US20080209581A1 - Induction of exon skipping in eukaryotic cells - Google Patents

Induction of exon skipping in eukaryotic cells Download PDF

Info

Publication number
US20080209581A1
US20080209581A1 US11/982,285 US98228507A US2008209581A1 US 20080209581 A1 US20080209581 A1 US 20080209581A1 US 98228507 A US98228507 A US 98228507A US 2008209581 A1 US2008209581 A1 US 2008209581A1
Authority
US
United States
Prior art keywords
exon
mrna
protein
cell
nucleic acid
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/982,285
Inventor
Garrit-Jan Boudewijn van Ommen
T. van Deutekom Judith Christina
Johannes Theodorus den Dunnen
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Leids Universitair Medisch Centrum LUMC
Original Assignee
Leids Universitair Medisch Centrum LUMC
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=8172043&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20080209581(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Leids Universitair Medisch Centrum LUMC filed Critical Leids Universitair Medisch Centrum LUMC
Priority to US11/982,285 priority Critical patent/US20080209581A1/en
Assigned to ACADEMISCH ZIEKENHUIS LEIDEN reassignment ACADEMISCH ZIEKENHUIS LEIDEN ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DEN DUNNEN, JOHANNES T., VAN DEUTEKOM, JUDITH C.T., VAN OMMEN, GARRIT-JAN B.
Publication of US20080209581A1 publication Critical patent/US20080209581A1/en
Assigned to ACADEMISCH ZIEKENHUIS LEIDEN H.O.D.N. LUMC reassignment ACADEMISCH ZIEKENHUIS LEIDEN H.O.D.N. LUMC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: VAN DEUTEKOM, JUDITH C., VAN OMMEN, GARRIT-JAN B., DEN DUNNEN, JOHANNES T.
Priority to US14/331,934 priority patent/US20140350076A1/en
Priority to US14/712,753 priority patent/US20150322434A1/en
Priority to US14/839,200 priority patent/US20150361424A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/04Drugs for skeletal disorders for non-specific disorders of the connective tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3212'-O-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/34Spatial arrangement of the modifications
    • C12N2310/346Spatial arrangement of the modifications having a combination of backbone and sugar modifications
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/33Alteration of splicing
    • 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
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14142Use of virus, viral particle or viral elements as a vector virus or viral particle as vehicle, e.g. encapsulating small organic molecule

Definitions

  • the invention relates to the fields of biotechnology and gene therapy.
  • Exon-skipping results in mature mRNA that does not contain the skipped exon and thus, when the exon codes for amino acids, can lead to the expression of an altered product.
  • Technology for exon-skipping is currently directed toward the use of so-called “Anti-sense Oligonucleotides” (AONs).
  • DMD Duchenne muscular dystrophy
  • the mdx mouse which carries a nonsense mutation in exon 23 of the dystrophin gene, has been used as an animal model of Duchenne muscular dystrophy.
  • mdx mutation which should preclude the synthesis of a functional dystrophin protein
  • rare, naturally occurring dystrophin-positive fibers have been observed in mdx muscle tissue. These dystrophin-positive fibers are thought to have arisen from an apparently naturally occurring exon-skipping mechanism, either due to somatic mutations or through alternative splicing.
  • AONs directed to, respectively, the 3′ and 5′ splice sites of introns 22 and 23 in dystrophin pre-mRNA have been shown to interfere with factors normally involved in removal of intron 23 so that exon 23 was also removed from the mRNA (Wilton, 1999).
  • Dunckley et al. (1998) showed that exon skipping using AONs directed to 3′ and 5′ splice sites can have unexpected results. They observed skipping of not only exon 23 but also of exons 24-29, thus resulting in an mRNA containing an exon 22-exon 30 junction.
  • the underlying mechanism for the appearance of the unexpected 22-30 splicing variant is not known. It could be due to the fact that splice sites contain consensus sequences leading to promiscuous hybridization of the oligos used to direct the exon skipping. Hybridization of the oligos to other splice sites than the sites of the exon to be skipped of course could easily interfere with the accuracy of the splicing process. On the other hand, the accuracy could be lacking due to the fact that two oligos (for the 5′ and the 3′ splice site) need to be used. Pre-mRNA containing one but not the other oligo could be prone to unexpected splicing variants.
  • a method for directing splicing of a pre-mRNA in a system capable of performing a splicing operation comprising contacting the pre-mRNA in the system with an agent capable of specifically inhibiting an exon inclusion signal of at least one exon in the pre-mRNA, the method further comprising allowing splicing of the pre-mRNA.
  • Interfering with an exon inclusion signal has the advantage that such elements are located within the exon.
  • EIS exon inclusion signal
  • the method further comprises allowing translation of mRNA produced from splicing of the pre-mRNA.
  • the mRNA encodes a functional protein.
  • the protein comprises two or more domains, wherein at least one of the domains is encoded by the mRNA as a result of skipping of at least part of an exon in the pre-mRNA.
  • Exon skipping will typically, though not necessarily, be of relevance for proteins in the wild-type configuration, having at least two functional domains that each performs a function, wherein the domains are generated from distinct parts of the primary amino acid sequence.
  • Examples are, for instance, transcription factors.
  • these factors comprise a DNA binding domain and a domain that interacts with other proteins in the cell.
  • Skipping of an exon that encodes a part of the primary amino acid sequence that lies between these two domains can lead to a shorter protein that comprises the same function, at least in part.
  • detrimental mutations in this intermediary region for instance, frame-shift or stop mutations
  • the system comprises a cell.
  • the cell is cultured in vitro or in the organism in vivo. Typically, though not necessarily, the organism comprises a human or a mouse.
  • a method for at least in part decreasing the production of an aberrant protein in a cell comprising pre-mRNA comprising exons coding for the protein, the method comprising providing the cell with an agent capable of specifically inhibiting an exon inclusion signal of at least one of the exons, the method further comprising allowing translation of mRNA produced from splicing of the pre-mRNA.
  • the agent comprises a nucleic acid or a functional equivalent thereof.
  • the nucleic acid is in single-stranded form.
  • Peptide nucleic acid and other molecules comprising the same nucleic acid binding characteristics in kind, but not necessarily in amount, are suitable equivalents.
  • Nucleic acid or an equivalent may comprise modifications to provide additional functionality. For instance, 2′-O-methyl oligoribonucleotides can be used. These ribonucleotides are more resistant to RNAse action than conventional oligonucleotides.
  • the exon inclusion signal is interfered with by an antisense nucleic acid directed to an exon recognition sequence (ERS).
  • ERS exon recognition sequence
  • These sequences are relatively purine-rich and can be distinguished by scrutinizing the sequence information of the exon to be skipped (Tanaka et al., 1994 , Mol. Cell. Biol. 14, p. 1347-1354).
  • Exon recognition sequences are thought to aid inclusion into mRNA of so-called weak exons (Achsel et al., 1996 , J. Biochem. 120, p. 53-60). These weak exons comprise, for instance, 5′ and or 3′ splice sites that are less efficiently recognized by the splicing machinery.
  • exon skipping can also be induced in so-called strong exons, i.e., exons which are normally efficiently recognized by the splicing machinery of the cell. From any given sequence, it is (almost) always possible to predict whether the sequence comprises putative exons and to determine whether these exons are strong or weak.
  • strong exons i.e., exons which are normally efficiently recognized by the splicing machinery of the cell. From any given sequence, it is (almost) always possible to predict whether the sequence comprises putative exons and to determine whether these exons are strong or weak.
  • Several algorithms for determining the strength of an exon exist. A useful algorithm can be found on the NetGene2 splice site prediction server (Brunak, et al., 1991 , J. Mol. Biol. 220, p. 49-65).
  • Exon skipping by a means of the invention can be induced in (almost) every exon, independent of whether the exon is a weak exon or a strong exon and also independent of whether the exon comprises an ERS.
  • an exon that is targeted for skipping is a strong exon.
  • an exon targeted for skipping does not comprise an ERS.
  • a method described herein is used to at least in part decrease the production of an aberrant protein.
  • Such proteins can, for instance, be oncoproteins or viral proteins.
  • an oncoprotein not only the presence of an oncoprotein but also its relative level of expression has been associated with the phenotype of the tumor cell.
  • viral proteins not only the presence of viral proteins but also the amount of viral protein in a cell determines the virulence of a particular virus.
  • the timing of expression in the life cycle and the balance in the amount of certain viral proteins in a cell determines whether viruses are efficiently or inefficiently produced.
