wherein x=2 and y=2.
15. The composition according to claim 1, wherein each N nucleotide of the guide strand is a 2′-deoxy-2′-fluoro nucleotide and each N nucleotide of the guide strand is a 2′-O-methyl nucleotide.
16. The composition according to claim 1, wherein 5, 6, 7, 8, 9, or 10 or more pyrimidine N nucleotides of the passenger strand are 2′-O-methyl nucleotides and 5, 6, 7, 8, 9, or 10 or more purine N nucleotides of the passenger strand are 2′-deoxy-2′-fluoro nucleotides.
17. The composition according to claim 1, wherein the 5′-terminal N position comprises a terminal phosphate group.
18. The composition according to claim 1, wherein B1 and B2 each comprise an inverted abasic 2′-deoxyribose moiety and B3 is optionally absent.
19. The composition according to claim 1, wherein B1 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, B2 comprises an inverted abasic 2′-deoxyribose moiety, and B3 is optionally absent.
20. The composition according to claim 1, wherein B1 comprises one or more galactosamine moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, B2 comprises an inverted abasic 2′-deoxyribose moiety, and B3 is optionally absent.
21. The composition according to claim 1, wherein B1 comprises one or more N-acetylgalactosamine moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, B2 comprises an inverted abasic 2′-deoxyribose moiety, and B3 is optionally absent.
22. The composition according to claim 1, wherein B1 comprises one or more folate moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, B2 comprises an inverted abasic 2′-deoxyribose moiety, and B3 is optionally absent.
23. The composition according to claim 1, wherein B1 comprises one or more cholesterol moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, B2 comprises an inverted abasic 2′-deoxyribose moiety, and B3 is optionally absent.
24. The composition according to claim 1, wherein B1 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, B2 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B3 is optionally absent.
25. The composition according to claim 1, wherein B1 comprises one or more galactosamine moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, B2 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B3 is optionally absent.
26. The composition according to claim 1, wherein B1 comprises one or more N-acetylgalactosamine moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, B2 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B3 is optionally absent.
27. The composition according to claim 1, wherein B1 comprises one or more folate moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, B2 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B3 is optionally absent.
28. The composition according to claim 1, wherein B1 comprises one or more cholesterol moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, B2 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B3 is optionally absent.
29. The composition according to claim 1, wherein B1 comprises a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, B2 comprises any of SEQ ID NOS: 54-528 optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B3 is optionally absent.
30. The composition according to claim 1, wherein B1 comprises one or more galactosamine moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, B2 comprises any of SEQ ID NOS: 54-528 optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B3 is optionally absent.
31. The composition according to claim 1, wherein B1 comprises one or more N-acetylgalactosamine moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, B2 comprises any of SEQ ID NOS: 54-528 optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B3 is optionally absent.
32. The composition according to claim 1, wherein B1 comprises one or more folate moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, B2 comprises any of SEQ ID NOS: 54-528 optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B3 is optionally absent.
33. The composition according to claim 1, wherein B1 comprises one or more cholesterol moieties optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, B2 comprises any of SEQ ID NOS: 54-528 optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety optionally attached via a linker to an inverted abasic 2′-deoxyribose moiety, and B3 is optionally absent.
34. The composition according to claim 1, wherein S1 and S2 are required and S3 is optional.
35. The composition according to claim 1, wherein S1 and S3 are required and S2 is optional.
36. The composition according to claim 1, wherein S1, S2, and S3 are required.
37. The composition according to claim 1, wherein B1 comprises:
Figure US20150299696A1-20151022-C00039
wherein each L is independently a linker and GalNAc is an N-acetyl galactosamine moiety.
38. The composition according to claim 1, wherein B1 comprises:
Figure US20150299696A1-20151022-C00040
39. The composition according to claim 1, wherein B1 comprises:
Figure US20150299696A1-20151022-C00041
wherein n is an integer from 1 to 20.
40. The composition according to claim 1, wherein B1 or B2 comprises:
Figure US20150299696A1-20151022-C00042
wherein X is —O—, —S—, —CH2— or —NH—; n is 1, 2, 3, or 4; and the bond with “
Figure US20150299696A1-20151022-P00001
” indicates the point of attachment optionally including one or more linkers, which may be the same or different; and further optionally including one or more targeting ligands, solubilizing agents, pharmacokinetics enhancing agents, lipids, and/or masking agents.
41. A composition comprising:
a double-stranded short interfering nucleic acid (siNA) molecule that modulates expression of a target RNA sequence via RNA interference, wherein the molecule has a passenger strand and a guide strand and comprises structure selected from the structures represented by
Figure US20150299696A1-20151022-C00043
wherein,
(a) the upper strand is the passenger strand and the lower strand is the guide strand of the double-stranded nucleic acid molecule; the guide strand is complementary to the target sequence and the passenger strand is complementary to the guide strand, wherein one or more mismatches between the guide strand and passenger strand and/or between the guide strand and the target sequence are tolerated so long as RNAi activity is maintained;
(b) B1 and B2, are each independently a terminal cap optionally including a ligand, polymer, protein or peptide transduction domain, nuclear localization sequence, cell penetrating peptide, receptor, steroid, vitamin, antibody, protamine, and/or hormone, optionally attached via a linker;
(c) every N nucleotide is a 2′-deoxy-2′-fluoro nucleotide and every N nucleotide is a 2′-O-methyl nucleotide;
(d) each S is a phosphorothioate or phosphorodithioate internucleotide linkage.
42-46. (canceled)
47. The composition according to claim 41, wherein B1 comprises:
Figure US20150299696A1-20151022-C00044
wherein each L is independently a linker and GalNAc is an N-acetyl galactosamine moiety.
48. The composition according to claim 41, wherein B1 comprises:
Figure US20150299696A1-20151022-C00045
49. The composition according to claim 41, wherein B1 comprises:
Figure US20150299696A1-20151022-C00046
wherein n is an integer from 1 to 20.
50. The composition according to claim 41, wherein B1 or B2 comprises:
Figure US20150299696A1-20151022-C00047
wherein X is —O—, —S—, —CH2— or —NH—; n is 1, 2, 3, or 4; and the bond with “
Figure US20150299696A1-20151022-P00001
” indicates the point of attachment optionally including one or more linkers, which may be the same or different; and further optionally including one or more targeting ligands, solubilizing agents, pharmacokinetics enhancing agents, lipids, and/or masking agents.
51. The composition according to claim 1, further comprising a terminal phosphate group at the 5′-end of the guide strand.
52. A composition comprising the double-stranded short interfering nucleic acid (siNA) molecule according to claim 1, and a pharmaceutically acceptable carrier or diluent.
53. The composition according to claim 41, further comprising a terminal phosphate group at the 5′-end of the guide strand.
54. A composition comprising the double-stranded short interfering nucleic acid (siNA) molecule according to claim 41, and a pharmaceutically acceptable carrier or diluent.
US14/398,300 2012-05-02 2013-04-26 SHORT INTERFERING NUCLEIC ACID (siNA) COMPOSITIONS Abandoned US20150299696A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/398,300 US20150299696A1 (en) 2012-05-02 2013-04-26 SHORT INTERFERING NUCLEIC ACID (siNA) COMPOSITIONS

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201261641626P 2012-05-02 2012-05-02
PCT/US2013/038305 WO2013165816A2 (en) 2012-05-02 2013-04-26 SHORT INTERFERING NUCLEIC ACID (siNA) COMPOSITIONS
US14/398,300 US20150299696A1 (en) 2012-05-02 2013-04-26 SHORT INTERFERING NUCLEIC ACID (siNA) COMPOSITIONS

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/038305 A-371-Of-International WO2013165816A2 (en) 2012-05-02 2013-04-26 SHORT INTERFERING NUCLEIC ACID (siNA) COMPOSITIONS

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/684,902 Continuation US10689648B2 (en) 2012-05-02 2017-08-23 Short interfering nucleic acid (siNA) compositions

Publications (1)

Publication Number Publication Date
US20150299696A1 true US20150299696A1 (en) 2015-10-22

Family

ID=49515013

Family Applications (4)

Application Number Title Priority Date Filing Date
US14/398,300 Abandoned US20150299696A1 (en) 2012-05-02 2013-04-26 SHORT INTERFERING NUCLEIC ACID (siNA) COMPOSITIONS
US15/684,902 Active US10689648B2 (en) 2012-05-02 2017-08-23 Short interfering nucleic acid (siNA) compositions
US16/793,790 Active US11530407B2 (en) 2012-05-02 2020-02-18 Short interfering nucleic acid (siNA) compositions
US17/984,486 Pending US20230348904A1 (en) 2012-05-02 2022-11-10 SHORT INTERFERING NUCLEIC ACID (siNA) COMPOSITIONS

Family Applications After (3)

Application Number Title Priority Date Filing Date
US15/684,902 Active US10689648B2 (en) 2012-05-02 2017-08-23 Short interfering nucleic acid (siNA) compositions
US16/793,790 Active US11530407B2 (en) 2012-05-02 2020-02-18 Short interfering nucleic acid (siNA) compositions
US17/984,486 Pending US20230348904A1 (en) 2012-05-02 2022-11-10 SHORT INTERFERING NUCLEIC ACID (siNA) COMPOSITIONS

Country Status (3)

Country Link
US (4) US20150299696A1 (en)
EP (4) EP3453762B1 (en)
WO (1) WO2013165816A2 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20170081425A1 (en) * 2013-11-06 2017-03-23 Merck Sharp & Dohme Corp. Peptide containing conjugates for dual molecular delivery of oligonucleotides
US20190078089A1 (en) * 2017-09-14 2019-03-14 Arrowhead Pharmaceuticals, Inc. RNAi Agents And Compositions for Inhibiting Expression of Angiopoietin-Like 3 (ANGPTL3), And Methods Of Use
WO2021072139A1 (en) 2019-10-11 2021-04-15 Massachusetts Institute Of Technology Formulations for gastrointestinal delivery of oligonucleotides
US11021503B2 (en) * 2017-05-23 2021-06-01 Hoffmann-La Roche Inc. Process for GalNAc oligonucleotide conjugates
EP4035659A1 (en) 2016-11-29 2022-08-03 PureTech LYT, Inc. Exosomes for delivery of therapeutic agents
US11530407B2 (en) 2012-05-02 2022-12-20 Sirna Therapeutics, Inc. Short interfering nucleic acid (siNA) compositions

