WO2009043353A2 - Micromirs - Google Patents

Micromirs Download PDF

Info

Publication number
WO2009043353A2
WO2009043353A2 PCT/DK2008/000344 DK2008000344W WO2009043353A2 WO 2009043353 A2 WO2009043353 A2 WO 2009043353A2 DK 2008000344 W DK2008000344 W DK 2008000344W WO 2009043353 A2 WO2009043353 A2 WO 2009043353A2
Authority
WO
WIPO (PCT)
Prior art keywords
mir
lna
oligomer
cells
nucleotide
Prior art date
Application number
PCT/DK2008/000344
Other languages
French (fr)
Other versions
WO2009043353A3 (en
Inventor
Susanna Obad
Sakari Kauppinen
Joacim Elmen
Morten Lindow
Marcus Heidenblad
Original Assignee
Santaris Pharma A/S
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=40526747&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2009043353(A2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to JP2010527323A priority Critical patent/JP6035010B2/en
Priority to CA2701547A priority patent/CA2701547C/en
Priority to EP18185844.0A priority patent/EP3492594A1/en
Priority to AU2008306327A priority patent/AU2008306327B2/en
Priority to EP12191738.9A priority patent/EP2623598B1/en
Priority to MX2010003299A priority patent/MX2010003299A/en
Priority to UAA201005202A priority patent/UA102529C2/en
Priority to CN200880110388.3A priority patent/CN101821391B/en
Priority to US12/681,587 priority patent/US8906871B2/en
Priority to DK08801378.4T priority patent/DK2205737T3/en
Priority to NZ583677A priority patent/NZ583677A/en
Application filed by Santaris Pharma A/S filed Critical Santaris Pharma A/S
Priority to EP12191739.7A priority patent/EP2623599B1/en
Priority to ES08801378T priority patent/ES2406686T3/en
Priority to EP08801378A priority patent/EP2205737B1/en
Priority to BRPI0817485-7A priority patent/BRPI0817485A2/en
Priority to KR1020107009969A priority patent/KR101889518B1/en
Priority to PL12191738T priority patent/PL2623598T3/en
Priority to EA201070421A priority patent/EA019939B1/en
Publication of WO2009043353A2 publication Critical patent/WO2009043353A2/en
Publication of WO2009043353A3 publication Critical patent/WO2009043353A3/en
Priority to IL204254A priority patent/IL204254A/en
Priority to ZA2010/02040A priority patent/ZA201002040B/en
Priority to US14/527,115 priority patent/US9790493B2/en
Priority to IL244498A priority patent/IL244498A0/en
Priority to US15/705,739 priority patent/US10450564B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • 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
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • 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
    • A61P31/14Antivirals for RNA viruses
    • 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
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/111General methods applicable to biologically active non-coding nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • 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
    • C12N15/1131Non-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 viruses
    • 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
    • C12N2310/11Antisense
    • C12N2310/113Antisense targeting other non-coding nucleic acids, e.g. antagomirs
    • 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
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • C12N2310/141MicroRNAs, miRNAs
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3212'-O-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3222'-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/323Chemical structure of the sugar modified ring structure
    • C12N2310/3231Chemical structure of the sugar modified ring structure having an additional ring, e.g. LNA, ENA
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/33Chemical structure of the base
    • C12N2310/334Modified C
    • C12N2310/33415-Methylcytosine
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/34Spatial arrangement of the modifications
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • 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
    • C12N2310/352Nature of the modification linked to the nucleic acid via a carbon atom
    • C12N2310/3525MOE, methoxyethoxy
    • 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
    • C12N2310/353Nature of the modification linked to the nucleic acid via an atom other than carbon
    • C12N2310/3533Halogen
    • 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
    • C12N2320/31Combination therapy
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to very short oligonucleotides which target and inhibit microRNAs in vivo, and their use in medicaments and pharmaceutical compositions.
  • MicroRNAs are an abundant class of short endogenous RNAs that act as post- transcriptional regulators of gene expression by base-pairing with their target mRNAs. They are processed from longer (ca 70-80 nt) hairpin-like precursors termed pre-miRNAs by the RNAse III enzyme Dicer. MicroRNAs assemble in ribonucleoprotein complexes termed miRNPs and recognize their target sites by antisense complementarity thereby mediating down-regulation of their target genes. Near-perfect or perfect complementarity between the miRNA and its target site results in target mRNA cleavage, whereas limited complementarity between the microRNA and the target site results in translational inhibition of the target gene.
  • WO2007/112754 and WO2007/112753 A summary of the role of_microRNAs in human diseases, and the inhibition of microRNAs using single stranded oligonucleotides is provided by WO2007/112754 and WO2007/112753, which are both hereby incorporated by reference in its entirety.
  • WO2008046911 hereby incorporated by reference, provides microRNA sequences which are associated with cancer. Numerous microRNAs have been associated with disease phenotypes and it is therefore desirable to provide substances capable of modulating the availability of microRNAs in vivo.
  • WO2007/112754 and WO2007/112753 disclose short single stranded oligonucleotides which are considered to form a strong duplex with their target miRNA.
  • SEQ ID NOs 1 - 45 are examples of anti microRNA oligonucleotides as disclosed in WO2007/112754 and WO2007/112753.
  • the present invention is based upon the discovery that the use of very short oligonucleotides which target microRNAs and which have a high proportion of nucleotide analogue nucleotides, such as LNA nucleotides, are highly effective in alleviating the repression of RNAs, such as an mRNA, by the targeted microRNAs in vivo.
  • the present invention provides an oligomer a contiguous sequence of 7, 8, 9 or 10 nucleotide units in length, for use in reducing the effective amount of a microRNA target in a cell or an organism, wherein at least 70%, such as at least 80% of the nucleotide units of the oligomer are selected from the group consisting of LNA units and 2' substituted nucleotide analogues.
  • the present invention provides an oligomer a contiguous sequence of 7, 8, 9 or 10 nucleotide units in length, for use in reducing the effective amount of a microRNA target in a cell or an organism, wherein at least 70% of the nucleotide units of the oligomer are selected from the group consisting of LNA units and 2' substituted nucleotide analogues, and wherein at least 50%, such as at least 60%, such as at least 70% of the nucleotide units of the oligomer are LNA units.
  • the invention provides oligomers of between 7-10 nucleotides in length which comprises a contiguous nucleotide sequence of a total of between 7-10 nucleotides, such as 7, 8, 9, nucleotide units, wherein at least 50% of the nucleotide units of the oligomer are nucleotide analogues.
  • the invention further provides for an oligomer of between 7-10 nucleotides in length which comprises a contiguous nucleotide sequence of a total of between 7-10 nucleotides, such as 7, 8, 9, or 10, nucleotide units, wherein the nucleotide sequence is complementary to a corresponding nucleotide sequence found in mammalian or viral microRNA, and wherein at least 50% of the nucleotide units of the oligomer are nucleotide analogues.
  • the present invention provides olgiomers according to the invention as a medicament.
  • the present invention provides pharmaceutical compositions comprising the oligomer of the invention and a pharmaceutically acceptable diluent, carrier, salt or adjuvant.
  • the invention provides for a conjugate comprising an oligomer according to the invention, conjugated to at least one non-nucleotide or polynucleotide entity, such as a sterol, such as cholesterol.
  • a conjugate comprising an oligomer according to the invention, conjugated to at least one non-nucleotide or polynucleotide entity, such as a sterol, such as cholesterol.
  • the invention provides for the use of an oligomer or a conjugate according to the invention, for the manufacture of a medicament for the treatment of a disease or medical disorder associated with the presence or over-expression of a microRNA, such as one or more of the microRNAs referred to herein.
  • the invention provides for the treatment of a disease or medical disorder associated with the presence or overexpression of the microRNA, comprising the step of administering a composition (such as the pharmaceutical composition) comprising an oligomer or conjugate according to the invention to a patient suffering from or likely to suffer from said disease or medical disorder.
  • a composition such as the pharmaceutical composition
  • the invention provides for a method for reducing the effective amount of a microRNA target in a cell or an organism, comprising administering the oligomer of the invention, or a composition (such as a pharmaceutical composition) comprising the oligomer or conjugate according to the invention to the cell or organism.
  • the invention provides for a method for reducing the effective amount of a microRNA target in a cell or an organism, comprising administering the oligomer or conjugate or pharmaceutical composition according to the invention to the cell or organism.
  • the invention provides for a method for de-repression of a target mRNA (or one ore mor RNAs) in a cell or an organism, comprising administering an oligomer or conjugate according to the invention, or a composition comprising said oligomer or conjugate, to said cell or organism.
  • the invention provides for the use of an oligomer or a conjugate according to the invention, for inhibiting the mircoRNA in a cell which comprises said microRNA, such as a human cell.
  • the use may be in vivo or in vitro.
  • FIG. 1 Schematic presentation of the miR-21, miR-155 and miR-122 8-mer LNA-antimiRs, indicating the targeting positions with the fully LNA-modified and phosphorothiolated LNA- antimiR. Preferred hybridisation positions for 7mer, 8mer, 9mer and 10mer LNA oligonucleotides on the mature microRNA are also indicated.
  • Figure 2. Assessment of miR-21 antagonism by SEQ ID #3205 and SEQ ID #3204 LNA- antimiRs in MCF-7 cells using a luciferase sensor assay.
  • FIG. 3 Assessment of miR-21 antagonism by SEQ ID #3205 and SEQ ID #3204 LNA- antimiRs in HeLa cells using a luciferase sensor assay.
  • FIG. 4 Assessment of miR-155 antagonism by SEQ ID #3206 and SEQ ID #3207 LNA- antimiRs in LPS-treated mouse RAW cells using a luciferase sensor assay.
  • RAW cells were co- transfected with miR-155 and the different LNA-antimiRs at different concentrations. After 24 hours, cells were harvested and luciferase activity measured. Shown are the mean of renilla/firefly, were all have been normalized against 0 nM psiCHECK2.
  • Figure 6. Schematic presentation of the miR-21 luciferase reporter constructs.
  • FIG. 1 Shown is also a schematic presentation of the miR-21 sequence and the design and position of the LNA-antimiRs. LNA nucleotides are indicated by ovals, and DNA residues are indicated by bars.
  • FIG. 10 Length assessment of fully LNA-substituted LNA-antimiRs antagonizing miR-21.
  • Figure 11. Determination of the most optimal position for an 8-mer LNA-antimiR within the miR target recognition sequence.
  • FIG. 12 Shown is also a schematic presentation of the miR-21 sequence and the design and position of the LNA-antimiRs.
  • Figure 12. Validation of interaction of the Pdcd4-3'-UTR and miR-21 by the 8-mer SEQ ID #3205 LNA-antimiR.
  • FIG. 13 Comparison of an 8-mer LNA-antimiR (SEQ ID #3207) with a 15-mer LNA-antimiR (SEQ ID #3206) in antagonizing miR-155 in mouse RAW cells.
  • FIG. 14 Assessment of c/EBPDAssessment of c/EBPer LNA-antimiR (SEQ ID #3207) with a 15-mer LNA-antimiR (SEQ ID #3206) in antagonizing miR-155 in mouse RAW cells.
  • Mouse RAW cells were co-transfected with luciferase reporter plasmids containing a perfect match for miR-155 and the diffter 20 hours, cells were harvested and western blot analysis of protein extracts from RAW cells was performed. The different isoforms of c/EBP ⁇ are indicated, and the ratios calculated on c/EBP ⁇ LIP and beta-tubulin are shown below.
  • Figure 15 Assessment of c/EBPDAssessment of c/EBPer LNA-antimiR (SEQ ID #3207) with a 15-mer LNA-antimiR (SEQ ID #3206) in antagonizing miR-155 in mouse RAW cells.
  • Mouse RAW cells were co-transfected with
  • FIG. 16 Shown is also a schematic presentation of the miR-106b sequence and the design and position of the LNA- antimiRs.
  • Figure 16. Antagonism of miR-19b by a fully LNA-modified 8-mer (SEQ ID #3222) LNA-antimiR and a 15-mer (SEQ ID #3229) mixmer antimiR.
  • HeLa cells were co-transfected with luciferase reporter plasmids containing a perfect match for miR-19a and the two LNA-antimiRs at different concentrations. After 24 hours, cells were harvested and luciferase activity measured.
  • Shown are the mean values of four replicate experiments, where the renilla/firefly ratios have been normalized against 0 nM empty vector without a miR-19a target site ( control). Shown is also a schematic presentation of the miR-19a sequence and the design and position of the LNA- antimiRs.
  • FIG. 17 Schematic presentation showing the mature human miR-221 and miR-222 sequences. Shown in the square is the seed sequence (7-mer) that is conserved in both miRNA sequences.
  • Figure 18. Targeting of a microRNA family using short, fully LNA-substituted LNA-antimiR. PC3 cells were co-transfected with luciferase reporter plasmids for miR-221 and miR-222 separately or together and with the different LNA-antimiRs at varying concentrations.
  • FIG. 19 Shown is also a schematic presentation of the miR-221/222 sequence and the design and position of the LNA-antimiRs.
  • Figure 19 Assessment of p27 protein levels as a functional readout for antagonism of the miR- 221/222 family by the 7-mer SEQ ID #3225 LNA-antimiR.
  • PC3 cells were transfected with the 7- mer LNA-antimiR SEQ ID #3225 targeting both miR-221 and miR-222 at varying concentrations. After 24 hours, cells were harvested and protein levels were measured on a western blot. Shown are the ratios of p27/tubulin.
  • Figure 20 Shown are the ratios of p27/tubulin.
  • FIG. 21 Validation of interaction of the Pdcd4 3 ' UTR and miR-21 by the 8-mer SEQ ID #3205 LNA-antimiR versus the 15-mer (SEQ ID #3204) and an 8-mer with two mismatches (SEQ ID #3218).
  • Huh-7 cells were co-transfected with a luciferase reporter plasmid containing part of the 3 ' UTR of Pdcd4 gene, pre-miR-21 (10 nM) and LNA-antimiRs at different concentrations. After 24 hours, cells were harvested and luciferase activity measured.
  • HeLa cells were transfected with 5 nM LNA-antimiR SEQ ID #3205 (perfect match), or SEQ ID NO:
  • FIG 23 ALT and AST levels in mice treated with SEQ ID #3205 (perfect match) or SEQ ID #3218 (mismatch control). Mice were sacrificed after 14 days and after receiving 25 mg/kg every other day.
  • Figure 24 Assessment of PU.1 protein levels as a functional readout for miR-155 antagonism by short LNA-antimiR (SEQ ID #3207).
  • THP-1 cells were co-transfected with pre- miR-155 (5 nmol) and different LNA oligonucleotides (5 nM) and 100 ng/ml LPS was added. After 24 hours, cells were harvested and western blot analysis of protein extracts from the THP-1 cells was performed. PU.1 and tubulin are indicated. Figure 25. Assessment of p27 protein levels as a functional readout for antagonism of the miR- 221/222 family by the 7-mer SEQ ID #3225 LNA-antimiR.
  • PC3 cells were transfected with the 7-mer LNA-antimiR SEQ ID #3225 targeting both miR-221 and miR-222 and a LNA scrambled control at 5 and 25 nM. After 24 hours, cells were harvested and protein levels were measured on a western blot. Shown are the ratios of p27/tubulin. Figure 26. Knock-down of miR-221 /222 by the 7-mer SEQ ID #3225 (perfect match) LNA- antimiR reduces colony formation in soft agar in PC3 cells.
  • PC3 cells were transfected with 25 nM of the 7-mer LNA-antimiR SEQ ID #3225 targeting both miR-221 and miR-222 or a 7-mer scrambled control ((SEQ ID #3231). After 24 hours, cells were harvested and seeded on soft agar. After 12 days, colonies were counted. One experiment has been done in triplicate.
  • Figure 27 Overview of the human let-7 family, and of tested antagonists,
  • the sequences represent the mature miRNA for each member and the box depicts nucleotides 2-16, the positions typically antagonized by LNA-antimiRs. Columns to the right show the number of nucleotide differences compared to let-7a, within the seed (S: position 2-8), extended seed (ES; position 2-9), and the remaining sequence typically targeted by LNA- antimiRs (NE; position 9-16), respectively. Nucleotides with inverted colors are altered compared to let-7a. (lower) Summary of tested antagonists against the let-7 family, including information on design, length and perfectly complementary targets. All compounds are fully phoshorothiolated.
  • FIG 28 Assessment of let-7 antagonism by six different LNA-antimiRs in Huh-7 cells using a luciferase sensor assay.
  • Huh-7 cells were co-transfected with luciferase sensor plasmids containing a partial HMGA2 3'UTR (with four let-7 binding sites), with or without let-7a precursor (grey and black bars, respectively), and with 6 different LNA-antimiRs at increasing concentrations. After 24 hours, cells were harvested and luciferase activity measured. Shown are the mean of renilla/firefly ratios for duplicate measurements and standard deviations for each assay. Within each LNA- antimiR group all ratios have been normalized to the average of wells containing no let-7a precursor (black bars).
  • FIG 29 Luciferase results from Huh-7 cells transfected with the HMGA2 3'UTR sensor plasmid, LNA-antimiRs SEQ ID #3226 (left) and SEQ ID #3227 (right), and pre-miRs for let-7a (A), let-7d (B), let-7e (C), and let-7i (D). Grey bars indicate the target de-repression after pre-mis inclusion, whereas black control bars represent the equivalent level without pre-miR addition. Each ratio is based on quadruplicate measurements and have been normalized against the average of wells containing no precursor (black bars) within each treatment group.
  • Figure 30 Luciferase results from Huh-7 cells transfected with the HMGA2 3'UTR sensor plasmid, LNA-antimiRs SEQ ID #3226 (left) and SEQ ID #3227 (right), and pre-miRs for let-7a (A), let-7d (B), let-7e (C), and let-7i (D). Grey bars
  • Luciferase results from HeLa cells transfected with the HMGA2 3'UTR sensor plasmid or control vector, and the LNA-antimiR SEQ ID #3227 at various concentrations. Each ratio is based on quadruplicate measurements normalized against untreated (0 nM) empty control vector (psi-CHECK-2; grey bars).
  • FIG 31 Assessment of miR-21 antagonism by 8mer (#3205) in HCT116 cells using a luciferase sensor assay.
  • Figure 32 Silencing of miR-21 by the 8-mer #3205 LNA-antimiR reduces colony formation in soft agar in PC3 cells.
  • FIG. 33 Knock-down of miR-21 by the 8-mer #3205 LNA-antimiR reduces colony formation in soft agar in HepG2 cells.
  • FIG. 34 Wound closure in the invasive human prostate cell line PC3 after treatment with #3205.
  • PC3 cells were transfected at day 3 with LNA-antimiR and control oligonucleotides at 25 nM, #3205 (8mer, perfect match) and #3219 (8mer, mismatch) and the following day a scratch was made. Pictures were taken after 24 hours in order to monitor the migration.
  • B The area in each timepoint has been measured with the software program Image J and normalized against respective 0 h time-point.
  • FIG 35 Length assessment of fully LNA-substituted LNA-antimiRs antagonizing miR-155.
  • Figure 36 Shown is also a schematic presentation of the miR sequence and the design and position of the LNA-antimiRs.
  • Pdcd4 protein levels in kidneys of LNA-antimiR-treated mice (dark grey bars), normalized against the average of equivalent saline controls (light grey bars), using Gapdh as loading control.
  • the molecules designed for diagnostic or reagent use are very different in design than those for in vivo or pharmaceutical use.
  • the terminal nucleotides of the reagent oligos are typically not LNA, but DNA, and the internucleoside linkages are typically other than phosphorothioate, the preferred linkage for use in the oligonucleotides of the present invention.
  • the invention therefore provides for a novel class of oligonucleotides (referred to herein as oligomers) per se.
  • oligomer of the invention refers to certain embodiments of the oligomer of the invention, which may be used in a pharmaceutical composition. Aspects which refer to the oligomer may also refer to the contiguous nucleotide sequence, and vice versa.
  • the oligomer of the invention is a single stranded oligonucleotide which comprises nucleotide analogues, such as LNA, which form part of, or the entire contiguous nucleotide sequence of the oligonucleotide.
  • the nucleotide sequence of the oligomer consists of a contiguous nucleotide sequence.
  • oligonucleotide (or simply "oligo"), which is used interchangeably with the term
  • oligomer refers, in the context of the present invention, to a molecule formed by covalent linkage of two or more nucleotides.
  • oligonucleotide may have, in one embodiment, for example have between 7 - 10 nucleotides, such as in individual embodiments, 7, 8, 9, or 10.
  • nucleotide refers to nucleotides, such as DNA and RNA, and nucleotide analogues. It should be recognised that, in some aspects, the term nucleobase may also be used to refer to a nucleotide which may be either naturally occurring or non-naturally occurring - in this respect the term nucleobase and nucleotide may be used interchangeably herein.
  • the contiguous nucleotide sequence consists of 7 nucleotide analogues. In some embodiments, the contiguous nucleotide sequence consists of 8 nucleotide H analogues. In some embodiments, the contiguous nucleotide sequence consists of 9 nucleotide analogues.
  • At least about 50% of the nucleotides of the oligomer are nucleotide analogues, such as at least about 55%, such as at least about 60%, or at least about 65% or at least about 70%, such as at least about 75%, such as at least about 80%, such as at least about 85%, such as at least about 90%, such as at least about 95% or such as 100%.
  • the oligonucleotide may comprise of a nucleotide sequence which consists of only nucleotide analogues.
  • the oligomer may comprise at least one LNA monomer, such as 2, 3, 4, 5, 6, 7, 8, 9 or 10 LNA monomers.
  • the contiguous nucleotide sequence may consist only of LNA units (including linkage groups, such as phosphorothioate linkages), or may conists of LNA and DNA units, or LNA and other nucleotide analogues.
  • the contiguous nucleotide sequence comprises either one or two DNA nucleotides, the remainder of the nucleotides being nucleotide analogues, such as LNA unit.
  • the contiguous nucleotide sequence consists of 6 nucleotide analogues and a single DNA nucleotide.
  • the contiguous nucleotide consists of 7 nucleotide analogues and a single DNA nucleotide. In some embodiments, the contiguous nucleotide sequence consists of 8 nucleotide analogues and a single DNA nucleotide. In some embodiments, the contiguous nucleotide sequence consists of 9 nucleotide analogues and a single DNA nucleotide. In some embodiments, the contiguous nucleotide sequence consists of 7 nucleotide analogues and two DNA nucleotides. In some embodiments, the contiguous nucleotide sequence consists of 8 nucleotide analogues and two DNA nucleotides.
  • the oligomer may consist of the contiguous nucleotide sequence.
  • all the nucleotide analogues are LNA.
  • all nucleotides of the oligomer are LNA.
  • all nucleotides of the oligomer are LNA and all internucleoside linkage groups are phosphothioate.
  • nucleobases A, C, T and G such as the DNA nucleobases A, C, T and G, the RNA nucleobases A, C, U and G, as well as non- DNA/RNA nucleobases, such as 5-methylcytosine ( Me C), isocytosine, pseudoisocytosine, 5- bromouracil, 5-propynyluracil, 5-propyny-6-fluoroluracil, 5-methylthiazoleuracil, 6-aminopurine, 2-aminopurine, inosine, 2,6-diaminopurine, 7-propyne-7-deazaadenine, 7-propyne-7- deazaguanine and 2-chloro-6-aminopurine, in particular Me C.
  • Me C 5-methylcytosine
  • isocytosine pseudoisocytosine
  • 5- bromouracil 5-propynyluracil
  • 5-propyny-6-fluoroluracil 5-methyl
  • non-DNA/RNA nucleobase will depend on the corresponding (or matching) nucleotide present in the microRNA strand which the oligonucleotide is intended to target.
  • the corresponding nucleotide is G it will normally be necessary to select a non-DNA/RNA nucleobase which is capable of establishing hydrogen bonds to G.
  • a typical example of a preferred non- DNA/RNA nucleobase is Me C.
  • corresponding to and “corresponds to” refer to the comparison between the nucleotide sequence of the oligomer or contiguous nucleotide sequence (a first sequence) and the equivalent contiguous nucleotide sequence of a further sequence selected from either i) a sub-sequence of the reverse complement of the microRNA nucleic acid target (such as a microRNA target selected from SEQ ID 40 - SEQ ID 976, and/or ii) the sequence of nucleotides provided herein such as the group consisting of SEQ ID NO 977 - 1913, or SEQ ID NO 1914- 2850, or SEQ ID NO 2851 - 3787.
  • Nucleotide analogues are compared directly to their equivalent or corresponding nucleotides.
  • a first sequence which corresponds to a further sequence under i) or ii) typically is identical to that sequence over the length of the first sequence (such as the contiguous nucleotide sequence).
  • the length of a nucleotide molecule corresponds to the number of monomer units, i.e. nucleotides, irrespective as to whether those monomer units are nucleotides or nucleotide analogues.
  • monomer and unit are used interchangeably herein.
  • hybridisation means hydrogen bonding, which may be Watson-Crick, Hoogsteen, reversed Hoogsteen hydrogen bonding, etc., between complementary nucleoside or nucleotide bases.
  • the four nucleobases commonly found in DNA are G, A, T and C of which G pairs with C, and A pairs with T.
  • RNA T is replaced with uracil (U), which then pairs with A.
  • the chemical groups in the nucleobases that participate in standard duplex formation constitute the Watson-Crick face.
  • Hoogsteen showed a couple of years later that the purine nucleobases (G and A) in addition to their Watson-Crick face have a Hoogsteen face that can be recognised from the outside of a duplex, and used to bind pyrimidine oligonucleotides via hydrogen bonding, thereby forming a triple helix structure.
  • "complementary” refers to the capacity for precise pairing between two nucleotides sequences with one another.
  • a nucleotide at a certain position of an oligonucleotide is capable of hydrogen bonding with a nucleotide at the corresponding position of a DNA or RNA molecule
  • the oligonucleotide and the DNA or RNA are considered to be complementary to each other at that position.
  • the DNA or RNA strand are considered complementary to each other when a sufficient number of nucleotides in the oligonucleotide can form hydrogen bonds with corresponding nucleotides in the target DNA or RNA to enable the formation of a stable complex.
  • the sequence of an oligonucleotide need not be 100% complementary to its target microRNA.
  • complementary and “specifically hybridisable” thus imply that the oligonucleotide binds sufficiently strong and specific to the target molecule to provide the desired interference with the normal function of the target whilst leaving the function of non-target RNAs unaffected.
  • complementary shall mean 100% complementary or fully complementary.
  • the oligonucleotide of the invention is 100% complementary to a miRNA sequence, such as a human microRNA sequence, or one of the microRNA sequences refered to herein.
  • the oligonucleotide of the invention comprises a contiguous sequence, which is 100% complementary to the seed region of the human microRNA sequence.
  • microRNA or "miRNA”, in the context of the present invention, means an RNA oligonucleotide consisting of between 18 to 25 nucleotides in length.
  • miRNAs are typically regulatory endogenous RNA molecules.
  • target microRNA or “target miRNA” refer to a microRNA with a biological role in human disease, e.g. an upregulated, oncogenic miRNA or a tumor suppressor miRNA in cancer, thereby being a target for therapeutic intervention of the disease in question.
  • target gene refers to regulatory mRNA targets of microRNAs, in which said "target gene” or “target mRNA” is regulated post-transcriptionally by the microRNA based on near-perfect or perfect complementarity between the miRNA and its target site resulting in target mRNA cleavage; or limited complementarity, often conferred to complementarity between the so-called seed sequence (nucleotides 2-7 of the miRNA) and the target site resulting in translational inhibition of the target mRNA.
  • the oligonucleotide is single stranded, this refers to the situation where the oligonucleotide is in the absence of a complementary oligonucleotide - i.e. it is not a double stranded oligonucleotide complex, such as an siRNA.
  • the composition according ot the invention does not comprise a further oligonucleotide which has a region of complementarity with the oligomer of 5 or more, such as 6, 7, 8, 9, or 10 consecutive nucleotides, such as eight or more. Length
  • RNA sequence Ae. perfectly complementary to a corresponding region of a mammalian, human or viral microRNA (miRNA) sequence, preferably a human or viral miRNA sequence.
  • miRNA mammalian, human or viral microRNA
  • the microRNA sequence may suitably be a mature microRNA.
  • the microRNA may be a microRNA precursor.
  • the human microRNA sequence may be selected from SEQ ID No 1 - 558 as disclosed in WO2008/046911 , which are all hereby and specifically incorporated by reference. As described in WO2008/046911, these microRNAs are associated with cancer.
  • the viral microRNA sequence may, in some embodiments, be selected from the group consisting of Herpes simplex virus 1 , Kaposi sarcoma-associated herpesvirus, Epstein Barr virus and Human cytomegalovirus.
  • the contiguous nucleotide sequence is complementary (such as 100% complementary) to a corresponding region of a miRNA sequence selected from the group of miRNAs listed in table 1.
  • Table 1 provides 7mer, 8mer and 9mer oligomers which target human and viral microRNAs published in miRBase (Release 12.0 - http://microma.sanger.ac.uk/sequences/).
  • the oligomers according to the invention may consist of or comprise a contiguous nucleotide sequence which is complementary to a corresponding microRNA sequence selected from the group consisting of miR-1 , miR-10b, miR-17-3p, miR-18, miR-19a, miR-19b, miR-20, miR-21 , miR-34a, miR-93, miR-106a, miR-106b, miR-122, miR-133, miR-134, miR-138, miR-155, miR-192, miR-194, miR-221 , miR-222, miR-375.
  • the miRNA (Le target miRNA) is selected from the group consisting of miR-1 , miR-1 Ob, miR-17-3p, miR-18, miR-19a, miR-19b, miR-20, miR-21 , miR- 34a, miR-93, miR-106a, miR-106b, miR-122, miR-133, miR-134, miR-138, miR-155, miR-192, miR-194, miR-221 , miR-222, and miR-375.
  • the miRNA target is a member of the miR 17 - 92 cluster, such as miR 17, miR 106a, miR 106b, miR 18, miR 19a, miR 19b/1 , miR 19b/2, miR20/93, miR92/1 , miR92/2 and miR25.
  • the contiguous nucleotide sequence is complementary to a corresponding region of a microRNA (miRNA) sequence selected from the group consisting of miR-21, miR-155, miR-221 , mir-222, and mir-122.
  • miRNA microRNA
  • said miRNA is selected from the group consisting of miR-1, miR- IOmiR-29, miR-125b,miR-126, miR-133, miR-141 , miR-143, miR-200b, miR-206, miR-208, miR- 302, miR-372, miR-373, miR-375, and miR-520c/e.
  • the contiguous nucleotide sequence is complementary to a corresponding region of a microRNA (miRNA) sequence present in the miR 17 - 92 cluster, such as a microRNA selected from the group consisting of miR-17-5p, miR-20a/b, miR-93, miR-106a/b, miR-18a/b, miR-19a/b, miR-25, miR-92a, , miR-363.
  • miRNA microRNA
  • the miRNA (Ae target miRNA) is miR-21 , such as hsa-miR-21.
  • the miRNA (Le target miRNA) is miR-122, such as hsa-miR-122.
  • the miRNA (Le target miRNA) is miR-19b, such as hsa-miR-19b.
  • the miRNA (Le target miRNA) is miR-155, such as hsa-miR-155.
  • the miRNA (Le target miRNA) is miR-375, such as hsa-miR-375.
  • the miRNA (Ae target miRNA) is miR-375, such as hsa-miR-106b.
  • the contiguous nucleotide sequence may be complementary to a corresponding region of the microRNA, such as a hsa-miR selected from the group consisting of 19b, 21 , 122, 155 and 375.
  • a hsa-miR selected from the group consisting of 19b, 21 , 122, 155 and 375.
  • oligonucleotides comprising or consisting of nucleotide analogues, such as high affinity nucleotide analogues such as locked nucleic acid (LNA) units, show significant silencing of microRNAs, resulting in reduced microRNA levels. It was found that tight binding of said oligonucleotides to the so- called seed sequence, typically nucleotides 2 to 8 or 2 to 7, counting from the 5' end, of the target microRNAs was important. Nucleotide 1 of the target microRNAs is a non-pairing base and is most likely hidden in a binding pocket in the Ago 2 protein.
  • nucleotide analogues such as high affinity nucleotide analogues such as locked nucleic acid (LNA) units
  • the present inventors consider that by selecting the seed region sequences, particularly with oligonucleotides that comprise LNA, preferably LNA units in the region which is complementary to the seed region, the duplex between miRNA and oligonucleotide is particularly effective in targeting miRNAs, avoiding off target effects, and possibly providing a further feature which prevents RISC directed miRNA function.
  • the inventors have found that microRNA silencing is even more enhanced when LNA- modified single stranded oligonucleotides do not contain a nucleotide at the 3' end corresponding to this non-paired nucleotide 1. It was further found that at least two LNA units in the 3' end of the oligonucleotides according to the present invention made said oligonucleotides highly nuclease resistant.
  • the first or second 3' nucleotide of the oligomer corresponds to the second 5' nucleotide of the microRNA sequence, and may be a nucleotide analogue, such as LNA.
  • nucleotide units 1 to 6 (inclusive) of the oligomer as measured from the 3' end the region of the oligomer are complementary to the microRNA seed region sequence, and may all be nucleotide analogues, such as LNA.
  • nucleotide units 1 to 7 (inclusive) of the oligomer as measured from the 3' end the region of the oligomer are complementary to the microRNA seed region sequence, and may all be nucleotide analogues, such as LNA.
  • nucleotide units 2 to 7 (inclusive) of the oligomer as measured from the 3' end the region of the oligomer are complementary to the microRNA seed region sequence, and may all be nucleotide analogues, such as LNA.
  • the oligomer comprises at least one nucleotide analogue unit, such as at least one LNA unit, in a position which is within the region complementary to the miRNA seed region.
  • the oligomer may, in one embodiment comprise at between one and 6 or between 1 and 7 nucleotide analogue units, such as between 1 and 6 and 1 and 7 LNA units, in a position which is within the region complementary to the miRNA seed region.
  • the contiguous nucleotide sequence consists of or comprises a sequence which is complementary (such as 100% complementary) to the seed sequence of said microRNA.
  • the contiguous nucleotide sequence consists of or comprises a sequence selected from any one of the seedmer sequences listed in table 1.
  • the 3' nucleotide of the seedmer forms the 3' most nucleotide of the contiguous nucleotide sequence, wherein the contiguous nucleotide sequence may, optionally, comprise one or two further nucleotide 5' to the seedmer sequence.
  • the oligomer does not comprise a nucleotide which corresponds to the first nucleotide present in the microRNA sequence counted from the 5' end.
  • the oligonucleotide according to the invention does not comprise a nucleotide at the 3' end that corresponds to the first 5' end nucleotide of the target microRNA.
  • oligonucleotides of 7, 8, 9, 10 nucleotides such as 7, 8 or 9 nucleotides, wherein at least 50%, such as 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or such as 100% of the nucleotide units of the oligomer are (preferably high affinity) nucleotide analogues, such as a Locked Nucleic Acid (LNA) nucleotide unit.
  • LNA Locked Nucleic Acid
  • the oligonucleotide of the invention is 7, 8 or 9 nucleotides long, and comprises a contiguous nucleotide sequence which is complementary to a seed region of a human or viral microRNA, and wherein at least 75 %, such as at least 80 %, such as at least 85%, such as at least 90%, such as at least 95%, such as 100% of the nucleotides are are Locked Nucleic Acid (LNA) nucleotide units.
  • LNA Locked Nucleic Acid
  • the linkage groups are other than phosphodiester linkages, such as are phosphorothioate linkages.
  • all of the nucleotide units of the contiguous nucleotide sequence are LNA nucleotide units.
  • the contiguous nucleotide sequence comprises or consists of 7, 8, 9 or 10, preferably contiguous, LNA nucleotide units.
  • the oligonucleotide of the invention is 7, 8 or 9 nucleotides long, and comprises a contiguous nucleotide sequence which is complementary to a seed region of a human or viral microRNA, and wherein at least 80 % of the nucleotides are LNA, and wherein at least 80%, such as 85%, such as 90%, such as 95%, such as 100% of the intern ucleotide bonds are phosphorothioate bonds. It will be recognised that the contiguous nucleotide sequence of the oligmer (a seedmer) may extend beyond the seed region.
  • the oligonucleotide of the invention is 7 nucleotides long, which are all LNA.
  • the oligonucleotide of the invention is 8 nucleotides long, of which up to 1 nucleotide may be other than LNA. In some embodiments, the oligonucleotide of the invention is 9 nucleotides long, of which up to 1 or 2 nucleotides may be other than LNA. In some embodiments, the oligonucleotide of the invention is 10 nucleotides long, of which 1 , 2 or 3 nucleotides may be other than LNA.
  • the nucleotides 'other than LNA may for example, be DNA, or a 2' substituted nucleotide analogues.
  • High affinity nucleotide analogues are nucleotide analogues which result in oligonucleotides which has a higher thermal duplex stability with a complementary RNA nucleotide than the binding affinity of an equivalent DNA nucleotide. This may be determined by measuring the T m .
  • the nucleotide analogue units present in the contiguous nucleotide sequence are selected, optionally independently, from the group consisting of 2'-O_alkyl-RNA unit, 2'-OMe-RNA unit, 2'-amino-DNA unit, 2'-fluoro-DNA unit, LNA unit, PNA unit, HNA unit, INA unit, and a 2'MOE RNA unit.
  • the nucleotide analogue units present in the contiguous nucleotide sequence are selected, optionally independently, from the group consisting of 2'-O_alkyl-RNA unit, 2'-OMe-RNA unit, 2'-amino-DNA unit, 2'-fluoro-DNA unit, LNA unit, and a 2'MOE RNA unit.
  • 2'fluoro-DNA refers to a DNA analogue with a substitution to fluorine at the 2' position (2'F).
  • 2'fluoro-DNA is a preferred form of 2'fluoro-nucleotide.
  • the oligomer comprises at least 4 nucleotide analogue units, such as at least 5 nucleotide analogue units, such as at least 6 nucleotide analogue units, such as at least 7 nucleotide analogue units, such as at least 8 nucleotide analogue units, such as at least 9 nucleotide analogue units, such as 10, nucleotide analogue units.
  • the oligomer comprises at least 3 LNA units, such as at least 4 LNA units, such as at least 5 LNA units, such as at least 6 LNA units, such as at least 7 LNA units, such as at least 8 LNA units, such as at least 9 LNA units, such as 10 LNA.
  • At least one of the nucleotide analogues, such as LNA units is either cytosine or guanine, such as between 1 - 10 of the of the nucleotide analogues, such as LNA units, is either cytosine or guanine, such as 2, 3, 4, 5, 6, 7, 8, or 9 of the of the nucleotide analogues, such as LNA units, is either cytosine or guanine.
  • At least two of the nucleotide analogues such as LNA units are either cytosine or guanine. In one embodiment at least three of the nucleotide analogues such as LNA units are either cytosine or guanine. In one embodiment at least four of the nucleotide analogues such as LNA units are either cytosine or guanine. In one embodiment at least five of the nucleotide analogues such as LNA units are either cytosine or guanine. In one embodiment at least six of the nucleotide analogues such as LNA units are either cytosine or guanine.
  • At least seven of the nucleotide analogues such as LNA units are either cytosine or guanine. In one embodiment at least eight of the nucleotide analogues such as LNA units are either cytosine or guanine.
  • the nucleotide analogues have a higher thermal duplex stability for a complementary RNA nucleotide than the binding affinity of an equivalent DNA nucleotide to said complementary RNA nucleotide.
  • the nucleotide analogues confer enhanced serum stability to the single stranded oligonucleotide.
  • sequence of nucleotides (bases) shown in the sequence listings may be of LNA such as LNA/PS, LNA or may be oligomers containing alternative backbone chemistry, such as sugar/linkage chemistry, whilst retaining the same base sequence (A, T, C or G).
  • the oligomers Whilst it is envisaged that other nucleotide analogues, such as 2'-MOE RNA or 2'-fluoro nucleotides may be useful in the oligomers according to the invention, it is preferred that the oligomers have a high proportion, such as at least 50%, LNA. nucleotides.
  • the nucleotide analogue may be a DNA analogue such as a DNA analogue where the 2'-H group is substituted with a substitution other than -OH (RNA) e.g. by substitution with -0-CH 3 , - 0-CH 2 -CH 2 -O-CH 3 , -0-CH 2 -CH 2 -CH 2 -NH 2 , -0-CH 2 -CH 2 -CH 2 -OH or -F.
  • the nucleotide analogue may be a RNA analogues such as a RNA analogue which have been modified in its 2'-OH group, e.g.
  • nucleotide analogue is "ENA”.
  • LNA unit LNA monomer
  • LNA residue locked nucleic acid unit
  • locked nucleic acid monomer or locked nucleic acid residue
  • LNA unit refers to a bicyclic nucleoside analogue.
  • LNA units are described in inter alia WO 99/14226, WO 00/56746, WO 00/56748, WO 01/25248, WO 02/28875, WO 03/006475 and WO 03/095467.
  • the LNA unit may also be defined with respect to its chemical formula.
  • an "LNA unit” has the chemical structure shown in Scheme 1 below:
  • X is selected from the group consisting of O, S and NR H , where R H is H or C 1-4 -alkyl; Y is (-CH 2 ) r , where r is an integer of 1-4; and B is a nitrogenous base.
  • r is 1 or 2, in particular 1 , i.e. a preferred LNA unit has the chemical structure shown in Scheme 2 below:
  • the LNA units incorporated in the oligonucleotides of the invention are independently selected from the group consisting of thio-LNA units, amino-LNA units and oxy-LNA units.
  • the thio-LNA unit may have the chemical structure shown in Scheme 3 below:
  • the thio-LNA unit is in its beta-D-form, i.e. having the structure shown in 3A above, likewise, the amino-LNA unit may have the chemical structure shown in Scheme 4 below:
  • the amino-LNA unit is in its beta-D-form, i.e. having the structure shown in 4A above.
  • the oxy-LNA unit may have the chemical structure shown in Scheme 5 below:
  • the oxy-LNA unit is in its beta-D-form, i.e. having the structure shown in 5A above.
  • B is a nitrogenous base which may be of natural or non-natural origin.
  • nitrogenous bases include adenine (A), cytosine (C), 5- methylcytosine ( Me C), isocytosine, pseudoisocytosine, guanine (G), thymine (T), uracil (U), 5- bromouracil, 5-propynyluracil, 5-propyny-6, 5-methylthiazoleuracil, 6-aminopurine, 2- aminopurine, inosine, 2,6-diaminopurine, 7-propyne-7-deazaadenine, 7-propyne-7- deazaguanine and 2-chloro-6-aminopurine.
  • thio-LNA unit refers to an LNA unit in which X in Scheme 1 is S.
  • a thio-LNA unit can be in both the beta-D form and in the alpha-L form.
  • the beta-D form of the thio-LNA unit is preferred.
  • the beta-D-form and alpha-L-form of a thio-LNA unit are shown in Scheme 3 as compounds 3A and 3B, respectively.
  • amino-LNA unit refers to an LNA unit in which X in Scheme 1 is NH or NR H , where R H is hydrogen or C 1-4 -alkyl.
  • An amino-LNA unit can be in both the beta-D form and in the alpha-L form.
  • the beta-D form of the amino-LNA unit is preferred.
  • the beta-D-form and alpha-L-form of an amino-LNA unit are shown in Scheme 4 as compounds 4A and 4B, respectively.
  • oxy-LNA unit refers to an LNA unit in which X in Scheme 1 is O.
  • An Oxy-LNA unit can be in both the beta-D form and in the alpha-L form.
  • the beta-D form of the oxy-LNA unit is preferred.
  • the beta-D form and the alpha-L form of an oxy-LNA unit are shown in Scheme 5 as compounds 5A and 5B, respectively.
  • Ci -6 -alkyl is intended to mean a linear or branched saturated hydrocarbon chain wherein the longest chains has from one to six carbon atoms, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isopentyl, neopentyl and hexyl.
  • a branched hydrocarbon chain is intended to mean a Ci -6 -alkyl substituted at any carbon with a hydrocarbon chain.
  • C 1-4 -alkyl is intended to mean a linear or branched saturated hydrocarbon chain wherein the longest chains has from one to four carbon atoms, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl and tert-butyl.
  • a branched hydrocarbon chain is intended to mean a C 1-4 -alkyl substituted at any carbon with a hydrocarbon chain.
  • C 1-6 -alkoxy is intended to mean C 1-6 -alkyl-oxy, such as methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, sec-butoxy, tert-butoxy, pentoxy, isopentoxy, neopentoxy and hexoxy.
  • C 2 - 6 -alkenyl is intended to mean a linear or branched hydrocarbon group having from two to six carbon atoms and containing one or more double bonds.
  • Illustrative examples of C 2 - 6 -alkenyl groups include allyl, homo-allyl, vinyl, crotyl, butenyl, butadienyl, pentenyl, pentadienyl, hexenyl and hexadienyl.
  • the position of the unsaturation may be at any position along the carbon chain.
  • C 2-6 -alkynyl is intended to mean linear or branched hydrocarbon groups containing from two to six carbon atoms and containing one or more triple bonds.
  • Illustrative examples of C 2-6 -alkynyl groups include acetylene, propynyl, butynyl, pentynyl and hexynyl.
  • the position of unsaturation may be at any position along the carbon chain. More than one bond may be unsaturated such that the "C 2 . 6 -alkynyl" is a di-yne or enedi-yne as is known to the person skilled in the art.
  • corresponding LNA unit is intended to mean that the DNA unit has been replaced by an LNA unit containing the same nitrogenous base as the DNA unit that it has replaced, e.g. the corresponding LNA unit of a DNA unit containing the nitrogenous base A also contains the nitrogenous base A.
  • the corresponding LNA unit may contain the base C or the base Me C, preferably Me C.
  • non-LNA unit refers to a nucleoside different from an LNA-unit, i.e. the term “non-LNA unit” includes a DNA unit as well as an RNA unit.
  • a preferred non-LNA unit is a DNA unit.
  • At least one encompasses an integer larger than or equal to 1 , such as 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20 and so forth.
  • a and “an” as used about a nucleotide, an agent, an LNA unit, etc. is intended to mean one or more.
  • the expression “a component (such as a nucleotide, an agent, an LNA unit, or the like) selected from the group consisting of " is intended to mean that one or more of the cited components may be selected.
  • expressions like “a component selected from the group consisting of A, B and C” is intended to include all combinations of A, B and C 1 i.e. A, B, C, A+B, A+C, B+C and A+B+C.
  • nucleoside linkage group is intended to mean a group capable of covalently coupling together two nucleotides, such as between DNA units, between DNA units and nucleotide analogues, between two non-LNA units, between a non-LNA unit and an LNA unit, and between two LNA units, etc.
  • examples include phosphate, phosphodiester groups and phosphorothioate groups.
  • At least one of, such as all of the internucleoside linkage in the oligomer is phosphodiester.
  • phosphorothioate linkages may be preferred.
  • Typical internucleoside linkage groups in oligonucleotides are phosphate groups, but these may be replaced by internucleoside linkage groups differing from phosphate.
  • the oligonucleotide of the invention is modified in its internucleoside linkage group structure, i.e. the modified oligonucleotide comprises an internucleoside linkage group which differs from phosphate. Accordingly, in a preferred embodiment, the oligonucleotide according to the present invention comprises at least one internucleoside linkage group which differs from phosphate.
  • internucleoside linkage groups which differ from phosphate
  • (-0-P(O) 2 -O-) include -0-P(O 1 S)-O-, -0-P(S) 2 -O-, -S-P(O) 2 -O-, -S-P(O 1 S)-O-, -S-P(S) 2 -O-, -0-P(O) 2 -S-, -0-P(O 1 S)-S-, -S-P(O) 2 -S-, -O-PO(R H )-O-, 0-PO(OCHs)-O-, -O-PO(NR H )-O-, -O- PO(OCH 2 CH 2 S-R)-O-, -O-PO(BH 3 )-O-, -O-PO(NHR H )-O-, -0-P(O) 2 -N R H -, -NR H -P(O) 2 -O-, -NR H -C0-0-, -
  • the internucleoside linkage group is preferably a phosphorothioate group (-0-P(O 1 S)-O- ).
  • all internucleoside linkage groups of the oligonucleotides according to the present invention are phosphorothioate.
  • the internucleoside linkage may be selected form the group consisting of: -0-P(O) 2 -O-,
  • -NR H -C0-NR H -, and/or the internucleoside linkage may be selected form the group consisting of: -O-CO-O-, -O-CO-NR H -, -NR H -CO-CH 2 -, -0-CH 2 -CO-NR 8 -, -O-CH 2 -CH 2 -NR H -, -CO-NR 1"1 -
  • R H is selected from hydrogen and C 1-4 -alkyl.
  • S sulphur
  • the internucleoside linkages may be independently selected, or all be the same, such as phosphorothioate linkages.
  • At least 75%, such as 80% or 85% or 90% or 95% or all of the internucleoside linkages present between the nucleotide units of the contiguous nucleotide sequence are phosphorothioate internucleoside linkages.
  • the contiguous nucleotide sequence is complementary to the corresponding sequence of at least two miRNA sequences such as 2, 3, 4, 5, 6, 7, 8, 9, or 10 miRNA sequence,.
  • the use of a single universal base may allow a single oligomer of the invention to target two independant microRNAs which either one or both have a single mismatch in the region which corresponds to oligomer at the position where the universal nucleotide is positioned.
  • the contiguous nucleotide sequence consists of or comprises a sequence which is complementary to the sequence of at least two miRNA seed region sequences such as 2, 3, 4, 5, 6, 7, 8, 9, or 10 miRNA seed region sequences.
  • the contiguous nucleotide sequence is complementary to the corresponding region of both miR-221 and miR-222.
  • the contiguous nucleotide sequence is complementary to the corresponding region of more than one member of the miR-17-92 cluster - such as two or more or all of miR-17-5p, miR-20a/b, miR-93, miR-106a/b; or two or more or all of miR-25, miR-92a and miR-363.
  • the contiguous nucleotide sequence consists of or comprises a sequence that is complementary to 5'GCTACAT3'. Oligomer Design
  • the first nucleotide of the oligomer according to the invention, counting from the 3 1 end is a nucleotide analogue, such as an LNA unit.
  • the last nucleotide of the oligomer according to the invention, counting from the 3' end is a nucleotide analogue, such as an LNA unit.
  • the second nucleotide of the oligomer according to the invention, counting from the 3' end is a nucleotide analogue, such as an LNA unit.
  • the ninth and/or the tenth nucleotide of the oligomer according to the invention, counting from the 3' end is a nucleotide analogue, such as an LNA unit.
  • the ninth nucleotide of the oligomer according to the invention, counting from the 3' end is a nucleotide analogue, such as an LNA unit.
  • the tenth nucleotide of the oligomer according to the invention, counting from the 3' end is a nucleotide analogue, such as an LNA unit.
  • both the ninth and the tenth nucleotide of the oligomer according to the invention, calculated from the 3' end is a nucleotide analogue, such as an LNA unit.
  • the oligomer according to the invention does not comprise a region of more than 3 consecutive DNA nucleotide units. In one embodiment, the oligomer according to the invention does not comprise a region of more than 2 consecutive DNA nucleotide units. In one embodiment, the oligomer comprises at least a region consisting of at least two consecutive nucleotide analogue units, such as at least two consecutive LNA units. In one embodiment, the oligomer comprises at least a region consisting of at least three consecutive nucleotide analogue units, such as at least three consecutive LNA units. Other Patterns of Nucleotide Analogues such as LNA in the Oligomer
  • oligomers containing at least 6 LNA, such as at least 7 nucleotide units may be preferable
  • the discovery that such short oligomers are highly effective at targeting microRNAs in vivo can be used to prepare shorter oligomers of the invention which comprise other nucleotide analogues, such as high affinity nucleotide analogues.
  • the combination of LNA with other high affinity nucleotide analogues are considered as part of the present invention.
  • nucleotide at positions 1 and/ or 2 may be a nucleotide analogue, such as a high affinity nucleotide analogue, such as LNA, or a nucleotide analogue selected from the group consisting of 2'-O-alkyl-RNA unit, 2'-OMe-RNA unit, 2'-amino-DNA unit, 2'-fluoro-DNA unit, 2'-MOE-RNA unit, LNA unit, PNA unit, HNA unit, INA unit.
  • a nucleotide analogue such as a high affinity nucleotide analogue, such as LNA
  • a nucleotide analogue selected from the group consisting of 2'-O-alkyl-RNA unit, 2'-OMe-RNA unit, 2'-amino-DNA unit, 2'-fluoro-DNA unit, 2'-MOE-RNA unit, LNA unit, PNA unit, HNA unit, INA unit.
  • the two 3' nucleotide may therefore be Xx, xX, XX or xx, wherein:
  • X is LNA and x is DNA or another nucleotide analogue, such as as a 2' substituted nucleotide analogue selected from the group consisting of 2'-O_alkyl-RNA unit, 2'-OMe-RNA unit, 2'-amino-DNA unit, 2'-fluoro-DNA unit, LNA, and a 2'MOE RNA unit.
  • Said non-LNA unit (x) may therefore be 2'MOE RNA or 2'-fluoro-DNA.
  • X is a nucleotide analogue
  • x is DNA.
  • nucleotides designated as X and x may be the same throughout the oligomer. It will be noted that when the oligomer is only 7 nucleotides in length the 8 th nucleotide counting from the 3' end should be discarded.
  • the LNA units in one embodiment, may be replaced with other nucleotide anlogues, such as those referred to herein.
  • X may, therefore be selected from the group consisting of 2'-O-alkyl-RNA unit, 2'-OMe-RNA unit, 2'-amino-DNA unit, 2'-fluoro-DNA unit, 2'-MOE-RNA unit, LNA unit, PNA unit, HNA unit, INA unit, "x" is preferably DNA or RNA, most preferably DNA. However, it is preferred that X is LNA.
  • the oligonucleotides of the invention are modified in positions 3 to 8, counting from the 3' end.
  • the design of this sequence may be defined by the number of non-LNA units present or by the number of LNA units present.
  • at least one, such as one, of the nucleotides in positions three to eight, counting from the 3' end is a non-LNA unit.
  • at least two, such as two, of the nucleotides in positions three to eight, counting from the 3' end are non-LNA units.
  • at least three, such as three, of the nucleotides in positions three to eight, counting from the 3' end are non-LNA units.
  • At least four, such as four, of the nucleotides in positions three to eight, counting from the 3' end, are non- LNA units.
  • at least five, such as five, of the nucleotides in positions three to eight, counting from the 3' end are non-LNA units.
  • all six nucleotides in positions three to eight, counting from the 3' end are non-LNA units.
  • the oligonucleotide according to the present invention comprises at least three LNA units in positions three to eight, counting from the 3' end. In an embodiment thereof, the oligonucleotide according to the present invention comprises three LNA units in positions three to eight, counting from the 3 1 end.
  • substitution pattern for the nucleotides in positions three to eight, counting from the 3' end may be selected from the group consisting of XXXxxx, xXXXxx, xxXXXx, xxxXXX, XXxXxx, XXxxxX, xXXxXx, xXXxxX, xxXXxX, XxxXXx, XxxxXX, XxxxXX, xXxXXx, xXxxXXX, xxXXX, xXXXx, xXxxXXX, xxXXX, xXxXxX and XxXxXx, wherein "X” denotes an LNA unit and "x" denotes a non-LNA unit.
  • the substitution pattern for the nucleotides in positions three to eight, counting from the 3" end is selected from the group consisting of XXxXxx, XXxxXx, XXxxxX, xXXxXx, xXXxxX, xxXXxX, XxxXXx, XxxxXX, xXxXXx, xXxxXX, xxXxXX, xXxXxX and XxXxXx, wherein "X” denotes an LNA unit and "x” denotes a non-LNA unit.