  • it is possible to lower the amount of aberrant protein in a cell such that, for instance, a tumor cell becomes less tumorigenic (metastatic) and/or a virus-infected cell produces less virus.
  • a method described herein is used to modify the aberrant protein into a functional protein.
  • the functional protein is capable of performing a function of a protein normally present in a cell but absent in the cells to be treated. Very often, even partial restoration of function results in significantly improved performance of the cell thus treated. Due to the better performance, such cells can also have a selective advantage over unmodified cells, thus aiding the efficacy of the treatment.
  • This aspect is particularly suited for the restoration of expression of defective genes. This is achieved by causing the specific skipping of targeted exons, thus bypassing or correcting deleterious mutations (typically stop-mutations or frame-shifting point mutations, single- or multi-exon deletions or insertions leading to translation termination).
  • deleterious mutations typically stop-mutations or frame-shifting point mutations, single- or multi-exon deletions or insertions leading to translation termination).
  • this novel form of splice-modulation gene therapy requires the administration of much smaller therapeutic reagents, typically, but not limited to, 14-40 nucleotides.
  • molecules of 14-25 nucleotides are used since these molecules are easier to produce and enter the cell more effectively.
  • the methods of the invention allow much more flexibility in the subsequent design of effective and safe administration systems.
  • An important additional advantage of this aspect is that it restores (at least some of) the activity of the endogenous gene, which still possesses most or all of its gene-regulatory circuitry, thus ensuring proper expression levels and the synthesis of tissue-specific isoforms.
  • This aspect can, in principle, be applied to any genetic disease or genetic predisposition to disease in which targeted skipping of specific exons would restore the translational reading frame when this has been disrupted by the original mutation, provided that translation of an internally slightly shorter protein is still fully or partly functional.
  • Preferred embodiments for which this application can be of therapeutic value are: predisposition to second hit mutations in tumor suppressor genes, e.g., those involved in breast cancer, colon cancer, tuberous sclerosis, neurofibromatosis etc., where (partial) restoration of activity would preclude the manifestation of nullosomy by second hit mutations and thus would protect against tumorigenesis.
  • Another preferred embodiment involves the (partial) restoration of defective gene products which have a direct disease causing effect, e.g., hemophilia A (clotting factor VIII deficiency), some forms of congenital hypothyroidism (due to thyroglobulin synthesis deficiency) and Duchenne muscular dystrophy (DMD), in which frame-shifting deletions, duplications and stop mutations in the X-linked dystrophin gene cause severe, progressive muscle degradation.
  • DMD is typically lethal in late adolescence or early adulthood, while non-frame-shifting deletions or duplications in the same gene cause the much milder Becker muscular dystrophy (BMD), compatible with a life expectancy between 35-40 years to normal.
  • the invention enables exon skipping to extend an existing deletion (or alter the mRNA product of an existing duplication) by as many adjacent exons as required to restore the reading frame and generate an internally slightly shortened, but still functional, protein. Based on the much milder clinical symptoms of BMD patients with the equivalent of this induced deletion, the disease in the DMD patients would have a much milder course after AON-therapy.
  • dystrophin gene can lead to a dysfunctional protein.
  • Table 1 comprises a non-limiting list of exons that can be skipped and lists for the mentioned exons some of the more frequently occurring dystrophin gene mutations that have been observed in humans and that can be treated with a method described herein. Skipping of the mentioned exon leads to a mutant dystrophin protein comprising at least the functionality of a Becker mutant.
  • a method described herein wherein the exon inclusion signal is present in exon numbers 2, 8, 19, 29, 43, 44, 45, 46, 50, 51, 52 or 53 of the human dystrophin gene.
  • the occurrence of certain deletion/insertion variations is more frequent than others.
  • By inducing skipping of exon 51 approximately 15% of DMD-deletion containing patients can be treated with a means or method described herein. Such treatment will result in the patient having at least some dystrophin-positive fibers.
  • the exon exclusion signal is present in exon 46 or exon 51.
  • the agent comprises a nucleic acid sequence according to hAON#4, hAON#6, hAON#8, hAON#9, hAON#11 and/or one or more of hAON#21-30 or a functional part, derivative and/or analogue of the hAON.
  • a functional part, derivative and/or analogue of the hAON comprises the same exon skipping activity in kind, but not necessarily in amount, in a method described herein.
  • a preferred but non-limiting example of such a case in the DMD deletion database is a 46-50 deletion. Patients comprising a 46-50 deletion do not produce functional dystrophin. However, an at least partially functional dystrophin can be generated by inducing skipping of both exon 45 and exon 51. Another preferred but non-limiting example is patients comprising a duplication of exon 2.
  • the invention therefore provides a method described herein further comprising providing the cell with another agent capable of inhibiting an exon inclusion signal in another exon of the pre-mRNA.
  • another agent capable of inhibiting an exon inclusion signal in another exon of the pre-mRNA.
  • a method for selecting the suitable agents for splice-therapy and their validation as specific exon-skipping agents in pilot experiments comprising providing a cell having a pre-mRNA containing the exon with the agent, culturing the cell to allow the formation of an mRNA from the pre-mRNA and determining whether the exon is absent the mRNA.
  • the agent comprises a nucleic acid or a functional equivalent thereof, the nucleic acid comprising complementarity to a part of the exon. Agents capable of inducing specific exon skipping can be identified with a method described herein.
  • RNA molecule it is possible to include a prescreen for agents by first identifying whether the agent is capable of binding with a relatively high affinity to an exon containing nucleic acid, preferably RNA.
  • a method for determining whether an agent is capable of specifically inhibiting an exon inclusion signal of an exon is provided, further comprising first determining in vitro the relative binding affinity of the nucleic acid or functional equivalent thereof to an RNA molecule comprising the exon.
  • an agent is provided that is obtainable by a method described herein.
  • the agent comprises a nucleic acid or a functional equivalent thereof.
  • the agent when used to induce exon skipping in a cell, is capable of at least in part reducing the amount of aberrant protein in the cell. More preferably, the exon skipping results in an mRNA encoding a protein that is capable of performing a function in the cell.
  • the pre-mRNA is derived from a dystrophin gene.
  • the functional protein comprises a mutant or normal dystrophin protein. In certain embodiments, the mutant dystrophin protein comprises at least the functionality of a dystrophin protein in a Becker patient.
  • the agent comprises the nucleic acid sequence of hAON#4, hAON#6, hAON#8, hAON#9, hAON#11 and/or one or more of hAON#21-30 or a functional part, derivative and/or analogue of the hAON.
  • a functional part, derivative and/or analogue of the hAON comprises the same exon skipping activity in kind, but not necessarily in amount, in a method described herein.
  • nucleic acid delivery methods have been widely developed. The artisan is well capable of determining whether a method of delivery is suitable for performing the invention.
  • the method includes the packaging of an agent of the invention into liposomes, the liposomes being provided to cells comprising a target pre-mRNA. Liposomes are particularly suited for delivery of nucleic acid to cells.
  • Antisense molecules capable of inducing exon skipping can be produced in a cell upon delivery of nucleic acid containing a transcription unit to produce antisense RNA.
  • suitable transcription units are small nuclear RNA (SNRP) or tRNA transcription units.
  • the invention therefore, further provides a nucleic acid delivery vehicle comprising a nucleic acid or functional equivalent thereof of the invention capable of inhibiting an exon inclusion signal.
  • the delivery vehicle is capable of expressing the nucleic acid of the invention.
  • single-stranded viruses are used as a vehicle, it is entirely within the scope of the invention when such a virus comprises only the antisense sequence of an agent of the invention.
  • AONs of the invention are encoded by small nuclear RNA or tRNA transcription units on viral nucleic encapsulated by the virus as vehicle.
  • a preferred single-stranded virus is adeno-associated virus.
  • a nucleic acid or a nucleic acid delivery vehicle of the invention for the preparation of a medicament.
  • the medicament is used for the treatment of an inherited disease. More preferably, the medicament is used for the treatment of Duchenne Muscular Dystrophy.
  • FIG. 1 Deletion of exon 45 is one of the most frequent DMD-mutations. Due to this deletion, exon 44 is spliced to exon 46, the translational reading frame is interrupted, and a stop codon is created in exon 46 leading to a dystrophin deficiency. Our aim is to artificially induce the skipping of an additional exon, exon 46, in order to reestablish the reading frame and restore the synthesis of a slightly shorter, but largely functional, dystrophin protein as found in the much milder affected Becker muscular dystrophy patients affected by a deletion of both exons 45 and 46.