Families Citing this family (53)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9657294B2 (en) 2002-02-20 2017-05-23 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US9181551B2 (en) 2002-02-20 2015-11-10 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
JP6106085B2 (en) 2010-08-24 2017-03-29 サーナ・セラピューティクス・インコーポレイテッドSirna Therapeutics,Inc. Single-stranded RNAi agent containing an internal non-nucleic acid spacer
US9260471B2 (en) 2010-10-29 2016-02-16 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using short interfering nucleic acids (siNA)
BR122019026068B8 (en) 2011-06-21 2022-10-18 Alnylam Pharmaceuticals DOUBLE-STRAND RIBONUCLEIC ACID (DSRNA) FOR INHIBITING ANGPTL3 EXPRESSION AND ITS USE, PHARMACEUTICAL COMPOSITION AND IN VITRO METHOD FOR INHIBITING ANGPTL3 EXPRESSION IN A CELL
EP2751270B1 (en) 2011-08-29 2018-08-22 Ionis Pharmaceuticals, Inc. Oligomer-conjugate complexes and their use
AR090905A1 (en) * 2012-05-02 2014-12-17 Merck Sharp & Dohme CONJUGATES CONTAINING TETRAGALNAC AND PEPTIDES AND PROCEDURES FOR THE ADMINISTRATION OF OLIGONUCLEOTIDES, PHARMACEUTICAL COMPOSITION
JP6373762B2 (en) * 2012-12-28 2018-08-15 国立大学法人東北大学 Peptides for intracellular introduction of target molecules
KR102212275B1 (en) 2013-05-01 2021-02-05 아이오니스 파마수티컬즈, 인코포레이티드 Compositions and methods for modulating hbv and ttr expression
US9926556B2 (en) 2014-04-28 2018-03-27 Ionis Pharmaceuticals, Inc. Linkage modified oligomeric compounds
EP3137604B1 (en) 2014-05-01 2020-07-15 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating growth hormone receptor expression
BR112016022593B1 (en) 2014-05-01 2022-04-26 Ionis Pharmaceuticals, Inc Oligomeric compounds, compositions comprising them, and uses thereof
HUE052709T2 (en) 2014-05-01 2021-05-28 Ionis Pharmaceuticals Inc Conjugates of modified antisense oligonucleotides and their use for modulating pkk expression
MX2016014102A (en) 2014-05-01 2017-05-03 Ionis Pharmaceuticals Inc Compositions and methods for modulating angiopoietin-like 3 expression.
US10570169B2 (en) 2014-05-22 2020-02-25 Ionis Pharmaceuticals, Inc. Conjugated antisense compounds and their use
EP3736334A1 (en) 2014-07-16 2020-11-11 Arrowhead Pharmaceuticals, Inc. Rnai compositions to treat apoc3-related diseases
JP2017535552A (en) * 2014-11-17 2017-11-30 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Alnylam Pharmaceuticals, Inc. Apolipoprotein C3 (APOC3) iRNA composition and methods of use thereof
US9809817B2 (en) 2015-04-03 2017-11-07 University Of Massachusetts Oligonucleotide compounds for targeting huntingtin mRNA
DK3277815T3 (en) 2015-04-03 2021-12-13 Univ Massachusetts OLIGONUCLEOTIDE COMPOUNDS FOR THE TREATMENT OF PRE-CLAMPSY AND OTHER ANGIOGENIC DISORDERS
JP6892433B2 (en) * 2015-04-03 2021-06-23 ユニバーシティ・オブ・マサチューセッツUniversity Of Massachusetts Well-stabilized asymmetric SIRNA
BR112017021053A2 (en) * 2015-04-13 2018-08-14 Alnylam Pharmaceuticals Inc angiopoietin type 3 (angpt13) irna compositions and methods of use thereof
EP4092119A3 (en) 2015-07-10 2023-03-22 Ionis Pharmaceuticals, Inc. Modulators of diacyglycerol acyltransferase 2 (dgat2)
US10633653B2 (en) 2015-08-14 2020-04-28 University Of Massachusetts Bioactive conjugates for oligonucleotide delivery
CN108513588A (en) 2015-09-24 2018-09-07 Ionis制药公司 The conditioning agent of KRAS expression
CN113952353A (en) 2015-11-06 2022-01-21 Ionis制药公司 Modulation of apolipoprotein (a) expression
EP4119569A1 (en) 2015-11-06 2023-01-18 Ionis Pharmaceuticals, Inc. Conjugated antisense compounds for use in therapy
JP6853193B2 (en) 2016-01-29 2021-03-31 協和キリン株式会社 Nucleic acid complex
JP2019503394A (en) 2016-01-31 2019-02-07 ユニバーシティ・オブ・マサチューセッツUniversity Of Massachusetts Branched oligonucleotide
JOP20170057B1 (en) 2016-03-07 2022-03-14 Arrowhead Pharmaceuticals Inc Targeting Ligands For Therapeutic Compounds
MA45470A (en) 2016-04-01 2019-02-06 Avidity Biosciences Llc KRAS NUCLEIC ACIDS AND THEIR USES
MA45469A (en) 2016-04-01 2019-02-06 Avidity Biosciences Llc BETA-CATENIN NUCLEIC ACIDS AND THEIR USES
HRP20221378T1 (en) 2016-07-15 2023-01-06 Ionis Pharmaceuticals, Inc. Compounds and methods for modulation of smn2
US11753638B2 (en) 2016-08-12 2023-09-12 University Of Massachusetts Conjugated oligonucleotides
JOP20170056B1 (en) 2016-09-02 2021-08-17 Arrowhead Pharmaceuticals Inc Targeting Ligands
US11400161B2 (en) 2016-10-06 2022-08-02 Ionis Pharmaceuticals, Inc. Method of conjugating oligomeric compounds
JOP20190215A1 (en) 2017-03-24 2019-09-19 Ionis Pharmaceuticals Inc Modulators of pcsk9 expression
PE20201283A1 (en) 2017-09-11 2020-11-24 Arrowhead Pharmaceuticals Inc IRNA AGENTS AND COMPOSITIONS TO INHIBIT THE EXPRESSION OF APOLIPOPROTEIN C-III (APOC3)
TW201934129A (en) 2018-01-15 2019-09-01 美商Ionis製藥公司 Modulators of DNM2 expression
US11332733B2 (en) 2018-02-12 2022-05-17 lonis Pharmaceuticals, Inc. Modified compounds and uses thereof
US20210355497A1 (en) 2018-05-09 2021-11-18 Ionis Pharmaceuticals, Inc. Compounds and methods for reducing fxi expression
WO2019226940A1 (en) * 2018-05-24 2019-11-28 Sirnaomics, Inc. Composition and methods of controllable co-coupling polypeptide nanoparticle delivery system for nucleic acid therapeutics
WO2020033748A1 (en) 2018-08-08 2020-02-13 Arcturus Therapeutics, Inc. Compositions and agents against nonalcoholic steatohepatitis
KR20210093227A (en) 2018-08-10 2021-07-27 유니버시티 오브 매사추세츠 Modified oligonucleotides targeting SNPs
TW202023573A (en) 2018-09-19 2020-07-01 美商Ionis製藥公司 Modulators of pnpla3 expression
JP2022552249A (en) 2019-10-14 2022-12-15 アストラゼネカ・アクチエボラーグ Modulators of PNPLA3 expression
JP2023514336A (en) 2020-02-18 2023-04-05 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 Apolipoprotein C3 (APOC3) iRNA compositions and methods of use thereof
AU2021225957A1 (en) 2020-02-28 2022-09-08 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating SMN2
CN116194073A (en) 2020-07-21 2023-05-30 化美有限责任公司 Diester cosmetic formulations and uses thereof
IL302817A (en) 2020-11-18 2023-07-01 Ionis Pharmaceuticals Inc Compounds and methods for modulating angiotensinogen expression
IL304880A (en) 2021-02-12 2023-10-01 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
IL305414A (en) 2021-03-04 2023-10-01 Alnylam Pharmaceuticals Inc Angiopoietin-like 3 (angptl3) irna compositions and methods of use thereof
IL309334A (en) 2021-06-23 2024-02-01 Univ Massachusetts Optimized anti-flt1 oligonucleotide compounds for treatment of preeclampsia and other angiogenic disorders
US11879125B2 (en) 2022-03-16 2024-01-23 Empirico Inc. GalNAc compositions for improving siRNA bioavailability

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090176725A1 (en) * 2005-08-17 2009-07-09 Sirna Therapeutics Inc. Chemically modified short interfering nucleic acid molecules that mediate rna interference

