US20080261304A1 - Methods and compositions for enhancing delivery of double-stranded rna or a double-stranded hybrid nucleic acid to regulate gene expression in mammalian cells - Google Patents

Methods and compositions for enhancing delivery of double-stranded rna or a double-stranded hybrid nucleic acid to regulate gene expression in mammalian cells Download PDF

Info

Publication number
US20080261304A1
US20080261304A1 US12/013,274 US1327408A US2008261304A1 US 20080261304 A1 US20080261304 A1 US 20080261304A1 US 1327408 A US1327408 A US 1327408A US 2008261304 A1 US2008261304 A1 US 2008261304A1
Authority
US
United States
Prior art keywords
cholesteryl
spermidine
carbamate
cholesterol
sirna
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
US12/013,274
Inventor
Mohammad Ahmadian
Kunyuan Cui
Lishan Chen
Shu-Chih Chen
Michael E. Houston
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.)
Marina Biotech Inc
Original Assignee
MDRNA Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to US12/013,274 priority Critical patent/US20080261304A1/en
Application filed by MDRNA Inc filed Critical MDRNA Inc
Publication of US20080261304A1 publication Critical patent/US20080261304A1/en
Assigned to EOS HOLDINGS LLC, AS AGENT reassignment EOS HOLDINGS LLC, AS AGENT SECURITY AGREEMENT Assignors: MDRNA RESEARCH, INC., MDRNA, INC., NASTECH PHARMACEUTICAL COMPANY, INC.
Assigned to MDRNA, INC. reassignment MDRNA, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CUI, KUNYUAN, AHMADIAN, MOHAMMAD, CHEN, LISHAN, HOUSTON, MICHAEL E., JR., CHEN, SHU-CHIH
Assigned to NASTECH PHARMACEUTICAL COMPANY, INC., MDRNA, INC., MDRNA RESEARCH, INC. reassignment NASTECH PHARMACEUTICAL COMPANY, INC. RELEASE OF SECURITY INTEREST Assignors: EOS HOLDINGS LLC, AS AGENT
Assigned to MDRNA, INC. reassignment MDRNA, INC. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: NASTECH PHARMACEUTICAL COMPANY, INC.
Assigned to CEQUENT PHARMACEUTICALS, INC. reassignment CEQUENT PHARMACEUTICALS, INC. SECURITY AGREEMENT (PATENTS) Assignors: MDRNA, INC. FKA NASTECH PHARMACEUTICAL COMPANY INC.
Assigned to MARINA BIOTECH, INC. (F/K/A MDRNA, INC.) reassignment MARINA BIOTECH, INC. (F/K/A MDRNA, INC.) RELEASE BY SECURED PARTY (SEE DOCUMENT FOR DETAILS). Assignors: CEQUENT PHARMACEUTICALS, INC.
Priority to US12/870,989 priority patent/US8940857B2/en
Priority to US14/566,695 priority patent/US20150252369A1/en
Priority to US14/970,529 priority patent/US20160206749A1/en
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
    • C12N15/111General methods applicable to biologically active non-coding nucleic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/554Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being a steroid plant sterol, glycyrrhetic acid, enoxolone or bile acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • 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
    • C12N15/113Non-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
    • C12N15/1137Non-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 against enzymes
    • 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
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • 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
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • C12N2310/3513Protein; Peptide
    • 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
    • C12N2310/3515Lipophilic moiety, e.g. cholesterol
    • 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
    • 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
    • C12YENZYMES
    • C12Y302/00Hydrolases acting on glycosyl compounds, i.e. glycosylases (3.2)
    • C12Y302/01Glycosidases, i.e. enzymes hydrolysing O- and S-glycosyl compounds (3.2.1)
    • C12Y302/01023Beta-galactosidase (3.2.1.23), i.e. exo-(1-->4)-beta-D-galactanase