  • Exon 46 contains a purine-rich region that is hypothesized to have a potential role in the regulation of its splicing in the pre-mRNA.
  • a series of overlapping 2′O-methyl phosphorothioate antisense oligoribonucleotides (AONs) was designed directed at this purine-rich region in mouse dystrophin exon 46.
  • the AONs differ both in length and sequence. The chemical modifications render the AONs resistant to endonucleases and RNaseH inside the muscle cells.
  • the AONs contained a 5′ fluorescein group which allowed identification of AON-positive cells.
  • FIG. 3 To determine the binding affinity of the different AONs to the target exon 46 RNA, we performed gel mobility shift assays. In this figure, the five mAONs (mAON#4, 6, 8, 9, and 11) with highest affinity for the target RNA are shown. Upon binding of the AONs to the RNA, a complex is formed that exhibits a retarded gel mobility as can be determined by the band shift. The binding of the AONs to the target was sequence-specific. A random mAON, i.e. not specific for exon 46, did not generate a band shift.
  • FIGS. 4A and 4B The mouse- and human-specific AONs which showed the highest binding affinity in the gel mobility shift assays were transfected into mouse and human myotube cultures.
  • FIG. 4A RT-PCR analysis showed a truncated product, of which the size corresponded to exon 45 directly spliced to exon 47, in the mouse cell cultures upon transfection with the different mAONs#4, 6, 9, and 11. No exon 46 skipping was detected following transfection with a random AON.
  • FIG. 4B RT-PCR analysis in the human muscle cell cultures derived from one unaffected individual (C) and two unrelated DMD patients (P 1 and P 2 ) revealed truncated products upon transfection with hAON#4 and hAON#8. In the control, this product corresponded to exon 45 spliced to exon 47, while in the patients, the fragment size corresponded to exon 44 spliced to exon 47. No exon 46 skipping was detected in the non-transfected cell cultures or following transfection with a random HAON. Highest exon 46 skipping efficiencies were obtained with hAON#8.
  • FIG. 5 Sequence data from the RT-PCR products obtained from patient DL279.1 (corresponding to P 1 in FIG. 4 ), which confirmed the deletion of exon 45 in this patient (upper panel), and the additional skipping of exon 46 following transfection with hAON#8 (lower panel). The skipping of exon 46 was specific, and exon 44 was exactly spliced to exon 47, which reestablishes the translational reading frame.
  • FIG. 6 Immunohistochemical analysis of the muscle cell culture from patient DL279.1 upon transfection with hAON#8.
  • Cells were subject to two different dystrophin antibodies raised against different regions of the protein, located proximally (ManDys-1, ex. 31-32) and distally (Dys-2, ex. 77-79) from the targeted exon 46.
  • the lower panel shows the absence of a dystrophin protein in the myotubes, whereas the hAON#8-induced skipping of exon 46 clearly restored the synthesis of a dystrophin protein as detected by both antibodies (upper panel).
  • FIG. 7A RT-PCR analysis of RNA isolated from human control muscle cell cultures treated with hAON#23, #24, #27, #28, or #29. An additional aberrant splicing product was obtained in cells treated with hAON#28 and #29. Sequence analysis revealed the utilization of an in-frame cryptic splice site within exon 51 that is used at a low frequency upon AON treatment. The product generated included a partial exon 51, which also had a restored reading frame, thereby confirming further the therapeutic value.
  • FIG. 7B A truncated product, with a size corresponding to exon 50 spliced to exon 52, was detected in cells treated with hAON#23 and #28. Sequence analysis of these products confirmed the precise skipping of exon 51.
  • FIG. 8A Gel mobility shift assays were performed to determine the binding affinity of the different h29AON#'s for the exon 29 target RNA. When compared to non-hybridized RNA (none), h29AON#1, #2, #4, #6, #9, #10, and #11 generated complexes with lower gel mobilities, indicating their binding to the RNA. A random AON derived from dystrophin exon 19 did not generate a complex.
  • FIG. 8B RT-PCR analysis of RNA isolated from human control muscle cell cultures treated with h29AON#1, #2, #4, #6, #9, #10, or #11 revealed a truncated product of which the size corresponded to exon 28 spliced to exon 30.
  • exon 29 can specifically be skipped using AONs directed to sequences either within (h29AON#1, #2, #4, or #6) or outside (h29AON#9, #10, or #11) the hypothesized ERS in exon 29.
  • An additional aberrant splicing product was observed that resulted from skipping of both exon 28 and exon 29 (confirmed by sequence data not shown). Although this product was also present in non-treated cells, suggesting that this alternative skipping event may occur naturally, it was enhanced by the AON-treatment.
  • AON 19, derived from dystrophin exon 19 did not induce exon 29 skipping.
  • FIG. 8C The specific skipping of exon 29 was confirmed by sequence data from the truncated RT-PCR fragments. Shown here is the sequence obtained from the exon 29 skipping product in cells treated with h29AON#1.
  • FIG. 9A RT-PCR analysis of RNA isolated from mouse gastrocnemius muscles two days post-injection of 5, 10, or 20 ⁇ g of either mAON#4, #6, or #11. Truncated products, with a size corresponding to exon 45 spliced to exon 47, were detected in all treated muscles. The samples -RT, -RNA, AD-1, and AD-2 were analyzed as negative controls for the RT-PCR reactions.
  • FIG. 9B Sequence analysis of the truncated products generated by mAON#4 and #6 (and #11, not shown) confirmed the precise skipping of exon 46.
  • exon 45 is one of the most frequently deleted exons in DMD, we initially aimed at inducing the specific skipping of exon 46 ( FIG. 1 ). This would produce the shorter, largely functional dystrophin found in BMD patients carrying a deletion of exons 45 and 46.
  • the system was initially set up for modulation of dystrophin pre-mRNA splicing of the mouse dystrophin gene. We later aimed for the human dystrophin gene with the intention to restore the translational reading frame and dystrophin synthesis in muscle cells from DMD patients affected by a deletion of exon 45.
  • a series of mouse- and human-specific AONs was designed, directed at an internal part of exon 46 that contains a stretch of purine-rich sequences and is hypothesized to have a putative regulatory role in the splicing process of exon 46 ( FIG. 2 ).
  • mAONs and hAONs mouse- and human-specific AONs
  • RNA oligonucleotides are known to be resistant to endonucleases and RNaseH, and to bind to RNA with high affinity.
  • the sequences of those AONs that were eventually effective and applied in muscle cells in vitro are shown below.
  • the corresponding mouse and human-specific AONs are highly homologous but not completely identical.
  • mAON#2 5′ GCAATGTTATCTGCTT (SEQ ID NO:1)
  • mAON#3 5′ GTTATCTGCTTCTTCC
  • mAON#4 5′ CTGCTTCTTCCAGCC
  • mAON#5 5′ TCTGCTTCTTCCAGC
  • mAON#6 5′ GTTATCTGCTTCTTCCAGCC
  • mAON#7 5′ CTTTTAGCTGCTGCTC
  • mAON#8 5′ GTTGTTCTTTTAGCTGCTGC (SEQ ID NO:7)
  • mAON#9 5′ TTAGCTGCTGCTCAT (SEQ ID NO:8)
  • mAON#10 5′ TTTAGCTGCTGCTCATCTCC
  • hAON#4 5′
  • the efficacy of the AONs is determined by their binding affinity for the target sequence. Notwithstanding recent improvements in computer simulation programs for the prediction of RNA-folding, it is difficult to speculate which of the designed AONs would be capable of binding the target sequence with a relatively high affinity. Therefore, we performed gel mobility shift assays (according to protocols described by Bruice et al., 1997).
  • the exon 46 target RNA fragment was generated by in vitro T7-transcription from a PCR fragment (amplified from either murine or human muscle mRNA using a sense primer that contains the T7 promoter sequence) in the presence of 32P-CTP.
  • the binding affinity of the individual AONs (0.5 ⁇ mol) for the target transcript fragments was determined by hybridization at 37° C.
  • exon 46-specific AONs which showed the highest target binding affinity in gel mobility shift assays were selected for analysis of their efficacy in inducing the skipping in muscle cells in vitro.
  • a non-specific AON as a negative control for the specific skipping of exon 46.
  • the system was first set up in mouse muscle cells. We used both proliferating myoblasts and post-mitotic myotube cultures (expressing higher levels of dystrophin) derived from the mouse muscle cell line C2C12. For the subsequent experiments in human-derived muscle cell cultures, we used primary muscle cell cultures isolated from muscle biopsies from one unaffected individual and two unrelated DMD patients carrying a deletion of exon 45.