  • the substitution pattern for the nucleotides in positions three to eight, counting from the 3' end is selected from the group consisting of xXXxXx, xXXxxX, xxXXxX, xXxXXx, xXxxXX, xxXxXX and xXxXxX, wherein "X” denotes an LNA unit and "x” denotes a non-LNA unit.
  • the substitution pattern for the nucleotides in positions three to eight, counting from the 3' end is xXxXxX or XxXxXx, wherein "X" denotes an LNA unit and "x” denotes a non-LNA unit.
  • the substitution pattern for the nucleotides in positions three to eight, counting from the 3" end is xXxXxX, wherein "X" denotes an LNA unit and "x" denotes a non-LNA unit.
  • the oligonucleotide according to the present invention comprises at least four LNA units in positions three to eight, counting from the 3' end. In an embodiment thereof, the oligonucleotide according to the present invention comprises four LNA units in positions three to eight, counting from the 3' end.
  • substitution pattern for the nucleotides in positions three to eight, counting from the 3' end may be selected from the group consisting of xxXXXX, xXxXXX, xXXxXX, xXXXxX, xXXXx, XxxXXX, XxXxX 1 XxXXxX, XxXXXx, XXxxXX, XXxXxX, XXxXx, XXxxX, XXXxXx and XXXXxx, wherein "X” denotes an LNA unit and "x" denotes a non-LNA unit.
  • the oligonucleotide according to the present invention comprises at least five LNA units in positions three to eight, counting from the 3 1 end. In an embodiment thereof, the oligonucleotide according to the present invention comprises five LNA units in positions three to eight, counting from the 3' end.
  • the substitution pattern for the nucleotides in positions three to eight, counting from the 3 1 end may be selected from the group consisting of xXXXXX, XxXXXX, XXxXXX, XXXxXX, XXXxX and XXXXx, wherein "X” denotes an LNA unit and "x" denotes a non-LNA unit.
  • the oligonucleotide according to the present invention comprises one or two
  • LNA units in positions three to eight, counting from the 3' end. This is considered advantageous for the stability of the A-helix formed by the oligo:microRNA duplex, a duplex resembling an RNA: RNA duplex in structure.
  • the oligonucleotide according to the present invention comprises at least six LNA units in positions three to eight, counting from the 3' end.
  • the oligonucleotide according to the present invention comprises at from three to six LNA units in positions three to eight, counting from the 3 1 end, and in addition from none to three other high affinity nucleotide analogues in the same region, such that the total amount of high affinity nucleotide analogues (including the LNA units) amount to six in the region from positions three to eight, counting from the 3' end.
  • said non-LNA unit (x) is another nucleotide analogue unit, such as a 2' substituted nucleotide analogue selected from the group consisting of 2'-O_alkyl-RNA unit, 2'-0Me-RNA unit, 2'-amino-DNA unit, 2'-fluoro-DNA unit, LNA, and a 2'MOE RNA unit.
  • Said non-LNA unit (x) may therefore be 2'MOE RNA or 2'-fluoro-DNA.
  • the nucleotide at positions 9 and/ or 10 may be a nucleotide analogue, such as a high affinity nucleotide analogue, such as LNA, or a nucleotide analogue selected from the group consisting of 2'-O-aIkyl-RNA unit, 2'-OMe-RNA unit, 2'-amino-DNA unit, 2'-fluoro-DNA unit, 2'-MOE-RNA unit, LNA unit, PNA unit, HNA unit, INA unit.
  • the two 5' nucleotides may therefore be any nucleotide analogue, such as a high affinity nucleotide analogue, such as LNA, or a nucleotide analogue selected from the group consisting of 2'-O-aIkyl-RNA unit, 2'-OMe-RNA unit, 2'-amino-DNA unit, 2'-fluoro-DNA unit, 2'-MOE-RNA unit, LNA unit, PNA unit, HNA
  • X is LNA and x is DNA or another nucleotide analogue, such as as a 2' substituted nucleotide analogue selected from the group consisting of 2'-O_alkyl-RNA unit, 2'-OMe-RNA unit, 2'-amino-DNA unit, 2'-fluoro-DNA unit, LNA, and a 2'MOE RNA unit.
  • Said non-LNA unit (x) may therefore be 2'MOE RNA or 2'-fluoro-DNA.
  • X is a nucleotide analogue
  • x is DNA.
  • nucleotides designated as X and x may be the same throughout the oligomer.
  • the oligonucleotide according to the present invention contains an LNA unit at the 5' end. In another preferred embodiment, the oligonucleotide according to the present invention contains an LNA unit at the first two positions, counting from the 5' end.
  • the invention further provides for an oligomer as described in the context of the pharmaceutical composition of the invention, or for use in vivo in an organism, such as a medicament, wherein said oligomer (or contiguous nucleotide sequence) comprises either i) at least one phosphorothioate linkage and/or ii) at least one 3' terminal LNA unit, and/or iii) at least one 5' teriminal LNA unit.
  • the oligomer may therefore contain at least one phosphorothioate linkage, such as all linkages being phosphorthioates, and at least one 3' terminal LNA unit, and at least one 5' teriminal LNA unit.
  • the oligonucleotide is fully phosphorothiolated - an exception being for therapeutic oligonucleotides for use in the CNS, such as in the brain or spine where phosphorothioation can be toxic, and due to the absence of nucleases, phosphodiester bonds may be used, even between consecutive DNA units.
  • the second 3' nucleotide, and/or the 9 th and 10 th (from the 3' end), if present, may also be LNA.
  • the oligomer comprises at least five nucleotide analogue units, such as at least five LNA units, in positions which are complementary to the miRNA seed region.
  • the nucleotide sequence of the oligomer which is complementary to the sequence of the microRNA seed region is selected from the group consisting of (X)xXXXXX, (X)XxXXXX, (X)XXxXXX, (X)XXXxXX, (X)XXXxX and (X)XXXXXx, wherein "X” denotes a nucleotide analogue, such as an LNA unit, (X) denotes an optional nucleotide analogue, such as an LNA unit, and "x" denotes a DNA or RNA nucleotide unit.
  • the oligomer comprises six or seven nucleotide analogue units, such as six or seven LNA units, in positions which are complementary to the miRNA seed region.
  • the nucleotide sequence of the oligomer which is complementary to the sequence of the microRNA seed region is selected from the group consisting of XXXXXX, XxXXXX, XXxXXX, XXXxXXX, XXXXxXXX, XXXXxX and XXXXXx, wherein "X” denotes a nucleotide analogue, such as an LNA unit, such as an LNA unit, and "x" denotes a DNA or RNA nucleotide unit.
  • the two nucleotide motif at position 7 to 8, counting from the 3' end of the oligomer is selected from the group consisting of xx, XX, xX and Xx, wherein "X” denotes a nucleotide analogue, such as an LNA unit, such as an LNA unit, and "x" denotes a DNA or RNA nucleotide unit.
  • the two nucleotide motif at position 7 to 8, counting from the 3' end of the oligomer is selected from the group consisting of XX, xX and Xx, wherein "X” denotes a nucleotide analogue, such as an LNA unit, such as an LNA unit, and "x" denotes a DNA or RNA nucleotide unit.
  • the oligomer comprises at 12 nucleotides and wherein the two nucleotide motif at position 11 to 12, counting from the 3' end of the oligomer is selected from the group consisting of xx, XX, xX and Xx, wherein "X” denotes a nucleotide analogue, such as an LNA unit, such as an LNA unit, and "x" denotes a DNA or RNA nucleotide unit.
  • the oligomer comprises 12 nucleotides and wherein the two nucleotide motif at position 11 to 12, counting from the 3' end of the oligomer is selected from the group consisting of XX, xX and Xx, wherein "X” denotes a nucleotide analogue, such as an LNA unit, such as an LNA unit, and "x" denotes a DNA or RNA nucleotide unit, such as a DNA unit.
  • the oligomer comprises a nucleotide analogue unit, such as an LNA unit, at the 5 1 end.
  • a nucleotide analogue unit such as an LNA unit
  • the nucleotide analogue units such as X, are independently selected form the group consisting of: 2'-O-alkyl-RNA unit, 2'-OMe-RNA unit, 2'-amino-DNA unit, 2'- fluoro-DNA unit, 2'-MOE-RNA unit, LNA unit, PNA unit, HNA unit, INA unit.
  • all the nucleotides of the oligomer of the invention are nucleotide analogue units.
  • the nucleotide analogue units such as X, are independently selected form the group consisting of: 2'-OMe-RNA units, 2'-fluoro-DNA units, and LNA units,
  • the oligomer comprises said at least one LNA analogue unit and at least one further nucleotide analogue unit other than LNA.
  • the non-LNA nucleotide analogue unit or units are independently selected from 2'-OMe RNA units and 2'-fluoro DNA units.
  • the oligomer consists of at least one sequence XYX or YXY, wherein X is LNA and Y is either a 2'-OMe RNA unit and 2'-fluoro DNA unit.
  • sequence of nucleotides of the oligomer consists of alternative X and Y units.
  • the oligomer comprises alternating LNA and DNA units (Xx) or (xX). In one embodiment, the oligomer comprises a motif of alternating LNA followed by 2 DNA units (Xxx), xXx or xxX.
  • At least one of the DNA or non-LNA nucleotide analogue units are replaced with a LNA nucleotide in a position selected from the positions identified as LNA nucleotide units in any one of the embodiments referred to above.
  • X donates an LNA unit.
  • Table 1 below provides non-limiting examples of short microRNA sequences that could advantageously be targeted with an oligonucleotide of the present invention.
  • oligonucleotides according to the invention may, in one embodiment, have a sequence of 7, 8, 9 or 10 LNA nucleotides 5' - 3' LLLLLLL(L)(L)(L), or have a sequence of nucleotides selected form the group consisting of, the first 7, 8, 9 or 10 nucleotides of the following motifs:
  • the invention provides for a pharmaceutical composition
  • a pharmaceutical composition comprising the oligomer according to the invention, and a pharmaceutically acceptable diluent, carrier, salt or adjuvant.
  • the invention further provides for the use of an oligonucleotide according to the invention, such as those which may form part of the pharmaceutical composition, for the manufacture of a medicament for the treatment of a disease or medical disorder associated with the presence or over-expression (upregulation) of the microRNA.
  • the invention further provides for a method for the treatment of a disease or medical disorder associated with the presence or over-expression of the microRNA, comprising the step of administering a composition (such as the pharmaceutical composition) according to the invention to a person in need of treatment.
  • a composition such as the pharmaceutical composition
  • the invention further provides for a method for reducing the effective amount of a miRNA in a cell or an organism, comprising administering a composition (such as the pharmaceutical composition) according to the invention or a oligomer according to the invention to the cell or the organism.
  • a composition such as the pharmaceutical composition
  • a oligomer according to the invention to the cell or the organism.
  • Reducing the effective amount in this context refers to the reduction of functional miRNA present in the cell or organism.
  • the preferred oligonucleotides according to the invention may not always significantly reduce the actual amount of miRNA in the cell or organism as they typically form very stable duplexes with their miRNA targets.
  • the reduction of the effective amount of the miRNA in a cell may, in one embodiment, be measured by detecting the level of de-repression of the miRNA's target in the cell.
  • the invention further provides for a method for de-repression of a target mRNA of a miRNA in a cell or an organism, comprising administering a composition (such as the pharmaceutical composition) or a oligomer according to the invention to the cell or the organism.
  • a composition such as the pharmaceutical composition
  • a oligomer according to the invention to the cell or the organism.
  • the invention further provides for the use of a oligomer of between 7 - 10 such as 7, 8, 9, or 10 nucleotides in length, for the manufacture of a medicament for the treatment of a disease or medical disorder associated with the presence or over-expression of the microRNA.
  • the medical condition is hepatitis C (HCV), and the miRNA is miR-122.
  • the pharmaceutical composition according to the invention is for use in the treatment of a medical disorder or disease selected from the group consisting of: hepatitis C virus infection and hypercholesterolemia and related disorders, and cancers.
  • the medical disorder or disease is a CNS disease, such as a CNS disease where one or more microRNAs are known to be indicated.
  • hypercholesterolemia related disorders refers to diseases such as atherosclerosis or hyperlipidemia.
  • related diseases also include different types of HDL/LDL cholesterol imbalance; dyslipidemias, e.g., familial combined hyperlipidemia (FCHL), acquired hyperlipidemia, statin-resistant hypercholesterolemia; coronary artery disease (CAD) coronary heart disease (CHD), atherosclerosis.
  • FCHL familial combined hyperlipidemia
  • CAD coronary artery disease
  • CHD coronary heart disease
  • the pharmaceutical composition according to the invention further comprises a second independent active ingredient that is an inhibitor of the VLDL assembly pathway, such as an ApoB inhibitor, or an MTP inhibitor (such as those disclosed in US 60/977,497, hereby incorporated by reference).
  • a second independent active ingredient that is an inhibitor of the VLDL assembly pathway, such as an ApoB inhibitor, or an MTP inhibitor (such as those disclosed in US 60/977,497, hereby incorporated by reference).
  • the invention further provides for a method for the treatment of a disease or medical disorder associated with the presence or over-expression of the microRNA, comprising the step of administering a composition (such as the pharmaceutical composition) comprising a oligomer of between between 7 - 10 such as 7, 8, 9, or 10 nucleotides in length, to a person in need of treatment.
  • a composition such as the pharmaceutical composition
  • a oligomer of between between 7 - 10 such as 7, 8, 9, or 10 nucleotides in length
  • the invention further provides for a method for reducing the effective amount of a miRNA target (i.e. 'available' miRNA) in a cell or an organism, comprising administering a composition (such as the pharmaceutical composition) comprising a oligomer of between 6 7 - 10 such as 7, 8, 9, or 10 nucleotides in length, to the cell or the organism.
  • a composition such as the pharmaceutical composition
  • oligomer of between 6 7 - 10 such as 7, 8, 9, or 10 nucleotides in length
  • reducing the effective amount of one or more microRNAs in a cell or organism refers to the inhibition of the microRNA function in the call or organism.
  • the cell is preferably amammalain cell or a human cell which expresses the microRNA or microRNAs.
  • the invention further provides for a method for de-repression of a target mRNA of a miRNA in a cell or an organism, comprising a oligomer of 7 - 10 such as 7, 8, 9, or 10 nucleotides in length, or (or a composition comprising said oligonucleotide) to the cell or the organism.
  • a fourth aspect of the invention relates to the use of an oligonucleotide as defined herein for the manufacture of a medicament for the treatment of a disease associated with the expression of microRNAs selected from the group consisting of spinal muscular atrophy, Tourette's syndrome, hepatitis C, fragile X mental retardation, DiGeorge syndrome and cancer, such as in non limiting example, chronic lymphocytic leukemia, breast cancer, lung cancer and colon cancer, in particular cancer.
  • the invention further provides for a method for the synthesis of an oligomer targeted against a human microRNA, such as an oligomer described herein, said method comprising the steps of: a. Optionally selecting a first nucleotide, counting from the 3' end, which is a nucleotide analogue, such as an LNA nucleotide. b. Optionally selecting a second nucleotide, counting from the 3' end, which is a nucleotide analogue, such as an LNA nucleotide. c. Selecting a region of the oligomer which corresponds to the miRNA seed region, wherein said region is as defined herein. d.
  • selecting one or two further 5' terminal of the oligomer is as defined herein; wherein the synthesis is performed by sequential synthesis of the regions defined in steps a - e, wherein said synthesis may be performed in either the 3'-5' ( a to f) or 5' - 3' (e to a)direction, and wherein said oligomer is complementary to a sequence of the miRNA target.
  • the invention further provides for a method for the preparation of an oligomer (such as an oligomer according to the invention), said method comprising the steps of a) comparing the sequences of two or more miRNA sequences to identifiy two or more miRNA sequences which comprise a common contiguous nucleotide sequence of at least 7 nucleotides in length, such as 7, 8, 9 or 10 nucleotides in length (i.e. a sequence found in both non-idnetical miRNAs), b) preparing an oligomer sequence which consists or comprises of a contiguous nucleotide sequence with is complementary to said common contiguous nucleotide sequence, wherein said oligomer is, as according to the oligomer of the invention.
  • the common contiguous nucleotide sequence consists or comprises of the seed region of each of said two or more miRNA sequences (which comprise a common contiguous nucleotide seqeunce of at least 6 nucleotides in length).
  • the seed regions of the two or more miRNAs are identical.
  • the oligomer consists or comprises a seedmer sequence of 7 or 8 nucleotides in length which comprises of a seqeunce which is complementary to said two or more miRNAs. This method may be used in conjunction with step c of the above method.
  • the method for the synthesis of the oligomer according to the invention may be performed using standard solid phase oligonucleotide systhesis. In one embodiment, the method for the synthesis of a oligomer targeted against a human microRNA, is performed in the 3' to 5' direction a - e.
  • a further aspect of the invention is a method to reduce the levels of target microRNA by contacting the target microRNA to an oligonucleotide as defined herein, wherein the oligonucleotide (i) is complementary to the target microRNA sequence (ii) does not contain a nucleotide at the 3' end that corresponds to the first 5' end nucleotide of the target microRNA.
  • Duplex stability and T m are examples of T m
  • the oligomer of the invention is capable of forming a duplex with a complementary single stranded RNA nucleic acid molecule (typically of about the same length of said single stranded oligonucleotide) with phosphodiester internucleoside linkages, wherein the duplex has a T m of between 3O 0 C and and 70 0 C or 80°C, such as between 30 0 C and 60°C ot 70 0 C, or between 30°C and 5O 0 C or 60°C. In one embodiment the T m is at least 4O 0 C.
  • T m may be determined by determining the T m of the oligomer and a complementary RNA target in the following buffer conditions: 10OmM NaCI, 0.1mM EDTA, 1OmM Na-phosphate, pH 7.0 (see examples for a detailed protocol).
  • a high affinity analogue may be defined as an analogue which, when used in the oligomer of the invention, results in an increase in the T m of the oligomer as compared to an identicial oligomer which has contains only DNA bases.
  • said oligomer is conjugated with one or more non-nucleotide (or polynucleotide) compounds.
  • conjugate is intended to indicate a heterogenous molecule formed by the covalent attachment (“conjugation") of the oligomer as described herein to one or more non-nucleotide, or non-polynucleotide moieties.
  • non-nucleotide or non- polynucleotide moieties include macromolecular agents such as proteins, fatty acid chains, sugar residues, glycoproteins, polymers, or combinations thereof.
  • proteins may be antibodies for a target protein.
  • Typical polymers may be polyethylene glycol.
  • the oligomer of the invention may comprise both a polynucleotide region which typically consists of a contiguous sequence of nucleotides, and a further non-nucleotide region.
  • the compound may comprise non-nucleotide components, such as a conjugate component.
  • the oligomeric compound is linked to ligands/conjugates, which may be used, e.g. to increase the cellular uptake of oligomeric compounds.
  • ligands/conjugates which may be used, e.g. to increase the cellular uptake of oligomeric compounds.
  • WO2007/031091 provides suitable ligands and conjugates, which are hereby incorporated by reference.
  • the invention also provides for a conjugate comprising the compound according to the invention as herein described, and at least one non-nucleotide or non-polynucleotide moiety covalently attached to said compound. Therefore, in various embodiments where the compound of the invention consists of a specified nucleic acid or nucleotide sequence, as herein disclosed, the compound may also comprise at least one non-nucleotide or non- polynucleotide moiety (e.g. not comprising one or more nucleotides or nucleotide analogues) covalently attached to said compound. Conjugation (to a conjugate moiety) may enhance the activity, cellular distribution or cellular uptake of the oligomer of the invention.
  • Such moieties include, but are not limited to, antibodies, polypeptides, lipid moieties such as a cholesterol moiety, cholic acid, a thioether, e.g. Hexyl-s-tritylthiol, a thiocholesterol, an aliphatic chain, e.g., dodecandiol or undecyl residues, a phospholipids, e.g., di-hexadecyl-rac-glycerol or triethylammonium 1 ,2-di-o- hexadecyl-rac-glycero-3-h-phosphonate, a polyamine or a polyethylene glycol chain, an adamantane acetic acid, a palmityl moiety, an octadecylamine or hexylamino-carbonyl- oxycholesterol moiety.
  • the oligomers of the invention may also be conjugated to active drug
  • the conjugated moiety is a sterol, such as cholesterol.
  • the conjugated moiety comprises or consists of a positively charged polymer, such as a positively charged peptides of, for example between 1 -50, such as 2 - 20 such as 3 - 10 amino acid residues in length, and/or polyalkylene oxide such as polyethylglycol(PEG) or polypropylene glycol - see WO 2008/034123, hereby incorporated by reference.
  • a positively charged polymer such as a positively charged peptides of, for example between 1 -50, such as 2 - 20 such as 3 - 10 amino acid residues in length
  • polyalkylene oxide such as polyethylglycol(PEG) or polypropylene glycol - see WO 2008/034123, hereby incorporated by reference.
  • the positively charged polymer, such as a polyalkylene oxide may be attached to the oligomer of the invention via a linker such as the releasable inker described in WO 2008/034123.
  • conjugate moieties may be used in the conjugates of the invention:
  • activated oligomers refers to an oligomer of the invention that is covalently linked (i.e., functionalized) to at least one functional moiety that permits covalent linkage of the oligomer to one or more conjugated moieties, i.e., moieties that are not themselves nucleic acids or monomers, to form the conjugates herein described.
  • a functional moiety will comprise a chemical group that is capable of covalently bonding to the oligomer via, e.g., a 3'-hydroxyl group or the exocyclic NH 2 group of the adenine base, a spacer that is preferably hydrophilic and a terminal group that is capable of binding to a conjugated moiety (e.g., an amino, sulfhydryl or hydroxyl group).
  • this terminal group is not protected, e.g., is an NH 2 group.
  • the terminal group is protected, for example, by any suitable protecting group such as those described in "Protective Groups in Organic Synthesis” by Theodora W Greene and Peter G M Wuts, 3rd edition (John Wiley & Sons, 1999).
  • suitable hydroxyl protecting groups include esters such as acetate ester, aralkyl groups such as benzyl, diphenylmethyl, or triphenylmethyl, and tetrahydropyranyl.
  • Suitable amino protecting groups include benzyl, alpha-methylbenzyl, diphenylmethyl, triphenylmethyl, benzyloxycarbonyl, tert-butoxycarbonyl, and acyl groups such as trichloroacetyl or trifluoroacetyl.
  • the functional moiety is self-cleaving. In other embodiments, the functional moiety is biodegradable. See e.g., U.S. Patent No. 7,087,229, which is incorporated by reference herein in its entirety.
  • oligomers of the invention are functionalized at the 5' end in order to allow covalent attachment of the conjugated moiety to the 5' end of the oligomer.
  • oligomers of the invention can be functionalized at the 3' end. In still other embodiments, oligomers of the invention can be functionalized along the backbone or on the 5 heterocyclic base moiety. In yet other embodiments, oligomers of the invention can be functionalized at more than one position independently selected from the 5' end, the 3' end, the backbone and the base.
  • activated oligomers of the invention are synthesized by incorporating during the synthesis one or more monomers that is covalently attached to a 10. functional moiety. In other embodiments, activated oligomers of the invention are synthesized with monomers that have not been functionalized, and the oligomer is functionalized upon completion of synthesis.
  • the oligomers are functionalized with a hindered ester containing an aminoalkyl linker, wherein the alkyl portion has the formula (CH 2 )w, wherein w is an integer ranging from 1 to 10, preferably about 6, wherein the alkyl portion of the 15 alkylamino group can be straight chain or branched chain, and wherein the functional group is attached to the oligomer via an ester group (-O-C(O)-(CH 2 ) W NH).
  • the oligomers are functionalized with a hindered ester containing a (CH 2 ) w -sulfhydryl (SH) linker, wherein w is an integer ranging from 1 to 10, preferably about 6, wherein the alkyl portion of the alkylamino group can be straight chain or branched chain, and 20 wherein the functional group attached to the oligomer via an ester group (-O-C(O)-(CH 2 ) W SH).
  • a hindered ester containing a (CH 2 ) w -sulfhydryl (SH) linker wherein w is an integer ranging from 1 to 10, preferably about 6, wherein the alkyl portion of the alkylamino group can be straight chain or branched chain, and 20 wherein the functional group attached to the oligomer via an ester group (-O-C(O)-(CH 2 ) W SH).
  • sulfhydryl-activated oligonucleotides are conjugated with polymer moieties such as polyethylene glycol or peptides (via formation of a disulfide bond).
  • Activated oligomers containing hindered esters as described above can be synthesized by any method known in the art, and in particular by methods disclosed in PCT Publication No. WO 5 2008/034122 and the examples therein, which is incorporated herein by reference in its entirety.
  • the oligomers of the invention are functionalized by introducing sulfhydryl, amino or hydroxyl groups into the oligomer by means of a functionalizing reagent substantially as described in U.S. Patent Nos. 4,962,029 and 4,914,210, i.e., a substantially linear reagent having a phosphoramidite at one end linked through a hydrophilic spacer chain to 0 the opposing end which comprises a protected or unprotected sulfhydryl, amino or hydroxyl group.
  • a functionalizing reagent substantially as described in U.S. Patent Nos. 4,962,029 and 4,914,210, i.e., a substantially linear reagent having a phosphoramidite at one end linked through a hydrophilic spacer chain to 0 the opposing end which comprises a protected or unprotected sulfhydryl, amino or hydroxyl group.
  • such activated oligomers have a functionalizing reagent coupled to a 5'-hydroxyl group of the oligomer. In other embodiments, the activated oligomers have a functionalizing reagent coupled to a 3"-hydroxyl group. In still other embodiments, the activated oligomers of 5 the invention have a functionalizing reagent coupled to a hydroxyl group on the backbone of the oligomer. In yet further embodiments, the oligomer of the invention is functionalized with more than one of the functionalizing reagents as described in U.S. Patent Nos. 4,962,029 and 4,914,210, incorporated herein by reference in their entirety. Methods of synthesizing such functionalizing reagents and incorporating them into monomers or oligomers are disclosed in U.S. Patent Nos. 4,962,029 and 4,914,210.
  • the 5'-terminus of a solid-phase bound oligomer is functionalized with a dienyl phosphoramidite derivative, followed by conjugation of the deprotected oligomer with, e.g., an amino acid or peptide via a Diels-Alder cycloaddition reaction.
  • the incorporation of monomers containing 2'-sugar modifications, such as a 2'-carbamate substituted sugar or a 2'-(O-pentyl-N-phthalimido)- deoxyribose sugar into the oligomer facilitates covalent attachment of conjugated moieties to the sugars of the oligomer.
  • an oligomer with an amino-containing linker at the 2'-position of one or more monomers is prepared using a reagent such as, for example, 5'-dimethoxytrityl-2'-0-(e-phthalimidylaminopentyl)-2 1 -deoxyadenosine-3'- N,N-diisopropyl- cyanoethoxy phosphoramidite. See, e.g., Manoharan, et al., Tetrahedron Letters, 1991 , 34, 7171.
  • the oligomers of the invention may have amine-containing functional moieties on the nucleotide, including on the N6 purine amino groups, on the exocyclic N2 of guanine, or on the N4 or 5 positions of cytosine.
  • such functionalization may be achieved by using a commercial reagent that is already functionalized in the oligomer synthesis.
  • Some functional moieties are commercially available, for example, heterobifunctional and homobifunctional linking moieties are available from the Pierce Co. (Rockford, III.).
  • Other commercially available linking groups are ⁇ '-Amino-Modifier C6 and 3'-Amino-Modifier reagents, both available from Glen Research Corporation (Sterling, Va.).
  • ⁇ '-Amino-Modifier C6 is also available from ABI (Applied Biosystems Inc., Foster City, Calif.) as Aminolink-2, and 3'-Amino- Modifier is also available from Clontech Laboratories Inc. (Palo Alto, Calif.).
  • the oligonucleotides of the invention will constitute suitable drugs with improved properties.
  • the design of a potent and safe drug requires the fine-tuning of various parameters such as affinity/specificity, stability in biological fluids, cellular uptake, mode of action, pharmacokinetic properties and toxicity.
  • the present invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising an oligonucleotide according to the invention and a pharmaceutically acceptable diluent, carrier or adjuvant.
  • a pharmaceutically acceptable diluent, carrier or adjuvant is saline or buffered saline.
  • the present invention relates to an oligonucleotide according to the present invention for use as a medicament.
  • dosing is dependent on severity and responsiveness of the disease state to be treated, and the course of treatment lasting from several days to several months, or until a cure is effected or a diminution of the disease state is achieved.
  • Optimal dosing schedules can be calculated from measurements of drug accumulation in the body of the patient.
  • Optimum dosages may vary depending on the relative potency of individual oligonucleotides. Generally it can be estimated based on EC50s found to be effective in in vitro and in vivo animal models.
  • dosage is from 0.01 ⁇ g to 1 g per kg of body weight, and may be given once or more daily, weekly, monthly or yearly, or even once every 2 to 10 years or by continuous infusion for hours up to several months.
  • the repetition rates for dosing can be estimated based on measured residence times and concentrations of the drug in bodily fluids or tissues.
  • the invention also relates to a pharmaceutical composition, which comprises at least one oligonucleotide of the invention as an active ingredient.
  • the pharmaceutical composition according to the invention optionally comprises a pharmaceutical carrier, and that the pharmaceutical composition optionally comprises further compounds, such as chemotherapeutic compounds, anti-inflammatory compounds, antiviral compounds and/or immuno-modulating compounds.
  • oligonucleotides of the invention can be used "as is” or in form of a variety of pharmaceutically acceptable salts.
  • pharmaceutically acceptable salts refers to salts that retain the desired biological activity of the herein-identified oligonucleotides and exhibit minimal undesired toxicological effects.
  • Non-limiting examples of such salts can be formed with organic amino acid and base addition salts formed with metal cations such as zinc, calcium, bismuth, barium, magnesium, aluminum, copper, cobalt, nickel, cadmium, sodium, potassium, and the like, or with a cation formed from ammonia, ⁇ /, ⁇ /-dibenzylethylene-diamine, D-glucosamine, tetraethylammonium, or ethylenediamine.
  • metal cations such as zinc, calcium, bismuth, barium, magnesium, aluminum, copper, cobalt, nickel, cadmium, sodium, potassium, and the like, or with a cation formed from ammonia, ⁇ /, ⁇ /-dibenzylethylene-diamine, D-glucosamine, tetraethylammonium, or ethylenediamine.
  • the oligonucleotide may be in the form of a prodrug.
  • Oligonucleotides are by virtue negatively charged ions. Due to the lipophilic nature of cell membranes the cellular uptake of oligonucleotides are reduced compared to neutral or lipophilic equivalents. This polarity "hindrance” can be avoided by using the pro-drug approach (see e.g. Crooke, R. M. (1998) in Crooke, S. T. Antisense research and Application. Springer-Verlag, Berlin, Germany, vol. 131 , pp. 103-140).
  • Pharmaceutically acceptable binding agents and adjuvants may comprise part of the formulated drug.
  • Examples of delivery methods for delivery of the therapeutic agents described herein, as well as details of pharmaceutical formulations, salts, may are well described elsewhere for example in US provisional application 60/838,710 and 60/788,995, which are hereby incorporated by reference, and Danish applications, PA 2006 00615 which is also hereby incorporated by reference.
  • compositions of the present invention include, but are not limited to, solutions, emulsions, and liposome-containing formulations. These compositions may be generated from a variety of components that include, but are not limited to, preformed liquids, self- emulsifying solids and self-emulsifying semisolids. Delivery of drug to tumour tissue may be enhanced by carrier-mediated delivery including, but not limited to, cationic liposomes, cyclodextrins, porphyrin derivatives, branched chain dendrimers, polyethylenimine polymers, nanoparticles and microspheres (Dass CR. J Pharm Pharmacol 2002; 54(1 ):3-27).
  • compositions of the present invention may be prepared according to conventional techniques well known in the pharmaceutical industry. Such techniques include the step of bringing into association the active ingredients with the pharmaceutical carrier(s) or excipient(s). In general the formulations are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product.
  • the compositions of the present invention may be formulated into any of many possible dosage forms such as, but not limited to, tablets, capsules, gel capsules, liquid syrups, soft gels and suppositories.
  • the compositions of the present invention may also be formulated as suspensions in aqueous, non-aqueous or mixed media.
  • Aqueous suspensions may further contain substances which increase the viscosity of the suspension including, for example, sodium carboxymethylcellulose, sorbitol and/or dextran.
  • the suspension may also contain stabilizers.
  • the compounds of the invention may also be conjugated to active drug substances, for example, aspirin, ibuprofen, a sulfa drug, an antidiabetic, an antibacterial or an antibiotic.
  • compositions of the invention may contain one or more oligonucleotide compounds, targeted to a first microRNA and one or more additional oligonucleotide compounds targeted to a second microRNA target. Two or more combined compounds may be used together or sequentially.
  • therapeutic methods of the invention include administration of a therapeutically effective amount of an oligonucleotide to a mammal, particularly a human.
  • the present invention provides pharmaceutical compositions containing (a) one or more compounds of the invention, and (b) one or more chemotherapeutic agents.
  • chemotherapeutic agents When used with the compounds of the invention, such chemotherapeutic agents may be used individually, sequentially, or in combination with one or more other such chemotherapeutic agents or in combination with radiotherapy. All chemotherapeutic agents known to a person skilled in the art are here incorporated as combination treatments with compound according to the invention.
  • anti-inflammatory drugs including but not limited to nonsteroidal anti-inflammatory drugs and corticosteroids, antiviral drugs, and immuno- modulating drugs may also be combined in compositions of the invention. Two or more combined compounds may be used together or sequentially.
  • TPM1 Tumor suppressor gene tropomysin 1
  • mtpn Myotrophin
  • the present invention relates to the use of an oligonucleotide according to the invention for the manufacture of a medicament for the treatment of a disease selected from the group consisting of: atherosclerosis, hypercholesterolemia and hyperlipidemia; cancer, glioblastoma, breast cancer, lymphoma, lung cancer; diabetes, metabolic disorders; myoblast differentiation; immune disorders.
  • a disease selected from the group consisting of: atherosclerosis, hypercholesterolemia and hyperlipidemia; cancer, glioblastoma, breast cancer, lymphoma, lung cancer; diabetes, metabolic disorders; myoblast differentiation; immune disorders.
  • the invention further refers to oligonucleotides according to the invention for the use in the treatment of from a disease selected from the group consisting of: atherosclerosis, hypercholesterolemia and hyperlipidemia; cancer, glioblastoma, breast cancer, lymphoma, lung cancer; diabetes, metabolic disorders; myoblast differentiation; immune disorders.
  • a disease selected from the group consisting of: atherosclerosis, hypercholesterolemia and hyperlipidemia; cancer, glioblastoma, breast cancer, lymphoma, lung cancer; diabetes, metabolic disorders; myoblast differentiation; immune disorders.
  • the invention provides for a method of treating a subject suffering from a disease or condition selected from from the group consisting of: atherosclerosis, hypercholesterolemia and hyperlipidemia; cancer, glioblastoma, breast cancer, lymphoma, lung cancer; diabetes, metabolic disorders; myoblast differentiation; immune disorders, the method comprising the step of administering an oligonucleotide or pharmaceutical composition of the invention to the subject in need thereof.
  • a disease or condition selected from from the group consisting of: atherosclerosis, hypercholesterolemia and hyperlipidemia; cancer, glioblastoma, breast cancer, lymphoma, lung cancer; diabetes, metabolic disorders; myoblast differentiation; immune disorders
  • the invention further provides for a kit comprising a pharmaceutical composition according to the invention, and a second independent active ingredient that is an inhibitor of the VLDL assembly pathway, such as an ApoB inhibitor, or an MTP inhibitor.
  • a pharmaceutical composition according to the invention and a second independent active ingredient that is an inhibitor of the VLDL assembly pathway, such as an ApoB inhibitor, or an MTP inhibitor.
  • the present invention relates to the use of an oligonucleotide according to the invention for the manufacture of a medicament for the treatment of cancer.
  • the present invention concerns a method for treatment of, or prophylaxis against, cancer, said method comprising administering an oligonucleotide of the invention or a pharmaceutical composition of the invention to a patient in need thereof.
  • Such cancers may include lymphoreticular neoplasia, lymphoblastic leukemia, brain tumors, gastric tumors, plasmacytomas, multiple myeloma, leukemia, connective tissue tumors, lymphomas, and solid tumors.
  • said cancer may suitably be in the form of a solid tumor.
  • said cancer in the method for treating cancer disclosed herein said cancer may suitably be in the form of a solid tumor.
  • said cancer is also suitably a carcinoma.
  • the carcinoma is typically selected from the group consisting of malignant melanoma, basal cell carcinoma, ovarian carcinoma, breast carcinoma, non-small cell lung cancer, renal cell carcinoma, bladder carcinoma, recurrent superficial bladder cancer, stomach carcinoma, prostatic carcinoma, pancreatic carcinoma, lung carcinoma, cervical carcinoma, cervical dysplasia, laryngeal papillomatosis, colon carcinoma, colorectal carcinoma and carcinoid tumors.
  • said carcinoma is selected from the group consisting of malignant melanoma, non-small cell lung cancer, breast carcinoma, colon carcinoma and renal cell carcinoma.
  • the malignant melanoma is typically selected from the group consisting of superficial spreading melanoma, nodular melanoma, lentigo maligna melanoma, acral melagnoma, amelanotic melanoma and desmoplastic melanoma.
  • the cancer may suitably be a sarcoma.
  • the sarcoma is typically in the form selected from the group consisting of osteosarcoma, Ewing's sarcoma, chondrosarcoma, malignant fibrous histiocytoma, fibrosarcoma and Kaposi's sarcoma.
  • the cancer may suitably be a glioma.
  • a further embodiment is directed to the use of an oligonucleotide according to the invention for the manufacture of a medicament for the treatment of cancer, wherein said medicament further comprises a chemotherapeutic agent selected from the group consisting of adrenocorticosteroids, such as prednisone, dexamethasone or decadron; altretamine (hexalen, hexamethylmelamine (HMM)); amifostine (ethyol); aminoglutethimide (cytadren); amsacrine (M- AMSA); anastrozole (arimidex); androgens, such as testosterone; asparaginase (elspar); bacillus calmette-gurin; bicalutamide (casodex); bleomycin (blenoxane); busulfan (myleran); carboplatin (paraplatin); carmustine (BCNU, BiCNU); chlorambucil (leukeran); chlorodeoxyadenosine (2-C
  • the invention is further directed to the use of an oligonucleotide according to the invention for the manufacture of a medicament for the treatment of cancer, wherein said treatment further comprises the administration of a further chemotherapeutic agent selected from the group consisting of adrenocorticosteroids, such as prednisone, dexamethasone or decadron; altretamine (hexalen, hexamethylmelamine (HMM)); amifostine (ethyol); aminoglutethimide (cytadren); amsacrine (M-AMSA); anastrozole (arimidex); androgens, such as testosterone; asparaginase (elspar); bacillus calmette-gurin; bicalutamide (casodex); bleomycin (blenoxane); busulfan (myleran); carboplatin (paraplatin); carmustine (BCNU, BiCNU); chlorambucil (leukeran); chlorodeoxyaden
  • the invention is furthermore directed to a method for treating cancer, said method comprising administering an oligonucleotide of the invention or a pharmaceutical composition according to the invention to a patient in need thereof and further comprising the administration of a further chemotherapeutic agent.
  • Said further administration may be such that the further chemotherapeutic agent is conjugated to the compound of the invention, is present in the pharmaceutical composition, or is administered in a separate formulation.
  • the compounds of the invention may be broadly applicable to a broad range of infectious diseases, such as diphtheria, tetanus, pertussis, polio, hepatitis B, hepatitis C, hemophilus influenza, measles, mumps, and rubella.
  • infectious diseases such as diphtheria, tetanus, pertussis, polio, hepatitis B, hepatitis C, hemophilus influenza, measles, mumps, and rubella.
  • Hsa-miR122 is indicated in hepatitis C infection and as such oligonucleotides according to the invention which target miR-122 may be used to treat Hepatitus C infection.
  • the present invention relates the use of an oligonucleotide according to the invention for the manufacture of a medicament for the treatment of an infectious disease, as well as to a method for treating an infectious disease, said method comprising administering an oligonucleotide according to the invention or a pharmaceutical composition according to the invention to a patient in need thereof.
  • the invention provides for a combination treatment providing an anti miR-122 oligomer in combination with an inhibitor of VLDL assembly, such as an inhibitor of apoB, or of MTP.
  • an inhibitor of VLDL assembly such as an inhibitor of apoB, or of MTP.
  • Inflammatory diseases The inflammatory response is an essential mechanism of defense of the organism against the attack of infectious agents, and it is also implicated in the pathogenesis of many acute and chronic diseases, including autoimmune disorders. In spite of being needed to fight pathogens, the effects of an inflammatory burst can be devastating. It is therefore often necessary to restrict the symptomatology of inflammation with the use of anti-inflammatory drugs. Inflammation is a complex process normally triggered by tissue injury that includes activation of a large array of enzymes, the increase in vascular permeability and extravasation of blood fluids, cell migration and release of chemical mediators, all aimed to both destroy and repair the injured tissue.
  • the present invention relates to the use of an oligonucleotide according to the invention for the manufacture of a medicament for the treatment of an inflammatory disease, as well as to a method for treating an inflammatory disease, said method comprising administering an oligonucleotide according to the invention or a pharmaceutical composition according to the invention to a patient in need thereof.
  • the inflammatory disease is a rheumatic disease and/or a connective tissue diseases, such as rheumatoid arthritis, systemic lupus erythematous (SLE) or Lupus, scleroderma, polymyositis, inflammatory bowel disease, dermatomyositis, ulcerative colitis, Crohn's disease, vasculitis, psoriatic arthritis, exfoliative psoriatic dermatitis, pemphigus vulgaris and Sjorgren's syndrome, in particular inflammatory bowel disease and Crohn's disease.
  • SLE systemic lupus erythematous
  • Lupus scleroderma
  • polymyositis inflammatory bowel disease
  • dermatomyositis ulcerative colitis
  • Crohn's disease vasculitis
  • psoriatic arthritis exfoliative psoriatic dermatitis
  • pemphigus vulgaris and Sjorgren's syndrome
  • the inflammatory disease may be a non-rheumatic inflammation, like bursitis, synovitis, capsulitis, tendinitis and/or other inflammatory lesions of traumatic and/or university origin.
  • a metabolic disease is a disorder caused by the accumulation of chemicals produced naturally in the body. These diseases are usually serious, some even life threatening. Others may slow physical development or cause mental retardation. Most infants with these disorders, at first, show no obvious signs of disease. Proper screening at birth can often discover these problems. With early diagnosis and treatment, metabolic diseases can often be managed effectively.
  • the present invention relates to the use of an oligonucleotide according to the invention or a conjugate thereof for the manufacture of a medicament for the treatment of a metabolic disease, as well as to a method for treating a metabolic disease, said method comprising administering an oligonucleotide according to the invention or a conjugate thereof, or a pharmaceutical composition according to the invention to a patient in need thereof.
  • the metabolic disease is selected from the group consisting of Amyloidosis, Biotinidase, OMIM (Online Mendelian Inheritance in Man), Crigler Najjar Syndrome, Diabetes, Fabry Support & Information Group, Fatty acid Oxidation Disorders, Galactosemia, Glucose-6-Phosphate Dehydrogenase (G6PD) deficiency, Glutaric aciduria, International Organization of Glutaric Acidemia, Glutaric Acidemia Type I, Glutaric Acidemia, Type II, Glutaric Acidemia Type I, Glutaric Acidemia Type-ll, F-HYPDRR - Familial Hypophosphatemia, Vitamin D Resistant Rickets, Krabbe Disease, Long chain 3 hydroxyacyl CoA dehydrogenase deficiency (LCHAD), Mannosidosis Group, Maple Syrup Urine Disease, Mitochondrial disorders, Mucopolysaccharidosis Syndromes: Niemann Pick, Organic acidemias, P
  • the present invention relates to the use of an oligonucleotide according to the invention or a conjugate thereof for the manufacture of a medicament for the treatment of a liver disorder, as well as to a method for treating a liver disorder, said method comprising administering an oligonucleotide according to the invention or a conjugate thereof, or a pharmaceutical composition according to the invention to a patient in need thereof.
  • the liver disorder is selected from the group consisting of Biliary Atresia, Alagille Syndrome, Alpha-1 Antitrypsin, Tyrosinemia, Neonatal Hepatitis, and Wilson Disease.
  • the oligonucleotides of the present invention can be utilized for as research reagents for diagnostics, therapeutics and prophylaxis.
  • the oligonucleotide may be used to specifically inhibit the synthesis of target genes in cells and experimental animals thereby facilitating functional analysis of the target or an appraisal of its usefulness as a target for therapeutic intervention.
  • the oligonucleotides may be used to detect and quantitate target expression in cell and tissues by Northern blotting, in-situ hybridisation or similar techniques.
  • an animal or a human, suspected of having a disease or disorder, which can be treated by modulating the expression of target is treated by administering the oligonucleotide compounds in accordance with this invention.
  • Still another aspect of the invention is use of the above described oligonucleotides targeting miR-122a for the preparation of a medicament for treatment of increased plasma cholesterol levels (or hypercholesterolemia and related disorders).
  • increased plasma cholesterol levels is undesireable as it increases the risk of various conditions, e.g. atherosclerosis.
  • Still another aspect of the invention is use of the above described oligonucleotides targeting miR-122a for upregulating the mRNA levels of Nrdg3, Aldo A, Bckdk or CD320.
  • a pharmaceutical composition comprising an oligomer of between 6-12 nucleotides in length, wherein said oligomer comprises a contiguous nucleotide sequence of a total of between 6-12 nucleotides, such as 6, 7, 8, 9, 10, 11 or 12 nucleotide units, wherein at least 50% of the nucleobase units of the oligomer are high affinity nucleotide analogue units, and a pharmaceutically acceptable diluent, carrier, salt or adjuvant.
  • composition according to embodiment 1 wherein the contiguous nucleotide sequence is complementary to a corresponding region of a mammalian, human or viral microRNA (miRNA) sequence.
  • miRNA microRNA
  • composition according to embodiment 2 wherein the contiguous nucleotide sequence is complementary to a corresponding region of a miRNA sequence selected from the group of miRNAs listed in any one of tables 3, 4 or 5.
  • composition according to embodiment 4 or 5 wherein the 3' nucleobase of the seedmer forms the 3' most nucleobase of the contiguous nucleotide sequence, wherein the contiguous nucleotide sequence may, optionally, comprise one or two further 5' nucleobases.
  • composition according to embodiment 8, wherein said miRNA is selected from the group consisting of miR-1 , miR-IOb, miR-17-3p, miR-18, miR-19a, miR-19b, miR-20, miR-21 , miR-34a, miR-93, miR-106a, miR-106b, miR-122, miR-133, miR-134, miR-138, miR-155, miR-192, miR-194, miR-221 , miR-222, and miR-375.
  • miRNA is selected from the group consisting of miR-1 , miR-IOb, miR-17-3p, miR-18, miR-19a, miR-19b, miR-20, miR-21 , miR-34a, miR-93, miR-106a, miR-106b, miR-122, miR-133, miR-134, miR-138, miR-155, miR-192, miR-194, miR-221 , miR-222, and mi
  • composition according to any one of embodiments 1-9, wherein at least 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or all of the nucleobase units of the contiguous nucleotide sequence are nucleotide analogue units.
  • nucleotide analogue units are selected from the group consisting of 2'-O_alkyl-RNA unit, 2'-OMe-RNA unit, 2'-amino-DNA unit, 2'-fluoro-DNA unit, LNA unit, PNA unit, HNA unit, INA unit, and a 2'MOE RNA unit.
  • composition according to embodiment 10 or 11 wherein at least 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or all of the nucleobase units of the contiguous nucleotide sequence are Locked Nucleic Acid (LNA) nucleobase units.
  • LNA Locked Nucleic Acid
  • composition according to any one of embodiments 1-14, wherein the oligomer consist of 7, 8, 9 or 10 contiguous nucleobase units and wherein at least 7 nucleobase units are nucleotide analogue units.
  • nucleotide analogue units are Locked Nucleic Acid (LNA) nucleobase units.
  • LNA Locked Nucleic Acid
  • nucleotide analogue units in the molecule consists of a mixture of at least 50% LNA units and up to 50 % other nucleotide analogue units.
  • composition according to embodiment 25 for use in the treatment of a medical disorder or disease selected from the group consisting of: hepatitis C virus infection and hypercholesterolemia and related disorders.
  • a medical disorder or disease selected from the group consisting of: hepatitis C virus infection and hypercholesterolemia and related disorders.
  • 27 The pharmaceutical composition according to embodiment 25 or 26, wherein the composition further comprises a second independent active ingredient that is an inhibitor of the VLDL assembly pathway, such as an ApoB inhibitor, or an MTP inhibitor.
  • kits comprising a pharmaceutical composition according to embodiment 25 or 26, and a second independent active ingredient that is an inhibitor of the VLDL assembly pathway, such as an ApoB inhibitor, or an MTP inhibitor.
  • a method for the treatment of a disease or medical disorder associated with the presence or overexpression of a microRNA comprising the step of administering a the pharmaceutical composition) according to any one of embodiments 1 - 28 to a patient who is suffering from, or is likely to siffer from said disease or medical disorder.
  • An oligomer as defined according to anyone of embodiments 1 - 25.
  • a conjugate comprising the oligomer according to embodiment 30, and at least one non- nucleobase compounds.
  • a method for reducing the amount, or effective amount, of a miRNA in a cell comprising administering an oligomer, a conjugate or a pharmaceutical composition, according to any one of the preceeding embodiments to the cell which is expressing said miRNA so as to reduce the amount, or effective amount of the miRNA in the cell.
  • a method for de-repression of a mRNA whose expression is repressed by a miRNA in a cell comprising administering an oligomer, a conjugate or a pharmaceutical composition, according to any one of the preceeding embodiments to the cell to the cell which expressed both said mRNA and said miRNA, in order to de-repress the expression of the mRNA.
  • LNA Monomer and oligonucleotide synthesis were performed using the methodology referred to in Examples 1 and 2 of WO2007/112754.
  • the stability of LNA oligonucletides in human or rat plasma is performed using the methodology referred to in Example 4 of WO2007/112754.
  • the treatment of in vitro cells with LNA anti-miR antisense oligonucleotide (targeting miR-122) is performed using the methodology referred to in Example 6 of WO2007/112754.
  • the analysis of Oligonucleotide Inhibition of miR expression by microRNA specific quantitative PCR in both an in vitro and in vivo model is performed using the methodology referred to in Example 7 of WO2007/112754.
  • the assessment of LNA antimir knock-down specificity using miRNA microarray expression profiling is performed using the methodology referred to in Example 8 of WO2007/112754.
  • the detection of microRNAs by in situ hybridization is performed using the methodology referred to in Example 9 of WO2007/112754.
  • the Isolation and analysis of mRNA expression (total RNA isolation and cDNA synthesis for mRNA analysis) in both an in vitro and in vivo model is performed using the methodology referred to in Example 10 of WO2007/112754.
  • In vivo Experiments using Oligomers of the invention targeting microRNA- 122. and subsequent analysis are performed using the methods disclosed in Examples 11 - 27 of WO2007/112754.
  • the above mentioned examples of WO2007/112754 are hereby specifically incorporated by reference.
  • Example 1 Design of the LNA antimiR oligonucleotides and melting temperatures
  • SEQ# is an identifier used throughout the examples and figures - the SEQ ID NO which is used in the sequence listing is also provided.
  • LNA oligonucleotides on target nucleic acid expression can be tested in any of a variety of cell types provided that the target nucleic acid is present at measurable levels.
  • Target can be expressed endogenously or by transient or stable transfection of a nucleic acid encoding said nucleic acid.
  • target nucleic acid can be routinely determined using, for example, Northern blot analysis (including microRNA northern), Quantitative PCR (including microRNA qPCR), Ribonuclease protection assays.
  • Northern blot analysis including microRNA northern
  • Quantitative PCR including microRNA qPCR
  • Ribonuclease protection assays The following cell types are provided for illustrative purposes, but other cell types can be routinely used, provided that the target is expressed in the cell type chosen.
  • Cells were cultured in the appropriate medium as described below and maintained at 37°C at 95-98% humidity and 5% CO 2 . Cells were routinely passaged 2-3 times weekly.