Definitions

  • RNA interference is the process of sequence-specific post transcriptional gene silencing in cells initiated by double-stranded RNA (dsRNA) that is homologous in sequence to a portion of a targeted mRNA.
  • dsRNA double-stranded RNA
  • Introduction of dsRNA into cells leads to the destruction of the endogenous RNAs that share the same sequence as the dsRNA.
  • the dsRNA molecules are cleaved by an RNase III family nuclease called Dicer into short-interfering RNAs (siRNA), which are 19-23 nucleotides (nt) in length.
  • siRNAs are incorporated into a multicomponent nuclease complex (RISC, RNA-induced silencing complex), which identifies mRNA substrates through their homology to the siRNA, binds to and destroys the targeted mRNA.
  • RISC RNA-induced silencing complex
  • dsRNAs longer than 30 base pairs can activate the dsRNA-dependent kinase PKR and 2′-5′-oligoadenylate synthetase, normally induced by interferon.
  • synthetic siRNA avoids activation of the interferon response.
  • the activated PKR inhibits general translation by phosphorylation of the translation factor eukaryotic initiation factor 2 ⁇ (eIF2 ⁇ ), while 2′-5′-oligoadenylate synthetase causes nonspecific mRNA degradation via activation of RNase L.
  • eIF2 ⁇ translation factor eukaryotic initiation factor 2 ⁇
  • 2′-5′-oligoadenylate synthetase causes nonspecific mRNA degradation via activation of RNase L.
  • siRNA can mediate selective gene silencing in the mammalian system
  • Hairpin RNA with a short loop and 19 to 27 base pairs in the stem also selectively silences expression of genes that are homologous to the sequence in the double-stranded stem.
  • Mammalian cells can convert short hairpin RNA into siRNA to mediate selective gene silencing.
  • RISC mediates cleavage of single stranded RNA having sequence complementary to the antisense strand of the siRNA duplex. Cleavage of the target RNA takes place in the middle of the region complementary to the antisense strand of the siRNA duplex.
  • siRNA duplexes are most active when containing two nucleotide 3′-overhangs. Furthermore, complete substitution of one or both siRNA strands with 2′-deoxy (2′-H) or 2′-O-methyl nucleotides abolishes RNAi activity, whereas substitution of the 3′-terminal siRNA overhang nucleotides with deoxy nucleotides (2′-H) was shown to be tolerated.
  • RNA interference is emerging as a promising means for reducing the expression of specific gene products, and thus may be useful for developing therapeutic drugs to treat viral infections, cancers, autoimmune diseases, and other diseases and conditions amenable to treatment by down-regulation of mRNA expression.
  • FIG. 1 illustrates serum effects on cellular uptake of a cholesterol-conjugated siRNA in complex with a delivery enhancing agent (comprising a permeabilizing peptide, PN73), and on an unconjugated siRNA in complex with PN73-expressed as percentage uptake.
  • a delivery enhancing agent comprising a permeabilizing peptide, PN73
  • FIG. 2 illustrates serum effects on cellular uptake of a cholesterol-conjugated siRNA in complex with PN73, and on an unconjugated siRNA in complex with PN73—expressed as mean fluorescence intensity (MFI).
  • MFI mean fluorescence intensity
  • FIG. 3 illustrates the effects of increasing concentrations of serum on cellular uptake of a cholesterol-conjugated siRNA in the presence or absence of a second delivery enhancing agent, lipofectamine—expressed as percentage uptake.
  • FIG. 4 illustrates the effects of increasing concentrations of serum on cellular uptake of a cholesterol-conjugated siRNA in the presence or absence of a second delivery enhancing agent, lipofectamine—expressed as MFI.
  • the present invention fulfills these needs and satisfies additional objects and advantages by providing double-stranded nucleic acids conjugated to a cholesterol moiety to facilitate delivery of the nucleic acids into a selected target cell or tissue.
  • the present invention is directed towards methods and compositions to administer double-stranded ribonucleic acid to a mammal so as to effectuate transfection of the double-stranded RNA into a desired tissue of the mammal.
  • the double-stranded RNA has 30 or fewer nucleotides, and is a short interfering RNA (siRNA).
  • siRNA/cholesterol moiety constructs increase the silencing effect of the targeted mRNA in comparison to siRNA having no cholesterol conjugated to it:
  • constructs listed above are embodiments of the present invention, as well as those constructs in which the ds nucleic acid is a siHybrid in which the sense strand is a DNA molecule.
  • the term “inverted repeat” refers to a nucleic acid sequence comprising a sense and an antisense element positioned so that they are able to form a double stranded siRNA when the repeat is transcribed.
  • the inverted repeat may optionally include a linker or a heterologous sequence such as a self-cleaving ribozyme between the two elements of the repeat.
  • the elements of the inverted repeat have a length sufficient to form a double stranded RNA.
  • each element of the inverted repeat is about 15 to about 100 nucleotides in length, preferably about 20-30 base nucleotides, preferably about 20-25 nucleotides in length, e.g., 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides in length.
  • “Silencing” refers to partial or complete loss-of-function through targeted inhibition of gene expression in a cell and may also be referred to as “knock down”. Depending on the circumstances and the biological problem to be addressed, it may be preferable to partially reduce gene expression. Alternatively, it might be desirable to reduce gene expression as much as possible. The extent of silencing may be determined by any method known in the art, some of which are summarized in International Publication No. WO 99/32619. Depending on the assay, quantitation of gene expression permits detection of various amounts of inhibition for example, greater than 10%, 33%, 50%, 90%, 95% or 99%.
  • inhibitors expression of a target gene refers to the ability of a siRNA of the invention to initiate gene silencing of the target gene.
  • samples or assays of the organism of interest or cells in culture expressing a particular construct are compared to control samples lacking expression of the construct.
  • Control samples (lacking construct expression) are assigned a relative value of 100%. Inhibition of expression of a target gene is achieved when the test value relative to the control is about 90%, preferably 50%, more preferably 25-0%.
  • Suitable assays include, e.g., examination of protein or mRNA levels using techniques known to those of skill in the art such as dot blots, northern blots, in situ hybridization, ELISA, immunoprecipitation, enzyme function, as well as phenotypic assays known to those of skill in the art.
  • Nucleic acid refers to deoxyribonucleotides or ribonucleotides and polymers thereof in single- or double-stranded form.
  • the term encompasses nucleic acids containing known nucleotide analogs or modified backbone residues or linkages, which are synthetic, naturally occurring, and non-naturally occurring, which have similar binding properties as the reference nucleic acid, and which are metabolized in a manner similar to the reference nucleotides. Examples of such analogs include, without limitation, phosphorothioates, phosphoramidates, methyl phosphonates, chiral-methyl phosphonates, 2-O-methyl ribonucleotides, peptide-nucleic acids (PNAs).
  • PNAs peptide-nucleic acids
  • “Large double-stranded RNA” refers to any double-stranded RNA having a size greater than about 40 base pairs (bp) for example, larger than 100 bp or more particularly larger than 300 bp.
  • the sequence of a large dsRNA may represent a segment of a mRNA or the entire mRNA. The maximum size of the large dsRNA is not limited herein.
  • the double-stranded RNA may include modified bases where the modification may be to the phosphate sugar backbone or to the nucleoside. Such modifications may include a nitrogen or sulfur heteroatom or any other modification known in the art.
  • the double-stranded structure may be formed by self-complementary RNA strand such as occurs for a hairpin or a micro RNA or by annealing of two distinct complementary RNA strands.
  • “Overlapping” refers to when two RNA fragments have sequences which overlap by a plurality of nucleotides on one strand, for example, where the plurality of nucleotides (nt) numbers as few as 2-5 nucleotides or by 5-10 nucleotides or more.
  • One or more dsRNAs refers to dsRNAs that differ from each other on the basis of sequence.
  • Target gene or mRNA refers to any gene or mRNA of interest. Any of the genes previously identified by genetics or by sequencing can be implemented as a target. Target genes or mRNA can include developmental genes and regulatory genes, as well as metabolic or structural genes or genes encoding enzymes. The target gene may be expressed in cells in which a phenotype is being investigated, or in an organism in a manner that directly or indirectly impacts a phenotypic characteristic. The target gene may be endogenous or exogenous. Such cells include any cell in the body of an adult or embryonic animal or plant including gamete or any isolated cell such as occurs in an immortal cell line or primary cell culture.
  • siRNA means a small interfering RNA that is a short-length double-stranded RNA that are not toxic in mammalian cells. The length is not limited to 21 to 23 bp long. There is no particular limitation in the length of siRNA as long as it does not show toxicity. “siRNAs” can be, for example, 15 to 49 bp, preferably 15 to 35 bp, and more preferably 21 to 30 bp long. Alternatively, the double-stranded RNA portion of a final transcription product of siRNA to be expressed can be, for example, 15 to 49 bp, preferably 15 to 35 bp, and more preferably 21 to 30 bp long.
  • the double-stranded RNA portions of siRNAs in which two RNA strands pair up are not limited to the completely paired ones, and may contain nonpairing portions due to mismatch (the corresponding nucleotides are not complementary), bulge (lacking in the corresponding complementary nucleotide on one strand), and the like. Nonpairing portions can be contained to the extent that they do not interfere with siRNA formation.
  • the “bulge” used herein preferably comprise 1 to 2 nonpairing nucleotides, and the double-stranded RNA region of siRNAs in which two RNA strands pair up contains preferably 1 to 7, more preferably 1 to 5 bulges.
  • the “mismatch” used herein is contained in the double-stranded RNA region of siRNAs in which two RNA strands pair up, preferably 1 to 7, more preferably 1 to 5, in number.
  • one of the nucleotides is guanine, and the other is uracil.
  • Such a mismatch is due to a mutation from C to T, G to A, or mixtures thereof in DNA coding for sense RNA, but not particularly limited to them.
  • the double-stranded RNA region of siRNAs in which two RNA strands pair up may contain both bulge and mismatched, which sum up to, preferably 1 to 7, more preferably 1 to 5 in number.
  • the terminal structure of siRNA may be either blunt or cohesive (overhanging) as long as siRNA enables to silence the target gene expression due to its RNAi effect.
  • the cohesive (overhanging) end structure is not limited only to the 3′ overhang as reported by Tuschl et al. (ibid.), and the 5′ overhanging structure may be included as long as it is capable of inducing the RNAi effect.
  • the number of overhanging nucleotides is not limited to the reported 2 or 3, but can be any numbers as long as the overhang is capable of inducing the RNAi effect.
  • the overhang may be 1 to 8, or 2 to 4 nucleotides.
  • the total length of siRNA having cohesive end structure is expressed as the sum of the length of the paired double-stranded portion and that of a pair comprising overhanging single-strands at both ends. For example, in the case of 19 bp double-stranded RNA portion with 4 nucleotide overhangs at both ends, the total length is expressed as 23 bp. Furthermore, since this overhanging sequence has low specificity to a target gene, it is not necessarily complementary (antisense) or identical (sense) to the target gene sequence.
  • the siRNA may comprise a low molecular weight RNA (which may be a natural RNA molecule such as tRNA, rRNA or viral RNA, or an artificial RNA molecule), for example, in the overhanging portion at its one end.
  • a low molecular weight RNA which may be a natural RNA molecule such as tRNA, rRNA or viral RNA, or an artificial RNA molecule
  • the terminal structure of the “siRNA” is necessarily the cut off structure at both ends as described above, and may have a stem-loop structure in which ends of one side of double-stranded RNA are connected by a linker RNA.
  • the length of the double-stranded RNA region (stem-loop portion) can be, for example, 15 to 49 bp, preferably 15 to 35 bp, and more preferably 21 to 30 bp long.
  • the length of the double-stranded RNA region that is a final transcription product of siRNAs to be expressed is, for example, 15 to 49 bp, preferably 15 to 35 bp, and more preferably 21 to 30 bp long.
  • the linker portion may have a clover-leaf tRNA structure.
  • the linker portion may include introns so that the introns are excised during processing of precursor RNA into mature RNA, thereby allowing pairing of the stem portion.
  • either end (head or tail) of RNA with no loop structure may have a low molecular weight RNA.
  • this low molecular weight RNA may be a natural RNA molecule such as tRNA, rRNA or viral RNA, or an artificial RNA molecule.
  • Antisense RNA is an RNA strand having a sequence complementary to a target gene mRNA, and thought to induce RNAi by binding to the target gene mRNA.
  • Sense RNA has a sequence complementary to the antisense RNA, and annealed to its complementary antisense RNA to form siRNA. These antisense and sense RNAs have been conventionally synthesized with an RNA synthesizer.
  • RNAi construct is a generic term used throughout the specification to include small interfering RNAs (siRNAs), hairpin RNAs, and other RNA species which can be cleaved in vivo to form siRNAs.
  • RNAi constructs herein also include expression vectors (also referred to as RNAi expression vectors) capable of giving rise to transcripts which form dsRNAs or hairpin RNAs in cells, and/or transcripts which can produce siRNAs in vivo.
  • the siRNA include single strands or double strands of siRNA.
  • siHybrid molecule is a double-stranded nucleic acid that has a similar function to siRNA.
  • a siHybrid is comprised of an RNA strand and a DNA strand.
  • the RNA strand is the antisense strand as that is the strand that binds to the target mRNA.
  • the siHybrid created by the hybridization of the DNA and RNA strands have a hybridized complementary portion and preferably at least one 3′overhanging end.
  • a cholesterol moiety is a cholesterol molecule, sterol or any compound derived from cholesterol including chlolestanol, ergosterol, stimastanol, stigmasterol, methyl-lithocholic acid, cortisol, corticosterone, ⁇ 5 -pregnenolone, progesterone, deoxycorticosterone, 17-OH-pregnenolone, 17-OH-progesterone, 11-dioxycortisol, dehydroepiandrosterone, dehydroepiandrosterone sulfate, androstenedione, aldosterone, 18-hydroxycorticosterone, tetrahydrocortisol, tetrahydrocortisone, cortisone, prednisone, 6 ⁇ -methylpredisone, 9 ⁇ -fluoro-16 ⁇ -hydroxyprednisolone, 9 ⁇ -fluoro-16 ⁇ -methylprednisolone, 9 ⁇ -fluorocort
  • a cholesterol-conjugated siRNA or siHybrid is formulated with, or delivered in a coordinate administration method with, one or more secondary delivery-enhancing agent(s) that is/are further effective to enhance delivery of the cholesterol-conjugated siRNA or siHybrid into mammalian cells.
  • the second delivery-enhancing agent(s) is/are effective to facilitate delivery of the cholesterol-conjugated siRNA or siHybrid across the plasma membrane and into the cytoplasm of a targeted mammalian cell.
  • the targeted cell may be any cell for which delivery of a cholesterol-conjugated siRNA or siHybrid into the cell for regulation of gene expression is desired.
  • target cells in this context include pulmonary alveolar or other airway cells, skin cells, hepatic cells, renal cells, pancreatic cells, endothelial cells, nucleated blood cells (e.g., lymphocytes, monocytes, macrophages, or dendritic cells), muscle cells (e.g., cardiac or smooth muscle cells), mammary cells, peripheral or central nervous system (CNS) cells, cells of the stomach or intestinal tract, tumor cells, and other cells that are amenable to gene regulation for therapeutic purposes according to the methods and compositions of the invention.
  • nucleated blood cells e.g., lymphocytes, monocytes, macrophages, or dendritic cells
  • muscle cells e.g., cardiac or smooth muscle cells
  • mammary cells e.g., peripheral or central nervous system (CNS) cells
  • CNS central nervous system
  • the cholesterol-conjugated siRNA or siHybrid are targeted for delivery to mucosal epithelial cells, for example nasal mucosal epithelial cells.
  • the secondary delivery-enhancing agent(s) may be selected from one or any combination of the following:
  • a membrane penetration-enhancing agent selected from (i) a surfactant, (ii) a bile salt, (iii) a phospholipid additive, mixed micelle, liposome, or carrier, (iv) an alcohol, (v) an enamine, (vi) an NO donor compound, (vii) a long-chain amphipathic molecule (viii) a small hydrophobic penetration enhancer; (ix) sodium or a salicylic acid derivative; (x) a glycerol ester of acetoacetic acid (xi) a cyclodextrin or beta-cyclodextrin derivative, (xii) a medium-chain fatty acid, (xiii) a chelating agent, (xiv) an amino acid or salt thereof, (xv) an N-acetylamino acid or salt thereof, (xvi) an enzyme degradative to a selected membrane component, (xvii) an inhibitor of fatty acid synthesis, or (xviii) an enzyme de
  • the delivery-enhancing agent(s) comprise(s) any one or any combination of two or more of the foregoing delivery-enhancing agents recited in (a)-(k), and the formulation of the cholesterol-conjugated siRNA or siHybrid with the delivery-enhancing agents provides for increased delivery of the cholesterol-conjugated siRNA or siHybrid into the cytoplasm of target cells for gene regulation by the cholesterol-conjugated siRNA or siHybrid.
  • any one or combination of the foregoing secondary delivery-enhancing agents may be added to a pharmaceutical composition comprising a cholesterol-conjugated siRNA or siHybrid as described herein, to yield a combinatorial formulation providing greater delivery enhancement in comparison to intracellular delivery of the cholesterol-conjugated siRNA or siHybrid without the secondary delivery-enhancing agent(s).
  • the cholesterol-conjugated siRNA or siHybrid is administered to a target cell, tissue, or individual in combination with one or more secondary delivery-enhancing agents in a coordinate administration protocol.
  • the cholesterol-conjugated siRNA or siHybrid is administered to the same cell, tissue, or individual as the secondary delivery-enhancing agent(s), prior to, simultaneous with, or after administration of the secondary delivery-enhancing agent(s), which similarly may be selected from any one or combination of the following:
  • a membrane penetration-enhancing agent selected from (i) a surfactant, (ii) a bile salt, (iii) a phospholipid additive, mixed micelle, liposome, or carrier, (iv) an alcohol, (v) an enamine, (vi) an NO donor compound, (vii) a long-chain amphipathic molecule (viii) a small hydrophobic penetration enhancer; (ix) sodium or a salicylic acid derivative; (x) a glycerol ester of acetoacetic acid (xi) a cyclodextrin or beta-cyclodextrin derivative, (xii) a medium-chain fatty acid, (xiii) a chelating agent, (xiv) an amino acid or salt thereof, (xv) an N-acetylamino acid or salt thereof, (xvi) an enzyme degradative to a selected membrane component, (xvii) an inhibitor of fatty acid synthesis, or (xviii) an enzyme de
  • the coordinate administration of the cholesterol-conjugated siRNA or siHybrid and secondary delivery-enhancing agent(s) provides for increased uptake of the cholesterol-conjugated siRNA or siHybrid into the cytoplasm of targeted cells, typically enhancing gene regulation (e.g., increasing knockdown of mRNA translation to thereby reduce expression of one or more selected protein(s), such as TNF- ⁇ , in the target cell.
  • a delivery-enhancing peptide is employed as the secondary delivery-enhancing agent.
  • the delivery-enhancing peptide may be conjugated to, combinatorially formulated with, or coordinately administered with, the cholesterol-conjugated siRNA or siHybrid to enhance intracellular uptake of the cholesterol-conjugated siRNA or siHybrid and improve gene regulation results achieved thereby.
  • Delivery-enhancing peptides in this context may include natural or synthetic, therapeutically or prophylactically active, peptides (comprised of two or more covalently linked amino acids), proteins, peptide or protein fragments, peptide or protein analogs, peptide or protein mimetics, and chemically modified derivatives or salts of active peptides or proteins.
  • delivery-enhancing peptide will often be intended to embrace all of these active species, i.e., peptides and proteins, peptide and protein fragments, peptide and protein analogs, peptide and protein mimetics, and chemically modified derivatives and salts of active peptides or proteins.
  • the delivery-enhancing peptide comprises a mutein that is readily obtainable by partial substitution, addition, or deletion of amino acids within a naturally occurring or native (e.g., wild-type, naturally occurring mutant, or allelic variant) peptide or protein sequence (e.g., a sequence of a naturally occurring “cell penetrating peptide” or peptide fragment of a native protein, such as a tight junction protein).
  • a naturally occurring or native e.g., wild-type, naturally occurring mutant, or allelic variant
  • biologically active fragments of native peptides or proteins are included. Such mutant derivatives and fragments substantially retain the desired cell penetrating or other delivery-enhancing activity of the corresponding native peptide or proteins.
  • biologically active variants marked by alterations in these carbohydrate species are also included within the invention.
  • the delivery-enhancing peptides, proteins, analogs and mimetics for use within the methods and compositions of the invention are may be conjugated to, or formulated with, the cholesterol-conjugated siRNA or siHybrid to yield a pharmaceutical composition that includes a delivery-enhancing effective amount of the delivery-enhancing peptide, protein, analog or mimetic (i.e., an amount of the peptide sufficient to detectably enhance intracellular delivery of the cholesterol-conjugated siRNA or siHybrid).
  • Exemplary delivery-enhancing peptides for use within the methods and compositions of the invention include any one or combination of the following peptides, or active fragments, muteins, conjugates, or complexes thereof:
  • Delivery-enhancing peptides of the invention may further include various modifications known in the art, e.g., for modifying the charge, membrane permeability, half-life, degradative potential, reactivity (e.g., to form conjugates), immunogenicity, or other desired properties of the subject peptide.
  • exemplary modified delivery-enhancing peptides in this context may include, for example, peptides modified by incorporation of one or more selected amino- or carboxy-terminal chemical modifications.
  • amino- and/or carboxy-terminal amide, BrAc, or maleimide groups may be included, as exemplified by the modified delivery-enhancing peptides shown in Table 1.
  • the + and ⁇ notations indicated in Table 1 for the listed peptides relate to activity of the peptides to enhance permeation of across epithelial monolayers—as determined by measurement of peptide-mediated changes in trans-epithelial electrical resistance (TEER).
  • TEER trans-epithelial electrical resistance
  • a + notation indicates that the subject peptide enhances epithelial permeation of macromolecules.
  • the peptides that exhibit permeation-enhancing activity can be tested and selected according to the methods herein to determine their utility for enhancing delivery of cholesterol-conjugated siRNA or siHybrid into the cytoplasm of targeted cells to enhance gene regulation
  • Unmodified siRNAs were synthesized according to the general strategy for solid-phase oligonucleotide synthesis. The syntheses proceeded from the 3′- to 5′-direction [current protocols in nucleic acid chemistry, chapter 3]. The first step involved attachment of a mononucleoside/tide to the surface of an insoluble solid support through a covalent bond. All unmodified siRNAs described here were synthesized starting with a CPG-bound deoxythymidine (purchased from Glen Research, Sterling Va.). The thymidine nucleoside is covalently attached to the solid support through 3′-hydroxyl group using a base labile linker.
  • the terminal-protecting group (dimethoxytrityl, DMT) on the nucleoside is removed. This exposes a free 5′-OH group where the next nucleotide unit can be added.
  • An excess of reagents is used to force the coupling reaction to occur on as many of the immobilized nucleotides as possible.
  • excess reagents are washed away.
  • the reaction is followed by a c capping step, to block off non-extended sites, and an oxidation step.
  • the process of terminal-protecting group removal and chain extension is then repeated using different bases until the desired sequence has been assembled.
  • Some or all of the protecting groups may optionally be removed, and then the covalent attachment to the support is hydrolyzed to release the product. Removal of the protecting groups were carried out with 3:1 mixture of concentrated ammonia:ethanol. After removal of any remaining protecting groups, the oligonucleotide is ready for purification and use.
  • RNA syntheses were carried out by Applied Biosystems 3400 using standard phosphoramidite chemistry.
  • 5′-dimethoxytrityl-N-dimethylformamidine-guanosine 2′-O-(t-butyldimethylsilyl)-3′-[(2-cyanoethyl)-(N,N-diisopropyl)]-phosphoramidite (dmf-G-CE phosphoramidite) (II)
  • CPG Controlled Pore glass
  • VI deoxytimidine
  • Other reagents and solvents were purchased from Glen Research (Sterling, Va.) and/or Applied Biosystems (Foster City, Calif.).
  • 3′-cholestery-labelled siRNAs was carried out using the modified support strategy.
  • a new modified solid phase synthesis support must be prepared for each 3′-reporter group or conjugate.
  • the solid phase support for attaching cholesteryl group to the 3′-termini of oligonucleotides is commercially available.
  • the synthesis of the 3′-cholesteryl-labelled oligonucleotides were accomplished using 1-dimethoyxytrityloxy-3-O—(N-cholesteryl-3-aminopropyl)-triethyleneglycol-glyceryl-2-O-succinoyl-long-chain-alkylamino-CPG (VII, Glen Research, Sterling Va.).
  • the designed 21 nucleotide sequence was then assembled on this modified solid support using standard phosphoramidite protocols for RNA synthesis as described herein above.
  • a protected oligonucleotide with a free hydroxyl group at the 5′-end, immobilized on the solid support may easily be obtained by solid phase synthesis using either methodologies described herein above.
  • the 5′-terminal hydroxyl can then be reacted with phosphoramidites.
  • Phosphoramidites often obtained from a molecule having a hydroxyl functionality allow the direct introduction of a functional group or ligand to the chain after oxidation and deprotection.
  • dimethoxytrityloxy-3-O—(N-cholesteryl-3-aminopropyl)-triethyleneglycol-glyceryl-2-O-(2-cyanoethyl)-(N,N-diisopropyl)-phosphoramidite VIII was purchased from Glen Research (Sterling, Va.).
  • the 5′-dimethoyxtrytyl protecting group was cleaved and VIII was coupled to the grown chain
  • Syntheses of 3′,5′-dicholesteryl-labeled siRNAs were accomplished using a combination of the methods described above.
  • the synthesis of such a molecule started with using VII as the “modified solid support”, and elongation and incorporation of the 5′-cholesteryl moiety were carried out as described above.
  • the transfection was performed with either regular siRNA or cholesterol-conjugated siRNA with lipofectamine (Invitrogen) on 9 L/beta-gal cells.
  • the siRNA was designed to specifically knock down beta-galactosidase mRNA and activities are expressed as percentage of beta-gal activities from control (transfected cells by lipofectamine alone).
  • Table 1 above provides results of transfection and mRNA silencing experiments using the siRNA constructs made using sense and antisense strands designated above.
  • the transfection and silencing assay results show cholesterol-enhanced delivery of exemplary siRNAs of the invention, and demonstrate silencing of the beta-galactosidase mRNA by the cholesterol-conjugated siRNAs.
  • the “Activity (% of control)” indicates the beta-galactosidase activity remaining after the transfection. The lower the percentage, the greater was the efficacy of the siRNA construct.
  • the double letters represent a double-stranded siRNA.
  • the exemplary constructs, BE, AF, BA, CA, CF, and AE are representative of the nature and activity of cholesterol conjugated dsRNAs of the present invention. These constructs show greater silencing efficacy than the corresponding unconjugated siRNAs.
  • siRNA constructs CE, AG, BF, DA, BG, DF, DE, CG and DG showed lower efficacy than the unconjugated siRNA construct AA.
  • AA is a siRNA construct with no cholesterol conjugated to any of the ends of the sense or antisense RNA strands. This construct was transfected into the cells resulting in silencing of the beta-galactosidase mRNA so that 23.12% of the activity of the beta-galactosidase mRNA remained.
  • BE is a siRNA construct having a cholesterol moiety linked to the 5′ end of the sense strand and a cholesterol moiety linked to the 5′ end of the antisense strand, and no cholesterol moiety linked to the other ends of the siRNA.
  • This construct was transfected into the cells resulting in silencing the beta-galactosidase mRNA so that only 10.27% of the activity of the beta-galactosidase mRNA remained. This is unexpectedly superior to the unconjugated siRNA.
  • AF is a siRNA construct having a cholesterol moiety linked to the 3′ end of the antisense strand and no cholesterol moiety linked to the other ends of the siRNA strands. This construct was transfected into the cells resulting in silencing the beta-galactosidase mRNA so that only 11.99% of the activity of the beta-galactosidase mRNA remained. This is unexpectedly superior to the unconjugated siRNA.
  • BA is a siRNA construct having a cholesterol moiety linked to the 5′ end of the sense strand and no cholesterol moiety linked to the other ends of the siRNA strands. This construct was transfected into the cells resulting in silencing the beta-galactosidase mRNA so that only 12.09% of the activity of the beta-galactosidase mRNA remained. This is unexpectedly superior to the unconjugated siRNA.
  • CA is a siRNA construct having a cholesterol moiety linked to the 3′ end of the sense strand and no cholesterol moiety linked to the other ends of the siRNA strands. This construct was transfected into the cells resulting in silencing the beta-galactosidase mRNA so that only 16.18% of the activity of the beta-galactosidase mRNA remained. This is unexpectedly superior to the unconjugated siRNA.
  • CF is a siRNA construct having a cholesterol moiety linked to the 3′ end of the sense strand, a cholesterol moiety linked to the 3′ end of the antisense strand, and no cholesterol moiety linked to the other ends of the siRNA strands.
  • This construct was transfected into the cells resulting in silencing the beta-galactosidase mRNA so that only 16.76% of the activity of the beta-galactosidase mRNA remained. This is unexpectedly superior to the unconjugated siRNA.
  • AE is a siRNA construct having a cholesterol moiety linked to the 5′ end of the antisense strand and no cholesterol moiety linked to the other ends of the siRNA strands. This construct was transfected into the cells resulting in silencing the beta-galactosidase mRNA so that only 19.02% of the activity of the beta-galactosidase mRNA remained. This is unexpectedly superior to the unconjugated siRNA.
  • constructs listed below showed lower ability to silence the beta-galactosidase reporter than was determined for the corresponding, unconjugated siRNA.
  • CE is a siRNA construct having a cholesterol moiety linked to the 3′ end of the sense strand, a cholesterol moiety linked 5′ end of the antisense strand, and no cholesterol moiety linked to the other ends of the siRNA strands.
  • This construct was transfected into the cells resulting in silencing the beta-galactosidase mRNA so that 27.62% of the activity of the beta-galactosidase mRNA remained. This silencing effect was lower than that observed for the corresponding, unconjugated siRNA.
  • siRNA construct having a cholesterol moiety linked to the 3′ end of the antisense strand, a cholesterol moiety linked to the 5′ end of the antisense strand, and no cholesterol moiety linked to the other ends of the siRNA strands.
  • This construct was transfected into the cells resulting in silencing the beta-galactosidase mRNA so that 29.87% of the activity of the beta-galactosidase mRNA remained. This silencing effect was lower than that observed for the corresponding, unconjugated siRNA.
  • BF is a siRNA construct having a cholesterol moiety linked to the 5′ end of the sense strand, a cholesterol moiety linked to the 3′ end of the antisense strand, and no cholesterol moiety linked to the other ends of the siRNA strands.
  • This construct was transfected into the cells resulting in silencing the beta-galactosidase mRNA so that 32.02% of the activity of the beta-galactosidase mRNA remained. This silencing effect was lower than that observed for the corresponding, unconjugated siRNA.
  • DA is a siRNA construct having a cholesterol moiety linked to 5′ end of the sense strand, a cholesterol moiety linked to the 3′ end of the sense strand, and no cholesterol moiety linked to the other ends of the siRNA strands.
  • This construct was transfected into the cells resulting in silencing the beta-galactosidase mRNA so that 33.99% of the activity of the beta-galactosidase mRNA remained. This silencing effect was lower than that observed for the corresponding, unconjugated siRNA.
  • BG is a siRNA construct having a cholesterol moiety linked to 5′ end of the sense strand, a cholesterol moiety linked to the 3′ end of the antisense strand, a cholesterol moiety linked to the 5′ end of the antisense strand, and no cholesterol moiety linked to the 3′ end of the sense strand.
  • This construct was transfected into the cells resulting in silencing the beta-galactosidase mRNA so that 46.39% of the activity of the beta-galactosidase mRNA remained. This silencing effect was lower than that observed for the corresponding, unconjugated siRNA.
  • DF is a siRNA construct having a cholesterol moiety linked to 5′ end of the sense strand, a cholesterol moiety linked to the 3′ end of the sense strand, a cholesterol moiety linked to the 3′ end of the antisense strand, and no cholesterol moiety linked to the 5′ end of the antisense strand.
  • This construct was transfected into the cells resulting in silencing the beta-galactosidase mRNA so that 65.40% of the activity of the beta-galactosidase mRNA remained. This silencing effect was lower than that observed for the corresponding, unconjugated siRNA.
  • DE is a siRNA construct having a cholesterol moiety linked to 5′ end of the sense strand, a cholesterol moiety linked to the 3′ end of the sense strand, a cholesterol moiety linked to the 5′ end of the antisense strand and no cholesterol moiety linked to the 3′ end of the antisense strand.
  • This construct was transfected into the cells resulting in silencing the beta-galactosidase mRNA so that 77.12% of the activity of the beta-galactosidase mRNA remained. This silencing effect was lower than that observed for the corresponding, unconjugated siRNA.
  • BG is a siRNA construct having a cholesterol moiety linked to 3′ end of the sense strand, a cholesterol moiety linked to the 3′ end of the sense strand, a cholesterol moiety linked to the 3′ end of the antisense strand, a cholesterol moiety linked to the 5′ end of the antisense strand, and no cholesterol moiety linked to the 5′ end of the sense strand.
  • This construct was transfected into the cells resulting in silencing the beta-galactosidase mRNA so that 77.80% of the activity of the beta-galactosidase mRNA remained. This silencing effect was lower than that observed for the corresponding, unconjugated siRNA.
  • DG is a siRNA construct having a cholesterol moiety on the 5′ end of the sense strand, a cholesterol moiety on the 3′ end of the sense strand, a cholesterol moiety on the 3′ end of the antisense strand, and a cholesterol moiety on the 5′ end of the antisense strand.
  • This construct was transfected into the cells resulting in silencing the beta-galactosidase mRNA so that 98.84% of the activity of the beta-galactosidase mRNA remained. This silencing effect was lower than that observed for the corresponding, unconjugated siRNA.
  • PBMC Peripheral blood mononuclear cells
  • MILTENYI BIOTEC GmbH Germany
  • the purity of the monocytes was greater than 95%, judged by flow cytometry stained with anti-CD14 antibody (BD Biosciences, CA). Purified human monocytes were maintained overnight in complete media before induction and knockdown assay.
  • Fluorescence activated cell sorting analysis were performed using Beckman Coulter FC500 cell analyzer (Fullerton, Calif.). The instrument was adjusted according to the fluorescence probes used (FAM or Cy5 for siRNA and FITC and PE for CD14). Propidium iodide (Fluka, St Lois, Mo.) and AnnexinV (R&D systems, Minneapolis, Minn.) were used as indicators for cell viability and cytotoxicity.
  • siRNA uptake analysis cells were washed with PBS, treated with trypsin (attached cells only), and then analyzed by flow cytometry. Uptake of the siRNA designated BA, described above, was also measured by intensity of Cy5 or FAM fluorescence in the cells and cellular viability assessed by addition of propidium iodide or AnnexinV-PE. In order to differentiate the cellular uptake from the membrane insertion of fluorescence labeled siRNA, trypan blue was used to quench the fluorescence on the cell membrane surface.
  • FIGS. 1 and 2 illustrate the effects of 5% serum on cellular uptake of a cholesterol-conjugated siRNA according to the invention in complex with a permeabilizing peptide delivery enhancing agent, PN73 (cholesterol siRNA+PN73), and on an unconjugated siRNA in complex with PN73 (siRNA+PN73).
  • PN73 cholesterol-conjugated siRNA and siRNA/PN73 complex were transfected into human monocytes in Opti-MEM® media (Invitrogen) as described above, with serum added in fixed or varied concentration(s).
  • the final concentration of siRNA for both cholesterol and complex were 0.2 ⁇ M.
  • the uptake efficiency and Mean fluorescence intensity were assessed by flow cytometry.
  • the cellular uptake values shown in FIGS. 1 and 2 were determined with variation of PN73 concentrations in the presence of a fixed, 5% concentration of serum.
  • FIGS. 3 and 4 illustrate the effects of varying concentrations of serum on cellular uptake of a cholesterol-conjugated siRNA in the presence or absence of a second delivery enhancing agent, lipofectamine, as determined by flow cytometry.
  • a permeabilizing peptide to a delivery formulation comprising a siRNA conjugated to a cholesterol moiety reduces the inhibitory effects of serum on cholesterol-siRNA uptake in a dose dependent manner.
  • additional delivery-enhancing agents including, but not limited to, Lipofectamine and PN73, can further enhance siRNA delivery to mammalian cells and tissues in vitro and in vivo.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • General Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Microbiology (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Virology (AREA)
  • Botany (AREA)
  • Immunology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