  • RNA was reverse transcribed using C. therm. polymerase (Roche) and an exon 48-specific reverse primer.
  • the cDNA was amplified by two rounds of PCR, including a nested amplification using primers in exons 44 and 47 (for the human system), or exons 45 and 47 (for the mouse system).
  • exons 44 and 47 for the human system
  • exons 45 and 47 for the mouse system.
  • truncated product of which the size corresponded to exon 45 directly spliced to exon 47 FIG. 4 .
  • Subsequent sequence analysis confirmed the specific skipping of exon 46 from these mouse dystrophin transcripts.
  • Exon 51 is an interesting target exon. The skipping of this exon is therapeutically applicable in patients carrying deletions spanning exon 50, exons 45-50, exons 48-50, exons 49-50, exon 52, and exons 52-63, which includes a total of 15% of patients from our Leiden database.
  • the RT-PCR and sequence analysis revealed that we were able to induce the specific skipping of exon 51 from the human dystrophin transcript.
  • hAON#21 5′ CCACAGGTTGTGTCACCAG (SEQ ID NO:16)
  • hAON#22 5′ TTTCCTTAGTAACCACAGGTT
  • hAON#23 5′ TGGCATTTCTAGTTTGG
  • hAON#24 5′ CCAGAGCAGGTACCTCCAACATC
  • hAON#25 5′ GGTAAGTTCTGTCCAAGCCC
  • hAON#26 5′ TCACCCTCTGTGATTTTAT (SEQ ID NO:21)
  • hAON#27 5′ CCCTCTGTGATTTT
  • SEQ ID NO:22 hAON#28: 5′ TCACCCACCATCACCCT
  • SEQ ID NO:23 hAON#29: 5′ TGATATCCTCAAGGTCACCC
  • hAON#30 5′ CTGCTTGATG
  • exon 46 or exon 51 restores the reading frame for a considerable number of different DMD mutations.
  • the range of mutations for which this strategy is applicable can be enlarged by the simultaneous skipping of more than one exon. For instance, in DMD patients with a deletion of exon 46 to exon 50, only the skipping of both the deletion-flanking exons 45 and 51 enables the reestablishment of the translational reading frame.
  • a mutation in exon 29 leads to the skipping of this exon in two Becker muscular dystrophy patients (Ginjaar at al., 2000, EJHG, vol. 8, p. 793-796).
  • the mutation is located in a purine-rich stretch that could be associated with ERS activity.
  • We designed a series of AONs (see below) directed to sequences both within (h29AON#1 to h29AON#6) and outside (h29AON#7 to h29AON#11) the hypothesized ERS.
  • Gel mobility shift assays were performed (as described) to identify those AONs with highest affinity for the target RNA ( FIG. 8 ).
  • h29AON#1, #2, #4, #6, #9, #10, and #11 were transfected into human control myotube cultures using the PEI transfection reagent.
  • RNA was isolated 24 hrs post-transfection, and cDNA was generated using an exon 31-specific reverse primer. PCR-amplification of the targeted region was performed using different primer combinations flanking exon 29.
  • This RT-PCR and subsequent sequence analysis revealed that we were able to induce the skipping of exon 29 from the human dystrophin transcript.
  • the AONs that facilitated this skipping were directed to sequences both within and outside the hypothesized ERS (h29AON#1, #2, #4, #6, #9, and #11).
  • h29AON#1 5′ TATCCTCTGAATGTCGCATC (SEQ ID NO:26)
  • h29AON#2 5′ GGTTATCCTCTGAATGTCGC (SEQ ID NO:27)
  • h29AON#3 5′ TCTGTTAGGGTCTGTGCC (SEQ ID NO:28)
  • h29AON#4 5′ CCATCTGTTAGGGTCTGTG (SEQ ID NO:29)
  • h29AON#5 5′ GTCTGTGCCAATATGCG (SEQ ID NO:30)
  • h29AON#6 5′ TCTGTGCCAATATGCGAATC (SEQ ID NO:31)
  • h29AON#7 5′ TGTCTCAAGTTCCTC
  • h29AON#8 5′ GAATTAAATGTCTCAAGTTC (SEQ ID NO:33)
  • h29AON#9 5′ TTAAATGTCTCAAGTTCC (SEQ ID NO:34)
  • mice dystrophin exon 46-specific AONs were tested in vivo by injecting them, linked to polyethylenimine (PEI), into the gastrocnemius muscles of control mice.
  • PEI polyethylenimine
  • mAON#4, #6, and #11 previously shown to be effective in mouse muscle cells in vitro, we were able to induce the skipping of exon 46 in muscle tissue in vivo as determined by both RT-PCR and sequence analysis ( FIG. 9 ).
  • the in vivo exon 46 skipping was dose-dependent with highest efficiencies (up to 10%) following injection of 20 ⁇ g per muscle per day for two subsequent days.

Abstract

Described is a method for at least in part decreasing the production of an aberrant protein in a cell, the cell comprising pre-mRNA comprising exons coding for the protein, by inducing so-called exon skipping in the cell. Exon-skipping results in mature mRNA that does not contain the skipped exon, which leads to an altered product of the exon codes for amino acids. Exon skipping is performed by providing a cell with an agent capable of specifically inhibiting an exon inclusion signal, for instance, an exon recognition sequence, of the exon. The exon inclusion signal can be interfered with by a nucleic acid comprising complementarity to a part of the exon. The nucleic acid, which is also herewith provided, can be used for the preparation of a medicament, for instance, for the treatment of an inherited disease.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a divisional of co-pending U.S. patent application Ser. No. 10/395,031, filed Mar. 21, 2003, now U.S. Pat. No. ______, which is a continuation of International Application PCT/NL01/00697, filed Sep. 21, 2001, designating the United States, published in English Mar. 28, 2002, as WO 02/024906 A1 and subsequently published with corrections Jan. 23, 2003, as WO 02/024906 C2, the contents of the entirety of each of which are hereby incorporated herein by this reference.
  • TECHNICAL FIELD
  • The invention relates to the fields of biotechnology and gene therapy.
  • BACKGROUND
  • Given the rapid advances of human genome research, professionals and the public expect that the near future will bring us, in addition to understanding of disease mechanisms and refined and reliable diagnostics, therapies for many devastating genetic diseases.
  • While it is hoped that for some (e.g., metabolic) diseases, the improved insights will bring easily administrable small-molecule therapies, it is likely that in most cases one or another form of gene therapy will ultimately be required, i.e., the correction, addition or replacement of the defective gene product.
  • In the past few years, research and development in this field have highlighted several technical difficulties which need to be overcome, e.g., related to the large size of many genes involved in genetic disease (limiting the choice of suitable systems to administer the therapeutic gene), the accessibility of the tissue in which the therapeutic gene should function (requiring the design of specific targeting techniques, either physically, by restricted injection, or biologically, by developing systems with tissue-specific affinities) and the safety to the patient of the administration system. These problems are to some extent interrelated, and it can be generally concluded that the smaller the therapeutic agent is, the easier it will become to develop efficient, targetable and safe administration systems.
  • BRIEF SUMMARY OF THE INVENTION
  • This problem is addressed by inducing so-called “exon-skipping” in cells. Exon-skipping results in mature mRNA that does not contain the skipped exon and thus, when the exon codes for amino acids, can lead to the expression of an altered product. Technology for exon-skipping is currently directed toward the use of so-called “Anti-sense Oligonucleotides” (AONs).
  • Much of this work is done in the mdx mouse model for Duchenne muscular dystrophy (DMD). The mdx mouse, which carries a nonsense mutation in exon 23 of the dystrophin gene, has been used as an animal model of Duchenne muscular dystrophy. Despite the mdx mutation, which should preclude the synthesis of a functional dystrophin protein, rare, naturally occurring dystrophin-positive fibers have been observed in mdx muscle tissue. These dystrophin-positive fibers are thought to have arisen from an apparently naturally occurring exon-skipping mechanism, either due to somatic mutations or through alternative splicing.
  • AONs directed to, respectively, the 3′ and 5′ splice sites of introns 22 and 23 in dystrophin pre-mRNA have been shown to interfere with factors normally involved in removal of intron 23 so that exon 23 was also removed from the mRNA (Wilton, 1999). In a similar study, Dunckley et al. (1998) showed that exon skipping using AONs directed to 3′ and 5′ splice sites can have unexpected results. They observed skipping of not only exon 23 but also of exons 24-29, thus resulting in an mRNA containing an exon 22-exon 30 junction.