Family Cites Families (416)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3126375A (en) 1964-03-24 Chioacyl
US2789118A (en) 1956-03-30 1957-04-16 American Cyanamid Co 16-alpha oxy-belta1, 4-pregnadienes
US2990401A (en) 1958-06-18 1961-06-27 American Cyanamid Co 11-substituted 16alpha, 17alpha-substituted methylenedioxy steroids
US3048581A (en) 1960-04-25 1962-08-07 Olin Mathieson Acetals and ketals of 16, 17-dihydroxy steroids
US3749712A (en) 1970-09-25 1973-07-31 Sigma Tau Ind Farmaceuti Triamcinolone acetonide esters and process for their preparation
US3996359A (en) 1972-05-19 1976-12-07 Ab Bofors Novel stereoisomeric component A of stereoisomeric mixtures of 2'-unsymmetrical 16,17-methylenedioxy steroid 21-acylates, compositions thereof, and method of treating therewith
SE378109B (en) 1972-05-19 1975-08-18 Bofors Ab
SE378110B (en) 1972-05-19 1975-08-18 Bofors Ab
US4294926A (en) 1979-06-15 1981-10-13 Merck & Co., Inc. Hypocholesteremic fermentation products and process of preparation
US4319039A (en) 1979-06-15 1982-03-09 Merck & Co., Inc. Preparation of ammonium salt of hypocholesteremic fermentation product
US4231938A (en) 1979-06-15 1980-11-04 Merck & Co., Inc. Hypocholesteremic fermentation products and process of preparation
US4444784A (en) 1980-08-05 1984-04-24 Merck & Co., Inc. Antihypercholesterolemic compounds
MX7065E (en) 1980-06-06 1987-04-10 Sankyo Co A MICROBIOLOGICAL PROCEDURE FOR PREPARING DERIVATIVES OF ML-236B
JPS5889191A (en) 1981-11-20 1983-05-27 Sankyo Co Ltd Preparation of 3-hydroxy-ml-236b derivative
US5354772A (en) 1982-11-22 1994-10-11 Sandoz Pharm. Corp. Indole analogs of mevalonolactone and derivatives thereof
US4501729A (en) 1982-12-13 1985-02-26 Research Corporation Aerosolized amiloride treatment of retained pulmonary secretions
US4911165A (en) 1983-01-12 1990-03-27 Ethicon, Inc. Pliabilized polypropylene surgical filaments
US4751180A (en) 1985-03-28 1988-06-14 Chiron Corporation Expression using fused genes providing for protein product
US4935233A (en) 1985-12-02 1990-06-19 G. D. Searle And Company Covalently linked polypeptide cell modulators
US4681893A (en) 1986-05-30 1987-07-21 Warner-Lambert Company Trans-6-[2-(3- or 4-carboxamido-substituted pyrrol-1-yl)alkyl]-4-hydroxypyran-2-one inhibitors of cholesterol synthesis
US4782084A (en) 1987-06-29 1988-11-01 Merck & Co., Inc. HMG-COA reductase inhibitors
US4885314A (en) 1987-06-29 1989-12-05 Merck & Co., Inc. Novel HMG-CoA reductase inhibitors
US4820850A (en) 1987-07-10 1989-04-11 Merck & Co., Inc. Process for α-C-alkylation of the 8-acyl group on mevinolin and analogs thereof
US5180589A (en) 1988-03-31 1993-01-19 E. R. Squibb & Sons, Inc. Pravastatin pharmaceuatical compositions having good stability
US5030447A (en) 1988-03-31 1991-07-09 E. R. Squibb & Sons, Inc. Pharmaceutical compositions having good stability
US4916239A (en) 1988-07-19 1990-04-10 Merck & Co., Inc. Process for the lactonization of mevinic acids and analogs thereof
EP0360390A1 (en) 1988-07-25 1990-03-28 Glaxo Group Limited Spirolactam derivatives
US5118853A (en) 1988-10-13 1992-06-02 Sandoz Ltd. Processes for the synthesis of 3-disubstituted aminoacroleins
US5290946A (en) 1988-10-13 1994-03-01 Sandoz Ltd. Processes for the synthesis of 3-(substituted indolyl-2-yl)propenaldehydes
WO1990005525A1 (en) 1988-11-23 1990-05-31 Pfizer Inc. Quinuclidine derivatives as substance p antagonists
US4929437A (en) 1989-02-02 1990-05-29 Merck & Co., Inc. Coenzyme Q10 with HMG-CoA reductase inhibitors
US5164372A (en) 1989-04-28 1992-11-17 Fujisawa Pharmaceutical Company, Ltd. Peptide compounds having substance p antagonism, processes for preparation thereof and pharmaceutical composition comprising the same
US5189164A (en) 1989-05-22 1993-02-23 Sandoz Ltd. Processes for the synthesis of syn-(E)-3,5-dihydroxy-7-substituted hept-6-enoic and heptanoic acids and derivatives and intermediates thereof
FI94339C (en) 1989-07-21 1995-08-25 Warner Lambert Co Process for the preparation of pharmaceutically acceptable [R- (R *, R *)] - 2- (4-fluorophenyl) -, - dihydroxy-5- (1-methylethyl) -3-phenyl-4 - [(phenylamino) carbonyl] -1H- for the preparation of pyrrole-1-heptanoic acid and its pharmaceutically acceptable salts
PH27357A (en) 1989-09-22 1993-06-21 Fujisawa Pharmaceutical Co Pyrazole derivatives and pharmaceutical compositions comprising the same
US5286634A (en) 1989-09-28 1994-02-15 Stadler Joan K Synergistic method for host cell transformation
FI97540C (en) 1989-11-06 1997-01-10 Sanofi Sa Process for the preparation of therapeutically useful aromatically substituted piperidine and piperazine derivatives
FR2654725B1 (en) 1989-11-23 1992-02-14 Rhone Poulenc Sante NEW ISOINDOLONE DERIVATIVES, THEIR PREPARATION AND THE PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
FR2654726B1 (en) 1989-11-23 1992-02-14 Rhone Poulenc Sante NEW ISOINDOLONE DERIVATIVES AND THEIR PREPARATION.
GB8929070D0 (en) 1989-12-22 1990-02-28 Fujisawa Pharmaceutical Co Peptide compounds,processes for preparation thereof and pharmaceutical composition comprising the same
US5232929A (en) 1990-11-28 1993-08-03 Pfizer Inc. 3-aminopiperidine derivatives and related nitrogen containing heterocycles and pharmaceutical compositions and use
UA41251C2 (en) 1990-01-04 2001-09-17 Пфайзер, Інк. Hydrogenated nitrogen-containing heterocyclic substances, piperidine derivatives, pharmaceutical composition and method for inhibiting activity of p substance
US6153737A (en) 1990-01-11 2000-11-28 Isis Pharmaceuticals, Inc. Derivatized oligonucleotides having improved uptake and other properties
US5321032A (en) 1990-02-15 1994-06-14 Fujisawa Pharmaceutical Co., Ltd. Peptide compounds and pharmaceutical compositions thereof
US5214136A (en) 1990-02-20 1993-05-25 Gilead Sciences, Inc. Anthraquinone-derivatives oligonucleotides
US5420245A (en) 1990-04-18 1995-05-30 Board Of Regents, The University Of Texas Tetrapeptide-based inhibitors of farnesyl transferase
DE69105131T2 (en) 1990-06-01 1995-03-23 Pfizer 3-AMINO-2-ARYLCHINUCLIDINE, METHOD FOR THE PRODUCTION THEREOF AND PHARMACEUTICAL PREPARATIONS CONTAINING THEM.
ATE116317T1 (en) 1990-07-23 1995-01-15 Pfizer QUINUCLIDINE DERIVATIVES.
US5218105A (en) 1990-07-27 1993-06-08 Isis Pharmaceuticals Polyamine conjugated oligonucleotides
US5138045A (en) 1990-07-27 1992-08-11 Isis Pharmaceuticals Polyamine conjugated oligonucleotides
BR9106905A (en) 1990-09-28 1993-08-17 Pfizer CONDENSATE RING ANALOGS OF NON-AROMATIC HETEROCYCLES CONTAINING NITROGEN
GB9023116D0 (en) 1990-10-24 1990-12-05 Fujisawa Pharmaceutical Co Peptide compounds,processes for preparation thereof and pharmaceutical composition comprising the same
DE69114117T2 (en) 1990-12-21 1996-03-21 Fujisawa Pharmaceutical Co New use of peptide derivative.
AU652407B2 (en) 1991-01-10 1994-08-25 Pfizer Inc. N-alkyl quinuclidinium salts as substance P antagonists
EP0499313B1 (en) 1991-02-11 1997-06-11 MERCK SHARP & DOHME LTD. Azabicyclic compounds, pharmaceutical compositions containing them and their use in therapy
US5373003A (en) 1991-03-01 1994-12-13 Pfizer Inc. 1-azabicyclo[3.2.2]nonan-3-amine derivatives
US5747469A (en) 1991-03-06 1998-05-05 Board Of Regents, The University Of Texas System Methods and compositions comprising DNA damaging agents and p53
JPH0794440B2 (en) 1991-03-26 1995-10-11 フアイザー・インコーポレイテツド Stereoselective production of substituted piperidines
FR2677361A1 (en) 1991-06-04 1992-12-11 Adir NOVEL PEPTIDES AND PSEUDOPEPTIDES, TACHYKININ DERIVATIVES, PROCESS FOR PREPARING THEM AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
FR2676055B1 (en) 1991-05-03 1993-09-03 Sanofi Elf AMINO POLYCYCLIC COMPOUNDS AND THEIR ENANTIOMERS, PROCESS FOR THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
FR2676053B1 (en) 1991-05-03 1993-08-27 Sanofi Elf NOVEL DIALKYLENEPIPERIDINO COMPOUNDS AND THEIR ENANTIOMERS, PROCESS FOR THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
FR2676446B1 (en) 1991-05-17 1993-08-06 Rhone Poulenc Rorer Sa NOVEL THIOPYRANOPYRROLE DERIVATIVES, THEIR PREPARATION AND THE PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
FR2676443B1 (en) 1991-05-17 1993-08-06 Rhone Poulenc Rorer Sa NOVEL PERHYDROISOINDOLE DERIVATIVES AND THEIR PREPARATION.
FR2676442B1 (en) 1991-05-17 1993-08-06 Rhone Poulenc Rorer Sa NEW PERHYDROISOINDOLE DERIVATIVES, THEIR PREPARATION AND THE PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
FR2676447B1 (en) 1991-05-17 1993-08-06 Rhone Poulenc Rorer Sa NOVEL THIOPYRANOPYRROLE DERIVATIVES AND THEIR PREPARATION.
AU658898B2 (en) 1991-05-22 1995-05-04 Pfizer Inc. Substituted 3-aminoquinuclidines
WO1992020661A1 (en) 1991-05-22 1992-11-26 Merck & Co., Inc. N, n-diacylpiperazines
DE122006000066I1 (en) 1991-05-31 2007-03-22 Pfizer quinuclidine derivatives
GB9113219D0 (en) 1991-06-19 1991-08-07 Fujisawa Pharmaceutical Co Peptide compound,processes for preparation thereof and pharmaceutical composition comprising the same
WO1993000331A1 (en) 1991-06-20 1993-01-07 Pfizer Inc. Fluoroalkoxybenzylamino derivatives of nitrogen containing heterocycles
TW202432B (en) 1991-06-21 1993-03-21 Pfizer
US5288730A (en) 1991-06-24 1994-02-22 Merck Sharp & Dohme Limited Azabicyclic compounds, pharmaceutical compositions containing them and their use in therapy
US5472978A (en) 1991-07-05 1995-12-05 Merck Sharp & Dohme Ltd. Aromatic compounds, pharmaceutical compositions containing them and their use in therapy
ATE133657T1 (en) 1991-07-05 1996-02-15 Merck Sharp & Dohme AROMATIC COMPOUNDS, PHARMACEUTICAL COMPOSITIONS CONTAINING THEM AND THEIR THERAPEUTIC USE
EP0536817A1 (en) 1991-07-05 1993-04-14 MERCK SHARP & DOHME LTD. Azabicyclic compounds as tachykinin antagonists
US5610183A (en) 1991-07-10 1997-03-11 Merck, Sharp & Dohme Ltd. Phenylglycine derivatives pharmaceutical compositions containing them and their use in therapy
US5495047A (en) 1991-07-10 1996-02-27 Merck, Sharp & Dohme (Ltd.) Fused tricyclic compounds, pharmaceutical compositions containing them and their use in therapy
MY110227A (en) 1991-08-12 1998-03-31 Ciba Geigy Ag 1-acylpiperindine compounds.
AU661711B2 (en) 1991-08-20 1995-08-03 Merck Sharp & Dohme Limited Azacyclic compounds, processes for their preparation and pharmaceutical compositions containing them
ES2149767T5 (en) 1991-09-20 2005-06-16 Glaxo Group Limited NEW MEDICAL USE FOR TAQUIQUININE ANTAGONISTS.
HU9400728D0 (en) 1991-09-26 1994-06-28 Pfizer Fused tricyclic nitrogen containing heterocycles as substance p receptor antagonists
US6335434B1 (en) 1998-06-16 2002-01-01 Isis Pharmaceuticals, Inc., Nucleosidic and non-nucleosidic folate conjugates
WO1993009116A1 (en) 1991-11-07 1993-05-13 Yoshitomi Pharmaceutical Industries, Ltd. Quinuclidine compound and medicinal use thereof
EP0613458B1 (en) 1991-11-12 1998-01-07 Pfizer Inc. Acyclic ethylenediamine derivatives as substance p receptor antagonists
EP0545478A1 (en) 1991-12-03 1993-06-09 MERCK SHARP & DOHME LTD. Heterocyclic compounds as tachykinin antagonists
HU217629B (en) 1991-12-12 2000-03-28 Novartis Ag. Process for producing stabilized pharmaceutical compositions comprising fluvastatin
GB9200535D0 (en) 1992-01-10 1992-02-26 Fujisawa Pharmaceutical Co New compound
GB9201179D0 (en) 1992-01-21 1992-03-11 Glaxo Group Ltd Chemical compounds
US5328927A (en) 1992-03-03 1994-07-12 Merck Sharpe & Dohme, Ltd. Hetercyclic compounds, processes for their preparation and pharmaceutical compositions containing them
JP2656702B2 (en) 1992-03-23 1997-09-24 ファイザー製薬株式会社 Peptide quinuclidine
FR2689888B1 (en) 1992-04-10 1994-06-10 Rhone Poulenc Rorer Sa NOVEL PERHYDROISOINDOLE DERIVATIVES, THEIR PREPARATION AND THE PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
CA2133077A1 (en) 1992-04-15 1993-10-28 Raymond Baker Azacyclic compounds
GB2266529A (en) 1992-05-01 1993-11-03 Merck Sharp & Dohme Tetrahydroisoquinoline derivatives
AU687736B2 (en) 1992-05-11 1998-03-05 Ribozyme Pharmaceuticals, Inc. Method and reagent for inhibiting viral replication
US5977343A (en) 1992-05-14 1999-11-02 Ribozyme Pharmaceuticals, Inc. Synthesis, deprotection, analysis and purification of RNA and ribozymes
PT641328E (en) 1992-05-18 2002-04-29 Pfizer AZA-BICYCLIC DERIVATIVES IN BRIDGE AS ANTAGONISTS OF SUBSTANCE P
GB9211193D0 (en) 1992-05-27 1992-07-08 Merck Sharp & Dohme Therapeutic agents
US5719147A (en) 1992-06-29 1998-02-17 Merck & Co., Inc. Morpholine and thiomorpholine tachykinin receptor antagonists
US5637699A (en) 1992-06-29 1997-06-10 Merck & Co., Inc. Process for preparing morpholine tachykinin receptor antagonists