  • the human prostate cancer cell line 15PC3 was kindly donated by Dr. F. Baas, Neurozintuigen Laboratory, AMC, The Netherlands and was cultured in DMEM (Sigma) + 10% fetal bovine serum (FBS) + Glutamax I + gentamicin.
  • PC3 The human prostate cancer cell line PC3 was purchased from ATCC and was cultured in
  • 518A2 The human melanoma cancer cell line 518A2 was kindly donated by Dr. B. Jansen, Section of experimental Oncology, Molecular Pharmacology, Department of Clinical
  • HeLa The cervical carcinoma cell line HeLa was cultured in MEM (Sigma) containing 10% fetal bovine serum gentamicin at 37°C, 95% humidity and 5% CO 2 .
  • MPC-11 The murine multiple myeloma cell line MPC-11 was purchased from ATCC and maintained in DMEM with 4mM Glutamax+ 10% Horse Serum.
  • the human prostate cancer cell line DU-145 was purchased from ATCC and maintained in RPMI with Glutamax + 10% FBS.
  • RCC-4 +/- VHL The human renal cancer cell line RCC4 stably transfected with plasmid expressing VHL or empty plasmid was purchased from ECACC and maintained according to manufacturers instructions.
  • the human renal cell carcinoma cell line 786-0 was purchased from ATCC and maintained according to manufacturers instructions
  • HUVEC The human umbilical vein endothelial cell line HUVEC was purchased from Camcrex and maintained in EGM-2 medium.
  • K562 The human chronic myelogenous leukaemia cell line K562 was purchased from ECACC and maintained in RPMI with Glutamax + 10% FBS.
  • U87MG The human glioblastoma cell line
  • U87MG was purchased from ATCC and maintained according to the manufacturers instructions.
  • B16 The murine melanoma cell line B16 was purchased from ATCC and maintained according to the manufacturers instructions.
  • LNCap The human prostate cancer cell line LNCap was purchased from ATCC and maintained in RPMI with Glutamax + 10% FBS
  • Huh-7 Human liver, epithelial like cultivated in Eagles MEM with 10 % FBS, 2mM Glutamax I, 1x non-essential amino acids, Gentamicin 25 ⁇ g/ml L428: (Deutsche Sammlung f ⁇ r Mikroorganismen (DSM, Braunschwieg, Germany)): Human B cell lymphoma maintained in RPMI 1640 supplemented with 10% FCS, L-glutamine and antibiotics.
  • L1236 (Deutsche Sammlung f ⁇ r Mikroorganismen (DSM, Braunschwieg, Germany)): Human B cell lymphoma maintained in RPMI 1640 supplemented with 10% FCS, L-glutamine and antibiotics.
  • Example 3 Design of a LNA antimiR library for all human microRNA sequences in miRBase microRNA database.
  • miRBase version used was version 12, as reported in Griffiths-Jones, S., Grocock, RJ. , van Dongen, S., Bateman, A., Enright, A.J. 2006. miRBase: microRNA sequences, targets and gene nomenclature. Nucleic Acids Res. 34: D140-4, and available via http://microrna. sanger.ac.uk/sequences/index.shtml.
  • Table 1 shows 7, 8 and 9mer nucleotide sequences comprising the seedmer sequence of micro RNA's according to the miRBase micro RNA database.
  • the seedmer sequence comprises the reverse complement of the microRNA seed region.
  • the oligomer of the invention has a contiguous nucleotide sequence selected from the 7mer, 8mer or 9mer sequences. With respect to the 7mer, 8mer and 9mer sequences, in some embodiments, all the intemucleoside linkages are phosphorothioate.
  • the 7mer, 8mer and 9mer nucleotide sequences may consist of sequence of nucleotide analogues as described herein, such as LNA nucleotide analogues.
  • LNA cytosines may be methyl-cytosine (5'methyl-cytosine). In some embodiments, the LNA is beta-D-oxy-LNA.
  • Table 3 provides a list of microRNAs grouped into those which can be targeted by the same seedmer oligomers, such as the 7, 8 or 9mers provided herein (see table 1 ).
  • the melting temperatures can be assessed towards the mature microRNA sequence, using a synthetic microRNA oligonucleotide (typically consisting of RNA nucleotides with a phosphodiester backbone). Typically measured T m s are higher than predicted T m s when using LNA oligomers against the RNA target.
  • Example 4 Assessment of miR-21 antagonism by SEQ ID #3205 LNA-antimiR in MCF-7 cells using a luciferase sensor assay.
  • luciferase sensor constructs were made containing a perfect match target site for the mature miR-21 and as control, a target site with two mutations in the seed (Fig. 6).
  • the breast carcinoma cell line MCF-7 was transfected with the different luciferase constructs together with the miR-21 antagonist SEQ ID #3205 at varying concentrations in comparison with a 15-mer LNA-antimiR SEQ ID #3204 against miR-21. After 24 hours, luciferase activity was measured.
  • the new fully LNA-modified 8-mer LNA-antimiR shows two-fold higher potency compared to SEQ ID #3204, as shown by de-repression of the Iuciferase activity.
  • the control miR-21 sensor construct with two mismatches in the miR-21 seed did not show any de-repression of the firefly Iuciferase activity, thereby demonstrating the specificity of the perfect match miR-21 sensor in monitoring miR-21 activity in cells.
  • the de-repression of Iuciferase activity by the 8-mer LNA-antimiR is clearly dose- dependent, which is not seen with SEQ ID #3204.
  • the new 8-mer is also much more potent at lower doses than SEQ ID #3204.
  • the 8-mer LNA-antimiR shows significantly improved potency in inhibition of miR-21 in vitro compared to the 15-mer LNA-antimiR SEQ ID #3204 targeting miR- 21.
  • MCF-7 The breast carcinoma cell line MCF-7 was purchased from ATCC (#HTB-22TM). MCF- 7 cells were cultured in EMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax, 1x NEAA and 25 ug/ml Gentamicin. Transfection: 400.000 cells were seeded per well in a 6-well plate the day before transfection in order to receive 50-70% confluency the next day.
  • MCF-7 cells were transfected with 0.8 ug miR-21 perfect match/psiCHECK2, miR-21.mm2/psiCHECK2 or empty psiCHECK2 vector (SDS Promega) together with 1 ⁇ l Lipofectamine2000 (Invitrogen) according to manufacturer's instructions. After 24 hours, cells were harvested for Iuciferase measurements. Lucif erase assay: The cells were washed with PBS and harvested with cell scraper, after which cells were centrifugated for 5 min at 10.000 rpm. The supernatant was discarded and 50 ⁇ l 1 x Passive Lysis Buffer (Promega) was added to the cell pellet, after which cells were put on ice for 30 min. The lysed cells were spinned at 10.000 rpm for 30 min after which 20 ⁇ l were transferred to a 96 well plate and Iuciferase measurements were performed according to manufacturer's instructions (Promega).
  • Example 5 Assessment of miR-21 antagonism by SEQ ID #3205 LNA-antimiR in HeLa cells using a Iuciferase sensor assay.
  • the cervix carcinoma cell line HeLa was also transfected with the previously described miR-21 Iuciferase sensor constructs alongside SEQ ID #3205 at varying concentrations as described in the above section ( Figure 3).
  • the SEQ ID #3205 shows complete de-repression of the miR-21 Iuciferase sensor construct in HeLa cells already at 5 nM compared to SEQ ID #3204, which did not show complete de-repression until the highest dose (50 nM).
  • antagonism of miR-21 by the SEQ ID #3205 shows complete de-repression of the miR-21 Iuciferase sensor construct in HeLa cells already at 5 nM compared to SEQ ID #3204, which did not show complete de-repression until the highest dose (50 nM).
  • the fully LNA-modified SEQ ID #3205 shows significantly improved potency in inhibition of miR-21 in vitro, in both MCF-7 and HeLa cells compared to the 15-mer LNA-antimiR SEQ ID #3204.
  • HeLa The human cervix carcinoma cell line HeLa was purchased from ECACC (#93021013). HeLa cells were cultured in EMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax, 1x NEAA and 25 ug/ml Gentamicin.
  • Transfection 60.000 cells were seeded per well in a 24 well plate the day before transfection in order to receive 50-70% confluency the next day. On the day of transfection, HeLa cells were transfected with 0.2 ug miR-21 perfect match/psiCHECK2, miR-21.mm2/psiCHECK2 or empty psiCHECK2 vector together with 0,7 ⁇ l Lipofectamine2000 (Invitrogen) according to manufacturer's instructions. After 24 hours, cells were harvested for luciferase measurements.
  • Luciferase assay The cells were washed with PBS and 100 ⁇ l 1 x Passive Lysis Buffer (Promega) was added to each well, after which the 24 well plates was put on an orbital shaker for 30 min. The cells were collected and transferred to an eppendorf tube and spinned at 10.000 rpm for 30 min after which 10 ⁇ l were transferred to a 96 well plate and luciferase measurements were performed according to manufacturer's instructions (Promega).
  • Example 6 Assessment of miR-155 antagonism by SEQ ID #3207 LNA-antimiR in mouse RAW cells using a luciferase sensor assay.
  • a perfect match target site for miR-155 was cloned into the same luciferase vector (psiCHECK2) and transfected into the mouse leukaemic monocyte macrophage RAW cell line. Because the endogenous levels of miR-155 are low in the RAW cell line, the cells were treated with 100 ng/ml LPS for 24 hours in order to induce miR-155 accumulation.
  • Luciferase measurements showed that the fully LNA-modified 8-mer LNA-antimiR SEQ ID #3207 targeting miR-155 was similarly effective in antagonizing miR-155 compared to the 15-mer LNA-antimiR SEQ ID #3206 ( Figure 4). Both LNA-antimirs showed a >50% derepression of the miR-155 luciferase sensor at 0.25 nM concentration and inhibited miR-155 in a dose-dependent manner.
  • the mouse leukaemic monocyte macrophage RAW 264.7 was purchased from ATCC (TIB-71 ). RAW cells were cultured in DMEM medium, supplemented with 10% fetal bovine serum, 4 mM Glutamax and 25 ug/ml Gentamicin. Transfection: 500.000 cells were seeded per well in a 6 well plate the day before transfection in order to receive 50% confluency the next day. On the day of transfection, MCF-7 cells were transfected with 0.3 ug miR-155 or empty psiCHECK2 vector together with 10 ⁇ l Lipofectamine2000 (Invitrogen) according to manufacturer's instructions. In order to induce miR- 155 accumulation, LPS (100 ng/ml) was added to the RAW cells after the 4 hour incubation with the transfection complexes. After another 24 hours, cells were harvested for luciferase measurements.
  • DMEM medium supplemented with 10% fetal bovine serum, 4 mM Glutamax and 25
  • Luciferase assay The cells were washed with PBS and harvested with cell scraper, after which cells were centrifugated for 5 min at 2.500 rpm. The supernatant were discarded and 50 ⁇ l 1 x Passive Lysis Buffer (Promega) was added to the cell pellet, after which cells were put on ice for 30 min. The lysed cells were spinned at 10.000 rpm for 30 min after which 20 ⁇ l were transferred to a 96 well plate and luciferase measurments were performed according to manufacturer's instructions (Promega).
  • Example 7 Assessment of miR-122 antagonism by SEQ ID #3208 LNA-antimiR in HuH-7 cells using a luciferase sensor assay.
  • the potency of the fully modified 8-mer LNA-antimiR SEQ ID #3208 against miR-122 was assessed in the human hepatoma cell line HuH-7.
  • the HuH-7 cells were transfected with luciferase sensor construct containing a perfect match miR-122 target site. After 24 hours luciferase measurements were performed (Figure 5).
  • the human hepatoma cell line HuH-7 was a kind gift from R. Bartenschlager,
  • Huh-7 cells were cultured in EMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax, 1x NEAA and 25 ug/ml Gentamicin.
  • Transfection 8.000 cells were seeded per well in a 96 well plate the day before transfection in order to receive 50-70% confluency the next day. On the day of transfection, HuH-7 cells were transfected with 57 ng miR-122 or empty psiCHECK2 vector together with 1 ⁇ l Lipofectamine2000 (Invitrogen). After 24 hours, cells were harvested for luciferase measurements.
  • Luciferase assay 50 ⁇ l 1 x Passive Lysis Buffer (Promega) was added to each well, after which the 96 well plate was put on an orbital shaker for 30 min. To each well the Dual-luciferase Reporter assay system (Promega) was added and luciferase measurements were performed according to manufacturer's instructions (Promega).
  • Example 8 Assessment of miR-21 antagonism by comparing an 8-mer (SEQ ID #3205) versus a 15-mer (SEQ ID #3204) LNA-antimiR in human prostate carcinoma cells (PC3).
  • PC3 human prostate carcinoma cells
  • luciferase reporter constructs were generated in which a perfect match target site for the mature miR-21 and a target site with two mismatches in the seed were cloned in the 3'UTR of Renilla luciferase gene ( Figure 7).
  • PC3 cells were transfected with the different luciferase constructs together with the miR-21 antagonist SEQ ID #3205 (8-mer) and for comparison with the 15-mer LNA-antimiR perfect match SEQ ID #3204 at varying concentrations. After 24 hours, luciferase activity was measured.
  • PC3 cell line The human prostate carcinoma PC3 cell line was purchased from ECACC (#90112714). PC3 cells were cultured in DMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax and 25 ug/ml Gentamicin.
  • Transfection 100.000 cells were seeded per well in a 12-well plate the day before transfection in order to receive 50% confluency the next day.
  • PC3 cells were transfected with 0.3 ⁇ g miR-21 or empty psiCHECK2 vector together with 1 ,2 ⁇ l Lipofectamine2000 (Invitrogen) according to manufacturer's instructions.
  • Transfected was also varying concentrations of LNA-antimiRs. After 24 hours, cells were harvested for luciferase measurements.
  • Luciferase assay The cells were washed with PBS and 250 ⁇ l 1 x Passive Lysis Buffer
  • Example 9 Specificity assessment of miR-21 antagonism by an 8-mer LNA-antimiR
  • an 8-mer mismatch control LNA-antimiR (SEQ ID #3218) containing 2 mismatches in the seed recognition sequence (see Figure 8).
  • the luciferase reporter constructs described in example 1 were transfected into the human cervix carcinoma cell line HeLa together with the LNA mismatch control oligo SEQ ID #3218 and its efficacy was compared with the 8-mer LNA- antimiR (SEQ ID #3205) targeting miR-21. After 24 hours, luciferase activity was measured.
  • HeLa The human cervix carcinoma cell line HeLa was purchased from ECACC (#93021013). HeLa cells were cultured in EMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax, 1x NEAA and 25 ug/ml Gentamicin. Transfection: 60.000 cells were seeded per well in a 24-well plate the day before transfection in order to receive 50-70% confluency the next day.
  • HeLa cells were transfected with 0.2 ug miR-21 perfect match/psiCHECK2, miR-21.mm2/psiCHECK2 or empty psiCHECK2 vector together with 0,7 ⁇ l Lipofectamine2000 (Invitrogen) according to manufacturer's instructions. Transfected was also varying concentrations of LNA-antimiRs. After 24 hours, cells were harvested for luciferase measurements. Luciferase assay: The cells were washed with PBS and 100 ⁇ l 1 x Passive Lysis Buffer
  • Example 10 Assessment of the shortest possible length of a fully LNA-modified LNA- antimiR that mediates effective antagonism of miR-21.
  • HeLa The human cervix carcinoma cell line HeLa was purchased from ECACC (#93021013). HeLa cells were cultured in EMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax, 1x NEAA and 25 ug/ml Gentamicin. Transfection: 60.000 cells were seeded per well in a 24 well plate the day before transfection in order to receive 50-70% confluency the next day.
  • HeLa cells were transfected with 0.2 ug miR-21 perfect match/psiCHECK2, miR-21.mm2/psiCHECK2 or empty psiCHECK2 vector together with 0,7 ⁇ l Lipofectamine2000 (Invitrogen) according to manufacturer's instructions. Transfected was also varying concentrations of LNA-antimiRs. After 24 hours, cells were harvested for luciferase measurements. Luciferase assay: The cells were washed with PBS and 100 ⁇ l 1 x Passive Lysis Buffer
  • Cell line The human cervix carcinoma cell line HeLa was purchased from ECACC (#93021013). HeLa cells were cultured in EMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax, 1x NEAA and 25 ug/ml Gentamicin.
  • Transfection 60.000 cells were seeded per well in a 24-well plate the day before transfection in order to achieve 50-70% confluency the next day. On the day of transfection, HeLa cells were transfected with 0.2 ug miR-21 perfect match/psiCHECK2, miR-21. mm2/psiCHECK2 or empty psiCHECK2 control vector without target site together with 0,7 ⁇ l Lipofectamine2000 (Invitrogen) according to manufacturer's instructions. Transfected was also varying concentrations of LNA-antimiRs. After 24 hours, cells were harvested for luciferase measurements.
  • Luciferase assay The cells were washed with PBS and 100 ⁇ l 1 x Passive Lysis Buffer (Promega) was added to each well, after which the 24-well plates were put on an orbital shaker for 30 min. The cells were collected and transferred to an eppendorf tube and spinned at 10.000 rpm for 30 min after which 10 ⁇ l were transferred to a 96-well plate and luciferase measurements were performed according to manufacturer's instructions (Promega).
  • Example 12 Determination of the most optimal position for an 8-mer LNA-antimiR within the miR target recognition sequence.
  • Our experiments have shown that the most potent fully LNA-modified phosphorothiolated LNA- antimiR is 8 nucleotides in length.
  • To assess the most optimal position for an 8-mer LNA- antimiR within the miR target recognition sequence we designed four different fully LNA- modified 8-mer LNA-antimiRs tiled across the mature miR-21 sequence as shown in Figure 11. The different LNA-antimiRs were co-transfected together with the miR-21 luciferase reporter constructs into HeLa cells. Luciferase measurements were performed after 24 hours.
  • HeLa cells were cultured in EMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax, 1x NEAA and 25 ug/ml Gentamicin.
  • Transfection 60.000 cells were seeded per well in a 24-well plate the day before transfection in order to achieve 50-70% confluency the next day. On the day of transfection, HeLa cells were transfected with 0.2 ug miR-21 perfect match/psiCHECK2, miR-21.mm2/psiCHECK2 or empty psiCHECK2 vector together with 0,7 ⁇ l Lipofectamine2000 (Invitrogen) according to the manufacturer's instructions. Transfected was also varying concentrations of LNA-antimiRs. After 24 hours, cells were harvested for luciferase measurements.
  • Luciferase assay The cells were washed with PBS and 100 ⁇ l 1 x Passive Lysis Buffer (Promega) was added to each well, after which the 24-well plates was put on an orbital shaker for 30 min. The cells were collected and transferred to an eppendorf tube and spinned at 10.000 rpm for 30 min after which 10 ⁇ l were transferred to a 96 well plate and luciferase measurements were performed according to manufacturer's instructions (Promega).
  • Example 13 Validation of interaction of the miR-21 target site in the Pdcd4-3 ' -UTR and miR-21 using the ⁇ -mer SEQ ID #3205 LNA-antimiR.
  • tumour suppressor protein Pdcd4 inhibits TPA-induced neoplastic transformation, tumour promotion and progression.
  • Pdcd4 has also been shown to be upregulated in apoptosis in response to different inducers.
  • downregulation of Pdcd4 in lung and colorectal cancer has also been associated with a poor patient prognosis.
  • Asangani efa/ and Frankel et al showed that the Pdcd4-3'-UTR contains a conserved target site for miR-21 , and transfecting cells with an antimiR-21 , resulted in an increase in Pdcd4 protein.
  • luciferase reporter plasmid harboring 313 nt of the 3 ' UTR region of Pdcd4 encompassing the aforementioned miR-21 target site, which was co-transfected together with different LNA-antimiRs into HeLa cells.
  • the different LNA-antimiRs were; SEQ ID #3205 (8-mer, perfect match) or SEQ ID #3218 (8-mer, mismatch). Luciferase measurements were performed after 24 hours.
  • HeLa The human cervix carcinoma cell line HeLa was purchased from ECACC (#93021013). HeLa cells were cultured in EMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax, 1x NEAA and 25 ug/ml Gentamicin. Transfection: 60.000 cells were seeded per well in a 24-well plate the day before transfection in order to achieve 50-70% confluency the next day. On the day of transfection, HeLa cells were transfected with 0.2 ug Pdcd4-3'UTR/psiCHECK2 or empty psiCHECK2 vector together with 0,7 ⁇ l Lipofectamine2000 (Invitrogen) according to the manufacturer's instructions. Varying concentrations of the LNA-antimiR oligonucleotides were also transfected. After 24 hours, cells were harvested for luciferase measurements.
  • Luciferase assay The cells were washed with PBS and 100 ⁇ l 1 x Passive Lysis Buffer (Promega) was added to each well, after which the 24-well plates was put on an orbital shaker for 30 min. The cells were collected and transferred to an eppendorf tube and spinned at 10.000 rpm for 30 min after which 10 ⁇ l were transferred to a 96 well plate and luciferase measurements were performed according to manufacturer's instructions (Promega).
  • Example 14 Comparison of an 8-mer LNA-antimiR (SEQ ID #3207) with a 15-mer LNA- antimiR (SEQ ID #3206) in antagonizing miR-155 in mouse RAW cells.
  • Luciferase measurements showed that the fully LNA-modified 8-mer LNA-antimiR SEQ ID #3207 targeting miR-155 was similarly effective in antagonizing miR-155 compared to the 15-mer LNA-antimiR SEQ ID #3206 ( Figure 13). Both LNA-antimiRs showed a >50% derepression of the miR-155 luciferase sensor at 0.25 nM concentration and inhibited miR-155 in a dose-dependent manner. Analysis of the miRBase microRNA sequence database showed that the miR-155 recognition sequence, of the LNA-antimiR SEQ ID #3207 is unique for microRNA-155.
  • Cell line The mouse macrophage RAW 264.7 cell line was purchased from ATCC (TIB-71 ). RAW cells were cultured in DMEM medium, supplemented with 10% fetal bovine serum, 4 mM Glutamax and 25 ug/ml Gentamicin.
  • Transfection 500.000 cells were seeded per well in a 6-well plate the day before transfection in order to receive 50% confluency the next day.
  • RAW 264.7 cells were transfected with 0.3 ug miR-155 perfect match/psiCHECK2 or empty psiCHECK2 vector together with 10 ⁇ l Lipofectamine2000 (Invitrogen) according to the manufacturer's instructions.
  • Transfected was also varying concentrations of LNA-antimiRs.
  • LPS 100 ng/ml was added to the RAW cells after the 4 hour incubation with the transfection complexes. After another 24 hours, cells were harvested for luciferase measurements.
  • Luciferase assay The cells were washed with PBS and harvested with cell scraper, after which cells were spinned for 5 min at 2.500 rpm. The supernatant was discarded and 50 ⁇ l 1 x Passive Lysis Buffer (Promega) was added to the cell pellet, after which cells were put on ice for 30 min. The lysed cells were spinned at 10.000 rpm for 30 min after which 20 ⁇ l were transferred to a 96-well plate and luciferase measurements were performed according to the manufacturer's instructions (Promega).
  • Example 15 Assessment of c/EBP ⁇ protein levels as a functional readout for miR-155 antagonism by short LNA-antimiR (SEQ ID #3207).
  • Transfection 500.000 cells were seeded per well in a 6-well plate the day before transfection in order to achieve 50% confluency the next day.
  • RAW 264.7 cells were transfected with 5 nmol pre-miR-155 (Ambion) and/or 5 nM LNA-antimiR together with 10 ⁇ l Lipofectamine2000 (Invitrogen) according to the manufacturer's instructions.
  • Transfected was also varying concentrations of LNA-antimiRs.
  • LPS 100 ng/ml was added to the RAW cells after the 4 hour incubation with the transfection complexes. After 16 hours, cells were harvested for protein extraction and western blot analysis.
  • Example 16 Antagonism of miR-106b by a fully LNA-modified 8-mer (SEQ ID #3221) LNA- antimiR
  • Cell line The human cervix carcinoma cell line HeLa was purchased from ECACC (#93021013). HeLa cells were cultured in EMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax, 1x NEAA and 25 ug/ml Gentamicin.
  • Transfection 5.200 cells were seeded per well in a 96-well plate the day before transfection in order to achieve 50-70% confluency the next day. On the day of transfection, HeLa cells were transfected with 57 ng miR-21 perfect match/psiCHECK2, miR-21.mm2/psiCHECK2 or empty psiCHECK2 vector together with 0,14 ⁇ l Lipofectamine2000 (Invitrogen) according to the manufacturer's instructions. Transfected was also varying concentrations of LNA-antimiRs. After 24 hours, cells were harvested for luciferase measurements.
  • Luciferase assay The cells were washed with PBS and 30 ⁇ l 1 x Passive Lysis Buffer (Promega) was added to each well, after which the 24-well plates was put on an orbital shaker for 30 min. The cells were collected and transferred to eppendorf tubes and spinned at 10.000 rpm for 30 min after which luciferase measurements were performed according to the manufacturer's instructions (Promega).
  • Example 17 Antagonism of miR-19a by a fully LNA-modified 8-mer (SEQ ID #3222) LNA- antimiR
  • SEQ ID #3222 LNA- antimiR
  • a perfect match target site for miR-19a was cloned in the 3'UTR of the luciferase gene in the psiCHECK2 vector.
  • the reporter plasmid was transfected into the human cervix carcinoma HeLa cell line together with a short LNA-antimiR (SEQ ID #3222) or with a 15- mer LNA-antimiR (SEQ ID #3229) targeting miR-19a at varying concentrations. Luciferase measurements were performed after 24 hours.
  • transfection of the 15-mer LNA-antimiR SEQ ID #3229 into HeLa efficiently antagonizes miR-19a as demonstrated by complete de-repression at 1 nM LNA-antimiR concentration.
  • transfection of the 8-mer LNA-antimiR SEQ ID #3222 resulted in effective miR-19a antagonism already at 0.5 nM concentration, indicating that this 8-mer LNA-antimiR is at least equally potent compared with a 15-mer LNA-antimiR in HeLa cells.
  • Targeting of miR-19a in HeLa cells shows that an 8-mer fully LNA-modified and phosphorothiolated LNA-antimiR is at least equally potent compared with a 15-mer LNA/DNA mixmer LNA-antimiR.
  • Cell line The human cervix carcinoma cell line HeLa was purchased from ECACC (#93021013). HeLa cells were cultured in EMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax, 1x NEAA and 25 ug/ml Gentamicin. Transfection: 5.200 cells were seeded per well in a 96-well plate the day before transfection in order to achieve 50-70% confluency the next day.
  • HeLa cells were transfected with 57 ng miR-21 perfect match/psiCHECK2, miR-21.mm2/psiCHECK2 or empty psiCHECK2 vector together with 0,14 ⁇ l Lipofectamine2000 (Invitrogen) according to manufacturer's instructions. Transfected was also varying concentrations of LNA-antimiRs. After 24 hours, cells were harvested for luciferase measurements. Luciferase assay: The cells were washed with PBS and 30 ⁇ l 1 x Passive Lysis Buffer
  • Example 18 Targeting of a microRNA family using short, fully LNA-substituted LNA- antimiR.
  • a microRNA family using a single short 7- mer LNA-antimiR complementary to the seed sequence that is common for all family members we focused on miR-221 and miR-222 that are overexpressed in solid tumors of the colon, pancreas, prostate and stomach. It has also been shown that miR-221 and miR-222 are the most significantly upregulated microRNAs in glioblastoma multiforme. Furthermore, overexpression of miR-221 and miR-222 may contribute to the growth and progression of prostate carcinoma, at least in part by blocking the tumor suppressor protein p27.
  • PC3 cells were cultured in DMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax and 25 ug/ml Gentamicin.
  • Transfection 100.000 cells were seeded per well in a 12-well plate the day before transfection in order to receive 50% confluency the next day.
  • PC3 cells were transfected with 0.3 ug of luciferase reporter plasmid for miR-221 or for miR-222 or with empty psiCHECK2 vector without miRNA target site as control together with 1 ,2 ⁇ l Lipofectamine2000 (Invitrogen) according to the manufacturer's instructions. After 24 hours, cells were harvested for luciferase measurements.
  • Luciferase assay The cells were washed with PBS and 250 ⁇ l 1 x Passive Lysis Buffer (Promega) was added to the wells.
  • the plates were placed on a shaker for 30 min., after which the cell lysates was transferred to eppendorf tubes.
  • the cell lysate was spinned for 10 min at 2.500 rpm after which 20 ⁇ l were transferred to a 96-well plate and luciferase measurements were performed according to the manufacturer's instructions (Promega).
  • Example 19 Assessment of p27 protein levels as a functional readout for antagonism of the miR-221/222 family by the 7-mer SEQ ID #3225 LNA-antimiR.
  • PC3 cells were cultured in DMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax and 25 ug/ml Gentamicin.
  • Transfection 250.000 cells were seeded per well in a 6-well plate the day before transfection in order to receive 50% confluency the next day.
  • PC3 cells were transfected with LNA-antimiRs at varying concentrations with Lipofectamine2000. Cells were harvested after 24 hours for protein extraction and western blot analysis.
  • Western blot Cells were washed with PBS, trypsinated, transferred to eppendorf tubes and 250 ⁇ l lysis buffer (IxRIPA) was added. The cell lysate was placed on ice for 20 min, then spinned at 10.000 rpm for 10 minutes.
  • the protein concentration was measured with Coomassie Plus according to the manufacturer ' s instructions and 100 ug was loaded onto a 4-12% BIS-TRIS gel. The membrane was incubated overnight at 4 0 C with the primary monoclonal mouse antibody p27 (BD Biosciences) at a 1 :1000 dilution. Immunoreactive bands were visualized with ECL Plus (Amersham).
  • Example 20 Duplex melting temperatures (T m ) of the LNA-antimiRs.
  • T m values increase with increasing the length of short fully modified LNA- antimiRs (see T m values for SEQ ID #3205, SEQ ID #3209-3214 in Table 7). Most optimal inhibitory effect was achieved with the 8-mer LNA-antimiR SEQ ID #3205 against miR-21 , whereas the very low Tm of the 6-mer SEQ ID #3209 is most likely not sufficient to mediate antagonism of the miR-21 target.
  • T m measurements The oligonucleotide:miR-21 RNA duplexes were diluted to 3 ⁇ M in 500 ⁇ l RNase free H 2 O and mixed with 500 ⁇ l 2x T m -buffer (200 mM NaCI, 0.2 mM EDTA, 20 mM Na- phosphate, pH 7,0). The solution was heated to 95°C for 3 min and then allowed to anneal in RT for 30 min.
  • the duplex melting temperatures (T m ) were measured on a Lambda 40 UV/VIS Spectrophotometer equipped with a Peltier temperature programmer PTP6 using PE Templab software (Perkin Elmer). The temperature was ramped up from 20 0 C to 95°C and then down to 25°C, recording absorption at 260 nm. First derivative and the local maximums of both the melting and annealing were used to assess the duplex melting temperatures.
  • Example 21 Assessment of miR-21 antagonism by comparing an 8-mer (SEQ ID #3205) versus a 15-mer (SEQ ID #3204) LNA-antimiR in human hepatocytic cell line HepG2.
  • HepG2 cells were transfected with the luciferase constructs together with the miR-21 antagonist SEQ ID #3205 (8-mer) and for comparison of specificity with the 8-mer LNA-antimiR mismatch (SEQ ID #3218) and for comparison of potency together with the 15-mer (SEQ ID #3204) at varying concentrations. After 24 hours, luciferase activity was measured. Results: The luciferase reporter experiments showed a dose-dependent de-repression of the luciferase miR-21 reporter activity with the 15-mer LNA-antimiR against miR-21 (SEQ ID #3204).
  • the 8-mer (SEQ ID #3205) is more potent than the 15-mer LNA-antimiR in targeting miR-21 and antagonism of miR-21 by SEQ ID #3205 is specific.
  • Ce// line The human hepatocytic HepG2 cell line was purchased from ECACC (#85011430). HepG2 cells were cultured in EMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax and 25 ug/ml Gentamicin.
  • Transfection 650.000 cells were seeded per well in a 6-well plate and reverse transfection were performed. HepG2 cells were transfected with 0.6 ⁇ g miR-21 or empty psiCHECK2 vector together with 2,55 ⁇ l Lipofectamine2000 (Invitrogen) according to manufacturer's instructions. Transfected was also varying concentrations of LNA-antimiRs. After 24 hours, cells were harvested for luciferase measurements.
  • Luciferase assay The cells were washed with PBS and 300 ⁇ l 1 x Passive Lysis Buffer (Promega) was added to the wells. The plates were placed on a shaker for 30 min., after which the cell lysates were transferred to eppendorf tubes. The cell lysate was centrifugated for 10 min at 2.500 rpm after which 50 ⁇ l were transferred to a 96 well plate and luciferase measurements were performed according to the manufacturer's instructions (Promega).
  • Example 22 Validation of interaction of the miR-21 target site in the Pdcd4 3 ' UTR and miR-21 using the 8-mer SEQ ID #3205 LNA-antimiR in human hepatocellular cell line Huh- 7.
  • tumour suppressor protein Pdcd4 inhibits tumour promotion and progression. Furthermore, downregulation of Pdcd4 in lung and colorectal cancer has also been associated with poor patient prognosis. Recently, Asangani et al (Oncogene 2007) and Frankel et al (J Biol Chem 2008) showed that the Pdcd4 3'UTR contains a conserved target site for miR-21 , and transfecting cells with an antimiR-21, resulted in an increase in Pdcd4 protein.
  • luciferase reporter plasmid harboring 313 nt of the 3'UTR region of Pdcd4 encompassing the aforementioned miR-21 target site, which was co-transfected together with different LNA-antimiRs and pre-miR-21 (10 nM) into Huh-7 cells.
  • the different LNA-antimiRs were; SEQ ID #3205 (8-mer, perfect match), SEQ ID #3218 (8-mer, mismatch) and SEQ ID #3204 (15-mer, DNA/LNA mixmer). Luciferase measurements were performed after 24 hours.
  • Cell line The human hepatoma cell line Huh-7 was a kind gift from R. Bartinschlager (Dept MoI Virology, University of Heidelberg). Huh-7 cells were cultured in DMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax, 1x NEAA and 25 ug/ml Gentamicin. Transfection: 11.000 cells were seeded per well in a 96-well plate the day before transfection in order to achieve 50-70% confluency the next day.
  • Huh-7 cells were transfected with 20 ng Pdcd4 3 ' UTR/psiCHECK2 or empty psiCHECK2 vector together with 10 nM pre-miR-21 (Ambion) and 0,14 ⁇ l Lipofectamine2000 (Invitrogen) according to the manufacturer's instructions. Varying concentrations of the LNA-antimiR oligonucleotides were also transfected. After 24 hours, cells were harvested for luciferase measurements. Luciferase assay: Cells were washed and 30 ⁇ l 1 x Passive Lysis Buffer (Promega) was added to each well, after which the 96-well plates was put on an orbital shaker.
  • Luciferase Assay Buffer Il Dual-Luciferase Reporter Assay System from Promega, Cat# E1910
  • Example 23 Assessment of Pdcd4 protein levels as a functional readout for miR-21 antagonism by the 8-mer LNA-antimiR (SEQ ID #3205).
  • HeLa The human cervix carcinoma cell line HeLa was purchased from ECACC (#93021013). HeLa cells were cultured in EMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax, 1x NEAA and 25 ug/ml Gentamicin.
  • Example 24 Assessment of potential hepatotoxicity of the 8-mer perfect match LNA- antimiR SEQ ID #3205 and the LNA mismatch control SEQ ID #3218.
  • Each compound was injected into female NMRI mice, at doses of 25 mg/kg, 5 mg/kg and 1 mg/kg, every other day for 2 weeks. The animals were sacrificed and serum was collected from whole blood for ALT and AST analyses. As seen in Figure 23, the ALT and AST levels were not elevated for SEQ ID #3205 compared to saline or SEQ ID #3218 (mismatch control). However, one mouse showed increased levels (marked red), since the serum samples were contaminated with red blood cells, which contain 6-8 times higher levels of ALT and AST compared to plasma. The mice that received 5 mg/kg and 1 mg/kg were also analyzed for ALT and AST levels and showed no changes compared to saline treated control animals (data not shown). Materials and Methods: Experimental design:
  • ALT and AST measurements were performed in 96-well plates using ALT and AST reagents from ABX Pentra (A11 A01627 - ALT, A11 A01629 - AST) according to the manufacturer's instructions.
  • Example 25 Assessment of PU.1 protein levels as a functional readout for miR-155 antagonism by short LNA-antimiR (SEQ ID #3207).
  • Cell line The human monocytic THP-1 cell line was purchased from ECACC (#88081201 ).
  • THP-1 cells were cultured in RPMI with L-glutamine, supplemented with 10% fetal bovine serum.
  • Transfection 200.000 cells were seeded per well in a 12-well plate the day before. On the day of transfection, THP-1 cells were transfected with 5 nmol pre-miR-155 (Ambion) and/or 5 nM LNA-antimiR together with Lipofectamine2000 (Invitrogen) according to the manufacturer's instructions. LPS (100 ng/ml) was added to the cells after the 4 hour incubation with the transfection complexes. After 24 hours, cells were harvested for protein extraction and western blot analysis.
  • TRIS gel TRIS gel.
  • the proteins were transferred to a nitrocellulose membrane using iBlot (Invitrogen) according to manufacturers instructions The membrane was incubated overnight at 4 0 C with the rabbit monoclonal PU.1 antibody (Cell Signaling) with a 1:2000 concentration. As equal loading, Tubulin (Thermo Scientific) was used at a 1 :5000 dilution. Immunoreactive bands were visualized with ECL Plus (Amersham).
  • Example 26 Assessment of p27 protein levels as a functional readout for antagonism of the miR-221/222 family by the 7-mer SEQ ID #3225 LNA-antimiR. Previous work has shown (Ie Sage et al. 2007, Galardi et al. 2007) that miR-221 and miR-222 post-transcriptionally regulate the expression of the tumour suppressor gene p27, which is involved in cell cycle regulation. In these studies, down-regulation of miR-221 and miR-222 was shown to increase expression levels of p27.
  • LNA-antimiR we determined the protein levels of p27 after transfection of the LNA-antimiR SEQ ID #3225 into PC3 cells.
  • PC3 cells were cultured in DMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax and 25 ug/ml Gentamicin.
  • Lipofectamine2000 Cells were harvested after 24 hours for protein extraction and western blot analysis.
  • Cell line The human prostate carcinoma PC3 cell line was purchased from ECACC
  • PC3 cells were cultured in DMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax and 25 ug/ml Gentamicin. Transfection: 250.000 cells were seeded per well in a 6-well plate the day before transfection in order to receive 50% confluency the next day. On the day of transfection, PC3 cells were transfected with 25 nM of different LNA oligonucleotides with Lipofectamine2000. Clonogenic growth in soft agar: 2.5x10 3 PC3 cells were seeded in 0.35% agar on the top of a base layer containing 0.5% agar. Cells were plated 24 hours after transfection. Plates were incubated in at 37°C, 5%CO 2 in a humified incubator for 12 days and stained with 0.005% crystal violet for 1 h, after which cells were counted. The assay was performed in triplicate.
  • Example 28 Assessment of let-7 antagonism by 6-9-mer LNA-antimiRs in Huh-7 cells transfected with let-7a precursor miRNA, and a luciferase sensor assay.
  • a luciferase sensor construct was made, containing some 800 bp of the HMGA2 3 1 UTR.
  • the sequence cloned into the vector contains four out of seven functional let-7 binding sites (sites 2-5), as previously demonstrated by Mayr et al. (Science, 2007) and Lee and Dutta (Genes Dev, 2007).
  • the hepatocellular carcinoma cell line Huh-7 (with low to non-existing levels of endogenous let-7) was transfected with the luciferase sensor construct, with let-7a precursor miRNA, and with the 6-9 mer let-7 antagonists SEQ ID #3232, -3233, -3227, -3234, -3235; see Figure 27) at increasing concentrations.
  • the 6-9-mer LNA-antimiRs were compared with SEQ ID #3226, a 15-mer against let-7a as a positive control. After 24 hours, luciferase activity was measured.
  • Huh-7 The hepatocellular carcinoma cell line Huh-7 was a kind gift from R. Bartinschlager (Dept MoI Virology, University of Heidelberg).Huh-7 cells were cultured in DMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax, 1x NEAA and 25 ug/ml Gentamicin.
  • Transfection 8,000 cells were seeded per well in a 96-well plate the day before transfection in order to receive 60-80% confluency the next day.
  • Huh-7 cells in each well were transfected with 20 ng HMGA2 3'UTR/psiCHECK2 plasmid, let-7a precursor miRNA (Dharmacon; 10 nM end-concentration), LNA-antimiRs SEQ ID #3232, -3233, -3227, - 3234, -3235, -3226; 0-50 nM end concentrations) together with 0.17 ⁇ l Lipofectamine2000 (Invitrogen) according to manufacturer's instructions. After 24 hours, cells were harvested for luciferase measurements.
  • Luciferase assay Growth media was discarded and 30 ⁇ l 1x Passive Lysis Buffer (Promega) was added to each well. After 15-30 minutes of incubation on an orbital shaker, renilla and firefly luciferase measurements were performed according to manufacturer's instructions (Promega).
  • Example 29 Assessment of entire let- 7 family antagonism by 8-, and 15-mer LNA- antimiRs in Huh-7 cells transfected with a luciferase sensor assay.
  • the positive control 15-mer shows potent antagonism of all precursors and gives nearly full de-repression at 0.5 nM.
  • the de-repression of luciferase activity by both the 8- and the 15-mer LNA-antimiRs is clearly dose-dependent, as seen in all four panels (Fig 29A-D).
  • the 8-mer LNA-antimiR (SEQ ID #3227), is a potent antagonist against four representative let-7 family members in vitro, and thus likely against the entire family. Compared to a 15-mer positive control antagonist, SEQ ID #3226, the 8-mer is equally potent for three of four targets, and slightly less potent for the fourth target, let-7e, explained by a terminal mismatch in this case. Materials and Methods:
  • Ce// line The hepatocellular carcinoma cell line Huh-7 was a kind gift from R. Bartinschlager (Dept MoI Virology, University of Heidelberg). Huh-7 cells were cultured in DMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax, 1x NEAA and 25 ug/ml Gentamicin.
  • Transfection 8,000 cells were seeded per well in a 96-well plate the day before transfection in order to receive 60-80% confluency the next day.
  • Huh-7 cells in each well were transfected with 20 ng HMGA2 3'UTR/psiCHECK2 plasmid, with let-7a, -7d, -7e, or -7i precursor miRNA (Dharmacon; 10 nM end-concentration), and with LNA-antimiRs SEQ ID #3227 and SEQ ID #3226; 0-50 nM end concentrations) together with 0.17 ⁇ l
  • Lipofectamine2000 (Invitrogen) according to manufacturer's instructions. After 24 hours, cells were harvested for luciferase measurements.
  • Luciferase assay Growth medium was discarded and 30 ⁇ l 1x Passive Lysis Buffer (Promega) was added to each well. After 15-30 minutes of incubation on an orbital shaker, renilla and firefly luciferase measurements were performed according to manufacturer's instructions (Promega).
  • Example 30 Assessment of endogenous let-7 antagonism by SEQ ID #3227, an 8-mer LNA-antimiRs, in HeLa cells transfected with a luciferase sensor assay.
  • SEQ ID #3227 an 8-mer LNA-antimiRs
  • the same luciferase sensor construct as described in previous two examples was co-transfected with SEQ ID #3227 into the cervical cancer cell line HeLa (that expresses moderate to high levels of let-7 as determined by Q-PCR; data not shown).
  • Empty psiCHECK-2 vector was included as a negative control.
  • the cervical cancer cell line HeLa was purchased from ATCC (#CCL-2TM). HeLa cells were cultured in Eagle's MEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax, 1x NEAA and 25 ug/ml Gentamicin.
  • Transfection 8,000 cells were seeded per well in a 96-well plate the day before transfection in order to receive 50-70% confluency the next day. On the day of transfection, HeLa cells in each well were co-transfected with 20 ng HMGA2 3'UTR/psiCHECK2 plasmid or psiCHECK-2 (empty vector), and with LNA-antimiR SEQ ID #3227 (0-50 nM, end concentrations) together with 0.17 ⁇ l Lipofectamine2000 (Invitrogen) according to manufacturer's instructions. After 24 hours, cells were harvested for luciferase measurements.
  • Luciferase assay Growth media was discarded and 30 ⁇ l 1x Passive Lysis Buffer (Promega) was added to each well. After 15-30 minutes of incubation on an orbital shaker, renilla and firefly luciferase measurements were performed according to manufacturer's instructions (Promega).
  • Example 31 Assessment of miR-21 antagonism by an 8-mer LNA-antimiR-21 (#3205) versus an 8-mer (#3219) scrambled control LNA in the human colon carcinoma cell line HCT116.
  • HCT116 cells were transfected with the luciferase constructs together with the miR-21 antagonist #3205 (8-mer) and for comparison of specificity with the 8-mer LNA scrambled control (#3219). After 24 hours, luciferase activity was measured. Results: The luciferase reporter experiments showed a dose-dependent de-repression of the luciferase miR-21 reporter activity with the 8-mer LNA-antimiR against miR-21 (#3205) and complete de-repression was obtained at 5 nM ( Figure 31 ).
  • HCT116 cell line The human colon carcinoma HCT116 cell line was purchased from ATCC (CCL-247). HCT116 cells were cultured in RPMI medium, supplemented with 10% fetal bovine serum, and 25 ug/ml Gentamicin. Transfection: 110.000 cells were seeded per well in a 12-well plate and transfection was performed. HCT116 cells were transfected with 0.3 ⁇ g miR-21 luciferase sensor plasmid or empty psiCHECK2 vector together with 1.2 ⁇ l Lipofectamine2000 (Invitrogen) according to the manufacturer's instructions. Transfected were also varying concentrations of LNA-antimiR and control oligonucleotides.
  • Luciferase assay The cells were washed with PBS and 250 ⁇ l 1 x Passive Lysis Buffer (Promega) was added to the wells. The plates were placed on a shaker for 30 min., after which the cell lysates were transferred to eppendorf tubes. The cell lysate was centrifugated for 10 min at 2.500 rpm after which 50 ⁇ i were transferred to a 96 well plate and luciferase measurements were performed according to the manufacturer's instructions (Promega).
  • Example 32 Knock-down of miR-21 by the 8-mer #3205 LNA-antimiR reduces colony formation of PC3 cells.
  • PC3 cells were cultured in DMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax and 25 ug/ml Gentamicin.
  • Transfection 250.000 cells were seeded per well in a 6-well plate the day before transfection in order to receive 50% confluency the next day. On the day of transfection, PC3 cells were transfected with 25 nM of different LNA oligonucleotides with Lipofectamine2000.
  • Clono ⁇ enic growth in soft agar 2.5x10 3 PC3 cells were seeded in 0.35% agar on the top of a base layer containing 0.5% agar. Cells were plated 24 hours after transfection. Plates were incubated in at 37°C, 5% CO 2 in a humified incubator for 12 days and stained with 0.005% crystal violet for 1 h, after which cells were counted. The assay was performed in triplicate.
  • miR-21 is overexpressed in the human hepatocellular carcinoma cell line HepG2 and we have previously shown that we are able to regulate the luciferase activity of a miR-21 sensor plasmid with #3205 in these cells.
  • HepG2 cells were transfected with #3205 and #3219 (scrambled 8-mer), and after 24 hours plated into soft agar. Colonies were counted after 17 days with a microscope.
  • HepG2 cell line The human hepatocytic HepG2 cell line was purchased from ECACC (#85011430). HepG2 cells were cultured in EMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax and 25 ug/ml Gentamicin.
  • Transfection 650.000 cells were seeded per well in a 6-well plate and reverse transfection was performed. HepG2 cells were transfected with 0.6 ⁇ g miR-21 luciferase sensor plasmid or empty psiCHECK2 vector together with 2,55 ⁇ l Lipofectamine2000 (Invitrogen) according to the manufacturer's instructions. Transfected were also LNA-antimiR and control oligonucleotides as varying concentrations. After 24 hours, the cells were harvested for luciferase measurements. Clonogenic growth in soft agar: 2.0x10 3 HepG2 cells were seeded in 0.35% agar on the top of a base layer containing 0.5% agar. Cells were plated 24 hours after transfection. Plates were incubated in at 37°C, 5% CO 2 in a humified incubator for 17 days and stained with 0.005% crystal violet for 1 h, after which cells were counted. The assay was performed in triplicate.
  • Example 34 Silencing of miR-21 by the 8-mer #3205 LNA-antimiR inhibits cell migration in PC3 cells.
  • PC3 cells had been treated with 25 nM #3205 (perfect match, miR-21), the control #3219 or left untransfected. Pictures were taken after 24 hours, and the area was calculated for the wound closure at respective time-point. The wound closure for the untransfected cells and for the control, #3219, was faster as compared to our LNA-antimiR against miR-21 , #3205, indicating that #3205 inhibits miR-21 and prevents the cells from migrating ( Figure 34B). Conclusion: The 8-mer (#3205) targeting miR-21 inhibits the cell migration of PC3 cells compared to untransfected and control transfected cells. Materials and Methods:
  • PC3 cell line The human prostate carcinoma PC3 cell line was purchased from ECACC (#90112714). PC3 cells were cultured in DMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax and 25 ug/ml Gentamicin. Scratch assay: 150.000 cells were seeded per well in a 6-well plate three days before transfection in order to receive 100% confluency the next day. At 24 hours after transfection, a scratch was made in the cell monolayer with a 200 ⁇ l tip. Pictures were taken at 0 h and after 24 hours by using a digital camera coupled to a microscope. The software program Image J was used to determine wound closure.
  • Example 35 Length assessment of fully LNA-substituted LNA-antimiRs antagonizing miR-155.
  • LNA-antimiRs LNA-antimiRs, and showed that the most potent LNA-antimiRs are 7-, 8- or 9 nt in length.
  • the same experiment was repeated with miR-155.
  • a perfect match target site for miR-155 was cloned into the 3'UTR of the luciferase gene in the reporter plasmid psiCHECK2 and transfected into the mouse RAW macrophage cell line together with fully LNA-substituted LNA-antimiRs of different lengths. Because the endogenous levels of miR-155 are low in the RAW cell line, the cells were treated with 100 ng/ml LPS for 24 hours in order to induce miR-155 accumulation. After 24 hours, luciferase analysis was performed.
  • RAW 264.7 cell line The mouse macrophage RAW 264.7 cell line was purchased from ATCC (TIB-71 ). RAW cells were cultured in DMEM medium, supplemented with 10% fetal bovine serum, 4 mM Glutamax and 25 ug/ml Gentamicin. Transfection: 500.000 cells were seeded per well in a 6-well plate the day before transfection in order to receive 50% confluency the next day. On the day of transfection, RAW 264.7 cells were transfected with 0.3 ug miR-155 perfect match/psiCHECK2 or empty psiCHECK2 vector together with 10 ⁇ l Lipofectamine2000 (Invitrogen) according to the manufacturer's instructions.
  • LPS 100 ng/ml was added to the RAW cells after the 4 hour incubation with the transfection complexes. After another 24 hours, cells were harvested for luciferase measurements.
  • Luciferase assay The cells were washed with PBS and harvested with cell scraper, after which cells were spinned for 5 min at 2.500 rpm. The supernatant was discarded and 50 ⁇ l 1 x
  • Passive Lysis Buffer (Promega) was added to the cell pellet, after which cells were put on ice for 30 min. The lysed cells were spinned at 10.000 rpm for 30 min after which 20 ⁇ l were transferred to a 96-well plate and luciferase measurements were performed according to the manufacturer's instructions (Promega).
  • Example 36 Plasma protein binding for the fully LNA-substituted 8-mer #3205 targeting miR-21 (LNA-antimiR-21).
  • the plasma proteins are not saturated with #3205 at the plasma concentrations in the experiment shown in Figure 36A.
  • #3205 concentrations in the plasma the protein binding is around 95% of the #3205 LNA-antimiR-21 in Figure 36B.
  • #3205 concentrations 50.1 ⁇ M (174 ⁇ g/mL) the binding capacity of plasma proteins for FAM-labeled #3205 has not been saturated.
  • Mouse plasma 100 ⁇ L was spiked with FAM-labeled #3205 to 0.167, 1.67, 5.01 , 10.02, 16.7, 25.05 and 50.1 ⁇ M concentrations. The solutions were incubated at 37°C for 30 minutes. The solutions were transferred to a Microcon Ultracel YM-30 filter (regenerated cellulose 30.000 MWCO). The filters were spun for 20 minutes at 200Og and at room temperature in a microcentrifuge. The filtrate was diluted 5, 10 and 20 times and 100 ⁇ L samples were transferred to a microtiter plate (Polystyrene Black NUNC-237108).
  • the fluorescence was detected using a FLUOstar Optima elisa reader with excitation 458 nm and emission 520 nm.
  • the amount of unbound FAM-labeled #3205 was calculated from a standard curve derived from filtrated plasma spiked with FAM-labeled #3205 at 12 different (0.45 - 1000 nM) concentrations. The numbers were corrected with the recovery number established from filtration experiments with #3205 concentrations 0.167, 1.67, 5.01 , 10.02, 16.7, 25.05 and 50.1 ⁇ M in filtrated plasma. The recovery of FAM-labeled #3205 was 86%.
  • Example 37 Quantitative whole body autoradiography study in female pigmented mice after single intravenous administration of 35 S-labelled #3205 LNA-antimiR-21.
  • Table 6(ii) Tissue to liver ratios after single intravenous administration of 35 S- labelled #3205 in female pigmented mice.
  • #3205 shows blood clearance of radioactivity with elimination half-lives of 8-10 hours. High levels of radioactivity were registered in the kidney cortex, lymph, liver, bone marrow, spleen, ovary and uterus. The highest level of radioactivity was registered in the kidney cortex showing five times higher levels than that of the liver for #3205. A strong retention of radioactivity was noticed in the kidney cortex, lymph, liver, bone marrow and spleen for #3205 LNA-antimiR-21. Materials and Methods:
  • mice All mice were weighed before administration. Nine female mice were given 10 mg/kg of 35 S-#3205 intravenously in a tail vein. The volume given to each animal was 10 mL/kg of the test formulation. The specific activity 75.7 ⁇ Ci/mg. Individual mice were killed 5 min, 15 min, 1 hour, 4 hours, 24 hours, 2 days, 4 days, 7 days and 21 days after administration of #3205.
  • Whole body autoradiography The mice were anaesthetized by sevoflurane, and then immediately immersed in heptane, cooled with dry ice to -80 0 C, ABR-SOP-0130.
  • the frozen carcasses were embedded in a gel of aqueous carboxymethyl cellulose (CMC), frozen in ethanol, cooled with dry ice (-80 0 C) and sectioned sag ittaly for whole body autoradiography, according to the standard method, ABR-SOP-0131. From each animal 20 ⁇ m sections were cut at different levels with a cryomicrotome (Leica CM 3600) at a temperature of about -20°C. The obtained sections were caught on tape (Minnesota Mining and Manufacturing Co., No. 810) and numbered consecutively with radioactive ink.
  • CMC carboxymethyl cellulose
  • Pharmacokinetic calculations The 35 S radioactivity measured in whole blood and tissues was expressed as nCi/g tissue and recalculated to nmol equiv/g tissue for the pharmacokinetic evaluation.
  • the pharmacokinetic parameters C max , t 1/2 and AUC were determined for the whole blood and tissues by non-compartmental analysis using WinNonlin Professional (Pharsight
  • Example 38 Assessment of let-7 inhibition in vivo by an 8-mer LNA-antimiR, as determined through Ras protein quantification in mouse lung and kidney
  • mice were intravenously (i.v.) injected with an 8-mer LNA-antimiR antagonist or with saline.
  • proteins were isolated from lungs and kidneys. Because the Ras family of proteins (N-Ras, K-Ras, and H-Ras), in particular N-Ras and K-Ras, has previously been shown to be regulated (repressed) by the let-7 family by Johnson et al. (Cell, 2005), the aim was to analyze whether these let-7 targets could be de-repressed in vivo.
  • Neomarkers and a secondary HRP-conjugated goat-anti-mouse antibody (P0447, Dako) was used.
  • Example 40 In vivo efficacy assessment of the 8-mer LNA-antimiR (#3205) in targeting miR-21, as determined by Pdcd4 protein up-regulation in mouse kidney.
  • 8-mer LNA-antimiR that is fully LNA-modified antagonizes miR-21 and has the ability to regulate the protein levels of the miR-21 target Pdcd4 in vitro.
  • the mice received 25 mg/kg of #3205 by i.v. injection every other day for 14 days (a total of 5 doses). The mice were sacrificed on day 14, the kidney was removed, and protein was isolated. In order to determine target regulation, Western blot analysis was performed.
  • mice with #3205 showed significantly increased Pdcd4 protein levels as compared to the saline control. While the normalized Pdcd4 versus Gapdh ratio was consistent in both saline samples, the protein up-regulation in the two LNA- antimiR-treated (#32059 mice were measured to 3.3- and 6.3-fold, respectively, demonstrating an in vivo pharmacological effect of the #3205 8-mer LNA-antimiR.
  • the animals were dozed with LNA-antimiR or saline (0.9% NaCI), receiving an injection of 25 mg/kg every other day for 14 days, a total of 5 doses. Animals were sacrificed on day 14.
  • Western blot analysis 80 ⁇ g kidney tissue from saline or LNA-treated mice was separated on NuPAGE Bis Tris 4-12% (Invitrogen). The proteins were transferred to a nitrocellulose membrane using iBlot (Invitrogen) according to the manufacturer's instructions. The membrane was incubated with Pdcd4 antibody (Bethyl Laboratories), followed by HRP-conjugated swine- anti-rabbit antibody (Dako). As equal loading control, GAPDH (Abeam) was used, followed by HRP-conjugated swine-anti-mouse antibody. The membranes were visualized by chemiluminiscence (ECL, Amersham).