  • The underlying mechanism for the appearance of the unexpected 22-30 splicing variant is not known. It could be due to the fact that splice sites contain consensus sequences leading to promiscuous hybridization of the oligos used to direct the exon skipping. Hybridization of the oligos to other splice sites than the sites of the exon to be skipped of course could easily interfere with the accuracy of the splicing process. On the other hand, the accuracy could be lacking due to the fact that two oligos (for the 5′ and the 3′ splice site) need to be used. Pre-mRNA containing one but not the other oligo could be prone to unexpected splicing variants.
  • To overcome these and other problems, provided is a method for directing splicing of a pre-mRNA in a system capable of performing a splicing operation comprising contacting the pre-mRNA in the system with an agent capable of specifically inhibiting an exon inclusion signal of at least one exon in the pre-mRNA, the method further comprising allowing splicing of the pre-mRNA. Interfering with an exon inclusion signal (EIS) has the advantage that such elements are located within the exon. By providing an antisense oligo for the interior of the exon to be skipped, it is possible to interfere with the exon inclusion signal, thereby effectively masking the exon from the splicing apparatus. The failure of the splicing apparatus to recognize the exon to be skipped thus leads to exclusion of the exon from the final mRNA.
  • The processes and compounds disclosed herein do not interfere directly with the enzymatic process of the splicing machinery (the joining of the exons). It is thought that this allows the method to be more robust and reliable. It is thought that an EIS is a particular structure of an exon that allows splice acceptor and donor to assume a particular spatial conformation. In this concept, it is the particular spatial conformation that enables the splicing machinery to recognize the exon. However, the invention is certainly not limited to this model.
  • It has been found that agents capable of binding to an exon can inhibit an EIS. Agents may specifically contact the exon at any point and still be able to specifically inhibit the EIS. The mRNA may be useful in itself. For instance, production of an undesired protein can be at least in part reduced by inhibiting inclusion of a required exon into the mRNA. In certain embodiments, the method further comprises allowing translation of mRNA produced from splicing of the pre-mRNA. In certain embodiments, the mRNA encodes a functional protein. In various embodiments, the protein comprises two or more domains, wherein at least one of the domains is encoded by the mRNA as a result of skipping of at least part of an exon in the pre-mRNA.
  • Exon skipping will typically, though not necessarily, be of relevance for proteins in the wild-type configuration, having at least two functional domains that each performs a function, wherein the domains are generated from distinct parts of the primary amino acid sequence. Examples are, for instance, transcription factors. Typically, these factors comprise a DNA binding domain and a domain that interacts with other proteins in the cell. Skipping of an exon that encodes a part of the primary amino acid sequence that lies between these two domains can lead to a shorter protein that comprises the same function, at least in part. Thus, detrimental mutations in this intermediary region (for instance, frame-shift or stop mutations) can be at least in part repaired by inducing exon skipping to allow synthesis of the shorter (partly) functional protein.
  • Using a method described herein, it is also possible to induce partial skipping of the exon. In this embodiment, the contacting results in activation of a cryptic splice site in a contacted exon. This embodiment broadens the potential for manipulation of the pre-mRNA leading to a functional protein. In certain embodiments, the system comprises a cell. In certain embodiments, the cell is cultured in vitro or in the organism in vivo. Typically, though not necessarily, the organism comprises a human or a mouse.
  • In certain embodiments, provided is a method for at least in part decreasing the production of an aberrant protein in a cell, the cell comprising pre-mRNA comprising exons coding for the protein, the method comprising providing the cell with an agent capable of specifically inhibiting an exon inclusion signal of at least one of the exons, the method further comprising allowing translation of mRNA produced from splicing of the pre-mRNA.
  • Any agent capable of specifically inhibiting an exon exclusion signal can be used for the invention. In certain embodiments, the agent comprises a nucleic acid or a functional equivalent thereof. In certain embodiments, but not necessarily, the nucleic acid is in single-stranded form. Peptide nucleic acid and other molecules comprising the same nucleic acid binding characteristics in kind, but not necessarily in amount, are suitable equivalents. Nucleic acid or an equivalent may comprise modifications to provide additional functionality. For instance, 2′-O-methyl oligoribonucleotides can be used. These ribonucleotides are more resistant to RNAse action than conventional oligonucleotides.
  • In various embodiments, the exon inclusion signal is interfered with by an antisense nucleic acid directed to an exon recognition sequence (ERS). These sequences are relatively purine-rich and can be distinguished by scrutinizing the sequence information of the exon to be skipped (Tanaka et al., 1994, Mol. Cell. Biol. 14, p. 1347-1354). Exon recognition sequences are thought to aid inclusion into mRNA of so-called weak exons (Achsel et al., 1996, J. Biochem. 120, p. 53-60). These weak exons comprise, for instance, 5′ and or 3′ splice sites that are less efficiently recognized by the splicing machinery. In the invention, it has been found that exon skipping can also be induced in so-called strong exons, i.e., exons which are normally efficiently recognized by the splicing machinery of the cell. From any given sequence, it is (almost) always possible to predict whether the sequence comprises putative exons and to determine whether these exons are strong or weak. Several algorithms for determining the strength of an exon exist. A useful algorithm can be found on the NetGene2 splice site prediction server (Brunak, et al., 1991, J. Mol. Biol. 220, p. 49-65). Exon skipping by a means of the invention can be induced in (almost) every exon, independent of whether the exon is a weak exon or a strong exon and also independent of whether the exon comprises an ERS. In certain embodiments, an exon that is targeted for skipping is a strong exon. In another preferred embodiment, an exon targeted for skipping does not comprise an ERS.
  • Methods of the invention can be used in many ways. In one embodiment, a method described herein is used to at least in part decrease the production of an aberrant protein. Such proteins can, for instance, be oncoproteins or viral proteins. In many tumors, not only the presence of an oncoprotein but also its relative level of expression has been associated with the phenotype of the tumor cell. Similarly, not only the presence of viral proteins but also the amount of viral protein in a cell determines the virulence of a particular virus. Moreover, for efficient multiplication and spread of a virus, the timing of expression in the life cycle and the balance in the amount of certain viral proteins in a cell determines whether viruses are efficiently or inefficiently produced. Using a method described herein, it is possible to lower the amount of aberrant protein in a cell such that, for instance, a tumor cell becomes less tumorigenic (metastatic) and/or a virus-infected cell produces less virus.
  • In certain embodiments, a method described herein is used to modify the aberrant protein into a functional protein. In one embodiment, the functional protein is capable of performing a function of a protein normally present in a cell but absent in the cells to be treated. Very often, even partial restoration of function results in significantly improved performance of the cell thus treated. Due to the better performance, such cells can also have a selective advantage over unmodified cells, thus aiding the efficacy of the treatment.
  • This aspect is particularly suited for the restoration of expression of defective genes. This is achieved by causing the specific skipping of targeted exons, thus bypassing or correcting deleterious mutations (typically stop-mutations or frame-shifting point mutations, single- or multi-exon deletions or insertions leading to translation termination).
  • Compared to gene-introduction strategies, this novel form of splice-modulation gene therapy requires the administration of much smaller therapeutic reagents, typically, but not limited to, 14-40 nucleotides. In certain embodiments, molecules of 14-25 nucleotides are used since these molecules are easier to produce and enter the cell more effectively. The methods of the invention allow much more flexibility in the subsequent design of effective and safe administration systems. An important additional advantage of this aspect is that it restores (at least some of) the activity of the endogenous gene, which still possesses most or all of its gene-regulatory circuitry, thus ensuring proper expression levels and the synthesis of tissue-specific isoforms.
  • This aspect can, in principle, be applied to any genetic disease or genetic predisposition to disease in which targeted skipping of specific exons would restore the translational reading frame when this has been disrupted by the original mutation, provided that translation of an internally slightly shorter protein is still fully or partly functional. Preferred embodiments for which this application can be of therapeutic value are: predisposition to second hit mutations in tumor suppressor genes, e.g., those involved in breast cancer, colon cancer, tuberous sclerosis, neurofibromatosis etc., where (partial) restoration of activity would preclude the manifestation of nullosomy by second hit mutations and thus would protect against tumorigenesis. Another preferred embodiment involves the (partial) restoration of defective gene products which have a direct disease causing effect, e.g., hemophilia A (clotting factor VIII deficiency), some forms of congenital hypothyroidism (due to thyroglobulin synthesis deficiency) and Duchenne muscular dystrophy (DMD), in which frame-shifting deletions, duplications and stop mutations in the X-linked dystrophin gene cause severe, progressive muscle degradation. DMD is typically lethal in late adolescence or early adulthood, while non-frame-shifting deletions or duplications in the same gene cause the much milder Becker muscular dystrophy (BMD), compatible with a life expectancy between 35-40 years to normal. In the embodiment as applied to DMD, the invention enables exon skipping to extend an existing deletion (or alter the mRNA product of an existing duplication) by as many adjacent exons as required to restore the reading frame and generate an internally slightly shortened, but still functional, protein. Based on the much milder clinical symptoms of BMD patients with the equivalent of this induced deletion, the disease in the DMD patients would have a much milder course after AON-therapy.