CA2099233A1 (en) 1992-06-29 1993-12-30 Conrad P. Dorn Morpholine and thiomorpholine tachykinin receptor antagonists
WO1994001402A1 (en) 1992-07-13 1994-01-20 Merck Sharp & Dohme Limited Heterocyclic amide derivatives as tachykinin derivatives
JPH07509133A (en) 1992-07-17 1995-10-12 リボザイム・ファーマシューティカルズ・インコーポレイテッド Methods and agents for the treatment of animal diseases
GB2268931A (en) 1992-07-22 1994-01-26 Merck Sharp & Dohme Azabicyclic tachykinin-receptor antagonists
AU675447B2 (en) 1992-07-28 1997-02-06 Merck Sharp & Dohme Limited Carboxamidomethyl piperidine and analogues
GB2269170A (en) 1992-07-29 1994-02-02 Merck Sharp & Dohme Azatricyclic tachykinin antagonists
AU4718093A (en) 1992-07-31 1994-03-03 Merck Sharp & Dohme Limited Substituted amines as tachykinin receptor antagonists
WO1994003445A1 (en) 1992-08-04 1994-02-17 Pfizer Inc. 3-benzylamino-2-phenyl-piperidine derivatives as substance p receptor antagonists
GB9216911D0 (en) 1992-08-10 1992-09-23 Merck Sharp & Dohme Therapeutic agents
EP0655055B1 (en) 1992-08-13 2000-11-29 Warner-Lambert Company Tachykinin antagonists
DK0655996T3 (en) 1992-08-19 2001-12-27 Pfizer Substituted benzylamino nitrogen-containing, non-aromatic heterocyclic compounds
US5387595A (en) 1992-08-26 1995-02-07 Merck & Co., Inc. Alicyclic compounds as tachykinin receptor antagonists
ATE161530T1 (en) 1992-09-04 1998-01-15 Takeda Chemical Industries Ltd CONDENSED HETEROCYCLIC COMPOUNDS, THEIR PRODUCTION AND USE
AU4973693A (en) 1992-09-10 1994-03-29 Merck Sharp & Dohme Limited Alcohols and ethers with aromatic substituents as tachykinin-antagonists
US6235886B1 (en) 1993-09-03 2001-05-22 Isis Pharmaceuticals, Inc. Methods of synthesis and use
GB9220286D0 (en) 1992-09-25 1992-11-11 Merck Sharp & Dohme Therapeutic agents
JP2656699B2 (en) 1992-10-21 1997-09-24 ファイザー製薬株式会社 Substituted benzylaminoquinuclidine
GB9222262D0 (en) 1992-10-23 1992-12-09 Merck Sharp & Dohme Therapeutic agents
GB9222486D0 (en) 1992-10-26 1992-12-09 Merck Sharp & Dohme Therapeutic agents
AU678409B2 (en) 1992-10-28 1997-05-29 Merck Sharp & Dohme Limited 4-arylmethyloxymethyl piperidines as tachykinin antagonists
JP2656700B2 (en) 1992-10-28 1997-09-24 ファイザー製薬株式会社 Substituted quinuclidine derivatives
AU5342894A (en) 1992-10-30 1994-05-24 Merck Sharp & Dohme Limited Tachykinin antagonists
ES2096312T3 (en) 1992-11-12 1997-03-01 Pfizer QUINUCLIDINE DERIVATIVE AS ANTAGONIST OF THE SUBSTANCE P.
US5261188A (en) 1992-11-23 1993-11-16 The Standard Products Company Belt weatherstrip with bulb
WO1994013663A1 (en) 1992-12-10 1994-06-23 Pfizer Inc. Aminomethylene substituted non-aromatic heterocycles and use as substance p antagonists
US5604260A (en) 1992-12-11 1997-02-18 Merck Frosst Canada Inc. 5-methanesulfonamido-1-indanones as an inhibitor of cyclooxygenase-2
AU682838B2 (en) 1992-12-14 1997-10-23 Merck Sharp & Dohme Limited 4-aminomethyl/thiomethyl/sulfonylmethyl-4-phenylpiperidines as tachykinin receptor antagonists
CA2111902A1 (en) 1992-12-21 1994-06-22 Jack Beuford Campbell Antitumor compositions and methods of treatment
GB9226581D0 (en) 1992-12-21 1993-02-17 Merck Sharp & Dohme Therapeutic agents
GB9300051D0 (en) 1993-01-04 1993-03-03 Merck Sharp & Dohme Therapeutic agents
CA2152792C (en) 1993-01-15 2000-02-15 Stephen R. Bertenshaw Novel 3,4-diaryl thiophenes and analogs thereof having use as antiinflammatory agents
EP0610793A1 (en) 1993-02-08 1994-08-17 Takeda Chemical Industries, Ltd. Tetracyclic morpholine derivatives and their use or analgesics
ES2116583T3 (en) 1993-02-18 1998-07-16 Merck Sharp & Dohme AZACICLIC COMPOUNDS, COMPOSITIONS CONTAINING THEM AND THEIR USE AS TACHYCIN ANTAGONISTS.
WO1994019320A1 (en) 1993-02-22 1994-09-01 Merck Sharp & Dohme Limited Aromatic compounds, compositions containing them and their use in therapy
WO1994019357A1 (en) 1993-02-23 1994-09-01 Merrell Dow Pharmaceuticals Inc. Farnesyl:protein transferase inhibitors as anticancer agents
US5298627A (en) 1993-03-03 1994-03-29 Warner-Lambert Company Process for trans-6-[2-(substituted-pyrrol-1-yl)alkyl]pyran-2-one inhibitors of cholesterol synthesis
US5688806A (en) 1993-03-04 1997-11-18 Pfizer Inc. Spiroazacyclic derivatives as substance P antagonists
US5409944A (en) 1993-03-12 1995-04-25 Merck Frosst Canada, Inc. Alkanesulfonamido-1-indanone derivatives as inhibitors of cyclooxygenase
EP0689424B1 (en) 1993-03-17 1998-10-14 Minnesota Mining And Manufacturing Company Aerosol formulation containing an ester-, amide-, or mercaptoester-derived dispersing aid
CA2118985A1 (en) 1993-04-02 1994-10-03 Dinesh V. Patel Heterocyclic inhibitors of farnesyl protein transferase
US5496833A (en) 1993-04-13 1996-03-05 Merck Sharp & Dohme Limited Piperidine tachykinin receptor antagonists
DE69405862T2 (en) 1993-05-06 1998-01-15 Merrell Pharma Inc SUBSTITUTED PYRROLIDIN-3-ALKYL-PIPERIDINE USEFUL AS TACHYKININ ANTAGONISTS
JPH09500615A (en) 1993-05-14 1997-01-21 ジェネンテク,インコーポレイテッド Ras-farnesyl transferase inhibitor
US5602098A (en) 1993-05-18 1997-02-11 University Of Pittsburgh Inhibition of farnesyltransferase
IL109646A0 (en) 1993-05-19 1994-08-26 Pfizer Heteroatom substituted alkyl benzylamino-quinuclidines
US5380738A (en) 1993-05-21 1995-01-10 Monsanto Company 2-substituted oxazoles further substituted by 4-fluorophenyl and 4-methylsulfonylphenyl as antiinflammatory agents
JPH08511522A (en) 1993-06-07 1996-12-03 メルク エンド カンパニー インコーポレーテッド Spiro-Substituted Aza Rings as Neurokinin Antagonists
US5474995A (en) 1993-06-24 1995-12-12 Merck Frosst Canada, Inc. Phenyl heterocycles as cox-2 inhibitors
US5436265A (en) 1993-11-12 1995-07-25 Merck Frosst Canada, Inc. 1-aroyl-3-indolyl alkanoic acids and derivatives thereof useful as anti-inflammatory agents
GB9602877D0 (en) 1996-02-13 1996-04-10 Merck Frosst Canada Inc 3,4-Diaryl-2-hydroxy-2,5- dihydrofurans as prodrugs to cox-2 inhibitors
EP0634402A1 (en) 1993-07-14 1995-01-18 Takeda Chemical Industries, Ltd. Isochinolinone derivatives, their production and use
US5869499A (en) 1993-07-15 1999-02-09 Pfizer Inc Benzyloxyquinuclidines as substance P antagonists
TW365603B (en) 1993-07-30 1999-08-01 Rhone Poulenc Rorer Sa Novel perhydroisoindole derivatives, their preparation and pharmaceutical compositions which contain them
GB9315808D0 (en) 1993-07-30 1993-09-15 Merck Sharp & Dohme Therapeutic agents
GB9317987D0 (en) 1993-08-26 1993-10-13 Glaxo Group Ltd Chemical compounds
EP0728139B1 (en) 1993-09-03 2003-08-13 Isis Pharmaceuticals, Inc. Amine-derivatized nucleosides and oligonucleosides
PT719253E (en) 1993-09-17 2004-07-30 Pfizer 3-AMINO-5-CARBOXY-SUBSTITUTED PIPERIDINS AND 3-AMINO-4-CARBOXY-SUBSTITUTED PYRROLIDINES AS TACHYCININ ANTAGONISTS
US5703065A (en) 1993-09-17 1997-12-30 Pfizer Inc. Heteroarylamino and heteroarylsulfonamido substituted 3-benyzlaminomethyl piperidines and related compounds
EP0670314A4 (en) 1993-09-22 1996-04-10 Kyowa Hakko Kogyo Kk Farnesyltransferase inhibitor.
IL111002A (en) 1993-09-22 1998-09-24 Glaxo Group Ltd Piperidine derivatives their preparation and pharmaceutical compositions containing them
US5661152A (en) 1993-10-15 1997-08-26 Schering Corporation Tricyclic sulfonamide compounds useful for inhibition of G-protein function and for treatment of proliferative diseases
NZ274749A (en) 1993-10-15 1998-05-27 Schering Corp Tricyclic carbamate derivatives useful for inhibition of g-protein function and for treating proliferative diseases, medicaments
AU698960B2 (en) 1993-10-15 1998-11-12 Schering Corporation Tricyclic sulfonamide compounds useful for inhibition of g-protein function and for treatment of proliferative diseases
US5719148A (en) 1993-10-15 1998-02-17 Schering Corporation Tricyclic amide and urea compounds useful for inhibition of g-protein function and for treatment of proliferative diseases
IL111235A (en) 1993-10-15 2001-03-19 Schering Plough Corp Pharmaceutical compositions for inhibition of g-protein function and for treatment of proliferative diseases containing tricyclic compounds some such compounds and process for preparing part of them
US5721236A (en) 1993-10-15 1998-02-24 Schering Corporation Tricyclic carbamate compounds useful for inhibition of G-protein function and for treatment of proliferative diseases
WO1995011917A1 (en) 1993-10-25 1995-05-04 Parke, Davis & Company Substituted tetra- and pentapeptide inhibitors of protein:farnesyl transferase
WO1995011880A1 (en) 1993-10-27 1995-05-04 Merck Sharp & Dohme Limited Substituted amides as tachykinin antagonists
US5344991A (en) 1993-10-29 1994-09-06 G.D. Searle & Co. 1,2 diarylcyclopentenyl compounds for the treatment of inflammation
ATE177420T1 (en) 1993-11-04 1999-03-15 Abbott Lab CYCLOBUTANE DERIVATIVES AS INHIBITORS OF SQUALENE SYNTHETASE AND PROTEIN FARNESYL TRANSFERASE
US5783593A (en) 1993-11-04 1998-07-21 Abbott Laboratories Inhibitors of squalene synthetase and protein farnesyltransferase
WO1995012612A1 (en) 1993-11-05 1995-05-11 Warner-Lambert Company Substituted di- and tripeptide inhibitors of protein:farnesyl transferase
US6403577B1 (en) 1993-11-17 2002-06-11 Eli Lilly And Company Hexamethyleneiminyl tachykinin receptor antagonists
US5466823A (en) 1993-11-30 1995-11-14 G.D. Searle & Co. Substituted pyrazolyl benzenesulfonamides
IT1271462B (en) 1993-12-03 1997-05-28 Menarini Farma Ind TACHYCHININ ANTAGONISTS, PROCEDURE FOR THEIR PREPARATION AND THEIR USE IN PHARMACEUTICAL FORMULATIONS.
US5484799A (en) 1993-12-09 1996-01-16 Abbott Laboratories Antifungal dorrigocin derivatives
IL111960A (en) 1993-12-17 1999-12-22 Merck & Co Inc Morpholines and thiomorpholines their preparation and pharmaceutical compositions containing them
AU1339795A (en) 1993-12-21 1995-07-10 Eli Lilly And Company Non-peptide tachykinin receptor antagonists
HU221139B1 (en) 1993-12-29 2002-08-28 Merck Sharp & Dohme Tachykinin antagonist substituted morpholine derivatives, preparation and use thereof
CN1046515C (en) 1993-12-29 1999-11-17 辉瑞大药厂 Diazabicyclic neurokinin antagonists
ATE170174T1 (en) 1994-01-13 1998-09-15 Merck Sharp & Dohme GEM-BIS-SUBSTITUTED AZAZYCLIC TACHYKININ ANTAGONISTS
CA2181376A1 (en) 1994-01-28 1995-08-03 Malcolm Maccoss Aralkylamino substituted azacyclic therapeutic agents
US5393790A (en) 1994-02-10 1995-02-28 G.D. Searle & Co. Substituted spiro compounds for the treatment of inflammation
GB9402688D0 (en) 1994-02-11 1994-04-06 Merck Sharp & Dohme Therapeutic agents
US5610165A (en) 1994-02-17 1997-03-11 Merck & Co., Inc. N-acylpiperidine tachykinin antagonists
IL112778A0 (en) 1994-03-04 1995-05-26 Merck & Co Inc Substituted heterocycles, their preparation and pharmaceutical compositions containing them
WO1995024612A1 (en) 1994-03-07 1995-09-14 International Business Machines Corporation Fast process and device for interpolating intermediate values from periodic phase-shifted signals and for detecting rotary body defects
EP0750609A4 (en) 1994-03-15 1997-09-24 Eisai Co Ltd Isoprenyl transferase inhibitors
FR2718136B1 (en) 1994-03-29 1996-06-21 Sanofi Sa Amino aromatic compounds, process for their preparation and pharmaceutical compositions containing them.
IL113196A0 (en) 1994-03-31 1995-06-29 Bristol Myers Squibb Co Imidazole derivatives and pharmaceutical compositions containing the same
US5523430A (en) 1994-04-14 1996-06-04 Bristol-Myers Squibb Company Protein farnesyl transferase inhibitors
US5610145A (en) 1994-04-15 1997-03-11 Warner-Lambert Company Tachykinin antagonists
NZ270985A (en) 1994-04-29 1997-06-24 Lilly Co Eli Substituted benzimidazole derivatives; medicaments and preparation of medicaments
US5747491A (en) 1994-05-05 1998-05-05 Merck Sharp & Dohme Ltd. Morpholine derivatives and their use as antagonists of tachikinins
EP0804463A1 (en) 1994-05-07 1997-11-05 Boehringer Ingelheim Kg Neurokinine (tachykinine) antagonists
US5510510A (en) 1994-05-10 1996-04-23 Bristol-Meyers Squibb Company Inhibitors of farnesyl protein transferase
US5563255A (en) 1994-05-31 1996-10-08 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of raf gene expression
EP0764163B1 (en) 1994-06-06 2001-10-10 Warner-Lambert Company Tachykinin (nk 1) receptor antagonists
CA2150992A1 (en) 1994-06-10 1995-12-11 Philip Arthur Hipskind Cyclohexyl tachykinin receptor antagonists
CA2192389A1 (en) 1994-06-10 1995-12-21 Bernard Baudoin Novel farnesyl transferase inhibitors, their preparation and pharmaceutical compositions containing same
US5571792A (en) 1994-06-30 1996-11-05 Warner-Lambert Company Histidine and homohistidine derivatives as inhibitors of protein farnesyltransferase