Landscapes

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

Abstract

The present invention relates to very short heavily modified oligonucleotides which target and inhibit microRNAs in vivo, and their use in medicaments and pharmaceutical compositions.

Description

MICROMIRs
FIELD OF THE INVENTION
The present invention relates to very short oligonucleotides which target and inhibit microRNAs in vivo, and their use in medicaments and pharmaceutical compositions.
BACKGROUND OF THE INVENTION
MicroRNAs (miRNAs) are an abundant class of short endogenous RNAs that act as post- transcriptional regulators of gene expression by base-pairing with their target mRNAs. They are processed from longer (ca 70-80 nt) hairpin-like precursors termed pre-miRNAs by the RNAse III enzyme Dicer. MicroRNAs assemble in ribonucleoprotein complexes termed miRNPs and recognize their target sites by antisense complementarity thereby mediating down-regulation of their target genes. Near-perfect or perfect complementarity between the miRNA and its target site results in target mRNA cleavage, whereas limited complementarity between the microRNA and the target site results in translational inhibition of the target gene.
A summary of the role of_microRNAs in human diseases, and the inhibition of microRNAs using single stranded oligonucleotides is provided by WO2007/112754 and WO2007/112753, which are both hereby incorporated by reference in its entirety. WO2008046911, hereby incorporated by reference, provides microRNA sequences which are associated with cancer. Numerous microRNAs have been associated with disease phenotypes and it is therefore desirable to provide substances capable of modulating the availability of microRNAs in vivo. WO2007/112754 and WO2007/112753 disclose short single stranded oligonucleotides which are considered to form a strong duplex with their target miRNA. SEQ ID NOs 1 - 45 are examples of anti microRNA oligonucleotides as disclosed in WO2007/112754 and WO2007/112753.
RELATED APPLICATIONS This application claims priority from four applications: US 60/977497 filed 4th October 2007, US 60/979217 filed 11th October 2007, US 61/028062, filed 12 February 2008, and EP08104780, filed 17th July 2008, all of which are hereby incorporated by reference. Furthermore we reference and incorporate by reference WO2007/112754 and WO2007/112753 which are earlier applications from the same applicants.
SUMMARY OF THE INVENTION
The present invention is based upon the discovery that the use of very short oligonucleotides which target microRNAs and which have a high proportion of nucleotide analogue nucleotides, such as LNA nucleotides, are highly effective in alleviating the repression of RNAs, such as an mRNA, by the targeted microRNAs in vivo.
The present invention provides an oligomer a contiguous sequence of 7, 8, 9 or 10 nucleotide units in length, for use in reducing the effective amount of a microRNA target in a cell or an organism, wherein at least 70%, such as at least 80% of the nucleotide units of the oligomer are selected from the group consisting of LNA units and 2' substituted nucleotide analogues.
The present invention provides an oligomer a contiguous sequence of 7, 8, 9 or 10 nucleotide units in length, for use in reducing the effective amount of a microRNA target in a cell or an organism, wherein at least 70% of the nucleotide units of the oligomer are selected from the group consisting of LNA units and 2' substituted nucleotide analogues, and wherein at least 50%, such as at least 60%, such as at least 70% of the nucleotide units of the oligomer are LNA units.
The invention provides oligomers of between 7-10 nucleotides in length which comprises a contiguous nucleotide sequence of a total of between 7-10 nucleotides, such as 7, 8, 9, nucleotide units, wherein at least 50% of the nucleotide units of the oligomer are nucleotide analogues.
The invention further provides for an oligomer of between 7-10 nucleotides in length which comprises a contiguous nucleotide sequence of a total of between 7-10 nucleotides, such as 7, 8, 9, or 10, nucleotide units, wherein the nucleotide sequence is complementary to a corresponding nucleotide sequence found in mammalian or viral microRNA, and wherein at least 50% of the nucleotide units of the oligomer are nucleotide analogues.
The present invention provides olgiomers according to the invention as a medicament.
The present invention provides pharmaceutical compositions comprising the oligomer of the invention and a pharmaceutically acceptable diluent, carrier, salt or adjuvant.
The invention provides for a conjugate comprising an oligomer according to the invention, conjugated to at least one non-nucleotide or polynucleotide entity, such as a sterol, such as cholesterol.
The invention provides for the use of an oligomer or a conjugate according to the invention, for the manufacture of a medicament for the treatment of a disease or medical disorder associated with the presence or over-expression of a microRNA, such as one or more of the microRNAs referred to herein.
The invention provides for the treatment of a disease or medical disorder associated with the presence or overexpression of the microRNA, comprising the step of administering a composition (such as the pharmaceutical composition) comprising an oligomer or conjugate according to the invention to a patient suffering from or likely to suffer from said disease or medical disorder. The invention provides for a method for reducing the effective amount of a microRNA target in a cell or an organism, comprising administering the oligomer of the invention, or a composition (such as a pharmaceutical composition) comprising the oligomer or conjugate according to the invention to the cell or organism. The invention provides for a method for reducing the effective amount of a microRNA target in a cell or an organism, comprising administering the oligomer or conjugate or pharmaceutical composition according to the invention to the cell or organism.
The invention provides for a method for de-repression of a target mRNA (or one ore mor RNAs) in a cell or an organism, comprising administering an oligomer or conjugate according to the invention, or a composition comprising said oligomer or conjugate, to said cell or organism.
The invention provides for the use of an oligomer or a conjugate according to the invention, for inhibiting the mircoRNA in a cell which comprises said microRNA, such as a human cell. The use may be in vivo or in vitro.
BRIEF DESCRIPTION OF THE DRAWINGS Figure 1. Schematic presentation of the miR-21, miR-155 and miR-122 8-mer LNA-antimiRs, indicating the targeting positions with the fully LNA-modified and phosphorothiolated LNA- antimiR. Preferred hybridisation positions for 7mer, 8mer, 9mer and 10mer LNA oligonucleotides on the mature microRNA are also indicated. Figure 2. Assessment of miR-21 antagonism by SEQ ID #3205 and SEQ ID #3204 LNA- antimiRs in MCF-7 cells using a luciferase sensor assay. MCF-7 cells were co-transfected with luciferase sensor plasmids containing a perfect match target site for miR-21 or a mismatch target site (.mm2) and LNA-antimiRs at different concentrations. After 24 hours, cells were harvested and luciferase activity measured. Shown are the mean of renilla/firefly ratios for three separate experiments (bars = s.e.m), were all have been normalized against 0 nM psiCHECK2 (=control).
Figure 3. Assessment of miR-21 antagonism by SEQ ID #3205 and SEQ ID #3204 LNA- antimiRs in HeLa cells using a luciferase sensor assay. HeLa cells were co-transfected with luciferase sensor plasmids containing a perfect match target site for miR-21 (mir-21 ) or a mismatch target site (mm2) and LNA-antimiRs at different concentrations. After 24 hours, cells were harvested and luciferase activity measured. Shown are the mean of renilla/firefly ratios for three separate experiments (bars = s.e.m), were all have been normalized against 0 nM psiCHECK2 (^control).
Figure 4. Assessment of miR-155 antagonism by SEQ ID #3206 and SEQ ID #3207 LNA- antimiRs in LPS-treated mouse RAW cells using a luciferase sensor assay. RAW cells were co- transfected with miR-155 and the different LNA-antimiRs at different concentrations. After 24 hours, cells were harvested and luciferase activity measured. Shown are the mean of renilla/firefly, were all have been normalized against 0 nM psiCHECK2. Figure 5. Assessment of miR-122 antagonism by SEQ ID #3208 and SEQ ID #4 LNA-antimiRs in HuH-7 cells using a luciferase sensor assay. HuH-7 cells were co-transfected with a miR-122 luciferase sensor containing a perfect match miR-122 target site and the different LNA-antimiRs at different concentrations. After 24 hours, cells were harvested and luciferase activity measured. Shown are the mean of renilla/firefly ratios for three separate experiments (bars = s.e.m), where all have been normalized against 0 nM psiCHECK2 (=control). Figure 6. Schematic presentation of the miR-21 luciferase reporter constructs. Figure 7. Assessment of miR-21 antagonism by an 8-mer LNA-antimiR (SEQ ID #3205) versus a 15-mer LNA-antimiR (SEQ ID #3204) in PC3 cells using a luciferase reporter assay. PC3 cells were co-transfected with luciferase reporter plasmids containing a perfect match target site for miR-21 or a mismatch target site and LNA-antimiRs at different concentrations. After 24 hours, cells were harvested and luciferase activity measured. Shown are the mean values (bars=s.e.m) of three independent experiments where the renilla/firefly ratios have been normalized against 0 nM empty vector without target site (=control). Shown is also a schematic presentation of the miR-21 sequence and the design and position of the LNA-antimiRs. LNA nucleotides are indicated by ovals, and DNA residues are indicated by bars. Figure 8. Specificity assessment of miR-21 antagonism by an 8-mer LNA-antimiR in HeLa cells using a luciferase reporter assay. HeLa cells were co-transfected with luciferase reporter plasmids containing a perfect match or a mismatched target site for miR-21 and LNA-antimiRs (SEQ ID #3205) or an 8-mer LNA mismatch control oligo (SEQ ID #3218) at different concentrations. After 24 hours, cells were harvested and luciferase activity was measured. Shown are the mean values (bars=s.e.m) for three independent experiments where the Renilla/firefly ratios have been normalized against 0 nM empty vector without target site
(=control). Shown is also a schematic presentation of the miR-21 sequence and the design and position of the LNA-antimiRs. Mismatches are indicated by filled ovals. Figure 9. Assessment of the shortest possible length of a fully LNA-modified LNA-antimiR that mediates effective antagonism of miR-21. HeLa cells were co-transfected with luciferase reporter plasmids containing a perfect match or a mismatch target site for miR-21 and the LNA- antimiRs at different concentrations (SEQ ID #3209 =6-mer and SEQ ID #3210=7-mer). After 24 hours, cells were harvested and luciferase activity measured. Shown are the mean values (bars=s.e.m) for three independent experiments where the renilla/firefly ratios have been normalized against 0 nM empty vector without target site (=control). Shown is also a schematic presentation of the miR-21 sequence and the design and position of the LNA-antimiRs.
Figure 10. Length assessment of fully LNA-substituted LNA-antimiRs antagonizing miR-21. HeLa cells were co-transfected with luciferase reporter plasmids containing a perfect match or a mismatch target site for miR-21 and LNA-antimiRs at different concentrations (SEQ ID #3211 =9-mer, SEQ ID #3212=10-mer, SEQ ID #3213=12-mer and SEQ ID #3214=14-mer). After 24 hours, cells were harvested and luciferase activity measured. Shown are the mean values (bars=s.e.m) for three independent experiments where the renilla/firefly ratios have been normalized against 0 nM empty vector without target site (=control). Shown is also a schematic presentation of the miR-21 sequence and the design and position of the LNA-antimiRs. Figure 11. Determination of the most optimal position for an 8-mer LNA-antimiR within the miR target recognition sequence. HeLa cells were co-transfected with luciferase reporter plasmids containing a perfect match or a mismatch target site for miR-21 and the LNA-antimiRs at different concentrations. After 24 hours, cells were harvested and luciferase activity measured. Shown are the mean values (bars=s.e.m) for three independent experiments where the renilla/firefly ratios have been normalized against 0 nM empty vector without target site (=control). Shown is also a schematic presentation of the miR-21 sequence and the design and position of the LNA-antimiRs. Figure 12. Validation of interaction of the Pdcd4-3'-UTR and miR-21 by the 8-mer SEQ ID #3205 LNA-antimiR. HeLa cells were co-transfected with a luciferase reporter plasmid containing part of the 3'UTR of Pdcd4 gene and LNA-antimiRs at different concentrations (SEQ ID #3205 = 8 mer, perfect match; SEQ ID #3218 = 8 mer, mismatch; SEQ ID #3204 = 15 mer, LNA/DNA mix; SEQ ID #3220 = 15 mer, gapmer). After 24 hours, cells were harvested and luciferase activity measured. Shown are renilla/firefly ratios that have been normalized against 0 nM. Shown is also a schematic presentation of the miR-21 sequence and the design and position of the LNA-antimiRs.
Figure 13. Comparison of an 8-mer LNA-antimiR (SEQ ID #3207) with a 15-mer LNA-antimiR (SEQ ID #3206) in antagonizing miR-155 in mouse RAW cells. Mouse RAW cells were co- transfected with luciferase reporter plasmids containing a perfect match for miR-155 and the different LNA-antimiRs at different concentrations. After 24 hours, cells were harvested and luciferase activity measured. Shown are the mean values (bars=s.e.m) of three independent experiments where the renilla/firefly ratios have been normalized against 0 nM empty vector without miR-155 target site (=control). Shown is also a schematic presentation of the miR-155 sequence and the design and position of the LNA-antimiRs.
Figure 14. Assessment of c/EBPDAssessment of c/EBPer LNA-antimiR (SEQ ID #3207) with a 15-mer LNA-antimiR (SEQ ID #3206) in antagonizing miR-155 in mouse RAW cells. Mouse RAW cells were co-transfected with luciferase reporter plasmids containing a perfect match for miR-155 and the diffter 20 hours, cells were harvested and western blot analysis of protein extracts from RAW cells was performed. The different isoforms of c/EBPβ are indicated, and the ratios calculated on c/EBPβ LIP and beta-tubulin are shown below. Figure 15. Antagonism of miR-106b by a fully LNA-modified 8-mer (SEQ ID #3221) LNA- antimiR or by a 15-mer mixmer (SEQ ID #3228) antimiR. HeLa cells were co-transfected with luciferase reporter plasmids containing a perfect match for miR-106b and the different LNA- antimiRs at different concentrations. After 24 hours, cells were harvested and luciferase activity measured. Shown are the mean values of four replicates where the renilla/firefly ratios have been normalized against 0 nM empty vector without miRNA target site (=control). Shown is also a schematic presentation of the miR-106b sequence and the design and position of the LNA- antimiRs. Figure 16. Antagonism of miR-19b by a fully LNA-modified 8-mer (SEQ ID #3222) LNA-antimiR and a 15-mer (SEQ ID #3229) mixmer antimiR. HeLa cells were co-transfected with luciferase reporter plasmids containing a perfect match for miR-19a and the two LNA-antimiRs at different concentrations. After 24 hours, cells were harvested and luciferase activity measured. Shown are the mean values of four replicate experiments, where the renilla/firefly ratios have been normalized against 0 nM empty vector without a miR-19a target site (=control). Shown is also a schematic presentation of the miR-19a sequence and the design and position of the LNA- antimiRs.
Figure 17. Schematic presentation showing the mature human miR-221 and miR-222 sequences. Shown in the square is the seed sequence (7-mer) that is conserved in both miRNA sequences. Figure 18. Targeting of a microRNA family using short, fully LNA-substituted LNA-antimiR. PC3 cells were co-transfected with luciferase reporter plasmids for miR-221 and miR-222 separately or together and with the different LNA-antimiRs at varying concentrations. When co-transfecting with the LNA-antimiRs (15-mers) SEQ ID #3223 (against miR-221 ) and SEQ ID #3224 (against miR-222), the total concentration was 2 nM (1 nM each), while transfecting the cells with SEQ ID #3225 (7-mer) the concentrations were 0, 1 , 5, 10 or 25 nM. After 24 hours, cells were harvested and luciferase activity measured. Shown are the mean values (bars=s.e.m) of three independent experiments where the renilla/firefly ratios have been normalized against 0 nM empty vector without a miRNA target site (=control). Shown is also a schematic presentation of the miR-221/222 sequence and the design and position of the LNA-antimiRs. Figure 19. Assessment of p27 protein levels as a functional readout for antagonism of the miR- 221/222 family by the 7-mer SEQ ID #3225 LNA-antimiR. PC3 cells were transfected with the 7- mer LNA-antimiR SEQ ID #3225 targeting both miR-221 and miR-222 at varying concentrations. After 24 hours, cells were harvested and protein levels were measured on a western blot. Shown are the ratios of p27/tubulin. Figure 20. Assessment of miR-21 antagonism by an 8-mer LNA-antimiR (SEQ ID #3205) versus a 15-mer LNA-antimiR (SEQ ID #3204) and an 8-mer with 2 mismatches (SEQ ID #3218) in HepG2 cells using a luciferase reporter assay. HepG2 cells were co-transfected with luciferase reporter plasmid containing a perfect match target site for miR-21 and LNA-antimiRs at different concentrations. After 24 hours, cells were harvested and luciferase activity measured. Shown are the mean values (bars=s.e.m) of three independent experiments where the renilla/firefly ratios have been normalized against 0 nM empty vector without target site (=control). Shown is also a schematic presentation of the miR- 21 sequence and the design and position of the LNA-antimiRs.
Figure 21. Validation of interaction of the Pdcd4 3'UTR and miR-21 by the 8-mer SEQ ID #3205 LNA-antimiR versus the 15-mer (SEQ ID #3204) and an 8-mer with two mismatches (SEQ ID #3218). Huh-7 cells were co-transfected with a luciferase reporter plasmid containing part of the 3'UTR of Pdcd4 gene, pre-miR-21 (10 nM) and LNA-antimiRs at different concentrations. After 24 hours, cells were harvested and luciferase activity measured. Shown are the mean values (bars=s.e.m) of three independent experiments where the renilla/firefly ratios have been normalized against 0 nM empty vector without target site (=control). Shown is also a schematic presentation of the miR-21 sequence and the design and position of the LNA-antimiRs.
Figure 22. Antagonism of miR-21 by SEQ ID #3205 leads to increased levels of Pdcd4 protein levels.
HeLa cells were transfected with 5 nM LNA-antimiR SEQ ID #3205 (perfect match), or SEQ ID
#3219 LNA scrambled (8mer) or SEQ ID #3218 (8-mer mismatch). Cells were harvested after 24 hours and subjected to Western blot with Pdcd4 antibody.
Figure 23. ALT and AST levels in mice treated with SEQ ID #3205 (perfect match) or SEQ ID #3218 (mismatch control). Mice were sacrificed after 14 days and after receiving 25 mg/kg every other day. Figure 24. Assessment of PU.1 protein levels as a functional readout for miR-155 antagonism by short LNA-antimiR (SEQ ID #3207).
THP-1 cells were co-transfected with pre- miR-155 (5 nmol) and different LNA oligonucleotides (5 nM) and 100 ng/ml LPS was added. After 24 hours, cells were harvested and western blot analysis of protein extracts from the THP-1 cells was performed. PU.1 and tubulin are indicated. Figure 25. Assessment of p27 protein levels as a functional readout for antagonism of the miR- 221/222 family by the 7-mer SEQ ID #3225 LNA-antimiR.
PC3 cells were transfected with the 7-mer LNA-antimiR SEQ ID #3225 targeting both miR-221 and miR-222 and a LNA scrambled control at 5 and 25 nM. After 24 hours, cells were harvested and protein levels were measured on a western blot. Shown are the ratios of p27/tubulin. Figure 26. Knock-down of miR-221 /222 by the 7-mer SEQ ID #3225 (perfect match) LNA- antimiR reduces colony formation in soft agar in PC3 cells.
PC3 cells were transfected with 25 nM of the 7-mer LNA-antimiR SEQ ID #3225 targeting both miR-221 and miR-222 or a 7-mer scrambled control ((SEQ ID #3231). After 24 hours, cells were harvested and seeded on soft agar. After 12 days, colonies were counted. One experiment has been done in triplicate.
Figure 27. Overview of the human let-7 family, and of tested antagonists, (upper) The sequences represent the mature miRNA for each member and the box depicts nucleotides 2-16, the positions typically antagonized by LNA-antimiRs. Columns to the right show the number of nucleotide differences compared to let-7a, within the seed (S: position 2-8), extended seed (ES; position 2-9), and the remaining sequence typically targeted by LNA- antimiRs (NE; position 9-16), respectively. Nucleotides with inverted colors are altered compared to let-7a. (lower) Summary of tested antagonists against the let-7 family, including information on design, length and perfectly complementary targets. All compounds are fully phoshorothiolated.
Figure 28. Assessment of let-7 antagonism by six different LNA-antimiRs in Huh-7 cells using a luciferase sensor assay. Huh-7 cells were co-transfected with luciferase sensor plasmids containing a partial HMGA2 3'UTR (with four let-7 binding sites), with or without let-7a precursor (grey and black bars, respectively), and with 6 different LNA-antimiRs at increasing concentrations. After 24 hours, cells were harvested and luciferase activity measured. Shown are the mean of renilla/firefly ratios for duplicate measurements and standard deviations for each assay. Within each LNA- antimiR group all ratios have been normalized to the average of wells containing no let-7a precursor (black bars).
Figure 29. Luciferase results from Huh-7 cells transfected with the HMGA2 3'UTR sensor plasmid, LNA-antimiRs SEQ ID #3226 (left) and SEQ ID #3227 (right), and pre-miRs for let-7a (A), let-7d (B), let-7e (C), and let-7i (D). Grey bars indicate the target de-repression after pre-mis inclusion, whereas black control bars represent the equivalent level without pre-miR addition. Each ratio is based on quadruplicate measurements and have been normalized against the average of wells containing no precursor (black bars) within each treatment group. Figure 30. Luciferase results from HeLa cells transfected with the HMGA2 3'UTR sensor plasmid or control vector, and the LNA-antimiR SEQ ID #3227 at various concentrations. Each ratio is based on quadruplicate measurements normalized against untreated (0 nM) empty control vector (psi-CHECK-2; grey bars).
Figure 31. Assessment of miR-21 antagonism by 8mer (#3205) in HCT116 cells using a luciferase sensor assay. HCT116 cells were co-transfected with luciferase sensor plasmids containing a perfect match target site for miR-21 (grey bars) and LNA-antimiR and control oigonucleotides at different concentrations. After 24 hours, cells were harvested and luciferase activity measured. Shown is one typical example of two where the renilla/firefly ratios have been normalized against 0 nM empty vector (=black bars). Figure 32. Silencing of miR-21 by the 8-mer #3205 LNA-antimiR reduces colony formation in soft agar in PC3 cells. PC3 cells were transfected with 25 nM of the 8-mer LNA-antimiR #3205 targeting miR-21. After 24 hours, cells were harvested and seeded on soft agar. After 12 days, colonies were counted. Shown is the mean of three separate experiments, each performed in triplicate, and normalised against 0 nM control (i.e. transfection but with no LNA). p=0.01898 for #3205.
Figure 33. Knock-down of miR-21 by the 8-mer #3205 LNA-antimiR reduces colony formation in soft agar in HepG2 cells. HepG2 cells were transfected with 25 nM of the 8-mer LNA-antimiR #3205 targeting miR-21. After 24 hours, cells were harvested and seeded on soft agar. After 17 days, colonies were counted. Shown is the mean of three replicates from one experiment (bars=SEM).
Figure 34. Wound closure in the invasive human prostate cell line PC3 after treatment with #3205. (A) PC3 cells were transfected at day 3 with LNA-antimiR and control oligonucleotides at 25 nM, #3205 (8mer, perfect match) and #3219 (8mer, mismatch) and the following day a scratch was made. Pictures were taken after 24 hours in order to monitor the migration. (B) The area in each timepoint has been measured with the software program Image J and normalized against respective 0 h time-point.
Figure 35. Length assessment of fully LNA-substituted LNA-antimiRs antagonizing miR-155. RAW cells were co-transfected with luciferase reporter plasmids containing a perfect match target site for miR-155 and with LNA-antimiR oligonucleotides at different concentrations. After 24 hours, cells were harvested and luciferase activity measured. Shown are the mean values (bars=s.e.m) for three independent experiments where the renilla/firefly ratios have been normalized against 0 nM empty vector without target site (=mock). Shown is also a schematic presentation of the miR sequence and the design and position of the LNA-antimiRs. Figure 36. Binding of 5'-FAM labeled LNA-antimiR-21 (#3205) to mouse plasma protein. (A)% unbound LNA-antimiR-21 compound as a function of oligonucleotide concentration in mouse plasma. (B) Concentration of unbound LNA-antimiR-21 compound #3205 as a function of #3205 concentration in mouse plasma. Figure 37. Quantification Ras protein levels by Western blot analysis. A. Gel image showing Ras and Tubulin (internal standard) protein in treated (anti-let-7; 8- mer) vs. untreated (saline) lung and kidney samples. B. Quantifications of Ras protein levels in the lung and kidney, respectively, of LNA-antimiR-treated mice (black bars), normalized against equivalent saline controls (grey bars), using tubulin as equal-loading control. B. Silencing of miR-21 by #3205 leads to increased levels of Pdcd4 protein levels in vivo. C. Mice were injected with saline or 25 mg/kg LNA-antimiR (#3205) over 14 days every other day, with a total of 5 doses. Mice were sacrificed and protein was isolated from kidney and subjected to Western blot analysis with Pdcd4 antibody. A. Gel image showing Pdcd4 and Gapdh (internal standard) protein in treated (antimiR-21 ; 8-mer) vs. untreated (saline) kidney samples (M1 , mouse 1 ; M2, mouse 2). B. Quantification of
Pdcd4 protein levels in kidneys of LNA-antimiR-treated mice (dark grey bars), normalized against the average of equivalent saline controls (light grey bars), using Gapdh as loading control.
DETAILED DESCRIPTION OF THE INVENTION Short oligonucleotides which incorporate LNA are known from the in vitro reagents area,
(see for example WO2005/098029 and WO 2006/069584). However the molecules designed for diagnostic or reagent use are very different in design than those for in vivo or pharmaceutical use. For example, the terminal nucleotides of the reagent oligos are typically not LNA, but DNA, and the internucleoside linkages are typically other than phosphorothioate, the preferred linkage for use in the oligonucleotides of the present invention. The invention therefore provides for a novel class of oligonucleotides (referred to herein as oligomers) per se.
The following embodiments refer to certain embodiments of the oligomer of the invention, which may be used in a pharmaceutical composition. Aspects which refer to the oligomer may also refer to the contiguous nucleotide sequence, and vice versa. The Oligomer
The oligomer of the invention is a single stranded oligonucleotide which comprises nucleotide analogues, such as LNA, which form part of, or the entire contiguous nucleotide sequence of the oligonucleotide. The nucleotide sequence of the oligomer consists of a contiguous nucleotide sequence. The term "oligonucleotide" (or simply "oligo"), which is used interchangeably with the term
"oligomer" refers, in the context of the present invention, to a molecule formed by covalent linkage of two or more nucleotides. When used in the context of the oligonucleotide of the invention (also referred to the single stranded oligonucleotide), the term "oligonucleotide" may have, in one embodiment, for example have between 7 - 10 nucleotides, such as in individual embodiments, 7, 8, 9, or 10.
The term 'nucleotide' refers to nucleotides, such as DNA and RNA, and nucleotide analogues. It should be recognised that, in some aspects, the term nucleobase may also be used to refer to a nucleotide which may be either naturally occurring or non-naturally occurring - in this respect the term nucleobase and nucleotide may be used interchangeably herein. In some embodiments, the contiguous nucleotide sequence consists of 7 nucleotide analogues. In some embodiments, the contiguous nucleotide sequence consists of 8 nucleotide H analogues. In some embodiments, the contiguous nucleotide sequence consists of 9 nucleotide analogues.
In one embodiment at least about 50% of the nucleotides of the oligomer are nucleotide analogues, such as at least about 55%, such as at least about 60%, or at least about 65% or at least about 70%, such as at least about 75%, such as at least about 80%, such as at least about 85%, such as at least about 90%, such as at least about 95% or such as 100%. It will also be apparent that the oligonucleotide may comprise of a nucleotide sequence which consists of only nucleotide analogues. Suitably, the oligomer may comprise at least one LNA monomer, such as 2, 3, 4, 5, 6, 7, 8, 9 or 10 LNA monomers. As described below, the contiguous nucleotide sequence may consist only of LNA units (including linkage groups, such as phosphorothioate linkages), or may conists of LNA and DNA units, or LNA and other nucleotide analogues. In some embodiments, the contiguous nucleotide sequence comprises either one or two DNA nucleotides, the remainder of the nucleotides being nucleotide analogues, such as LNA unit. In some embodiments, the contiguous nucleotide sequence consists of 6 nucleotide analogues and a single DNA nucleotide. In some embodiments, the contiguous nucleotide consists of 7 nucleotide analogues and a single DNA nucleotide. In some embodiments, the contiguous nucleotide sequence consists of 8 nucleotide analogues and a single DNA nucleotide. In some embodiments, the contiguous nucleotide sequence consists of 9 nucleotide analogues and a single DNA nucleotide. In some embodiments, the contiguous nucleotide sequence consists of 7 nucleotide analogues and two DNA nucleotides. In some embodiments, the contiguous nucleotide sequence consists of 8 nucleotide analogues and two DNA nucleotides.
The oligomer may consist of the contiguous nucleotide sequence. In a specially preferred embodiment, all the nucleotide analogues are LNA. In a further preferred embodiment, all nucleotides of the oligomer are LNA. In a further preferred embodiment, all nucleotides of the oligomer are LNA and all internucleoside linkage groups are phosphothioate.
Herein, the term "nitrogenous base" is intended to cover purines and pyrimidines, such as the DNA nucleobases A, C, T and G, the RNA nucleobases A, C, U and G, as well as non- DNA/RNA nucleobases, such as 5-methylcytosine (MeC), isocytosine, pseudoisocytosine, 5- bromouracil, 5-propynyluracil, 5-propyny-6-fluoroluracil, 5-methylthiazoleuracil, 6-aminopurine, 2-aminopurine, inosine, 2,6-diaminopurine, 7-propyne-7-deazaadenine, 7-propyne-7- deazaguanine and 2-chloro-6-aminopurine, in particular MeC. It will be understood that the actual selection of the non-DNA/RNA nucleobase will depend on the corresponding (or matching) nucleotide present in the microRNA strand which the oligonucleotide is intended to target. For example, in case the corresponding nucleotide is G it will normally be necessary to select a non-DNA/RNA nucleobase which is capable of establishing hydrogen bonds to G. In this specific case, where the corresponding nucleotide is G, a typical example of a preferred non- DNA/RNA nucleobase is MeC.
It should be recognised that the term in 'one embodiment' should not necessarily be limited to refer to one specific embodiment, but may refer to a feature which may be present in 'some embodiments', or even as a generic feature of the invention. Likewise, the use of the term 'some emboidments' may be used to describe a feature of one specific embodiment, or a collection of embodiments, or even as a generic feature of the invention.
The terms "corresponding to" and "corresponds to" refer to the comparison between the nucleotide sequence of the oligomer or contiguous nucleotide sequence (a first sequence) and the equivalent contiguous nucleotide sequence of a further sequence selected from either i) a sub-sequence of the reverse complement of the microRNA nucleic acid target (such as a microRNA target selected from SEQ ID 40 - SEQ ID 976, and/or ii) the sequence of nucleotides provided herein such as the group consisting of SEQ ID NO 977 - 1913, or SEQ ID NO 1914- 2850, or SEQ ID NO 2851 - 3787. Nucleotide analogues are compared directly to their equivalent or corresponding nucleotides. A first sequence which corresponds to a further sequence under i) or ii) typically is identical to that sequence over the length of the first sequence (such as the contiguous nucleotide sequence).
When referring to the length of a nucleotide molecule as referred to herein, the length corresponds to the number of monomer units, i.e. nucleotides, irrespective as to whether those monomer units are nucleotides or nucleotide analogues. With respect to nucleotides or nucleobases, the terms monomer and unit are used interchangeably herein.
It should be understood that when the term "about" is used in the context of specific values or ranges of values, the disclosure should be read as to include the specific value or range referred to.
As used herein, "hybridisation" means hydrogen bonding, which may be Watson-Crick, Hoogsteen, reversed Hoogsteen hydrogen bonding, etc., between complementary nucleoside or nucleotide bases. The four nucleobases commonly found in DNA are G, A, T and C of which G pairs with C, and A pairs with T. In RNA T is replaced with uracil (U), which then pairs with A. The chemical groups in the nucleobases that participate in standard duplex formation constitute the Watson-Crick face. Hoogsteen showed a couple of years later that the purine nucleobases (G and A) in addition to their Watson-Crick face have a Hoogsteen face that can be recognised from the outside of a duplex, and used to bind pyrimidine oligonucleotides via hydrogen bonding, thereby forming a triple helix structure. In the context of the present invention "complementary" refers to the capacity for precise pairing between two nucleotides sequences with one another. For example, if a nucleotide at a certain position of an oligonucleotide is capable of hydrogen bonding with a nucleotide at the corresponding position of a DNA or RNA molecule, then the oligonucleotide and the DNA or RNA are considered to be complementary to each other at that position. The DNA or RNA strand are considered complementary to each other when a sufficient number of nucleotides in the oligonucleotide can form hydrogen bonds with corresponding nucleotides in the target DNA or RNA to enable the formation of a stable complex. To be stable in vitro or in vivo the sequence of an oligonucleotide need not be 100% complementary to its target microRNA. The terms "complementary" and "specifically hybridisable" thus imply that the oligonucleotide binds sufficiently strong and specific to the target molecule to provide the desired interference with the normal function of the target whilst leaving the function of non-target RNAs unaffected. However, in one preferred embodiment the term complementary shall mean 100% complementary or fully complementary.
In a preferred example the oligonucleotide of the invention is 100% complementary to a miRNA sequence, such as a human microRNA sequence, or one of the microRNA sequences refered to herein. In a preferred example, the oligonucleotide of the invention comprises a contiguous sequence, which is 100% complementary to the seed region of the human microRNA sequence.
Preferably, the term "microRNA" or "miRNA", in the context of the present invention, means an RNA oligonucleotide consisting of between 18 to 25 nucleotides in length. In functional terms miRNAs are typically regulatory endogenous RNA molecules.
The terms "target microRNA" or "target miRNA" refer to a microRNA with a biological role in human disease, e.g. an upregulated, oncogenic miRNA or a tumor suppressor miRNA in cancer, thereby being a target for therapeutic intervention of the disease in question.
The terms "target gene" or "target mRNA" refer to regulatory mRNA targets of microRNAs, in which said "target gene" or "target mRNA" is regulated post-transcriptionally by the microRNA based on near-perfect or perfect complementarity between the miRNA and its target site resulting in target mRNA cleavage; or limited complementarity, often conferred to complementarity between the so-called seed sequence (nucleotides 2-7 of the miRNA) and the target site resulting in translational inhibition of the target mRNA. In the context of the present invention the oligonucleotide is single stranded, this refers to the situation where the oligonucleotide is in the absence of a complementary oligonucleotide - i.e. it is not a double stranded oligonucleotide complex, such as an siRNA. In one embodiment, the composition according ot the invention does not comprise a further oligonucleotide which has a region of complementarity with the oligomer of 5 or more, such as 6, 7, 8, 9, or 10 consecutive nucleotides, such as eight or more. Length
Surprisingly we have found that such short 'antimiRs' provide an improved specific inhibition of microRNAs in vivo, whilst retaining remarkable specificity for the microRNA target. A further benefit has been found to be the ability to inhibit several microRNAs simultaneously due to the conservation of homologous short sequences between microRNA species - such as the seed regions as described herein. According to the present invention, it has been found that it is particularly advantageous to have short oligonucleotides of 7, 8, 9, 10 nucleotides, such as 7, 8 or 9 nucleotides. Sequences The contiguous nucleotide sequence is complementary (such as 100% complementary -
Ae. perfectly complementary) to a corresponding region of a mammalian, human or viral microRNA (miRNA) sequence, preferably a human or viral miRNA sequence.
The microRNA sequence may suitably be a mature microRNA. In some embodiments the microRNA may be a microRNA precursor. The human microRNA sequence may be selected from SEQ ID No 1 - 558 as disclosed in WO2008/046911 , which are all hereby and specifically incorporated by reference. As described in WO2008/046911, these microRNAs are associated with cancer.
The viral microRNA sequence may, in some embodiments, be selected from the group consisting of Herpes simplex virus 1 , Kaposi sarcoma-associated herpesvirus, Epstein Barr virus and Human cytomegalovirus.
In one embodiment, the contiguous nucleotide sequence is complementary (such as 100% complementary) to a corresponding region of a miRNA sequence selected from the group of miRNAs listed in table 1. Table 1 provides 7mer, 8mer and 9mer oligomers which target human and viral microRNAs published in miRBase (Release 12.0 - http://microma.sanger.ac.uk/sequences/).
In some embodiments, the oligomers according to the invention may consist of or comprise a contiguous nucleotide sequence which is complementary to a corresponding microRNA sequence selected from the group consisting of miR-1 , miR-10b, miR-17-3p, miR-18, miR-19a, miR-19b, miR-20, miR-21 , miR-34a, miR-93, miR-106a, miR-106b, miR-122, miR-133, miR-134, miR-138, miR-155, miR-192, miR-194, miR-221 , miR-222, miR-375.
Therefore, in one embodiment, the miRNA (Le target miRNA) is selected from the group consisting of miR-1 , miR-1 Ob, miR-17-3p, miR-18, miR-19a, miR-19b, miR-20, miR-21 , miR- 34a, miR-93, miR-106a, miR-106b, miR-122, miR-133, miR-134, miR-138, miR-155, miR-192, miR-194, miR-221 , miR-222, and miR-375. In one embodiment, the miRNA target is a member of the miR 17 - 92 cluster, such as miR 17, miR 106a, miR 106b, miR 18, miR 19a, miR 19b/1 , miR 19b/2, miR20/93, miR92/1 , miR92/2 and miR25. In some embodiments the contiguous nucleotide sequence is complementary to a corresponding region of a microRNA (miRNA) sequence selected from the group consisting of miR-21, miR-155, miR-221 , mir-222, and mir-122.
In some embodiments said miRNA is selected from the group consisting of miR-1, miR- IOmiR-29, miR-125b,miR-126, miR-133, miR-141 , miR-143, miR-200b, miR-206, miR-208, miR- 302, miR-372, miR-373, miR-375, and miR-520c/e.
In some embodiments the contiguous nucleotide sequence is complementary to a corresponding region of a microRNA (miRNA) sequence present in the miR 17 - 92 cluster, such as a microRNA selected from the group consisting of miR-17-5p, miR-20a/b, miR-93, miR-106a/b, miR-18a/b, miR-19a/b, miR-25, miR-92a, , miR-363.
In one embodiment, the miRNA (Ae target miRNA) is miR-21 , such as hsa-miR-21. In one embodiment, the miRNA (Le target miRNA) is miR-122, such as hsa-miR-122. In one embodiment, the miRNA (Le target miRNA) is miR-19b, such as hsa-miR-19b. In one embodiment, the miRNA (Le target miRNA) is miR-155, such as hsa-miR-155. In one embodiment, the miRNA (Le target miRNA) is miR-375, such as hsa-miR-375. In one embodiment, the miRNA (Ae target miRNA) is miR-375, such as hsa-miR-106b.
Suitably, the contiguous nucleotide sequence may be complementary to a corresponding region of the microRNA, such as a hsa-miR selected from the group consisting of 19b, 21 , 122, 155 and 375. The Seed Region and Seedmers
The inventors have found that carefully designed short single stranded oligonucleotides comprising or consisting of nucleotide analogues, such as high affinity nucleotide analogues such as locked nucleic acid (LNA) units, show significant silencing of microRNAs, resulting in reduced microRNA levels. It was found that tight binding of said oligonucleotides to the so- called seed sequence, typically nucleotides 2 to 8 or 2 to 7, counting from the 5' end, of the target microRNAs was important. Nucleotide 1 of the target microRNAs is a non-pairing base and is most likely hidden in a binding pocket in the Ago 2 protein. Whilst not wishing to be bound to a specific theory, the present inventors consider that by selecting the seed region sequences, particularly with oligonucleotides that comprise LNA, preferably LNA units in the region which is complementary to the seed region, the duplex between miRNA and oligonucleotide is particularly effective in targeting miRNAs, avoiding off target effects, and possibly providing a further feature which prevents RISC directed miRNA function.
The inventors have found that microRNA silencing is even more enhanced when LNA- modified single stranded oligonucleotides do not contain a nucleotide at the 3' end corresponding to this non-paired nucleotide 1. It was further found that at least two LNA units in the 3' end of the oligonucleotides according to the present invention made said oligonucleotides highly nuclease resistant. In one embodiment, the first or second 3' nucleotide of the oligomer corresponds to the second 5' nucleotide of the microRNA sequence, and may be a nucleotide analogue, such as LNA.
In one embodiment, nucleotide units 1 to 6 (inclusive) of the oligomer as measured from the 3' end the region of the oligomer are complementary to the microRNA seed region sequence, and may all be nucleotide analogues, such as LNA.
In one embodiment, nucleotide units 1 to 7 (inclusive) of the oligomer as measured from the 3' end the region of the oligomer are complementary to the microRNA seed region sequence, and may all be nucleotide analogues, such as LNA. In one embodiment, nucleotide units 2 to 7 (inclusive) of the oligomer as measured from the 3' end the region of the oligomer are complementary to the microRNA seed region sequence, and may all be nucleotide analogues, such as LNA.
In one embodiment, the oligomer comprises at least one nucleotide analogue unit, such as at least one LNA unit, in a position which is within the region complementary to the miRNA seed region. The oligomer may, in one embodiment comprise at between one and 6 or between 1 and 7 nucleotide analogue units, such as between 1 and 6 and 1 and 7 LNA units, in a position which is within the region complementary to the miRNA seed region.
In one embodiment, the contiguous nucleotide sequence consists of or comprises a sequence which is complementary (such as 100% complementary) to the seed sequence of said microRNA.
In one embodiment, the contiguous nucleotide sequence consists of or comprises a sequence selected from any one of the seedmer sequences listed in table 1.
In one embodiment, the 3' nucleotide of the seedmer forms the 3' most nucleotide of the contiguous nucleotide sequence, wherein the contiguous nucleotide sequence may, optionally, comprise one or two further nucleotide 5' to the seedmer sequence.
In one embodiment, the oligomer does not comprise a nucleotide which corresponds to the first nucleotide present in the microRNA sequence counted from the 5' end.
In one embodiment, the oligonucleotide according to the invention does not comprise a nucleotide at the 3' end that corresponds to the first 5' end nucleotide of the target microRNA. Nucleotide Analogues
According to the present invention, it has been found that it is particularly advantageous to have short oligonucleotides of 7, 8, 9, 10 nucleotides, such as 7, 8 or 9 nucleotides, wherein at least 50%, such as 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or such as 100% of the nucleotide units of the oligomer are (preferably high affinity) nucleotide analogues, such as a Locked Nucleic Acid (LNA) nucleotide unit.
In some embodiments, the oligonucleotide of the invention is 7, 8 or 9 nucleotides long, and comprises a contiguous nucleotide sequence which is complementary to a seed region of a human or viral microRNA, and wherein at least 75 %, such as at least 80 %, such as at least 85%, such as at least 90%, such as at least 95%, such as 100% of the nucleotides are are Locked Nucleic Acid (LNA) nucleotide units.
In such oligomers, in some embodiments, the linkage groups are other than phosphodiester linkages, such as are phosphorothioate linkages.
In one embodiment, all of the nucleotide units of the contiguous nucleotide sequence are LNA nucleotide units.
In one embodiment, the contiguous nucleotide sequence comprises or consists of 7, 8, 9 or 10, preferably contiguous, LNA nucleotide units. In a further preferred embodiment, the oligonucleotide of the invention is 7, 8 or 9 nucleotides long, and comprises a contiguous nucleotide sequence which is complementary to a seed region of a human or viral microRNA, and wherein at least 80 % of the nucleotides are LNA, and wherein at least 80%, such as 85%, such as 90%, such as 95%, such as 100% of the intern ucleotide bonds are phosphorothioate bonds. It will be recognised that the contiguous nucleotide sequence of the oligmer (a seedmer) may extend beyond the seed region.
In some embodiments, the oligonucleotide of the invention is 7 nucleotides long, which are all LNA.
In some embodiments, the oligonucleotide of the invention is 8 nucleotides long, of which up to 1 nucleotide may be other than LNA. In some embodiments, the oligonucleotide of the invention is 9 nucleotides long, of which up to 1 or 2 nucleotides may be other than LNA. In some embodiments, the oligonucleotide of the invention is 10 nucleotides long, of which 1 , 2 or 3 nucleotides may be other than LNA. The nucleotides 'other than LNA, may for example, be DNA, or a 2' substituted nucleotide analogues.
High affinity nucleotide analogues are nucleotide analogues which result in oligonucleotides which has a higher thermal duplex stability with a complementary RNA nucleotide than the binding affinity of an equivalent DNA nucleotide. This may be determined by measuring the Tm.
In some embodiments, the nucleotide analogue units present in the contiguous nucleotide sequence are selected, optionally independently, from the group consisting of 2'-O_alkyl-RNA unit, 2'-OMe-RNA unit, 2'-amino-DNA unit, 2'-fluoro-DNA unit, LNA unit, PNA unit, HNA unit, INA unit, and a 2'MOE RNA unit.
In some embodiments, the nucleotide analogue units present in the contiguous nucleotide sequence are selected, optionally independently, from the group consisting of 2'-O_alkyl-RNA unit, 2'-OMe-RNA unit, 2'-amino-DNA unit, 2'-fluoro-DNA unit, LNA unit, and a 2'MOE RNA unit. The term 2'fluoro-DNA refers to a DNA analogue with a substitution to fluorine at the 2' position (2'F). 2'fluoro-DNA is a preferred form of 2'fluoro-nucleotide. In some embodiments, the oligomer comprises at least 4 nucleotide analogue units, such as at least 5 nucleotide analogue units, such as at least 6 nucleotide analogue units, such as at least 7 nucleotide analogue units, such as at least 8 nucleotide analogue units, such as at least 9 nucleotide analogue units, such as 10, nucleotide analogue units. In one embodiment, the oligomer comprises at least 3 LNA units, such as at least 4 LNA units, such as at least 5 LNA units, such as at least 6 LNA units, such as at least 7 LNA units, such as at least 8 LNA units, such as at least 9 LNA units, such as 10 LNA.
In one embodiment wherein at least one of the nucleotide analogues, such as LNA units, is either cytosine or guanine, such as between 1 - 10 of the of the nucleotide analogues, such as LNA units, is either cytosine or guanine, such as 2, 3, 4, 5, 6, 7, 8, or 9 of the of the nucleotide analogues, such as LNA units, is either cytosine or guanine.
In one embodiment at least two of the nucleotide analogues such as LNA units are either cytosine or guanine. In one embodiment at least three of the nucleotide analogues such as LNA units are either cytosine or guanine. In one embodiment at least four of the nucleotide analogues such as LNA units are either cytosine or guanine. In one embodiment at least five of the nucleotide analogues such as LNA units are either cytosine or guanine. In one embodiment at least six of the nucleotide analogues such as LNA units are either cytosine or guanine. In one embodiment at least seven of the nucleotide analogues such as LNA units are either cytosine or guanine. In one embodiment at least eight of the nucleotide analogues such as LNA units are either cytosine or guanine.
In a preferred embodiment the nucleotide analogues have a higher thermal duplex stability for a complementary RNA nucleotide than the binding affinity of an equivalent DNA nucleotide to said complementary RNA nucleotide.
In one embodiment, the nucleotide analogues confer enhanced serum stability to the single stranded oligonucleotide.
Whilst the specific SEQ IDs in the sequence listing and table 1 refer to oligomers of LNA monomers with phosphorothioate (PS) backbone, it will be recognised that the invention also encompasses the use of other nucleotide analogues and/or linkages, either as an alternative to, or in combination with LNA. As such, the sequence of nucleotides (bases) shown in the sequence listings may be of LNA such as LNA/PS, LNA or may be oligomers containing alternative backbone chemistry, such as sugar/linkage chemistry, whilst retaining the same base sequence (A, T, C or G).
Whilst it is envisaged that other nucleotide analogues, such as 2'-MOE RNA or 2'-fluoro nucleotides may be useful in the oligomers according to the invention, it is preferred that the oligomers have a high proportion, such as at least 50%, LNA. nucleotides.
The nucleotide analogue may be a DNA analogue such as a DNA analogue where the 2'-H group is substituted with a substitution other than -OH (RNA) e.g. by substitution with -0-CH3, - 0-CH2-CH2-O-CH3, -0-CH2-CH2-CH2-NH2, -0-CH2-CH2-CH2-OH or -F. The nucleotide analogue may be a RNA analogues such as a RNA analogue which have been modified in its 2'-OH group, e.g. by substitution with a group other than -H (DNA), for example -0-CH3, -O- CH2-CH2-O-CH3, -0-CH2-CH2-CH2-NH2, -0-CH2-CH2-CH2-OH or -F. In one emdodiment the nucleotide analogue is "ENA". LNA
When used in the present context, the terms "LNA unit", "LNA monomer", "LNA residue", "locked nucleic acid unit", "locked nucleic acid monomer" or "locked nucleic acid residue", refer to a bicyclic nucleoside analogue. LNA units are described in inter alia WO 99/14226, WO 00/56746, WO 00/56748, WO 01/25248, WO 02/28875, WO 03/006475 and WO 03/095467. The LNA unit may also be defined with respect to its chemical formula. Thus, an "LNA unit", as used herein, has the chemical structure shown in Scheme 1 below:
Scheme 1
Figure imgf000020_0001
1A 1B wherein
X is selected from the group consisting of O, S and NRH, where RH is H or C1-4-alkyl; Y is (-CH2)r, where r is an integer of 1-4; and B is a nitrogenous base.
In a preferred embodiment of the invention, r is 1 or 2, in particular 1 , i.e. a preferred LNA unit has the chemical structure shown in Scheme 2 below:
Scheme 2
Figure imgf000020_0002
2A 2B wherein X and B are as defined above.
In an interesting embodiment, the LNA units incorporated in the oligonucleotides of the invention are independently selected from the group consisting of thio-LNA units, amino-LNA units and oxy-LNA units.
Thus, the thio-LNA unit may have the chemical structure shown in Scheme 3 below:
Scheme 3
Figure imgf000021_0001
3A 3B wherein B is as defined above.
Preferably, the thio-LNA unit is in its beta-D-form, i.e. having the structure shown in 3A above, likewise, the amino-LNA unit may have the chemical structure shown in Scheme 4 below:
Scheme 4
Figure imgf000021_0002
4A 4B wherein B and RH are as defined above. Preferably, the amino-LNA unit is in its beta-D-form, i.e. having the structure shown in 4A above. The oxy-LNA unit may have the chemical structure shown in Scheme 5 below:
Scheme 5
Figure imgf000021_0003
5A 5B wherein B is as defined above.
Preferably, the oxy-LNA unit is in its beta-D-form, i.e. having the structure shown in 5A above. As indicated above, B is a nitrogenous base which may be of natural or non-natural origin. Specific examples of nitrogenous bases include adenine (A), cytosine (C), 5- methylcytosine (MeC), isocytosine, pseudoisocytosine, guanine (G), thymine (T), uracil (U), 5- bromouracil, 5-propynyluracil, 5-propyny-6, 5-methylthiazoleuracil, 6-aminopurine, 2- aminopurine, inosine, 2,6-diaminopurine, 7-propyne-7-deazaadenine, 7-propyne-7- deazaguanine and 2-chloro-6-aminopurine.
The term "thio-LNA unit" refers to an LNA unit in which X in Scheme 1 is S. A thio-LNA unit can be in both the beta-D form and in the alpha-L form. Generally, the beta-D form of the thio-LNA unit is preferred. The beta-D-form and alpha-L-form of a thio-LNA unit are shown in Scheme 3 as compounds 3A and 3B, respectively. The term "amino-LNA unit" refers to an LNA unit in which X in Scheme 1 is NH or NRH, where RH is hydrogen or C1-4-alkyl. An amino-LNA unit can be in both the beta-D form and in the alpha-L form. Generally, the beta-D form of the amino-LNA unit is preferred. The beta-D-form and alpha-L-form of an amino-LNA unit are shown in Scheme 4 as compounds 4A and 4B, respectively.
The term "oxy-LNA unit" refers to an LNA unit in which X in Scheme 1 is O. An Oxy-LNA unit can be in both the beta-D form and in the alpha-L form. Generally, the beta-D form of the oxy-LNA unit is preferred. The beta-D form and the alpha-L form of an oxy-LNA unit are shown in Scheme 5 as compounds 5A and 5B, respectively. In the present context, the term "Ci-6-alkyl" is intended to mean a linear or branched saturated hydrocarbon chain wherein the longest chains has from one to six carbon atoms, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isopentyl, neopentyl and hexyl. A branched hydrocarbon chain is intended to mean a Ci-6-alkyl substituted at any carbon with a hydrocarbon chain. In the present context, the term "C1-4-alkyl" is intended to mean a linear or branched saturated hydrocarbon chain wherein the longest chains has from one to four carbon atoms, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl and tert-butyl. A branched hydrocarbon chain is intended to mean a C1-4-alkyl substituted at any carbon with a hydrocarbon chain. When used herein the term "C1-6-alkoxy" is intended to mean C1-6-alkyl-oxy, such as methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, sec-butoxy, tert-butoxy, pentoxy, isopentoxy, neopentoxy and hexoxy.
In the present context, the term "C2-6-alkenyl" is intended to mean a linear or branched hydrocarbon group having from two to six carbon atoms and containing one or more double bonds. Illustrative examples of C2-6-alkenyl groups include allyl, homo-allyl, vinyl, crotyl, butenyl, butadienyl, pentenyl, pentadienyl, hexenyl and hexadienyl. The position of the unsaturation (the double bond) may be at any position along the carbon chain.
In the present context the term "C2-6-alkynyl" is intended to mean linear or branched hydrocarbon groups containing from two to six carbon atoms and containing one or more triple bonds. Illustrative examples of C2-6-alkynyl groups include acetylene, propynyl, butynyl, pentynyl and hexynyl. The position of unsaturation (the triple bond) may be at any position along the carbon chain. More than one bond may be unsaturated such that the "C2.6-alkynyl" is a di-yne or enedi-yne as is known to the person skilled in the art.
When referring to substituting a DNA unit by its corresponding LNA unit in the context of the present invention, the term "corresponding LNA unit" is intended to mean that the DNA unit has been replaced by an LNA unit containing the same nitrogenous base as the DNA unit that it has replaced, e.g. the corresponding LNA unit of a DNA unit containing the nitrogenous base A also contains the nitrogenous base A. The exception is that when a DNA unit contains the base C, the corresponding LNA unit may contain the base C or the base MeC, preferably MeC.
Herein, the term "non-LNA unit" refers to a nucleoside different from an LNA-unit, i.e. the term "non-LNA unit" includes a DNA unit as well as an RNA unit. A preferred non-LNA unit is a DNA unit.
The terms "unit", "residue" and "monomer" are used interchangeably herein.
The term "at least one" encompasses an integer larger than or equal to 1 , such as 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20 and so forth.
The terms "a" and "an" as used about a nucleotide, an agent, an LNA unit, etc., is intended to mean one or more. In particular, the expression "a component (such as a nucleotide, an agent, an LNA unit, or the like) selected from the group consisting of ..." is intended to mean that one or more of the cited components may be selected. Thus, expressions like "a component selected from the group consisting of A, B and C" is intended to include all combinations of A, B and C1 i.e. A, B, C, A+B, A+C, B+C and A+B+C. lnternucleoside Linkages
The term "internucleoside linkage group" is intended to mean a group capable of covalently coupling together two nucleotides, such as between DNA units, between DNA units and nucleotide analogues, between two non-LNA units, between a non-LNA unit and an LNA unit, and between two LNA units, etc. Examples include phosphate, phosphodiester groups and phosphorothioate groups.
In some embodiments, at least one of, such as all of the internucleoside linkage in the oligomer is phosphodiester. However for in vivo use, phosphorothioate linkages may be preferred.
Typical internucleoside linkage groups in oligonucleotides are phosphate groups, but these may be replaced by internucleoside linkage groups differing from phosphate. In a further interesting embodiment of the invention, the oligonucleotide of the invention is modified in its internucleoside linkage group structure, i.e. the modified oligonucleotide comprises an internucleoside linkage group which differs from phosphate. Accordingly, in a preferred embodiment, the oligonucleotide according to the present invention comprises at least one internucleoside linkage group which differs from phosphate.
Specific examples of internucleoside linkage groups which differ from phosphate
(-0-P(O)2-O-) include -0-P(O1S)-O-, -0-P(S)2-O-, -S-P(O)2-O-, -S-P(O1S)-O-, -S-P(S)2-O-, -0-P(O)2-S-, -0-P(O1S)-S-, -S-P(O)2-S-, -O-PO(RH)-O-, 0-PO(OCHs)-O-, -O-PO(NRH)-O-, -O- PO(OCH2CH2S-R)-O-, -O-PO(BH3)-O-, -O-PO(NHRH)-O-, -0-P(O)2-N RH-, -NRH-P(O)2-O-, -NRH-C0-0-, -NRH-CO-NRH-, -0-C0-0-, -O-CO-NRH-, -NR1^CO-CH2-, -0-CH2-CO-NR1"1-, -
0-CH2-CH2-NR1"1-, -CO-NR1^CH2-, -CH2-NRH-CO-, -0-CH2-CH2-S-, -S-CH2-CH2-O-, -S-CH2-CH2- S-, -CH2-SO2-CH2-, -CH2-CO-NR1"1-, -0-CH2-CH2-N RH-CO -, -CH2-NCH3-O-CH2-, where RH is hydrogen or C1-4-alkyl.
When the internucleoside linkage group is modified, the internucleoside linkage group is preferably a phosphorothioate group (-0-P(O1S)-O- ). In a preferred embodiment, all internucleoside linkage groups of the oligonucleotides according to the present invention are phosphorothioate.
The internucleoside linkage may be selected form the group consisting of: -0-P(O)2-O-,
-0-P(O1S)-O-, -0-P(S)2-O-, -S-P(O)2-O-, -S-P(O1S)-O-, -S-P(S)2-O-, -0-P(O)2-S-, -0-P(O1S)-S-,
-S-P(O)2-S-, -0-P0(RH)-0-, O-PO(OCH3)-O-, -O-PO(NRH)-O-, -0-PO(OCH2CH2S-R)-O-, -O-PO(BH3)-O-, -O-PO(NHRH)-O-, -0-P(O)2-N RH-, -NRH-P(0)2-0-, -NRH-C0-0-,
-NRH-C0-NRH-, and/or the internucleoside linkage may be selected form the group consisting of: -O-CO-O-, -O-CO-NRH-, -NRH-CO-CH2-, -0-CH2-CO-NR8-, -O-CH2-CH2-NRH-, -CO-NR1"1-
CH2-, -CH2-NRH-C0-, -0-CH2-CH2-S-, -S-CH2-CH2-O-, -S-CH2-CH2-S-, -CH2-SO2-CH2-, -CH2-
C0-NRH-, -O-CH2-CH2-NRH-CO -, -CH2-NCH3-O-CH2-, where RH is selected from hydrogen and C1-4-alkyl. Suitably, in some embodiments, sulphur (S) containing internucleoside linkages as provided above may be preferred. The internucleoside linkages may be independently selected, or all be the same, such as phosphorothioate linkages.
In one embodiment, at least 75%, such as 80% or 85% or 90% or 95% or all of the internucleoside linkages present between the nucleotide units of the contiguous nucleotide sequence are phosphorothioate internucleoside linkages.
Micromir oligonucleotides targeting more than one microRNA
In one embodiment, the contiguous nucleotide sequence is complementary to the corresponding sequence of at least two miRNA sequences such as 2, 3, 4, 5, 6, 7, 8, 9, or 10 miRNA sequence,. The use of a single universal base may allow a single oligomer of the invention to target two independant microRNAs which either one or both have a single mismatch in the region which corresponds to oligomer at the position where the universal nucleotide is positioned.
In one embodiment, the contiguous nucleotide sequence consists of or comprises a sequence which is complementary to the sequence of at least two miRNA seed region sequences such as 2, 3, 4, 5, 6, 7, 8, 9, or 10 miRNA seed region sequences.
In one embodiment, the contiguous nucleotide sequence is complementary to the corresponding region of both miR-221 and miR-222.
In one embodiment, the contiguous nucleotide sequence is complementary to the corresponding region of more than one member of the miR-17-92 cluster - such as two or more or all of miR-17-5p, miR-20a/b, miR-93, miR-106a/b; or two or more or all of miR-25, miR-92a and miR-363. In one embodiment, the contiguous nucleotide sequence consists of or comprises a sequence that is complementary to 5'GCTACAT3'. Oligomer Design
In one embodiment, the first nucleotide of the oligomer according to the invention, counting from the 31 end, is a nucleotide analogue, such as an LNA unit. In one embodiment, which may be the same or different, the last nucleotide of the oligomer according to the invention, counting from the 3' end, is a nucleotide analogue, such as an LNA unit.
In one embodiment, the second nucleotide of the oligomer according to the invention, counting from the 3' end, is a nucleotide analogue, such as an LNA unit. In one embodiment, the ninth and/or the tenth nucleotide of the oligomer according to the invention, counting from the 3' end, is a nucleotide analogue, such as an LNA unit.
In one embodiment, the ninth nucleotide of the oligomer according to the invention, counting from the 3' end is a nucleotide analogue, such as an LNA unit.
In one embodiment, the tenth nucleotide of the oligomer according to the invention, counting from the 3' end is a nucleotide analogue, such as an LNA unit.
In one embodiment, both the ninth and the tenth nucleotide of the oligomer according to the invention, calculated from the 3' end is a nucleotide analogue, such as an LNA unit.
In one embodiment, the oligomer according to the invention does not comprise a region of more than 3 consecutive DNA nucleotide units. In one embodiment, the oligomer according to the invention does not comprise a region of more than 2 consecutive DNA nucleotide units. In one embodiment, the oligomer comprises at least a region consisting of at least two consecutive nucleotide analogue units, such as at least two consecutive LNA units. In one embodiment, the oligomer comprises at least a region consisting of at least three consecutive nucleotide analogue units, such as at least three consecutive LNA units. Other Patterns of Nucleotide Analogues such as LNA in the Oligomer
Whilst it is envisaged that oligomers containing at least 6 LNA, such as at least 7 nucleotide units may be preferable, the discovery that such short oligomers are highly effective at targeting microRNAs in vivo can be used to prepare shorter oligomers of the invention which comprise other nucleotide analogues, such as high affinity nucleotide analogues. Indeed, the combination of LNA with other high affinity nucleotide analogues are considered as part of the present invention.
Modification of nucleotides in positions 1 to 2, counting from the 3' end. The nucleotide at positions 1 and/ or 2 may be a nucleotide analogue, such as a high affinity nucleotide analogue, such as LNA, or a nucleotide analogue selected from the group consisting of 2'-O-alkyl-RNA unit, 2'-OMe-RNA unit, 2'-amino-DNA unit, 2'-fluoro-DNA unit, 2'-MOE-RNA unit, LNA unit, PNA unit, HNA unit, INA unit. The two 3' nucleotide may therefore be Xx, xX, XX or xx, wherein: In one embodiment X is LNA and x is DNA or another nucleotide analogue, such as as a 2' substituted nucleotide analogue selected from the group consisting of 2'-O_alkyl-RNA unit, 2'-OMe-RNA unit, 2'-amino-DNA unit, 2'-fluoro-DNA unit, LNA, and a 2'MOE RNA unit. Said non-LNA unit (x) may therefore be 2'MOE RNA or 2'-fluoro-DNA. Alternatively X is a nucleotide analogue, and x is DNA.
The above modification at the 2 3' terminal nucleotides may be combined with modification of nucleotides in positions 3 - 8 counting from the 3' end, as described below. In this respect nucleotides designated as X and x may be the same throughout the oligomer. It will be noted that when the oligomer is only 7 nucleotides in length the 8th nucleotide counting from the 3' end should be discarded. In the following embodiments which refer to the modification of nucleotides in positions 3 to 8, counting from the 3" end, the LNA units, in one embodiment, may be replaced with other nucleotide anlogues, such as those referred to herein. "X" may, therefore be selected from the group consisting of 2'-O-alkyl-RNA unit, 2'-OMe-RNA unit, 2'-amino-DNA unit, 2'-fluoro-DNA unit, 2'-MOE-RNA unit, LNA unit, PNA unit, HNA unit, INA unit, "x" is preferably DNA or RNA, most preferably DNA. However, it is preferred that X is LNA.
In one embodiment of the invention, the oligonucleotides of the invention are modified in positions 3 to 8, counting from the 3' end. The design of this sequence may be defined by the number of non-LNA units present or by the number of LNA units present. In a preferred embodiment of the former, at least one, such as one, of the nucleotides in positions three to eight, counting from the 3' end, is a non-LNA unit. In another embodiment, at least two, such as two, of the nucleotides in positions three to eight, counting from the 3' end, are non-LNA units. In yet another embodiment, at least three, such as three, of the nucleotides in positions three to eight, counting from the 3' end, are non-LNA units. In still another embodiment, at least four, such as four, of the nucleotides in positions three to eight, counting from the 3' end, are non- LNA units. In a further embodiment, at least five, such as five, of the nucleotides in positions three to eight, counting from the 3' end, are non-LNA units. In yet a further embodiment, all six nucleotides in positions three to eight, counting from the 3' end, are non-LNA units.
Alternatively defined, in an embodiment, the oligonucleotide according to the present invention comprises at least three LNA units in positions three to eight, counting from the 3' end. In an embodiment thereof, the oligonucleotide according to the present invention comprises three LNA units in positions three to eight, counting from the 31 end. The substitution pattern for the nucleotides in positions three to eight, counting from the 3' end, may be selected from the group consisting of XXXxxx, xXXXxx, xxXXXx, xxxXXX, XXxXxx, XXxxXx, XXxxxX, xXXxXx, xXXxxX, xxXXxX, XxXXxx, XxxXXx, XxxxXX, xXxXXx, xXxxXX, xxXxXX, xXxXxX and XxXxXx, wherein "X" denotes an LNA unit and "x" denotes a non-LNA unit. In a preferred embodiment, the substitution pattern for the nucleotides in positions three to eight, counting from the 3" end, is selected from the group consisting of XXxXxx, XXxxXx, XXxxxX, xXXxXx, xXXxxX, xxXXxX, XxXXxx, XxxXXx, XxxxXX, xXxXXx, xXxxXX, xxXxXX, xXxXxX and XxXxXx, wherein "X" denotes an LNA unit and "x" denotes a non-LNA unit. In a more preferred embodiment, the substitution pattern for the nucleotides in positions three to eight, counting from the 3' end, is selected from the group consisting of xXXxXx, xXXxxX, xxXXxX, xXxXXx, xXxxXX, xxXxXX and xXxXxX, wherein "X" denotes an LNA unit and "x" denotes a non-LNA unit. In an embodiment, the substitution pattern for the nucleotides in positions three to eight, counting from the 3' end, is xXxXxX or XxXxXx, wherein "X" denotes an LNA unit and "x" denotes a non-LNA unit. In an embodiment, the substitution pattern for the nucleotides in positions three to eight, counting from the 3" end, is xXxXxX, wherein "X" denotes an LNA unit and "x" denotes a non-LNA unit. In a further embodiment, the oligonucleotide according to the present invention comprises at least four LNA units in positions three to eight, counting from the 3' end. In an embodiment thereof, the oligonucleotide according to the present invention comprises four LNA units in positions three to eight, counting from the 3' end. The substitution pattern for the nucleotides in positions three to eight, counting from the 3' end, may be selected from the group consisting of xxXXXX, xXxXXX, xXXxXX, xXXXxX, xXXXXx, XxxXXX, XxXxXX1 XxXXxX, XxXXXx, XXxxXX, XXxXxX, XXxXXx, XXXxxX, XXXxXx and XXXXxx, wherein "X" denotes an LNA unit and "x" denotes a non-LNA unit.
In yet a further embodiment, the oligonucleotide according to the present invention comprises at least five LNA units in positions three to eight, counting from the 31 end. In an embodiment thereof, the oligonucleotide according to the present invention comprises five LNA units in positions three to eight, counting from the 3' end. The substitution pattern for the nucleotides in positions three to eight, counting from the 31 end, may be selected from the group consisting of xXXXXX, XxXXXX, XXxXXX, XXXxXX, XXXXxX and XXXXXx, wherein "X" denotes an LNA unit and "x" denotes a non-LNA unit. Preferably, the oligonucleotide according to the present invention comprises one or two
LNA units in positions three to eight, counting from the 3' end. This is considered advantageous for the stability of the A-helix formed by the oligo:microRNA duplex, a duplex resembling an RNA: RNA duplex in structure.
In yet a further embodiment, the oligonucleotide according to the present invention comprises at least six LNA units in positions three to eight, counting from the 3' end. In an embodiment thereof, the oligonucleotide according to the present invention comprises at from three to six LNA units in positions three to eight, counting from the 31 end, and in addition from none to three other high affinity nucleotide analogues in the same region, such that the total amount of high affinity nucleotide analogues (including the LNA units) amount to six in the region from positions three to eight, counting from the 3' end.
In some embodiments, such as when X is LNA, said non-LNA unit (x) is another nucleotide analogue unit, such as a 2' substituted nucleotide analogue selected from the group consisting of 2'-O_alkyl-RNA unit, 2'-0Me-RNA unit, 2'-amino-DNA unit, 2'-fluoro-DNA unit, LNA, and a 2'MOE RNA unit. Said non-LNA unit (x) may therefore be 2'MOE RNA or 2'-fluoro-DNA.
For oligomers which have 9 or 10 nucleotides, the nucleotide at positions 9 and/ or 10 may be a nucleotide analogue, such as a high affinity nucleotide analogue, such as LNA, or a nucleotide analogue selected from the group consisting of 2'-O-aIkyl-RNA unit, 2'-OMe-RNA unit, 2'-amino-DNA unit, 2'-fluoro-DNA unit, 2'-MOE-RNA unit, LNA unit, PNA unit, HNA unit, INA unit. The two 5' nucleotides may therefore be
Xx, xX, XX or xx, wherein: In one embodiment X is LNA and x is DNA or another nucleotide analogue, such as as a 2' substituted nucleotide analogue selected from the group consisting of 2'-O_alkyl-RNA unit, 2'-OMe-RNA unit, 2'-amino-DNA unit, 2'-fluoro-DNA unit, LNA, and a 2'MOE RNA unit. Said non-LNA unit (x) may therefore be 2'MOE RNA or 2'-fluoro-DNA. Alternatively X is a nucleotide analogue, and x is DNA.
The above modification at the 2 5' terminal nucleotides may be combined with modification of nucleotides in positions 3 - 8 counting from the 3' end, and/or the 2 3' nucleotitides as described above. In this respect nucleotides designated as X and x may be the same throughout the oligomer.
In a preferred embodiment of the invention, the oligonucleotide according to the present invention contains an LNA unit at the 5' end. In another preferred embodiment, the oligonucleotide according to the present invention contains an LNA unit at the first two positions, counting from the 5' end.
In one embodiment, the invention further provides for an oligomer as described in the context of the pharmaceutical composition of the invention, or for use in vivo in an organism, such as a medicament, wherein said oligomer (or contiguous nucleotide sequence) comprises either i) at least one phosphorothioate linkage and/or ii) at least one 3' terminal LNA unit, and/or iii) at least one 5' teriminal LNA unit.
The oligomer may therefore contain at least one phosphorothioate linkage, such as all linkages being phosphorthioates, and at least one 3' terminal LNA unit, and at least one 5' teriminal LNA unit.
It is preferable for most therapeutic uses that the oligonucleotide is fully phosphorothiolated - an exception being for therapeutic oligonucleotides for use in the CNS, such as in the brain or spine where phosphorothioation can be toxic, and due to the absence of nucleases, phosphodiester bonds may be used, even between consecutive DNA units. As referred to herein, other in one aspect of the oligonucleotide according to the invention is that the second 3' nucleotide, and/or the 9th and 10th (from the 3' end), if present, may also be LNA. In one embodiment, the oligomer comprises at least five nucleotide analogue units, such as at least five LNA units, in positions which are complementary to the miRNA seed region.
In one embodiment, the nucleotide sequence of the oligomer which is complementary to the sequence of the microRNA seed region, is selected from the group consisting of (X)xXXXXX, (X)XxXXXX, (X)XXxXXX, (X)XXXxXX, (X)XXXXxX and (X)XXXXXx, wherein "X" denotes a nucleotide analogue, such as an LNA unit, (X) denotes an optional nucleotide analogue, such as an LNA unit, and "x" denotes a DNA or RNA nucleotide unit.
In one embodiment, the oligomer comprises six or seven nucleotide analogue units, such as six or seven LNA units, in positions which are complementary to the miRNA seed region. In one embodiment, the nucleotide sequence of the oligomer which is complementary to the sequence of the microRNA seed region, is selected from the group consisting of XXXXXX, XxXXXXX, XXxXXXX, XXXxXXX, XXXXxXX, XXXXXxX and XXXXXXx, wherein "X" denotes a nucleotide analogue, such as an LNA unit, such as an LNA unit, and "x" denotes a DNA or RNA nucleotide unit. In one embodiment, the two nucleotide motif at position 7 to 8, counting from the 3' end of the oligomer is selected from the group consisting of xx, XX, xX and Xx, wherein "X" denotes a nucleotide analogue, such as an LNA unit, such as an LNA unit, and "x" denotes a DNA or RNA nucleotide unit.
In one embodiment, the two nucleotide motif at position 7 to 8, counting from the 3' end of the oligomer is selected from the group consisting of XX, xX and Xx, wherein "X" denotes a nucleotide analogue, such as an LNA unit, such as an LNA unit, and "x" denotes a DNA or RNA nucleotide unit.
In one embodiment, the oligomer comprises at 12 nucleotides and wherein the two nucleotide motif at position 11 to 12, counting from the 3' end of the oligomer is selected from the group consisting of xx, XX, xX and Xx, wherein "X" denotes a nucleotide analogue, such as an LNA unit, such as an LNA unit, and "x" denotes a DNA or RNA nucleotide unit.
In one embodiment, the oligomer comprises 12 nucleotides and wherein the two nucleotide motif at position 11 to 12, counting from the 3' end of the oligomer is selected from the group consisting of XX, xX and Xx, wherein "X" denotes a nucleotide analogue, such as an LNA unit, such as an LNA unit, and "x" denotes a DNA or RNA nucleotide unit, such as a DNA unit.
In one embodiment, the oligomer comprises a nucleotide analogue unit, such as an LNA unit, at the 51 end.
In one embodiment, the nucleotide analogue units, such as X, are independently selected form the group consisting of: 2'-O-alkyl-RNA unit, 2'-OMe-RNA unit, 2'-amino-DNA unit, 2'- fluoro-DNA unit, 2'-MOE-RNA unit, LNA unit, PNA unit, HNA unit, INA unit. In one embodiment, all the nucleotides of the oligomer of the invention are nucleotide analogue units.
In one embodiment, the nucleotide analogue units, such as X, are independently selected form the group consisting of: 2'-OMe-RNA units, 2'-fluoro-DNA units, and LNA units, In one embodiment, the oligomer comprises said at least one LNA analogue unit and at least one further nucleotide analogue unit other than LNA.