A double-stranded RNA, preferably a small, interfering (si)RNA or a siHybrid, to which cholesterol moieties are linked.

Description

    CROSS REFERENCES TO RELATED APPLICATIONS
  • This application is a continuation claiming the benefit under 35 U.S.C. § 120 of copending U.S. patent application Ser. No. 11/107,371, filed Apr. 15, 2005, which claimed the benefit under 35 U.S.C. § 119(e) of U.S. Provisional Application No. 60/564,543, filed Apr. 20, 2004, each of which is incorporated herein by reference in its entirety.
  • BACKGROUND OF THE INVENTION
  • RNA interference is the process of sequence-specific post transcriptional gene silencing in cells initiated by double-stranded RNA (dsRNA) that is homologous in sequence to a portion of a targeted mRNA. Introduction of dsRNA into cells leads to the destruction of the endogenous RNAs that share the same sequence as the dsRNA. The dsRNA molecules are cleaved by an RNase III family nuclease called Dicer into short-interfering RNAs (siRNA), which are 19-23 nucleotides (nt) in length. The siRNAs are incorporated into a multicomponent nuclease complex (RISC, RNA-induced silencing complex), which identifies mRNA substrates through their homology to the siRNA, binds to and destroys the targeted mRNA. In mammalian cells, dsRNAs longer than 30 base pairs can activate the dsRNA-dependent kinase PKR and 2′-5′-oligoadenylate synthetase, normally induced by interferon. By virtue of its small size, synthetic siRNA avoids activation of the interferon response. The activated PKR inhibits general translation by phosphorylation of the translation factor eukaryotic initiation factor 2α (eIF2α), while 2′-5′-oligoadenylate synthetase causes nonspecific mRNA degradation via activation of RNase L.
  • In contrast to the nonspecific effect of long dsRNA, siRNA can mediate selective gene silencing in the mammalian system Hairpin RNA with a short loop and 19 to 27 base pairs in the stem also selectively silences expression of genes that are homologous to the sequence in the double-stranded stem. Mammalian cells can convert short hairpin RNA into siRNA to mediate selective gene silencing.
  • RISC mediates cleavage of single stranded RNA having sequence complementary to the antisense strand of the siRNA duplex. Cleavage of the target RNA takes place in the middle of the region complementary to the antisense strand of the siRNA duplex.
  • Studies have shown that 21 nucleotide siRNA duplexes are most active when containing two nucleotide 3′-overhangs. Furthermore, complete substitution of one or both siRNA strands with 2′-deoxy (2′-H) or 2′-O-methyl nucleotides abolishes RNAi activity, whereas substitution of the 3′-terminal siRNA overhang nucleotides with deoxy nucleotides (2′-H) was shown to be tolerated.
  • Studies have shown that replacing the 3′-overhanging segments of a 21-mer siRNA duplex having 2 nucleotide 3′ overhangs with deoxyribonucleotides does not have an adverse effect on RNAi activity. Replacing up to 4 nucleotides on each end of the siRNA with deoxyribonucleotides has been reported to be well tolerated whereas complete substitution with deoxyribonucleotides results in no RNAi activity.
  • RNA interference is emerging as a promising means for reducing the expression of specific gene products, and thus may be useful for developing therapeutic drugs to treat viral infections, cancers, autoimmune diseases, and other diseases and conditions amenable to treatment by down-regulation of mRNA expression. However, there remains an important need in the art for additional tools and methods to design, produce, formulate, deliver, and use siRNAs as therapeutic tools, including for therapies targeted to specific tissues and cells.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 illustrates serum effects on cellular uptake of a cholesterol-conjugated siRNA in complex with a delivery enhancing agent (comprising a permeabilizing peptide, PN73), and on an unconjugated siRNA in complex with PN73-expressed as percentage uptake.
  • FIG. 2 illustrates serum effects on cellular uptake of a cholesterol-conjugated siRNA in complex with PN73, and on an unconjugated siRNA in complex with PN73—expressed as mean fluorescence intensity (MFI).
  • FIG. 3 illustrates the effects of increasing concentrations of serum on cellular uptake of a cholesterol-conjugated siRNA in the presence or absence of a second delivery enhancing agent, lipofectamine—expressed as percentage uptake.
  • FIG. 4 illustrates the effects of increasing concentrations of serum on cellular uptake of a cholesterol-conjugated siRNA in the presence or absence of a second delivery enhancing agent, lipofectamine—expressed as MFI.
  • DESCRIPTION OF EXEMPLARY EMBODIMENTS OF THE INVENTION
  • The present invention fulfills these needs and satisfies additional objects and advantages by providing double-stranded nucleic acids conjugated to a cholesterol moiety to facilitate delivery of the nucleic acids into a selected target cell or tissue. In particular the present invention is directed towards methods and compositions to administer double-stranded ribonucleic acid to a mammal so as to effectuate transfection of the double-stranded RNA into a desired tissue of the mammal. In certain embodiments the double-stranded RNA has 30 or fewer nucleotides, and is a short interfering RNA (siRNA).
  • It has been surprisingly discovered that selectively conjugating a cholesterol moiety to a siRNA at selective ends of the siRNA sense and/or antisense strands increases the silencing of the targeted mRNA. For example, the following siRNA/cholesterol moiety constructs increase the silencing effect of the targeted mRNA in comparison to siRNA having no cholesterol conjugated to it:
      • 1. A siRNA construct having a cholesterol moiety linked to the 5′ end of the sense strand and the 5′end of the antisense, and no cholesterol moiety at the other ends;
      • 2. A siRNA construct having a cholesterol moiety linked to the 3′ end of the antisense strand, and no cholesterol moiety linked to the other ends of the siRNA strands;
      • 3. A siRNA construct having cholesterol moiety linked to the 5′ end of the sense strand, and no cholesterol moiety linked to the other ends of the siRNA strands;
      • 4. A siRNA construct having a cholesterol moiety linked to the 3′ end of the sense strand and no cholesterol moiety linked to the other ends of the siRNA strands;
      • 5. A siRNA construct having a cholesterol moiety linked to the 3′ end of the sense strand, a cholesterol moiety linked to the 3′ end of the antisense strand and no cholesterol moiety linked to the other ends of the siRNA strands; and
      • 6. A siRNA construct having a cholesterol moiety linked to the 5′ end of the antisense strand and no cholesterol moiety linked to the other ends of the siRNA strands.
  • Thus, the constructs listed above are embodiments of the present invention, as well as those constructs in which the ds nucleic acid is a siHybrid in which the sense strand is a DNA molecule.
  • The following constructs showed a progressively decreased silencing of the targeted mRNA in comparison to a siRNA having no cholesterol moieties conjugated to any of its ends:
    • 1. A siRNA construct having a cholesterol moiety linked to the 3′ end of the sense strand, a cholesterol moiety linked 5′ end of the antisense strand and no cholesterol moiety linked to the other ends of the siRNA strands;
    • 2. A siRNA construct having a cholesterol moiety linked to the 3′ end of the antisense strand, a cholesterol moiety linked to the 5′ end of the antisense strand and no cholesterol moiety linked to the other ends of the siRNA strands;
    • 3. A siRNA construct having a cholesterol moiety linked to the 5′ end of the sense strand a cholesterol moiety linked to the 3′ end of the antisense strand and no cholesterol moiety linked to the other ends of the siRNA strands;
    • 4. A siRNA construct having a cholesterol moiety linked to 5′ end of the sense strand, a cholesterol moiety linked to the 3′ end of the sense strand, and no cholesterol moiety linked to the other ends of the siRNA strands;
    • 5. A siRNA construct having a cholesterol moiety linked to 5′ end of the sense strand, a cholesterol moiety linked to the 3′ end of the antisense strand, a cholesterol moiety linked to the 5′ end of the antisense strand and no cholesterol moiety linked to the 3′ end of the sense strand;
    • 6. A siRNA construct having a cholesterol moiety linked to 5′ end of the sense strand, a cholesterol moiety linked to the 3′ end of the sense strand, a cholesterol moiety linked to the 3′ end of the antisense strand and no cholesterol moiety linked to the 5′ end of the antisense strand;
    • 7. A siRNA construct having a cholesterol moiety linked to 5′ end of the sense strand, a cholesterol moiety linked to the 3′ end of the sense strand, a cholesterol moiety linked to the 5′ end of the antisense strand and no cholesterol moiety linked to the 3′ end of the antisense strand;
    • 8. A siRNA construct having a cholesterol moiety linked to 3′ end of the sense strand, a cholesterol moiety linked to the 3′ end of the sense strand, a cholesterol moiety linked to the 3′ end of the antisense strand, a cholesterol moiety linked to the 5′ end of the antisense strand, and no cholesterol moiety linked to the 5′ end of the sense strand;
    • 9. A siRNA construct having a cholesterol moiety on the 5′ end of the sense strand, a cholesterol moiety on the 3′ end of the sense strand, a cholesterol moiety on the 3′ end of the antisense strand and a cholesterol moiety on the 5′ end of the antisense strand.
    DEFINITIONS
  • As used herein, the term “inverted repeat” refers to a nucleic acid sequence comprising a sense and an antisense element positioned so that they are able to form a double stranded siRNA when the repeat is transcribed. The inverted repeat may optionally include a linker or a heterologous sequence such as a self-cleaving ribozyme between the two elements of the repeat. The elements of the inverted repeat have a length sufficient to form a double stranded RNA. Typically, each element of the inverted repeat is about 15 to about 100 nucleotides in length, preferably about 20-30 base nucleotides, preferably about 20-25 nucleotides in length, e.g., 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides in length.
  • “Silencing” refers to partial or complete loss-of-function through targeted inhibition of gene expression in a cell and may also be referred to as “knock down”. Depending on the circumstances and the biological problem to be addressed, it may be preferable to partially reduce gene expression. Alternatively, it might be desirable to reduce gene expression as much as possible. The extent of silencing may be determined by any method known in the art, some of which are summarized in International Publication No. WO 99/32619. Depending on the assay, quantitation of gene expression permits detection of various amounts of inhibition for example, greater than 10%, 33%, 50%, 90%, 95% or 99%.
  • The phrase “inhibiting expression of a target gene” refers to the ability of a siRNA of the invention to initiate gene silencing of the target gene. To examine the extent of gene silencing, samples or assays of the organism of interest or cells in culture expressing a particular construct are compared to control samples lacking expression of the construct. Control samples (lacking construct expression) are assigned a relative value of 100%. Inhibition of expression of a target gene is achieved when the test value relative to the control is about 90%, preferably 50%, more preferably 25-0%. Suitable assays include, e.g., examination of protein or mRNA levels using techniques known to those of skill in the art such as dot blots, northern blots, in situ hybridization, ELISA, immunoprecipitation, enzyme function, as well as phenotypic assays known to those of skill in the art.
  • “Nucleic acid” refers to deoxyribonucleotides or ribonucleotides and polymers thereof in single- or double-stranded form. The term encompasses nucleic acids containing known nucleotide analogs or modified backbone residues or linkages, which are synthetic, naturally occurring, and non-naturally occurring, which have similar binding properties as the reference nucleic acid, and which are metabolized in a manner similar to the reference nucleotides. Examples of such analogs include, without limitation, phosphorothioates, phosphoramidates, methyl phosphonates, chiral-methyl phosphonates, 2-O-methyl ribonucleotides, peptide-nucleic acids (PNAs).
  • “Large double-stranded RNA” refers to any double-stranded RNA having a size greater than about 40 base pairs (bp) for example, larger than 100 bp or more particularly larger than 300 bp. The sequence of a large dsRNA may represent a segment of a mRNA or the entire mRNA. The maximum size of the large dsRNA is not limited herein. The double-stranded RNA may include modified bases where the modification may be to the phosphate sugar backbone or to the nucleoside. Such modifications may include a nitrogen or sulfur heteroatom or any other modification known in the art.
  • The double-stranded structure may be formed by self-complementary RNA strand such as occurs for a hairpin or a micro RNA or by annealing of two distinct complementary RNA strands.
  • “Overlapping” refers to when two RNA fragments have sequences which overlap by a plurality of nucleotides on one strand, for example, where the plurality of nucleotides (nt) numbers as few as 2-5 nucleotides or by 5-10 nucleotides or more.
  • “One or more dsRNAs” refers to dsRNAs that differ from each other on the basis of sequence.
  • “Target gene or mRNA” refers to any gene or mRNA of interest. Any of the genes previously identified by genetics or by sequencing can be implemented as a target. Target genes or mRNA can include developmental genes and regulatory genes, as well as metabolic or structural genes or genes encoding enzymes. The target gene may be expressed in cells in which a phenotype is being investigated, or in an organism in a manner that directly or indirectly impacts a phenotypic characteristic. The target gene may be endogenous or exogenous. Such cells include any cell in the body of an adult or embryonic animal or plant including gamete or any isolated cell such as occurs in an immortal cell line or primary cell culture.
  • In this specification and the appended claims, the singular forms of “a”, “an” and “the” include plural reference unless the context clearly dictates otherwise.
  • “siRNA” means a small interfering RNA that is a short-length double-stranded RNA that are not toxic in mammalian cells. The length is not limited to 21 to 23 bp long. There is no particular limitation in the length of siRNA as long as it does not show toxicity. “siRNAs” can be, for example, 15 to 49 bp, preferably 15 to 35 bp, and more preferably 21 to 30 bp long. Alternatively, the double-stranded RNA portion of a final transcription product of siRNA to be expressed can be, for example, 15 to 49 bp, preferably 15 to 35 bp, and more preferably 21 to 30 bp long. The double-stranded RNA portions of siRNAs in which two RNA strands pair up are not limited to the completely paired ones, and may contain nonpairing portions due to mismatch (the corresponding nucleotides are not complementary), bulge (lacking in the corresponding complementary nucleotide on one strand), and the like. Nonpairing portions can be contained to the extent that they do not interfere with siRNA formation. The “bulge” used herein preferably comprise 1 to 2 nonpairing nucleotides, and the double-stranded RNA region of siRNAs in which two RNA strands pair up contains preferably 1 to 7, more preferably 1 to 5 bulges. In addition, the “mismatch” used herein is contained in the double-stranded RNA region of siRNAs in which two RNA strands pair up, preferably 1 to 7, more preferably 1 to 5, in number. In a preferable mismatch, one of the nucleotides is guanine, and the other is uracil. Such a mismatch is due to a mutation from C to T, G to A, or mixtures thereof in DNA coding for sense RNA, but not particularly limited to them. Furthermore, in the present invention, the double-stranded RNA region of siRNAs in which two RNA strands pair up may contain both bulge and mismatched, which sum up to, preferably 1 to 7, more preferably 1 to 5 in number.
  • The terminal structure of siRNA may be either blunt or cohesive (overhanging) as long as siRNA enables to silence the target gene expression due to its RNAi effect. The cohesive (overhanging) end structure is not limited only to the 3′ overhang as reported by Tuschl et al. (ibid.), and the 5′ overhanging structure may be included as long as it is capable of inducing the RNAi effect. In addition, the number of overhanging nucleotides is not limited to the reported 2 or 3, but can be any numbers as long as the overhang is capable of inducing the RNAi effect. For example, the overhang may be 1 to 8, or 2 to 4 nucleotides. Herein, the total length of siRNA having cohesive end structure is expressed as the sum of the length of the paired double-stranded portion and that of a pair comprising overhanging single-strands at both ends. For example, in the case of 19 bp double-stranded RNA portion with 4 nucleotide overhangs at both ends, the total length is expressed as 23 bp. Furthermore, since this overhanging sequence has low specificity to a target gene, it is not necessarily complementary (antisense) or identical (sense) to the target gene sequence. Furthermore, as long as the siRNA is able to maintain its gene silencing effect on the target gene, it may comprise a low molecular weight RNA (which may be a natural RNA molecule such as tRNA, rRNA or viral RNA, or an artificial RNA molecule), for example, in the overhanging portion at its one end.
  • In addition, the terminal structure of the “siRNA” is necessarily the cut off structure at both ends as described above, and may have a stem-loop structure in which ends of one side of double-stranded RNA are connected by a linker RNA. The length of the double-stranded RNA region (stem-loop portion) can be, for example, 15 to 49 bp, preferably 15 to 35 bp, and more preferably 21 to 30 bp long. Alternatively, the length of the double-stranded RNA region that is a final transcription product of siRNAs to be expressed is, for example, 15 to 49 bp, preferably 15 to 35 bp, and more preferably 21 to 30 bp long. Furthermore, there is no particular limitation in the length of the linker as long as it has a length so as not to hinder the pairing of the stem portion. For example, for stable pairing of the stem portion and suppression of the recombination between DNAs coding for the portion, the linker portion may have a clover-leaf tRNA structure. Even though the linker has a length that hinders pairing of the stem portion, it is possible, for example, to construct the linker portion to include introns so that the introns are excised during processing of precursor RNA into mature RNA, thereby allowing pairing of the stem portion. In the case of a stem-loop siRNA, either end (head or tail) of RNA with no loop structure may have a low molecular weight RNA. As described above, this low molecular weight RNA may be a natural RNA molecule such as tRNA, rRNA or viral RNA, or an artificial RNA molecule.
  • “Antisense RNA” is an RNA strand having a sequence complementary to a target gene mRNA, and thought to induce RNAi by binding to the target gene mRNA. “Sense RNA” has a sequence complementary to the antisense RNA, and annealed to its complementary antisense RNA to form siRNA. These antisense and sense RNAs have been conventionally synthesized with an RNA synthesizer.
  • As used herein, the term “RNAi construct” is a generic term used throughout the specification to include small interfering RNAs (siRNAs), hairpin RNAs, and other RNA species which can be cleaved in vivo to form siRNAs. RNAi constructs herein also include expression vectors (also referred to as RNAi expression vectors) capable of giving rise to transcripts which form dsRNAs or hairpin RNAs in cells, and/or transcripts which can produce siRNAs in vivo. Optionally, the siRNA include single strands or double strands of siRNA.
  • An siHybrid molecule is a double-stranded nucleic acid that has a similar function to siRNA. Instead of a double-stranded RNA molecule, a siHybrid is comprised of an RNA strand and a DNA strand. Preferably, the RNA strand is the antisense strand as that is the strand that binds to the target mRNA. The siHybrid created by the hybridization of the DNA and RNA strands have a hybridized complementary portion and preferably at least one 3′overhanging end.
  • A cholesterol moiety is a cholesterol molecule, sterol or any compound derived from cholesterol including chlolestanol, ergosterol, stimastanol, stigmasterol, methyl-lithocholic acid, cortisol, corticosterone, Δ5-pregnenolone, progesterone, deoxycorticosterone, 17-OH-pregnenolone, 17-OH-progesterone, 11-dioxycortisol, dehydroepiandrosterone, dehydroepiandrosterone sulfate, androstenedione, aldosterone, 18-hydroxycorticosterone, tetrahydrocortisol, tetrahydrocortisone, cortisone, prednisone, 6α-methylpredisone, 9α-fluoro-16α-hydroxyprednisolone, 9α-fluoro-16α-methylprednisolone, 9α-fluorocortisol, testosterone, dihydrotestosterone, androstenediol, androstenedione, androstenedione, 3α,5α-androstanediol, estrone, estradiol, estrogen, spermidine cholesterol carbamate, N4-spermidine cholesteryl carbamate, N4-spermidine cholesteryl carbamate di HCl salt, N4-spermidine-7 dehydro cholesteryl carbamate, N4-spermine cholesteryl carbamate, N,N bis(3-aminopropyl) cholesteryl carbamate, N,N bis(6-aminohexyl) cholesteryl carbamate, N4-spermidine dihydrocholesteryl carbamate, N4-spermidine lithocholic carbamate methyl ester, N1,N8-bis(3-aminopropyl-N4-spermidine cholesteryl carbamate, N(N4-3-aminopropylspermidine) cholesteryl carbamate, N,N-bis(4-aminobutyl) cholesteryl carbamate, N4-spermidine cholesteryl urea, N4-spermine cholesteryl urea, N4-spermidine dihydro cholesteryl urea, N4-spermine dihydro cholesteryl urea, N,N-bis(N′-3-aminopropyl-N″4-aminobutyl) cholesteryl carbamate, N4spermidine cholesteryl carboxamide, and N—[N1,N4,N8-tris (3-aminopropyl)spermidine] cholesteryl carbamate, lumisterol, cholic acid, desoxycholic acid, chenodesoxycholic acid and lithocholic acid and derivatives thereof (see, e.g., U.S. Pat. No. 6,331,524).
  • The following exemplary cholesterol-RNA constructs are illustrative of various embodiments of the invention:
      • 1. A siRNA or siHybrid construct having a cholesterol moiety linked to the 5′ end of the sense strand and the 5′ end of the antisense and no cholesterol moiety at the other ends;
      • 2. A siRNA or siHybrid construct having a cholesterol moiety linked to the 3′ end of the antisense strand and no cholesterol moiety linked to the other ends of the siRNA or siHybrid strands;
      • 3. A siRNA or siHybrid construct having cholesterol moiety linked to the 5′ end of the sense strand and no cholesterol moiety linked to the other ends of the siRNA or siHybrid strands;
      • 4. A siRNA or siHybrid construct having a cholesterol moiety linked to the 3′ end of the sense strand and no cholesterol moiety linked to the other ends of the siRNA or siHybrid strands;
      • 5. A siRNA or siHybrid construct having a cholesterol moiety linked to the 3′ end of the sense strand, a cholesterol moiety linked to the 3′ end of the antisense strand and no cholesterol moiety linked to the other ends of the siRNA or siHybrid strands; and
      • 6. A siRNA or siHybrid construct having a cholesterol moiety linked to the 5′ end of the antisense strand and no cholesterol moiety linked to the other ends of the siRNA or siHybrid strands.
  • In more detailed embodiments of the invention, a cholesterol-conjugated siRNA or siHybrid is formulated with, or delivered in a coordinate administration method with, one or more secondary delivery-enhancing agent(s) that is/are further effective to enhance delivery of the cholesterol-conjugated siRNA or siHybrid into mammalian cells. Typically the second delivery-enhancing agent(s) is/are effective to facilitate delivery of the cholesterol-conjugated siRNA or siHybrid across the plasma membrane and into the cytoplasm of a targeted mammalian cell. The targeted cell may be any cell for which delivery of a cholesterol-conjugated siRNA or siHybrid into the cell for regulation of gene expression is desired. Exemplary target cells in this context include pulmonary alveolar or other airway cells, skin cells, hepatic cells, renal cells, pancreatic cells, endothelial cells, nucleated blood cells (e.g., lymphocytes, monocytes, macrophages, or dendritic cells), muscle cells (e.g., cardiac or smooth muscle cells), mammary cells, peripheral or central nervous system (CNS) cells, cells of the stomach or intestinal tract, tumor cells, and other cells that are amenable to gene regulation for therapeutic purposes according to the methods and compositions of the invention.
  • In on exemplary embodiment, the cholesterol-conjugated siRNA or siHybrid are targeted for delivery to mucosal epithelial cells, for example nasal mucosal epithelial cells.
  • Within these and related aspects of the invention, the secondary delivery-enhancing agent(s) may be selected from one or any combination of the following:
  • (a) an aggregation inhibitory agent;
  • (b) a charge modifying agent;
  • (c) a pH control agent;
  • (d) a degradative enzyme inhibitory agent;
  • (e) a mucolytic or mucus clearing agent;
  • (f) a ciliostatic agent;
  • (g) a membrane penetration-enhancing agent selected from (i) a surfactant, (ii) a bile salt, (iii) a phospholipid additive, mixed micelle, liposome, or carrier, (iv) an alcohol, (v) an enamine, (vi) an NO donor compound, (vii) a long-chain amphipathic molecule (viii) a small hydrophobic penetration enhancer; (ix) sodium or a salicylic acid derivative; (x) a glycerol ester of acetoacetic acid (xi) a cyclodextrin or beta-cyclodextrin derivative, (xii) a medium-chain fatty acid, (xiii) a chelating agent, (xiv) an amino acid or salt thereof, (xv) an N-acetylamino acid or salt thereof, (xvi) an enzyme degradative to a selected membrane component, (xvii) an inhibitor of fatty acid synthesis, or (xviii) an inhibitor of cholesterol synthesis; or (xix) any combination of the membrane penetration enhancing agents recited in (g)(i)-(xix);
  • (h) a delivery-enhancing peptide;
  • (i) a vasodilator agent;
  • (j) a selective transport-enhancing agent; and
  • (k) a stabilizing delivery vehicle, carrier, support or complex-forming species with which the cholesterol-conjugated siRNA or siHybrid is effectively combined, associated, contained, encapsulated or bound resulting in stabilization of the siRNA or siHybrid for enhanced delivery.
  • In additional aspects of the invention, the delivery-enhancing agent(s) comprise(s) any one or any combination of two or more of the foregoing delivery-enhancing agents recited in (a)-(k), and the formulation of the cholesterol-conjugated siRNA or siHybrid with the delivery-enhancing agents provides for increased delivery of the cholesterol-conjugated siRNA or siHybrid into the cytoplasm of target cells for gene regulation by the cholesterol-conjugated siRNA or siHybrid.
  • Any one or combination of the foregoing secondary delivery-enhancing agents may be added to a pharmaceutical composition comprising a cholesterol-conjugated siRNA or siHybrid as described herein, to yield a combinatorial formulation providing greater delivery enhancement in comparison to intracellular delivery of the cholesterol-conjugated siRNA or siHybrid without the secondary delivery-enhancing agent(s).
  • Within coordinate administration methods of the invention, the cholesterol-conjugated siRNA or siHybrid is administered to a target cell, tissue, or individual in combination with one or more secondary delivery-enhancing agents in a coordinate administration protocol. Within these coordinate administration methods, the cholesterol-conjugated siRNA or siHybrid is administered to the same cell, tissue, or individual as the secondary delivery-enhancing agent(s), prior to, simultaneous with, or after administration of the secondary delivery-enhancing agent(s), which similarly may be selected from any one or combination of the following:
  • (a) an aggregation inhibitory agent;
  • (b) a charge modifying agent;
  • (c) a pH control agent;
  • (d) a degradative enzyme inhibitory agent;
  • (e) a mucolytic or mucus clearing agent;
  • (f) a ciliostatic agent;