  • Many different mutations in the dystrophin gene can lead to a dysfunctional protein. (For a comprehensive inventory see WorldWideWeb.dmd.nl, the internationally accepted database for DMD and related disorders.) The precise exon to be skipped to generate a functional dystrophin protein varies from mutation to mutation. Table 1 comprises a non-limiting list of exons that can be skipped and lists for the mentioned exons some of the more frequently occurring dystrophin gene mutations that have been observed in humans and that can be treated with a method described herein. Skipping of the mentioned exon leads to a mutant dystrophin protein comprising at least the functionality of a Becker mutant. Thus, in one embodiment, provided is a method described herein wherein the exon inclusion signal is present in exon numbers 2, 8, 19, 29, 43, 44, 45, 46, 50, 51, 52 or 53 of the human dystrophin gene. The occurrence of certain deletion/insertion variations is more frequent than others. In the invention, it was found that by inducing skipping of exon 46 with a means or a method described herein, approximately 7% of DMD-deletion containing patients can be treated, resulting in the patients to comprise dystrophin-positive muscle fibers. By inducing skipping of exon 51, approximately 15% of DMD-deletion containing patients can be treated with a means or method described herein. Such treatment will result in the patient having at least some dystrophin-positive fibers. Thus, with either skipping of exon 46 or 51 using a method described herein, approximately 22% of the patients containing a deletion in the dystrophin gene can be treated. Thus, In various embodiments, the exon exclusion signal is present in exon 46 or exon 51. In a particularly preferred embodiment, the agent comprises a nucleic acid sequence according to hAON#4, hAON#6, hAON#8, hAON#9, hAON#11 and/or one or more of hAON#21-30 or a functional part, derivative and/or analogue of the hAON. A functional part, derivative and/or analogue of the hAON comprises the same exon skipping activity in kind, but not necessarily in amount, in a method described herein.
  • TABLE 1
    Therapeutic for
    DMD-deletions Frequency in
    Exon to be skipped (exons) WorldWideWeb.dmd.nl (%)
    2 3-7 2
    8 3-7 4
    4-7
    5-7
    6-7
    43 44 5
    44-47
    44 35-43 8
    45
    45-54
    45 18-44 13
    46-47
    44
    46-48
    46-49
    46-51
    46-53
    46 45 7
    50 51 5
    51-55
    51 50 15
    45-50
    48-50
    49-50
    52
    52-63
    52 51 3
    53
    53-55
    53 45-52 9
    48-52
    49-52
    50-52
    52
  • It can be advantageous to induce exon skipping of more than one exon in the pre-mRNA. For instance, considering the wide variety of mutations and the fixed nature of exon lengths and amino acid sequence flanking such mutations, the situation can occur that for restoration of function more than one exon needs to be skipped. A preferred but non-limiting example of such a case in the DMD deletion database is a 46-50 deletion. Patients comprising a 46-50 deletion do not produce functional dystrophin. However, an at least partially functional dystrophin can be generated by inducing skipping of both exon 45 and exon 51. Another preferred but non-limiting example is patients comprising a duplication of exon 2. By providing one agent capable of inhibiting an EIS of exon 2, it is possible to partly skip either one or both exons 2, thereby regenerating the wild-type protein next to the truncated or double exon 2 skipped protein. Another preferred but non-limiting example is the skipping of exons 45 through 50. This generates an in-frame Becker-like variant. This Becker-like variant can be generated to cure any mutation localized in exons 45, 46, 47, 48, 49, and/or 50 or combinations thereof. In one aspect, the invention therefore provides a method described herein further comprising providing the cell with another agent capable of inhibiting an exon inclusion signal in another exon of the pre-mRNA. Of course, it is completely within the scope of the invention to use two or more agents for the induction of exon skipping in pre-mRNA of two or more different genes.
  • In another aspect, provided is a method for selecting the suitable agents for splice-therapy and their validation as specific exon-skipping agents in pilot experiments. A method is provided for determining whether an agent is capable of specifically inhibiting an exon inclusion signal of an exon, comprising providing a cell having a pre-mRNA containing the exon with the agent, culturing the cell to allow the formation of an mRNA from the pre-mRNA and determining whether the exon is absent the mRNA. In certain embodiments, the agent comprises a nucleic acid or a functional equivalent thereof, the nucleic acid comprising complementarity to a part of the exon. Agents capable of inducing specific exon skipping can be identified with a method described herein. It is possible to include a prescreen for agents by first identifying whether the agent is capable of binding with a relatively high affinity to an exon containing nucleic acid, preferably RNA. To this end, a method for determining whether an agent is capable of specifically inhibiting an exon inclusion signal of an exon is provided, further comprising first determining in vitro the relative binding affinity of the nucleic acid or functional equivalent thereof to an RNA molecule comprising the exon.
  • In yet another aspect, an agent is provided that is obtainable by a method described herein. In certain embodiments, the agent comprises a nucleic acid or a functional equivalent thereof. Preferably the agent, when used to induce exon skipping in a cell, is capable of at least in part reducing the amount of aberrant protein in the cell. More preferably, the exon skipping results in an mRNA encoding a protein that is capable of performing a function in the cell. In a particularly preferred embodiment, the pre-mRNA is derived from a dystrophin gene. In certain embodiments, the functional protein comprises a mutant or normal dystrophin protein. In certain embodiments, the mutant dystrophin protein comprises at least the functionality of a dystrophin protein in a Becker patient. In a particularly preferred embodiment, the agent comprises the nucleic acid sequence of hAON#4, hAON#6, hAON#8, hAON#9, hAON#11 and/or one or more of hAON#21-30 or a functional part, derivative and/or analogue of the hAON. A functional part, derivative and/or analogue of the hAON comprises the same exon skipping activity in kind, but not necessarily in amount, in a method described herein.
  • The art describes many ways to deliver agents to cells. Particularly, nucleic acid delivery methods have been widely developed. The artisan is well capable of determining whether a method of delivery is suitable for performing the invention. In a non-limiting example, the method includes the packaging of an agent of the invention into liposomes, the liposomes being provided to cells comprising a target pre-mRNA. Liposomes are particularly suited for delivery of nucleic acid to cells. Antisense molecules capable of inducing exon skipping can be produced in a cell upon delivery of nucleic acid containing a transcription unit to produce antisense RNA. Non-limiting examples of suitable transcription units are small nuclear RNA (SNRP) or tRNA transcription units. The invention, therefore, further provides a nucleic acid delivery vehicle comprising a nucleic acid or functional equivalent thereof of the invention capable of inhibiting an exon inclusion signal. In one embodiment, the delivery vehicle is capable of expressing the nucleic acid of the invention. Of course, in case, for instance, single-stranded viruses are used as a vehicle, it is entirely within the scope of the invention when such a virus comprises only the antisense sequence of an agent of the invention. In another embodiment of single strand viruses, AONs of the invention are encoded by small nuclear RNA or tRNA transcription units on viral nucleic encapsulated by the virus as vehicle. A preferred single-stranded virus is adeno-associated virus.
  • In yet another embodiment, provided is the use of a nucleic acid or a nucleic acid delivery vehicle of the invention for the preparation of a medicament. In certain embodiments, the medicament is used for the treatment of an inherited disease. More preferably, the medicament is used for the treatment of Duchenne Muscular Dystrophy.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1. Deletion of exon 45 is one of the most frequent DMD-mutations. Due to this deletion, exon 44 is spliced to exon 46, the translational reading frame is interrupted, and a stop codon is created in exon 46 leading to a dystrophin deficiency. Our aim is to artificially induce the skipping of an additional exon, exon 46, in order to reestablish the reading frame and restore the synthesis of a slightly shorter, but largely functional, dystrophin protein as found in the much milder affected Becker muscular dystrophy patients affected by a deletion of both exons 45 and 46.