In one embodiment, the non-LNA nucleotide analogue unit or units are independently selected from 2'-OMe RNA units and 2'-fluoro DNA units.
In one embodiment, the oligomer consists of at least one sequence XYX or YXY, wherein X is LNA and Y is either a 2'-OMe RNA unit and 2'-fluoro DNA unit.
In one embodiment, the sequence of nucleotides of the oligomer consists of alternative X and Y units.
In one embodiment, the oligomer comprises alternating LNA and DNA units (Xx) or (xX). In one embodiment, the oligomer comprises a motif of alternating LNA followed by 2 DNA units (Xxx), xXx or xxX.
In one embodiment, at least one of the DNA or non-LNA nucleotide analogue units are replaced with a LNA nucleotide in a position selected from the positions identified as LNA nucleotide units in any one of the embodiments referred to above. In one embodiment,"X" donates an LNA unit. Further Designs for Oligomers of the invention
Table 1 below provides non-limiting examples of short microRNA sequences that could advantageously be targeted with an oligonucleotide of the present invention.
The oligonucleotides according to the invention, such as those disclosed in table 1 may, in one embodiment, have a sequence of 7, 8, 9 or 10 LNA nucleotides 5' - 3' LLLLLLL(L)(L)(L)(L), or have a sequence of nucleotides selected form the group consisting of, the first 7, 8, 9 or 10 nucleotides of the following motifs:
LdLddL(L)(d)(d)(L)(d)(L)(d)(L)(L), LdLdLL(L)(d)(d)(L)(L)(L)(d)(L)(L), LMLMML(L)(M)(M)(LKM)(L)(M)(L)(L)1 LMLMLL(L)(M)(M)(L)(L)(LXM)(L)(L)1
LFLFFL(L)(F)(F)(L)(F)(L)(F)(L)(L), LFLFLL(L)(F)(F)(L)(L)(L)(F)(L)(L), and every third designs such as; LddLdd(L)(d)(d)(L)(d)(d)(L)(d)(d)(L)(d) 'dLddLd(d)(L)(d)(d)(L)(d)(d)(L)(d)(d)(L), ddLddL(d)(d)(L)(d)(d)(L)(d)(d)(L)(d)(d), LMMLMM(L)(M)(M)(L)(M)(M)(L)(M)(M)(L)(M), MLMMLM(M)(L)(M)(M)(L)(M)(M)(L)(M)(M)(L), MMLMML(M)(M)(L)(M)(M)(L)(M)(M)(L)(M)(M), LFFLFF(L)(F)(F)(L)(F)(F)(L)(F)(F)(L)(F)1 FLFFLF(F)(L)(F)(F)(L)(F)(F)(L)(F)(F)(L), FFLFFL(F)(F)(L)(F)(F)(L)(F)(F)(L)(F)(F), and dLdLdL(d)(L)(d)(L)(d)(L)(d)(L)(d)(L)(d) and an every second design, such as; LdLdLd(L)(d)(L)(d)(L)(d)(L)(d)(L)(d)(L), MLMLML(M)(L)(M)(L)(M)(L)(M)(L)(M)(L)(M), LMLMLM(L)(M)(L)(M)(L)(M)(L)(M)(L)(M)(L), FLFLFL(F)(L)(F)(L)(F)(L)(F)(L)(F)(L)(F), and LFLFLF(L)(F)(L)(F)(L)(F)(L)(F)(L)(F)(L); wherein L = LNA unit, d= DNA units, M = 2'MOE RNA, F = 2'Fluoro and residues in brackets are optional. Pharmaceutical Composition and Medical Application
The invention provides for a pharmaceutical composition comprising the oligomer according to the invention, and a pharmaceutically acceptable diluent, carrier, salt or adjuvant.
The invention further provides for the use of an oligonucleotide according to the invention, such as those which may form part of the pharmaceutical composition, for the manufacture of a medicament for the treatment of a disease or medical disorder associated with the presence or over-expression (upregulation) of the microRNA.
The invention further provides for a method for the treatment of a disease or medical disorder associated with the presence or over-expression of the microRNA, comprising the step of administering a composition (such as the pharmaceutical composition) according to the invention to a person in need of treatment.
The invention further provides for a method for reducing the effective amount of a miRNA in a cell or an organism, comprising administering a composition (such as the pharmaceutical composition) according to the invention or a oligomer according to the invention to the cell or the organism. Reducing the effective amount in this context refers to the reduction of functional miRNA present in the cell or organism. It is recognised that the preferred oligonucleotides according to the invention may not always significantly reduce the actual amount of miRNA in the cell or organism as they typically form very stable duplexes with their miRNA targets. The reduction of the effective amount of the miRNA in a cell may, in one embodiment, be measured by detecting the level of de-repression of the miRNA's target in the cell.
The invention further provides for a method for de-repression of a target mRNA of a miRNA in a cell or an organism, comprising administering a composition (such as the pharmaceutical composition) or a oligomer according to the invention to the cell or the organism. The invention further provides for the use of a oligomer of between 7 - 10 such as 7, 8, 9, or 10 nucleotides in length, for the manufacture of a medicament for the treatment of a disease or medical disorder associated with the presence or over-expression of the microRNA.
In one embodiment the medical condition (or disease) is hepatitis C (HCV), and the miRNA is miR-122. In one embodiment, the pharmaceutical composition according to the invention is for use in the treatment of a medical disorder or disease selected from the group consisting of: hepatitis C virus infection and hypercholesterolemia and related disorders, and cancers.
In one embodiment the medical disorder or disease is a CNS disease, such as a CNS disease where one or more microRNAs are known to be indicated. In the context of hypercholesterolemia related disorders refers to diseases such as atherosclerosis or hyperlipidemia. Further examples of related diseases also include different types of HDL/LDL cholesterol imbalance; dyslipidemias, e.g., familial combined hyperlipidemia (FCHL), acquired hyperlipidemia, statin-resistant hypercholesterolemia; coronary artery disease (CAD) coronary heart disease (CHD), atherosclerosis.
In one embodiment, the pharmaceutical composition according to the invention further comprises a second independent active ingredient that is an inhibitor of the VLDL assembly pathway, such as an ApoB inhibitor, or an MTP inhibitor (such as those disclosed in US 60/977,497, hereby incorporated by reference).
The invention further provides for a method for the treatment of a disease or medical disorder associated with the presence or over-expression of the microRNA, comprising the step of administering a composition (such as the pharmaceutical composition) comprising a oligomer of between between 7 - 10 such as 7, 8, 9, or 10 nucleotides in length, to a person in need of treatment.
The invention further provides for a method for reducing the effective amount of a miRNA target (i.e. 'available' miRNA) in a cell or an organism, comprising administering a composition (such as the pharmaceutical composition) comprising a oligomer of between 6 7 - 10 such as 7, 8, 9, or 10 nucleotides in length, to the cell or the organism.
It should be recognised that "reducing the effective amount" of one or more microRNAs in a cell or organism, refers to the inhibition of the microRNA function in the call or organism. The cell is preferably amammalain cell or a human cell which expresses the microRNA or microRNAs. The invention further provides for a method for de-repression of a target mRNA of a miRNA in a cell or an organism, comprising a oligomer of 7 - 10 such as 7, 8, 9, or 10 nucleotides in length, or (or a composition comprising said oligonucleotide) to the cell or the organism.
As mentioned above, microRNAs are related to a number of diseases. Hence, a fourth aspect of the invention relates to the use of an oligonucleotide as defined herein for the manufacture of a medicament for the treatment of a disease associated with the expression of microRNAs selected from the group consisting of spinal muscular atrophy, Tourette's syndrome, hepatitis C, fragile X mental retardation, DiGeorge syndrome and cancer, such as in non limiting example, chronic lymphocytic leukemia, breast cancer, lung cancer and colon cancer, in particular cancer.
Methods of Synthesis
The invention further provides for a method for the synthesis of an oligomer targeted against a human microRNA, such as an oligomer described herein, said method comprising the steps of: a. Optionally selecting a first nucleotide, counting from the 3' end, which is a nucleotide analogue, such as an LNA nucleotide. b. Optionally selecting a second nucleotide, counting from the 3' end, which is a nucleotide analogue, such as an LNA nucleotide. c. Selecting a region of the oligomer which corresponds to the miRNA seed region, wherein said region is as defined herein. d. Selecting a seventh and optionally an eight nucleotideas defined herein. e. Optionally selecting one or two further 5' terminal of the oligomer is as defined herein; wherein the synthesis is performed by sequential synthesis of the regions defined in steps a - e, wherein said synthesis may be performed in either the 3'-5' ( a to f) or 5' - 3' (e to a)direction, and wherein said oligomer is complementary to a sequence of the miRNA target.
The invention further provides for a method for the preparation of an oligomer (such as an oligomer according to the invention), said method comprising the steps of a) comparing the sequences of two or more miRNA sequences to identifiy two or more miRNA sequences which comprise a common contiguous nucleotide sequence of at least 7 nucleotides in length, such as 7, 8, 9 or 10 nucleotides in length (i.e. a sequence found in both non-idnetical miRNAs), b) preparing an oligomer sequence which consists or comprises of a contiguous nucleotide sequence with is complementary to said common contiguous nucleotide sequence, wherein said oligomer is, as according to the oligomer of the invention. In a preferred example, the common contiguous nucleotide sequence consists or comprises of the seed region of each of said two or more miRNA sequences (which comprise a common contiguous nucleotide seqeunce of at least 6 nucleotides in length). In one embodiment, the seed regions of the two or more miRNAs are identical. Suitably the oligomer consists or comprises a seedmer sequence of 7 or 8 nucleotides in length which comprises of a seqeunce which is complementary to said two or more miRNAs. This method may be used in conjunction with step c of the above method.
The method for the synthesis of the oligomer according to the invention may be performed using standard solid phase oligonucleotide systhesis. In one embodiment, the method for the synthesis of a oligomer targeted against a human microRNA, is performed in the 3' to 5' direction a - e.
A further aspect of the invention is a method to reduce the levels of target microRNA by contacting the target microRNA to an oligonucleotide as defined herein, wherein the oligonucleotide (i) is complementary to the target microRNA sequence (ii) does not contain a nucleotide at the 3' end that corresponds to the first 5' end nucleotide of the target microRNA. Duplex stability and Tm
In one embodiment, the oligomer of the invention is capable of forming a duplex with a complementary single stranded RNA nucleic acid molecule (typically of about the same length of said single stranded oligonucleotide) with phosphodiester internucleoside linkages, wherein the duplex has a Tm of between 3O0C and and 700C or 80°C, such as between 300C and 60°C ot 700C, or between 30°C and 5O0C or 60°C. In one embodiment the Tm is at least 4O0C. Tm may be determined by determining the Tm of the oligomer and a complementary RNA target in the following buffer conditions: 10OmM NaCI, 0.1mM EDTA, 1OmM Na-phosphate, pH 7.0 (see examples for a detailed protocol). A high affinity analogue may be defined as an analogue which, when used in the oligomer of the invention, results in an increase in the Tm of the oligomer as compared to an identicial oligomer which has contains only DNA bases. Conjugates
In one embodiment, said oligomer is conjugated with one or more non-nucleotide (or polynucleotide) compounds.
In the context the term "conjugate" is intended to indicate a heterogenous molecule formed by the covalent attachment ("conjugation") of the oligomer as described herein to one or more non-nucleotide, or non-polynucleotide moieties. Examples of non-nucleotide or non- polynucleotide moieties include macromolecular agents such as proteins, fatty acid chains, sugar residues, glycoproteins, polymers, or combinations thereof. Typically proteins may be antibodies for a target protein. Typical polymers may be polyethylene glycol.
Therefore, in various embodiments, the oligomer of the invention may comprise both a polynucleotide region which typically consists of a contiguous sequence of nucleotides, and a further non-nucleotide region. When referring to the oligomer of the invention consisting of a contiguous nucleotide sequence, the compound may comprise non-nucleotide components, such as a conjugate component.
In various embodiments of the invention the oligomeric compound is linked to ligands/conjugates, which may be used, e.g. to increase the cellular uptake of oligomeric compounds. WO2007/031091 provides suitable ligands and conjugates, which are hereby incorporated by reference.
The invention also provides for a conjugate comprising the compound according to the invention as herein described, and at least one non-nucleotide or non-polynucleotide moiety covalently attached to said compound. Therefore, in various embodiments where the compound of the invention consists of a specified nucleic acid or nucleotide sequence, as herein disclosed, the compound may also comprise at least one non-nucleotide or non- polynucleotide moiety (e.g. not comprising one or more nucleotides or nucleotide analogues) covalently attached to said compound. Conjugation (to a conjugate moiety) may enhance the activity, cellular distribution or cellular uptake of the oligomer of the invention. Such moieties include, but are not limited to, antibodies, polypeptides, lipid moieties such as a cholesterol moiety, cholic acid, a thioether, e.g. Hexyl-s-tritylthiol, a thiocholesterol, an aliphatic chain, e.g., dodecandiol or undecyl residues, a phospholipids, e.g., di-hexadecyl-rac-glycerol or triethylammonium 1 ,2-di-o- hexadecyl-rac-glycero-3-h-phosphonate, a polyamine or a polyethylene glycol chain, an adamantane acetic acid, a palmityl moiety, an octadecylamine or hexylamino-carbonyl- oxycholesterol moiety. The oligomers of the invention may also be conjugated to active drug substances, for example, aspirin, ibuprofen, a sulfa drug, an antidiabetic, an antibacterial or an antibiotic.
In certain embodiments the conjugated moiety is a sterol, such as cholesterol.
In various embodiments, the conjugated moiety comprises or consists of a positively charged polymer, such as a positively charged peptides of, for example between 1 -50, such as 2 - 20 such as 3 - 10 amino acid residues in length, and/or polyalkylene oxide such as polyethylglycol(PEG) or polypropylene glycol - see WO 2008/034123, hereby incorporated by reference. Suitably the positively charged polymer, such as a polyalkylene oxide may be attached to the oligomer of the invention via a linker such as the releasable inker described in WO 2008/034123.
By way of example, the following conjugate moieties may be used in the conjugates of the invention:
5'- OLIGOMER -3'
Figure imgf000035_0001
5'- OLIGOMER -S'
Figure imgf000035_0002
Activated oligomers The term "activated oligomer," as used herein, refers to an oligomer of the invention that is covalently linked (i.e., functionalized) to at least one functional moiety that permits covalent linkage of the oligomer to one or more conjugated moieties, i.e., moieties that are not themselves nucleic acids or monomers, to form the conjugates herein described. Typically, a functional moiety will comprise a chemical group that is capable of covalently bonding to the oligomer via, e.g., a 3'-hydroxyl group or the exocyclic NH2 group of the adenine base, a spacer that is preferably hydrophilic and a terminal group that is capable of binding to a conjugated moiety (e.g., an amino, sulfhydryl or hydroxyl group). In some embodiments, this terminal group is not protected, e.g., is an NH2 group. In other embodiments, the terminal group is protected, for example, by any suitable protecting group such as those described in "Protective Groups in Organic Synthesis" by Theodora W Greene and Peter G M Wuts, 3rd edition (John Wiley & Sons, 1999). Examples of suitable hydroxyl protecting groups include esters such as acetate ester, aralkyl groups such as benzyl, diphenylmethyl, or triphenylmethyl, and tetrahydropyranyl. Examples of suitable amino protecting groups include benzyl, alpha-methylbenzyl, diphenylmethyl, triphenylmethyl, benzyloxycarbonyl, tert-butoxycarbonyl, and acyl groups such as trichloroacetyl or trifluoroacetyl. In some embodiments, the functional moiety is self-cleaving. In other embodiments, the functional moiety is biodegradable. See e.g., U.S. Patent No. 7,087,229, which is incorporated by reference herein in its entirety. In some embodiments, oligomers of the invention are functionalized at the 5' end in order to allow covalent attachment of the conjugated moiety to the 5' end of the oligomer. In other embodiments, oligomers of the invention can be functionalized at the 3' end. In still other embodiments, oligomers of the invention can be functionalized along the backbone or on the 5 heterocyclic base moiety. In yet other embodiments, oligomers of the invention can be functionalized at more than one position independently selected from the 5' end, the 3' end, the backbone and the base.
In some embodiments, activated oligomers of the invention are synthesized by incorporating during the synthesis one or more monomers that is covalently attached to a 10. functional moiety. In other embodiments, activated oligomers of the invention are synthesized with monomers that have not been functionalized, and the oligomer is functionalized upon completion of synthesis. In some embodiments, the oligomers are functionalized with a hindered ester containing an aminoalkyl linker, wherein the alkyl portion has the formula (CH2)w, wherein w is an integer ranging from 1 to 10, preferably about 6, wherein the alkyl portion of the 15 alkylamino group can be straight chain or branched chain, and wherein the functional group is attached to the oligomer via an ester group (-O-C(O)-(CH2)WNH).
In other embodiments, the oligomers are functionalized with a hindered ester containing a (CH2)w-sulfhydryl (SH) linker, wherein w is an integer ranging from 1 to 10, preferably about 6, wherein the alkyl portion of the alkylamino group can be straight chain or branched chain, and 20 wherein the functional group attached to the oligomer via an ester group (-O-C(O)-(CH2)WSH).
In some embodiments, sulfhydryl-activated oligonucleotides are conjugated with polymer moieties such as polyethylene glycol or peptides (via formation of a disulfide bond).
Activated oligomers containing hindered esters as described above can be synthesized by any method known in the art, and in particular by methods disclosed in PCT Publication No. WO 5 2008/034122 and the examples therein, which is incorporated herein by reference in its entirety.
In still other embodiments, the oligomers of the invention are functionalized by introducing sulfhydryl, amino or hydroxyl groups into the oligomer by means of a functionalizing reagent substantially as described in U.S. Patent Nos. 4,962,029 and 4,914,210, i.e., a substantially linear reagent having a phosphoramidite at one end linked through a hydrophilic spacer chain to 0 the opposing end which comprises a protected or unprotected sulfhydryl, amino or hydroxyl group. Such reagents primarily react with hydroxyl groups of the oligomer. In some embodiments, such activated oligomers have a functionalizing reagent coupled to a 5'-hydroxyl group of the oligomer. In other embodiments, the activated oligomers have a functionalizing reagent coupled to a 3"-hydroxyl group. In still other embodiments, the activated oligomers of 5 the invention have a functionalizing reagent coupled to a hydroxyl group on the backbone of the oligomer. In yet further embodiments, the oligomer of the invention is functionalized with more than one of the functionalizing reagents as described in U.S. Patent Nos. 4,962,029 and 4,914,210, incorporated herein by reference in their entirety. Methods of synthesizing such functionalizing reagents and incorporating them into monomers or oligomers are disclosed in U.S. Patent Nos. 4,962,029 and 4,914,210.
In some embodiments, the 5'-terminus of a solid-phase bound oligomer is functionalized with a dienyl phosphoramidite derivative, followed by conjugation of the deprotected oligomer with, e.g., an amino acid or peptide via a Diels-Alder cycloaddition reaction.
In various embodiments, the incorporation of monomers containing 2'-sugar modifications, such as a 2'-carbamate substituted sugar or a 2'-(O-pentyl-N-phthalimido)- deoxyribose sugar into the oligomer facilitates covalent attachment of conjugated moieties to the sugars of the oligomer. In other embodiments, an oligomer with an amino-containing linker at the 2'-position of one or more monomers is prepared using a reagent such as, for example, 5'-dimethoxytrityl-2'-0-(e-phthalimidylaminopentyl)-21-deoxyadenosine-3'- N,N-diisopropyl- cyanoethoxy phosphoramidite. See, e.g., Manoharan, et al., Tetrahedron Letters, 1991 , 34, 7171. In still further embodiments, the oligomers of the invention may have amine-containing functional moieties on the nucleotide, including on the N6 purine amino groups, on the exocyclic N2 of guanine, or on the N4 or 5 positions of cytosine. In various embodiments, such functionalization may be achieved by using a commercial reagent that is already functionalized in the oligomer synthesis. Some functional moieties are commercially available, for example, heterobifunctional and homobifunctional linking moieties are available from the Pierce Co. (Rockford, III.). Other commercially available linking groups are δ'-Amino-Modifier C6 and 3'-Amino-Modifier reagents, both available from Glen Research Corporation (Sterling, Va.). δ'-Amino-Modifier C6 is also available from ABI (Applied Biosystems Inc., Foster City, Calif.) as Aminolink-2, and 3'-Amino- Modifier is also available from Clontech Laboratories Inc. (Palo Alto, Calif.).
Therapy and pharmaceutical compositions - formulation and administration
As explained initially, the oligonucleotides of the invention will constitute suitable drugs with improved properties. The design of a potent and safe drug requires the fine-tuning of various parameters such as affinity/specificity, stability in biological fluids, cellular uptake, mode of action, pharmacokinetic properties and toxicity.
Accordingly, in a further aspect the present invention relates to a pharmaceutical composition comprising an oligonucleotide according to the invention and a pharmaceutically acceptable diluent, carrier or adjuvant. Preferably said carrier is saline or buffered saline.
In a still further aspect the present invention relates to an oligonucleotide according to the present invention for use as a medicament. As will be understood, dosing is dependent on severity and responsiveness of the disease state to be treated, and the course of treatment lasting from several days to several months, or until a cure is effected or a diminution of the disease state is achieved. Optimal dosing schedules can be calculated from measurements of drug accumulation in the body of the patient. Optimum dosages may vary depending on the relative potency of individual oligonucleotides. Generally it can be estimated based on EC50s found to be effective in in vitro and in vivo animal models. In general, dosage is from 0.01 μg to 1 g per kg of body weight, and may be given once or more daily, weekly, monthly or yearly, or even once every 2 to 10 years or by continuous infusion for hours up to several months. The repetition rates for dosing can be estimated based on measured residence times and concentrations of the drug in bodily fluids or tissues. Following successful treatment, it may be desirable to have the patient undergo maintenance therapy to prevent the recurrence of the disease state.
As indicated above, the invention also relates to a pharmaceutical composition, which comprises at least one oligonucleotide of the invention as an active ingredient. It should be understood that the pharmaceutical composition according to the invention optionally comprises a pharmaceutical carrier, and that the pharmaceutical composition optionally comprises further compounds, such as chemotherapeutic compounds, anti-inflammatory compounds, antiviral compounds and/or immuno-modulating compounds.
The oligonucleotides of the invention can be used "as is" or in form of a variety of pharmaceutically acceptable salts. As used herein, the term "pharmaceutically acceptable salts" refers to salts that retain the desired biological activity of the herein-identified oligonucleotides and exhibit minimal undesired toxicological effects. Non-limiting examples of such salts can be formed with organic amino acid and base addition salts formed with metal cations such as zinc, calcium, bismuth, barium, magnesium, aluminum, copper, cobalt, nickel, cadmium, sodium, potassium, and the like, or with a cation formed from ammonia, Λ/,Λ/-dibenzylethylene-diamine, D-glucosamine, tetraethylammonium, or ethylenediamine.
In one embodiment of the invention, the oligonucleotide may be in the form of a prodrug. Oligonucleotides are by virtue negatively charged ions. Due to the lipophilic nature of cell membranes the cellular uptake of oligonucleotides are reduced compared to neutral or lipophilic equivalents. This polarity "hindrance" can be avoided by using the pro-drug approach (see e.g. Crooke, R. M. (1998) in Crooke, S. T. Antisense research and Application. Springer-Verlag, Berlin, Germany, vol. 131 , pp. 103-140).
Pharmaceutically acceptable binding agents and adjuvants may comprise part of the formulated drug. Examples of delivery methods for delivery of the therapeutic agents described herein, as well as details of pharmaceutical formulations, salts, may are well described elsewhere for example in US provisional application 60/838,710 and 60/788,995, which are hereby incorporated by reference, and Danish applications, PA 2006 00615 which is also hereby incorporated by reference.
Pharmaceutical compositions of the present invention include, but are not limited to, solutions, emulsions, and liposome-containing formulations. These compositions may be generated from a variety of components that include, but are not limited to, preformed liquids, self- emulsifying solids and self-emulsifying semisolids. Delivery of drug to tumour tissue may be enhanced by carrier-mediated delivery including, but not limited to, cationic liposomes, cyclodextrins, porphyrin derivatives, branched chain dendrimers, polyethylenimine polymers, nanoparticles and microspheres (Dass CR. J Pharm Pharmacol 2002; 54(1 ):3-27). The pharmaceutical formulations of the present invention, which may conveniently be presented in unit dosage form, may be prepared according to conventional techniques well known in the pharmaceutical industry. Such techniques include the step of bringing into association the active ingredients with the pharmaceutical carrier(s) or excipient(s). In general the formulations are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product. The compositions of the present invention may be formulated into any of many possible dosage forms such as, but not limited to, tablets, capsules, gel capsules, liquid syrups, soft gels and suppositories. The compositions of the present invention may also be formulated as suspensions in aqueous, non-aqueous or mixed media. Aqueous suspensions may further contain substances which increase the viscosity of the suspension including, for example, sodium carboxymethylcellulose, sorbitol and/or dextran. The suspension may also contain stabilizers. The compounds of the invention may also be conjugated to active drug substances, for example, aspirin, ibuprofen, a sulfa drug, an antidiabetic, an antibacterial or an antibiotic. In another embodiment, compositions of the invention may contain one or more oligonucleotide compounds, targeted to a first microRNA and one or more additional oligonucleotide compounds targeted to a second microRNA target. Two or more combined compounds may be used together or sequentially.
The compounds disclosed herein are useful for a number of therapeutic applications as indicated above. In general, therapeutic methods of the invention include administration of a therapeutically effective amount of an oligonucleotide to a mammal, particularly a human. In a certain embodiment, the present invention provides pharmaceutical compositions containing (a) one or more compounds of the invention, and (b) one or more chemotherapeutic agents. When used with the compounds of the invention, such chemotherapeutic agents may be used individually, sequentially, or in combination with one or more other such chemotherapeutic agents or in combination with radiotherapy. All chemotherapeutic agents known to a person skilled in the art are here incorporated as combination treatments with compound according to the invention. Other active agents, such as anti-inflammatory drugs, including but not limited to nonsteroidal anti-inflammatory drugs and corticosteroids, antiviral drugs, and immuno- modulating drugs may also be combined in compositions of the invention. Two or more combined compounds may be used together or sequentially.
Examples of therapeutic indications which may be treated by the pharmaceutical compositions of the invention:
Figure imgf000040_0001
Tumor suppressor gene tropomysin 1 (TPM1 ) mRNA has been indicated as a target of miR-21. Myotrophin (mtpn) mRNA has been indicated as a target of miR 375.
In an even further aspect, the present invention relates to the use of an oligonucleotide according to the invention for the manufacture of a medicament for the treatment of a disease selected from the group consisting of: atherosclerosis, hypercholesterolemia and hyperlipidemia; cancer, glioblastoma, breast cancer, lymphoma, lung cancer; diabetes, metabolic disorders; myoblast differentiation; immune disorders.
The invention further refers to oligonucleotides according to the invention for the use in the treatment of from a disease selected from the group consisting of: atherosclerosis, hypercholesterolemia and hyperlipidemia; cancer, glioblastoma, breast cancer, lymphoma, lung cancer; diabetes, metabolic disorders; myoblast differentiation; immune disorders.
The invention provides for a method of treating a subject suffering from a disease or condition selected from from the group consisting of: atherosclerosis, hypercholesterolemia and hyperlipidemia; cancer, glioblastoma, breast cancer, lymphoma, lung cancer; diabetes, metabolic disorders; myoblast differentiation; immune disorders, the method comprising the step of administering an oligonucleotide or pharmaceutical composition of the invention to the subject in need thereof.
The invention further provides for a kit comprising a pharmaceutical composition according to the invention, and a second independent active ingredient that is an inhibitor of the VLDL assembly pathway, such as an ApoB inhibitor, or an MTP inhibitor.
Cancer
In an even further aspect, the present invention relates to the use of an oligonucleotide according to the invention for the manufacture of a medicament for the treatment of cancer. In another aspect, the present invention concerns a method for treatment of, or prophylaxis against, cancer, said method comprising administering an oligonucleotide of the invention or a pharmaceutical composition of the invention to a patient in need thereof.
Such cancers may include lymphoreticular neoplasia, lymphoblastic leukemia, brain tumors, gastric tumors, plasmacytomas, multiple myeloma, leukemia, connective tissue tumors, lymphomas, and solid tumors.
In the use of a compound of the invention for the manufacture of a medicament for the treatment of cancer, said cancer may suitably be in the form of a solid tumor. Analogously, in the method for treating cancer disclosed herein said cancer may suitably be in the form of a solid tumor. Furthermore, said cancer is also suitably a carcinoma. The carcinoma is typically selected from the group consisting of malignant melanoma, basal cell carcinoma, ovarian carcinoma, breast carcinoma, non-small cell lung cancer, renal cell carcinoma, bladder carcinoma, recurrent superficial bladder cancer, stomach carcinoma, prostatic carcinoma, pancreatic carcinoma, lung carcinoma, cervical carcinoma, cervical dysplasia, laryngeal papillomatosis, colon carcinoma, colorectal carcinoma and carcinoid tumors. More typically, said carcinoma is selected from the group consisting of malignant melanoma, non-small cell lung cancer, breast carcinoma, colon carcinoma and renal cell carcinoma. The malignant melanoma is typically selected from the group consisting of superficial spreading melanoma, nodular melanoma, lentigo maligna melanoma, acral melagnoma, amelanotic melanoma and desmoplastic melanoma.
Alternatively, the cancer may suitably be a sarcoma. The sarcoma is typically in the form selected from the group consisting of osteosarcoma, Ewing's sarcoma, chondrosarcoma, malignant fibrous histiocytoma, fibrosarcoma and Kaposi's sarcoma. Alternatively, the cancer may suitably be a glioma. A further embodiment is directed to the use of an oligonucleotide according to the invention for the manufacture of a medicament for the treatment of cancer, wherein said medicament further comprises a chemotherapeutic agent selected from the group consisting of adrenocorticosteroids, such as prednisone, dexamethasone or decadron; altretamine (hexalen, hexamethylmelamine (HMM)); amifostine (ethyol); aminoglutethimide (cytadren); amsacrine (M- AMSA); anastrozole (arimidex); androgens, such as testosterone; asparaginase (elspar); bacillus calmette-gurin; bicalutamide (casodex); bleomycin (blenoxane); busulfan (myleran); carboplatin (paraplatin); carmustine (BCNU, BiCNU); chlorambucil (leukeran); chlorodeoxyadenosine (2-CDA, cladribine, leustatin); cisplatin (platinol); cytosine arabinoside (cytarabine); dacarbazine (DTIC); dactinomycin (actinomycin-D, cosmegen); daunorubicin (cerubidine); docetaxel (taxotere); doxorubicin (adriomycin); epirubicin; estramustine (emcyt); estrogens, such as diethylstilbestrol (DES); etopside (VP-16, VePesid, etopophos); fludarabine (fludara); flutamide (eulexin); 5-FUDR (floxuridine); 5-fluorouracil (5-FU); gemcitabine (gemzar); goserelin (zodalex); herceptin (trastuzumab); hydroxyurea (hydrea); idarubicin (idamycin); ifosfamide; IL-2 (proleukin, aldesleukin); interferon alpha (intron A, roferon A); irinotecan (camptosar); leuprolide (lupron); levamisole (ergamisole); lomustine (CCNU); mechlorathamine (mustargen, nitrogen mustard); melphalan (alkeran); mercaptopurine (purinethol, 6-MP); methotrexate (mexate); mitomycin-C (mutamucin); mitoxantrone (novantrone); octreotide (sandostatin); pentostatin (2-deoxycoformycin, nipent); plicamycin (mithramycin, mithracin); prorocarbazine (matulane); streptozocin; tamoxifin (nolvadex); taxol (paclitaxel); teniposide (vumon, VM-26); thiotepa; topotecan (hycamtin); tretinoin (vesanoid, all-trans retinoic acid); vinblastine (valban); vincristine (oncovin) and vinorelbine (navelbine). Suitably, the further chemotherapeutic agent is selected from taxanes such as Taxol, Paclitaxel or Docetaxel.
Similarly, the invention is further directed to the use of an oligonucleotide according to the invention for the manufacture of a medicament for the treatment of cancer, wherein said treatment further comprises the administration of a further chemotherapeutic agent selected from the group consisting of adrenocorticosteroids, such as prednisone, dexamethasone or decadron; altretamine (hexalen, hexamethylmelamine (HMM)); amifostine (ethyol); aminoglutethimide (cytadren); amsacrine (M-AMSA); anastrozole (arimidex); androgens, such as testosterone; asparaginase (elspar); bacillus calmette-gurin; bicalutamide (casodex); bleomycin (blenoxane); busulfan (myleran); carboplatin (paraplatin); carmustine (BCNU, BiCNU); chlorambucil (leukeran); chlorodeoxyadenosine (2-CDA, cladribine, leustatin); cisplatin (platinol); cytosine arabinoside (cytarabine); dacarbazine (DTIC); dactinomycin (actinomycin-D, cosmegen); daunorubicin (cerubidine); docetaxel (taxotere); doxorubicin (adriomycin); epirubicin; estramustine (emcyt); estrogens, such as diethylstilbestrol (DES); etopside (VP-16, VePesid, etopophos); fludarabine (fludara); flutamide (eulexin); 5-FUDR (floxuridine); 5- fluorouracil (5-FU); gemcitabine (gemzar); goserelin (zodalex); herceptin (trastuzumab); hydroxyurea (hydrea); idarubicin (idamycin); ifosfamide; IL-2 (proleukin, aldesleukin); interferon alpha (intron A, roferon A); irinotecan (camptosar); leuprolide (lupron); levamisole (ergamisole); lomustine (CCNU); mechlorathamine (mustargen, nitrogen mustard); melphalan (alkeran); mercaptopurine (purinethol, 6-MP); methotrexate (mexate); mitomycin-C (mutamucin); mitoxantrone (novantrone); octreotide (sandostatin); pentostatin (2-deoxycoformycin, nipent); plicamycin (mithramycin, mithracin); prorocarbazine (matulane); streptozocin; tamoxifin (nolvadex); taxol (paclitaxel); teniposide (vumon, VM-26); thiotepa; topotecan (hycamtin); tretinoin (vesanoid, all-trans retinoic acid); vinblastine (valban); vincristine (oncovin) and vinorelbine (navelbine). Suitably, said treatment further comprises the administration of a further chemotherapeutic agent selected from taxanes, such as Taxol, Paclitaxel or Docetaxel.
Alternatively stated, the invention is furthermore directed to a method for treating cancer, said method comprising administering an oligonucleotide of the invention or a pharmaceutical composition according to the invention to a patient in need thereof and further comprising the administration of a further chemotherapeutic agent. Said further administration may be such that the further chemotherapeutic agent is conjugated to the compound of the invention, is present in the pharmaceutical composition, or is administered in a separate formulation. Infectious diseases
It is contemplated that the compounds of the invention may be broadly applicable to a broad range of infectious diseases, such as diphtheria, tetanus, pertussis, polio, hepatitis B, hepatitis C, hemophilus influenza, measles, mumps, and rubella.
Hsa-miR122 is indicated in hepatitis C infection and as such oligonucleotides according to the invention which target miR-122 may be used to treat Hepatitus C infection.
Accordingly, in yet another aspect the present invention relates the use of an oligonucleotide according to the invention for the manufacture of a medicament for the treatment of an infectious disease, as well as to a method for treating an infectious disease, said method comprising administering an oligonucleotide according to the invention or a pharmaceutical composition according to the invention to a patient in need thereof.
In a preferred embodiment, the invention provides for a combination treatment providing an anti miR-122 oligomer in combination with an inhibitor of VLDL assembly, such as an inhibitor of apoB, or of MTP.
Inflammatory diseases The inflammatory response is an essential mechanism of defense of the organism against the attack of infectious agents, and it is also implicated in the pathogenesis of many acute and chronic diseases, including autoimmune disorders. In spite of being needed to fight pathogens, the effects of an inflammatory burst can be devastating. It is therefore often necessary to restrict the symptomatology of inflammation with the use of anti-inflammatory drugs. Inflammation is a complex process normally triggered by tissue injury that includes activation of a large array of enzymes, the increase in vascular permeability and extravasation of blood fluids, cell migration and release of chemical mediators, all aimed to both destroy and repair the injured tissue.
In yet another aspect, the present invention relates to the use of an oligonucleotide according to the invention for the manufacture of a medicament for the treatment of an inflammatory disease, as well as to a method for treating an inflammatory disease, said method comprising administering an oligonucleotide according to the invention or a pharmaceutical composition according to the invention to a patient in need thereof.
In one preferred embodiment of the invention, the inflammatory disease is a rheumatic disease and/or a connective tissue diseases, such as rheumatoid arthritis, systemic lupus erythematous (SLE) or Lupus, scleroderma, polymyositis, inflammatory bowel disease, dermatomyositis, ulcerative colitis, Crohn's disease, vasculitis, psoriatic arthritis, exfoliative psoriatic dermatitis, pemphigus vulgaris and Sjorgren's syndrome, in particular inflammatory bowel disease and Crohn's disease.
Alternatively, the inflammatory disease may be a non-rheumatic inflammation, like bursitis, synovitis, capsulitis, tendinitis and/or other inflammatory lesions of traumatic and/or sportive origin.
Metabolic diseases
A metabolic disease is a disorder caused by the accumulation of chemicals produced naturally in the body. These diseases are usually serious, some even life threatening. Others may slow physical development or cause mental retardation. Most infants with these disorders, at first, show no obvious signs of disease. Proper screening at birth can often discover these problems. With early diagnosis and treatment, metabolic diseases can often be managed effectively.
In yet another aspect, the present invention relates to the use of an oligonucleotide according to the invention or a conjugate thereof for the manufacture of a medicament for the treatment of a metabolic disease, as well as to a method for treating a metabolic disease, said method comprising administering an oligonucleotide according to the invention or a conjugate thereof, or a pharmaceutical composition according to the invention to a patient in need thereof. In one preferred embodiment of the invention, the metabolic disease is selected from the group consisting of Amyloidosis, Biotinidase, OMIM (Online Mendelian Inheritance in Man), Crigler Najjar Syndrome, Diabetes, Fabry Support & Information Group, Fatty acid Oxidation Disorders, Galactosemia, Glucose-6-Phosphate Dehydrogenase (G6PD) deficiency, Glutaric aciduria, International Organization of Glutaric Acidemia, Glutaric Acidemia Type I, Glutaric Acidemia, Type II, Glutaric Acidemia Type I, Glutaric Acidemia Type-ll, F-HYPDRR - Familial Hypophosphatemia, Vitamin D Resistant Rickets, Krabbe Disease, Long chain 3 hydroxyacyl CoA dehydrogenase deficiency (LCHAD), Mannosidosis Group, Maple Syrup Urine Disease, Mitochondrial disorders, Mucopolysaccharidosis Syndromes: Niemann Pick, Organic acidemias, PKU, Pompe disease, Porphyria, Metabolic Syndrome, Hyperlipidemia and inherited lipid disorders, Trimethylaminuria: the fish malodor syndrome, and Urea cycle disorders.
Liver disorders In yet another aspect, the present invention relates to the use of an oligonucleotide according to the invention or a conjugate thereof for the manufacture of a medicament for the treatment of a liver disorder, as well as to a method for treating a liver disorder, said method comprising administering an oligonucleotide according to the invention or a conjugate thereof, or a pharmaceutical composition according to the invention to a patient in need thereof. In one preferred embodiment of the invention, the liver disorder is selected from the group consisting of Biliary Atresia, Alagille Syndrome, Alpha-1 Antitrypsin, Tyrosinemia, Neonatal Hepatitis, and Wilson Disease.
Other uses
The oligonucleotides of the present invention can be utilized for as research reagents for diagnostics, therapeutics and prophylaxis. In research, the oligonucleotide may be used to specifically inhibit the synthesis of target genes in cells and experimental animals thereby facilitating functional analysis of the target or an appraisal of its usefulness as a target for therapeutic intervention. In diagnostics the oligonucleotides may be used to detect and quantitate target expression in cell and tissues by Northern blotting, in-situ hybridisation or similar techniques. For therapeutics, an animal or a human, suspected of having a disease or disorder, which can be treated by modulating the expression of target is treated by administering the oligonucleotide compounds in accordance with this invention. Further provided are methods of treating an animal particular mouse and rat and treating a human, suspected of having or being prone to a disease or condition, associated with expression of target by administering a therapeutically or prophylactically effective amount of one or more of the oligonucleotide compounds or compositions of the invention.
Therapeutic use of oligonucleotides targeting miR-122a
We have demonstrated that a LNA-antimiR, targeting miR-122a reduces plasma cholesterol levels. Therefore, another aspect of the invention is use of the above described oligonucleotides targeting miR-122a as medicine.
Still another aspect of the invention is use of the above described oligonucleotides targeting miR-122a for the preparation of a medicament for treatment of increased plasma cholesterol levels (or hypercholesterolemia and related disorders). The skilled man will appreciate that increased plasma cholesterol levels is undesireable as it increases the risk of various conditions, e.g. atherosclerosis. Still another aspect of the invention is use of the above described oligonucleotides targeting miR-122a for upregulating the mRNA levels of Nrdg3, Aldo A, Bckdk or CD320. EMBODIMENTS
The following embodiments of the present invention may be used in combination with the other embodiments described herein.
1. A pharmaceutical composition comprising an oligomer of between 6-12 nucleotides in length, wherein said oligomer comprises a contiguous nucleotide sequence of a total of between 6-12 nucleotides, such as 6, 7, 8, 9, 10, 11 or 12 nucleotide units, wherein at least 50% of the nucleobase units of the oligomer are high affinity nucleotide analogue units, and a pharmaceutically acceptable diluent, carrier, salt or adjuvant.
2. The pharmaceutical composition according to embodiment 1 , wherein the contiguous nucleotide sequence is complementary to a corresponding region of a mammalian, human or viral microRNA (miRNA) sequence.
3. The pharmaceutical composition according to embodiment 2, wherein the contiguous nucleotide sequence is complementary to a corresponding region of a miRNA sequence selected from the group of miRNAs listed in any one of tables 3, 4 or 5.
4. The pharmaceutical composition according to embodiment 2 or 3, wherein the contiguous nucleotide sequence consists of or comprises a sequence which is complementary to the seed sequence of said microRNA. 5. The pharmaceutical composition according to any one of embodiments 2 - 4, wherein the contiguous nucleotide sequence consists of or comprises a sequence selected from any one of the sequences listed in table 3 or 4.
6. The pharmaceutical composition according to embodiment 4 or 5, wherein the 3' nucleobase of the seedmer forms the 3' most nucleobase of the contiguous nucleotide sequence, wherein the contiguous nucleotide sequence may, optionally, comprise one or two further 5' nucleobases.
7. The pharmaceutical composition according to any one of embodiments 1-6, wherein said contiguous nucleotide sequence does not comprise a nucleotide which corresponds to the first nucleotide present in the micro RNA sequence counted from the 5' end. 8. The pharmaceutical composition according to any one of embodiments 1-7, wherein the contiguous nucleotide sequence is complementary to a corresponding nucleotide sequence present in a miRNA selected from those shown in table 3 or 4 or 5. 9. The pharmaceutical composition according to embodiment 8, wherein said miRNA is selected from the group consisting of miR-1 , miR-IOb, miR-17-3p, miR-18, miR-19a, miR-19b, miR-20, miR-21 , miR-34a, miR-93, miR-106a, miR-106b, miR-122, miR-133, miR-134, miR-138, miR-155, miR-192, miR-194, miR-221 , miR-222, and miR-375. 10. The pharmaceutical composition according to any one of embodiments 1-9, wherein at least 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or all of the nucleobase units of the contiguous nucleotide sequence are nucleotide analogue units.
11. The pharmaceutical composition according to embodiment 10, wherein the nucleotide analogue units are selected from the group consisting of 2'-O_alkyl-RNA unit, 2'-OMe-RNA unit, 2'-amino-DNA unit, 2'-fluoro-DNA unit, LNA unit, PNA unit, HNA unit, INA unit, and a 2'MOE RNA unit.
12. The pharmaceutical composition according to embodiment 10 or 11 , wherein at least 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or all of the nucleobase units of the contiguous nucleotide sequence are Locked Nucleic Acid (LNA) nucleobase units.
13. The pharmaceutical composition according to embodiment 12, wherein all of the nucleobase units of the contiguous nucleotide sequence are LNA nucleobase units.
14. The pharmaceutical composition according to any one of embodiments 1 - 13, wherein the contiguous nucleotide sequence comprises or consists of 7, 8, 9 or 10, preferably contiguous, LNA nucleobase units.
15. The pharmaceutical composition according to any one of embodiments 1-14, wherein the oligomer consist of 7, 8, 9 or 10 contiguous nucleobase units and wherein at least 7 nucleobase units are nucleotide analogue units.
16. The pharmaceutical composition according to embodiment 15, wherein the nucleotide analogue units are Locked Nucleic Acid (LNA) nucleobase units.
17. The pharmaceutical composition according to embodiment 15, wherein the nucleotide analogue units in the molecule consists of a mixture of at least 50% LNA units and up to 50 % other nucleotide analogue units.
18. The pharmaceutical composition according to any one of embodiments 1 - 17, wherein at least 75%, such as 80% or 85% or 90% or 95% or all of the internucleoside linkages present between the nucleobase units of the contiguous nucleotide sequence are phosphorothioate internucleoside linkages.
19. The pharmaceutical composition according to any one of embodiments 1 - 18, wherein said oligomer is conjugated with one or more non-nucleobase compounds. 20. The pharmaceutical composition according to any one of embodiments 1 - 19, wherein the contiguous nucleotide sequence is complementary to the corresponding sequence of at least two miRNA sequences such as 2, 3, 4, 5, 6, 7, 8, 9, or 10 miRNA sequences. 21. The pharmaceutical composition according to any one of embodiments 1 - 20, wherein the contiguous nucleotide sequence consists or comprises of a sequence which is complementary to the sequence of at least two miRNA seed region sequences such as 2, 3, 4, 5, 6, 7, 8, 9, or 10 miRNA seed region sequences. 22. The pharmaceutical composition according to any one of embodiments 20 or 21 , wherein the contiguous nucleotide sequence is complementary to the corresponding region of both miR-221 and miR-222.
23. The pharmaceutical composition according to embodiment 22, wherein the contiguous nucleotide sequence consists or comprises of a sequence that is complementary to
5'GCUACAU3'.
24. The pharmaceutical composition according to any one of embodiments 1 - 23, wherein the oligomer is constituted as a prodrug.
25. The pharmaceutical composition according to any one of embodiments 1 - 24, wherein the contiguous nucleotide sequence is complementary to a corresponding region of has-miR-
122.
26. The pharmaceutical composition according to embodiment 25, for use in the treatment of a medical disorder or disease selected from the group consisting of: hepatitis C virus infection and hypercholesterolemia and related disorders. 27. The pharmaceutical composition according to embodiment 25 or 26, wherein the composition further comprises a second independent active ingredient that is an inhibitor of the VLDL assembly pathway, such as an ApoB inhibitor, or an MTP inhibitor.
28. A kit comprising a pharmaceutical composition according to embodiment 25 or 26, and a second independent active ingredient that is an inhibitor of the VLDL assembly pathway, such as an ApoB inhibitor, or an MTP inhibitor.
29. A method for the treatment of a disease or medical disorder associated with the presence or overexpression of a microRNA, comprising the step of administering a the pharmaceutical composition) according to any one of embodiments 1 - 28 to a patient who is suffering from, or is likely to siffer from said disease or medical disorder. 30. An oligomer, as defined according to anyone of embodiments 1 - 25.
31. A conjugate comprising the oligomer according to embodiment 30, and at least one non- nucleobase compounds.
32. The use of an oligomer or a conjugate as defined in any one of embodiments 30 - 31 , for the manufacture of a medicament for the treatment of a disease or medical disorder associated with the presence or over-expression of the microRNA.
33. A method for reducing the amount, or effective amount, of a miRNA in a cell, comprising administering an oligomer, a conjugate or a pharmaceutical composition, according to any one of the preceeding embodiments to the cell which is expressing said miRNA so as to reduce the amount, or effective amount of the miRNA in the cell. 34. A method for de-repression of a mRNA whose expression is repressed by a miRNA in a cell comprising administering an oligomer, a conjugate or a pharmaceutical composition, according to any one of the preceeding embodiments to the cell to the cell which expressed both said mRNA and said miRNA, in order to de-repress the expression of the mRNA. References: Details of the reference are provided in the priority documents.
EXAMPLES LNA Monomer and oligonucleotide synthesis were performed using the methodology referred to in Examples 1 and 2 of WO2007/112754. The stability of LNA oligonucletides in human or rat plasma is performed using the methodology referred to in Example 4 of WO2007/112754. The treatment of in vitro cells with LNA anti-miR antisense oligonucleotide (targeting miR-122) is performed using the methodology referred to in Example 6 of WO2007/112754. The analysis of Oligonucleotide Inhibition of miR expression by microRNA specific quantitative PCR in both an in vitro and in vivo model is performed using the methodology referred to in Example 7 of WO2007/112754. The assessment of LNA antimir knock-down specificity using miRNA microarray expression profiling is performed using the methodology referred to in Example 8 of WO2007/112754. The detection of microRNAs by in situ hybridization is performed using the methodology referred to in Example 9 of WO2007/112754. The Isolation and analysis of mRNA expression (total RNA isolation and cDNA synthesis for mRNA analysis) in both an in vitro and in vivo model is performed using the methodology referred to in Example 10 of WO2007/112754. In vivo Experiments using Oligomers of the invention targeting microRNA- 122. and subsequent analysis are performed using the methods disclosed in Examples 11 - 27 of WO2007/112754. The above mentioned examples of WO2007/112754 are hereby specifically incorporated by reference.
Example 1: Design of the LNA antimiR oligonucleotides and melting temperatures
Table 2 - Oligomers used in the examples and figures. The SEQ# is an identifier used throughout the examples and figures - the SEQ ID NO which is used in the sequence listing is also provided.
Figure imgf000049_0001
Figure imgf000050_0001
Example 2: In vitro model: Cell culture
The effect of LNA oligonucleotides on target nucleic acid expression (amount) can be tested in any of a variety of cell types provided that the target nucleic acid is present at measurable levels. Target can be expressed endogenously or by transient or stable transfection of a nucleic acid encoding said nucleic acid.
The expression level of target nucleic acid can be routinely determined using, for example, Northern blot analysis (including microRNA northern), Quantitative PCR (including microRNA qPCR), Ribonuclease protection assays. The following cell types are provided for illustrative purposes, but other cell types can be routinely used, provided that the target is expressed in the cell type chosen. Cells were cultured in the appropriate medium as described below and maintained at 37°C at 95-98% humidity and 5% CO2. Cells were routinely passaged 2-3 times weekly.
15PC3: The human prostate cancer cell line 15PC3 was kindly donated by Dr. F. Baas, Neurozintuigen Laboratory, AMC, The Netherlands and was cultured in DMEM (Sigma) + 10% fetal bovine serum (FBS) + Glutamax I + gentamicin.
PC3: The human prostate cancer cell line PC3 was purchased from ATCC and was cultured in
F12 Coon's with glutamine (Gibco) + 10% FBS + gentamicin.
518A2: The human melanoma cancer cell line 518A2 was kindly donated by Dr. B. Jansen, Section of experimental Oncology, Molecular Pharmacology, Department of Clinical
Pharmacology, University of Vienna and was cultured in DMEM (Sigma) + 10% fetal bovine serum (FBS) + Glutamax I + gentamicin.
HeLa: The cervical carcinoma cell line HeLa was cultured in MEM (Sigma) containing 10% fetal bovine serum gentamicin at 37°C, 95% humidity and 5% CO2. MPC-11 : The murine multiple myeloma cell line MPC-11 was purchased from ATCC and maintained in DMEM with 4mM Glutamax+ 10% Horse Serum.
DU-145: The human prostate cancer cell line DU-145 was purchased from ATCC and maintained in RPMI with Glutamax + 10% FBS.
RCC-4 +/- VHL: The human renal cancer cell line RCC4 stably transfected with plasmid expressing VHL or empty plasmid was purchased from ECACC and maintained according to manufacturers instructions.
786-0: The human renal cell carcinoma cell line 786-0 was purchased from ATCC and maintained according to manufacturers instructions
HUVEC: The human umbilical vein endothelial cell line HUVEC was purchased from Camcrex and maintained in EGM-2 medium.
K562: The human chronic myelogenous leukaemia cell line K562 was purchased from ECACC and maintained in RPMI with Glutamax + 10% FBS. U87MG: The human glioblastoma cell line
U87MG was purchased from ATCC and maintained according to the manufacturers instructions. B16: The murine melanoma cell line B16 was purchased from ATCC and maintained according to the manufacturers instructions.
LNCap: The human prostate cancer cell line LNCap was purchased from ATCC and maintained in RPMI with Glutamax + 10% FBS
Huh-7: Human liver, epithelial like cultivated in Eagles MEM with 10 % FBS, 2mM Glutamax I, 1x non-essential amino acids, Gentamicin 25 μg/ml L428: (Deutsche Sammlung fϋr Mikroorganismen (DSM, Braunschwieg, Germany)): Human B cell lymphoma maintained in RPMI 1640 supplemented with 10% FCS, L-glutamine and antibiotics.
L1236: (Deutsche Sammlung fϋr Mikroorganismen (DSM, Braunschwieg, Germany)): Human B cell lymphoma maintained in RPMI 1640 supplemented with 10% FCS, L-glutamine and antibiotics.
Example 3: Design of a LNA antimiR library for all human microRNA sequences in miRBase microRNA database.
The miRBase version used was version 12, as reported in Griffiths-Jones, S., Grocock, RJ. , van Dongen, S., Bateman, A., Enright, A.J. 2006. miRBase: microRNA sequences, targets and gene nomenclature. Nucleic Acids Res. 34: D140-4, and available via http://microrna. sanger.ac.uk/sequences/index.shtml.
Table 1 shows 7, 8 and 9mer nucleotide sequences comprising the seedmer sequence of micro RNA's according to the miRBase micro RNA database. The seedmer sequence comprises the reverse complement of the microRNA seed region. In some emboidments the oligomer of the invention has a contiguous nucleotide sequence selected from the 7mer, 8mer or 9mer sequences. With respect to the 7mer, 8mer and 9mer sequences, in some embodiments, all the intemucleoside linkages are phosphorothioate. The 7mer, 8mer and 9mer nucleotide sequences may consist of sequence of nucleotide analogues as described herein, such as LNA nucleotide analogues. LNA cytosines may be methyl-cytosine (5'methyl-cytosine). In some embodiments, the LNA is beta-D-oxy-LNA.
Table 3 provides a list of microRNAs grouped into those which can be targeted by the same seedmer oligomers, such as the 7, 8 or 9mers provided herein (see table 1 ).
Table 3 hsa-let-7a*, hsa-let-7f-r hsa-let-7a, hsa-let-7b, hsa-let-7c, hsa-Iet-7d, hsa-let-7f, hsa-miR-98, hsa-let-7g, hsa-let-7i hsa-miR-1 , hsa-miR-206 hsa-miR-103, hsa-miR-107 hsa-miR-1 Oa, hsa-miR-1 Ob hsa-miR-125b, hsa-miR-125a-5p hsa-miR-129*, hsa-miR-129-3p hsa-miR-130a, hsa-miR-301a, hsa-miR-130b, hsa-miR-454, hsa-miR-301 b hsa-miR-133a, hsa-miR-133b hsa-miR-135a, hsa-miR-135b hsa-miR-141 , hsa-miR-200a hsa-miR-146a, hsa-miR-146b-5p hsa-miR-152, hsa-miR-148b hsa-miR-154*, hsa-miR-487a hsa-miR-15a, hsa-miR-16, hsa-miR-15b, hsa-miR-195, hsa-miR-497 hsa-miR-17, hsa-miR-20a, hsa-miR-93, hsa-miR-106a, hsa-miR-106b, hsa-miR-20b, hsa-miR-526b* hsa-miR-181a, hsa-miR-181c hsa-miR-181 b, hsa-miR-181d hsa-miR-18a, hsa-miR-18b hsa-miR-190, hsa-miR-190b hsa-miR-192, hsa-miR-215 hsa-miR-196a, hsa-miR-196b hsa-miR-199a-3p, hsa-miR-199b-3p hsa-miR-199a-5p, hsa-miR-199b-5p hsa-miR-19a*, hsa-miR-19b-1*, hsa-miR-19b-2* hsa-miR-19a, hsa-miR-19b hsa-miR-200b, hsa-miR-200c hsa-miR-204, hsa-miR-211 hsa-miR-208a, hsa-miR-208b hsa-miR-212, hsa-miR-132 hsa-miR-23a*, hsa-miR-23b* hsa-miR-23a, hsa-miR-23b, hsa-miR-130a* hsa-miR-24-1*, hsa-miR-24-2* hsa-miR-25, hsa-miR-92a, hsa-miR-367, hsa-miR-92b hsa-miR-26a, hsa-miR-26b hsa-miR-26a-1*, hsa-miR-26a-2* hsa-miR-27a, hsa-miR-27b hsa-miR-29a, hsa-miR-29b, hsa-miR-29c hsa-miR-302a, hsa-miR-302b, hsa-miR-302c, hsa-miR-302d, hsa-miR-373, hsa-miR-520e, hsa-miR-
520a-3p, hsa-miR-520b, hsa-miR-520c-3p, hsa-miR-520d-3p hsa-miR-302b*, hsa-miR-302d* hsa-miR-30a*, hsa-miR-30d*, hsa-miR-30e* hsa-tniR-30a, hsa-miR-30c, hsa-miR-30d, hsa-miR-30b, hsa-miR-30e hsa-miR-330-5p, hsa-miR-326 hsa-miR-34a, hsa-miR-34c-5p, hsa-miR-449a, hsa-miR-449b hsa-miR-362-3p, hsa-miR-329 hsa-miR-374a, hsa-miR-374b hsa-miR-376a, hsa-miR-376b hsa-miR-378, hsa-miR-422a hsa-miR-379*, hsa-miR-411* hsa-miR-381, hsa-miR-300 hsa-miR-509-5p, hsa-miR-509-3-5p hsa-miR-515-5p, hsa-miR-519e* hsa-miR-516b*, hsa-miR-516a-3p hsa-miR-517a, hsa-miR-517c hsa-miR-518a-5p, hsa-miR-527 hsa-miR-518f, hsa-miR-518b, hsa-miR-518c, hsa-miR-518a-3p, hsa-miR-518d-3p hsa-miR-519c-3p, hsa-miR-519b-3p, hsa-miR-519a hsa-miR-519c-5p, hsa-miR-519b-5p, hsa-miR-523*, hsa-miR-518f*, hsa-miR-526a, hsa-miR-520c- 5p, hsa-miR-518e*, hsa-miR-518d-5p, hsa-miR-522*. hsa-miR-519a* hsa-miR-519e, hsa-miR-33b* hsa-miR-520a-5p, hsa-miR-525-5p hsa-miR-520g, hsa-miR-520h hsa-miR-524-5p, hsa-miR-520d-5p hsa-miR-525-3p, hsa-miR-524-3p hsa-miR-548b-5p, hsa-miR-548a-5p, hsa-miR-548c-5p, hsa-miR-548d-5p hsa-miR-7-1*, hsa-miR-7-2* hsa-miR-99a, hsa-miR-100, hsa-miR-99b
We have constructed an 8-mer LNA-antimiR against miR-21 , miR-155 and miR-122 (designated here as micromiR) that is fully LNA modified and phosphorothiolated (see figure 1 and Table 6). Our results from repeated experiments in MCF-7, HeLa, Raw and Huh-7 cells using a luciferase sensor plasmid for miR-21 , miR-155 and miR-122 demonstrate that the fully LNA-modified short LNA-antimiRs are highly potent in antagonizing microRNAs.
Figure imgf000054_0001
The melting temperatures can be assessed towards the mature microRNA sequence, using a synthetic microRNA oligonucleotide (typically consisting of RNA nucleotides with a phosphodiester backbone). Typically measured Tms are higher than predicted Tms when using LNA oligomers against the RNA target.
Example 4: Assessment of miR-21 antagonism by SEQ ID #3205 LNA-antimiR in MCF-7 cells using a luciferase sensor assay.
In order to assess the efficiency of a fully LNA-modified 8-mer LNA-antimiR (SEQ ID #3205) oligonucleotide in targeting and antagonizing miR-21, luciferase sensor constructs were made containing a perfect match target site for the mature miR-21 and as control, a target site with two mutations in the seed (Fig. 6). In order to monitor microRNA-21 inhibition, the breast carcinoma cell line MCF-7 was transfected with the different luciferase constructs together with the miR-21 antagonist SEQ ID #3205 at varying concentrations in comparison with a 15-mer LNA-antimiR SEQ ID #3204 against miR-21. After 24 hours, luciferase activity was measured.
Results: As seen in Figure 2, the new fully LNA-modified 8-mer LNA-antimiR (SEQ ID #3205) shows two-fold higher potency compared to SEQ ID #3204, as shown by de-repression of the Iuciferase activity. By contrast, the control miR-21 sensor construct with two mismatches in the miR-21 seed did not show any de-repression of the firefly Iuciferase activity, thereby demonstrating the specificity of the perfect match miR-21 sensor in monitoring miR-21 activity in cells. The de-repression of Iuciferase activity by the 8-mer LNA-antimiR is clearly dose- dependent, which is not seen with SEQ ID #3204. Moreover, the new 8-mer is also much more potent at lower doses than SEQ ID #3204.
To conclude, the 8-mer LNA-antimiR (SEQ ID #3205) shows significantly improved potency in inhibition of miR-21 in vitro compared to the 15-mer LNA-antimiR SEQ ID #3204 targeting miR- 21.
Materials and Methods:
Cell line: The breast carcinoma cell line MCF-7 was purchased from ATCC (#HTB-22™). MCF- 7 cells were cultured in EMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax, 1x NEAA and 25 ug/ml Gentamicin. Transfection: 400.000 cells were seeded per well in a 6-well plate the day before transfection in order to receive 50-70% confluency the next day. On the day of transfection, MCF-7 cells were transfected with 0.8 ug miR-21 perfect match/psiCHECK2, miR-21.mm2/psiCHECK2 or empty psiCHECK2 vector (SDS Promega) together with 1 μl Lipofectamine2000 (Invitrogen) according to manufacturer's instructions. After 24 hours, cells were harvested for Iuciferase measurements. Lucif erase assay: The cells were washed with PBS and harvested with cell scraper, after which cells were centrifugated for 5 min at 10.000 rpm. The supernatant was discarded and 50 μl 1 x Passive Lysis Buffer (Promega) was added to the cell pellet, after which cells were put on ice for 30 min. The lysed cells were spinned at 10.000 rpm for 30 min after which 20 μl were transferred to a 96 well plate and Iuciferase measurements were performed according to manufacturer's instructions (Promega).
Example 5: Assessment of miR-21 antagonism by SEQ ID #3205 LNA-antimiR in HeLa cells using a Iuciferase sensor assay.
To further assess the efficiency of the fully LNA-modified 8-mer LNA-antimiR SEQ ID #3205 in targeting miR-21 , the cervix carcinoma cell line HeLa was also transfected with the previously described miR-21 Iuciferase sensor constructs alongside SEQ ID #3205 at varying concentrations as described in the above section (Figure 3).
Results: The SEQ ID #3205 shows complete de-repression of the miR-21 Iuciferase sensor construct in HeLa cells already at 5 nM compared to SEQ ID #3204, which did not show complete de-repression until the highest dose (50 nM). In addition, antagonism of miR-21 by the
8-mer SEQ ID #3205 LNA-antimiR is dose-dependent. To demonstrate the specificity of the miR-21 luciferase sensor assay, a mismatched miR-21 target site (2 mismatches in seed) was also transfected into HeLa cells, but did not show any de-repression of the firefly luciferase activity.
To conclude, the fully LNA-modified SEQ ID #3205 shows significantly improved potency in inhibition of miR-21 in vitro, in both MCF-7 and HeLa cells compared to the 15-mer LNA-antimiR SEQ ID #3204.
Materials and Methods:
Cell line: The human cervix carcinoma cell line HeLa was purchased from ECACC (#93021013). HeLa cells were cultured in EMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax, 1x NEAA and 25 ug/ml Gentamicin.
Transfection: 60.000 cells were seeded per well in a 24 well plate the day before transfection in order to receive 50-70% confluency the next day. On the day of transfection, HeLa cells were transfected with 0.2 ug miR-21 perfect match/psiCHECK2, miR-21.mm2/psiCHECK2 or empty psiCHECK2 vector together with 0,7 μl Lipofectamine2000 (Invitrogen) according to manufacturer's instructions. After 24 hours, cells were harvested for luciferase measurements. Luciferase assay: The cells were washed with PBS and 100 μl 1 x Passive Lysis Buffer (Promega) was added to each well, after which the 24 well plates was put on an orbital shaker for 30 min. The cells were collected and transferred to an eppendorf tube and spinned at 10.000 rpm for 30 min after which 10 μl were transferred to a 96 well plate and luciferase measurements were performed according to manufacturer's instructions (Promega).
Example 6: Assessment of miR-155 antagonism by SEQ ID #3207 LNA-antimiR in mouse RAW cells using a luciferase sensor assay.
To ask whether a fully LNA-modified 8-mer LNA-antimiR can effectively antagonize miR-155, a perfect match target site for miR-155 was cloned into the same luciferase vector (psiCHECK2) and transfected into the mouse leukaemic monocyte macrophage RAW cell line. Because the endogenous levels of miR-155 are low in the RAW cell line, the cells were treated with 100 ng/ml LPS for 24 hours in order to induce miR-155 accumulation.
Results: Luciferase measurements showed that the fully LNA-modified 8-mer LNA-antimiR SEQ ID #3207 targeting miR-155 was similarly effective in antagonizing miR-155 compared to the 15-mer LNA-antimiR SEQ ID #3206 (Figure 4). Both LNA-antimirs showed a >50% derepression of the miR-155 luciferase sensor at 0.25 nM concentration and inhibited miR-155 in a dose-dependent manner.
Conclusion: These data further support the results from antagonizing miR-21, as shown in examples 1 and 2, demonstrating that a fully thiolated 8-mer LNA-antimiR is highly potent in microRNA targeting. Materials and Methods:
Cell line: The mouse leukaemic monocyte macrophage RAW 264.7 was purchased from ATCC (TIB-71 ). RAW cells were cultured in DMEM medium, supplemented with 10% fetal bovine serum, 4 mM Glutamax and 25 ug/ml Gentamicin. Transfection: 500.000 cells were seeded per well in a 6 well plate the day before transfection in order to receive 50% confluency the next day. On the day of transfection, MCF-7 cells were transfected with 0.3 ug miR-155 or empty psiCHECK2 vector together with 10 μl Lipofectamine2000 (Invitrogen) according to manufacturer's instructions. In order to induce miR- 155 accumulation, LPS (100 ng/ml) was added to the RAW cells after the 4 hour incubation with the transfection complexes. After another 24 hours, cells were harvested for luciferase measurements.
Luciferase assay: The cells were washed with PBS and harvested with cell scraper, after which cells were centrifugated for 5 min at 2.500 rpm. The supernatant were discarded and 50 μl 1 x Passive Lysis Buffer (Promega) was added to the cell pellet, after which cells were put on ice for 30 min. The lysed cells were spinned at 10.000 rpm for 30 min after which 20 μl were transferred to a 96 well plate and luciferase measurments were performed according to manufacturer's instructions (Promega).
Example 7: Assessment of miR-122 antagonism by SEQ ID #3208 LNA-antimiR in HuH-7 cells using a luciferase sensor assay.
The potency of the fully modified 8-mer LNA-antimiR SEQ ID #3208 against miR-122 was assessed in the human hepatoma cell line HuH-7. The HuH-7 cells were transfected with luciferase sensor construct containing a perfect match miR-122 target site. After 24 hours luciferase measurements were performed (Figure 5). Results: The fully LNA-modified 8-mer LNA-antimiR SEQ ID #3208 is more potent than the 15- mer LNA-antimiR SEQ ID #4 at low concentration, as shown by de-repression of the miR-122 luciferase sensor. Both LNA-antimiRs inhibit miR-122 in a dose-dependet manner (Figure 5).
Conclusion: The fully LNA-modified 8-mer LNA-antimiR SEQ ID #3208 targeting miR-122 shows improved potency in inhibition of miR-122 in vitro. Materials and Methods:
Cell line: The human hepatoma cell line HuH-7 was a kind gift from R. Bartenschlager,
Heidelberg. Huh-7 cells were cultured in EMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax, 1x NEAA and 25 ug/ml Gentamicin.
Transfection: 8.000 cells were seeded per well in a 96 well plate the day before transfection in order to receive 50-70% confluency the next day. On the day of transfection, HuH-7 cells were transfected with 57 ng miR-122 or empty psiCHECK2 vector together with 1 μl Lipofectamine2000 (Invitrogen). After 24 hours, cells were harvested for luciferase measurements.
Luciferase assay: 50 μl 1 x Passive Lysis Buffer (Promega) was added to each well, after which the 96 well plate was put on an orbital shaker for 30 min. To each well the Dual-luciferase Reporter assay system (Promega) was added and luciferase measurements were performed according to manufacturer's instructions (Promega).
Example 8. Assessment of miR-21 antagonism by comparing an 8-mer (SEQ ID #3205) versus a 15-mer (SEQ ID #3204) LNA-antimiR in human prostate carcinoma cells (PC3). We have previously shown (patent application 1051 ), that an 8-mer LNA-antimiR that is fully LNA-modified and phosphorothiolated is able to completely de-repress the miR-21 luciferase reporter levels in the human cervix carcinoma cell line HeLa and partly de-repress the miR-21 luciferase reporter levels in the human breast carcinoma cell line MCF-7. We next extended this screening approach to the human prostate cancer cell line PC3. To assess the efficiency of the different LNA-antimiR oligonucleotides against miR-21 , luciferase reporter constructs were generated in which a perfect match target site for the mature miR-21 and a target site with two mismatches in the seed were cloned in the 3'UTR of Renilla luciferase gene (Figure 7). In order to monitor miR-21 inhibition, PC3 cells were transfected with the different luciferase constructs together with the miR-21 antagonist SEQ ID #3205 (8-mer) and for comparison with the 15-mer LNA-antimiR perfect match SEQ ID #3204 at varying concentrations. After 24 hours, luciferase activity was measured.
Results: The luciferase reporter experiments showed a dose-dependent de-repression of the luciferase miR-21 reporter activity with the 15-mer LNA-antimiR against miR-21 (SEQ ID #3204). However, complete de-repression of the luciferase reporter was not obtained even at the highest concentrations (Figure 7). In contrast, the cells that were transfected with the 8-mer fully LNA substituted LNA-antimiR showed complete de-repression already at 1 nM, indicating significantly improved potency compared to the 15-mer LNA-antimiR. The luciferase control reporter harboring a mismatch target site for miR-21 was not affected by either LNA-antimiR, demonstrating high specificity of both LNA-antimiRs. Conclusion: The micromer is far more potent than the 15-mer LNA-antimiR in targeting miR-21 and has so far shown to be most potent in prostate carcinoma cells. Materials and Methods:
Cell line: The human prostate carcinoma PC3 cell line was purchased from ECACC (#90112714). PC3 cells were cultured in DMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax and 25 ug/ml Gentamicin.
Transfection: 100.000 cells were seeded per well in a 12-well plate the day before transfection in order to receive 50% confluency the next day. On the day of transfection, PC3 cells were transfected with 0.3 μg miR-21 or empty psiCHECK2 vector together with 1 ,2 μl Lipofectamine2000 (Invitrogen) according to manufacturer's instructions. Transfected was also varying concentrations of LNA-antimiRs. After 24 hours, cells were harvested for luciferase measurements. Luciferase assay: The cells were washed with PBS and 250 μl 1 x Passive Lysis Buffer
(Promega) was added to the wells. The plates were placed on a shaker for 30 min., after which the cell lysates were transferred to eppendorf tubes. The cell lysate was centrifugated for 10 min at 2.500 rpm after which 20 μl were transferred to a 96 well plate and luciferase measurements were performed according to manufacturer's instructions (Promega).
Example 9. Specificity assessment of miR-21 antagonism by an 8-mer LNA-antimiR To investigate the specificity of our short LNA-antimiR targeting miR-21 , we designed an 8-mer mismatch control LNA-antimiR (SEQ ID #3218) containing 2 mismatches in the seed recognition sequence (see Figure 8). The luciferase reporter constructs described in example 1 were transfected into the human cervix carcinoma cell line HeLa together with the LNA mismatch control oligo SEQ ID #3218 and its efficacy was compared with the 8-mer LNA- antimiR (SEQ ID #3205) targeting miR-21. After 24 hours, luciferase activity was measured. Results: As shown in Figure 8, transfection of the fully LNA-modified 8-mer LNA-antimiR in HeLa cells resulted in complete de-repression of the luciferase miR-21 reporter already at 5 nM. In contrast, when the cells were transfected with the 8-mer LNA mismatch control oligo, combined with the results obtained with the control miR-21 luciferase reporter having two mismatches in the miR-21 seed, these data demonstrate high specificity of the fully LNA- subsituted 8-mer LNA-antimiR in targeting miR-21 in HeIa cells. Analysis of the miRBase microRNA sequence database showed that the miR-21 recognition sequence, of the LNA-antimiR SEQ ID #3205 is unique for microRNA-21. However, when decreasing the micromer length to 7 nt, it is not specific for only miR-21 , since ath-miR-844, mmu-miR-590-3p and has-miR-590-3p are also targeted.
Conclusion:Exhanging two nucleotide positions within the 8-mer LNA-antimiR with two mismatching nucleotides completely abolished the antagonizing activity of the LNA-antimiR for miR-21.
Materials and Methods:
Cell line: The human cervix carcinoma cell line HeLa was purchased from ECACC (#93021013). HeLa cells were cultured in EMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax, 1x NEAA and 25 ug/ml Gentamicin. Transfection: 60.000 cells were seeded per well in a 24-well plate the day before transfection in order to receive 50-70% confluency the next day. On the day of transfection, HeLa cells were transfected with 0.2 ug miR-21 perfect match/psiCHECK2, miR-21.mm2/psiCHECK2 or empty psiCHECK2 vector together with 0,7 μl Lipofectamine2000 (Invitrogen) according to manufacturer's instructions. Transfected was also varying concentrations of LNA-antimiRs. After 24 hours, cells were harvested for luciferase measurements. Luciferase assay: The cells were washed with PBS and 100 μl 1 x Passive Lysis Buffer
(Promega) was added to each well, after which the 24-well plates were put on an orbital shaker for 30 min. The cells were collected and transferred to an eppendorf tube and spinned at 10.000 rpm for 30 min after which 10 μl were transferred to a 96-well plate and luciferase measurements were performed according to manufacturer's instructions (Promega).