  • (g) a membrane penetration-enhancing agent selected from (i) a surfactant, (ii) a bile salt, (iii) a phospholipid additive, mixed micelle, liposome, or carrier, (iv) an alcohol, (v) an enamine, (vi) an NO donor compound, (vii) a long-chain amphipathic molecule (viii) a small hydrophobic penetration enhancer; (ix) sodium or a salicylic acid derivative; (x) a glycerol ester of acetoacetic acid (xi) a cyclodextrin or beta-cyclodextrin derivative, (xii) a medium-chain fatty acid, (xiii) a chelating agent, (xiv) an amino acid or salt thereof, (xv) an N-acetylamino acid or salt thereof, (xvi) an enzyme degradative to a selected membrane component, (xvii) an inhibitor of fatty acid synthesis, or (xviii) an inhibitor of cholesterol synthesis; or (xix) any combination of the membrane penetration enhancing agents recited in (g)(i)-(xix);
  • (h) a delivery-enhancing peptide;
  • (i) a vasodilator agent;
  • (j) a selective transport-enhancing agent; and
  • (k) a stabilizing delivery vehicle, carrier, support or complex-forming species with which the cholesterol-conjugated siRNA or siHybrid is effectively combined, associated, contained, encapsulated or bound resulting in stabilization of the siRNA or siHybrid for enhanced intracellular delivery. The coordinate administration of the cholesterol-conjugated siRNA or siHybrid and secondary delivery-enhancing agent(s) provides for increased uptake of the cholesterol-conjugated siRNA or siHybrid into the cytoplasm of targeted cells, typically enhancing gene regulation (e.g., increasing knockdown of mRNA translation to thereby reduce expression of one or more selected protein(s), such as TNF-α, in the target cell.
  • Additional detailed description pertaining to secondary delivery-enhancing agents, for use within the instant invention is provided, for example, in United States Provisional Patent Applications Nos. 60/612,121, filed Sep. 21, 2004; 60/667,835, filed Apr. 1, 2005; 60/612,285, filed Sep. 21, 2004; 60/667,871, filed Apr. 1, 2005; 60/613,416, filed Sep. 27, 2004; and 60/667,833, filed Apr. 1, 2005, each incorporated herein by reference.
  • Within exemplary embodiments of the invention, a delivery-enhancing peptide is employed as the secondary delivery-enhancing agent. The delivery-enhancing peptide may be conjugated to, combinatorially formulated with, or coordinately administered with, the cholesterol-conjugated siRNA or siHybrid to enhance intracellular uptake of the cholesterol-conjugated siRNA or siHybrid and improve gene regulation results achieved thereby. Delivery-enhancing peptides in this context may include natural or synthetic, therapeutically or prophylactically active, peptides (comprised of two or more covalently linked amino acids), proteins, peptide or protein fragments, peptide or protein analogs, peptide or protein mimetics, and chemically modified derivatives or salts of active peptides or proteins. Thus, as used herein, the term “delivery-enhancing peptide” will often be intended to embrace all of these active species, i.e., peptides and proteins, peptide and protein fragments, peptide and protein analogs, peptide and protein mimetics, and chemically modified derivatives and salts of active peptides or proteins. Often, the delivery-enhancing peptide comprises a mutein that is readily obtainable by partial substitution, addition, or deletion of amino acids within a naturally occurring or native (e.g., wild-type, naturally occurring mutant, or allelic variant) peptide or protein sequence (e.g., a sequence of a naturally occurring “cell penetrating peptide” or peptide fragment of a native protein, such as a tight junction protein). Additionally, biologically active fragments of native peptides or proteins are included. Such mutant derivatives and fragments substantially retain the desired cell penetrating or other delivery-enhancing activity of the corresponding native peptide or proteins. In the case of peptides or proteins having carbohydrate chains, biologically active variants marked by alterations in these carbohydrate species are also included within the invention.
  • The delivery-enhancing peptides, proteins, analogs and mimetics for use within the methods and compositions of the invention are may be conjugated to, or formulated with, the cholesterol-conjugated siRNA or siHybrid to yield a pharmaceutical composition that includes a delivery-enhancing effective amount of the delivery-enhancing peptide, protein, analog or mimetic (i.e., an amount of the peptide sufficient to detectably enhance intracellular delivery of the cholesterol-conjugated siRNA or siHybrid).
  • Exemplary delivery-enhancing peptides for use within the methods and compositions of the invention include any one or combination of the following peptides, or active fragments, muteins, conjugates, or complexes thereof:
  • (SEQ ID NO: 1)
    RKKRRQRRRPPQCAAVALLPAVLLALLAP;
    (SEQ ID NO: 2)
    RQIKIWFQNRRMKWKK;
    (SEQ ID NO: 3)
    GWTLNSAGYLLGKINLKALAALAKKIL;
    (SEQ ID NO: 4)
    KLALKLALKALKAALKLA;
    (SEQ ID NO: 7)
    KLWSAWPSLWSSLWKP;
    (SEQ ID NO: 8)
    AAVALLPAVLLALLAPRKKRRQRRRPPQ;
    (SEQ ID NO: 9)
    LLETLLKPFQCRICMRNFSTRQARRNHRRRHRR
    (SEQ ID NO: 10)
    RRRQRRKRGGDIMGEWGNEIFGAIAGFLG;
    (SEQ ID NO: 11)
    KETWWETWWTEWSQPGRKKRRQRRRPPQ;
    (SEQ ID NO: 12)
    GLGSLLKKAGKKLKQPKSKRKV;
    and
    (SEQ ID NO: 13)
    KGSKKAVTKAQKKDGKKRKRSRKESYSVYVYKVLKQ
  • Delivery-enhancing peptides of the invention may further include various modifications known in the art, e.g., for modifying the charge, membrane permeability, half-life, degradative potential, reactivity (e.g., to form conjugates), immunogenicity, or other desired properties of the subject peptide. Exemplary modified delivery-enhancing peptides in this context may include, for example, peptides modified by incorporation of one or more selected amino- or carboxy-terminal chemical modifications. For example, amino- and/or carboxy-terminal amide, BrAc, or maleimide groups may be included, as exemplified by the modified delivery-enhancing peptides shown in Table 1.
  • TABLE 1
    Peptide Sequences Effects
    PN0028 RKKRRQRRRPPQCAAVALLPAVLLALLAP-amide +
    (SEQ ID NO: 1)
    PN0058 RQIKIWFQNRRMKWKK-amide +
    (SEQ ID NO: 2)
    PN0064 BrAc-GWTLNSAGYLLGKINLKALAALAKKILamide +
    (SEQ ID NO: 3)
    PN0068 BrAc-KLALKLALKALKAALKLA-amide +
    (SEQ ID NO: 4)
    PN0069 GRKKRRQRRRPQ-amide
    (SEQ ID NO: 5)
    PN0071 RRRRRRR
    (SEQ ID NO: 6)
    PN0228 NH2-KLWSAWPSLWSSLWKP-amide +/−
    (SEQ ID NO: 7)
    PN027 NH2-AAVALLPAVLLALLAPRKKRRQRRRPPQ-amide +
    (SEQ ID NO: 8)
    PN202 NH2-LLETLLKPFQCRICMRNFSTRQARRNHRRRHRR-amide +
    (SEQ ID NO: 9)
    PN250 NH2-RRRQRRKRGGDIMGEWGNEIFGAIAGFLG-amide +
    (SEQ ID NO: 10)
    PN183 NH2-KETWWETWWTEWSQPGRKKRRQRRRPPQ-amide +
    (SEQ ID NO: 11)
    PN283 Maleimide-GLGSLLKKAGKKLKQPKSKRKV-amide +
    (SEQ ID NO: 12)
    PN073 KGSKKAVTKAQKKDGKKRKRSRKESYSVYVYKVLKQ-amide +
    (SEQ ID NO: 13)
    Assay medium only
  • The + and − notations indicated in Table 1 for the listed peptides relate to activity of the peptides to enhance permeation of across epithelial monolayers—as determined by measurement of peptide-mediated changes in trans-epithelial electrical resistance (TEER). A + notation indicates that the subject peptide enhances epithelial permeation of macromolecules. The peptides that exhibit permeation-enhancing activity can be tested and selected according to the methods herein to determine their utility for enhancing delivery of cholesterol-conjugated siRNA or siHybrid into the cytoplasm of targeted cells to enhance gene regulation
  • The above disclosure generally describes the present invention, which is further exemplified by the following examples. These examples are described solely for purposes of illustration, and are not intended to limit the scope of the invention. Although specific terms and values have been employed herein, such terms and values will likewise be understood as exemplary and non-limiting to the scope of the invention.
  • Example 1 Synthesis and Purification of Cholesteryl-Labeled siRNA
  • Synthesis of Unmodified siRNAs:
  • Unmodified siRNAs were synthesized according to the general strategy for solid-phase oligonucleotide synthesis. The syntheses proceeded from the 3′- to 5′-direction [current protocols in nucleic acid chemistry, chapter 3]. The first step involved attachment of a mononucleoside/tide to the surface of an insoluble solid support through a covalent bond. All unmodified siRNAs described here were synthesized starting with a CPG-bound deoxythymidine (purchased from Glen Research, Sterling Va.). The thymidine nucleoside is covalently attached to the solid support through 3′-hydroxyl group using a base labile linker. Before chain elongation can proceed, the terminal-protecting group (dimethoxytrityl, DMT) on the nucleoside is removed. This exposes a free 5′-OH group where the next nucleotide unit can be added. An excess of reagents is used to force the coupling reaction to occur on as many of the immobilized nucleotides as possible. After the coupling reaction, excess reagents are washed away. The reaction is followed by a c capping step, to block off non-extended sites, and an oxidation step. The process of terminal-protecting group removal and chain extension is then repeated using different bases until the desired sequence has been assembled. Some or all of the protecting groups may optionally be removed, and then the covalent attachment to the support is hydrolyzed to release the product. Removal of the protecting groups were carried out with 3:1 mixture of concentrated ammonia:ethanol. After removal of any remaining protecting groups, the oligonucleotide is ready for purification and use.
  • RNA syntheses were carried out by Applied Biosystems 3400 using standard phosphoramidite chemistry. The corresponding building blocks, 5′-dimethoxytrityl-N-benzoyladenosine-2′-O-(t-butyldimethylsilyl)-3′-[(2-cyanoethyl)-(N,N-diisopropyl)]-phosphoramidite (Bz-A-CE phosphoramidite) (I), 5′-dimethoxytrityl-N-dimethylformamidine-guanosine, 2′-O-(t-butyldimethylsilyl)-3′-[(2-cyanoethyl)-(N,N-diisopropyl)]-phosphoramidite (dmf-G-CE phosphoramidite) (II), 5′-dimethoxytrytiyl-N-acetylcytidine-2′-O-(t-butyldimethylsilyl)-3′-[(2-cyanoethyl)-(N,N-diisopropyl)]-phosphoramidite (Ac-C-CE phosphoramidite) (III), 5-dimethoxytrityluridine-2′-O-(t-butyldimethylsilyl)-3′-[(2-cyanoethyl)-(N,N-diisopropyl)]-phosphoramidite (U-CE phosphoramidite) (IV) and 5′-dimethoxytrityl-2′-deoxythymidine-3′-[(2-cyanoethyl)-(N,N-diisopropyl)]-phosphoramidite (V) were purchased from Glen Research Inc. (Sterling Va.). For un-modified sequences, the syntheses started on Controlled Pore glass (CPG) bound deoxytimidine (VI) (Applied Biosystems, Foster City, Calif.) in 0.2 or 1.0 μmol scale. Other reagents and solvents were purchased from Glen Research (Sterling, Va.) and/or Applied Biosystems (Foster City, Calif.).
  • Figure US20080261304A1-20081023-C00001
    Figure US20080261304A1-20081023-C00002
  • DMT=4,4′-dimethoxytrytyl
  • Commonly used protected phosphoramidites for the synthesis of RNA
  • Synthesis of 3′-cholesteryl-Labeled siRNA
  • The synthesis of 3′-cholestery-labelled siRNAs was carried out using the modified support strategy. In this method a new modified solid phase synthesis support must be prepared for each 3′-reporter group or conjugate. The solid phase support for attaching cholesteryl group to the 3′-termini of oligonucleotides is commercially available. The synthesis of the 3′-cholesteryl-labelled oligonucleotides were accomplished using 1-dimethoyxytrityloxy-3-O—(N-cholesteryl-3-aminopropyl)-triethyleneglycol-glyceryl-2-O-succinoyl-long-chain-alkylamino-CPG (VII, Glen Research, Sterling Va.). The designed 21 nucleotide sequence was then assembled on this modified solid support using standard phosphoramidite protocols for RNA synthesis as described herein above.
  • Synthesis of 5′-cholesteryl-Labeled siRNA
  • A protected oligonucleotide with a free hydroxyl group at the 5′-end, immobilized on the solid support, may easily be obtained by solid phase synthesis using either methodologies described herein above. The 5′-terminal hydroxyl can then be reacted with phosphoramidites. Phosphoramidites often obtained from a molecule having a hydroxyl functionality allow the direct introduction of a functional group or ligand to the chain after oxidation and deprotection. To incorporated a cholesteryl group to the 5′-end of siRNA molecules, dimethoxytrityloxy-3-O—(N-cholesteryl-3-aminopropyl)-triethyleneglycol-glyceryl-2-O-(2-cyanoethyl)-(N,N-diisopropyl)-phosphoramidite (VIII) was purchased from Glen Research (Sterling, Va.). During the solid support synthesis of siRNA, after the incorporation of the last nucleoside/tide, the 5′-dimethoyxtrytyl protecting group was cleaved and VIII was coupled to the grown chain
  • Figure US20080261304A1-20081023-C00003
  • 1-dimethoyxytrityloxy-3-O—(N-cholesteryl-3-aminopropyl)-triethyleneglycol-glyceryl-2-O-succinoyl-long-chain-alkylamino-CPG (VII)
  • Figure US20080261304A1-20081023-C00004
  • dimethoxytrityloxy-3-O—(N-cholesteryl-3-aminopropyl)-triethyleneglycol-glyceryl-2-O-(2-cyanoethyl)-(N,N-diisopropyl)-phosphoramidite (VIII)
    Syntheses of 3′ and 5′-dicholesteryl-Labeled siRNAs
  • Syntheses of 3′,5′-dicholesteryl-labeled siRNAs were accomplished using a combination of the methods described above. The synthesis of such a molecule started with using VII as the “modified solid support”, and elongation and incorporation of the 5′-cholesteryl moiety were carried out as described above.
  • Example 2 Cholesterol-Enhanced Uptake of siRNA and Silencing of Beta-Galactosidase mRNA Expression Transfection of 9 L/LacZ Cells: Day 0:
      • a) Take saturated 9 L/LacZ culture from T75 flask, detach cell and dilute into 10 ml with complete medium (DMEM, 1×PS, 1×Na Pyruvate, 1×NEAA).
      • b) Further dilute the cell to 1:15, and seed 100 μl into each 96 well, which should give 50% confluence cell the next day for transfection. Remember to leave the edge well empty and fill with 250 μl water, do not stack up plates in the incubator.
      • c) Incubate overnight at 37° C., 5% CO2 incubator.
    Day 1:
      • a) Prepare the transfection complex in Opti-MEM, 50 μl each well.
      • b) Dump the medium in plates, wash each well once with 200 μl PBS or Opti-MEM.
      • c) Blot the plates dry completely with tissue by inversion.
      • d) Add the transfection mixture (50 μl/well) into each well, add 250 μl water into wells on the edge to prevent wells from drying.
      • e) Incubate for at least 3 hours at 37° C., 5% CO2 incubator.
      • f) Dump the transfection mixture, replace with 100 μl of complete medium (DMEM, 1×PS, 1×Na Pyruvate, 1×NEAA).
    β-Gal/BCA Assay in 96 Well Format Cell Lysis
      • a) Dump the medium, wash once with 200 μl PBS, blot the plate dry with inversion.
      • b) Add 30 μl lysis buffer from β-Gal Kit into each well.
      • c) Freeze-Thaw the cells twice to generate lysate.
        β-Gal assay
      • a) Prepare assay mix (50 μl 1× buffer, 17 μl ONPG each well)
      • b) Take new plate and add 65 ul assay mix into each well.
      • c) Add 10 μl of cell lysate into each well. There should be blank wells for subtraction of the background activities.
      • d) Incubate at 37° C. for about 20 minutes, prevent long incubation which will use up all ONPG and biased the high expression.
      • e) Add 100 μl of the Stop solution.
      • f) Measure the OD at 420 nm.
    BCA Assay
      • a) Prepare BSA standard (150 ul per well), every points should be duplicated on each plate.
      • b) Put 145 μl of water into each well, add 5 ul of cell lysate into each well.
      • c) Prepare Assay Reagent (A:B:C: 25:24:1), mix right before use.
      • d) Add 150 μl of Assay Reagent into each well.
      • e) Incubate at 37° C. for about 20 minutes.
      • f) Measure the OD at 562 nm.
        Flow Cytometry Measurement of FITC/FAM Conjugated siRNA
      • a) After transfection, incubate cell for at least 3 hours.
      • b) Wash with 200 μl PBS.
      • c) Detach cell with 15 μl TE, incubate at 37° C.
      • d) Re-suspend five wells with 30 μl FACS solution (PBS with 0.5% BSA, and 0.1% sodium Azide)
      • e) Combine all five wells into a tube.
      • f) Add PI 5 μl into each tube.
      • g) Analyze the cells with fluorescence activated cell sorting (FCAS) with BD FACscan instrument according to manufacture's instruction.
    Results
  • Cholesterol Conjugation of siRNA
  • The transfection was performed with either regular siRNA or cholesterol-conjugated siRNA with lipofectamine (Invitrogen) on 9 L/beta-gal cells. The siRNA was designed to specifically knock down beta-galactosidase mRNA and activities are expressed as percentage of beta-gal activities from control (transfected cells by lipofectamine alone).
  • 1. siRNA Sequence and Structure Information of Cholesterol-Conjugated siRNA
  • C.U.A.C.A.C.A.A.A.U.C.A.G.C.G.A.U.U.U.dT.dT (Sense) (SEQ ID NO: 14)
  • A.A.A.U.C.G.C.U.G.A.U.U.U.G.U.G.U.A.G.dT.dT (Antisense) (SEQ ID NO: 15)
  • Designation of Cholesterol Conjugated siRNA
  • A. regular sense or antisense strand
  • B. 5′ end labeled sense strand
  • C. 3′ end labeled sense strand
  • D. both ends labeled sense strand
  • E. 5′ end labeled antisense strand
  • F. 3′ end labeled antisense strand
  • G. both end labeled antisense strand
  • Figure US20080261304A1-20081023-C00005
  • Figure US20080261304A1-20081023-C00006
  • Figure US20080261304A1-20081023-C00007
  • Figure US20080261304A1-20081023-C00008
  • Figure US20080261304A1-20081023-C00009
  • Figure US20080261304A1-20081023-C00010
  • TABLE 1
    Cholesterol siRNA Activities Post-Transfection
    Activity (% of Activity (% of
    Duplexes control Duplexes control
    AA 23.12 AG 29.87
    BE 10.27 BF 32.02
    AF 11.99 DA 33.99
    BA 12.09 BG 46.39
    CA 16.18 DF 65.4
    CF 16.76 DE 77.12
    AE 19.02 CG 77.80
    CE 27.62 DG 98.84
  • Table 1 above provides results of transfection and mRNA silencing experiments using the siRNA constructs made using sense and antisense strands designated above. The transfection and silencing assay results show cholesterol-enhanced delivery of exemplary siRNAs of the invention, and demonstrate silencing of the beta-galactosidase mRNA by the cholesterol-conjugated siRNAs. The “Activity (% of control)” indicates the beta-galactosidase activity remaining after the transfection. The lower the percentage, the greater was the efficacy of the siRNA construct. The double letters represent a double-stranded siRNA. Thus, the exemplary constructs, BE, AF, BA, CA, CF, and AE are representative of the nature and activity of cholesterol conjugated dsRNAs of the present invention. These constructs show greater silencing efficacy than the corresponding unconjugated siRNAs. siRNA constructs CE, AG, BF, DA, BG, DF, DE, CG and DG showed lower efficacy than the unconjugated siRNA construct AA.
  • AA is a siRNA construct with no cholesterol conjugated to any of the ends of the sense or antisense RNA strands. This construct was transfected into the cells resulting in silencing of the beta-galactosidase mRNA so that 23.12% of the activity of the beta-galactosidase mRNA remained.
  • BE is a siRNA construct having a cholesterol moiety linked to the 5′ end of the sense strand and a cholesterol moiety linked to the 5′ end of the antisense strand, and no cholesterol moiety linked to the other ends of the siRNA. This construct was transfected into the cells resulting in silencing the beta-galactosidase mRNA so that only 10.27% of the activity of the beta-galactosidase mRNA remained. This is unexpectedly superior to the unconjugated siRNA.
  • AF is a siRNA construct having a cholesterol moiety linked to the 3′ end of the antisense strand and no cholesterol moiety linked to the other ends of the siRNA strands. This construct was transfected into the cells resulting in silencing the beta-galactosidase mRNA so that only 11.99% of the activity of the beta-galactosidase mRNA remained. This is unexpectedly superior to the unconjugated siRNA.
  • BA is a siRNA construct having a cholesterol moiety linked to the 5′ end of the sense strand and no cholesterol moiety linked to the other ends of the siRNA strands. This construct was transfected into the cells resulting in silencing the beta-galactosidase mRNA so that only 12.09% of the activity of the beta-galactosidase mRNA remained. This is unexpectedly superior to the unconjugated siRNA.
  • CA is a siRNA construct having a cholesterol moiety linked to the 3′ end of the sense strand and no cholesterol moiety linked to the other ends of the siRNA strands. This construct was transfected into the cells resulting in silencing the beta-galactosidase mRNA so that only 16.18% of the activity of the beta-galactosidase mRNA remained. This is unexpectedly superior to the unconjugated siRNA.
  • CF is a siRNA construct having a cholesterol moiety linked to the 3′ end of the sense strand, a cholesterol moiety linked to the 3′ end of the antisense strand, and no cholesterol moiety linked to the other ends of the siRNA strands. This construct was transfected into the cells resulting in silencing the beta-galactosidase mRNA so that only 16.76% of the activity of the beta-galactosidase mRNA remained. This is unexpectedly superior to the unconjugated siRNA.
  • AE is a siRNA construct having a cholesterol moiety linked to the 5′ end of the antisense strand and no cholesterol moiety linked to the other ends of the siRNA strands. This construct was transfected into the cells resulting in silencing the beta-galactosidase mRNA so that only 19.02% of the activity of the beta-galactosidase mRNA remained. This is unexpectedly superior to the unconjugated siRNA.
  • The constructs listed below showed lower ability to silence the beta-galactosidase reporter than was determined for the corresponding, unconjugated siRNA.
  • CE is a siRNA construct having a cholesterol moiety linked to the 3′ end of the sense strand, a cholesterol moiety linked 5′ end of the antisense strand, and no cholesterol moiety linked to the other ends of the siRNA strands. This construct was transfected into the cells resulting in silencing the beta-galactosidase mRNA so that 27.62% of the activity of the beta-galactosidase mRNA remained. This silencing effect was lower than that observed for the corresponding, unconjugated siRNA.
  • AG is a siRNA construct having a cholesterol moiety linked to the 3′ end of the antisense strand, a cholesterol moiety linked to the 5′ end of the antisense strand, and no cholesterol moiety linked to the other ends of the siRNA strands. This construct was transfected into the cells resulting in silencing the beta-galactosidase mRNA so that 29.87% of the activity of the beta-galactosidase mRNA remained. This silencing effect was lower than that observed for the corresponding, unconjugated siRNA.
  • BF is a siRNA construct having a cholesterol moiety linked to the 5′ end of the sense strand, a cholesterol moiety linked to the 3′ end of the antisense strand, and no cholesterol moiety linked to the other ends of the siRNA strands. This construct was transfected into the cells resulting in silencing the beta-galactosidase mRNA so that 32.02% of the activity of the beta-galactosidase mRNA remained. This silencing effect was lower than that observed for the corresponding, unconjugated siRNA.
  • DA is a siRNA construct having a cholesterol moiety linked to 5′ end of the sense strand, a cholesterol moiety linked to the 3′ end of the sense strand, and no cholesterol moiety linked to the other ends of the siRNA strands. This construct was transfected into the cells resulting in silencing the beta-galactosidase mRNA so that 33.99% of the activity of the beta-galactosidase mRNA remained. This silencing effect was lower than that observed for the corresponding, unconjugated siRNA.
  • BG is a siRNA construct having a cholesterol moiety linked to 5′ end of the sense strand, a cholesterol moiety linked to the 3′ end of the antisense strand, a cholesterol moiety linked to the 5′ end of the antisense strand, and no cholesterol moiety linked to the 3′ end of the sense strand. This construct was transfected into the cells resulting in silencing the beta-galactosidase mRNA so that 46.39% of the activity of the beta-galactosidase mRNA remained. This silencing effect was lower than that observed for the corresponding, unconjugated siRNA.
  • DF is a siRNA construct having a cholesterol moiety linked to 5′ end of the sense strand, a cholesterol moiety linked to the 3′ end of the sense strand, a cholesterol moiety linked to the 3′ end of the antisense strand, and no cholesterol moiety linked to the 5′ end of the antisense strand. This construct was transfected into the cells resulting in silencing the beta-galactosidase mRNA so that 65.40% of the activity of the beta-galactosidase mRNA remained. This silencing effect was lower than that observed for the corresponding, unconjugated siRNA.
  • DE is a siRNA construct having a cholesterol moiety linked to 5′ end of the sense strand, a cholesterol moiety linked to the 3′ end of the sense strand, a cholesterol moiety linked to the 5′ end of the antisense strand and no cholesterol moiety linked to the 3′ end of the antisense strand. This construct was transfected into the cells resulting in silencing the beta-galactosidase mRNA so that 77.12% of the activity of the beta-galactosidase mRNA remained. This silencing effect was lower than that observed for the corresponding, unconjugated siRNA.
  • BG is a siRNA construct having a cholesterol moiety linked to 3′ end of the sense strand, a cholesterol moiety linked to the 3′ end of the sense strand, a cholesterol moiety linked to the 3′ end of the antisense strand, a cholesterol moiety linked to the 5′ end of the antisense strand, and no cholesterol moiety linked to the 5′ end of the sense strand. This construct was transfected into the cells resulting in silencing the beta-galactosidase mRNA so that 77.80% of the activity of the beta-galactosidase mRNA remained. This silencing effect was lower than that observed for the corresponding, unconjugated siRNA.
  • DG is a siRNA construct having a cholesterol moiety on the 5′ end of the sense strand, a cholesterol moiety on the 3′ end of the sense strand, a cholesterol moiety on the 3′ end of the antisense strand, and a cholesterol moiety on the 5′ end of the antisense strand. This construct was transfected into the cells resulting in silencing the beta-galactosidase mRNA so that 98.84% of the activity of the beta-galactosidase mRNA remained. This silencing effect was lower than that observed for the corresponding, unconjugated siRNA.
  • Example 3 Serum Inhibition of Cholesterol-Enhanced siRNA Uptake, and Rescue of Cholesterol Enhancement of Uptake by Additional Delivery-Enhancing Agents Human Monocyte Isolation and Purity
  • Fresh human blood samples from healthy donors were purchased from Golden West Biologicals (Temecula, Calif.). For isolation of monocytes, blood samples were diluted with PBS at 1:1 ratio immediately after receiving. Peripheral blood mononuclear cells (PBMC) were first isolated by Ficoll (Amersham, Calif., USA) gradient from whole blood. Then monocytes were further purified from PBMCs using Miltenyi CD14 positive selection kit (MILTENYI BIOTEC GmbH, Germany) by following the manufacturer's instructions. The purity of the monocytes was greater than 95%, judged by flow cytometry stained with anti-CD14 antibody (BD Biosciences, CA). Purified human monocytes were maintained overnight in complete media before induction and knockdown assay.
  • Flow Cytometry
  • Fluorescence activated cell sorting (FACS) analysis were performed using Beckman Coulter FC500 cell analyzer (Fullerton, Calif.). The instrument was adjusted according to the fluorescence probes used (FAM or Cy5 for siRNA and FITC and PE for CD14). Propidium iodide (Fluka, St Lois, Mo.) and AnnexinV (R&D systems, Minneapolis, Minn.) were used as indicators for cell viability and cytotoxicity.
  • For siRNA uptake analysis, cells were washed with PBS, treated with trypsin (attached cells only), and then analyzed by flow cytometry. Uptake of the siRNA designated BA, described above, was also measured by intensity of Cy5 or FAM fluorescence in the cells and cellular viability assessed by addition of propidium iodide or AnnexinV-PE. In order to differentiate the cellular uptake from the membrane insertion of fluorescence labeled siRNA, trypan blue was used to quench the fluorescence on the cell membrane surface.
  • TABLE 2
    Higher MFI with PN73 compared with cholesterol siRNA alone
    Unconjugated siRNA with
    Serum Cholesterol siRNA alone 20 μM PN73
     0 24.8 32.9
     5% 1.55 11.5
    10% 1.34 6.39
    20% 1.19 5.85
  • The data in Table 2 show that the presence of serum significantly reduces cellular uptake of the siRNA conjugated to a cholesterol moiety according to the invention. Serum also inhibits unconjugated siRNA uptake in the presence of an exemplary delivery-enhancing peptide, PN73
  • (KGSKKAVTKAQKKDGKKRKRSRKESYSVYVYKVLKQ-amide; SEQ ID NO: 13),