  • FIG. 2. Exon 46 contains a purine-rich region that is hypothesized to have a potential role in the regulation of its splicing in the pre-mRNA. A series of overlapping 2′O-methyl phosphorothioate antisense oligoribonucleotides (AONs) was designed directed at this purine-rich region in mouse dystrophin exon 46. The AONs differ both in length and sequence. The chemical modifications render the AONs resistant to endonucleases and RNaseH inside the muscle cells. To determine the transfection efficiency in our in vitro studies, the AONs contained a 5′ fluorescein group which allowed identification of AON-positive cells.
  • FIG. 3. To determine the binding affinity of the different AONs to the target exon 46 RNA, we performed gel mobility shift assays. In this figure, the five mAONs ( mAON# 4, 6, 8, 9, and 11) with highest affinity for the target RNA are shown. Upon binding of the AONs to the RNA, a complex is formed that exhibits a retarded gel mobility as can be determined by the band shift. The binding of the AONs to the target was sequence-specific. A random mAON, i.e. not specific for exon 46, did not generate a band shift.
  • FIGS. 4A and 4B. The mouse- and human-specific AONs which showed the highest binding affinity in the gel mobility shift assays were transfected into mouse and human myotube cultures.
  • FIG. 4A. RT-PCR analysis showed a truncated product, of which the size corresponded to exon 45 directly spliced to exon 47, in the mouse cell cultures upon transfection with the different mAONs# 4, 6, 9, and 11. No exon 46 skipping was detected following transfection with a random AON.
  • FIG. 4B. RT-PCR analysis in the human muscle cell cultures derived from one unaffected individual (C) and two unrelated DMD patients (P1 and P2) revealed truncated products upon transfection with hAON#4 and hAON#8. In the control, this product corresponded to exon 45 spliced to exon 47, while in the patients, the fragment size corresponded to exon 44 spliced to exon 47. No exon 46 skipping was detected in the non-transfected cell cultures or following transfection with a random HAON. Highest exon 46 skipping efficiencies were obtained with hAON#8.
  • FIG. 5. Sequence data from the RT-PCR products obtained from patient DL279.1 (corresponding to P1 in FIG. 4), which confirmed the deletion of exon 45 in this patient (upper panel), and the additional skipping of exon 46 following transfection with hAON#8 (lower panel). The skipping of exon 46 was specific, and exon 44 was exactly spliced to exon 47, which reestablishes the translational reading frame.
  • FIG. 6. Immunohistochemical analysis of the muscle cell culture from patient DL279.1 upon transfection with hAON#8. Cells were subject to two different dystrophin antibodies raised against different regions of the protein, located proximally (ManDys-1, ex. 31-32) and distally (Dys-2, ex. 77-79) from the targeted exon 46. The lower panel shows the absence of a dystrophin protein in the myotubes, whereas the hAON#8-induced skipping of exon 46 clearly restored the synthesis of a dystrophin protein as detected by both antibodies (upper panel).
  • FIG. 7A. RT-PCR analysis of RNA isolated from human control muscle cell cultures treated with hAON#23, #24, #27, #28, or #29. An additional aberrant splicing product was obtained in cells treated with hAON#28 and #29. Sequence analysis revealed the utilization of an in-frame cryptic splice site within exon 51 that is used at a low frequency upon AON treatment. The product generated included a partial exon 51, which also had a restored reading frame, thereby confirming further the therapeutic value.
  • FIG. 7B. A truncated product, with a size corresponding to exon 50 spliced to exon 52, was detected in cells treated with hAON#23 and #28. Sequence analysis of these products confirmed the precise skipping of exon 51.
  • FIG. 8A. Gel mobility shift assays were performed to determine the binding affinity of the different h29AON#'s for the exon 29 target RNA. When compared to non-hybridized RNA (none), h29AON#1, #2, #4, #6, #9, #10, and #11 generated complexes with lower gel mobilities, indicating their binding to the RNA. A random AON derived from dystrophin exon 19 did not generate a complex.
  • FIG. 8B. RT-PCR analysis of RNA isolated from human control muscle cell cultures treated with h29AON#1, #2, #4, #6, #9, #10, or #11 revealed a truncated product of which the size corresponded to exon 28 spliced to exon 30. These results indicate that exon 29 can specifically be skipped using AONs directed to sequences either within (h29AON#1, #2, #4, or #6) or outside (h29AON#9, #10, or #11) the hypothesized ERS in exon 29. An additional aberrant splicing product was observed that resulted from skipping of both exon 28 and exon 29 (confirmed by sequence data not shown). Although this product was also present in non-treated cells, suggesting that this alternative skipping event may occur naturally, it was enhanced by the AON-treatment. AON 19, derived from dystrophin exon 19, did not induce exon 29 skipping.
  • FIG. 8C. The specific skipping of exon 29 was confirmed by sequence data from the truncated RT-PCR fragments. Shown here is the sequence obtained from the exon 29 skipping product in cells treated with h29AON#1.
  • FIG. 9A. RT-PCR analysis of RNA isolated from mouse gastrocnemius muscles two days post-injection of 5, 10, or 20 μg of either mAON#4, #6, or #11. Truncated products, with a size corresponding to exon 45 spliced to exon 47, were detected in all treated muscles. The samples -RT, -RNA, AD-1, and AD-2 were analyzed as negative controls for the RT-PCR reactions.
  • FIG. 9B. Sequence analysis of the truncated products generated by mAON#4 and #6 (and #11, not shown) confirmed the precise skipping of exon 46.
  • DETAILED DESCRIPTION OF THE INVENTION Examples Example 1
  • Since exon 45 is one of the most frequently deleted exons in DMD, we initially aimed at inducing the specific skipping of exon 46 (FIG. 1). This would produce the shorter, largely functional dystrophin found in BMD patients carrying a deletion of exons 45 and 46. The system was initially set up for modulation of dystrophin pre-mRNA splicing of the mouse dystrophin gene. We later aimed for the human dystrophin gene with the intention to restore the translational reading frame and dystrophin synthesis in muscle cells from DMD patients affected by a deletion of exon 45.
  • Design of mAONs and hAONs
  • A series of mouse- and human-specific AONs (mAONs and hAONs) was designed, directed at an internal part of exon 46 that contains a stretch of purine-rich sequences and is hypothesized to have a putative regulatory role in the splicing process of exon 46 (FIG. 2). For the initial screening of the AONs in the gel mobility shift assays (see below), we used non-modified DNA-oligonucleotides (synthesized by EuroGentec, Belgium). For the actual transfection experiments in muscle cells, we used 2′-O-methyl-phosphorothioate oligoribonucleotides (also synthesized by EuroGentec, Belgium). These modified RNA oligonucleotides are known to be resistant to endonucleases and RNaseH, and to bind to RNA with high affinity. The sequences of those AONs that were eventually effective and applied in muscle cells in vitro are shown below. The corresponding mouse and human-specific AONs are highly homologous but not completely identical.
  • The listing below refers to the deoxy-form used for testing, in the finally used 2-O-methyl ribonucleotides all T's should be read as U's.
  • mAON#2: 5′ GCAATGTTATCTGCTT (SEQ ID NO:1)
    mAON#3: 5′ GTTATCTGCTTCTTCC (SEQ ID NO:2)
    mAON#4: 5′ CTGCTTCTTCCAGCC (SEQ ID NO:3)
    mAON#5: 5′ TCTGCTTCTTCCAGC (SEQ ID NO:4)
    mAON#6: 5′ GTTATCTGCTTCTTCCAGCC (SEQ ID NO:5)
    mAON#7: 5′ CTTTTAGCTGCTGCTC (SEQ ID NO:6)
    mAON#8: 5′ GTTGTTCTTTTAGCTGCTGC (SEQ ID NO:7)
    mAON#9: 5′ TTAGCTGCTGCTCAT (SEQ ID NO:8)
    mAON#10: 5′ TTTAGCTGCTGCTCATCTCC (SEQ ID NO:9)
    mAON#11: 5′ CTGCTGCTCATCTCC (SEQ ID NO:10)
    hAON#4: 5′ CTGCTTCCTCCAACC (SEQ ID NO:11)
    hAON#6: 5′ GTTATCTGCTTCCTCCAACC (SEQ ID NO:12)
    hAON#8: 5′ GCTTTTCTTTTAGTTGCTGC (SEQ ID NO:13)
    hAON#9: 5′ TTAGTTGCTGCTCTT (SEQ ID NO:14)
    hAON#11: 5′ TTGCTGCTCTTTTCC (SEQ ID NO:15)
  • Gel Mobility Shift Assays
  • The efficacy of the AONs is determined by their binding affinity for the target sequence. Notwithstanding recent improvements in computer simulation programs for the prediction of RNA-folding, it is difficult to speculate which of the designed AONs would be capable of binding the target sequence with a relatively high affinity. Therefore, we performed gel mobility shift assays (according to protocols described by Bruice et al., 1997). The exon 46 target RNA fragment was generated by in vitro T7-transcription from a PCR fragment (amplified from either murine or human muscle mRNA using a sense primer that contains the T7 promoter sequence) in the presence of 32P-CTP. The binding affinity of the individual AONs (0.5 μmol) for the target transcript fragments was determined by hybridization at 37° C. for 30 minutes and subsequent polyacrylamide (8%) gel electrophoresis. We performed these assays for the screening of both the mouse and human-specific AONs (FIG. 3). At least 5 different mouse-specific AONs ( mAON# 4, 6, 8, 9 and 11) and four corresponding human-specific AONs ( hAON# 4, 6, 8, and 9) generated a mobility shift, demonstrating their binding affinity for the target RNA.