Example 10. Assessment of the shortest possible length of a fully LNA-modified LNA- antimiR that mediates effective antagonism of miR-21.
To further investigate the LNA-antimiR length requirements, we designed a 7-mer and a 6-mer LNA-antimiR targeting miR-21, both fully LNA-modified and phosphorothiolated oligonucleotides. The miR-21 luciferase reporter constructs were transfected into HeLa cells along with the LNA-antimiRs at varying concentrations. Luciferase measurements were performed after 24 hours.
Results: As seen in Figure 9, the 7-mer LNA-antimiR mediates de-repression of the miR-21 luciferase reporter plasmid, but at lower potency compared to the 8-mer LNA-antimiR (SEQ ID #3205). Nevertheless, a dose-dependent trend can still be observed. By contrast, the 6-mer LNA-antimiR did not show any inhibitory activity.
Conclusion: To conclude, the shortest possible length of an LNA-antimiR which is able to mediate miR-21 inhibition is 7 nucleotides. However, the 7-mer LNA-antimiR is less potent compared to the 8-mer LNA-antimiR for miR-21. Materials and Methods:
Cell line: The human cervix carcinoma cell line HeLa was purchased from ECACC (#93021013). HeLa cells were cultured in EMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax, 1x NEAA and 25 ug/ml Gentamicin. Transfection: 60.000 cells were seeded per well in a 24 well plate the day before transfection in order to receive 50-70% confluency the next day. On the day of transfection, HeLa cells were transfected with 0.2 ug miR-21 perfect match/psiCHECK2, miR-21.mm2/psiCHECK2 or empty psiCHECK2 vector together with 0,7 μl Lipofectamine2000 (Invitrogen) according to manufacturer's instructions. Transfected was also varying concentrations of LNA-antimiRs. After 24 hours, cells were harvested for luciferase measurements. Luciferase assay: The cells were washed with PBS and 100 μl 1 x Passive Lysis Buffer
(Promega) was added to each well, after which the 24-well plates was put on an orbital shaker for 30 min. The cells were collected and transferred to an eppendorf tube and spinned at 10.000 rpm for 30 min after which 10 μl were transferred to a 96-well plate and luciferase measurements were performed according to manufacturer's instructions (Promega).
Example 11. Length assessment of fully LNA-substituted LNA-antimiRs antagonizing miR-21
Next, we investigated the effect of increasing the length from a 9-mer to a 14-mer fully LNA substituted LNA-antimiRs on antagonizing miR-21 in HeLa cells. The resulting LNA-antimiRs were transfected into HeLa cells together with the miR-21 luciferase reporter constructs (Figure 10). Luciferase measurements were performed after 24 hours.
Results: The 9-mer LNA-antimiR SEQ ID #3211 (9-mer) showed dose-dependent derepression of the miR-21 luciferase reporter which did not reach complete de-repression, as demonstrated for the 7-mer LNA-antimiR (SEQ ID #3210). Increasing the length to 10-mer to 14-mer (SEQ ID #3212, SEQ ID #3213 and SEQ ID #3214) decreased the potency as shown by less efficient de-repression of the miR-21 reporter.
Conclusion: As shown in Figure 10, the longest fully LNA-modified and phosphorothiolated LNA-antimiR which is still able to mediate miR-21 inhibition is a 9-mer LNA-antimiR SEQ ID #3211. However, it is clearly less efficient than the 7-mer and 8-mer LNA-antimiRs. Materials and Methods: Cell line: The human cervix carcinoma cell line HeLa was purchased from ECACC (#93021013). HeLa cells were cultured in EMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax, 1x NEAA and 25 ug/ml Gentamicin. Transfection: 60.000 cells were seeded per well in a 24-well plate the day before transfection in order to achieve 50-70% confluency the next day. On the day of transfection, HeLa cells were transfected with 0.2 ug miR-21 perfect match/psiCHECK2, miR-21. mm2/psiCHECK2 or empty psiCHECK2 control vector without target site together with 0,7 μl Lipofectamine2000 (Invitrogen) according to manufacturer's instructions. Transfected was also varying concentrations of LNA-antimiRs. After 24 hours, cells were harvested for luciferase measurements. Luciferase assay: The cells were washed with PBS and 100 μl 1 x Passive Lysis Buffer (Promega) was added to each well, after which the 24-well plates were put on an orbital shaker for 30 min. The cells were collected and transferred to an eppendorf tube and spinned at 10.000 rpm for 30 min after which 10 μl were transferred to a 96-well plate and luciferase measurements were performed according to manufacturer's instructions (Promega).
Example 12. Determination of the most optimal position for an 8-mer LNA-antimiR within the miR target recognition sequence. Our experiments have shown that the most potent fully LNA-modified phosphorothiolated LNA- antimiR is 8 nucleotides in length. To assess the most optimal position for an 8-mer LNA- antimiR within the miR target recognition sequence, we designed four different fully LNA- modified 8-mer LNA-antimiRs tiled across the mature miR-21 sequence as shown in Figure 11. The different LNA-antimiRs were co-transfected together with the miR-21 luciferase reporter constructs into HeLa cells. Luciferase measurements were performed after 24 hours. Results: The only LNA-antimiR that mediated efficient silencing of miR-21 as measured by the luciferase reporter was SEQ ID #3205, which targets the seed region of miR-21. Neither SEQ ID #3215 which was designed to cover the 3'end of the seed (50% seed targeting) did not show any effect, nor did the other two LNA-antimiRs SEQ ID #3216 or SEQ ID #3217, which were positioned to target the central region and the 3'end of the mature miR-21 , respectively. Conclusion: The only 8-mer LNA-antimiR mediating potent silencing of miR-21 is the one targeting the seed of the miR-21. Materials and Methods: Cell line: The human cervix carcinoma cell line HeLa was purchased from ECACC
(#93021013). HeLa cells were cultured in EMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax, 1x NEAA and 25 ug/ml Gentamicin.
Transfection: 60.000 cells were seeded per well in a 24-well plate the day before transfection in order to achieve 50-70% confluency the next day. On the day of transfection, HeLa cells were transfected with 0.2 ug miR-21 perfect match/psiCHECK2, miR-21.mm2/psiCHECK2 or empty psiCHECK2 vector together with 0,7 μl Lipofectamine2000 (Invitrogen) according to the manufacturer's instructions. Transfected was also varying concentrations of LNA-antimiRs. After 24 hours, cells were harvested for luciferase measurements. Luciferase assay: The cells were washed with PBS and 100 μl 1 x Passive Lysis Buffer (Promega) was added to each well, after which the 24-well plates was put on an orbital shaker for 30 min. The cells were collected and transferred to an eppendorf tube and spinned at 10.000 rpm for 30 min after which 10 μl were transferred to a 96 well plate and luciferase measurements were performed according to manufacturer's instructions (Promega).
Example 13. Validation of interaction of the miR-21 target site in the Pdcd4-3'-UTR and miR-21 using the δ-mer SEQ ID #3205 LNA-antimiR.
The tumour suppressor protein Pdcd4 inhibits TPA-induced neoplastic transformation, tumour promotion and progression. Pdcd4 has also been shown to be upregulated in apoptosis in response to different inducers. Furthermore, downregulation of Pdcd4 in lung and colorectal cancer has also been associated with a poor patient prognosis. Recently, Asangani efa/ and Frankel et al showed that the Pdcd4-3'-UTR contains a conserved target site for miR-21 , and transfecting cells with an antimiR-21 , resulted in an increase in Pdcd4 protein. We therefore constructed a luciferase reporter plasmid, harboring 313 nt of the 3'UTR region of Pdcd4 encompassing the aforementioned miR-21 target site, which was co-transfected together with different LNA-antimiRs into HeLa cells. The different LNA-antimiRs were; SEQ ID #3205 (8-mer, perfect match) or SEQ ID #3218 (8-mer, mismatch). Luciferase measurements were performed after 24 hours.
Results: As shown in Figure 12, in cells transfected with the Pdcd43'UTR luciferase reporter and SEQ ID #3205, an increase in luciferase activity was observed, indicating interaction between the Pdcd4 3'UTR and miR-21. However, transfecting the cells with the mismatch compound, SEQ ID #3218, no change in luciferase activity was observed, which was expected since the compound does not antagonize miR-21. When comparing the 8-mer LNA-antimiR against two longer designed LNA-antimiRs, the short fully LNA-modified and phosphorothiolated LNA-antimiR was significantly more potent, confirming previous luciferase assay data. Conclusion: These data conclude that SEQ ID #3205, which antagonizes miR-21, can regulate the interaction between Pdcd4 3'UTR and miR-21. Materials and Methods:
Cell line: The human cervix carcinoma cell line HeLa was purchased from ECACC (#93021013). HeLa cells were cultured in EMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax, 1x NEAA and 25 ug/ml Gentamicin. Transfection: 60.000 cells were seeded per well in a 24-well plate the day before transfection in order to achieve 50-70% confluency the next day. On the day of transfection, HeLa cells were transfected with 0.2 ug Pdcd4-3'UTR/psiCHECK2 or empty psiCHECK2 vector together with 0,7 μl Lipofectamine2000 (Invitrogen) according to the manufacturer's instructions. Varying concentrations of the LNA-antimiR oligonucleotides were also transfected. After 24 hours, cells were harvested for luciferase measurements.
Luciferase assay: The cells were washed with PBS and 100 μl 1 x Passive Lysis Buffer (Promega) was added to each well, after which the 24-well plates was put on an orbital shaker for 30 min. The cells were collected and transferred to an eppendorf tube and spinned at 10.000 rpm for 30 min after which 10 μl were transferred to a 96 well plate and luciferase measurements were performed according to manufacturer's instructions (Promega).
Example 14. Comparison of an 8-mer LNA-antimiR (SEQ ID #3207) with a 15-mer LNA- antimiR (SEQ ID #3206) in antagonizing miR-155 in mouse RAW cells.
To ask whether our approach of using short LNA-antimiRs could be adapted to targeting other miRNAs we designed a fully LNA-modified 8-mer LNA-antimiR against microRNA-155. A perfect match target site for miR-155 was cloned into the 3'UTR of the luciferase gene in the reporter plasmid psiCHECK2 and transfected into the mouse RAW macrophage cell line together with an 8-mer or a 15-mer LNA-antimiR. Because the endogenous levels of miR-155 are low in the RAW cell line, the cells were treated with 100 ng/ml LPS for 24 hours in order to induce miR- 155 accumulation. After 24 hours, luciferase analysis was performed. Results: Luciferase measurements showed that the fully LNA-modified 8-mer LNA-antimiR SEQ ID #3207 targeting miR-155 was similarly effective in antagonizing miR-155 compared to the 15-mer LNA-antimiR SEQ ID #3206 (Figure 13). Both LNA-antimiRs showed a >50% derepression of the miR-155 luciferase sensor at 0.25 nM concentration and inhibited miR-155 in a dose-dependent manner. Analysis of the miRBase microRNA sequence database showed that the miR-155 recognition sequence, of the LNA-antimiR SEQ ID #3207 is unique for microRNA-155. However, when decreasing the LNA-antimiR length to 7 nt, it is not specific for only miR-155, mdv1-miR-M4 and kshv-miR-K12-11 is also targeted. .Conclusion: A fully LNA-modified and phosphorothiolated 8-mer LNA-antimiR is equally potent compared with a 15-mer LNA-antimiR of a mixed LNA/DNA design in antagonizing miR-155. Thus, our approach of using short LNA-antimiRs can be readily adapted to targeting of other miRNAs
Materials and Methods: Cell line: The mouse macrophage RAW 264.7 cell line was purchased from ATCC (TIB-71 ). RAW cells were cultured in DMEM medium, supplemented with 10% fetal bovine serum, 4 mM Glutamax and 25 ug/ml Gentamicin.
Transfection: 500.000 cells were seeded per well in a 6-well plate the day before transfection in order to receive 50% confluency the next day. On the day of transfection, RAW 264.7 cells were transfected with 0.3 ug miR-155 perfect match/psiCHECK2 or empty psiCHECK2 vector together with 10 μl Lipofectamine2000 (Invitrogen) according to the manufacturer's instructions. Transfected was also varying concentrations of LNA-antimiRs. In order to induce miR-155 accumulation, LPS (100 ng/ml) was added to the RAW cells after the 4 hour incubation with the transfection complexes. After another 24 hours, cells were harvested for luciferase measurements. Luciferase assay: The cells were washed with PBS and harvested with cell scraper, after which cells were spinned for 5 min at 2.500 rpm. The supernatant was discarded and 50 μl 1 x Passive Lysis Buffer (Promega) was added to the cell pellet, after which cells were put on ice for 30 min. The lysed cells were spinned at 10.000 rpm for 30 min after which 20 μl were transferred to a 96-well plate and luciferase measurements were performed according to the manufacturer's instructions (Promega). Example 15. Assessment of c/EBPβ protein levels as a functional readout for miR-155 antagonism by short LNA-antimiR (SEQ ID #3207).
As a functional readout for miR-155 antagonism by short LNA-antimiR (SEQ ID #3207) we determined the protein levels of a novel miR-155 target, c/EBPβ. The mouse macrophage RAW cell line was transfected together with either an 8-mer (SEQ ID #3207) or a 15-mer (SEQ ID #3206) LNA-antimiR in the absence or presence of pre-miR-155. As mismatch controls for the 15-mer, SEQ ID #4 was used, which targets miR-122 and for the 8-mer SEQ ID #3205 was used, which targets miR-21. These two control miRNAs do not regulate c/EBPβ expression levels. LPS was used to induce miR-155 accumulation and cells were harvested after 16 hours with LPS. c/EBPβ has three isoforms; LIP, LAP and LAP* that were detected by Western blot analysis and the same membranes were re-probed with beta-tubulin as loading control. Results: Ratios were calculated for c/EBPβ LIP and beta-tubulin as indicated in Figure 14. RAW cells that were transfected with the 15-mer LNA-antimiR and no pre-miR-155 all showed equal c/EBPβ LIP/beta-tubulin ratios, due to inhibition of miR-155 increases the c/EBPβ LIP levels (Figure 14, left panel). By comparison, transfection of pre-miR-155 in RAW cells resulted in decreased c/EBPβ LIP levels as expected, if c/EBPβ was a miR-155 target, as shown in lanes with protein extracts from RAW cells treated with no LNA or a mismatch. However, protein extracts from RAW cells transfected with LNA-antimiR against miR-155, showed an increase of c/EBPβ LIP levels. The same experiments were also carried out with the 8-mer LNA-antimiR- 155 (SEQ ID #3207) and as shown in Figure 14 (right panel) comparable results to those with the 15-mer LNA-antimiR SEQ ID #3206 were obtained.
Conclusion: Antagonism of miR-155 using either an 8-mer or a 15-mer LNA-antimiR leads to de-repression of the direct target c/EBPβ. Materials and Methods: Cell line: The mouse macrophage RAW 264.7 cell line was purchased from ATCC (TIB-71 ). RAW cells were cultured in DMEM medium, supplemented with 10% fetal bovine serum, 4 mM Glutamax and 25 ug/ml Gentamicin.
Transfection: 500.000 cells were seeded per well in a 6-well plate the day before transfection in order to achieve 50% confluency the next day. On the day of transfection, RAW 264.7 cells were transfected with 5 nmol pre-miR-155 (Ambion) and/or 5 nM LNA-antimiR together with 10 μl Lipofectamine2000 (Invitrogen) according to the manufacturer's instructions. Transfected was also varying concentrations of LNA-antimiRs. In order to induce miR-155 accumulation, LPS (100 ng/ml) was added to the RAW cells after the 4 hour incubation with the transfection complexes. After 16 hours, cells were harvested for protein extraction and western blot analysis. Western blot: Cells were washed with PBS, trypsinated, transferred to eppendorf tubes and 250 μl lysis buffer (IxRIPA) was added. The cell lysate was placed on ice for 20 min and spinned at 10.000 rpm for 10 minutes. The protein concentration was measured with Coomassie Plus according to the manufacturer's instructions and 80 ug was loaded onto a 4-12% BIS-TRIS gel. The membrane was incubated overnight at 40C with the primary monoclonal mouse antibody C/EBP β (Santa Cruz) with a 1 :100 concentration. Immunoreactive bands were visualized with ECL Plus (Amersham).
Example 16. Antagonism of miR-106b by a fully LNA-modified 8-mer (SEQ ID #3221) LNA- antimiR
To confirm that our approach of using short LNA-antimiRs could be adapted to targeting of other miRNAs we designed a fully LNA-modified 8-mer LNA-antimiR against microRNA-106b. A perfect match target site for miR-106b was cloned into the 3'UTR of the luciferase gene in the vector (psiCHECK2) and transfected into the human cervix carcinoma HeLa cell line together with a short LNA-antimiR (SEQ ID #3221 ) or with a 15-mer LNA-antimiR (SEQ ID #3228) at varying concentrations. Luciferase measurements were performed after 24 hours. Results: Transfection of the 8-mer LNA-antimiR SEQ ID #3221 against miR-106b resulted in dose-dependent inhibition of miR-106b as shown by de-repression of the luciferase reporter, which was completely de-repressed at 1 nM LNA-antimiR concentration (Figure 15). Comparable results were obtained using the 15-mer LNA-antimiR SEQ ID #3228 demonstrating that an 8-mer LNA-antimiR is similarly potent to a 15-mer. Conclusion: Targeting of miR-106b in HeLa cells shows that an 8-mer fully LNA-modified and phosphorotiolated LNA-antimiR is equally potent compared with a 15-mer LNA/DNA mixmer LNA-antimiR. Materials and Methods: Cell line: The human cervix carcinoma cell line HeLa was purchased from ECACC (#93021013). HeLa cells were cultured in EMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax, 1x NEAA and 25 ug/ml Gentamicin.
Transfection: 5.200 cells were seeded per well in a 96-well plate the day before transfection in order to achieve 50-70% confluency the next day. On the day of transfection, HeLa cells were transfected with 57 ng miR-21 perfect match/psiCHECK2, miR-21.mm2/psiCHECK2 or empty psiCHECK2 vector together with 0,14 μl Lipofectamine2000 (Invitrogen) according to the manufacturer's instructions. Transfected was also varying concentrations of LNA-antimiRs. After 24 hours, cells were harvested for luciferase measurements.
Luciferase assay: The cells were washed with PBS and 30 μl 1 x Passive Lysis Buffer (Promega) was added to each well, after which the 24-well plates was put on an orbital shaker for 30 min. The cells were collected and transferred to eppendorf tubes and spinned at 10.000 rpm for 30 min after which luciferase measurements were performed according to the manufacturer's instructions (Promega).
Example 17. Antagonism of miR-19a by a fully LNA-modified 8-mer (SEQ ID #3222) LNA- antimiR To further confirm that our approach of using short LNA-antimiRs can be readily adapted to targeting of other miRNAs we designed a fully LNA-modified 8-mer LNA-antimiR against microRNA-19a. A perfect match target site for miR-19a was cloned in the 3'UTR of the luciferase gene in the psiCHECK2 vector. The reporter plasmid was transfected into the human cervix carcinoma HeLa cell line together with a short LNA-antimiR (SEQ ID #3222) or with a 15- mer LNA-antimiR (SEQ ID #3229) targeting miR-19a at varying concentrations. Luciferase measurements were performed after 24 hours.
Results: As shown in Figure 16, transfection of the 15-mer LNA-antimiR SEQ ID #3229 into HeLa efficiently antagonizes miR-19a as demonstrated by complete de-repression at 1 nM LNA-antimiR concentration. By comparison, transfection of the 8-mer LNA-antimiR SEQ ID #3222 resulted in effective miR-19a antagonism already at 0.5 nM concentration, indicating that this 8-mer LNA-antimiR is at least equally potent compared with a 15-mer LNA-antimiR in HeLa cells.
Conclusion: Targeting of miR-19a in HeLa cells shows that an 8-mer fully LNA-modified and phosphorothiolated LNA-antimiR is at least equally potent compared with a 15-mer LNA/DNA mixmer LNA-antimiR.
Materials and Methods: Cell line: The human cervix carcinoma cell line HeLa was purchased from ECACC (#93021013). HeLa cells were cultured in EMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax, 1x NEAA and 25 ug/ml Gentamicin. Transfection: 5.200 cells were seeded per well in a 96-well plate the day before transfection in order to achieve 50-70% confluency the next day. On the day of transfection, HeLa cells were transfected with 57 ng miR-21 perfect match/psiCHECK2, miR-21.mm2/psiCHECK2 or empty psiCHECK2 vector together with 0,14 μl Lipofectamine2000 (Invitrogen) according to manufacturer's instructions. Transfected was also varying concentrations of LNA-antimiRs. After 24 hours, cells were harvested for luciferase measurements. Luciferase assay: The cells were washed with PBS and 30 μl 1 x Passive Lysis Buffer
(Promega) was added to each well, after which the 24-well plates was put on an orbital shaker for 30 min. The cells were collected and transferred to eppendorf tubes and spinned at 10.000 rpm for 30 min after which luciferase measurements were performed according to the manufacturer's instructions (Promega).
Example 18. Targeting of a microRNA family using short, fully LNA-substituted LNA- antimiR. Next, we investigated whether it is possible to target a microRNA family using a single short 7- mer LNA-antimiR complementary to the seed sequence that is common for all family members (see Figure 17). In this experiment, we focused on miR-221 and miR-222 that are overexpressed in solid tumors of the colon, pancreas, prostate and stomach. It has also been shown that miR-221 and miR-222 are the most significantly upregulated microRNAs in glioblastoma multiforme. Furthermore, overexpression of miR-221 and miR-222 may contribute to the growth and progression of prostate carcinoma, at least in part by blocking the tumor suppressor protein p27. A perfect match target site for both miR-221 and miR-222, respectively, was cloned into the 3'UTR of the luciferase gene resulting in two reporter constructs. These constructs were then transfected either separate or combined into the prostate carcinoma cell line, PC3. In addition to the 7-mer, targeting both miR-221 and miR-222, we also co-transfected a 15-mer LNA-antimiR (15mer) targeting either miR-221 (SEQ ID #3223) or miR-222 (SEQ ID #3224), each transfected separately or together (see Figure 18 left). Results: As shown in Figure 18, transfection of PC3 cells with the LNA-antimiR SEQ ID #3223 against miR-221 resulted in efficient inhibition of miR-221 at 1 nM LNA-antimiR concentration. An inhibitory effect is also observed when using the luciferase reporter plasmid for miR-222 as well as when co-transfecting both luciferase reporters for miR-221 and miR-222 simultaneously into PC3 cells. This inhibitory effect is most likely due to the shared seed sequence between miR-221 and miR-222. Similarly, transfection of PC3 cells with the LNA-antimiR SEQ ID #3224 against miR-222 resulted in efficient inhibition of miR-222 at 1 nM LNA-antimiR concentration as shown by complete de-repression of the luciferase reporter for miR-222. An inhibitory effect is also observed when using the luciferase reporter plasmid for miR-222 as well as when co- transfecting both luciferase reporters for miR-221 and miR-222 simultaneously into PC3 cells. Co-tranfection of both LNA-antimiR compounds SEQ ID #3223 and SEQ ID #3224 against miR- 221 and miR-222, respectively, (see Figure 18 left), resulted in effective inhibition of both miRNAs as shown by complete de-repression of the luciferase reporter plasmids both when separately transfected and when co-transfected into PC3 cells. Interestingly, transfection of a single fully LNA-modified 7-mer LNA-antimiR (SEQ ID #3225) targeting the seed sequence of miR-221 and miR-222 into PC3 cells resulted in efficient, dose-dependent antagonism of miR- 221 and miR-222 simultaneously as shown by complete de-repression of the luciferase reporter plasmids both when separately transfected and when co-transfected into PC3 cells. This demonstrates that a single, short LNA-substituted LNA-antimiR can effectively target seed sequences thereby antagonizing entire microRNA families simultaneously. Analysis of the miRBase microRNA sequence database showed that the miR-221 /222 seed recognition sequence, of the LNA-antimiR SEQ ID #3225 is unique for both miRNAs. Conclusion: Our results demonstrate that LNA enables design and synthesis of short fully LNA-substituted LNA-antimiR oligonucleotides that can effectively target microRNA seed sequences thereby antagonizing entire microRNA families simultaneously. Materials and Methods: Ce// line: The human prostate carcinoma PC3 cell line was purchased from ECACC
(#90112714) PC3 cells were cultured in DMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax and 25 ug/ml Gentamicin.
Transfection: 100.000 cells were seeded per well in a 12-well plate the day before transfection in order to receive 50% confluency the next day. On the day of transfection, PC3 cells were transfected with 0.3 ug of luciferase reporter plasmid for miR-221 or for miR-222 or with empty psiCHECK2 vector without miRNA target site as control together with 1 ,2 μl Lipofectamine2000 (Invitrogen) according to the manufacturer's instructions. After 24 hours, cells were harvested for luciferase measurements. Luciferase assay: The cells were washed with PBS and 250 μl 1 x Passive Lysis Buffer (Promega) was added to the wells. The plates were placed on a shaker for 30 min., after which the cell lysates was transferred to eppendorf tubes. The cell lysate was spinned for 10 min at 2.500 rpm after which 20 μl were transferred to a 96-well plate and luciferase measurements were performed according to the manufacturer's instructions (Promega).
Example 19. Assessment of p27 protein levels as a functional readout for antagonism of the miR-221/222 family by the 7-mer SEQ ID #3225 LNA-antimiR.
Previous work has shown (Ie Sage et al. 2007, Galardi et al. 2007) that miR-221 and miR-222 post-transcriptionally regulate the expression of the tumour suppressor gene p27, which is involved in cell cycle regulation. In these studies, down-regulation of miR-221 and miR-222 was shown to increase expression levels of p27. Thus, as a functional readout for antagonism of the miR-221/222 family by the 7-mer SEQ ID #3225 LNA-antimiR we determined the protein levels of p27 after transfection of the LNA-antimiR SEQ ID #3225 into PC3 cells in comparison with an 8-mer LNA mismatch control. After 24 hours the cells were harvested for western blot analysis (Figure 19). Results: As shown in Figure 19, transfection of the 7-mer LNA-antimiR SEQ ID #3225 targeting the seed sequence in miR-221 and miR-222 resulted in dose-dependent increase of the p27 protein levels compared to either untransfected or LNA mismatch control transfected PC3 cells. These results clearly demonstrate that the 7-mer LNA-antimiR is able to effectively antagonize the miR-221/222 family leading to de-repression of the direct target p27 at the protein level. Conclusion: A fully LNA-modified 7-mer LNA-antimiR targeting the seed sequence in the miR- 221/222 family effectively antagonized both miRNAs leading to de-repression of the direct target p27 at the protein level. Materials and Methods: Cell line: The human prostate carcinoma PC3 cell line was purchased from ECACC
(#90112714) PC3 cells were cultured in DMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax and 25 ug/ml Gentamicin.
Transfection: 250.000 cells were seeded per well in a 6-well plate the day before transfection in order to receive 50% confluency the next day. On the day of transfection, PC3 cells were transfected with LNA-antimiRs at varying concentrations with Lipofectamine2000. Cells were harvested after 24 hours for protein extraction and western blot analysis. Western blot: Cells were washed with PBS, trypsinated, transferred to eppendorf tubes and 250 μl lysis buffer (IxRIPA) was added. The cell lysate was placed on ice for 20 min, then spinned at 10.000 rpm for 10 minutes. The protein concentration was measured with Coomassie Plus according to the manufacturer's instructions and 100 ug was loaded onto a 4-12% BIS-TRIS gel. The membrane was incubated overnight at 40C with the primary monoclonal mouse antibody p27 (BD Biosciences) at a 1 :1000 dilution. Immunoreactive bands were visualized with ECL Plus (Amersham).
Example 20. Duplex melting temperatures (Tm) of the LNA-antimiRs.
As shown in Table 5, Tm values increase with increasing the length of short fully modified LNA- antimiRs (see Tm values for SEQ ID #3205, SEQ ID #3209-3214 in Table 7). Most optimal inhibitory effect was achieved with the 8-mer LNA-antimiR SEQ ID #3205 against miR-21 , whereas the very low Tm of the 6-mer SEQ ID #3209 is most likely not sufficient to mediate antagonism of the miR-21 target. On the other hand, increasing the length beyond a 10-mer (SEQ ID #3212) significantly increases the Tm, while simultaneously decreasing the inhibitory activity as measured using the luciferase miR-21 reporter, which is most likely due to high propensity of the fully modified 12- and 14-mer LNA-antimiRs to form homodimers. The experiments using a sliding window of fully LNA-modified 8-mer LNA-antimirs across the mir-21 recognition sequence clearly demonstrate that in addition to adequate Tm value of the LNA- antimiR, the seed region is most critical for miRNA function and, thus, the most optimal region to be targeted by an LNA-antimiR. Table 5: Tm values for miR-21 LNA-antimiRs, measured against a complementary RNA oligonucleotide
Figure imgf000071_0001
Conclusion: The Tm values along with experimental data obtained with luciferase reporters show that potent antagonism by LNA-antimiR is not only dependent on Tm but also depends on the positioning of the LNA-antimiR within the microRNA recognition sequence. Materials and Methods:
Tm measurements: The oligonucleotide:miR-21 RNA duplexes were diluted to 3 μM in 500 μl RNase free H2O and mixed with 500 μl 2x Tm-buffer (200 mM NaCI, 0.2 mM EDTA, 20 mM Na- phosphate, pH 7,0). The solution was heated to 95°C for 3 min and then allowed to anneal in RT for 30 min. The duplex melting temperatures (Tm) were measured on a Lambda 40 UV/VIS Spectrophotometer equipped with a Peltier temperature programmer PTP6 using PE Templab software (Perkin Elmer). The temperature was ramped up from 200C to 95°C and then down to 25°C, recording absorption at 260 nm. First derivative and the local maximums of both the melting and annealing were used to assess the duplex melting temperatures.
Example 21. Assessment of miR-21 antagonism by comparing an 8-mer (SEQ ID #3205) versus a 15-mer (SEQ ID #3204) LNA-antimiR in human hepatocytic cell line HepG2.
We have previously shown in this application, that an 8-mer LNA-antimiR that is fully LNA- modified and phosphorothiolated effectively antagonizes miR-21 in the human cervix carcinoma cell line HeLa, the human breast carcinoma cell line MCF-7 and the human prostate cancer cell line PC3. We extended this screening approach to the human hepatocellular cancer cell line HepG2. To assess the efficiency of the 8-mer LNA-antimiR oligonucleotide against miR-21 , luciferase reporter constructs were generated in which a perfect match target site for the mature miR-21 was cloned into the 3'UTR of the Renilla luciferase gene. In order to monitor miR-21 inhibition, HepG2 cells were transfected with the luciferase constructs together with the miR-21 antagonist SEQ ID #3205 (8-mer) and for comparison of specificity with the 8-mer LNA-antimiR mismatch (SEQ ID #3218) and for comparison of potency together with the 15-mer (SEQ ID #3204) at varying concentrations. After 24 hours, luciferase activity was measured. Results: The luciferase reporter experiments showed a dose-dependent de-repression of the luciferase miR-21 reporter activity with the 15-mer LNA-antimiR against miR-21 (SEQ ID #3204). However, complete de-repression of the luciferase reporter was not obtained, not even at the higher concentrations (Figure 20). In contrast, the cells that were transfected with the 8- mer fully LNA modified LNA-antimiR (SEQ ID #3205) showed complete de-repression already at 5 nM, indicating significantly improved potency compared to the 15-mer LNA-antimiR. Comparing the specificity of the 8-mer perfect match and the 8-mer mismatch, the mismatch LNA-antimiR (SEQ ID #3218) did not show any de-repression at all, demonstrating high specificity of the LNA-antimiR compound against miR-21.
Conclusion: The 8-mer (SEQ ID #3205) is more potent than the 15-mer LNA-antimiR in targeting miR-21 and antagonism of miR-21 by SEQ ID #3205 is specific. Materials and Methods: Ce// line: The human hepatocytic HepG2 cell line was purchased from ECACC (#85011430). HepG2 cells were cultured in EMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax and 25 ug/ml Gentamicin.
Transfection: 650.000 cells were seeded per well in a 6-well plate and reverse transfection were performed. HepG2 cells were transfected with 0.6 μg miR-21 or empty psiCHECK2 vector together with 2,55 μl Lipofectamine2000 (Invitrogen) according to manufacturer's instructions. Transfected was also varying concentrations of LNA-antimiRs. After 24 hours, cells were harvested for luciferase measurements.
Luciferase assay: The cells were washed with PBS and 300 μl 1 x Passive Lysis Buffer (Promega) was added to the wells. The plates were placed on a shaker for 30 min., after which the cell lysates were transferred to eppendorf tubes. The cell lysate was centrifugated for 10 min at 2.500 rpm after which 50 μl were transferred to a 96 well plate and luciferase measurements were performed according to the manufacturer's instructions (Promega).
Example 22. Validation of interaction of the miR-21 target site in the Pdcd4 3'UTR and miR-21 using the 8-mer SEQ ID #3205 LNA-antimiR in human hepatocellular cell line Huh- 7.
The tumour suppressor protein Pdcd4 inhibits tumour promotion and progression. Furthermore, downregulation of Pdcd4 in lung and colorectal cancer has also been associated with poor patient prognosis. Recently, Asangani et al (Oncogene 2007) and Frankel et al (J Biol Chem 2008) showed that the Pdcd4 3'UTR contains a conserved target site for miR-21 , and transfecting cells with an antimiR-21, resulted in an increase in Pdcd4 protein. We therefore constructed a luciferase reporter plasmid, harboring 313 nt of the 3'UTR region of Pdcd4 encompassing the aforementioned miR-21 target site, which was co-transfected together with different LNA-antimiRs and pre-miR-21 (10 nM) into Huh-7 cells. The different LNA-antimiRs were; SEQ ID #3205 (8-mer, perfect match), SEQ ID #3218 (8-mer, mismatch) and SEQ ID #3204 (15-mer, DNA/LNA mixmer). Luciferase measurements were performed after 24 hours. Results: As shown in Figure 21 , cells transfected with the Pdcd4 3'UTR luciferase reporter and SEQ ID #3205, an increase in luciferase activity was observed, indicating interaction between the Pdcd4 3'UTR and miR-21. However, transfecting the cells with the mismatch compound, SEQ ID #3218, no change in luciferase activity was observed, which was expected since the compound does not antagonize miR-21. When comparing the 8-mer LNA-antimiR against the 15-mer LNA-antimiR (SEQ ID #3204), the short fully LNA-modified and phosphorothiolated LNA-antimiR was significantly more potent, confirming previous data. Materials and Methods: Cell line: The human hepatoma cell line Huh-7 was a kind gift from R. Bartinschlager (Dept MoI Virology, University of Heidelberg). Huh-7 cells were cultured in DMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax, 1x NEAA and 25 ug/ml Gentamicin. Transfection: 11.000 cells were seeded per well in a 96-well plate the day before transfection in order to achieve 50-70% confluency the next day. On the day of transfection, Huh-7 cells were transfected with 20 ng Pdcd4 3'UTR/psiCHECK2 or empty psiCHECK2 vector together with 10 nM pre-miR-21 (Ambion) and 0,14 μl Lipofectamine2000 (Invitrogen) according to the manufacturer's instructions. Varying concentrations of the LNA-antimiR oligonucleotides were also transfected. After 24 hours, cells were harvested for luciferase measurements. Luciferase assay: Cells were washed and 30 μl 1 x Passive Lysis Buffer (Promega) was added to each well, after which the 96-well plates was put on an orbital shaker. After 30 min., 50 μl luciferase substrate dissolved in Luciferase Assay Buffer Il (Dual-Luciferase Reporter Assay System from Promega, Cat# E1910) was added to the wells with lysated cells and luciferase measurements were performed according to the manufacturer's instructions (Promega).
Example 23. Assessment of Pdcd4 protein levels as a functional readout for miR-21 antagonism by the 8-mer LNA-antimiR (SEQ ID #3205).
In addition, we also transfected HeLa cells with SEQ ID #3205 (perfect match), SEQ ID #3218 (mismatch), SEQ ID #3219 (scrambled) and analyzed Pdcd4 protein levels after 24 hours with Western blot (Figure 22). As shown, in the protein extracts from cells where SEQ ID #3205 had been added, the Pdcd4 protein levels increase, due to antagonism of mir-21 by SEQ ID #3205 in contrast to the two control LNA oligonucleotides. Conclusion: Antagonism of miR-21 using an 8-mer (SEQ ID #3205) leads to derepression of the direct target Pdcd4ntagonism of miR-21 Materials and Methods:
Cell line: The human cervix carcinoma cell line HeLa was purchased from ECACC (#93021013). HeLa cells were cultured in EMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax, 1x NEAA and 25 ug/ml Gentamicin.
Transfection: 200.000 cells were seeded per well in a 6-well plate the day before transfection in order to receive 50-70% confluency the next day. On the day of transfection, HeLa cells were transfected with 5 nM LNA oligonucleotides and 2,5 μg/ml Lipofectamine2000 (Invitrogen) according to the manufacturer's instructions. After 24 hours, cells were harvested for Western blot analysis.
Western blot: Cells were washed with PBS, trypsinated, transferred to eppendorf tubes and 50 μl lysis buffer (IxRIPA) was added. The cell lysate was placed on ice for 20 min and spinned at 10.000 rpm for 10 minutes. Equal amounts (15 μl cell lysate) were loaded onto a 4-12% BIS- TRIS gel. The proteins were transferred to a nitrocellulose membrane using iBlot (Invitrogen) according to manufacturers instructions. The membrane was incubated overnight at 40C with the primary affinity purified rabbit serum antibody Pdcd4 (Rockland) with a 1 :2000 concentration. As control, anti- beta tubulin antibodies (Thermo Scientific) were used at a 1 :5000 dilution. Immunoreactive bands were visualized with ECL Plus (Amersham).
Example 24. Assessment of potential hepatotoxicity of the 8-mer perfect match LNA- antimiR SEQ ID #3205 and the LNA mismatch control SEQ ID #3218. Each compound was injected into female NMRI mice, at doses of 25 mg/kg, 5 mg/kg and 1 mg/kg, every other day for 2 weeks. The animals were sacrificed and serum was collected from whole blood for ALT and AST analyses. As seen in Figure 23, the ALT and AST levels were not elevated for SEQ ID #3205 compared to saline or SEQ ID #3218 (mismatch control). However, one mouse showed increased levels (marked red), since the serum samples were contaminated with red blood cells, which contain 6-8 times higher levels of ALT and AST compared to plasma. The mice that received 5 mg/kg and 1 mg/kg were also analyzed for ALT and AST levels and showed no changes compared to saline treated control animals (data not shown). Materials and Methods: Experimental design:
Figure imgf000074_0001
Figure imgf000075_0001
Sacrifice; The animals was sacrificed by cervical dislocation.
Sampling of serum for ALT/AST; The animals were anaesthetised with 70% CO2-30% O2 before collection of retro orbital sinus blood. The blood was collected into S-monovette Serum-Gel vials. The serum samples were harvested and stored from each individual mouse. The blood samples were stored at room temperature for two hours and thereafter centrifuged 10 min, 3000 rpm, at room temp. The serum fractions were harvested into Eppendorf tubes on wet ice. ALT and AST measurements; ALT and AST measurements were performed in 96-well plates using ALT and AST reagents from ABX Pentra (A11 A01627 - ALT, A11 A01629 - AST) according to the manufacturer's instructions. In short, serum samples were diluted 2.5 fold with H2O and each sample was assayed in duplicate. After addition of 50 μl diluted sample or standard (multical from ABX Pentra - A11 A01652) to each well, 200 μl of 37 0C AST or ALT reagent mix was added to each well. Kinetic measurements were performed for 5 min with an interval of 30s at 340 nm and 37 0C.
Example 25. Assessment of PU.1 protein levels as a functional readout for miR-155 antagonism by short LNA-antimiR (SEQ ID #3207).
We have previously shown that the 8-mer (SEQ ID #3207) antagonizing miR-155 leads to derepression of the miR-155 target c/EBPbeta in the mouse macrophage RAW cells. To further verify the potency of SEQ ID #3207 we determined the protein levels of another miR-155 target, PU.1 As a functional readout for miR-155 antagonism by short LNA-antimiR (SEQ ID #3207) we performed Western blot. The antagonism was verified in the human monocytic THP-1 cell line which was transfected together with either an 8-mer (SEQ ID #3207) perfect match or a 8-mer control LNA in the absence or presence of pre-miR-155. LPS was used to induce miR-155 accumulation and cells were harvested after 24 hours. Results: THP-1 cells that were transfected with pre-miR-155 shows a decrease in PU.1 levels (Figure 24). Transfecting the cells with the fully LNA-modified and phosphorothiolated SEQ ID #3207 effectively antagonizes miR-155, leading to unaltered levels of PU.1 protein. By comparison, transfecting the cells with an 8-mer LNA control, PU.1 levels decreased, indicating that antagonism of miR-155 by SEQ ID #3207 LNA-antimiR is specific. Conclusion: Antagonism of miR-155 using an 8-mer leads to de-repression of the direct target
PU.1 in human THP-1 cells.
Materials and Methods: Cell line: The human monocytic THP-1 cell line was purchased from ECACC (#88081201 ).
THP-1 cells were cultured in RPMI with L-glutamine, supplemented with 10% fetal bovine serum.
Transfection: 200.000 cells were seeded per well in a 12-well plate the day before. On the day of transfection, THP-1 cells were transfected with 5 nmol pre-miR-155 (Ambion) and/or 5 nM LNA-antimiR together with Lipofectamine2000 (Invitrogen) according to the manufacturer's instructions. LPS (100 ng/ml) was added to the cells after the 4 hour incubation with the transfection complexes. After 24 hours, cells were harvested for protein extraction and western blot analysis.
Western blot: Cells were washed with PBS, trypsinated, transferred to eppendorf tubes and 50 μl lysis buffer (IxRIPA) was added. The cell lysate was placed on ice for 20 min and spinned at
10.000 rpm for 10 minutes. Equal amounts (15 μl cell lysate) were loaded onto a 4-12% BIS-
TRIS gel. The proteins were transferred to a nitrocellulose membrane using iBlot (Invitrogen) according to manufacturers instructions The membrane was incubated overnight at 40C with the rabbit monoclonal PU.1 antibody (Cell Signaling) with a 1:2000 concentration. As equal loading, Tubulin (Thermo Scientific) was used at a 1 :5000 dilution. Immunoreactive bands were visualized with ECL Plus (Amersham).
Example 26. Assessment of p27 protein levels as a functional readout for antagonism of the miR-221/222 family by the 7-mer SEQ ID #3225 LNA-antimiR. Previous work has shown (Ie Sage et al. 2007, Galardi et al. 2007) that miR-221 and miR-222 post-transcriptionally regulate the expression of the tumour suppressor gene p27, which is involved in cell cycle regulation. In these studies, down-regulation of miR-221 and miR-222 was shown to increase expression levels of p27. Thus, as a functional readout for antagonism of the miR-221/222 family by the 7-mer SEQ ID #3225 LNA-antimiR we determined the protein levels of p27 after transfection of the LNA-antimiR SEQ ID #3225 into PC3 cells.
Results: As shown in Figure 25, transfection of the 7-mer LNA-antimiR SEQ ID #3225 targeting the seed sequence of miR-221 and miR-222 resulted in dose-dependent increase of the p27 protein levels compared to either untransfected or our LNA scrambled control transfected PC3 cells. These results clearly demonstrate that the 7-mer LNA-antimiR is able to effectively antagonize the miR-221/222 family leading to de-repression of the direct target p27 at the protein level at concentrations as low as 5 nM. Conclusion: A fully LNA-modified 7-mer LNA-antimiR targeting the seed sequence in the miR- 221/222 family at 5 nM can effectively antagonize both miRNAs leading to de-repression of the direct target p27 at protein level. Materials and Methods: Ce// line: The human prostate carcinoma PC3 cell line was purchased from ECACC
(#90112714). PC3 cells were cultured in DMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax and 25 ug/ml Gentamicin.
Transfection: 250.000 cells were seeded per well in a 6-well plate the day before transfection in order to receive 50% confluency the next day. On the day of transfection, PC3 cells were transfected with LNA-oligonucleotides at varying concentrations (see Figure 25) with
Lipofectamine2000. Cells were harvested after 24 hours for protein extraction and western blot analysis.
Western blot: Cells were washed with PBS, trypsinated, transferred to eppendorf tubes and 50 μl lysis buffer (IxRIPA) was added. The cell lysate was placed on ice for 20 min, then spinned at 10.000 rpm for 10 minutes. Equal amounts (15 μl cell lysate) were loaded onto a 4-12% BIS- TRIS gel. The proteins were transferred to a nitrocellulose membrane using iBlot (Invitrogen) according to manufacturers instructions. The membrane was incubated overnight at 4°C with the primary monoclonal mouse antibody p27 (BD Biosciences) at a 1 :1000 dilution. As loading control, Tubulin (Thermo Scientific) was used at a 1 :5000 dilution. Immunoreactive bands were visualized with ECL Plus (Amersham).
Example 27. Knock-down of miR-221/222 by the 7-mer SEQ ID #3225 LNA-antimiR reduces colony formation of PC3 cells
A hallmark of cellular transformation is the ability for tumour cells to grow in an anchorage- independent way in semisolid medium. We have therefore performed soft agar assay which is a phenotypic assay that is relevant for cancer, given that it measures the decrease of tumour cells. We transfected SEQ ID #3225 (perfect match) and SEQ ID #3231 (scrambled) into PC3 cells, and after 24 hours plated cells in soft agar. Colonies were counted after 12 days. We show in Figure 26 that inhibition of miR-221 and miR-222 by SEQ ID #3225 can reduce the amount of colonies growing in soft agar compared to the scrambled control LNA-antimiR, indicating decrease of tumour cells.
Conclusion: The 7-mer (SEQ ID #3225) targeting the miR-221/222 family reduces the number of colonies in soft agar, indicating proliferation arrest of PC3 cells.
Materials and Methods: Cell line: The human prostate carcinoma PC3 cell line was purchased from ECACC
(#90112714). PC3 cells were cultured in DMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax and 25 ug/ml Gentamicin. Transfection: 250.000 cells were seeded per well in a 6-well plate the day before transfection in order to receive 50% confluency the next day. On the day of transfection, PC3 cells were transfected with 25 nM of different LNA oligonucleotides with Lipofectamine2000. Clonogenic growth in soft agar: 2.5x103 PC3 cells were seeded in 0.35% agar on the top of a base layer containing 0.5% agar. Cells were plated 24 hours after transfection. Plates were incubated in at 37°C, 5%CO2 in a humified incubator for 12 days and stained with 0.005% crystal violet for 1 h, after which cells were counted. The assay was performed in triplicate.
Example 28: Assessment of let-7 antagonism by 6-9-mer LNA-antimiRs in Huh-7 cells transfected with let-7a precursor miRNA, and a luciferase sensor assay.
In order to assess the efficiency of fully LNA-modified 6-9-mer oligonucleotides in targeting and antagonizing the let-7 family of miRNAs, a luciferase sensor construct was made, containing some 800 bp of the HMGA2 31UTR. The sequence cloned into the vector contains four out of seven functional let-7 binding sites (sites 2-5), as previously demonstrated by Mayr et al. (Science, 2007) and Lee and Dutta (Genes Dev, 2007). In order to monitor let-7 inhibition, the hepatocellular carcinoma cell line Huh-7 (with low to non-existing levels of endogenous let-7) was transfected with the luciferase sensor construct, with let-7a precursor miRNA, and with the 6-9 mer let-7 antagonists SEQ ID #3232, -3233, -3227, -3234, -3235; see Figure 27) at increasing concentrations. The 6-9-mer LNA-antimiRs were compared with SEQ ID #3226, a 15-mer against let-7a as a positive control. After 24 hours, luciferase activity was measured. Results: As seen in Figure 28, the fully LNA-modified 8- and 9-mer LNA-antimiRs (SEQ ID #3227, SEQ ID #3234, and SEQ ID #3235) show similar potencies in de-repressing the let-7 targets in the luciferase sensor assay, as the positive control 15-mer SEQ ID #3226. Full target de-repression for these highly potent compounds is achieved already at 1-5 nM, whereas the 7- mer SEQ ID #3233 needs to be present at slightly higher concentrations (10 nM) to generate the same effect. However, the 6-mer SEQ ID #3232 shows no effect even at as high concentrations as 50 nM. The de-repression of luciferase activity by the 7-9- and the 15-mer LNA-antimiRs is dose-dependent, which is particularly clear in the case of the slightly less potent SEQ ID #3233. Conclusion: To conclude, the 8-9-mer LNA-antimiRs (SEQ ID #3227, SEQ ID #3234, and SEQ ID #3235) show equal antagonist potencies in inhibition of let-7a in vitro compared to the 15- mer LNA-antimiR SEQ ID #3226 targeting let-7a. A potent effect, albeit at slightly higher concentrations is also seen for the 7-mer SEQ ID #3233, whereas a 6-mer has no effect at tested concentrations. Materials and Methods:
Cell line: The hepatocellular carcinoma cell line Huh-7 was a kind gift from R. Bartinschlager (Dept MoI Virology, University of Heidelberg).Huh-7 cells were cultured in DMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax, 1x NEAA and 25 ug/ml Gentamicin.
Transfection: 8,000 cells were seeded per well in a 96-well plate the day before transfection in order to receive 60-80% confluency the next day. On the day of transfection, Huh-7 cells in each well were transfected with 20 ng HMGA2 3'UTR/psiCHECK2 plasmid, let-7a precursor miRNA (Dharmacon; 10 nM end-concentration), LNA-antimiRs SEQ ID #3232, -3233, -3227, - 3234, -3235, -3226; 0-50 nM end concentrations) together with 0.17 μl Lipofectamine2000 (Invitrogen) according to manufacturer's instructions. After 24 hours, cells were harvested for luciferase measurements. Luciferase assay: Growth media was discarded and 30 μl 1x Passive Lysis Buffer (Promega) was added to each well. After 15-30 minutes of incubation on an orbital shaker, renilla and firefly luciferase measurements were performed according to manufacturer's instructions (Promega).
Example 29: Assessment of entire let- 7 family antagonism by 8-, and 15-mer LNA- antimiRs in Huh-7 cells transfected with a luciferase sensor assay.
In order to assess the efficiency of a fully LNA-modified 8-mer oligonucleotide in antagonizing the entire let-7 family of miRNAs, the same luciferase sensor construct as described in the previous example was used. Again, Huh-7 cells (with low to non-existing levels of endogenous let-7) were transfected with the sensor construct, with one of the family-representative let-7a, let-7d, let-7e, or let-7i precursors, and with the antagonist SEQ ID #3227 at increasing concentrations. The 8-mer LNA-antimiR was compared to SEQ ID #3226, a 15-mer against let- 7a as a positive and potent control. After 24 hours, luciferase activity was measured. Results: As seen in Figure 29 the fully LNA-modified 8-mer LNA-antimiRs (SEQ ID #3227) show similar potencies in de-repressing the various let-7 targets in the luciferase sensor assay, as the positive control 15-mer SEQ ID #3226. Nearly full target de-repression for the 8-mer is achieved already at 0.5-1 nM, except in the case with let-7e premiR (Fig. 29C), to which only 7 out of 8 nucleotides of SEQ ID #3227 hybridizes to the target. However, despite the terminal mismatch in this case, SEQ ID #3227 generates full target de-repression at 5 nM. The positive control 15-mer shows potent antagonism of all precursors and gives nearly full de-repression at 0.5 nM. The de-repression of luciferase activity by both the 8- and the 15-mer LNA-antimiRs is clearly dose-dependent, as seen in all four panels (Fig 29A-D).
Conclusion: To conclude, the 8-mer LNA-antimiR (SEQ ID #3227), is a potent antagonist against four representative let-7 family members in vitro, and thus likely against the entire family. Compared to a 15-mer positive control antagonist, SEQ ID #3226, the 8-mer is equally potent for three of four targets, and slightly less potent for the fourth target, let-7e, explained by a terminal mismatch in this case. Materials and Methods:
Ce// line: The hepatocellular carcinoma cell line Huh-7 was a kind gift from R. Bartinschlager (Dept MoI Virology, University of Heidelberg). Huh-7 cells were cultured in DMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax, 1x NEAA and 25 ug/ml Gentamicin.
Transfection: 8,000 cells were seeded per well in a 96-well plate the day before transfection in order to receive 60-80% confluency the next day. On the day of transfection, Huh-7 cells in each well were transfected with 20 ng HMGA2 3'UTR/psiCHECK2 plasmid, with let-7a, -7d, -7e, or -7i precursor miRNA (Dharmacon; 10 nM end-concentration), and with LNA-antimiRs SEQ ID #3227 and SEQ ID #3226; 0-50 nM end concentrations) together with 0.17 μl
Lipofectamine2000 (Invitrogen) according to manufacturer's instructions. After 24 hours, cells were harvested for luciferase measurements.
Luciferase assay: Growth medium was discarded and 30 μl 1x Passive Lysis Buffer (Promega) was added to each well. After 15-30 minutes of incubation on an orbital shaker, renilla and firefly luciferase measurements were performed according to manufacturer's instructions (Promega).
Example 30. Assessment of endogenous let-7 antagonism by SEQ ID #3227, an 8-mer LNA-antimiRs, in HeLa cells transfected with a luciferase sensor assay. In order to determine the efficiency of a fully LNA-modified 8-mer oligonucleotide in targeting and antagonizing endogenous let-7, the same luciferase sensor construct as described in previous two examples, was co-transfected with SEQ ID #3227 into the cervical cancer cell line HeLa (that expresses moderate to high levels of let-7 as determined by Q-PCR; data not shown). Empty psiCHECK-2 vector was included as a negative control. Results: As seen in Figure 30, the fully LNA-modified 8-mer LNA-antimiR SEQ ID #3227 shows potent antagonism of endogenous let-7, and gives full target de-repression at concentrations of 5-10 nM. The de-repression of luciferase activity is dose-dependent, starting around 1 nM and reaching a plateau at approximately 10 nM. Conclusion: To conclude, the 8-mer LNA-antimiR (SEQ ID #3227), is a potent antagonist against also endogenous let-7 in vitro, and thus provides definite evidence that entire miRNA families can be successfully targeted by short and fully LNA-modified antagonists. Materials and Methods:
Cell line: The cervical cancer cell line HeLa was purchased from ATCC (#CCL-2™). HeLa cells were cultured in Eagle's MEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax, 1x NEAA and 25 ug/ml Gentamicin.
Transfection: 8,000 cells were seeded per well in a 96-well plate the day before transfection in order to receive 50-70% confluency the next day. On the day of transfection, HeLa cells in each well were co-transfected with 20 ng HMGA2 3'UTR/psiCHECK2 plasmid or psiCHECK-2 (empty vector), and with LNA-antimiR SEQ ID #3227 (0-50 nM, end concentrations) together with 0.17 μl Lipofectamine2000 (Invitrogen) according to manufacturer's instructions. After 24 hours, cells were harvested for luciferase measurements. Luciferase assay: Growth media was discarded and 30 μl 1x Passive Lysis Buffer (Promega) was added to each well. After 15-30 minutes of incubation on an orbital shaker, renilla and firefly luciferase measurements were performed according to manufacturer's instructions (Promega).
Example 31. Assessment of miR-21 antagonism by an 8-mer LNA-antimiR-21 (#3205) versus an 8-mer (#3219) scrambled control LNA in the human colon carcinoma cell line HCT116.
We have previously shown in this application, that an 8-mer LNA-antimiR that is fully LNA- modified and phosphorothiolated effectively antagonizes miR-21 in the human cervix carcinoma cell line HeLa, the human breast carcinoma cell line MCF-7, the human prostate cancer cell line PC3 and human hepatocellular carcinoma HepG2 cell line. We extended this screening approach to the human colon carcinoma cell line HCT116. To assess the efficiency of the 8-mer LNA-antimiR oligonucleotide against miR-21, luciferase reporter constructs were generated in which a perfect match target site for the mature miR-21 was cloned into the 3'UTR of the Renilla luciferase gene. In order to monitor miR-21 inhibition, HCT116 cells were transfected with the luciferase constructs together with the miR-21 antagonist #3205 (8-mer) and for comparison of specificity with the 8-mer LNA scrambled control (#3219). After 24 hours, luciferase activity was measured. Results:The luciferase reporter experiments showed a dose-dependent de-repression of the luciferase miR-21 reporter activity with the 8-mer LNA-antimiR against miR-21 (#3205) and complete de-repression was obtained at 5 nM (Figure 31 ). When comparing the specificity of the 8-mer perfect match and the 8-mer scrambled control, the scrambled control LNA-antimiR (#3219) did not show any de-repression at all, demonstrating high specificity of the LNA-antimiR compound against miR-21. Conclusion: The 8-mer (#3205) is potent in targeting miR-21 and antagonism of miR-21 by #3205 is specific. Materials and Methods:
Ce// line: The human colon carcinoma HCT116 cell line was purchased from ATCC (CCL-247). HCT116 cells were cultured in RPMI medium, supplemented with 10% fetal bovine serum, and 25 ug/ml Gentamicin. Transfection: 110.000 cells were seeded per well in a 12-well plate and transfection was performed. HCT116 cells were transfected with 0.3 μg miR-21 luciferase sensor plasmid or empty psiCHECK2 vector together with 1.2 μl Lipofectamine2000 (Invitrogen) according to the manufacturer's instructions. Transfected were also varying concentrations of LNA-antimiR and control oligonucleotides. After 24 hours, cells were harvested for luciferase measurements. Luciferase assay: The cells were washed with PBS and 250 μl 1 x Passive Lysis Buffer (Promega) was added to the wells. The plates were placed on a shaker for 30 min., after which the cell lysates were transferred to eppendorf tubes. The cell lysate was centrifugated for 10 min at 2.500 rpm after which 50 μi were transferred to a 96 well plate and luciferase measurements were performed according to the manufacturer's instructions (Promega).
Example 32. Knock-down of miR-21 by the 8-mer #3205 LNA-antimiR reduces colony formation of PC3 cells.
A hallmark of cellular transformation is the ability for tumour cells to grow in an anchorage- independent way in semisolid medium. We therefore performed soft agar assay which is a phenotypic assay that is relevant for cancer, given that it measures the decrease of tumour cells. We transfected #3205 (perfect match LNA-antimiR-21 ) and #3219 (LNA scrambled control) into PC3 cells, and after 24 hours plated cells in soft agar. Colonies were counted after 12 days. We show in Figure 32 that inhibition of miR-21 by #3205 can reduce the amount of colonies growing in soft agar compared to the scrambled control LNA treated or untreated control (transfected, but with no LNA), demonstrating decrease of tumour cells. Conclusion: The 8-mer (#3205) targeting the miR-21 family reduces the number of colonies in soft agar, demonstrating proliferation arrest of PC3 cells. Materials and Methods: Cell line: The human prostate carcinoma PC3 cell line was purchased from ECACC
(#90112714). PC3 cells were cultured in DMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax and 25 ug/ml Gentamicin.
Transfection: 250.000 cells were seeded per well in a 6-well plate the day before transfection in order to receive 50% confluency the next day. On the day of transfection, PC3 cells were transfected with 25 nM of different LNA oligonucleotides with Lipofectamine2000.
Clonoαenic growth in soft agar: 2.5x103 PC3 cells were seeded in 0.35% agar on the top of a base layer containing 0.5% agar. Cells were plated 24 hours after transfection. Plates were incubated in at 37°C, 5% CO2 in a humified incubator for 12 days and stained with 0.005% crystal violet for 1 h, after which cells were counted. The assay was performed in triplicate. Example 33. Silencing of miR-21 by the 8-mer #3205 LNA-antimiR reduces colony formation of HepG2 cells. miR-21 is overexpressed in the human hepatocellular carcinoma cell line HepG2 and we have previously shown that we are able to regulate the luciferase activity of a miR-21 sensor plasmid with #3205 in these cells. HepG2 cells were transfected with #3205 and #3219 (scrambled 8-mer), and after 24 hours plated into soft agar. Colonies were counted after 17 days with a microscope.
Results: We show in Figure 33 that inhibition of miR-21 by #3205 can reduce the amount of colonies growing in soft agar, showing that proliferation arrest has occurred. In addition, our scrambled 8-mer control, #3219, had no significant effect on the number of colonies. Conclusion: The 8-mer (#3205) targeting the miR-21 reduces the number of colonies in soft agar, indicating proliferation arrest of HepG2 cells. Materials and Methods:
Cell line: The human hepatocytic HepG2 cell line was purchased from ECACC (#85011430). HepG2 cells were cultured in EMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax and 25 ug/ml Gentamicin.
Transfection: 650.000 cells were seeded per well in a 6-well plate and reverse transfection was performed. HepG2 cells were transfected with 0.6 μg miR-21 luciferase sensor plasmid or empty psiCHECK2 vector together with 2,55 μl Lipofectamine2000 (Invitrogen) according to the manufacturer's instructions. Transfected were also LNA-antimiR and control oligonucleotides as varying concentrations. After 24 hours, the cells were harvested for luciferase measurements. Clonogenic growth in soft agar: 2.0x103 HepG2 cells were seeded in 0.35% agar on the top of a base layer containing 0.5% agar. Cells were plated 24 hours after transfection. Plates were incubated in at 37°C, 5% CO2 in a humified incubator for 17 days and stained with 0.005% crystal violet for 1 h, after which cells were counted. The assay was performed in triplicate.
Example 34. Silencing of miR-21 by the 8-mer #3205 LNA-antimiR inhibits cell migration in PC3 cells.
Cell migration can be monitored by performing a wound healing assay (=scratch assay) where a "scratch" is made in a cell monolayer, and images are captured at the beginning and at regular intervals during cell migration. By comparing the images, quantification of the migration rate of the cells can be determined. This was done in the human prostate cancer cell line PC3. Cells were seeded, and on day 3 the cells were transfected, and the next day, when 100% confluency was reached, a scratch (=wound) was made. When the scratch was made, pictures were taken in order to document the initial wound. Afterwards the area of the wound closure is measured at different time points with the free software program Image J. As shown in Figure 34A, PC3 cells had been treated with 25 nM #3205 (perfect match, miR-21), the control #3219 or left untransfected. Pictures were taken after 24 hours, and the area was calculated for the wound closure at respective time-point. The wound closure for the untransfected cells and for the control, #3219, was faster as compared to our LNA-antimiR against miR-21 , #3205, indicating that #3205 inhibits miR-21 and prevents the cells from migrating (Figure 34B). Conclusion: The 8-mer (#3205) targeting miR-21 inhibits the cell migration of PC3 cells compared to untransfected and control transfected cells. Materials and Methods:
Cell line: The human prostate carcinoma PC3 cell line was purchased from ECACC (#90112714). PC3 cells were cultured in DMEM medium, supplemented with 10% fetal bovine serum, 2 mM Glutamax and 25 ug/ml Gentamicin. Scratch assay: 150.000 cells were seeded per well in a 6-well plate three days before transfection in order to receive 100% confluency the next day. At 24 hours after transfection, a scratch was made in the cell monolayer with a 200 μl tip. Pictures were taken at 0 h and after 24 hours by using a digital camera coupled to a microscope. The software program Image J was used to determine wound closure.
Example 35. Length assessment of fully LNA-substituted LNA-antimiRs antagonizing miR-155.
We have previously shown a length assessment for miR-21 regarding fully LNA-substituted
LNA-antimiRs, and showed that the most potent LNA-antimiRs are 7-, 8- or 9 nt in length. The same experiment was repeated with miR-155. A perfect match target site for miR-155 was cloned into the 3'UTR of the luciferase gene in the reporter plasmid psiCHECK2 and transfected into the mouse RAW macrophage cell line together with fully LNA-substituted LNA-antimiRs of different lengths. Because the endogenous levels of miR-155 are low in the RAW cell line, the cells were treated with 100 ng/ml LPS for 24 hours in order to induce miR-155 accumulation. After 24 hours, luciferase analysis was performed.
Results: As shown in Figure 35, the most potent LNA-antimiRs are #3207(8 nt) and #3241 (9 nt), reaching almost a 80% de-repression at only 0.25 nM LNA concentration. The 6-mer (#3244) shows no significant de-repression. Increasing the length to 12-mer to 14-mer (#3242 and #3243) decreased the potency as shown by less efficient de-repression of the miR-155 reporter.
Conclusion:The most potent fully LNA-substituted LNA-antimiRs targeting miR-155 were an 8- and 9-mer (#3207and #3241).
Materials and Methods:
Cell line: The mouse macrophage RAW 264.7 cell line was purchased from ATCC (TIB-71 ). RAW cells were cultured in DMEM medium, supplemented with 10% fetal bovine serum, 4 mM Glutamax and 25 ug/ml Gentamicin. Transfection: 500.000 cells were seeded per well in a 6-well plate the day before transfection in order to receive 50% confluency the next day. On the day of transfection, RAW 264.7 cells were transfected with 0.3 ug miR-155 perfect match/psiCHECK2 or empty psiCHECK2 vector together with 10 μl Lipofectamine2000 (Invitrogen) according to the manufacturer's instructions. Transfected was also varying concentrations of LNA-antimiRs. In order to induce miR-155 accumulation, LPS (100 ng/ml) was added to the RAW cells after the 4 hour incubation with the transfection complexes. After another 24 hours, cells were harvested for luciferase measurements.
Luciferase assay: The cells were washed with PBS and harvested with cell scraper, after which cells were spinned for 5 min at 2.500 rpm. The supernatant was discarded and 50 μl 1 x
Passive Lysis Buffer (Promega) was added to the cell pellet, after which cells were put on ice for 30 min. The lysed cells were spinned at 10.000 rpm for 30 min after which 20 μl were transferred to a 96-well plate and luciferase measurements were performed according to the manufacturer's instructions (Promega).
Example 36. Plasma protein binding for the fully LNA-substituted 8-mer #3205 targeting miR-21 (LNA-antimiR-21).
The plasma proteins are not saturated with #3205 at the plasma concentrations in the experiment shown in Figure 36A. In a wide range of #3205 concentrations in the plasma the protein binding is around 95% of the #3205 LNA-antimiR-21 in Figure 36B. At #3205 concentrations 50.1 μM (174 μg/mL) the binding capacity of plasma proteins for FAM-labeled #3205 has not been saturated.
Materials and Methods: Mouse plasma (100 μL) was spiked with FAM-labeled #3205 to 0.167, 1.67, 5.01 , 10.02, 16.7, 25.05 and 50.1 μM concentrations. The solutions were incubated at 37°C for 30 minutes. The solutions were transferred to a Microcon Ultracel YM-30 filter (regenerated cellulose 30.000 MWCO). The filters were spun for 20 minutes at 200Og and at room temperature in a microcentrifuge. The filtrate was diluted 5, 10 and 20 times and 100μL samples were transferred to a microtiter plate (Polystyrene Black NUNC-237108). The fluorescence was detected using a FLUOstar Optima elisa reader with excitation 458 nm and emission 520 nm. The amount of unbound FAM-labeled #3205 was calculated from a standard curve derived from filtrated plasma spiked with FAM-labeled #3205 at 12 different (0.45 - 1000 nM) concentrations. The numbers were corrected with the recovery number established from filtration experiments with #3205 concentrations 0.167, 1.67, 5.01 , 10.02, 16.7, 25.05 and 50.1 μM in filtrated plasma. The recovery of FAM-labeled #3205 was 86%. Example 37. Quantitative whole body autoradiography study in female pigmented mice after single intravenous administration of 35S-labelled #3205 LNA-antimiR-21.
In order to determine the biodistribution of a short fully LNA-modified LNA-antimiR (#3205, 8- mer) a whole body tissue distribution of radioactively labeled compound was done in mice. 35S- labelled #3205 was dosed to mice with a single intravenous administration and mice were sacrificed at different time-points, ranging from 5 min to 21 days.
Table 6(i). Individual tissue concentrations (μg #3205/g tissue) after a single intravenous administration of 35S- labelled #3205 in female pigmented mice. The figures are mean values of three measurements for each tissue and ratio. The coefficient of variation (CV) is generally about 10%.
Figure imgf000086_0001
Table 6(ii) Tissue to liver ratios after single intravenous administration of 35S- labelled #3205 in female pigmented mice.
Figure imgf000086_0002
Figure imgf000087_0001
Conclusions: #3205 shows blood clearance of radioactivity with elimination half-lives of 8-10 hours. High levels of radioactivity were registered in the kidney cortex, lymph, liver, bone marrow, spleen, ovary and uterus. The highest level of radioactivity was registered in the kidney cortex showing five times higher levels than that of the liver for #3205. A strong retention of radioactivity was noticed in the kidney cortex, lymph, liver, bone marrow and spleen for #3205 LNA-antimiR-21. Materials and Methods:
Dose administration: All mice were weighed before administration. Nine female mice were given 10 mg/kg of 35S-#3205 intravenously in a tail vein. The volume given to each animal was 10 mL/kg of the test formulation. The specific activity 75.7 μCi/mg. Individual mice were killed 5 min, 15 min, 1 hour, 4 hours, 24 hours, 2 days, 4 days, 7 days and 21 days after administration of #3205.Whole body autoradiography: The mice were anaesthetized by sevoflurane, and then immediately immersed in heptane, cooled with dry ice to -800C, ABR-SOP-0130. The frozen carcasses were embedded in a gel of aqueous carboxymethyl cellulose (CMC), frozen in ethanol, cooled with dry ice (-800C) and sectioned sag ittaly for whole body autoradiography, according to the standard method, ABR-SOP-0131. From each animal 20 μm sections were cut at different levels with a cryomicrotome (Leica CM 3600) at a temperature of about -20°C. The obtained sections were caught on tape (Minnesota Mining and Manufacturing Co., No. 810) and numbered consecutively with radioactive ink. After being freeze-dried at -200C for about 24 hours, selected sections were covered with a thin layer of mylar foil, and put on imaging plates (Fuji, Japan). Exposure took place in light tight cassettes in a lead shielding box at -200C, to protect the image plates from environmental radiation. After exposure the imaging plates were scanned at a pixel size of 50 μm and analyzed by radioluminography using a bioimaging analysis system (Bas 2500, Fuji, Japan), and described in ABR-SOP-0214. A water-soluble standard test solution of 35S radioactivity was mixed with whole blood and used for production of a calibration scale, ABR-SOP-0251. However, the different blood standards were dissolved in 500 uL Soluene-35. 4.5 ml_ Ultima Gold was then added to the dissolved samples. As 35S and 14C have very similar energy spectra, a standard 14C-programme (Packard 2200CA) was used when the radioactivity for the different blood samples was settled.
Pharmacokinetic calculations: The 35S radioactivity measured in whole blood and tissues was expressed as nCi/g tissue and recalculated to nmol equiv/g tissue for the pharmacokinetic evaluation. The pharmacokinetic parameters Cmax, t1/2 and AUC were determined for the whole blood and tissues by non-compartmental analysis using WinNonlin Professional (Pharsight
Corporation, Mountain View, CA, USA). After intravenous administration, the concentration was extrapolated back to zero and expressed as (C0). The elimination rate constant λ was estimated by linear regression analysis of the terminal slope of the logarithmic plasma concentration-time curve. The elimination half-life, ti/2, was calculated using the equation, ti/2 = In2/λ. The last three time-points above LOQ were used in the elimination half-life calculations, if not stated otherwise.
Example 38. Assessment of let-7 inhibition in vivo by an 8-mer LNA-antimiR, as determined through Ras protein quantification in mouse lung and kidney In order to investigate the possibility to antagonize the abundantly expressed let-7 family in vivo, mice were intravenously (i.v.) injected with an 8-mer LNA-antimiR antagonist or with saline. To measure treatment effect, proteins were isolated from lungs and kidneys. Because the Ras family of proteins (N-Ras, K-Ras, and H-Ras), in particular N-Ras and K-Ras, has previously been shown to be regulated (repressed) by the let-7 family by Johnson et al. (Cell, 2005), the aim was to analyze whether these let-7 targets could be de-repressed in vivo.
Results: As seen in Figure 37, the 8-mer LNA-antimiR potently de-repressed Ras protein levels in the kidneys of treated mice, normalized against saline controls. The up-regulation in this organ was more than 3-fold, showing a clear in vivo effect. In the lungs, however, only a minimal (1.2-fold) Ras de-repression was observed (Fig 1 B), suggesting that insufficient amounts of LNA-antimiR has entered this organ in order to inhibit its massive amounts of let-7, as previously described by Johnson et al. (Cancer Research, 2007). Conclusion: The 8-mer LNA-antimiR shows a clear effect in regulating target let-7 miRNA in vivo, as evaluated based on Ras protein levels in treated vs. control mice. Whereas the effect seems to be smaller in lungs, Ras levels in the kidney show a substantial up-regulation upon antimiRs-treatment. Materials and Methods: Animals and dosing: C57BL/6 female mice were treated with 10 mg/kg LNA-antimiR or saline for three consecutive days (0, 1, and 2) and sacrificed on day 4. Tissue samples from lungs and kidneys were snapfrozen and stored at -800C until further processing. Western blot analysis: Lung and kidney proteins from saline and LNA-antimiR-treated mice were separated on NuPAGE Bis Tris 4-12% (Invitrogen), using 100 μg per sample. The proteins were transferred to a nitrocellulose membrane using iBlot (Invitrogen) according to the manufacturer's instructions. Blocking, antibody dilution and detection was performed according to the manufacturer's specifications. For Ras detection, a primary rabbit-anti Ras antibody (SC- 3339, Santa Cruz Biotechnology) and a secondary HRP-conjugated swine-anti-rabbit antibody (P0399, Dako) was used, and for tubulin detection, a primary tubulin alpha (MS-581-P1 ,
Neomarkers) and a secondary HRP-conjugated goat-anti-mouse antibody (P0447, Dako) was used.
Example 40. In vivo efficacy assessment of the 8-mer LNA-antimiR (#3205) in targeting miR-21, as determined by Pdcd4 protein up-regulation in mouse kidney. We have shown that an 8-mer LNA-antimiR that is fully LNA-modified antagonizes miR-21 and has the ability to regulate the protein levels of the miR-21 target Pdcd4 in vitro. We therefore injected the LNA-antimiR into mice to determine the effects of the LNA-antimiR in vivo. The mice received 25 mg/kg of #3205 by i.v. injection every other day for 14 days (a total of 5 doses). The mice were sacrificed on day 14, the kidney was removed, and protein was isolated. In order to determine target regulation, Western blot analysis was performed.
Results: As shown in Figure 37, treating mice with #3205 showed significantly increased Pdcd4 protein levels as compared to the saline control. While the normalized Pdcd4 versus Gapdh ratio was consistent in both saline samples, the protein up-regulation in the two LNA- antimiR-treated (#32059 mice were measured to 3.3- and 6.3-fold, respectively, demonstrating an in vivo pharmacological effect of the #3205 8-mer LNA-antimiR.
Conclusion: The fully LNA-modified 8-mer LNA-antimiR #3205 antagonizes miR-21 in vivo, as demonstrated through its ability to de-repress (up-regulate) mouse kidney levels of Pdcd4, a validated miR-21 target. Materials and Methods: Animals and dosing: C57BL/6 female mice with average of 20 g body weight at first dosing were used in all experiments and received regular chow diet (Altromin no 1324, Brogaarden, Gentofte, Denmark). Substances were formulated in physiological saline (0.9% NaCI). The animals were dozed with LNA-antimiR or saline (0.9% NaCI), receiving an injection of 25 mg/kg every other day for 14 days, a total of 5 doses. Animals were sacrificed on day 14. Western blot analysis: 80 μg kidney tissue from saline or LNA-treated mice was separated on NuPAGE Bis Tris 4-12% (Invitrogen). The proteins were transferred to a nitrocellulose membrane using iBlot (Invitrogen) according to the manufacturer's instructions. The membrane was incubated with Pdcd4 antibody (Bethyl Laboratories), followed by HRP-conjugated swine- anti-rabbit antibody (Dako). As equal loading control, GAPDH (Abeam) was used, followed by HRP-conjugated swine-anti-mouse antibody. The membranes were visualized by chemiluminiscence (ECL, Amersham).
Figure imgf000091_0001
Figure imgf000092_0001
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0001
Figure imgf000098_0001
Figure imgf000099_0001
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000105_0001
Figure imgf000106_0001
Figure imgf000107_0001
Figure imgf000108_0001