    but to a lesser extent than the inhibition noted for the cholesterol-conjugated siRNA.
  • FIGS. 1 and 2 illustrate the effects of 5% serum on cellular uptake of a cholesterol-conjugated siRNA according to the invention in complex with a permeabilizing peptide delivery enhancing agent, PN73 (cholesterol siRNA+PN73), and on an unconjugated siRNA in complex with PN73 (siRNA+PN73). For these and related uptake assays, cholesterol-conjugated siRNA and siRNA/PN73 complex were transfected into human monocytes in Opti-MEM® media (Invitrogen) as described above, with serum added in fixed or varied concentration(s). The final concentration of siRNA for both cholesterol and complex were 0.2 μM. The uptake efficiency and Mean fluorescence intensity were assessed by flow cytometry. The cellular uptake values shown in FIGS. 1 and 2 were determined with variation of PN73 concentrations in the presence of a fixed, 5% concentration of serum.
  • FIGS. 3 and 4 illustrate the effects of varying concentrations of serum on cellular uptake of a cholesterol-conjugated siRNA in the presence or absence of a second delivery enhancing agent, lipofectamine, as determined by flow cytometry.
  • The foregoing studies demonstrate that cholesterol-conjugation of siRNAs can significantly enhance their cellular uptake. However, uptake of cholesterol-conjugated siRNAs can be substantially diminished or even eliminated by the presence of serum. This is likely due to binding of the cholesterol moiety with serum proteins—inhibiting the ability of the cholesterol-bound siRNAs to enter target cells. In the presence of a selected delivery enhancing agent, Lipofectamine, this inhibitory effect of serum on cholesterol-siRNA uptake can be effectively diminished. In addition, the presence of a different kind of delivery enhancing agent, exemplified by the permeabilizing peptide PN73, can also mediate rescue of siRNA delivery blocked by serum. More specifically, the addition of a permeabilizing peptide to a delivery formulation comprising a siRNA conjugated to a cholesterol moiety reduces the inhibitory effects of serum on cholesterol-siRNA uptake in a dose dependent manner. This discovery indicates that, although cholesterol conjugation to siRNA alone may not optimize siRNA delivery, additional delivery-enhancing agents including, but not limited to, Lipofectamine and PN73, can further enhance siRNA delivery to mammalian cells and tissues in vitro and in vivo.
  • Although the foregoing invention has been described in detail by way of example for purposes of clarity of understanding, it will be apparent to the artisan that certain changes and modifications may be practiced within the scope of the appended claims which are presented by way of illustration not limitation. In this context, various publications and other references have been cited within the foregoing disclosure for economy of description. Each of these references is incorporated herein by reference in its entirety for all purposes. It is noted, however, that the various publications discussed herein are incorporated solely for their disclosure prior to the filing date of the present application, and the inventors reserve the right to antedate such disclosure by virtue of prior invention.