CZ288434B6 (en) 1994-07-12 2001-06-13 Lilly Co Eli Dihydrochloride trihydrate of (R)-3-(1H-indol-3-yl)-1-[N-(2-methoxybenzyl)acetylamino]-2-[N-/2-(4-piperid-1-yl)piperid-1-yl)acetyl/-amino]propane, pharmaceutical preparation in which it is comprised and use thereof
CA2154116A1 (en) 1994-07-22 1996-01-23 Philip Arthur Hipskind 1-aryl-2-acetamidopentanone derivatives for use as tachykinin receptor antagonists
GB9415997D0 (en) 1994-08-08 1994-09-28 Merck Sharp & Dohme Therapeutic agents
GB9415996D0 (en) 1994-08-08 1994-09-28 Merck Sharp & Dohme Therapeutic agents
TW432061B (en) 1994-08-09 2001-05-01 Pfizer Res & Dev Lactams
WO1996005529A1 (en) 1994-08-09 1996-02-22 Micron Optics, Inc. Temperature compensated fiber fabry-perot filters
CA2155448A1 (en) 1994-08-11 1996-02-12 Katerina Leftheris Inhibitors of farnesyl protein transferase
EP0776884B1 (en) 1994-08-11 2000-01-05 Banyu Pharmaceutical Co., Ltd. Substituted amide derivative
EP0805154A1 (en) 1994-08-12 1997-11-05 Banyu Pharmaceutical Co., Ltd. N,n-disubstituted amic acid derivative
DE69521208T2 (en) 1994-08-15 2001-11-08 Merck Sharp & Dohme MORPHOLINE DERIVATIVES AND THEIR USE AS THERAPEUTIC AGENTS
DE4429506B4 (en) 1994-08-19 2007-09-13 Degussa Gmbh Process for the extraction of natural carotenoid dyes
DE4429653C2 (en) 1994-08-20 1997-04-03 Anton Dr More Converter and method for refining molten metals, in particular from pig iron to steel
JPH10504580A (en) 1994-08-25 1998-05-06 メレルファーマスーティカルズ インコーポレイテッド New substituted piperidines useful for the treatment of allergic diseases
DE69405864T2 (en) 1994-08-29 1998-03-26 Akzo Nobel Nv Process for the production of quaternary diesters
GB9417956D0 (en) 1994-09-02 1994-10-26 Merck Sharp & Dohme Therapeutic agents
GB9418545D0 (en) 1994-09-15 1994-11-02 Merck Sharp & Dohme Therapeutic agents
US5457107A (en) 1994-09-16 1995-10-10 Merck & Co., Inc. Polymorphic form of a tachykinin receptor antagonist
PT783490E (en) 1994-09-30 2002-06-28 Novartis Erfind Verwalt Gmbh COMPOUNDS OF 1-ACYL-4-ALIFATILAMINOPIPERIDINE
TW397825B (en) 1994-10-14 2000-07-11 Novartis Ag Aroyl-piperidine derivatives
FR2725986B1 (en) 1994-10-21 1996-11-29 Adir NOVEL PIPERIDINE DERIVATIVES, THEIR PREPARATION PROCESS AND THE PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
DE69534213T2 (en) 1994-10-25 2006-01-12 Astrazeneca Ab Therapeutically effective heterocycles
GB9421709D0 (en) 1994-10-27 1994-12-14 Zeneca Ltd Therapeutic compounds
DE69507284T2 (en) 1994-11-22 1999-07-01 Philips Electronics Nv SEMICONDUCTORS WITH A CARRIER ON WHICH A SUBSTRATE WITH A SEMICONDUCTOR ELEMENT IS FASTENED BY MEANS OF AN ADHESIVE LAYER AND A PATTERN PATTERN
EP0714891A1 (en) 1994-11-22 1996-06-05 Eli Lilly And Company Heterocyclic tachykinin receptor antagonists
FR2727411B1 (en) 1994-11-30 1997-01-03 Rhone Poulenc Rorer Sa NOVEL PERHYDROISOINDOLE DERIVATIVES, THEIR PREPARATION AND THE PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
JPH10510261A (en) 1994-12-09 1998-10-06 ワーナー−ランバート・カンパニー Protein: substituted tetra- and pentapeptide inhibitors of farnesyltransferase
PE38997A1 (en) 1994-12-13 1997-10-02 Novartis Ag TACHYCININE ANTAGONIST
GB9426103D0 (en) 1994-12-23 1995-02-22 Merck Sharp & Dohme Therapeutic agents
BR9606896A (en) 1995-01-09 1997-10-21 Magla Int Ltd Water resistant image printing on latex surfaces
JP3929069B2 (en) 1995-01-12 2007-06-13 ユニバーシティ オブ ピッツバーグ Inhibitors of prenyltransferase
AU4437896A (en) 1995-01-12 1996-07-31 Glaxo Group Limited Piperidine derivatives having tachykinin antagonist activity
FR2729390A1 (en) 1995-01-18 1996-07-19 Rhone Poulenc Rorer Sa NOVEL FARNESYL TRANSFERASE INHIBITORS, THEIR PREPARATION AND THE PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
FR2729951B1 (en) 1995-01-30 1997-04-18 Sanofi Sa NOVEL HETEROCYCLIC COMPOUNDS, PROCESS FOR THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THE SAME
FR2730492B1 (en) 1995-02-09 1997-03-14 Rhone Poulenc Rorer Sa NOVEL FARNESYL TRANSFERASE INHIBITORS, THEIR PREPARATION AND THE PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
FR2730491B1 (en) 1995-02-09 1997-03-14 Rhone Poulenc Rorer Sa NOVEL FARNESYL TRANSFERASE INHIBITORS, THEIR PREPARATION AND THE PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
US5633272A (en) 1995-02-13 1997-05-27 Talley; John J. Substituted isoxazoles for the treatment of inflammation
GB9505492D0 (en) 1995-03-18 1995-05-03 Merck Sharp & Dohme Therapeutic agents
GB9505491D0 (en) 1995-03-18 1995-05-03 Merck Sharp & Dohme Therapeutic agents
US5554641A (en) 1995-03-20 1996-09-10 Horwell; David C. Nonpeptides as tachykinin antagonists
GB9505692D0 (en) 1995-03-21 1995-05-10 Glaxo Group Ltd Chemical compounds
US5700806A (en) 1995-03-24 1997-12-23 Schering Corporation Tricyclic amide and urea compounds useful for inhibition of G-protein function and for treatment of proliferative diseases
DE69628484T2 (en) 1995-03-24 2004-05-19 Takeda Chemical Industries, Ltd. Cyclic compounds, their preparation and their use as tachykinin receptor antagonists
US5684013A (en) 1995-03-24 1997-11-04 Schering Corporation Tricyclic compounds useful for inhibition of g-protein function and for treatment of proliferative diseases
IL117580A0 (en) 1995-03-29 1996-07-23 Merck & Co Inc Inhibitors of farnesyl-protein transferase and pharmaceutical compositions containing them
US5565568A (en) 1995-04-06 1996-10-15 Eli Lilly And Company 2-acylaminopropanamides as tachykinin receptor antagonists
US5891872A (en) 1995-04-07 1999-04-06 Schering Corporation Tricyclic compounds
EP0820452B1 (en) 1995-04-07 2003-06-04 Schering Corporation Carbonyl-piperazinyl and piperidinil compounds which inhibit farnesyl protein transferase
IL117798A (en) 1995-04-07 2001-11-25 Schering Plough Corp Tricyclic compounds useful for inhibition of g-protein function and for treatment of proliferative diseases and pharmaceutical compositions comprising them
US5712280A (en) 1995-04-07 1998-01-27 Schering Corporation Tricyclic compounds useful for inhibition of G-protein function and for treatment of proliferative diseases
KR100414321B1 (en) 1995-04-13 2004-02-18 아벤티스 파마슈티칼스 인크. Novel Substituted Piperazine Derivatives Having Tachykinin Receptor Antagonists Activity
DE69622166T2 (en) 1995-04-14 2003-04-03 Smithkline Beecham Corp DOSING INHALATOR FOR ALBUTEROL
US5831115A (en) 1995-04-21 1998-11-03 Abbott Laboratories Inhibitors of squalene synthase and protein farnesyltransferase
IL118101A0 (en) 1995-05-03 1996-09-12 Abbott Lab Inhibitors of farnesyltransferase
AU706021B2 (en) 1995-05-25 1999-06-03 Fujisawa Pharmaceutical Co., Ltd. Piperazine derivatives
US5889136A (en) 1995-06-09 1999-03-30 The Regents Of The University Of Colorado Orthoester protecting groups in RNA synthesis
WO1997000252A1 (en) 1995-06-16 1997-01-03 Warner-Lambert Company Tricyclic inhibitors of protein farnesyltransferase
GB9513117D0 (en) 1995-06-28 1995-08-30 Merck Sharp & Dohme Therapeutic agents
GB9513121D0 (en) 1995-06-28 1995-08-30 Merck Sharp & Dohme Therapeutic agents
GB9513118D0 (en) 1995-06-28 1995-08-30 Merck Sharp & Dohme Therapeutic agents
EP0840732B1 (en) 1995-07-07 2001-03-07 Pfizer Inc. Substituted benzolactam compounds as substance p antagonists
FR2736641B1 (en) 1995-07-10 1997-08-22 Rhone Poulenc Rorer Sa NOVEL FARNESYL TRANSFERASE INHIBITORS, THEIR PREPARATION AND THE PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
AT402617B (en) 1995-07-11 1997-07-25 Datacon Schweitzer & Zeindl Gm SYSTEM FOR AUTOMATED, HERMETIC SYSTEM FOR AUTOMATED, HERMETIC LOCKING OF HOUSINGS LOCKING OF HOUSINGS
FR2736638B1 (en) 1995-07-12 1997-08-22 Rhone Poulenc Rorer Sa NOVEL FARNESYL TRANSFERASE INHIBITORS, THEIR PREPARATION AND THE PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
CH690163A5 (en) 1995-07-28 2000-05-31 Symphar Sa Derivatives substituted gem-diphosphonates useful as anti-cancer.
TW340842B (en) 1995-08-24 1998-09-21 Pfizer Substituted benzylaminopiperidine compounds
US6020343A (en) 1995-10-13 2000-02-01 Merck Frosst Canada, Inc. (Methylsulfonyl)phenyl-2-(5H)-furanones as COX-2 inhibitors
AU722883B2 (en) 1995-10-18 2000-08-10 Merck & Co., Inc. Cyclopentyl tachykinin receptor antagonists
EP0873123B1 (en) 1995-11-06 2003-04-09 University Of Pittsburgh Inhibitors of protein isoprenyl transferases
DE19541283A1 (en) 1995-11-06 1997-05-07 Boehringer Ingelheim Kg Novel amino acid derivatives, processes for their preparation and pharmaceutical compositions containing these compounds
GB9523244D0 (en) 1995-11-14 1996-01-17 Merck Sharp & Dohme Therapeutic agents
JP4183099B2 (en) 1995-11-17 2008-11-19 ゲゼルシャフト・フュア・ビオテヒノロジッシェ・フォルシュング・ミット・ベシュレンクテル・ハフツング(ゲー・ベー・エフ) Epothilones C and D, production methods and compositions
EP0862435A4 (en) 1995-11-22 1999-02-03 Merck & Co Inc Inhibitors of farnesyl-protein transferase
WO1997019084A1 (en) 1995-11-23 1997-05-29 Merck Sharp & Dohme Limited Spiro-piperidine derivatives and their use as tachykinin antagonists
GB9524157D0 (en) 1995-11-25 1996-01-24 Pfizer Ltd Therapeutic agents
RU2135494C1 (en) 1995-12-01 1999-08-27 Санкио Компани Лимитед Heterocyclic compounds and composition on said showing antagonistic effect with respect to tachykinin receptors
PT1162201E (en) 1995-12-08 2006-08-31 Janssen Pharmaceutica Nv (IMIDAZOL-5-IL) METHYL-2-KINOLINONE DERIVATIVES AS FARNESIL PROTEIN INHIBITORS TRANSFERASE
GB9525296D0 (en) 1995-12-11 1996-02-07 Merck Sharp & Dohme Therapeutic agents
SK86198A3 (en) 1995-12-22 1999-02-11 Schering Corp Tricyclic amides useful for inhibition of g-protein function and for treatment of proliferative diseases
US5998203A (en) 1996-04-16 1999-12-07 Ribozyme Pharmaceuticals, Inc. Enzymatic nucleic acids containing 5'-and/or 3'-cap structures
WO1997026246A1 (en) 1996-01-16 1997-07-24 Warner-Lambert Company Substituted histidine inhibitors of protein farnesyltransferase
US6673927B2 (en) 1996-02-16 2004-01-06 Societe De Conseils De Recherches Et D'applications Scientifiques, S.A.S. Farnesyl transferase inhibitors
JP2001519766A (en) 1996-04-03 2001-10-23 メルク エンド カンパニー インコーポレーテッド Inhibitors of farnesyl protein transferase
CN1098256C (en) 1996-04-12 2003-01-08 G·D·瑟尔公司 Substd. benzenesulfonamide derivs as prodrugs of COX-2 inhibitors
JP2000511527A (en) 1996-05-22 2000-09-05 ワーナー―ランバート・コンパニー Inhibition of protein farnesyltransferase
CA2260216A1 (en) 1996-07-15 1998-01-22 Bristol-Myers Squibb Company Thiadioxobenzodiazepine inhibitors of farnesyl protein transferase
US5861419A (en) 1996-07-18 1999-01-19 Merck Frosst Canad, Inc. Substituted pyridines as selective cyclooxygenase-2 inhibitors
AU756699B2 (en) 1996-12-03 2003-01-23 Sloan-Kettering Institute For Cancer Research Synthesis of epothilones, intermediates thereto, analogues and uses thereof
AU5719598A (en) 1996-12-30 1998-07-31 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
AU6013998A (en) 1996-12-30 1998-07-31 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
US6001311A (en) 1997-02-05 1999-12-14 Protogene Laboratories, Inc. Apparatus for diverse chemical synthesis using two-dimensional array
US6126919A (en) 1997-02-07 2000-10-03 3M Innovative Properties Company Biocompatible compounds for pharmaceutical drug delivery systems
AR013269A1 (en) 1997-08-04 2000-12-13 Scras PRODUCT CONTAINING AT LEAST A DOUBLE FILAMENT RNA COMBINED WITH AT LEAST AN ANTI-VIRAL AGENT, FOR THERAPEUTIC USE IN THE TREATMENT OF A VIRAL DISEASE, ESPECIALLY OF VIRAL HEPATITIS
US6054576A (en) 1997-10-02 2000-04-25 Ribozyme Pharmaceuticals, Inc. Deprotection of RNA
AU1922999A (en) 1997-12-16 1999-07-05 Baylor College Of Medicine Needle-free injection of formulated nucleic acid molecules
US6506559B1 (en) 1997-12-23 2003-01-14 Carnegie Institute Of Washington Genetic inhibition by double-stranded RNA
US6111086A (en) 1998-02-27 2000-08-29 Scaringe; Stephen A. Orthoester protecting groups
AU3751299A (en) 1998-04-20 1999-11-08 Ribozyme Pharmaceuticals, Inc. Nucleic acid molecules with novel chemical compositions capable of modulating gene expression
EP1073763A4 (en) 1998-04-28 2002-08-07 Univ Washington Methods for transducing fusion molecules
AR020078A1 (en) 1998-05-26 2002-04-10 Syngenta Participations Ag METHOD FOR CHANGING THE EXPRESSION OF AN OBJECTIVE GENE IN A PLANT CELL
GB9827152D0 (en) 1998-07-03 1999-02-03 Devgen Nv Characterisation of gene function using double stranded rna inhibition
US6995259B1 (en) 1998-10-23 2006-02-07 Sirna Therapeutics, Inc. Method for the chemical synthesis of oligonucleotides
CA2354044A1 (en) 1998-12-10 2000-06-15 Washington University Protein transduction system and methods of use thereof