Claims

1. An oligomer of a contiguous sequence of 7, 8, 9 or 10 nucleotide units in length, for use in reducing the effective amount of a microRNA target in a cell or an organism, wherein at least 70% of the nucleotide units of the oligomer are selected from the group consisting of LNA units and 2' substituted nucleotide analogues, and wherein at least 50% of the nucleotide units of the oligomer are LNA units, and wherein at least one of the internucleoside linkages present between the nucleotide units of the contiguous nucleotide sequence is a phosphorothioate internucleoside linkage.
2. The oligomer according to claim 1 , wherein all the internucleoside linkages present between the nucleotide units of the contiguous nucleotide sequence are phosphorothioate internucleoside linkages.
3. The oligomer according to claim 2, wherein the length of the oligomer is 7, 8 or 9 contiguous nucleotides, wherein the contiguous nucleotide units are independently selected from the group consisting of LNA units and 2' substituted nucleotide analogues.
4. The oligomer according to any one of claims 1 - 3, wherein at least 70% of the nucleotide units of the oligomer are LNA units.
5. The oligomer according to any one of claims 1 - 3, wherein all the nucleotide units of the oligomer are LNA units.
6. The oligomer according to any one of claims 1 - 5, wherein the contiguous nucleotide sequence is complementary to a corresponding region of a microRNA (miRNA) sequence selected from the group consisting of miR-21 , miR-155, miR-221 , miR-222, and miR-122.
7. The oligomer according to any one of claims 1 - 5, wherein said miRNA is selected from the group consisting of miR-1 , miR-10b, miR-29, miR-125b,miR-126, miR-133, miR-141 , miR- 143, miR-200b, miR-206, miR-208, miR-302, miR-372, miR-373, miR-375, and miR-520c/e.
8. The oligomer according to any one of claims 1 - 5, wherein the contiguous nucleotide sequence is complementary to a corresponding region of a microRNA (miRNA) sequence present in the miR 17 - 92 cluster, such as a microRNA selected from the group consisting of miR-17-5p, miR-20a/b, miR-93, miR-106a/b, miR-18a/b, miR-19a/b, miR-25, miR-92a, , miR-363.
9. The oligomer according to any one of claims 1 - 5, wherein the contiguous nucleotide sequence is complementary to a corresponding region of a mammalian, human or viral microRNA (miRNA) sequence selected from the group of miRNAs listed in table 1.
10. The oligomer according to any one of claims 1 - 5, wherein the contiguous nucleotide sequence is complementary to a corresponding region of a mammalian, human or viral microRNA (miRNA) sequence selected from the group of miRNAs from SEQ ID No 1 - 558 as disclosed in WO2008/046911.
11. The oligomer according to any one of claims 1 - 10, wherein the contiguous nucleotide sequence of the oligomer consists of or comprises a sequence which is complementary to the seed sequence of said microRNA.
12. The oligomer according to any one of claims 1 - 11 , wherein the contiguous nucleotide sequence of the oligomer consists of or comprises a sequence selected from any one of the 7mer, 8mer or 9mer seedmer sequences listed in table 1.
13. The oligomer according to claim 11 or 12, wherein the 3' nucleotide of the seedmer forms the 3' most nucleotide of the contiguous nucleotide sequence, wherein the contiguous nucleotide sequence may, optionally, comprise one or two further 5' nucleotides.
14. The oligomer according to any one of claims 1 - 13, wherein said contiguous nucleotide sequence of the oligomer does not comprise a nucleotide which corresponds to the first nucleotide present in the micro-RNA sequence counted from the 5' end.
15. The oligomer according to any one of claims 1 - 14, wherein the nucleotide analogue units are selected from the group consisting of 2'-O_alkyl-RNA unit, 2'-OMe-RNA unit, 2'- amino-DNA unit, 2'-fluoro-DNA unit, LNA unit, and a 2'-MOE RNA unit.
16. The oligomer according to any one of claims 1 - 15, wherein the nucleotide analogue units are Locked Nucleic Acid (LNA) nucleotide analogue units.
17. The oligomer according to any one of claims 1 - 16, wherein the contiguous nucleotide sequence of the oligomer is complementary to the corresponding sequence of at least two miRNA sequences such as 2, 3, 4, 5, 6, 7, 8, 9, or 10 miRNA sequences, optionally with the use of a single universal nucleotide within the oligomer contiguous nucleotide sequence.
18. The oligomer according to claim 17, wherein the contiguous nucleotide sequence of the oligomer consists or comprises of a sequence which is complementary to the sequence of at least two miRNA seed region sequences such as 2, 3, 4, 5, 6, 7, 8, 9, or 10 miRNA seed region sequences.
19. The oligomer according to any one of claims 17 or 18, wherein the contiguous nucleotide sequence is complementary to the corresponding region of both miR-221 and miR-222.
20. The oligomer according to claim 19, wherein the contiguous nucleotide sequence consists or comprises of a sequence that is complementary to 5'GCUACAU3\
21. The oligomer according to any one of claims 1 - 20, wherein the contiguous nucleotide sequence is complementary to a corresponding region of hsa-miR-122.
22. The oligomer according to claim 21 , for use in the treatment of a medical disorder or disease selected from the group consisting of: hepatitis C virus infection and hypercholesterolemia and related disorders.
23. The oligomer according to any one of claims 1 - 22 as a medicament.
24. The oligomer according to any one of claims 1 - 23, for use in medicine, such as for the treatment of a disease or medical disorder associated with the presence or over-expression of the microRNA.
25. A pharmaceutical composition comprising the oligomer according to any one of claims 1 - 23, and a pharmaceutically acceptable diluent, carrier, salt of adjuvant.
26. The pharmaceutical composition according to claim 25, wherein the oligomer is as according to claim 21 or 22 and the composition further comprises a second independent active ingredient that is an inhibitor of the VLDL assembly pathway, such as an ApoB inhibitor, or an MTP inhibitor.
27. A kit comprising a pharmaceutical composition comprising the oligomer according to claim 21 or 22, and a second independent active ingredient that is an inhibitor of the VLDL assembly pathway, such as an ApoB inhibitor, or an MTP inhibitor.
28. A method for the treatment of a disease or medical disorder associated with the presence or over-expression of a microRNA, comprising the step of administering a the pharmaceutical composition according to any one of claims 25 - 26 to a patient who is suffering from, or is likely to suffer from said disease or medical disorder.
29. A conjugate comprising the oligomer according to any one of claims 1 - 24 and at least one non-nucleotide compounds.
30. The use of an oligomer or a conjugate as defined in any one of the proceeding claims, for the manufacture of a medicament for the treatment of a disease or medical disorder associated with the presence or over-expression of the microRNA.
31. The use of an oligomer or a conjugate as defined in any one of the proceeding claims, for inhibiting the mircoRNA in a cell which comprises said microRNA.
32. A method for reducing the amount, or effective amount, of a miRNA in a cell, comprising administering an oligomer, a conjugate or a pharmaceutical composition, according to any one of the proceeding claims to the cell which is expressing said miRNA so as to reduce the amount, or effective amount of the miRNA in the cell.
33. A method for de-repression of one or more mRNAs whose expression is repressed by a miRNA in a cell comprising administering an oligomer, a conjugate or a pharmaceutical composition, according to any one of the preceeding claims to the cell which expresses both said mRNA and said miRNA, in order to de-repress the expression of the mRNA.
PCT/DK2008/000344 2007-10-04 2008-10-03 Micromirs WO2009043353A2 (en)

Priority Applications (23)

Application Number Priority Date Filing Date Title
EP12191739.7A EP2623599B1 (en) 2007-10-04 2008-10-03 Micromirs
ES08801378T ES2406686T3 (en) 2007-10-04 2008-10-03 Micromirs
CA2701547A CA2701547C (en) 2007-10-04 2008-10-03 Oligonucleotides which target and inhibit micrornas
EP08801378A EP2205737B1 (en) 2007-10-04 2008-10-03 Micromirs
EP12191738.9A EP2623598B1 (en) 2007-10-04 2008-10-03 Micromirs
MX2010003299A MX2010003299A (en) 2007-10-04 2008-10-03 Micromirs.
UAA201005202A UA102529C2 (en) 2007-10-04 2008-10-03 Microribonucleic acids
CN200880110388.3A CN101821391B (en) 2007-10-04 2008-10-03 Micromirs
US12/681,587 US8906871B2 (en) 2007-10-04 2008-10-03 MicromiRs
DK08801378.4T DK2205737T3 (en) 2007-10-04 2008-10-03 Micromirers
NZ583677A NZ583677A (en) 2007-10-04 2008-10-03 MicroRNAs comprising Locked Nucleic Acid (LNA) units
JP2010527323A JP6035010B2 (en) 2007-10-04 2008-10-03 Micro MIR
EA201070421A EA019939B1 (en) 2007-10-04 2008-10-03 Modified oligomer for reducing amount of microrna in a cell
EP18185844.0A EP3492594A1 (en) 2007-10-04 2008-10-03 Micromirs
AU2008306327A AU2008306327B2 (en) 2007-10-04 2008-10-03 Micromirs
BRPI0817485-7A BRPI0817485A2 (en) 2007-10-04 2008-10-03 OLIGOMERS, AS WELL AS AN IN VITRO METHOD TO REDUCE THE EFFECTIVE AMOUNT OF A MICRNA TARGET IN A CELL
KR1020107009969A KR101889518B1 (en) 2007-10-04 2008-10-03 MICROMIRs
PL12191738T PL2623598T3 (en) 2007-10-04 2008-10-03 Micromirs
IL204254A IL204254A (en) 2007-10-11 2010-03-02 Oligomers comprising locked nucleic acid units, pharmaceutical compositions comprising them and use of same
ZA2010/02040A ZA201002040B (en) 2007-10-04 2010-03-23 Micromirs
US14/527,115 US9790493B2 (en) 2007-10-04 2014-10-29 MICROMIRs
IL244498A IL244498A0 (en) 2007-10-04 2016-03-08 Micromirs
US15/705,739 US10450564B2 (en) 2007-10-04 2017-09-15 Micromirs

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US97749707P 2007-10-04 2007-10-04
US60/977,497 2007-10-04
US97921707P 2007-10-11 2007-10-11
US60/979,217 2007-10-11
US2806208P 2008-02-12 2008-02-12
US61/028,062 2008-02-12
EP08104780.5 2008-07-17
EP08104780 2008-07-17

Related Child Applications (4)

Application Number Title Priority Date Filing Date
EP12191739.7A Previously-Filed-Application EP2623599B1 (en) 2007-10-04 2008-10-03 Micromirs
EP12191738.9A Previously-Filed-Application EP2623598B1 (en) 2007-10-04 2008-10-03 Micromirs
US12/681,587 A-371-Of-International US8906871B2 (en) 2007-10-04 2008-10-03 MicromiRs
US14/527,115 Continuation US9790493B2 (en) 2007-10-04 2014-10-29 MICROMIRs

Publications (2)

Publication Number Publication Date
WO2009043353A2 true WO2009043353A2 (en) 2009-04-09
WO2009043353A3 WO2009043353A3 (en) 2009-08-20

Family

ID=40526747

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/DK2008/000345 WO2009043354A2 (en) 2007-10-04 2008-10-03 Combination treatment for the treatment of hepatitis c virus infection
PCT/DK2008/000344 WO2009043353A2 (en) 2007-10-04 2008-10-03 Micromirs

Family Applications Before (1)

Application Number Title Priority Date Filing Date
PCT/DK2008/000345 WO2009043354A2 (en) 2007-10-04 2008-10-03 Combination treatment for the treatment of hepatitis c virus infection

Country Status (17)

Country Link
US (5) US8440637B2 (en)
EP (5) EP2623598B1 (en)
JP (2) JP6035010B2 (en)
KR (1) KR101889518B1 (en)
CN (1) CN101821391B (en)
AU (1) AU2008306327B2 (en)
CA (1) CA2701547C (en)
DK (2) DK2623599T3 (en)
EA (1) EA019939B1 (en)
ES (3) ES2406686T3 (en)
IL (2) IL204254A (en)
MX (1) MX2010003299A (en)
MY (1) MY156951A (en)
NZ (1) NZ583677A (en)
PL (1) PL2623598T3 (en)
WO (2) WO2009043354A2 (en)
ZA (1) ZA201002040B (en)

Cited By (128)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010133970A1 (en) 2009-05-20 2010-11-25 Eth Zurich Targeting micrornas for metabolic disorders
WO2010135714A3 (en) * 2009-05-22 2011-01-13 The Methodist Hospital Research Institute Methods for modulating adipocyte expression using microrna compositions
WO2011026909A1 (en) * 2009-09-02 2011-03-10 L'oreal Epidermal differentiation microrna signature and uses thereof
CN102002494A (en) * 2010-07-08 2011-04-06 浙江理工大学 microRNA biomarker and application thereof
WO2011048125A1 (en) 2009-10-20 2011-04-28 Santaris Pharma A/S Oral delivery of therapeutically effective lna oligonucleotides
EP2315832A1 (en) 2008-08-01 2011-05-04 Santaris Pharma A/S Micro-rna mediated modulation of colony stimulating factors
WO2011060100A1 (en) * 2009-11-11 2011-05-19 Sanford-Burnham Medical Research Institute Method for generation and regulation of ips cells and compositions thereof
WO2011131354A1 (en) * 2010-04-20 2011-10-27 Febit Holding Gmbh Complex mirna sets as novel biomarkers for an acute coronary syndrome
WO2012010711A1 (en) 2010-07-23 2012-01-26 Santaris Pharma A/S Process
EP2440566A1 (en) 2009-06-08 2012-04-18 Miragen Therapeutics CHEMICAL MODIFICATION MOTIFS FOR miRNA INHIBITORS AND MIMETICS
US8163708B2 (en) 2006-04-03 2012-04-24 Santaris Pharma A/S Pharmaceutical composition comprising anti-mirna antisense oligonucleotide
US8202848B2 (en) 2008-03-17 2012-06-19 Board Of Regents, The University Of Texas System Identification of micro-RNAS involved in neuromuscular synapse maintenance and regeneration
CN102643811A (en) * 2011-02-18 2012-08-22 中国科学院上海药物研究所 Human miR-1229 antisense oligonucleotide and application thereof
CN102643806A (en) * 2011-02-18 2012-08-22 中国科学院上海药物研究所 Antisense oligodeoxyncleotide of human miR-1913 and application thereof
CN102643813A (en) * 2011-02-18 2012-08-22 中国科学院上海药物研究所 Human miR-504 antisense oligonucleotide and application thereof
CN102643814A (en) * 2011-02-18 2012-08-22 中国科学院上海药物研究所 Antisense oligonucleotide for human miR-431 and application thereof
CN102643809A (en) * 2011-02-18 2012-08-22 中国科学院上海药物研究所 Antisense oligodeoxyncleotide of human miR-1274b and application thereof
CN102643810A (en) * 2011-02-18 2012-08-22 中国科学院上海药物研究所 Human miR-299-5p antisense oligonucleotide and application thereof
CN102666878A (en) * 2009-11-13 2012-09-12 江苏命码生物科技有限公司 Markers, methods, biochips and kits for milk qualit y detection
US8288356B2 (en) 2007-10-04 2012-10-16 Santaris Pharma A/S MicroRNAs
WO2012145374A1 (en) 2011-04-19 2012-10-26 Regulus Therapeutics Inc. TARGETING miR-378 FAMILY MEMBERS FOR THE TREATMENT OF METABOLIC DISORDERS
WO2012148952A1 (en) * 2011-04-25 2012-11-01 Regulus Therapeutics Inc Microrna compounds and methods for modulating mir-21 activity
WO2012175357A1 (en) * 2011-06-24 2012-12-27 Syddansk Universitet Modulation of microrna-138 for the treatment of bone loss
WO2012175733A1 (en) 2011-06-23 2012-12-27 Santaris Pharma A/S Hcv combination therapy
CN102851282A (en) * 2011-06-30 2013-01-02 上海市肿瘤研究所 MicroRNA markers for discriminating constitutional lung cancer tissue and paracancerous tissue
WO2013000855A1 (en) 2011-06-30 2013-01-03 Santaris Pharma A/S Hcv combination therapy
WO2013000856A1 (en) 2011-06-30 2013-01-03 Santaris Pharma A/S Hcv combination therapy
US8361980B2 (en) 2008-03-07 2013-01-29 Santaris Pharma A/S Pharmaceutical compositions for treatment of microRNA related diseases
US20130102546A1 (en) * 2010-03-12 2013-04-25 The Brigham And Women's Hospital, Inc. Methods of treating vascular inflammatory disorders
US8440636B2 (en) 2007-07-31 2013-05-14 The Board Of Regents, The University Of Texas System Micro-RNA family that modulates fibrosis and uses thereof
WO2013068348A1 (en) 2011-11-07 2013-05-16 Santaris Pharma A/S Lna oligomers for improvement in hepatic function
WO2013068347A1 (en) 2011-11-07 2013-05-16 Santaris Pharma A/S Prognostic method for checking efficacy of micro rna-122 inhibitors in hcv+ patients
US8481507B2 (en) 2007-07-31 2013-07-09 The Board Of Regents, The University Of Texas System Micro-RNAs that control myosin expression and myofiber identity
US8513209B2 (en) 2007-11-09 2013-08-20 The Board Of Regents, The University Of Texas System Micro-RNAS of the MIR-15 family modulate cardiomyocyte survival and cardiac repair
CN103290010A (en) * 2013-05-16 2013-09-11 南京市妇幼保健院 Parental serum/plasma miRNA marker mir-375 relevant to fetus congenital heart disease and application thereof
WO2013134403A1 (en) * 2012-03-06 2013-09-12 The Washington University Method of treating neurodegenerative diseases with microrna regulators
WO2013163258A1 (en) * 2012-04-25 2013-10-31 Regulus Therapeutics Inc. Microrna compounds and methods for modulating mir-21 activity
US8629119B2 (en) 2009-02-04 2014-01-14 The Board Of Regents, The University Of Texas System Dual targeting of MIR-208 and MIR-499 in the treatment of cardiac disorders
US8642751B2 (en) 2010-12-15 2014-02-04 Miragen Therapeutics MicroRNA inhibitors comprising locked nucleotides
US20140080899A1 (en) * 2011-04-28 2014-03-20 New York University miR-33 Inhibitors and Uses Thereof to Decrease Inflammation
US8680067B2 (en) 2007-10-29 2014-03-25 Regulus Therapeutics, Inc. Targeting microRNAs for the treatment of liver cancer
US8697663B2 (en) 2003-07-31 2014-04-15 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAs
US8729250B2 (en) 2006-04-03 2014-05-20 Joacim Elmén Antisense oligonucleotides for inhibition of microRNA-21
WO2014076195A1 (en) 2012-11-15 2014-05-22 Santaris Pharma A/S Oligonucleotide conjugates
WO2014118272A1 (en) 2013-01-30 2014-08-07 Santaris Pharma A/S Antimir-122 oligonucleotide carbohydrate conjugates
WO2014118267A1 (en) 2013-01-30 2014-08-07 Santaris Pharma A/S Lna oligonucleotide carbohydrate conjugates
US8815826B2 (en) 2010-07-23 2014-08-26 Regulus Therapeutics, Inc. Targeting microRNAs for the treatment of fibrosis
WO2014135653A1 (en) * 2013-03-06 2014-09-12 Royal College Of Surgeons In Ireland Diagnosis and treatment of metabolic disorders
US8859519B2 (en) 2010-08-25 2014-10-14 The General Hospital Corporation Methods targeting miR-33 microRNAs for regulating lipid metabolism
US20140357699A1 (en) * 2009-08-06 2014-12-04 Universiteit Maastricht Means and methods for counteracting, delaying and/or preventing adverse energy metabolism switches in heart disease
WO2014201314A1 (en) * 2013-06-14 2014-12-18 Joslin Diabetes Center, Inc. Microrna and uses in brown fat differentiation
US20150025232A1 (en) * 2011-08-29 2015-01-22 Masayuki Nashimoto Heptamer-type small guide nucleic acids inducing apoptosis of human leukemia cells
WO2014179446A3 (en) * 2013-05-01 2015-02-19 Regulus Therapeutics Inc. Microrna compounds and methods for modulating mir-122
EP2816114A3 (en) * 2011-12-15 2015-02-25 Oncostamen S.r.l. MicroRNAs and uses thereof
US9012423B2 (en) 2012-10-09 2015-04-21 Regulus Therapeutics Inc. Methods for treatment of alport syndrome
US9034837B2 (en) 2009-04-24 2015-05-19 Roche Innovation Center Copenhagen A/S Pharmaceutical compositions for treatment of HCV patients that are poor-responders to interferon
EP2755663A4 (en) * 2011-09-13 2015-10-07 Ottawa Hospital Res Inst Microrna inhibitors
US9163235B2 (en) 2012-06-21 2015-10-20 MiRagen Therapeutics, Inc. Inhibitors of the miR-15 family of micro-RNAs
EP2785355A4 (en) * 2011-11-30 2015-12-02 Cedars Sinai Medical Center Targeting micrornas mir-409-5p, mir-379 and mir-154* to treat prostate cancer bone metastasis and drug resistant lung cancer
US20160032286A1 (en) * 2014-08-04 2016-02-04 MiRagen Therapeutics, Inc. Inhibitors of myh7b and uses thereof
US9267139B2 (en) 2012-10-11 2016-02-23 Georgia Regents Research Institute, Inc. Compositions and methods for treating musculoskeletal diseases and disorders
US9328346B2 (en) 2010-11-12 2016-05-03 The General Hospital Corporation Polycomb-associated non-coding RNAs
US9334498B2 (en) 2012-05-10 2016-05-10 Uab Research Foundation Methods and compositions for modulating MIR-204 activity
WO2016100716A1 (en) 2014-12-18 2016-06-23 Vasant Jadhav Reversirtm compounds
US9388408B2 (en) 2012-06-21 2016-07-12 MiRagen Therapeutics, Inc. Oligonucleotide-based inhibitors comprising locked nucleic acid motif
US9428749B2 (en) 2011-10-06 2016-08-30 The Board Of Regents, The University Of Texas System Control of whole body energy homeostasis by microRNA regulation
US9441222B2 (en) 2011-01-11 2016-09-13 Interna Technologies B.V. MiRNA for treating diseases and conditions associated with neo-angiogenesis
US9487783B2 (en) 2014-08-07 2016-11-08 Regulus Therapeutics Inc. Targeting microRNAs for metabolic disorders
US9506030B2 (en) 2013-05-01 2016-11-29 Regulus Therapeutics Inc. Compounds and methods for enhanced cellular uptake
US9580708B2 (en) 2011-09-14 2017-02-28 Rana Therapeutics, Inc. Multimeric oligonucleotides compounds
WO2017032726A1 (en) 2015-08-24 2017-03-02 Roche Innovation Center Copenhagen A/S Lna-g process
WO2017067970A1 (en) 2015-10-22 2017-04-27 Roche Innovation Center Copenhagen A/S In vitro toxicity screening assay
US9682095B2 (en) 2010-07-06 2017-06-20 Interna Technologies B.V. MiRNA and its diagnostic and therapeutic uses in diseases or conditions associated with melanoma, or in diseases or conditions associated with activated BRAF pathway
US9745578B2 (en) 2011-11-30 2017-08-29 Cedars-Sinai Medical Center Targeting microRNA miR-409-3P to treat prostate cancer
WO2017157672A1 (en) 2016-03-18 2017-09-21 Roche Innovation Center Copenhagen A/S Acyl-protected l-lna-guanosine monomers
US9771585B2 (en) 2015-06-05 2017-09-26 MiRagen Therapeutics, Inc. miR-155 inhibitors for treating cutaneous T cell lymphoma (CTCL)
US9790494B2 (en) 2012-09-14 2017-10-17 Translate Bio Ma, Inc. Multimeric oligonucleotide compounds having non-nucleotide based cleavable linkers
KR101800243B1 (en) 2011-06-22 2017-12-20 (주)아모레퍼시픽 Composition for regulating expression of pigmentation-related genes containing microRNA
WO2017216325A1 (en) 2016-06-17 2017-12-21 F. Hoffmann-La Roche Ag In vitro nephrotoxicity screening assay
WO2017216340A1 (en) 2016-06-17 2017-12-21 F. Hoffmann-La Roche Ag In vitro nephrotoxicity screening assay
US9885042B2 (en) 2015-01-20 2018-02-06 MiRagen Therapeutics, Inc. miR-92 inhibitors and uses thereof
US9920317B2 (en) 2010-11-12 2018-03-20 The General Hospital Corporation Polycomb-associated non-coding RNAs
US9994846B2 (en) 2013-10-25 2018-06-12 Regulus Therapeutics Inc. MicroRNA compounds and methods for modulating miR-21 activity
WO2018106566A1 (en) * 2016-12-05 2018-06-14 Regulus Therapeutics Inc. Modified oligonucleotides for treatment of polycystic kidney disease
US10058623B2 (en) 2012-05-16 2018-08-28 Translate Bio Ma, Inc. Compositions and methods for modulating UTRN expression
US10059941B2 (en) 2012-05-16 2018-08-28 Translate Bio Ma, Inc. Compositions and methods for modulating SMN gene family expression
US10077446B2 (en) 2013-06-25 2018-09-18 The United States Of America, As Represented By The Secretary, Dept. Of Health And Human Services Glucan-encapsulated siRNA for treating type 2 diabetes mellitus
WO2018177825A1 (en) 2017-03-29 2018-10-04 Roche Innovation Center Copenhagen A/S Orthogonal protecting groups for the preparation of stereodefined phosphorothioate oligonucleotides
WO2018177881A1 (en) 2017-03-29 2018-10-04 Roche Innovation Center Copenhagen A/S Unylinker rapid cleavage
EP2850189B1 (en) 2012-05-16 2018-11-07 Translate Bio MA, Inc. Compositions and methods for modulating gene expression
EP3303589A4 (en) * 2015-06-05 2019-01-02 Miragen Therapeutics, Inc. Mir-155 inhibitors for treating amyotrophic lateral sclerosis (als)
WO2019002237A1 (en) 2017-06-28 2019-01-03 Roche Innovation Center Copenhagen A/S Multiple coupling & oxidation method
US10174323B2 (en) 2012-05-16 2019-01-08 The General Hospital Corporation Compositions and methods for modulating ATP2A2 expression
US10174315B2 (en) 2012-05-16 2019-01-08 The General Hospital Corporation Compositions and methods for modulating hemoglobin gene family expression
WO2019073018A1 (en) 2017-10-13 2019-04-18 Roche Innovation Center Copenhagen A/S Methods for identifying improved stereodefined phosphorothioate oligonucleotide variants of antisense oligonucleotides utilising sub-libraries of partially stereodefined oligonucleotides
US20190175660A1 (en) * 2014-11-05 2019-06-13 Emory University Compositions of Ascorbic Acid and Bone Morphogenetic Protein 4 (BMP-4) for Cell Growth and Uses Related Thereto
WO2019122279A1 (en) 2017-12-22 2019-06-27 Roche Innovation Center Copenhagen A/S Oligonucleotides comprising a phosphorodithioate internucleoside linkage
WO2019122277A1 (en) 2017-12-22 2019-06-27 Roche Innovation Center Copenhagen A/S Novel thiophosphoramidites
WO2019122282A1 (en) 2017-12-22 2019-06-27 Roche Innovation Center Copenhagen A/S Gapmer oligonucleotides comprising a phosphorodithioate internucleoside linkage
WO2019219723A1 (en) 2018-05-18 2019-11-21 F. Hoffmann-La Roche Ag Pharmaceutical compositions for treatment of microrna related diseases
US10487324B2 (en) 2015-02-25 2019-11-26 Washington University Methods to detect motor neuron disease comprising micro-RNAs
WO2020025527A1 (en) 2018-07-31 2020-02-06 Roche Innovation Center Copenhagen A/S Oligonucleotides comprising a phosphorotrithioate internucleoside linkage
WO2020025563A1 (en) 2018-07-31 2020-02-06 Roche Innovation Center Copenhagen A/S Oligonucleotides comprising a phosphorotrithioate internucleoside linkage
WO2020038968A1 (en) 2018-08-23 2020-02-27 Roche Innovation Center Copenhagen A/S Microrna-134 biomarker
US10577388B2 (en) 2015-10-02 2020-03-03 Roche Innovation Center Copenhagen A/S Oligonucleotide conjugation process
EP3620519A1 (en) 2018-09-04 2020-03-11 F. Hoffmann-La Roche AG Use of isolated milk extracellular vesicles for delivering oligonucleotides orally
US10655128B2 (en) 2012-05-16 2020-05-19 Translate Bio Ma, Inc. Compositions and methods for modulating MECP2 expression
WO2020152303A1 (en) 2019-01-25 2020-07-30 F. Hoffmann-La Roche Ag Lipid vesicle for oral drug delivery
WO2020169695A1 (en) 2019-02-20 2020-08-27 Roche Innovation Center Copenhagen A/S Phosphonoacetate gapmer oligonucleotides
WO2020169696A1 (en) 2019-02-20 2020-08-27 Roche Innovation Center Copenhagen A/S Novel phosphoramidites
US10758558B2 (en) 2015-02-13 2020-09-01 Translate Bio Ma, Inc. Hybrid oligonucleotides and uses thereof
US10837014B2 (en) 2012-05-16 2020-11-17 Translate Bio Ma, Inc. Compositions and methods for modulating SMN gene family expression
US10858650B2 (en) 2014-10-30 2020-12-08 The General Hospital Corporation Methods for modulating ATRX-dependent gene repression
US10900036B2 (en) 2015-03-17 2021-01-26 The General Hospital Corporation RNA interactome of polycomb repressive complex 1 (PRC1)
US11174220B2 (en) 2019-12-13 2021-11-16 Inspirna, Inc. Metal salts and uses thereof
WO2021226663A1 (en) * 2020-05-11 2021-11-18 The Florey Institute Of Neuroscience And Mental Health Compositions and methods for treating disorders associated with loss-of-function mutations in syngap1
US11214536B2 (en) 2017-11-21 2022-01-04 Inspirna, Inc. Polymorphs and uses thereof
US11253601B2 (en) 2016-07-11 2022-02-22 Translate Bio Ma, Inc. Nucleic acid conjugates and uses thereof
US11261209B2 (en) 2016-05-12 2022-03-01 Roche Innovation Center Copenhagen A/S Enhanced coupling of stereodefined oxazaphospholidine phosphoramidite monomers to nucleoside or oligonucleotide
WO2022245981A3 (en) * 2021-05-18 2022-12-29 Cornell University Use of microrna mimics to inhibit or treat liver disease
WO2023067038A1 (en) 2021-10-22 2023-04-27 Roche Innovation Center Copenhagen A/S Process for oligonucleotide purification
EP3963063A4 (en) * 2019-04-30 2023-09-27 The Trustees of The University of Pennsylvania Compositions useful for treatment of pompe disease
WO2024033446A1 (en) * 2022-08-09 2024-02-15 Geg Tech Transient expression system for rna, for gene editing
WO2024033441A1 (en) * 2022-08-09 2024-02-15 Geg Tech Transient expression system for rna
WO2024033444A1 (en) * 2022-08-09 2024-02-15 Geg Tech Transient expression system for rna, for cosmetic uses
WO2024033448A1 (en) * 2022-08-09 2024-02-15 Geg Tech Transient expression system for rna, for vaccination
US12005120B2 (en) 2018-05-08 2024-06-11 Regulus Therapeutics Inc. Galnac conjugated modified oligonucleotides as miR-122 inhibitor having HCV antiviral activity with reduced hyperbilirubinemia side-effect
US12018260B2 (en) 2017-08-17 2024-06-25 Alnylam Pharmaceuticals, Inc. Tunable Reversir™ compounds

Families Citing this family (116)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2338996A3 (en) 2004-05-04 2013-12-18 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for reducing viral genome amounts in a target cell
EP2468899B1 (en) 2006-01-05 2015-03-11 The Ohio State University Research Foundation MicroRNA-based methods for the diagnosis of stomach cancers
AU2007227423B2 (en) 2006-03-20 2013-11-07 The Ohio State University Research Foundation MicroRNA fingerprints during human megakaryocytopoiesis
WO2008061537A2 (en) * 2006-11-23 2008-05-29 Querdenker Aps Oligonucleotides for modulating target rna activity
US8697672B2 (en) 2007-05-16 2014-04-15 California Institute Of Technology Microrna inhibition for the treatment of inflammation and myeloproliferative disorders
CA2690643A1 (en) * 2007-06-14 2008-12-18 Mirx Therapeutics Aps Oligonucleotides for modulation of target rna activity
CN103866008B (en) 2007-08-03 2016-06-29 俄亥俄州立大学研究基金会 The super conservative region of coding NCRNA
US20110257244A1 (en) * 2008-01-18 2011-10-20 Alnylam Pharmaceuticals, Inc. Chemically modified oligonucleotides and small molecules for use in reducing micro rna activity levels and uses thereof
US20110034538A1 (en) * 2008-02-28 2011-02-10 The Ohio State University Research Foundation MicroRNA-Based Methods and Compositions for the Diagnosis, Prognosis and Treatment of Gastric Cancer
CN104031984A (en) * 2008-02-28 2014-09-10 俄亥俄州立大学研究基金会 Microrna-based methods and compositions for the diagnosis, prognosis and treatment of prostate related disorders
EP2274423A2 (en) 2008-04-04 2011-01-19 Isis Pharmaceuticals, Inc. Oligomeric compounds comprising bicyclic nucleosides and having reduced toxicity
US9068232B2 (en) * 2008-08-06 2015-06-30 Rosetta Genomics Ltd. Gene expression signature for classification of kidney tumors
EP2949752B1 (en) 2008-09-22 2017-12-20 RXi Pharmaceuticals Corporation Reduced size self-delivering rnai compounds
ES2572361T3 (en) * 2008-12-31 2016-05-31 Roche Innovation Ct Copenhagen As Use of ApoB antisense ANB oligomers for the treatment of acute coronary syndromes
EP2401406A4 (en) * 2009-02-25 2012-09-19 Cepheid Methods of detecting lung cancer
WO2011017089A1 (en) * 2009-07-27 2011-02-10 The Regents Of The University Of Colorado, A Body Corporate Mirna inhibition of six1 expression
US8822144B2 (en) 2009-08-19 2014-09-02 Rosetta Genomics Ltd. Compositions and methods for prognosis and treatment of prostate cancer
JPWO2011027893A1 (en) * 2009-09-07 2013-02-04 善基 村上 Method for predicting therapeutic effect of chronic hepatitis C
US20120231970A1 (en) * 2009-09-30 2012-09-13 Japan Health Sciences Foundation Colon cancer marker and method for testing for colon cancer
CN102803511A (en) 2009-11-23 2012-11-28 俄亥俄州立大学 Materials and methods useful for affecting tumor cell growth, migration and invasion
US20120302626A1 (en) * 2009-12-04 2012-11-29 Sandeep Dave Microrna and use thereof in identification of b cell malignancies
WO2011085276A2 (en) * 2010-01-09 2011-07-14 The Translational Genomics Research Institute Methods and kits to predict prognostic and therapeutic outcome in small cell lung cancer
US8841269B2 (en) * 2010-02-23 2014-09-23 Creighton University Polynucleotides for use in treating and diagnosing cancers
EP2539357B1 (en) * 2010-02-26 2017-06-14 Memorial Sloan-Kettering Cancer Center Methods and compositions for the detection and treatment of cancer involving mirnas and mirna inhibitors and targets
CN102140462B (en) * 2010-04-29 2013-06-12 苏州吉玛基因股份有限公司 Human miR-1260 antisense nucleic acid and application thereof
WO2011146937A1 (en) * 2010-05-21 2011-11-24 The Translational Genomics Research Institute Methods and kits useful in diagnosing nsclc
AU2011261213B2 (en) * 2010-06-04 2015-05-21 Board Of Regents, The University Of Texas System Regulation of metabolism by miR-378
US20120015830A1 (en) * 2010-06-21 2012-01-19 Diogenix, Inc. Microrna profiles for evaluating multiple sclerosis
CN102399852A (en) * 2010-09-08 2012-04-04 上海市公共卫生临床中心 Blood plasma miRNA spectrum and detection kit used for predicting chronic hepatitis b curative effect of interferon
WO2012048113A2 (en) 2010-10-07 2012-04-12 The General Hospital Corporation Biomarkers of cancer
AU2011326032B2 (en) 2010-11-12 2016-10-06 The Ohio State University Research Foundation Materials and methods related to microRNA-21, mismatch repair, and colorectal cancer
BR112013011942A2 (en) 2010-11-15 2016-11-01 Univ Michigan formulation, drug dosage form for oral transmucosal administration, transmucosal drug delivery system, method of treatment and prophylaxis of a disease or disorder, method of treatment, formulation, method for treatment or prevention of head and neck squamous cell carcinoma (hnscc), method for chemoprevention of an oral cancer or precancerous condition, method for increasing the concentration of a retinide composition, method of treatment and prophylaxis of a disease or condition, ratification method of a subject presenting a symptomatic medical condition , method of treating an oral cancer or precancerous condition in a patient, method for making an oral drug delivery system, method for increasing the release and permeation of a retinide composition.
US9045749B2 (en) 2011-01-14 2015-06-02 The General Hospital Corporation Methods targeting miR-128 for regulating cholesterol/lipid metabolism
CA2825522A1 (en) 2011-01-26 2012-08-02 Cepheid Methods of detecting lung cancer
CN102643808B (en) * 2011-02-18 2016-01-20 中国科学院上海药物研究所 The antisense oligonucleotide of people miR-1539 and application thereof
JP2014510155A (en) * 2011-04-06 2014-04-24 ザ トラスティーズ オブ プリンストン ユニバーシティ Antiviral combination therapy
WO2013013165A2 (en) * 2011-07-21 2013-01-24 The Trustees Of The University Of Pennsylvania Use of lna compounds for therapeutic inhibition of mir-21 in systemic lupus erythematosus
CA2851280C (en) 2011-10-11 2021-05-18 The Brigham And Women's Hospital, Inc. Micrornas in neurodegenerative disorders
AU2012323924A1 (en) 2011-10-14 2014-05-29 The Ohio State University Methods and materials related to ovarian cancer
JP2015501645A (en) * 2011-12-10 2015-01-19 オハイオ・ステイト・イノベーション・ファウンデーション MiRNAs and related compositions and methods useful for reducing tumor development and chemotherapy resistance in lung cancer
CN104619353A (en) 2011-12-13 2015-05-13 俄亥俄州国家创新基金会 Methods and compositions related to miR-21 and miR-29a, exosome inhibition, and cancer metastasis
US10519506B2 (en) * 2011-12-22 2019-12-31 Baylor Research Institute Identification of metastasis-specific miRNA and hypomethylation signatures in human colorectal cancer
AU2013209477B2 (en) 2012-01-20 2016-12-08 The Ohio State University Breast cancer biomarker signatures for invasiveness and prognosis
WO2014028461A2 (en) 2012-08-13 2014-02-20 The Rockefeller University Treatment and diagnosis of melanoma
CN103627704A (en) * 2012-08-30 2014-03-12 苏州博泰安生物科技有限公司 Prostatic cancer molecular marker miR-19a and applications thereof
US9493838B2 (en) 2012-10-10 2016-11-15 Beth Israel Deaconess Medical Center, Inc. Biomarkers and treatments for heart failure
US10942184B2 (en) 2012-10-23 2021-03-09 Caris Science, Inc. Aptamers and uses thereof
US9944991B2 (en) 2012-11-02 2018-04-17 Dana-Farber Cancer Institute, Inc. Compositions and methods for diagnosis, prognosis and treatment of hematological malignancies
WO2014078749A1 (en) * 2012-11-15 2014-05-22 The Regents Of The University Of California Splice modulating oligonucleotides that inhibit cancer
CN104903468B (en) 2012-11-16 2019-04-23 萨尔大学 New diagnosis MiRNA marker for Parkinson's disease
EP2733219B1 (en) 2012-11-16 2017-09-20 Siemens Aktiengesellschaft Diagnostic miRNA markers for Alzheimer
EP2733220B1 (en) * 2012-11-16 2017-10-18 Siemens Aktiengesellschaft Novel miRNA as a diagnostic marker for Alzheimer's Disease
EP2943570B1 (en) 2013-01-14 2018-01-03 Pierfrancesco Tassone Inhibitors of mirnas 221 and 222 for anti-tumor activity in multiple myeloma
PL2759595T3 (en) * 2013-01-24 2017-09-29 Pierre Fabre Médicament S.A.S. Composition comprising an encapsulated antagomir
KR101465319B1 (en) * 2013-03-08 2014-11-28 서울대학교산학협력단 MicroRNA decay element and its use
US9752143B2 (en) 2013-03-15 2017-09-05 MiRagen Therapeutics, Inc. Locked nucleic acid inhibitor of miR-145 and uses thereof
WO2014145142A2 (en) * 2013-03-15 2014-09-18 Miles Gregory Method of improving survival in cancer
US10053690B2 (en) 2013-06-12 2018-08-21 New York University Anti-miR-27b and anti-miR-148a oligonucleotides as therapeutic tools for treating dyslipidemias and cardiovascular diseases
DK3013959T3 (en) 2013-06-27 2020-02-17 Roche Innovation Ct Copenhagen As ANTISENSE OLIGOMERS AND CONJUGATES TARGETED PCSK9
US11478504B2 (en) 2013-07-08 2022-10-25 International Dehydrated Foods, Inc. Compositions and methods for preventing/treating metabolic syndrome
US10555967B2 (en) 2013-07-08 2020-02-11 International Dehydrated Foods, Inc. Compositions prepared from poultry and methods of their use
CN104342439B (en) * 2013-07-23 2017-06-23 中国科学院遗传与发育生物学研究所 MiR 7 and its application
JP2016534035A (en) 2013-10-04 2016-11-04 ラナ セラピューティクス インコーポレイテッド Compositions and methods for treating amyotrophic lateral sclerosis
AU2015205322B2 (en) * 2014-01-08 2019-08-08 Hadasit Medical Research Services And Development Ltd. MiR-122* as an active micro-RNA, compositions comprising the same and uses thereof
CN104774914B (en) * 2014-01-09 2018-01-09 复旦大学附属儿科医院 The microRNA serum marks analyte detection and its application of Biliary atresia and silt courage type INFANT HEPATITIS SYNDROME
EP3091970B1 (en) 2014-01-10 2020-10-28 Rgenix, Inc. Lxr agonists and uses thereof
CN104974987B (en) * 2014-04-04 2018-06-19 宁夏医科大学 Purposes of the small molecule RNA-1247-5p as novel tumor treatment molecular target
CN103937888B (en) * 2014-04-14 2016-08-17 上海交通大学 Differentiate screening and the application of the blood plasma microRNA mark of gastric cancer
CN103940998B (en) * 2014-05-04 2016-04-20 山东大学 The application of the early diagnosis marker that serum microRNA shifts as hepatocellular carcinoma
JP6841661B2 (en) * 2014-06-13 2021-03-10 ウニヴェルズィテート・フューア・ボーデンクルトゥーア・ウィーン Compositions and Methods for Diagnosis and Treatment of Fractures and Bone Disorders
WO2015200697A1 (en) 2014-06-25 2015-12-30 The General Hospital Corporation Targeting human satellite ii (hsatii)
EP3177722A4 (en) * 2014-07-31 2018-01-17 Agency For Science, Technology And Research Modified antimir-138 oligonucleotides
US20180291374A1 (en) * 2014-11-12 2018-10-11 The General Hospital Corporation INHIBITORS OF MICRORNAs miR-155, miR-103, miR-105 and miR-107 THAT REGULATE PRODUCTION OF ATRIAL NATRIURETIC PEPTIDE (ANP) AS THERAPEUTICS AND USES THEREOF
CA2968541A1 (en) * 2014-11-21 2016-05-26 Caris Science, Inc. Oligonucleotide probes and uses thereof
CN107429251A (en) * 2015-03-09 2017-12-01 肯塔基大学研究基金会 For treating the miRNA of breast cancer
WO2016160882A1 (en) * 2015-04-01 2016-10-06 Baylor Research Institute Methods for diagnosing and treating colorectal cancer using mirna549a
US10961532B2 (en) 2015-04-07 2021-03-30 The General Hospital Corporation Methods for reactivating genes on the inactive X chromosome
US20180161429A1 (en) 2015-06-26 2018-06-14 Beth Israel Deaconess Medical Center Inc. Cancer therapy targeting tetraspanin 33 (tspan33) in myeloid derived suppressor cells
CN108701922B (en) 2015-07-07 2020-02-14 Afci亚洲私人有限公司 Electrical connector
ES2876930T3 (en) 2015-08-20 2021-11-15 Asociacion Centro De Investig Cooperativa En Biociencias Cic Biogune Methods and compositions for treating liver diseases and conditions
WO2017066712A2 (en) 2015-10-16 2017-04-20 The Children's Medical Center Corporation Modulators of telomere disease
AU2016355178B9 (en) 2015-11-19 2019-05-30 Massachusetts Institute Of Technology Lymphocyte antigen CD5-like (CD5L)-interleukin 12B (p40) heterodimers in immunity
WO2017147087A1 (en) 2016-02-25 2017-08-31 The Brigham And Women's Hospital, Inc. Treatment methods for fibrosis targeting smoc2
WO2017192959A2 (en) * 2016-05-05 2017-11-09 The Research Foundation For The State University Of New York Therapeutically modulating apob and apoai
EP3254683A1 (en) 2016-06-06 2017-12-13 Medizinische Hochschule Hannover Medicament for the treatment of acute liver failure
EP3532638A4 (en) 2016-10-31 2020-07-29 University of Massachusetts Targeting microrna-101-3p in cancer therapy
WO2018165532A1 (en) 2017-03-10 2018-09-13 Baylor Research Institute Methods for diagnosing and treating gastric cancer using mirna expression
US20200384115A1 (en) 2017-04-21 2020-12-10 The Broad Institute , Inc. Targeted delivery to beta cells
KR101871920B1 (en) 2017-08-04 2018-07-02 (주)아모레퍼시픽 Composition containing microRNA
US11710917B2 (en) 2017-10-30 2023-07-25 Amphenol Fci Asia Pte. Ltd. Low crosstalk card edge connector
EP3703669A4 (en) 2017-11-01 2021-11-10 Dana-Farber Cancer Institute, Inc. Methods of treating cancers
US10601181B2 (en) 2017-12-01 2020-03-24 Amphenol East Asia Ltd. Compact electrical connector
CN113116918B (en) * 2017-12-29 2022-06-14 中国科学院上海药物研究所 Application of PCSK 9-targeted microRNA in treatment of LDLC-related metabolic diseases
CA3088112A1 (en) 2018-01-12 2019-07-18 Bristol-Myers Squibb Company Antisense oligonucleotides targeting alpha-synuclein and uses thereof
EP3765610A4 (en) * 2018-03-14 2022-01-19 Beth Israel Deaconess Medical Center Micro-rna and obesity
US20210071180A1 (en) * 2018-04-23 2021-03-11 Board Of Regents, The Universy Of Texas System Microrna 584-5p compositions and methods for treating cancer
CN109321655B (en) * 2018-05-17 2022-07-05 福建省肿瘤医院(福建省肿瘤研究所、福建省癌症防治中心) NKIRAS2 gene regulatory region sequence, regulatory sequence and application thereof in nasopharyngeal carcinoma
CA3110661A1 (en) 2018-08-29 2020-03-05 University Of Massachusetts Inhibition of protein kinases to treat friedreich ataxia
US20220056443A1 (en) * 2018-09-06 2022-02-24 Aptamir Therapeutics, Inc. Metabolic benefits of short mir-22 mirna antagomir therapies
WO2020073460A1 (en) 2018-10-09 2020-04-16 Amphenol Commercial Products (Chengdu) Co. Ltd. High-density edge connector
TWM576774U (en) 2018-11-15 2019-04-11 香港商安費諾(東亞)有限公司 Metal case with anti-displacement structure and connector thereof
TWM582251U (en) * 2019-04-22 2019-08-11 香港商安費諾(東亞)有限公司 Connector set with hidden locking mechanism and socket connector thereof
CN110283794B (en) * 2019-05-30 2021-04-23 伍泽堂 Recombinant oncolytic virus, preparation method, application and medicament thereof
WO2020261227A1 (en) 2019-06-26 2020-12-30 Biorchestra Co., Ltd. Micellar nanoparticles and uses thereof
US10844383B1 (en) 2019-09-18 2020-11-24 Dasman Diabetes Institute Microrna dyslipidemia inhibitor
US11588277B2 (en) 2019-11-06 2023-02-21 Amphenol East Asia Ltd. High-frequency electrical connector with lossy member
TW202127754A (en) 2019-11-06 2021-07-16 香港商安費諾(東亞)有限公司 High-frequency electrical connector with interlocking segments
US11058710B1 (en) 2020-02-14 2021-07-13 Dasman Diabetes Institute MicroRNA ANGPTL3 inhibitor
CA3174443A1 (en) * 2020-03-04 2021-09-10 The Trustees Of Indiana University Methods to re-engage a fetal wound healing pathway for adult skin repair
WO2021207644A1 (en) * 2020-04-09 2021-10-14 Biovista, Inc. Compositions and methods for treating viral infections
US11652307B2 (en) 2020-08-20 2023-05-16 Amphenol East Asia Electronic Technology (Shenzhen) Co., Ltd. High speed connector
CN212874843U (en) 2020-08-31 2021-04-02 安费诺商用电子产品(成都)有限公司 Electrical connector
WO2023122805A1 (en) 2021-12-20 2023-06-29 Vestaron Corporation Sorbitol driven selection pressure method
WO2023170659A1 (en) 2022-03-11 2023-09-14 Consejo Nacional De Investigaciones Cientificas Y Tecnicas (Conicet) Breast cancer diagnostic and treatment
WO2023212296A1 (en) * 2022-04-29 2023-11-02 University Of Massachusetts Selective rna-modulating agents
CN114990159A (en) * 2022-05-18 2022-09-02 昆明理工大学 Establishment method for inhibiting HCV proliferation by microRNA 206

Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4914210A (en) 1987-10-02 1990-04-03 Cetus Corporation Oligonucleotide functionalizing reagents
US4962029A (en) 1987-10-02 1990-10-09 Cetus Corporation Covalent oligonucleotide-horseradish peroxidase conjugate
WO1999014226A2 (en) 1997-09-12 1999-03-25 Exiqon A/S Bi- and tri-cyclic nucleoside, nucleotide and oligonucleotide analogues
WO2000056746A2 (en) 1999-03-24 2000-09-28 Exiqon A/S Improved synthesis of [2.2.1]bicyclo nucleosides
WO2000056748A1 (en) 1999-03-18 2000-09-28 Exiqon A/S Xylo-lna analogues
WO2001025248A2 (en) 1999-10-04 2001-04-12 Exiqon A/S Design of high affinity rnase h recruiting oligonucleotide
WO2002028875A2 (en) 2000-10-04 2002-04-11 Cureon A/S Improved synthesis of purine locked nucleic acid analogues
WO2003006475A2 (en) 2001-07-12 2003-01-23 Santaris Pharma A/S Method for preparation of lna phosphoramidites
WO2003095467A1 (en) 2002-05-08 2003-11-20 Santaris Pharma A/S Synthesis of locked nucleic acid derivatives
WO2005098029A2 (en) 2004-04-07 2005-10-20 Exiqon A/S Methods for quantification of micrornas and small interfering rnas
WO2006069584A2 (en) 2004-12-29 2006-07-06 Exiqon A/S NOVEL OLIGONUCLEOTIDE COMPOSITIONS AND PROBE SEQUENCES USEFUL FOR DETECTION AND ANALYSIS OF microRNAs AND THEIR TARGET mRNAs
US7087229B2 (en) 2003-05-30 2006-08-08 Enzon Pharmaceuticals, Inc. Releasable polymeric conjugates based on aliphatic biodegradable linkers
WO2007112753A2 (en) 2006-04-03 2007-10-11 Santaris Pharma A/S Pharmaceutical composition comprising anti-mirna antisense oligonucleotides
US20080034122A1 (en) 2006-08-02 2008-02-07 Robert Akira Kubo Apparatus and Method to Detect Miscabling in a Storage Area Network
WO2008034123A2 (en) 2006-09-15 2008-03-20 Enzon Pharmaceuticals, Inc. Polymeric conjugates containing positively-charged moieties
WO2008046911A2 (en) 2006-10-20 2008-04-24 Exiqon A/S Novel human micrornas associated with cancer

Family Cites Families (89)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2806208A (en) 1951-09-29 1957-09-10 Columbia Broadcasting Syst Inc Vacuum tube voltmeter
US4920115A (en) * 1988-12-28 1990-04-24 Virginia Commonwealth University Method of lowering LDL cholesterol in blood
JPH06311885A (en) 1992-08-25 1994-11-08 Mitsubishi Kasei Corp Anti-sense compound complementary with gene of hepatitis c virus
US6423489B1 (en) * 1992-09-10 2002-07-23 Isis Pharmaceuticals, Inc. Compositions and methods for treatment of Hepatitis C virus-associated diseases
EP0662157B1 (en) 1992-09-10 2001-06-20 Isis Pharmaceuticals, Inc. Compositions and methods for treatment of hepatitis c virus-associated diseases
US6433159B1 (en) * 1992-09-10 2002-08-13 Isis Pharmaceuticals, Inc. Compositions and methods for treatment of Hepatitis C virus associated diseases
WO1995030746A1 (en) 1994-05-10 1995-11-16 The General Hospital Corporation Antisense inhibition of hepatitis c virus
ATE233734T1 (en) * 1995-06-07 2003-03-15 Pfizer BIPHENYL-2-CARBONIC ACID-TETRAHYDRO-ISOCHINOLIN-6 YL AMIDE DERIVATIVES, THEIR PREPARATION AND THEIR USE AS INHIBITORS OF THE MICROSOMAL TRIGLYCERIDE TRANSFER PROTEIN AND/OR THE APOLIPOPROTEIN B (APO B) SECRETION
SK65499A3 (en) * 1996-11-27 2001-05-10 Pfizer Apo b-secretion/mtp inhibitory amides
EP0975809A4 (en) * 1997-03-05 2002-10-30 Univ Washington Novel screening methods to identify agents that selectively inhibit hepatitis c virus replication
US6264458B1 (en) * 1998-02-20 2001-07-24 Decoma International Corp. Mold clamp
PT1178999E (en) * 1999-05-04 2007-06-26 Santaris Pharma As L-ribo-lna analogues
US6617442B1 (en) 1999-09-30 2003-09-09 Isis Pharmaceuticals, Inc. Human Rnase H1 and oligonucleotide compositions thereof
IL139450A0 (en) 1999-11-10 2001-11-25 Pfizer Prod Inc Methods of administering apo b-secretion/mtp inhibitors
AU2001277170A1 (en) 2000-07-25 2002-02-05 Authentisure, Inc. Unified trust model providing secure identification, authentication and validation of physical products and entities, and processing, storage, and exchange of information
AU2002250236A1 (en) 2001-03-02 2002-09-19 Medimmune, Inc. Cd2 antagonists for treatment of autoimmune or inflammatory disease
EP1383782A1 (en) 2001-03-26 2004-01-28 Sirna Therpeutics, Inc. Oligonucleotide mediated inhibition of hepatitis b virus and hepatitis c virus replication
WO2003070750A2 (en) 2002-02-20 2003-08-28 Sirna Therapeutics, Inc Rna interference mediated inhibition of hepatitis c virus
US20030125241A1 (en) 2001-05-18 2003-07-03 Margit Wissenbach Therapeutic uses of LNA-modified oligonucleotides in infectious diseases
US7407943B2 (en) 2001-08-01 2008-08-05 Isis Pharmaceuticals, Inc. Antisense modulation of apolipoprotein B expression
EP2385123B1 (en) 2001-09-28 2018-04-25 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Microrna molecules
US7459461B2 (en) * 2001-10-19 2008-12-02 Ortho-Mcneil Pharmaceutical, Inc. Phosphonic acid compounds as inhibitors of serine proteases
WO2004014313A2 (en) * 2002-08-12 2004-02-19 Bristol-Myers Squibb Company Combination pharmaceutical agents as inhibitors of hcv replication
UA79300C2 (en) * 2002-08-12 2007-06-11 Janssen Pharmaceutica Nv N-aryl piperidine substituted biphenylcarboxamides as inhibitors of apolipoprotein b secretion
JP4986109B2 (en) 2002-11-13 2012-07-25 ジェンザイム・コーポレーション Antisense regulation of apolipoprotein B expression
DK2284269T3 (en) 2002-11-18 2017-10-23 Roche Innovation Ct Copenhagen As Antisense design
US8124582B2 (en) * 2002-12-06 2012-02-28 Fibrogen, Inc. Treatment of diabetes
FR2848572B1 (en) * 2002-12-12 2005-12-09 Univ Joseph Fourier INHIBITOR MOLECULES OF PROTEIN SYNTHESIS OF HEPATITIS C VIRUS AND METHOD FOR SCREENING THESE INHIBITORY MOLECULES
DK1592793T3 (en) 2003-02-10 2009-10-26 Santaris Pharma As Oligomeric Compounds for Modulation of Survivin Expression
EP1592791A2 (en) 2003-02-10 2005-11-09 National Institute of Advanced Industrial Science and Technology Regulation of gene expression by dna interference
US7683036B2 (en) 2003-07-31 2010-03-23 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAs
WO2005013905A2 (en) 2003-08-07 2005-02-17 Avi Biopharma, Inc. SENSE ANTIVIRAL COMPOUND AND METHOD FOR TREATING ssRNA VIRAL INFECTION
US20050142581A1 (en) 2003-09-04 2005-06-30 Griffey Richard H. Microrna as ligands and target molecules
GB2442373B (en) 2003-11-26 2008-10-22 Univ Massachusetts Sequence -specific inhibition of small rna function
UA83510C2 (en) 2003-12-09 2008-07-25 Янссен Фармацевтика Н.В. N-aryl piperidine substituted biphenylcarboxamides as inhibitors of apolipoprotein b
US7622453B2 (en) 2003-12-23 2009-11-24 Santaris Pharma A/S Oligomeric compounds for the modulation of Bcl-2
EP2295604B1 (en) 2004-02-09 2015-04-08 Thomas Jefferson University Diagnosis and treatment of cancers with microRNA located in or near cancer-associated chromosomal features
US20050182005A1 (en) 2004-02-13 2005-08-18 Tuschl Thomas H. Anti-microRNA oligonucleotide molecules
AU2005214904B2 (en) 2004-02-13 2011-07-21 Rockefeller University Anti-microRNA oligonucleotide molecules
DE102004015101B4 (en) 2004-03-27 2012-04-26 Eastman Kodak Co. Method and sensor device for controlling an endless, seam-containing transport medium for a printing press
US7365058B2 (en) * 2004-04-13 2008-04-29 The Rockefeller University MicroRNA and methods for inhibiting same
KR20070004957A (en) 2004-04-20 2007-01-09 제나코 바이오메디컬 프로덕츠, 인코포레이티드 Method for detecting ncrna
EP2338996A3 (en) * 2004-05-04 2013-12-18 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for reducing viral genome amounts in a target cell
WO2006093526A2 (en) 2004-07-21 2006-09-08 Alnylam Pharmaceuticals, Inc. Oligonucleotides comprising a modified or non-natural nucleobase
US20080213891A1 (en) 2004-07-21 2008-09-04 Alnylam Pharmaceuticals, Inc. RNAi Agents Comprising Universal Nucleobases
FR2873694B1 (en) 2004-07-27 2006-12-08 Merck Sante Soc Par Actions Si NEW AZA-INDOLES INHIBITORS OF MTP AND APOB
AU2005330637B2 (en) 2004-08-04 2012-09-20 Alnylam Pharmaceuticals, Inc. Oligonucleotides comprising a ligand tethered to a modified or non-natural nucleobase
EP1791567B1 (en) 2004-08-10 2015-07-29 Alnylam Pharmaceuticals Inc. Chemically modified oligonucleotides
PT1786472E (en) * 2004-08-10 2013-03-06 Genzyme Corp Antisense modulation of apolipoprotein b expression
WO2006027776A1 (en) 2004-09-07 2006-03-16 Yissum Research Development Company Of The Hebrew University Of Jerusalem Agents, compositions and methods for treating pathologies in which regulating an ache-associated biological pathway is beneficial
NZ554214A (en) 2004-09-24 2009-10-30 Alnylam Pharmaceuticals Inc Rnai modulation of apob and uses thereof
US8173611B2 (en) 2004-11-12 2012-05-08 Asuragen Inc. Methods and compositions involving miRNA and miRNA inhibitor molecules
AU2005306533B2 (en) 2004-11-17 2012-05-31 Arbutus Biopharma Corporation siRNA silencing of apolipoprotein B
US20060185027A1 (en) * 2004-12-23 2006-08-17 David Bartel Systems and methods for identifying miRNA targets and for altering miRNA and target expression
US20070065840A1 (en) * 2005-03-23 2007-03-22 Irena Naguibneva Novel oligonucleotide compositions and probe sequences useful for detection and analysis of microRNAS and their target mRNAS
EP1877369B1 (en) 2005-04-19 2013-07-10 Surface Logix, Inc. Inhibitors of microsomal triglyceride transfer protein and apo-b secretion
US20060265771A1 (en) 2005-05-17 2006-11-23 Lewis David L Monitoring microrna expression and function
WO2006133022A2 (en) 2005-06-03 2006-12-14 The Johns Hopkins University Compositions and methods for decreasing microrna expression for the treatment of neoplasia
US20070123482A1 (en) 2005-08-10 2007-05-31 Markus Stoffel Chemically modified oligonucleotides for use in modulating micro RNA and uses thereof
US20070213292A1 (en) 2005-08-10 2007-09-13 The Rockefeller University Chemically modified oligonucleotides for use in modulating micro RNA and uses thereof
DK1931780T3 (en) 2005-08-29 2016-01-25 Regulus Therapeutics Inc Antisense-forbindelser med forøget anti-microrna-aktivitet
WO2007027775A2 (en) * 2005-08-29 2007-03-08 Isis Pharmaceuticals, Inc. Methods for use in modulating mir-122a
KR20080068019A (en) 2005-09-15 2008-07-22 산타리스 팔마 에이/에스 Rna antagonist compounds for the inhibition of apo-b100 expression
WO2007031091A2 (en) 2005-09-15 2007-03-22 Santaris Pharma A/S Rna antagonist compounds for the modulation of p21 ras expression
JP5213723B2 (en) 2006-01-27 2013-06-19 アイシス ファーマシューティカルズ, インコーポレーテッド Oligomer compounds and compositions for use in modulating microRNA
EP3502255A1 (en) * 2006-04-03 2019-06-26 Roche Innovation Center Copenhagen A/S Pharmaceutical composition
CN103554205A (en) 2006-05-05 2014-02-05 Isis制药公司 Compounds and methods for modulating expression of gccr
AU2007249349B2 (en) 2006-05-11 2012-03-08 Isis Pharmaceuticals, Inc. 5'-Modified bicyclic nucleic acid analogs
US20080199961A1 (en) 2006-08-25 2008-08-21 Avi Biopharma, Inc. ANTISENSE COMPOSITION AND METHOD FOR INHIBITION OF miRNA BIOGENESIS
MX2009002859A (en) 2006-09-15 2009-03-30 Enzon Pharmaceuticals Inc Hindered ester-based biodegradable linkers for oligonucleotide delivery.
WO2008057234A2 (en) 2006-10-24 2008-05-15 The Board Of Trustees Of The Leland Stanford Junior University Modulation of t cell signaling threshold and t cell sensitivity to antigens
CN101554074B (en) 2006-10-30 2012-08-22 诺基亚公司 Method, apparatus and system providing operator controlled mobility for user equipment
WO2008061537A2 (en) * 2006-11-23 2008-05-29 Querdenker Aps Oligonucleotides for modulating target rna activity
WO2008074328A2 (en) 2006-12-21 2008-06-26 Exiqon A/S Microrna target site blocking oligos and uses thereof
DK2149605T3 (en) 2007-03-22 2013-09-30 Santaris Pharma As Short RNA antagonist compounds to modulate the desired mRNA
WO2008124384A2 (en) 2007-04-03 2008-10-16 Aegerion Pharmaceuticals, Inc. Combinations of mtp inhibitors with cholesterol absorption inhibitors or interferon for treating hepatitis c
US8278425B2 (en) 2007-05-30 2012-10-02 Isis Pharmaceuticals, Inc. N-substituted-aminomethylene bridged bicyclic nucleic acid analogs
EP2173760B2 (en) 2007-06-08 2015-11-04 Isis Pharmaceuticals, Inc. Carbocyclic bicyclic nucleic acid analogs
CA2690643A1 (en) 2007-06-14 2008-12-18 Mirx Therapeutics Aps Oligonucleotides for modulation of target rna activity
US20090082297A1 (en) 2007-06-25 2009-03-26 Lioy Daniel T Compositions and Methods for Regulating Gene Expression
US20110009466A1 (en) 2007-08-29 2011-01-13 President And Fellows Of Harvard College Methods of increasing gene expression through rna protection
US8440637B2 (en) 2007-10-04 2013-05-14 Santaris Pharma A/S Combination treatment for the treatment of hepatitis C virus infection
CA2717792A1 (en) 2008-03-07 2009-09-11 Santaris Pharma A/S Pharmaceutical compositions for treatment of microrna related diseases
WO2010000665A1 (en) 2008-06-30 2010-01-07 Santaris Pharma A/S Antidote oligomers
WO2010012667A1 (en) 2008-08-01 2010-02-04 Santaris Pharma A/S Micro-rna mediated modulation of colony stimulating factors
WO2010013999A1 (en) 2008-08-01 2010-02-04 Twister B.V. Cyclonic separator with a volute outlet duct
EP2421970B1 (en) * 2009-04-24 2016-09-07 Roche Innovation Center Copenhagen A/S Pharmaceutical compositions for treatment of hcv patients that are non-responders to interferon
US8942209B2 (en) 2009-08-12 2015-01-27 Qualcomm Incorporated Systems and methods for supporting an enhanced serving cell change when moving among different cell types
US9364495B2 (en) 2009-10-20 2016-06-14 Roche Innovation Center Copenhagen A/S Oral delivery of therapeutically effective LNA oligonucleotides

Patent Citations (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4914210A (en) 1987-10-02 1990-04-03 Cetus Corporation Oligonucleotide functionalizing reagents
US4962029A (en) 1987-10-02 1990-10-09 Cetus Corporation Covalent oligonucleotide-horseradish peroxidase conjugate
WO1999014226A2 (en) 1997-09-12 1999-03-25 Exiqon A/S Bi- and tri-cyclic nucleoside, nucleotide and oligonucleotide analogues
WO2000056748A1 (en) 1999-03-18 2000-09-28 Exiqon A/S Xylo-lna analogues
WO2000056746A2 (en) 1999-03-24 2000-09-28 Exiqon A/S Improved synthesis of [2.2.1]bicyclo nucleosides
WO2001025248A2 (en) 1999-10-04 2001-04-12 Exiqon A/S Design of high affinity rnase h recruiting oligonucleotide
WO2002028875A2 (en) 2000-10-04 2002-04-11 Cureon A/S Improved synthesis of purine locked nucleic acid analogues
WO2003006475A2 (en) 2001-07-12 2003-01-23 Santaris Pharma A/S Method for preparation of lna phosphoramidites
WO2003095467A1 (en) 2002-05-08 2003-11-20 Santaris Pharma A/S Synthesis of locked nucleic acid derivatives
US7087229B2 (en) 2003-05-30 2006-08-08 Enzon Pharmaceuticals, Inc. Releasable polymeric conjugates based on aliphatic biodegradable linkers
WO2005098029A2 (en) 2004-04-07 2005-10-20 Exiqon A/S Methods for quantification of micrornas and small interfering rnas
WO2006069584A2 (en) 2004-12-29 2006-07-06 Exiqon A/S NOVEL OLIGONUCLEOTIDE COMPOSITIONS AND PROBE SEQUENCES USEFUL FOR DETECTION AND ANALYSIS OF microRNAs AND THEIR TARGET mRNAs
WO2007112753A2 (en) 2006-04-03 2007-10-11 Santaris Pharma A/S Pharmaceutical composition comprising anti-mirna antisense oligonucleotides
WO2007112754A2 (en) 2006-04-03 2007-10-11 Santaris Pharma A/S Pharmaceutical compositions comprising anti-mirna antisense oligonucleotides
US20080034122A1 (en) 2006-08-02 2008-02-07 Robert Akira Kubo Apparatus and Method to Detect Miscabling in a Storage Area Network
WO2008034123A2 (en) 2006-09-15 2008-03-20 Enzon Pharmaceuticals, Inc. Polymeric conjugates containing positively-charged moieties
WO2008046911A2 (en) 2006-10-20 2008-04-24 Exiqon A/S Novel human micrornas associated with cancer

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
CROOKE, R. M.: "Crooke, S. T. Antisense research and Application", vol. 131, 1998, SPRINGER-VERLAG, pages: 103 - 140
DASS CR., J PHARM PHARMACOL, vol. 54, no. 1, 2002, pages 3 - 27
MANOHARAN ET AL., TETRAHEDRON LETTERS, vol. 34, 1991, pages 7171

Cited By (267)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9752146B2 (en) 2003-07-31 2017-09-05 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAs
US10093926B2 (en) 2003-07-31 2018-10-09 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAs
US8809294B2 (en) 2003-07-31 2014-08-19 Regulus Therapeutics Inc. Method of inhibiting miR-33 using a modified oligonucleotide
US9528108B2 (en) 2003-07-31 2016-12-27 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAS
US10072265B2 (en) 2003-07-31 2018-09-11 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAs
US8765701B2 (en) 2003-07-31 2014-07-01 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAs
US10584336B2 (en) 2003-07-31 2020-03-10 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAs
US9447413B2 (en) 2003-07-31 2016-09-20 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAs
US9447412B2 (en) 2003-07-31 2016-09-20 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAS
US9139832B2 (en) 2003-07-31 2015-09-22 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAs
US8859521B2 (en) 2003-07-31 2014-10-14 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAs
US8697663B2 (en) 2003-07-31 2014-04-15 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAs
US9663787B2 (en) 2003-07-31 2017-05-30 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAs
US9267138B2 (en) 2003-07-31 2016-02-23 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAs
US8729250B2 (en) 2006-04-03 2014-05-20 Joacim Elmén Antisense oligonucleotides for inhibition of microRNA-21
US8163708B2 (en) 2006-04-03 2012-04-24 Santaris Pharma A/S Pharmaceutical composition comprising anti-mirna antisense oligonucleotide
US8940712B2 (en) 2007-07-31 2015-01-27 The Board Of Regents, The University Of Texas System Micro-RNA family that modulates fibrosis and uses thereof
US10392618B2 (en) 2007-07-31 2019-08-27 The Board Of Regents, The University Of Texas System Micro-RNA family that modulates extracellular matrix genes and uses thereof
US9719086B2 (en) 2007-07-31 2017-08-01 The Board Of Regents, The University Of Texas System Micro-RNA family that modulates fibrosis and uses thereof
US8481507B2 (en) 2007-07-31 2013-07-09 The Board Of Regents, The University Of Texas System Micro-RNAs that control myosin expression and myofiber identity
US8440636B2 (en) 2007-07-31 2013-05-14 The Board Of Regents, The University Of Texas System Micro-RNA family that modulates fibrosis and uses thereof
US9719088B2 (en) 2007-07-31 2017-08-01 The Board Of Regents, The University Of Texas System Micro-RNA family that modulates fibrosis and uses thereof
US8962588B2 (en) 2007-07-31 2015-02-24 The Board Of Regents, The University Of Texas System Micro-RNAS that control myosin expression and myofiber identity
US8940711B2 (en) 2007-07-31 2015-01-27 The Board Of Regents, The University Of Texas System Micro-RNA family that modulates fibrosis and uses thereof
US8940713B2 (en) 2007-07-31 2015-01-27 The Board Of Regents, The University Of Texas System Micro-RNA family that modulates fibrosis and uses thereof
US9719087B2 (en) 2007-07-31 2017-08-01 The Board Of Regents, The University Of Texas System Micro-RNA family that modulates fibrosis and uses thereof
US8440637B2 (en) 2007-10-04 2013-05-14 Santaris Pharma A/S Combination treatment for the treatment of hepatitis C virus infection
US8906871B2 (en) 2007-10-04 2014-12-09 Santaris Pharma A/S MicromiRs
US8288356B2 (en) 2007-10-04 2012-10-16 Santaris Pharma A/S MicroRNAs
US10450564B2 (en) 2007-10-04 2019-10-22 Roche Innovation Center Copenhagen A/S Micromirs
US9845470B2 (en) 2007-10-29 2017-12-19 Regulus Therapeutics Inc. Targeting microRNAS for the treatment of liver cancer
US10301627B2 (en) 2007-10-29 2019-05-28 Regulus Therapeutics Inc. Targeting microRNAs for the treatment of liver cancer
US9506062B2 (en) 2007-10-29 2016-11-29 Regulus Therapeutics Inc. Targeting microRNAs for the treatment of liver cancer
US8680067B2 (en) 2007-10-29 2014-03-25 Regulus Therapeutics, Inc. Targeting microRNAs for the treatment of liver cancer
US9078919B2 (en) 2007-11-09 2015-07-14 The Board Of Regents, The University Of Texas System Micro-RNAs of the miR-15 family modulate cardiomyocyte survival and cardiac repair
US8513209B2 (en) 2007-11-09 2013-08-20 The Board Of Regents, The University Of Texas System Micro-RNAS of the MIR-15 family modulate cardiomyocyte survival and cardiac repair
US8361980B2 (en) 2008-03-07 2013-01-29 Santaris Pharma A/S Pharmaceutical compositions for treatment of microRNA related diseases
US8404659B2 (en) 2008-03-07 2013-03-26 Santaris Pharma A/S Pharmaceutical compositions for treatment of MicroRNA related diseases
US8728724B2 (en) 2008-03-17 2014-05-20 Board Of Regents, The University Of Texas System Identification of micro-RNAs involved in neuromuscular synapse maintenance and regeneration
US8202848B2 (en) 2008-03-17 2012-06-19 Board Of Regents, The University Of Texas System Identification of micro-RNAS involved in neuromuscular synapse maintenance and regeneration
US8492357B2 (en) 2008-08-01 2013-07-23 Santaris Pharma A/S Micro-RNA mediated modulation of colony stimulating factors
EP2315832B1 (en) 2008-08-01 2015-04-08 Roche Innovation Center Copenhagen A/S Micro-rna mediated modulation of colony stimulating factors
EP2315832A1 (en) 2008-08-01 2011-05-04 Santaris Pharma A/S Micro-rna mediated modulation of colony stimulating factors
US8629119B2 (en) 2009-02-04 2014-01-14 The Board Of Regents, The University Of Texas System Dual targeting of MIR-208 and MIR-499 in the treatment of cardiac disorders
US9034837B2 (en) 2009-04-24 2015-05-19 Roche Innovation Center Copenhagen A/S Pharmaceutical compositions for treatment of HCV patients that are poor-responders to interferon
US8877730B2 (en) 2009-05-20 2014-11-04 Eth Zurich Targeting microRNAs for metabolic disorders
US9932595B2 (en) 2009-05-20 2018-04-03 Eth Zurich Targeting microRNAs for metabolic disorders
US10287589B2 (en) 2009-05-20 2019-05-14 Eth Zurich Targeting microRNAs for metabolic disorders
US9243249B2 (en) 2009-05-20 2016-01-26 Eth Zurich Targeting MicroRNAs for metabolic disorders
JP2016106077A (en) * 2009-05-20 2016-06-16 イーティーエイチ チューリッヒ Targeting micrornas for metabolic disorders
US8592388B2 (en) 2009-05-20 2013-11-26 Eth Zurich Targeting MicroRNAs for metabolic disorders
WO2010133970A1 (en) 2009-05-20 2010-11-25 Eth Zurich Targeting micrornas for metabolic disorders
US9550996B2 (en) 2009-05-20 2017-01-24 Eth Zurich Targeting microRNAs for metabolic disorders
WO2010135714A3 (en) * 2009-05-22 2011-01-13 The Methodist Hospital Research Institute Methods for modulating adipocyte expression using microrna compositions
EP2440566A1 (en) 2009-06-08 2012-04-18 Miragen Therapeutics CHEMICAL MODIFICATION MOTIFS FOR miRNA INHIBITORS AND MIMETICS
JP2012529295A (en) * 2009-06-08 2012-11-22 ミラゲン セラピューティクス miRNA inhibitors and mimetic chemical modification motifs
EP2440566A4 (en) * 2009-06-08 2013-10-16 Miragen Therapeutics CHEMICAL MODIFICATION MOTIFS FOR miRNA INHIBITORS AND MIMETICS
US20140357699A1 (en) * 2009-08-06 2014-12-04 Universiteit Maastricht Means and methods for counteracting, delaying and/or preventing adverse energy metabolism switches in heart disease
US9611480B2 (en) * 2009-08-06 2017-04-04 Universiteit Maastricht Means and methods for counteracting, delaying and/or preventing adverse energy metabolism switches in heart disease
US9090937B2 (en) 2009-09-02 2015-07-28 L'oreal Epidermal differentiation microrna signature and uses thereof
CN102625853A (en) * 2009-09-02 2012-08-01 欧莱雅 Epidermal differentiation microRNA signature and uses thereof
KR101873440B1 (en) 2009-09-02 2018-07-02 로레알 Epidermal differentiation microrna signature and uses thereof
WO2011026909A1 (en) * 2009-09-02 2011-03-10 L'oreal Epidermal differentiation microrna signature and uses thereof
WO2011048125A1 (en) 2009-10-20 2011-04-28 Santaris Pharma A/S Oral delivery of therapeutically effective lna oligonucleotides
US9364495B2 (en) 2009-10-20 2016-06-14 Roche Innovation Center Copenhagen A/S Oral delivery of therapeutically effective LNA oligonucleotides
US9566293B2 (en) 2009-10-20 2017-02-14 Roche Innovation Center Copenhagen A/S Oral delivery of therapeutically effective LNA oligonucleotides
WO2011060100A1 (en) * 2009-11-11 2011-05-19 Sanford-Burnham Medical Research Institute Method for generation and regulation of ips cells and compositions thereof
CN102666878A (en) * 2009-11-13 2012-09-12 江苏命码生物科技有限公司 Markers, methods, biochips and kits for milk qualit y detection
US9534219B2 (en) 2010-03-12 2017-01-03 The Brigham And Women's Hospital, Inc. Methods of treating vascular inflammatory disorders
US20130102546A1 (en) * 2010-03-12 2013-04-25 The Brigham And Women's Hospital, Inc. Methods of treating vascular inflammatory disorders
US8906875B2 (en) * 2010-03-12 2014-12-09 The Brigham And Women's Hospital, Inc. Methods of treating vascular inflammatory disorders
WO2011131354A1 (en) * 2010-04-20 2011-10-27 Febit Holding Gmbh Complex mirna sets as novel biomarkers for an acute coronary syndrome
US9611511B2 (en) 2010-04-20 2017-04-04 Comprehensive Biomarker Center Gmbh Complex miRNA sets as novel biomarkers for an acute coronary syndrome
EP2865765A1 (en) * 2010-04-20 2015-04-29 Comprehensive Biomarker Center GmbH Complex miRNA sets as novel biomarkers for an acute coronary syndrome
US9682095B2 (en) 2010-07-06 2017-06-20 Interna Technologies B.V. MiRNA and its diagnostic and therapeutic uses in diseases or conditions associated with melanoma, or in diseases or conditions associated with activated BRAF pathway
CN102002494A (en) * 2010-07-08 2011-04-06 浙江理工大学 microRNA biomarker and application thereof
WO2012010711A1 (en) 2010-07-23 2012-01-26 Santaris Pharma A/S Process
US8815826B2 (en) 2010-07-23 2014-08-26 Regulus Therapeutics, Inc. Targeting microRNAs for the treatment of fibrosis
US9181548B2 (en) 2010-07-23 2015-11-10 Regulus Therapeutics Inc. Targeting micrornas for the treatment of fibrosis
US9970007B2 (en) 2010-07-23 2018-05-15 Regulus Therapeutics Inc. Targeting microRNAs for the treatment of fibrosis
US9556435B2 (en) 2010-07-23 2017-01-31 Regulus Therapeutics Inc. Targeting microRNAs for the treatment of fibrosis
US8859519B2 (en) 2010-08-25 2014-10-14 The General Hospital Corporation Methods targeting miR-33 microRNAs for regulating lipid metabolism
US9328346B2 (en) 2010-11-12 2016-05-03 The General Hospital Corporation Polycomb-associated non-coding RNAs
US10119144B2 (en) 2010-11-12 2018-11-06 The General Hospital Corporation Polycomb-associated non-coding RNAs
US10358644B2 (en) 2010-11-12 2019-07-23 The General Hospital Corporation Polycomb-associated non-coding RNAs
US10053694B2 (en) 2010-11-12 2018-08-21 The General Hospital Corporation Polycomb-associated non-coding RNAS
US9920317B2 (en) 2010-11-12 2018-03-20 The General Hospital Corporation Polycomb-associated non-coding RNAs
US9816094B2 (en) 2010-11-12 2017-11-14 The General Hospital Corporation Polycomb-associated non-coding RNAs
US9856479B2 (en) 2010-11-12 2018-01-02 The General Hospital Corporation Polycomb-associated non-coding RNAs
US11066673B2 (en) 2010-11-12 2021-07-20 The General Hospital Corporation Polycomb-associated non-coding RNAs
US8642751B2 (en) 2010-12-15 2014-02-04 Miragen Therapeutics MicroRNA inhibitors comprising locked nucleotides
US9441222B2 (en) 2011-01-11 2016-09-13 Interna Technologies B.V. MiRNA for treating diseases and conditions associated with neo-angiogenesis
CN102643811B (en) * 2011-02-18 2015-12-09 中国科学院上海药物研究所 The antisense oligonucleotide of people miR-1229 and application thereof
CN102643813B (en) * 2011-02-18 2015-07-29 中国科学院上海药物研究所 The antisense oligonucleotide of people miR-504 and application thereof
CN102643809B (en) * 2011-02-18 2016-01-20 中国科学院上海药物研究所 The antisense oligonucleotide of people miR-1274b and application thereof
CN102643814B (en) * 2011-02-18 2015-06-17 中国科学院上海药物研究所 Antisense oligonucleotide for human miR-431 and application thereof
CN102643806B (en) * 2011-02-18 2016-08-03 中国科学院上海药物研究所 The antisense oligonucleotide of people miR-1913 and application thereof
CN102643810B (en) * 2011-02-18 2015-12-16 中国科学院上海药物研究所 The antisense oligonucleotide of people miR-299-5p and application thereof
CN102643810A (en) * 2011-02-18 2012-08-22 中国科学院上海药物研究所 Human miR-299-5p antisense oligonucleotide and application thereof
CN102643811A (en) * 2011-02-18 2012-08-22 中国科学院上海药物研究所 Human miR-1229 antisense oligonucleotide and application thereof
CN102643806A (en) * 2011-02-18 2012-08-22 中国科学院上海药物研究所 Antisense oligodeoxyncleotide of human miR-1913 and application thereof
CN102643809A (en) * 2011-02-18 2012-08-22 中国科学院上海药物研究所 Antisense oligodeoxyncleotide of human miR-1274b and application thereof
CN102643813A (en) * 2011-02-18 2012-08-22 中国科学院上海药物研究所 Human miR-504 antisense oligonucleotide and application thereof
CN102643814A (en) * 2011-02-18 2012-08-22 中国科学院上海药物研究所 Antisense oligonucleotide for human miR-431 and application thereof
WO2012145374A1 (en) 2011-04-19 2012-10-26 Regulus Therapeutics Inc. TARGETING miR-378 FAMILY MEMBERS FOR THE TREATMENT OF METABOLIC DISORDERS
AU2012249851B2 (en) * 2011-04-25 2017-06-08 Sanofi MicroRNA compounds and methods for modulating miR-21 activity
US9790496B2 (en) 2011-04-25 2017-10-17 Regulus Therapeutics Inc. Microrna compounds and methods for modulating MIR-21 activity
KR102055172B1 (en) * 2011-04-25 2020-01-23 레굴루스 테라퓨틱스 인크 Microrna compounds and methods for modulating mir-21 activity
US9181547B2 (en) 2011-04-25 2015-11-10 Regulus Therapeutics Inc. MicroRNA compounds and methods for modulating MIR-21 activity
WO2012148952A1 (en) * 2011-04-25 2012-11-01 Regulus Therapeutics Inc Microrna compounds and methods for modulating mir-21 activity
EP3933040A1 (en) * 2011-04-25 2022-01-05 Sanofi Microrna compounds and methods for modulating mir-21 activity
EP3211082A1 (en) * 2011-04-25 2017-08-30 Regulus Therapeutics Inc. Microrna compounds and methods for modulating mir-21 activity
KR20140033062A (en) * 2011-04-25 2014-03-17 레굴루스 테라퓨틱스 인크 Microrna compounds and methods for modulating mir-21 activity
CN103620035B (en) * 2011-04-25 2021-04-23 赛诺菲 microRNA compounds and methods for modulating MIR-21 activity
EA025894B1 (en) * 2011-04-25 2017-02-28 Регулус Терапьютикс Инк. MICRORNA COMPOUNDS AND METHODS FOR MODULATING miR-21 ACTIVITY
CN103620035A (en) * 2011-04-25 2014-03-05 莱古路斯治疗法股份有限公司 Microrna compounds and methods for modulating mir-21 activity
US8912161B2 (en) 2011-04-25 2014-12-16 Regulus Therapeutics, Inc. MicroRNA compounds and methods for modulating miR-21 activity
US20140080899A1 (en) * 2011-04-28 2014-03-20 New York University miR-33 Inhibitors and Uses Thereof to Decrease Inflammation
US9241950B2 (en) * 2011-04-28 2016-01-26 New York University MiR-33 inhibitors and uses thereof to decrease inflammation
KR101800243B1 (en) 2011-06-22 2017-12-20 (주)아모레퍼시픽 Composition for regulating expression of pigmentation-related genes containing microRNA
WO2012175733A1 (en) 2011-06-23 2012-12-27 Santaris Pharma A/S Hcv combination therapy
WO2012175357A1 (en) * 2011-06-24 2012-12-27 Syddansk Universitet Modulation of microrna-138 for the treatment of bone loss
WO2013000856A1 (en) 2011-06-30 2013-01-03 Santaris Pharma A/S Hcv combination therapy
WO2013000855A1 (en) 2011-06-30 2013-01-03 Santaris Pharma A/S Hcv combination therapy
CN102851282A (en) * 2011-06-30 2013-01-02 上海市肿瘤研究所 MicroRNA markers for discriminating constitutional lung cancer tissue and paracancerous tissue
US9617538B2 (en) * 2011-08-29 2017-04-11 Niigata University Of Pharmacy And Applied Life Science Heptamer-type small guide nucleic acids inducing apoptosis of human leukemia cells
US20150025232A1 (en) * 2011-08-29 2015-01-22 Masayuki Nashimoto Heptamer-type small guide nucleic acids inducing apoptosis of human leukemia cells
EP2755663A4 (en) * 2011-09-13 2015-10-07 Ottawa Hospital Res Inst Microrna inhibitors
US9580708B2 (en) 2011-09-14 2017-02-28 Rana Therapeutics, Inc. Multimeric oligonucleotides compounds
US10093924B2 (en) 2011-09-14 2018-10-09 Translate Bio Ma, Inc. Multimetric oligonucleotide compounds
US9732340B2 (en) 2011-09-14 2017-08-15 Translate Bio Ma, Inc. Multimeric oligonucleotides compounds having cleavable linkers
US10704046B2 (en) 2011-09-14 2020-07-07 Translate Bio Ma, Inc. Multimeric oligonucleotide compounds
US9732341B2 (en) 2011-09-14 2017-08-15 Translate Bio Ma, Inc. Methods of delivering multiple targeting oligonucleotides to a cell using cleavable linkers
US9428749B2 (en) 2011-10-06 2016-08-30 The Board Of Regents, The University Of Texas System Control of whole body energy homeostasis by microRNA regulation
WO2013068347A1 (en) 2011-11-07 2013-05-16 Santaris Pharma A/S Prognostic method for checking efficacy of micro rna-122 inhibitors in hcv+ patients
WO2013068348A1 (en) 2011-11-07 2013-05-16 Santaris Pharma A/S Lna oligomers for improvement in hepatic function
EP2785355A4 (en) * 2011-11-30 2015-12-02 Cedars Sinai Medical Center Targeting micrornas mir-409-5p, mir-379 and mir-154* to treat prostate cancer bone metastasis and drug resistant lung cancer
US9745578B2 (en) 2011-11-30 2017-08-29 Cedars-Sinai Medical Center Targeting microRNA miR-409-3P to treat prostate cancer
EP3106168A3 (en) * 2011-11-30 2017-02-22 Cedars-Sinai Medical Center Targeting micrornas mir-409-5p, mir-379 and mir-154* to treat prostate cancer bone metastasis and drug resistant lung cancer
EP2816114A3 (en) * 2011-12-15 2015-02-25 Oncostamen S.r.l. MicroRNAs and uses thereof
WO2013134403A1 (en) * 2012-03-06 2013-09-12 The Washington University Method of treating neurodegenerative diseases with microrna regulators
KR20200064164A (en) * 2012-04-25 2020-06-05 사노피 Microrna compounds and methods for modulating mir-21 activity
US8969317B2 (en) 2012-04-25 2015-03-03 Regulus Therapeutics Inc. MicroRNA compounds and methods for modulating miR-21 activity
US9970009B2 (en) 2012-04-25 2018-05-15 Regulus Therapeutics Inc. MicroRNA compounds and methods for modulating miR-21 activity
US9688985B2 (en) 2012-04-25 2017-06-27 Regulus Therapeutics Inc. MicroRNA compounds and methods for modulating miR-21 activity
EA030211B1 (en) * 2012-04-25 2018-07-31 Регьюлэс Терапьютикс Инк. MICRORNA COMPOUNDS AND METHODS FOR MODULATING miR-21 ACTIVITY
WO2013163258A1 (en) * 2012-04-25 2013-10-31 Regulus Therapeutics Inc. Microrna compounds and methods for modulating mir-21 activity
US9267137B2 (en) 2012-04-25 2016-02-23 Regulus Therapeutics Inc. MicroRNA compounds and methods for modulating miR-21 activity
US9334498B2 (en) 2012-05-10 2016-05-10 Uab Research Foundation Methods and compositions for modulating MIR-204 activity
US11788089B2 (en) 2012-05-16 2023-10-17 The General Hospital Corporation Compositions and methods for modulating MECP2 expression
US10059941B2 (en) 2012-05-16 2018-08-28 Translate Bio Ma, Inc. Compositions and methods for modulating SMN gene family expression
US10174323B2 (en) 2012-05-16 2019-01-08 The General Hospital Corporation Compositions and methods for modulating ATP2A2 expression
US10837014B2 (en) 2012-05-16 2020-11-17 Translate Bio Ma, Inc. Compositions and methods for modulating SMN gene family expression
US10174315B2 (en) 2012-05-16 2019-01-08 The General Hospital Corporation Compositions and methods for modulating hemoglobin gene family expression
EP2850189B1 (en) 2012-05-16 2018-11-07 Translate Bio MA, Inc. Compositions and methods for modulating gene expression
US10058623B2 (en) 2012-05-16 2018-08-28 Translate Bio Ma, Inc. Compositions and methods for modulating UTRN expression
US10655128B2 (en) 2012-05-16 2020-05-19 Translate Bio Ma, Inc. Compositions and methods for modulating MECP2 expression
US9163235B2 (en) 2012-06-21 2015-10-20 MiRagen Therapeutics, Inc. Inhibitors of the miR-15 family of micro-RNAs
EP2863956A4 (en) * 2012-06-21 2016-01-20 Miragen Therapeutics Inc Inhibitors of the mir-15 family of micro-rnas
US9803202B2 (en) 2012-06-21 2017-10-31 MiRagen Therapeutics, Inc. Oligonucleotide-based inhibitors comprising locked nucleic acid motif
US10337005B2 (en) 2012-06-21 2019-07-02 MiRagen Therapeutics, Inc. Oligonucleotide-based inhibitors comprising locked nucleic acid motif
US9388408B2 (en) 2012-06-21 2016-07-12 MiRagen Therapeutics, Inc. Oligonucleotide-based inhibitors comprising locked nucleic acid motif
US10844375B2 (en) 2012-09-14 2020-11-24 Translate Bio Ma, Inc. Multimeric oligonucleotide compounds having non-nucleotide based cleavable linkers
US9790494B2 (en) 2012-09-14 2017-10-17 Translate Bio Ma, Inc. Multimeric oligonucleotide compounds having non-nucleotide based cleavable linkers
US9359609B2 (en) 2012-10-09 2016-06-07 Regulus Therapeutics Inc. Methods for treatment of alport syndrome
US9970011B2 (en) 2012-10-09 2018-05-15 Regulus Therapeutics Inc. Methods for treatment of alport syndrome
US9012423B2 (en) 2012-10-09 2015-04-21 Regulus Therapeutics Inc. Methods for treatment of alport syndrome
US9688986B2 (en) 2012-10-09 2017-06-27 Regulus Therapeutis Inc. Methods for treatment of alport syndrome
US9267139B2 (en) 2012-10-11 2016-02-23 Georgia Regents Research Institute, Inc. Compositions and methods for treating musculoskeletal diseases and disorders
US11155816B2 (en) 2012-11-15 2021-10-26 Roche Innovation Center Copenhagen A/S Oligonucleotide conjugates
EP3406718A1 (en) 2012-11-15 2018-11-28 Roche Innovation Center Copenhagen A/S Oligonucleotide conjugates
WO2014076195A1 (en) 2012-11-15 2014-05-22 Santaris Pharma A/S Oligonucleotide conjugates
US10077443B2 (en) 2012-11-15 2018-09-18 Roche Innovation Center Copenhagen A/S Oligonucleotide conjugates
WO2014118272A1 (en) 2013-01-30 2014-08-07 Santaris Pharma A/S Antimir-122 oligonucleotide carbohydrate conjugates
WO2014118267A1 (en) 2013-01-30 2014-08-07 Santaris Pharma A/S Lna oligonucleotide carbohydrate conjugates
WO2014135653A1 (en) * 2013-03-06 2014-09-12 Royal College Of Surgeons In Ireland Diagnosis and treatment of metabolic disorders
US9157083B2 (en) 2013-05-01 2015-10-13 Regulus Therapeutics Inc. MicroRNA compounds and methods for modulating miR-122
US10150967B2 (en) 2013-05-01 2018-12-11 Regulus Therapeutics Inc. MicroRNA compounds and methods for modulating miR-122
EA034731B1 (en) * 2013-05-01 2020-03-13 Регьюлэс Терапьютикс Инк. MICRORNA COMPOUNDS AND METHODS FOR MODULATING miR-122 ACTIVITY
US9506030B2 (en) 2013-05-01 2016-11-29 Regulus Therapeutics Inc. Compounds and methods for enhanced cellular uptake
US10240151B2 (en) 2013-05-01 2019-03-26 Regulus Therapeutics Inc. Compounds and methods for enhanced cellular uptake
US9574194B2 (en) 2013-05-01 2017-02-21 Regulus Therapeutics Inc. MicroRNA compounds and methods for modulating miR-122
WO2014179446A3 (en) * 2013-05-01 2015-02-19 Regulus Therapeutics Inc. Microrna compounds and methods for modulating mir-122
US10941400B2 (en) 2013-05-01 2021-03-09 Regulus Therapeutics Inc. Compounds and methods for enhanced cellular uptake
CN105378080A (en) * 2013-05-01 2016-03-02 莱古路斯治疗法股份有限公司 MicroRNA compounds and methods for modulating MIR-122
US9309513B2 (en) 2013-05-01 2016-04-12 Regulus Therapeutics Inc. MicroRNA compounds and methods for modulating miR-122
AU2014259954B2 (en) * 2013-05-01 2019-11-07 Regulus Therapeutics Inc. MicroRNA compounds and methods for modulating miR-122
CN103290010A (en) * 2013-05-16 2013-09-11 南京市妇幼保健院 Parental serum/plasma miRNA marker mir-375 relevant to fetus congenital heart disease and application thereof
WO2014201314A1 (en) * 2013-06-14 2014-12-18 Joslin Diabetes Center, Inc. Microrna and uses in brown fat differentiation
US9976140B2 (en) * 2013-06-14 2018-05-22 Joslin Diabetes Center, Inc. Microrna and uses in brown fat differentiation
US20160138017A1 (en) * 2013-06-14 2016-05-19 Joslin Diabetes Center, Inc. Microrna and uses in brown fat differentiation
EP3013958B1 (en) * 2013-06-25 2019-12-25 The United States of America, as represented by the Secretary, Department of Health and Human Services Glucan-encapsulated sirna for treating type 2 diabetes mellitus
US10077446B2 (en) 2013-06-25 2018-09-18 The United States Of America, As Represented By The Secretary, Dept. Of Health And Human Services Glucan-encapsulated siRNA for treating type 2 diabetes mellitus
US9994846B2 (en) 2013-10-25 2018-06-12 Regulus Therapeutics Inc. MicroRNA compounds and methods for modulating miR-21 activity
US9752144B2 (en) * 2014-08-04 2017-09-05 MiRagen Therapeutics, Inc. Inhibitors of MYH7B and uses thereof
US10144930B2 (en) 2014-08-04 2018-12-04 MiRagen Therapeutics, Inc. Inhibitors of MYH7B and uses thereof
US20160032286A1 (en) * 2014-08-04 2016-02-04 MiRagen Therapeutics, Inc. Inhibitors of myh7b and uses thereof
CN108064175A (en) * 2014-08-04 2018-05-22 米拉根医疗股份有限公司 Inhibitor of MYH7B and application thereof
US9487783B2 (en) 2014-08-07 2016-11-08 Regulus Therapeutics Inc. Targeting microRNAs for metabolic disorders
US10138484B2 (en) 2014-08-07 2018-11-27 Regulus Therapeutics Inc. Targeting microRNAs for metabolic disorders
US9862950B2 (en) 2014-08-07 2018-01-09 Regulus Therapeutics Inc. Targeting microRNAs for metabolic disorders
US10858650B2 (en) 2014-10-30 2020-12-08 The General Hospital Corporation Methods for modulating ATRX-dependent gene repression
US20190175660A1 (en) * 2014-11-05 2019-06-13 Emory University Compositions of Ascorbic Acid and Bone Morphogenetic Protein 4 (BMP-4) for Cell Growth and Uses Related Thereto
EP3234141A4 (en) * 2014-12-18 2018-06-20 Alnylam Pharmaceuticals, Inc. Reversir tm compounds
WO2016100716A1 (en) 2014-12-18 2016-06-23 Vasant Jadhav Reversirtm compounds
US9885042B2 (en) 2015-01-20 2018-02-06 MiRagen Therapeutics, Inc. miR-92 inhibitors and uses thereof
US10280422B2 (en) 2015-01-20 2019-05-07 MiRagen Therapeutics, Inc. MiR-92 inhibitors and uses thereof
US10758558B2 (en) 2015-02-13 2020-09-01 Translate Bio Ma, Inc. Hybrid oligonucleotides and uses thereof
US11225663B2 (en) 2015-02-25 2022-01-18 Washington University Methods to detect motor neuron disease comprising micro-RNAs
US10487324B2 (en) 2015-02-25 2019-11-26 Washington University Methods to detect motor neuron disease comprising micro-RNAs
US10900036B2 (en) 2015-03-17 2021-01-26 The General Hospital Corporation RNA interactome of polycomb repressive complex 1 (PRC1)
US9771585B2 (en) 2015-06-05 2017-09-26 MiRagen Therapeutics, Inc. miR-155 inhibitors for treating cutaneous T cell lymphoma (CTCL)
EP3303589A4 (en) * 2015-06-05 2019-01-02 Miragen Therapeutics, Inc. Mir-155 inhibitors for treating amyotrophic lateral sclerosis (als)
US9994852B2 (en) 2015-06-05 2018-06-12 MiRagen Therapeutics, Inc. Oligonucleotide compositions and uses thereof
US10316318B2 (en) 2015-06-05 2019-06-11 MiRagen Therapeutics, Inc. Oligonucleotide compositions and uses thereof
US11591594B2 (en) 2015-08-24 2023-02-28 Roche Innovation Center Copenhagen A/S LNA-G process
WO2017032726A1 (en) 2015-08-24 2017-03-02 Roche Innovation Center Copenhagen A/S Lna-g process
US11555050B2 (en) 2015-10-02 2023-01-17 Roche Innovation Center Copenhagen A/S Oligonucleotide conjugation process
US10577388B2 (en) 2015-10-02 2020-03-03 Roche Innovation Center Copenhagen A/S Oligonucleotide conjugation process
WO2017067970A1 (en) 2015-10-22 2017-04-27 Roche Innovation Center Copenhagen A/S In vitro toxicity screening assay
US10955407B2 (en) 2015-10-22 2021-03-23 Roche Innovation Center Copenhagen A/S In vitro toxicity screening assay
US11267843B2 (en) 2016-03-18 2022-03-08 Roche Innovation Center Copenhagen A/S Stereodefining L-monomers
WO2017157672A1 (en) 2016-03-18 2017-09-21 Roche Innovation Center Copenhagen A/S Acyl-protected l-lna-guanosine monomers
US11261209B2 (en) 2016-05-12 2022-03-01 Roche Innovation Center Copenhagen A/S Enhanced coupling of stereodefined oxazaphospholidine phosphoramidite monomers to nucleoside or oligonucleotide
WO2017216340A1 (en) 2016-06-17 2017-12-21 F. Hoffmann-La Roche Ag In vitro nephrotoxicity screening assay
WO2017216325A1 (en) 2016-06-17 2017-12-21 F. Hoffmann-La Roche Ag In vitro nephrotoxicity screening assay
US11105794B2 (en) 2016-06-17 2021-08-31 Hoffmann-La Roche Inc. In vitro nephrotoxicity screening assay
US11479818B2 (en) 2016-06-17 2022-10-25 Hoffmann-La Roche Inc. In vitro nephrotoxicity screening assay
US11253601B2 (en) 2016-07-11 2022-02-22 Translate Bio Ma, Inc. Nucleic acid conjugates and uses thereof
CN110036019A (en) * 2016-12-05 2019-07-19 莱古路斯治疗法股份有限公司 For treating the oligonucleotide of the modification of polycystic kidney disease
AU2017370560B2 (en) * 2016-12-05 2021-11-18 Regulus Therapeutics Inc. Modified oligonucleotides for treatment of polycystic kidney disease
AU2017370560C1 (en) * 2016-12-05 2022-08-11 Regulus Therapeutics Inc. Modified oligonucleotides for treatment of polycystic kidney disease
WO2018106566A1 (en) * 2016-12-05 2018-06-14 Regulus Therapeutics Inc. Modified oligonucleotides for treatment of polycystic kidney disease
US11542294B2 (en) 2017-03-29 2023-01-03 Roche Innovation Center Copenhagen A/S Rapid unylinker cleavage
WO2018177825A1 (en) 2017-03-29 2018-10-04 Roche Innovation Center Copenhagen A/S Orthogonal protecting groups for the preparation of stereodefined phosphorothioate oligonucleotides
US11591362B2 (en) 2017-03-29 2023-02-28 Roche Innovation Center Copenhagen A/S Orthogonal protecting groups for the preparation of stereodefined phosphorothioate oligonucleotides
WO2018177881A1 (en) 2017-03-29 2018-10-04 Roche Innovation Center Copenhagen A/S Unylinker rapid cleavage
WO2019002237A1 (en) 2017-06-28 2019-01-03 Roche Innovation Center Copenhagen A/S Multiple coupling & oxidation method
US11814407B2 (en) 2017-06-28 2023-11-14 Roche Innovation Center Copenhagen A/S Multiple coupling and oxidation method
US12018260B2 (en) 2017-08-17 2024-06-25 Alnylam Pharmaceuticals, Inc. Tunable Reversir™ compounds
WO2019073018A1 (en) 2017-10-13 2019-04-18 Roche Innovation Center Copenhagen A/S Methods for identifying improved stereodefined phosphorothioate oligonucleotide variants of antisense oligonucleotides utilising sub-libraries of partially stereodefined oligonucleotides
US11214536B2 (en) 2017-11-21 2022-01-04 Inspirna, Inc. Polymorphs and uses thereof
WO2019122279A1 (en) 2017-12-22 2019-06-27 Roche Innovation Center Copenhagen A/S Oligonucleotides comprising a phosphorodithioate internucleoside linkage
EP4092117A1 (en) 2017-12-22 2022-11-23 Roche Innovation Center Copenhagen A/S Gapmer oligonucleotides comprising a phosphorodithioate internucleoside linkage
WO2019122282A1 (en) 2017-12-22 2019-06-27 Roche Innovation Center Copenhagen A/S Gapmer oligonucleotides comprising a phosphorodithioate internucleoside linkage
WO2019122277A1 (en) 2017-12-22 2019-06-27 Roche Innovation Center Copenhagen A/S Novel thiophosphoramidites
US11597926B2 (en) 2017-12-22 2023-03-07 Roche Innovation Center Copenhagen A/S Thiophosphoramidites
US12005120B2 (en) 2018-05-08 2024-06-11 Regulus Therapeutics Inc. Galnac conjugated modified oligonucleotides as miR-122 inhibitor having HCV antiviral activity with reduced hyperbilirubinemia side-effect
WO2019219723A1 (en) 2018-05-18 2019-11-21 F. Hoffmann-La Roche Ag Pharmaceutical compositions for treatment of microrna related diseases
WO2020025563A1 (en) 2018-07-31 2020-02-06 Roche Innovation Center Copenhagen A/S Oligonucleotides comprising a phosphorotrithioate internucleoside linkage
WO2020025527A1 (en) 2018-07-31 2020-02-06 Roche Innovation Center Copenhagen A/S Oligonucleotides comprising a phosphorotrithioate internucleoside linkage
WO2020038968A1 (en) 2018-08-23 2020-02-27 Roche Innovation Center Copenhagen A/S Microrna-134 biomarker
EP3620519A1 (en) 2018-09-04 2020-03-11 F. Hoffmann-La Roche AG Use of isolated milk extracellular vesicles for delivering oligonucleotides orally
WO2020152303A1 (en) 2019-01-25 2020-07-30 F. Hoffmann-La Roche Ag Lipid vesicle for oral drug delivery
WO2020169695A1 (en) 2019-02-20 2020-08-27 Roche Innovation Center Copenhagen A/S Phosphonoacetate gapmer oligonucleotides
WO2020169696A1 (en) 2019-02-20 2020-08-27 Roche Innovation Center Copenhagen A/S Novel phosphoramidites
EP3963063A4 (en) * 2019-04-30 2023-09-27 The Trustees of The University of Pennsylvania Compositions useful for treatment of pompe disease
US11459292B2 (en) 2019-12-13 2022-10-04 Inspirna, Inc. Metal salts and uses thereof
US11878956B2 (en) 2019-12-13 2024-01-23 Inspirna, Inc. Metal salts and uses thereof
US11174220B2 (en) 2019-12-13 2021-11-16 Inspirna, Inc. Metal salts and uses thereof
WO2021226663A1 (en) * 2020-05-11 2021-11-18 The Florey Institute Of Neuroscience And Mental Health Compositions and methods for treating disorders associated with loss-of-function mutations in syngap1
WO2022245981A3 (en) * 2021-05-18 2022-12-29 Cornell University Use of microrna mimics to inhibit or treat liver disease
WO2023067038A1 (en) 2021-10-22 2023-04-27 Roche Innovation Center Copenhagen A/S Process for oligonucleotide purification
WO2024033446A1 (en) * 2022-08-09 2024-02-15 Geg Tech Transient expression system for rna, for gene editing
WO2024033441A1 (en) * 2022-08-09 2024-02-15 Geg Tech Transient expression system for rna
WO2024033444A1 (en) * 2022-08-09 2024-02-15 Geg Tech Transient expression system for rna, for cosmetic uses
WO2024033448A1 (en) * 2022-08-09 2024-02-15 Geg Tech Transient expression system for rna, for vaccination

Also Published As

Publication number Publication date
PL2623598T3 (en) 2018-12-31
US20100298410A1 (en) 2010-11-25
WO2009043354A3 (en) 2009-08-20
US8906871B2 (en) 2014-12-09
AU2008306327B2 (en) 2014-05-15
IL204254A (en) 2016-03-31
US20090143326A1 (en) 2009-06-04
US10450564B2 (en) 2019-10-22
EP2623598A1 (en) 2013-08-07
MY156951A (en) 2016-04-15
EP3492594A1 (en) 2019-06-05
AU2008306327A1 (en) 2009-04-09
JP6231029B2 (en) 2017-11-15
US20180201928A1 (en) 2018-07-19
JP2015130862A (en) 2015-07-23
EP2203559A2 (en) 2010-07-07
US8440637B2 (en) 2013-05-14
EP2205737B1 (en) 2013-02-13
EP2623599B1 (en) 2019-01-02
KR101889518B1 (en) 2018-08-17
WO2009043353A3 (en) 2009-08-20
EA019939B1 (en) 2014-07-30
US9790493B2 (en) 2017-10-17
NZ583677A (en) 2012-06-29
ES2689508T3 (en) 2018-11-14
ES2463665T3 (en) 2014-05-28
JP2010539959A (en) 2010-12-24
US20150299699A1 (en) 2015-10-22
EP2623598B1 (en) 2018-08-01
CA2701547C (en) 2020-03-10
US20100280099A1 (en) 2010-11-04
CA2701547A1 (en) 2009-04-09
ES2406686T3 (en) 2013-06-07
EA201070421A1 (en) 2010-10-29
MX2010003299A (en) 2010-04-14
CN101821391A (en) 2010-09-01
EP2205737A2 (en) 2010-07-14
KR20100100774A (en) 2010-09-15
DK2623599T3 (en) 2019-04-08
JP6035010B2 (en) 2016-11-30
EP2203559B1 (en) 2014-02-26
ZA201002040B (en) 2012-01-25
US8288356B2 (en) 2012-10-16
EP2623599A1 (en) 2013-08-07
CN101821391B (en) 2016-04-27
IL244498A0 (en) 2016-04-21
WO2009043354A2 (en) 2009-04-09
DK2205737T3 (en) 2013-05-21

Similar Documents

Publication Publication Date Title
US10450564B2 (en) Micromirs
EP3099797B1 (en) Poly oligomer compound with biocleavable conjugates
US8404659B2 (en) Pharmaceutical compositions for treatment of MicroRNA related diseases
CA2648132C (en) Pharmaceutical composition comprising anti-mirna antisense oligonucleotides
EP2666859A2 (en) Pharmaceutical composition comprising anti-miRNA antisense oligonucleotides
AU2013254923A1 (en) Pharmaceutical compositions comprising anti-miRNA antisense oligonucleotide
AU2013273821B2 (en) Micromirs
AU2007234192B2 (en) Pharmaceutical compositions comprising anti-miRNA antisense oligonucleotides
BRPI0817485A2 (en) OLIGOMERS, AS WELL AS AN IN VITRO METHOD TO REDUCE THE EFFECTIVE AMOUNT OF A MICRNA TARGET IN A CELL

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200880110388.3

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 08801378

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 204254

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2008306327

Country of ref document: AU

Ref document number: 583677

Country of ref document: NZ

Ref document number: 12010500500

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 999/KOLNP/2010

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: MX/A/2010/003299

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2008306327

Country of ref document: AU

Date of ref document: 20081003

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2701547

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2010040540

Country of ref document: EG

WWE Wipo information: entry into national phase

Ref document number: 2010527323

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: PI 2010001033

Country of ref document: MY

WWE Wipo information: entry into national phase

Ref document number: 2008801378

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 201070421

Country of ref document: EA

ENP Entry into the national phase

Ref document number: 20107009969

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: A201005202

Country of ref document: UA

WWE Wipo information: entry into national phase

Ref document number: 12681587

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 244498

Country of ref document: IL

ENP Entry into the national phase

Ref document number: PI0817485

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20100401