Claims (13)

1. A double-stranded RNA which is active for RNA interference having a sense and an anti-sense strand, each strand having a 5′ end and a 3′ end, wherein a cholesterol moiety is linked to the 5′ end of the antisense strand.
2. The double-stranded RNA of claim 1 having a length of 19 to 21 base pairs.
3. The double-stranded RNA of claim 1, wherein the cholesterol moiety is selected from the group consisting of cholesterol, sterol, a compound derived from cholesterol, chlolestanol, ergosterol, stimastanol, stigmasterol, methyl-lithocholic acid, cortisol, corticosterone, Δ5-pregnenolone, progesterone, deoxycorticosterone, 17-OH-pregnenolone, 17-OH-progesterone, 11-dioxycortisol, dehydroepiandrosterone, dehydroepiandrosterone sulfate, androstenedione, aldosterone, 18-hydroxycorticosterone, tetrahydrocortisol, tetrahydrocortisone, cortisone, prednisone, 6α-methylpredisone, 9α-fluoro-16α-hydroxyprednisolone, 9α-fluoro-16α-methylprednisolone, 9α-fluorocortisol, testosterone, dihydrotestosterone, androstenediol, androstenedione, androstenedione, 3α,5α-androstanediol, estrone, estradiol, estrogen, spermidine cholesterol carbamate, N4-spermidine cholesteryl carbamate, N4-spermidine cholesteryl carbamate di HCl salt, N4-spermidine-7 dehydro cholesteryl carbamate, N4-spermine cholesteryl carbamate, N,N bis(3-aminopropyl) cholesteryl carbamate, N,N bis(6-aminohexyl) cholesteryl carbamate, N4-spermidine dihydrocholesteryl carbamate, N4-spermidine lithocholic carbamate methyl ester, N1,N8-bis(3-aminopropyl-N4-spermidine cholesteryl carbamate, N(N4-3aminopropylspermidine) cholesteryl carbamate, N,N-bis(4-aminobutyl) cholesteryl carbamate, N4-spermidine cholesteryl urea, N4-spermine cholesteryl urea, N4-spermidine dihydro cholesteryl urea, N4-spermine dihydro cholesteryl urea, N,N-bis(N′-3-aminopropyl-N″4-aminobutyl) cholesteryl carbamate, N4spermidine cholesteryl carboxamide, and N—[N1,N4,N8-tris (3-aminopropyl)spermidine] cholesteryl carbamate, lumisterol, cholic acid, desoxycholic acid, chenodesoxycholic acid, lithocholic acid, and derivatives thereof.
4. An siHybrid having a sense strand and an antisense strand, each strand having a 5′ end and a 3′ end, wherein a cholesterol moiety is linked to the 3′ end of the antisense strand.
5. The siHybrid of claim 4 having a length of 19 to 21 base pairs.
6. The siHybrid of claim 4, wherein the sense strand is a DNA strand.
7. The siHybrid of claim 4, wherein the cholesterol moiety is selected from the group consisting of cholesterol, sterol, a compound derived from cholesterol, chlolestanol, ergosterol, stimastanol, stigmasterol, methyl-lithocholic acid, cortisol, corticosterone, Δ5-pregnenolone, progesterone, deoxycorticosterone, 17-OH-pregnenolone, 17-OH-progesterone, 11-dioxycortisol, dehydroepiandrosterone, dehydroepiandrosterone sulfate, androstenedione, aldosterone, 18-hydroxycorticosterone, tetrahydrocortisol, tetrahydrocortisone, cortisone, prednisone, 6α-methylpredisone, 9α-fluoro-16α-hydroxyprednisolone, 9α-fluoro-16α-methylprednisolone, 9α-fluorocortisol, testosterone, dihydrotestosterone, androstenediol, androstenedione, androstenedione, 3α,5α-androstanediol, estrone, estradiol, estrogen, spermidine cholesterol carbamate, N4-spermidine cholesteryl carbamate, N4-spermidine cholesteryl carbamate di HCl salt, N4-spermidine-7 dehydro cholesteryl carbamate, N4-spermine cholesteryl carbamate, N,N bis(3-aminopropyl) cholesteryl carbamate, N,N bis(6-aminohexyl) cholesteryl carbamate, N4-spermidine dihydrocholesteryl carbamate, N4-spermidine lithocholic carbamate methyl ester, N1,N8-bis(3-aminopropyl-N4-spermidine cholesteryl carbamate, N(N4-3aminopropylspermidine) cholesteryl carbamate, N,N-bis(4-aminobutyl) cholesteryl carbamate, N4-spermidine cholesteryl urea, N4-spermine cholesteryl urea, N4-spermidine dihydro cholesteryl urea, N4-spermine dihydro cholesteryl urea, N,N-bis(N′-3-aminopropyl-N″4-aminobutyl) cholesteryl carbamate, N4spermidine cholesteryl carboxamide, and N—[N1,N4,N8-tris (3-aminopropyl)spermidine] cholesteryl carbamate, lumisterol, cholic acid, desoxycholic acid, chenodesoxycholic acid, lithocholic acid, and derivatives thereof.
8. A composition for delivering a double-stranded RNA to a mammalian cell comprising a double-stranded RNA having a sense and an anti-sense strand, each strand having a 5′ end and a 3′ end, wherein a cholesterol moiety is linked to the 5′ end of the antisense strand, and one or more delivery-enhancing agents selected from:
(a) an aggregation inhibitory agent;
(b) a charge modifying agent;
(c) a pH control agent;
(d) a degradative enzyme inhibitory agent;
(e) a mucolytic or mucus clearing agent;
(f) a ciliostatic agent;
(g) a membrane penetration-enhancing agent selected from (i) a surfactant, (ii) a bile salt, (iii) a phospholipid additive, mixed micelle, liposome, or carrier, (iv) an alcohol, (v) an enamine, (vi) an NO donor compound, (vii) a long-chain amphipathic molecule (viii) a small hydrophobic penetration enhancer; (ix) sodium or a salicylic acid derivative; (x) a glycerol ester of acetoacetic acid (xi) a cyclodextrin or beta-cyclodextrin derivative, (xii) a medium-chain fatty acid, (xiii) a chelating agent, (xiv) an amino acid or salt thereof, (xv) an N-acetylamino acid or salt thereof, (xvi) an enzyme degradative to a selected membrane component, (xvii) an inhibitor of fatty acid synthesis, or (xviii) an inhibitor of cholesterol synthesis; or (xix) any combination of the membrane penetration enhancing agents recited in (i)-(xviii);
(h) a delivery-enhancing peptide;
(i) a vasodilator agent;
(j) a selective transport-enhancing agent; and
(k) a RNA-stabilizing vehicle or carrier.
9. The composition of claim 8, wherein the double-stranded RNA has a length of 19 to 21 base pairs.
10. The composition of claim 8, wherein the cholesterol moiety is selected from the group consisting of cholesterol, sterol, a compound derived from cholesterol, chlolestanol, ergosterol, stimastanol, stigmasterol, methyl-lithocholic acid, cortisol, corticosterone, Δ5-pregnenolone, progesterone, deoxycorticosterone, 17-OH-pregnenolone, 17-OH-progesterone, 11-dioxycortisol, dehydroepiandrosterone, dehydroepiandrosterone sulfate, androstenedione, aldosterone, 18-hydroxycorticosterone, tetrahydrocortisol, tetrahydrocortisone, cortisone, prednisone, 6α-methylpredisone, 9α-fluoro-16α-hydroxyprednisolone, 9α-fluoro-16α-methylprednisolone, 9α-fluorocortisol, testosterone, dihydrotestosterone, androstenediol, androstenedione, androstenedione, 3α,5α-androstanediol, estrone, estradiol, estrogen, spermidine cholesterol carbamate, N4-spermidine cholesteryl carbamate, N4-spermidine cholesteryl carbamate di HCl salt, N4-spermidine-7 dehydro cholesteryl carbamate, N4-spermine cholesteryl carbamate, N,N bis(3-aminopropyl) cholesteryl carbamate, N,N bis(6-aminohexyl) cholesteryl carbamate, N4-spermidine dihydrocholesteryl carbamate, N4-spermidine lithocholic carbamate methyl ester, N1,N8-bis(3-aminopropyl-N4-spermidine cholesteryl carbamate, N(N4-3aminopropylspermidine) cholesteryl carbamate, N,N-bis(4-aminobutyl) cholesteryl carbamate, N4-spermidine cholesteryl urea, N4-spermine cholesteryl urea, N4-spermidine dihydro cholesteryl urea, N4-spermine dihydro cholesteryl urea, N,N-bis(N′-3-aminopropyl-N″4-aminobutyl) cholesteryl carbamate, N4spermidine cholesteryl carboxamide, and N—[N1,N4,N8-tris (3-aminopropyl)spermidine] cholesteryl carbamate, lumisterol, cholic acid, desoxycholic acid, chenodesoxycholic acid, lithocholic acid, and derivatives thereof.
11. The composition of claim 8, wherein the delivery-enhancing agent is a delivery-enhancing peptide having the amino acid sequence selected from the group of:
(SEQ ID NO: 1) RKKRRQRRRPPQCAAVALLPAVLLALLAP; (SEQ ID NO: 2) RQIKIWFQNRRMKWKK; (SEQ ID NO: 3) GWTLNSAGYLLGKINLKALAALAKKIL; (SEQ ID NO: 4) KLALKLALKALKAALKLA; (SEQ ID NO: 7) KLWSAWPSLWSSLWKP; (SEQ ID NO: 8) AAVALLPAVLLALLAPRKKRRQRRRPPQ; (SEQ ID NO: 9) LLETLLKPFQCRICMRNFSTRQARRNHRRRHRR (SEQ ID NO: 10) RRRQRRKRGGDIMGEWGNEIFGAIAGFLG; (SEQ ID NO: 11) KETWWETWWTEWSQPGRKKRRQRRRPPQ; (SEQ ID NO: 12) GLGSLLKKAGKKLKQPKSKRKV; and (SEQ ID NO: 13) KGSKKAVTKAQKKDGKKRKRSRKESYSVYVYKVLKQ.
12. The composition of claim 11, wherein the delivery-enhancing peptide is conjugated to the double-stranded RNA.
13. The composition of claim 8, wherein the mammalian cell is selected from a pulmonary alveolar cell, a skin cell, an hepatic cell, a renal cell, a pancreatic cell, an endothelial cell, a nucleated blood cell, a muscle cell, a mammary cell, a peripheral cell, a central nervous system cell, a gastrointestinal cell, and a tumor cell.
US12/013,274 2004-04-20 2008-01-11 Methods and compositions for enhancing delivery of double-stranded rna or a double-stranded hybrid nucleic acid to regulate gene expression in mammalian cells Abandoned US20080261304A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US12/013,274 US20080261304A1 (en) 2004-04-20 2008-01-11 Methods and compositions for enhancing delivery of double-stranded rna or a double-stranded hybrid nucleic acid to regulate gene expression in mammalian cells
US12/870,989 US8940857B2 (en) 2004-04-20 2010-08-30 Methods and compositions for enhancing delivery of double-stranded RNA or a double-stranded hybrid nucleic acid to regulate gene expression in mammalian cells
US14/566,695 US20150252369A1 (en) 2004-04-20 2014-12-10 Methods and compositions for enhancing delivery of double-stranded rna or a double-stranded hybrid nucleic acid to regulate gene expression in mammalian cells
US14/970,529 US20160206749A1 (en) 2004-04-20 2015-12-15 Methods and compositions for enhancing delivery of double-stranded rna or a double-stranded hybrid nucleic acid to regulate gene expression in mammalian cells