CA2361201A1 (en) 1999-01-28 2000-08-03 Medical College Of Georgia Research Institute, Inc. Composition and method for in vivo and in vitro attenuation of gene expression using double stranded rna
JP2002536968A (en) 1999-01-29 2002-11-05 イムクローン システムズ インコーポレイティド Antibodies specific for KDR and uses thereof
DE19956568A1 (en) 1999-01-30 2000-08-17 Roland Kreutzer Method and medicament for inhibiting the expression of a given gene
GB9904387D0 (en) 1999-02-25 1999-04-21 Pharmacia & Upjohn Spa Antitumour synergistic composition
WO2000062067A1 (en) 1999-02-28 2000-10-19 Washington University Novel transduction molecules and methods for using same
AU4972900A (en) 1999-04-08 2000-11-14 Arch Development Corporation Use of anti-vegf antibody to enhance radiation in cancer therapy
AU1086501A (en) 1999-10-15 2001-04-30 Carnegie Institution Of Washington Rna interference pathway genes as tools for targeted genetic interference
US6545004B1 (en) 1999-10-27 2003-04-08 Cytokinetics, Inc. Methods and compositions utilizing quinazolinones
CA2388646C (en) 1999-10-27 2009-01-27 Cytokinetics, Inc. Methods and compositions utilizing quinazolinones
GB9927444D0 (en) 1999-11-19 2000-01-19 Cancer Res Campaign Tech Inhibiting gene expression
US20050032733A1 (en) 2001-05-18 2005-02-10 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (SiNA)
US8202979B2 (en) 2002-02-20 2012-06-19 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid
US7491805B2 (en) 2001-05-18 2009-02-17 Sirna Therapeutics, Inc. Conjugates and compositions for cellular delivery
US6401406B1 (en) 2000-02-11 2002-06-11 Domald K. Komara Retainment device for concrete block inspection plates
US8273866B2 (en) 2002-02-20 2012-09-25 Merck Sharp & Dohme Corp. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (SINA)
EP1268412B8 (en) 2000-03-20 2007-02-21 MERCK SHARP & DOHME LTD. Sulphonamido-substituted bridged bicycloalkyl derivatives
EP2028278B1 (en) 2000-03-30 2014-03-19 Whitehead Institute For Biomedical Research RNA sequence-specific mediators of RNA interference
GB0012671D0 (en) 2000-05-24 2000-07-19 Merck Sharp & Dohme Therapeutic agents
ATE506372T1 (en) 2000-09-01 2011-05-15 Ribozyme Pharm Inc METHOD FOR PRODUCING NUCLEOSIDE DERIVATIVES
GB0025173D0 (en) 2000-10-13 2000-11-29 Merck Sharp & Dohme Therapeutic agents
JP3880051B2 (en) 2000-11-02 2007-02-14 メルク シャープ エンド ドーム リミテッド Sulfamide as a γ-secretase inhibitor
UA74849C2 (en) 2000-11-17 2006-02-15 Lilly Co Eli Lactam
DK2813582T3 (en) 2000-12-01 2017-07-31 Max-Planck-Gesellschaft Zur Förderung Der Wss E V Small RNA molecules that mediate RNA interference
US6401408B1 (en) 2001-01-29 2002-06-11 Plastics Research Corporation Molded plastic stake with multiple shoulders
GB0108591D0 (en) 2001-04-05 2001-05-23 Merck Sharp & Dohme Therapeutic agents
GB0108592D0 (en) 2001-04-05 2001-05-23 Merck Sharp & Dohme Therapeutic agents
WO2002083675A2 (en) 2001-04-10 2002-10-24 Merck Sharp & Dohme Limited Inhibitors of akt activity
WO2002083138A1 (en) 2001-04-10 2002-10-24 Merck & Co., Inc. Inhibitors of akt activity
WO2002083140A1 (en) 2001-04-10 2002-10-24 Merck & Co., Inc. Inhibitors of akt activity
JP2004527531A (en) 2001-04-10 2004-09-09 メルク エンド カムパニー インコーポレーテッド How to treat cancer
WO2002083139A1 (en) 2001-04-10 2002-10-24 Merck & Co., Inc. Inhibitors of akt activity
US20070042983A1 (en) 2001-05-18 2007-02-22 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using short interfering nucleic acid (siNA)
GB0119152D0 (en) 2001-08-06 2001-09-26 Merck Sharp & Dohme Therapeutic agents
WO2003013526A1 (en) 2001-08-08 2003-02-20 Merck & Co. Inc. Anticoagulant compounds
DK1421062T3 (en) 2001-08-21 2008-02-18 Merck Sharp & Dohme Cyclohexyl sulfones
FR2830766B1 (en) 2001-10-12 2004-03-12 Optis France Sa DEVICE   OF   ISSUE   OF   DRUGS   THROUGH   IONTOPHORESIS   TRANSPALPEBRALE
FR2830767B1 (en) 2001-10-12 2004-03-12 Optis France Sa DEVICE FOR DELIVERING DRUGS BY IONTOPHORESIS OR INTROCULAR ELECTROPORATION
EP1465896A4 (en) 2001-12-06 2006-01-11 Merck & Co Inc Mitotic kinesin inhibitors
AU2002357043B2 (en) 2001-12-06 2008-04-24 Merck Sharp & Dohme Corp. Mitotic kinesin inhibitors
JP4467979B2 (en) 2001-12-06 2010-05-26 メルク・シャープ・エンド・ドーム・コーポレイション Mitotic kinesin inhibitor
ES2291543T3 (en) 2001-12-06 2008-03-01 MERCK & CO., INC. INHIBITION OF MYTHICAL KINESINA.
EP1458726B1 (en) 2001-12-06 2009-07-15 Merck & Co., Inc. Mitotic kinesin inhibitors
AU2003207708A1 (en) 2002-02-20 2003-09-09 Sirna Therapeutics, Inc. Rna interference mediated inhibition of map kinase genes
US9181551B2 (en) 2002-02-20 2015-11-10 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
AU2003249597B2 (en) 2002-03-08 2007-06-28 Merck Sharp & Dohme Corp. Mitotic kinesin inhibitors
WO2003086279A2 (en) 2002-04-08 2003-10-23 Merck & Co., Inc. Inhibitors of akt activity
EP1494676B1 (en) 2002-04-08 2013-05-08 Merck Sharp & Dohme Corp. Fused quinoxaline derivatives as inhibitors of akt activity
US20060142178A1 (en) 2002-04-08 2006-06-29 Barnett Stanley F Method of treating cancer
US7223738B2 (en) 2002-04-08 2007-05-29 Merck & Co., Inc. Inhibitors of Akt activity
JP4451136B2 (en) 2002-04-08 2010-04-14 メルク エンド カムパニー インコーポレーテッド Akt activity inhibitor
GB0209995D0 (en) 2002-05-01 2002-06-12 Merck Sharp & Dohme Therapeutic agents
GB0209997D0 (en) 2002-05-01 2002-06-12 Merck Sharp & Dohme Therapeutic agents
EP1503998B1 (en) 2002-05-01 2009-07-01 MERCK SHARP & DOHME LTD. Heteroaryl substituted spirocyclic sufamides for inhibition of gamma secretase
GB0209991D0 (en) 2002-05-01 2002-06-12 Merck Sharp & Dohme Therapeutic agents
AU2003299517A1 (en) 2002-05-23 2004-05-25 Merck & Co., Inc. Mitotic kinesin inhibitors
WO2003099211A2 (en) 2002-05-23 2003-12-04 Merck & Co., Inc. Mitotic kinesin inhibitors
CA2486215A1 (en) 2002-06-14 2003-12-24 Merck & Co., Inc. Mitotic kinesin inhibitors
US7348440B2 (en) 2002-06-14 2008-03-25 Merck & Co., Inc. Mitotic kinesin inhibitors
CA2487712A1 (en) 2002-06-28 2004-01-08 Nastech Pharmaceutical Company Inc. Compositions and methods for modulating physiology of epithelial junctional adhesion molecules for enhanced mucosal delivery of therapeutic compounds
US6989442B2 (en) 2002-07-12 2006-01-24 Sirna Therapeutics, Inc. Deprotection and purification of oligonucleotides and their derivatives
US7923547B2 (en) 2002-09-05 2011-04-12 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
GB0223039D0 (en) 2002-10-04 2002-11-13 Merck Sharp & Dohme Therapeutic compounds
GB0223040D0 (en) 2002-10-04 2002-11-13 Merck Sharp & Dohme Therapeutic compounds
GB0223038D0 (en) 2002-10-04 2002-11-13 Merck Sharp & Dohme Therapeutic compounds
AU2003287057B2 (en) 2002-10-18 2008-08-21 Merck Sharp & Dohme Corp. Mitotic kinesin inhibitors
DE60336576D1 (en) 2002-10-30 2011-05-12 Merck Sharp & Dohme HEMMER OF ACT ACTIVITY
US20040102360A1 (en) 2002-10-30 2004-05-27 Barnett Stanley F. Combination therapy
GB0225474D0 (en) 2002-11-01 2002-12-11 Merck Sharp & Dohme Therapeutic agents
GB0225475D0 (en) 2002-11-01 2002-12-11 Merck Sharp & Dohme Therapeutic agents
US9150605B2 (en) 2002-11-05 2015-10-06 Isis Pharmaceuticals, Inc. Compositions comprising alternating 2′-modified nucleosides for use in gene modulation
CA2508956A1 (en) 2002-12-20 2004-07-15 Merck & Co., Inc. Mitotic kinesin inhibitors
US7622468B2 (en) 2002-12-20 2009-11-24 Merck & Co. Inc. Mitotic kinesin inhibitors
KR100536594B1 (en) 2002-12-30 2005-12-14 삼성전자주식회사 A photosensitive polymer containing silicon and resist compositions using the same
US7816337B2 (en) 2003-02-18 2010-10-19 Roche Madison Inc. Reversible attachment of a membrane active polymer to a polynucleotide
GB0308318D0 (en) 2003-04-10 2003-05-14 Merck Sharp & Dohme Therapeutic agents
DE602004026047D1 (en) 2003-04-24 2010-04-29 Merck Sharp & Dohme HEMMER OF ACT ACTIVITY
EP1622616B1 (en) 2003-04-24 2011-06-15 Merck Sharp & Dohme Corp. Inhibitors of akt activity
CA2522430A1 (en) 2003-04-24 2004-11-11 Merck & Co., Inc. Inhibitors of akt activity
EP1631548B1 (en) 2003-04-24 2009-10-28 Merck & Co., Inc. Inhibitors of akt activity
DE602004011767T2 (en) 2003-05-16 2009-02-19 Merck Sharp & Dohme Ltd., Hoddesdon CYCLOHEXYLSULFONES AS GAMMA SECRETASE INHIBITORS
US20060222657A1 (en) 2003-06-20 2006-10-05 Dowdy Steven F Polypeptide transduction and fusogenic peptides
GB0318447D0 (en) 2003-08-05 2003-09-10 Merck Sharp & Dohme Therapeutic agents
US7307085B2 (en) 2003-08-13 2007-12-11 Merck & Co., Inc. Mitotic kinesin inhibitors
PE20050730A1 (en) 2003-08-15 2005-09-20 Merck & Co Inc DERIVATIVES OF 2,5-DIHYDROPYRROL 2,2-DISUSTITUTED AS INHIBITORS OF MYTHOTIC KINESINS
AU2004264533B2 (en) 2003-08-15 2009-01-22 Merck Sharp & Dohme Corp. Mitotic kinesin inhibitors
CA2533889A1 (en) 2003-08-15 2005-03-03 Merck & Co., Inc. Mitotic kinesin inhibitors
ES2319429T3 (en) 2003-08-15 2009-05-07 MERCK & CO., INC. INHIBITORS OF MITOTIC CINESINE.
EP1667984B1 (en) 2003-09-24 2011-05-18 Merck Sharp & Dohme Ltd. Gamma-secretase inhibitors
BRPI0416128B8 (en) 2003-11-03 2021-06-22 Glaxo Group Ltd fluid dispensing device
US7294640B2 (en) 2004-02-06 2007-11-13 Merck & Co., Inc. Mitotic kinesin inhibitors
EP2636739B1 (en) 2004-03-12 2014-12-10 Alnylam Pharmaceuticals Inc. iRNA agents targeting VEGF
AU2005233569B2 (en) 2004-04-09 2010-08-19 Merck Sharp & Dohme Corp. Inhibitors of Akt activity
EP1737843B1 (en) 2004-04-09 2011-02-23 Merck Sharp & Dohme Corp. Inhibitors of akt activity
CA2564616C (en) 2004-04-20 2016-08-30 Nastech Pharmaceutical Company Inc. Methods and compositions for enhancing delivery of double-stranded rna or a double-stranded hybrid nucleic acid to regulate gene expression in mammalian cells
US20060062758A1 (en) 2004-09-21 2006-03-23 Nastech Pharmaceutical Comapny Inc. Tight junction modulator peptide PN159 for enhanced mucosal delivery of therapeutic compounds
US7404969B2 (en) 2005-02-14 2008-07-29 Sirna Therapeutics, Inc. Lipid nanoparticle based compositions and methods for the delivery of biologically active molecules
KR20080036650A (en) 2005-08-17 2008-04-28 시르나 쎄러퓨틱스 인코퍼레이티드 Chemically modified short interfering nucleic acid molecules that mediate rna interference
WO2007095152A2 (en) 2006-02-10 2007-08-23 The Regents Of The University Of California TRANSDUCIBLE DELIVERY OF sIRNA BY dsRNA BINDING DOMAIN FUSIONS TO PTD/CPPS
CN101506368B (en) 2006-07-12 2017-02-08 加利福尼亚大学董事会 Transducible delivery of nucleic acids by reversible phosphotriester charge neutralization protecting groups
CA2660842C (en) 2006-08-18 2012-03-13 F. Hoffmann-La Roche Ag Polyconjugates for in vivo delivery of polynucleotides
US20100120895A1 (en) * 2007-05-02 2010-05-13 Merck & Co., Inc. RNA Interference Mediated Inhibition of Cyclic Nucleotide Type 4 Phosphodiesterase (PDE4B) Gene Expression Using Short Interfering Nucleic Acid (siNA)
EP2173358B1 (en) 2007-06-22 2015-10-21 Isis Pharmaceuticals, Inc. Double strand compositions comprising differentially modified strands for use in gene modulation
WO2009041902A1 (en) 2007-09-25 2009-04-02 Loefgren Kajsa Prion protein derived cell penetrating peptides and their uses
CA2713379A1 (en) 2008-01-31 2009-11-05 Alnylam Pharmaceuticals, Inc. Optimized methods for delivery of dsrna targeting the pcsk9 gene
US20120072123A1 (en) 2009-05-29 2012-03-22 Merck & Co., Inc. Methods of Predicting The Probability of Modulation of Transcript Levels By RNAI Compounds
US9096850B2 (en) 2009-08-24 2015-08-04 Sirna Therapeutics, Inc. Segmented micro RNA mimetics
WO2011084345A1 (en) 2009-12-17 2011-07-14 Merck Sharp & Dohme Corp. A method for rapidly evaluating performance of short interfering rna with novel chemical modifications
WO2011088554A1 (en) 2010-01-20 2011-07-28 Magna International Inc. Bi-metallic component and method of making the same
EP2601293B1 (en) 2010-08-02 2017-12-06 Sirna Therapeutics, Inc. RNA INTERFERENCE MEDIATED INHIBITION OF CATENIN (CADHERIN-ASSOCIATED PROTEIN), BETA 1 (CTNNB1) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
JP6106085B2 (en) 2010-08-24 2017-03-29 サーナ・セラピューティクス・インコーポレイテッドSirna Therapeutics,Inc. Single-stranded RNAi agent containing an internal non-nucleic acid spacer
CA2812046A1 (en) 2010-09-15 2012-03-22 Alnylam Pharmaceuticals, Inc. Modified irna agents
US9260471B2 (en) 2010-10-29 2016-02-16 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using short interfering nucleic acids (siNA)
EP2780454A2 (en) * 2011-11-18 2014-09-24 Alnylam Pharmaceuticals, Inc. Modified rnai agents
EP3453762B1 (en) 2012-05-02 2021-04-21 Sirna Therapeutics, Inc. Short interfering nucleic acid (sina) compositions