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US56454304P 2004-04-20 2004-04-20
US11/107,371 US20060014289A1 (en) 2004-04-20 2005-04-15 Methods and compositions for enhancing delivery of double-stranded RNA or a double-stranded hybrid nucleic acid to regulate gene expression in mammalian cells
US12/013,274 US20080261304A1 (en) 2004-04-20 2008-01-11 Methods and compositions for enhancing delivery of double-stranded rna or a double-stranded hybrid nucleic acid to regulate gene expression in mammalian cells

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/107,371 Continuation US20060014289A1 (en) 2004-04-20 2005-04-15 Methods and compositions for enhancing delivery of double-stranded RNA or a double-stranded hybrid nucleic acid to regulate gene expression in mammalian cells

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/870,989 Continuation US8940857B2 (en) 2004-04-20 2010-08-30 Methods and compositions for enhancing delivery of double-stranded RNA or a double-stranded hybrid nucleic acid to regulate gene expression in mammalian cells

Publications (1)

Publication Number Publication Date
US20080261304A1 true US20080261304A1 (en) 2008-10-23

Family

ID=35907831

Family Applications (5)

Application Number Title Priority Date Filing Date
US11/107,371 Abandoned US20060014289A1 (en) 2004-04-20 2005-04-15 Methods and compositions for enhancing delivery of double-stranded RNA or a double-stranded hybrid nucleic acid to regulate gene expression in mammalian cells
US12/013,274 Abandoned US20080261304A1 (en) 2004-04-20 2008-01-11 Methods and compositions for enhancing delivery of double-stranded rna or a double-stranded hybrid nucleic acid to regulate gene expression in mammalian cells
US12/870,989 Expired - Fee Related US8940857B2 (en) 2004-04-20 2010-08-30 Methods and compositions for enhancing delivery of double-stranded RNA or a double-stranded hybrid nucleic acid to regulate gene expression in mammalian cells
US14/566,695 Abandoned US20150252369A1 (en) 2004-04-20 2014-12-10 Methods and compositions for enhancing delivery of double-stranded rna or a double-stranded hybrid nucleic acid to regulate gene expression in mammalian cells
US14/970,529 Abandoned US20160206749A1 (en) 2004-04-20 2015-12-15 Methods and compositions for enhancing delivery of double-stranded rna or a double-stranded hybrid nucleic acid to regulate gene expression in mammalian cells

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US11/107,371 Abandoned US20060014289A1 (en) 2004-04-20 2005-04-15 Methods and compositions for enhancing delivery of double-stranded RNA or a double-stranded hybrid nucleic acid to regulate gene expression in mammalian cells

Family Applications After (3)

Application Number Title Priority Date Filing Date
US12/870,989 Expired - Fee Related US8940857B2 (en) 2004-04-20 2010-08-30 Methods and compositions for enhancing delivery of double-stranded RNA or a double-stranded hybrid nucleic acid to regulate gene expression in mammalian cells
US14/566,695 Abandoned US20150252369A1 (en) 2004-04-20 2014-12-10 Methods and compositions for enhancing delivery of double-stranded rna or a double-stranded hybrid nucleic acid to regulate gene expression in mammalian cells
US14/970,529 Abandoned US20160206749A1 (en) 2004-04-20 2015-12-15 Methods and compositions for enhancing delivery of double-stranded rna or a double-stranded hybrid nucleic acid to regulate gene expression in mammalian cells

Country Status (7)

Country Link
US (5) US20060014289A1 (en)
EP (2) EP2145957B1 (en)
JP (1) JP4635046B2 (en)
CA (1) CA2564616C (en)
HK (1) HK1136848A1 (en)
MX (1) MXPA06012076A (en)
WO (1) WO2006019430A2 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070213257A1 (en) * 2005-08-12 2007-09-13 Nastech Pharmaceutical Company Inc. Compositions and methods for complexes of nucleic acids and peptides
WO2010053548A2 (en) * 2008-11-04 2010-05-14 Anchor Therapeutics, Inc. Pthr1 receptor compounds
CN105709229A (en) * 2008-11-10 2016-06-29 阿尔尼拉姆医药品有限公司 Novel lipids and compositions for the delivery of therapeutics

Families Citing this family (72)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MXPA06012076A (en) * 2004-04-20 2007-01-25 Nastech Pharm Co Methods and compositions for enhancing delivery of double-stranded rna or a double-stranded hybrid nucleic acid to regulate gene expression in mammalian cells.
US20060035815A1 (en) * 2004-05-04 2006-02-16 Nastech Pharmaceutical Company Inc. Pharmaceutical compositions for delivery of ribonucleic acid to a cell
US20060040882A1 (en) * 2004-05-04 2006-02-23 Lishan Chen Compostions and methods for enhancing delivery of nucleic acids into cells and for modifying expression of target genes in cells
US20100184209A1 (en) * 2006-02-17 2010-07-22 Dharmacon, Inc. Compositions and methods for inhibiting gene silencing by rna interference
US20070281900A1 (en) * 2006-05-05 2007-12-06 Nastech Pharmaceutical Company Inc. COMPOSITIONS AND METHODS FOR LIPID AND POLYPEPTIDE BASED siRNA INTRACELLULAR DELIVERY
WO2008022046A2 (en) * 2006-08-18 2008-02-21 Nastech Pharmaceutical Company Inc. Dicer substrate rna peptide conjugates and methods for rna therapeutics
PT2062049E (en) 2006-09-06 2014-07-28 Univ California Molecular diagnosis and classification of malignant melanoma
WO2008036841A2 (en) * 2006-09-22 2008-03-27 Dharmacon, Inc. Tripartite oligonucleotide complexes and methods for gene silencing by rna interference
DE102007008596B4 (en) 2007-02-15 2010-09-02 Friedrich-Schiller-Universität Jena Biologically active molecules based on PNA and siRNA, methods for their cell-specific activation and application kit for administration
JP5555626B2 (en) 2007-08-13 2014-07-23 バクスター・インターナショナル・インコーポレイテッド IVIG modulation of chemokines to treat multiple sclerosis, Alzheimer's disease and Parkinson's disease
EP2212693B1 (en) 2007-10-22 2015-04-22 The Regents of The University of California Biomarkers for prenatal diagnosis of congenital cytomegalovirus
US10131904B2 (en) 2008-02-11 2018-11-20 Rxi Pharmaceuticals Corporation Modified RNAi polynucleotides and uses thereof
DE102009043743B4 (en) 2009-03-13 2016-10-13 Friedrich-Schiller-Universität Jena Cell-specific molecules based on siRNA as well as application kits for their production and use
EP2518509B1 (en) 2008-03-05 2014-05-14 The Regents of the University of California Molecular prognosis and classification of malignant melanoma based upon markers selected from the list consisting of RGS1, NCOA3, SPP1, PHIP.
ES2605618T3 (en) * 2008-03-31 2017-03-15 National Institute Of Advanced Industrial Science And Technology Double-stranded, lipid-modified RNA with high RNA interference effect
WO2010008582A2 (en) 2008-07-18 2010-01-21 Rxi Pharmaceuticals Corporation Phagocytic cell drug delivery system
US9433684B2 (en) 2008-08-19 2016-09-06 Nektar Therapeutics Conjugates of small-interfering nucleic acids
EP3336188B1 (en) * 2008-09-22 2020-05-06 Phio Pharmaceuticals Corp. Reduced size self-delivering rnai compounds
NZ609908A (en) 2008-10-27 2014-07-25 Baxter Int Models of thrombotic thrombocytopenic purpura and methods of use thereof
WO2010059226A2 (en) 2008-11-19 2010-05-27 Rxi Pharmaceuticals Corporation Inhibition of map4k4 through rnai
JP5592897B2 (en) * 2008-12-26 2014-09-17 サムヤン バイオファーマシューティカルズ コーポレイション Anionic drug-containing pharmaceutical composition and method for producing the same
US9493774B2 (en) 2009-01-05 2016-11-15 Rxi Pharmaceuticals Corporation Inhibition of PCSK9 through RNAi
US9745574B2 (en) 2009-02-04 2017-08-29 Rxi Pharmaceuticals Corporation RNA duplexes with single stranded phosphorothioate nucleotide regions for additional functionality
JP2012520685A (en) 2009-03-19 2012-09-10 メルク・シャープ・エンド・ドーム・コーポレイション RNA interference-mediated inhibition of GATA binding protein 3 (GATA3) gene expression using small interfering nucleic acids (siNA)
CN102439151A (en) 2009-03-19 2012-05-02 默沙东公司 Rna interference mediated inhibition of btb and cnc homology 1, basic leucine zipper transcription factor 1 (bach 1) gene expression using short interfering nucleic acid (sina) sequence listing
US20120016011A1 (en) 2009-03-19 2012-01-19 Merck Sharp & Dohme Corp. RNA Interference Mediated Inhibition of Connective Tissue Growth Factor (CTGF) Gene Expression Using Short Interfering Nucleic Acid (siNA)
EP2408458A1 (en) 2009-03-19 2012-01-25 Merck Sharp&Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF SIGNAL TRANSDUCER AND ACTIVATOR OF TRANSCRIPTION 6 (STAT6) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
WO2010111468A2 (en) 2009-03-27 2010-09-30 Merck Sharp & Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF THE NERVE GROWTH FACTOR BETA CHAIN (NGFß) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (SINA)
EP2411019A2 (en) 2009-03-27 2012-02-01 Merck Sharp&Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF SIGNAL TRANSDUCER AND ACTIVATOR OF TRANSCRIPTION 1 (STAT1) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
WO2010111497A2 (en) 2009-03-27 2010-09-30 Merck Sharp & Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF THE INTERCELLULAR ADHESION MOLECULE 1 (ICAM-1)GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
WO2010111464A1 (en) 2009-03-27 2010-09-30 Merck Sharp & Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF APOPTOSIS SIGNAL-REGULATING KINASE 1 (ASK1) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
JP2012521764A (en) 2009-03-27 2012-09-20 メルク・シャープ・エンド・ドーム・コーポレイション RNA interference-mediated inhibition of thymic stromal lymphocyte formation factor (TSLP) gene expression using small interfering nucleic acids (siNA)
WO2011035065A1 (en) 2009-09-17 2011-03-24 Nektar Therapeutics Monoconjugated chitosans as delivery agents for small interfering nucleic acids
WO2011094759A2 (en) 2010-02-01 2011-08-04 The Regents Of The University Of California Novel diagnostic and therapeutic targets associated with or regulated by n-cadherin expression and/or epithelial to mesenchymal transition (emt) in prostate cancer and other malignancies
EP2550001B1 (en) 2010-03-24 2019-05-22 Phio Pharmaceuticals Corp. Rna interference in ocular indications
KR20180044433A (en) 2010-03-24 2018-05-02 알엑스아이 파마슈티칼스 코포레이션 Rna interference in dermal and fibrotic indications
US9080171B2 (en) 2010-03-24 2015-07-14 RXi Parmaceuticals Corporation Reduced size self-delivering RNAi compounds
WO2011120023A1 (en) 2010-03-26 2011-09-29 Marina Biotech, Inc. Nucleic acid compounds for inhibiting survivin gene expression uses thereof
WO2011133584A2 (en) 2010-04-19 2011-10-27 Marina Biotech, Inc. Nucleic acid compounds for inhibiting hras gene expression and uses thereof
WO2011139842A2 (en) 2010-04-28 2011-11-10 Marina Biotech, Inc. Nucleic acid compounds for inhibiting fgfr3 gene expression and uses thereof
WO2012018754A2 (en) 2010-08-02 2012-02-09 Merck Sharp & Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF CATENIN (CADHERIN-ASSOCIATED PROTEIN), BETA 1 (CTNNB1) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
US9029341B2 (en) 2010-08-17 2015-05-12 Sirna Therapeutics, Inc. RNA interference mediated inhibition of hepatitis B virus (HBV) gene expression using short interfering nucleic acid (siNA)
WO2012027206A1 (en) 2010-08-24 2012-03-01 Merck Sharp & Dohme Corp. SINGLE-STRANDED RNAi AGENTS CONTAINING AN INTERNAL, NON-NUCLEIC ACID SPACER
EP2609106A4 (en) 2010-08-26 2014-03-19 Merck Sharp & Dohme RNA INTERFERENCE MEDIATED INHIBITION OF PROLYL HYDROXYLASE DOMAIN 2 (PHD2) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
WO2012044979A2 (en) 2010-10-01 2012-04-05 The Goverment Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Manipulation of stem cell function by p53 isoforms
US9260471B2 (en) 2010-10-29 2016-02-16 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using short interfering nucleic acids (siNA)
RU2599449C1 (en) 2011-12-15 2016-10-10 Байонир Корпорейшн New oligonucleotides conjugates and use thereof
CN104114704A (en) * 2012-01-05 2014-10-22 株式会社百奥尼 High-efficiency nanoparticle-type double-helical oligo-RNA structure and method for preparing same
EP3453762B1 (en) 2012-05-02 2021-04-21 Sirna Therapeutics, Inc. Short interfering nucleic acid (sina) compositions
WO2014005596A1 (en) * 2012-07-03 2014-01-09 Aarhus Universitet Modified payload molecules and their interactions and uses
KR20150090917A (en) 2012-12-06 2015-08-06 머크 샤프 앤드 돔 코포레이션 Disulfide masked prodrug compositions and methods
JP6276390B2 (en) * 2013-05-21 2018-02-07 成都先導薬物開発有限公司 Method of cell membrane permeation of compounds
CN104177465B (en) * 2013-05-21 2017-09-29 成都先导药物开发有限公司 A kind of compound administration precursor and drug carriers formulations
RU2744194C2 (en) 2013-12-02 2021-03-03 Фио Фармасьютикалс Корп Cancer immunotherapy
US11279934B2 (en) 2014-04-28 2022-03-22 Phio Pharmaceuticals Corp. Methods for treating cancer using nucleic acids targeting MDM2 or MYCN
JP6836987B2 (en) 2014-09-05 2021-03-03 フィオ ファーマシューティカルズ コーポレーションPhio Pharmaceuticals Corp. Methods for treating aging and skin disorders with nucleic acids targeting TYR or MMP1
EP3277811B1 (en) * 2015-04-03 2020-12-23 University of Massachusetts Fully stabilized asymmetric sirna
PT3277815T (en) 2015-04-03 2021-11-11 Beth Israel Deaconess Medical Ct Inc Oligonucleotide compounds for treatment of preeclampsia and other angiogenic disorders
CA2980337A1 (en) 2015-04-03 2016-10-06 University Of Massachusetts Oligonucleotide compounds for targeting huntingtin mrna
WO2016196366A1 (en) 2015-05-29 2016-12-08 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Extension of replicative lifespan in diseases of premature aging using p53 isoforms
US11001845B2 (en) 2015-07-06 2021-05-11 Phio Pharmaceuticals Corp. Nucleic acid molecules targeting superoxide dismutase 1 (SOD1)
WO2017007825A1 (en) 2015-07-06 2017-01-12 Rxi Pharmaceuticals Corporation Methods for treating neurological disorders using a synergistic small molecule and nucleic acids therapeutic approach
CA2995110A1 (en) 2015-08-14 2017-02-23 University Of Massachusetts Bioactive conjugates for oligonucleotide delivery
EP3365446A4 (en) 2015-10-19 2019-06-26 Phio Pharmaceuticals Corp. Reduced size self-delivering nucleic acid compounds targeting long non-coding rna
US10478503B2 (en) 2016-01-31 2019-11-19 University Of Massachusetts Branched oligonucleotides
WO2018031933A2 (en) 2016-08-12 2018-02-15 University Of Massachusetts Conjugated oligonucleotides
KR101797167B1 (en) * 2016-11-09 2017-11-13 주식회사 바이오셀트란 Novel protein transduction domain and the usage thereof
MX2021001590A (en) 2018-08-10 2021-07-02 Univ Massachusetts Modified oligonucleotides targeting snps.
EP3954395A4 (en) * 2019-04-08 2023-06-28 National University Corporation Tokyo Medical and Dental University Pharmaceutical composition for muscle disease treatment
WO2020222315A1 (en) * 2019-04-29 2020-11-05 주식회사 바이오셀트란 Composition for improving or treating skin wrinkles having excellent skin or cell permeability
EP4359534A1 (en) * 2021-06-21 2024-05-01 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating pompe disease
WO2022271786A1 (en) 2021-06-23 2022-12-29 University Of Massachusetts Optimized anti-flt1 oligonucleotide compounds for treatment of preeclampsia and other angiogenic disorders