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090176725A1 (en) * 2005-08-17 2009-07-09 Sirna Therapeutics Inc. Chemically modified short interfering nucleic acid molecules that mediate rna interference

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11530407B2 (en) 2012-05-02 2022-12-20 Sirna Therapeutics, Inc. Short interfering nucleic acid (siNA) compositions
US20170081425A1 (en) * 2013-11-06 2017-03-23 Merck Sharp & Dohme Corp. Peptide containing conjugates for dual molecular delivery of oligonucleotides
US10239957B2 (en) * 2013-11-06 2019-03-26 Merck Sharp & Dohme Corp. Peptide containing conjugates for dual molecular delivery of oligonucleotides
EP4035659A1 (en) 2016-11-29 2022-08-03 PureTech LYT, Inc. Exosomes for delivery of therapeutic agents
US11021503B2 (en) * 2017-05-23 2021-06-01 Hoffmann-La Roche Inc. Process for GalNAc oligonucleotide conjugates
US20190078089A1 (en) * 2017-09-14 2019-03-14 Arrowhead Pharmaceuticals, Inc. RNAi Agents And Compositions for Inhibiting Expression of Angiopoietin-Like 3 (ANGPTL3), And Methods Of Use
US10995335B2 (en) * 2017-09-14 2021-05-04 Arrowhead Pharmaceuticals, Inc. RNAi agents and compositions for inhibiting expression of angiopoietin-like 3 (ANGPTL3), and methods of use
WO2021072139A1 (en) 2019-10-11 2021-04-15 Massachusetts Institute Of Technology Formulations for gastrointestinal delivery of oligonucleotides