Citations (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5334711A (en) * 1991-06-20 1994-08-02 Europaisches Laboratorium Fur Molekularbiologie (Embl) Synthetic catalytic oligonucleotide structures
US5679559A (en) * 1996-07-03 1997-10-21 University Of Utah Research Foundation Cationic polymer and lipoprotein-containing system for gene delivery
US6001311A (en) * 1997-02-05 1999-12-14 Protogene Laboratories, Inc. Apparatus for diverse chemical synthesis using two-dimensional array
US6228642B1 (en) * 1998-10-05 2001-05-08 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of tumor necrosis factor-(α) (TNF-α) expression
US6300074B1 (en) * 1990-06-11 2001-10-09 Gilead Sciences, Inc. Systematic evolution of ligands by exponential enrichment: Chemi-SELEX
US6447796B1 (en) * 1994-05-16 2002-09-10 The United States Of America As Represented By The Secretary Of The Army Sustained release hydrophobic bioactive PLGA microspheres
US20020130430A1 (en) * 2000-12-29 2002-09-19 Castor Trevor Percival Methods for making polymer microspheres/nanospheres and encapsulating therapeutic proteins and other products
US6506559B1 (en) * 1997-12-23 2003-01-14 Carnegie Institute Of Washington Genetic inhibition by double-stranded RNA
US20030060399A1 (en) * 2001-08-23 2003-03-27 Colleen Brophy Reagents and methods for smooth muscle therapies
US20030119725A1 (en) * 2000-07-31 2003-06-26 Gilles Divita Peptide-mediated transfection agents and methods of use
US20030130186A1 (en) * 2001-07-20 2003-07-10 Chandra Vargeese Conjugates and compositions for cellular delivery
US20040019008A1 (en) * 2002-05-28 2004-01-29 Lewis David L. Compositions and processes using siRNA, amphipathic compounds and polycations
US20040110296A1 (en) * 2001-05-18 2004-06-10 Ribozyme Pharmaceuticals, Inc. Conjugates and compositions for cellular delivery
US20040204377A1 (en) * 2002-11-26 2004-10-14 University Of Massachusetts Delivery of siRNAs
US20050064595A1 (en) * 2003-07-16 2005-03-24 Protiva Biotherapeutics, Inc. Lipid encapsulated interfering RNA
US20050147993A1 (en) * 2003-10-24 2005-07-07 Shaharyar Khan Methods and compositions for delivering polynucleotides
US20050153913A1 (en) * 2001-04-10 2005-07-14 Kosak Kenneth M. Nucleic acid carrier compositions and methods for their synthesis
US20060014289A1 (en) * 2004-04-20 2006-01-19 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

Family Cites Families (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX9203290A (en) * 1983-09-19 1992-08-01 Liposome Co Inc LOCALIZED SUPPLY USING FIBRONECTIN CONJUGATES.
US4904582A (en) * 1987-06-11 1990-02-27 Synthetic Genetics Novel amphiphilic nucleic acid conjugates
US6071890A (en) * 1994-12-09 2000-06-06 Genzyme Corporation Organ-specific targeting of cationic amphiphile/DNA complexes for gene therapy
US6331524B1 (en) 1994-12-09 2001-12-18 Genzyme Corporation Organ-specific targeting of cationic amphiphile / DNA complexes for gene therapy
US5744335A (en) * 1995-09-19 1998-04-28 Mirus Corporation Process of transfecting a cell with a polynucleotide mixed with an amphipathic compound and a DNA-binding protein
US6001991A (en) * 1996-10-04 1999-12-14 Isis Pharmaceuticals Inc. Antisense oligonucleotide modulation of MDR P-glycoprotein gene expression
US6080580A (en) * 1998-10-05 2000-06-27 Isis Pharmaceuticals Inc. Antisense oligonucleotide modulation of tumor necrosis factor-α (TNF-α) expression
US20070026394A1 (en) * 2000-02-11 2007-02-01 Lawrence Blatt Modulation of gene expression associated with inflammation proliferation and neurite outgrowth using nucleic acid based technologies
EP1873259B1 (en) * 2000-12-01 2012-01-25 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. RNA interference mediated by 21 and 22nt RNAs
US7101995B2 (en) * 2001-08-27 2006-09-05 Mirus Bio Corporation Compositions and processes using siRNA, amphipathic compounds and polycations
AU2003261231A1 (en) * 2002-07-26 2004-02-16 Chiron Corporation Modified small interfering rna molecules and methods of use
US20040053289A1 (en) * 2002-09-09 2004-03-18 The Regents Of The University Of California Short interfering nucleic acid hybrids and methods thereof
EP2000160A3 (en) * 2002-10-30 2009-03-11 Gambro Lundia AB Method and apparatuses for determining the efficiency of dialysis
US20080188429A1 (en) * 2002-12-27 2008-08-07 Iyer Radhakrishnan P Synthetic siRNA compounds and methods for the downregulation of gene expression
DE10302421A1 (en) * 2003-01-21 2004-07-29 Ribopharma Ag New double-stranded interfering RNA, useful for inhibiting hepatitis C virus, has one strand linked to a lipophilic group to improve activity and eliminate the need for transfection auxiliaries
US20040147027A1 (en) * 2003-01-28 2004-07-29 Troy Carol M. Complex for facilitating delivery of dsRNA into a cell and uses thereof
US20040198640A1 (en) * 2003-04-02 2004-10-07 Dharmacon, Inc. Stabilized polynucleotides for use in RNA interference
DK1620544T3 (en) * 2003-04-17 2019-01-14 Alnylam Pharmaceuticals Inc MODIFIED iRNA AGENTS
ITMI20030860A1 (en) 2003-04-29 2004-10-30 Univ Bologna METHOD FOR SELECTIVE INHIBITION OF THE N-MYC GENE

Patent Citations (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6300074B1 (en) * 1990-06-11 2001-10-09 Gilead Sciences, Inc. Systematic evolution of ligands by exponential enrichment: Chemi-SELEX
US5334711A (en) * 1991-06-20 1994-08-02 Europaisches Laboratorium Fur Molekularbiologie (Embl) Synthetic catalytic oligonucleotide structures
US6447796B1 (en) * 1994-05-16 2002-09-10 The United States Of America As Represented By The Secretary Of The Army Sustained release hydrophobic bioactive PLGA microspheres
US5679559A (en) * 1996-07-03 1997-10-21 University Of Utah Research Foundation Cationic polymer and lipoprotein-containing system for gene delivery
US6001311A (en) * 1997-02-05 1999-12-14 Protogene Laboratories, Inc. Apparatus for diverse chemical synthesis using two-dimensional array
US6506559B1 (en) * 1997-12-23 2003-01-14 Carnegie Institute Of Washington Genetic inhibition by double-stranded RNA
US6228642B1 (en) * 1998-10-05 2001-05-08 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of tumor necrosis factor-(α) (TNF-α) expression
US20030119725A1 (en) * 2000-07-31 2003-06-26 Gilles Divita Peptide-mediated transfection agents and methods of use
US20020130430A1 (en) * 2000-12-29 2002-09-19 Castor Trevor Percival Methods for making polymer microspheres/nanospheres and encapsulating therapeutic proteins and other products
US20050153913A1 (en) * 2001-04-10 2005-07-14 Kosak Kenneth M. Nucleic acid carrier compositions and methods for their synthesis
US20040110296A1 (en) * 2001-05-18 2004-06-10 Ribozyme Pharmaceuticals, Inc. Conjugates and compositions for cellular delivery
US20030130186A1 (en) * 2001-07-20 2003-07-10 Chandra Vargeese Conjugates and compositions for cellular delivery
US20030060399A1 (en) * 2001-08-23 2003-03-27 Colleen Brophy Reagents and methods for smooth muscle therapies
US20040019008A1 (en) * 2002-05-28 2004-01-29 Lewis David L. Compositions and processes using siRNA, amphipathic compounds and polycations
US20040204377A1 (en) * 2002-11-26 2004-10-14 University Of Massachusetts Delivery of siRNAs
US20050064595A1 (en) * 2003-07-16 2005-03-24 Protiva Biotherapeutics, Inc. Lipid encapsulated interfering RNA
US20050147993A1 (en) * 2003-10-24 2005-07-07 Shaharyar Khan Methods and compositions for delivering polynucleotides
US20060014289A1 (en) * 2004-04-20 2006-01-19 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

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070213257A1 (en) * 2005-08-12 2007-09-13 Nastech Pharmaceutical Company Inc. Compositions and methods for complexes of nucleic acids and peptides
WO2010053548A2 (en) * 2008-11-04 2010-05-14 Anchor Therapeutics, Inc. Pthr1 receptor compounds
WO2010053548A3 (en) * 2008-11-04 2010-07-15 Anchor Therapeutics, Inc. Pthr1 receptor compounds
CN105709229A (en) * 2008-11-10 2016-06-29 阿尔尼拉姆医药品有限公司 Novel lipids and compositions for the delivery of therapeutics

Also Published As

Publication number Publication date
US8940857B2 (en) 2015-01-27
WO2006019430A2 (en) 2006-02-23
JP4635046B2 (en) 2011-02-16
CA2564616C (en) 2016-08-30
CA2564616A1 (en) 2006-02-23
EP1773998A2 (en) 2007-04-18
US20150252369A1 (en) 2015-09-10
US20060014289A1 (en) 2006-01-19
WO2006019430B1 (en) 2006-10-26
EP2145957A1 (en) 2010-01-20
WO2006019430A3 (en) 2006-06-01
HK1136848A1 (en) 2010-07-09
EP2145957B1 (en) 2013-12-25
MXPA06012076A (en) 2007-01-25
US20160206749A1 (en) 2016-07-21
US20100316707A1 (en) 2010-12-16
JP2007533323A (en) 2007-11-22

Similar Documents

Publication Publication Date Title
US8940857B2 (en) Methods and compositions for enhancing delivery of double-stranded RNA or a double-stranded hybrid nucleic acid to regulate gene expression in mammalian cells
US10167474B2 (en) Cell-specific internalizing RNA aptamers against human CCR5 and uses therefore
JP5816556B2 (en) UNA oligomer structure for therapeutic agents
US10287582B2 (en) Organic compositions to treat HSF1-related diseases
JP6060178B2 (en) High-efficiency nanoparticle-type double-stranded oligo RNA structure and method for producing the same
AU2014282666A1 (en) Double-stranded antisense nucleic acid with exon-skipping effect
CA2873833A1 (en) Compositions and methods for inhibiting expression of eg5 gene
EP1934359A2 (en) Pharmaceutical compositions for delivery of ribonucleic acid to a cell
KR20220008383A (en) ORGANIC COMPOSITIONS TO TREAT BETA-ENaC-RELATED DISEASES
JP2022517742A (en) Compositions and Methods for Inhibiting HMGB1 Expression
KR20200014320A (en) Nucleic Acids Inhibit Expression of APCS

Legal Events

Date Code Title Description
AS Assignment

Owner name: EOS HOLDINGS LLC, AS AGENT, CALIFORNIA

Free format text: SECURITY AGREEMENT;ASSIGNORS:MDRNA, INC.;MDRNA RESEARCH, INC.;NASTECH PHARMACEUTICAL COMPANY, INC.;REEL/FRAME:023708/0389

Effective date: 20091222

Owner name: EOS HOLDINGS LLC, AS AGENT,CALIFORNIA

Free format text: SECURITY AGREEMENT;ASSIGNORS:MDRNA, INC.;MDRNA RESEARCH, INC.;NASTECH PHARMACEUTICAL COMPANY, INC.;REEL/FRAME:023708/0389

Effective date: 20091222

AS Assignment

Owner name: MDRNA, INC., WASHINGTON

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:AHMADIAN, MOHAMMAD;CUI, KUNYUAN;CHEN, LISHAN;AND OTHERS;REEL/FRAME:023826/0763;SIGNING DATES FROM 20050502 TO 20050505

AS Assignment

Owner name: MDRNA, INC.,WASHINGTON

Free format text: RELEASE OF SECURITY INTEREST;ASSIGNOR:EOS HOLDINGS LLC, AS AGENT;REEL/FRAME:023973/0286

Effective date: 20100217

Owner name: MDRNA RESEARCH, INC.,WASHINGTON

Free format text: RELEASE OF SECURITY INTEREST;ASSIGNOR:EOS HOLDINGS LLC, AS AGENT;REEL/FRAME:023973/0286

Effective date: 20100217

Owner name: NASTECH PHARMACEUTICAL COMPANY, INC.,WASHINGTON

Free format text: RELEASE OF SECURITY INTEREST;ASSIGNOR:EOS HOLDINGS LLC, AS AGENT;REEL/FRAME:023973/0286

Effective date: 20100217

Owner name: MDRNA, INC., WASHINGTON

Free format text: RELEASE OF SECURITY INTEREST;ASSIGNOR:EOS HOLDINGS LLC, AS AGENT;REEL/FRAME:023973/0286

Effective date: 20100217

Owner name: MDRNA RESEARCH, INC., WASHINGTON

Free format text: RELEASE OF SECURITY INTEREST;ASSIGNOR:EOS HOLDINGS LLC, AS AGENT;REEL/FRAME:023973/0286

Effective date: 20100217

Owner name: NASTECH PHARMACEUTICAL COMPANY, INC., WASHINGTON

Free format text: RELEASE OF SECURITY INTEREST;ASSIGNOR:EOS HOLDINGS LLC, AS AGENT;REEL/FRAME:023973/0286

Effective date: 20100217

AS Assignment

Owner name: MDRNA, INC.,WASHINGTON

Free format text: CHANGE OF NAME;ASSIGNOR:NASTECH PHARMACEUTICAL COMPANY, INC.;REEL/FRAME:023993/0447

Effective date: 20080610

Owner name: MDRNA, INC., WASHINGTON

Free format text: CHANGE OF NAME;ASSIGNOR:NASTECH PHARMACEUTICAL COMPANY, INC.;REEL/FRAME:023993/0447

Effective date: 20080610

AS Assignment

Owner name: CEQUENT PHARMACEUTICALS, INC.,MASSACHUSETTS

Free format text: SECURITY AGREEMENT (PATENTS);ASSIGNOR:MDRNA, INC. FKA NASTECH PHARMACEUTICAL COMPANY INC.;REEL/FRAME:024300/0825

Effective date: 20100331

Owner name: CEQUENT PHARMACEUTICALS, INC., MASSACHUSETTS

Free format text: SECURITY AGREEMENT (PATENTS);ASSIGNOR:MDRNA, INC. FKA NASTECH PHARMACEUTICAL COMPANY INC.;REEL/FRAME:024300/0825

Effective date: 20100331

AS Assignment

Owner name: MARINA BIOTECH, INC. (F/K/A MDRNA, INC.), WASHINGT

Free format text: RELEASE BY SECURED PARTY;ASSIGNOR:CEQUENT PHARMACEUTICALS, INC.;REEL/FRAME:024767/0466

Effective date: 20100728

STCB Information on status: application discontinuation

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