Also Published As

Publication number Publication date
EP3358013B1 (en) 2020-06-24
US11530407B2 (en) 2022-12-20
US20230348904A1 (en) 2023-11-02
EP3358013A1 (en) 2018-08-08
EP2844261B1 (en) 2018-10-17
EP3919620A1 (en) 2021-12-08
US10689648B2 (en) 2020-06-23
WO2013165816A3 (en) 2014-01-16
WO2013165816A2 (en) 2013-11-07
EP3453762A1 (en) 2019-03-13
EP2844261A2 (en) 2015-03-11
US20180105812A1 (en) 2018-04-19
EP2844261A4 (en) 2015-12-30
US20200392499A1 (en) 2020-12-17
EP3453762B1 (en) 2021-04-21

Similar Documents

Publication Publication Date Title
US11530407B2 (en) Short interfering nucleic acid (siNA) compositions
US11932854B2 (en) RNA interference mediated inhibition of gene expression using short interfering nucleic acids (siNA)
US10407682B2 (en) RNA interference mediated inhibition of hepatitis B Virus (HBV) gene expression using short interfering nucleic acid (siNA)
US9850491B2 (en) RNA interference mediated inhibition of catenin (cadherin-associated protein), beta 1 (CTNNB1) gene expression using short interfering nucleic acid (siNA)
US9322020B2 (en) RNA interference mediated inhibition of isocitrate dehydrogenase (IDH1) gene expression
AU2019264591B2 (en) RNA interference mediated inhibition of hepatitis B virus (HBV) gene expression using short interfering nucleic acid (siNA)

Legal Events

Date Code Title Description
AS Assignment

Owner name: MERCK SHARP & DOHME CORP., NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CARR, BRIAN ALLEN;JADHAV, VASANT R.;KENSKI, DENISE M.;AND OTHERS;SIGNING DATES FROM 20161107 TO 20170605;REEL/FRAME:042761/0213

Owner name: SIRNA THERAPEUTICS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:MERCK SHARP & DOHME CORP.;REEL/FRAME:042761/0226

Effective date: 20140324

STCB Information on status: application discontinuation

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