  • Transfection into Muscle Cell Cultures
  • The exon 46-specific AONs which showed the highest target binding affinity in gel mobility shift assays were selected for analysis of their efficacy in inducing the skipping in muscle cells in vitro. In all transfection experiments, we included a non-specific AON as a negative control for the specific skipping of exon 46. As mentioned, the system was first set up in mouse muscle cells. We used both proliferating myoblasts and post-mitotic myotube cultures (expressing higher levels of dystrophin) derived from the mouse muscle cell line C2C12. For the subsequent experiments in human-derived muscle cell cultures, we used primary muscle cell cultures isolated from muscle biopsies from one unaffected individual and two unrelated DMD patients carrying a deletion of exon 45. These heterogeneous cultures contained approximately 20-40% myogenic cells. The different AONs (at a concentration of 1 μM) were transfected into the cells using the cationic polymer PEI (MBI Fermentas) at a ratio-equivalent of 3. The AONs transfected in these experiments contained a 5′ fluorescein group which allowed us to determine the transfection efficiencies by counting the number of fluorescent nuclei. Typically, more than 60% of cells showed specific nuclear uptake of the AONs. To facilitate RT-PCR analysis, RNA was isolated 24 hours post-transfection using RNAzol B (CamPro Scientific, The Netherlands).
  • RT-PCR and Sequence Analysis
  • RNA was reverse transcribed using C. therm. polymerase (Roche) and an exon 48-specific reverse primer. To facilitate the detection of skipping of dystrophin exon 46, the cDNA was amplified by two rounds of PCR, including a nested amplification using primers in exons 44 and 47 (for the human system), or exons 45 and 47 (for the mouse system). In the mouse myoblast and myotube cell cultures, we detected a truncated product of which the size corresponded to exon 45 directly spliced to exon 47 (FIG. 4). Subsequent sequence analysis confirmed the specific skipping of exon 46 from these mouse dystrophin transcripts. The efficiency of exon skipping was different for the individual AONs, with mAON#4 and #11 showing the highest efficiencies. Following these promising results, we focused on inducing a similar modulation of dystrophin splicing in the human-derived muscle cell cultures. Accordingly, we detected a truncated product in the control muscle cells, corresponding to exon 45 spliced to exon 47. Interestingly, in the patient-derived muscle cells, a shorter fragment was detected, which consisted of exon 44 spliced to exon 47. The specific skipping of exon 46 from the human dystrophin transcripts was confirmed by sequence data. This splicing modulation of both the mouse and human dystrophin transcript was neither observed in non-transfected cell cultures nor in cultures transfected with a non-specific AON.
  • Immunohistochemical Analysis
  • We intended to induce the skipping of exon 46 in muscle cells from patients carrying an exon 45 deletion in order to restore the translation and synthesis of a dystrophin protein. To detect a dystrophin product upon transfection with hAON#8, the two patient-derived muscle cell cultures were subject to immunocytochemistry using two different dystrophin monoclonal antibodies (Mandys-1 and Dys-2) raised against domains of the dystrophin protein located proximal and distal of the targeted region respectively. Fluorescent analysis revealed restoration of dystrophin synthesis in both patient-derived cell cultures (FIG. 5). Approximately at least 80% of the fibers stained positive for dystrophin in the treated samples.
  • Our results show, for the first time, the restoration of dystrophin synthesis from the endogenous DMD gene in muscle cells from DMD patients. This is a proof of principle of the feasibility of targeted modulation of dystrophin pre-mRNA splicing for therapeutic purposes.
  • Targeted Skipping of Exon 51 Simultaneous Skipping of Dystrophin Exons
  • The targeted skipping of exon 51. We demonstrated the feasibility of AON-mediated modulation of dystrophin exon 46 splicing, in mouse and human muscle cells in vitro. These findings warranted further studies to evaluate AONs as therapeutic agents for DMD. Since most DMD-causing deletions are clustered in two mutation hot spots, the targeted skipping of one particular exon can restore the reading frame in series of patients with different mutations (see Table 1). Exon 51 is an interesting target exon. The skipping of this exon is therapeutically applicable in patients carrying deletions spanning exon 50, exons 45-50, exons 48-50, exons 49-50, exon 52, and exons 52-63, which includes a total of 15% of patients from our Leiden database.
  • We designed a series of ten human-specific AONs (hAON#21-30, see below) directed at different purine-rich regions within dystrophin exon 51. These purine-rich stretches suggested the presence of a putative exon splicing regulatory element that we aimed to block in order to induce the elimination of that exon during the splicing process. All experiments were performed according to protocols as described for the skipping of exon 46 (see above). Gel mobility shift assays were performed to identify those hAONs with high binding affinity for the target RNA. We selected the five hAONs that showed the highest affinity. These hAONs were transfected into human control muscle cell cultures in order to test the feasibility of skipping exon 51 in vitro. RNA was isolated 24 hours post-transfection, and cDNA was generated using an exon 53- or 65-specific reverse primer. PCR-amplification of the targeted region was performed using different primer combinations flanking exon 51. The RT-PCR and sequence analysis revealed that we were able to induce the specific skipping of exon 51 from the human dystrophin transcript. We subsequently transfected two hAONs (#23 and #29) shown to be capable of inducing skipping of the exon into six different muscle cell cultures derived from DMD-patients carrying one of the mutations mentioned above. The skipping of exon 51 in these cultures was confirmed by RT-PCR and sequence analysis (FIG. 7). More importantly, immunohistochemical analysis, using multiple antibodies raised against different parts of the dystrophin protein, showed in all cases that, due to the skipping of exon 51, the synthesis of a dystrophin protein was restored.
  • Exon 51-specific hAONs:
  • hAON#21: 5′ CCACAGGTTGTGTCACCAG (SEQ ID NO:16)
    hAON#22: 5′ TTTCCTTAGTAACCACAGGTT (SEQ ID NO:17)
    hAON#23: 5′ TGGCATTTCTAGTTTGG (SEQ ID NO:18)
    hAON#24: 5′ CCAGAGCAGGTACCTCCAACATC (SEQ ID NO:19)
    hAON#25: 5′ GGTAAGTTCTGTCCAAGCCC (SEQ ID NO:20)
    hAON#26: 5′ TCACCCTCTGTGATTTTAT (SEQ ID NO:21)
    hAON#27: 5′ CCCTCTGTGATTTT (SEQ ID NO:22)
    hAON#28: 5′ TCACCCACCATCACCCT (SEQ ID NO:23)
    hAON#29: 5′ TGATATCCTCAAGGTCACCC (SEQ ID NO:24)
    hAON#30: 5′ CTGCTTGATGATCATCTCGTT (SEQ ID NO:25)
  • Simultaneous Skipping of Multiple Dystrophin Exons
  • The skipping of one additional exon, such as exon 46 or exon 51, restores the reading frame for a considerable number of different DMD mutations. The range of mutations for which this strategy is applicable can be enlarged by the simultaneous skipping of more than one exon. For instance, in DMD patients with a deletion of exon 46 to exon 50, only the skipping of both the deletion-flanking exons 45 and 51 enables the reestablishment of the translational reading frame.
  • ERS-Independent Exon Skipping
  • A mutation in exon 29 leads to the skipping of this exon in two Becker muscular dystrophy patients (Ginjaar at al., 2000, EJHG, vol. 8, p. 793-796). We studied the feasibility of directing the skipping of exon 29 through targeting the site of mutation by AONs. The mutation is located in a purine-rich stretch that could be associated with ERS activity. We designed a series of AONs (see below) directed to sequences both within (h29AON#1 to h29AON#6) and outside (h29AON#7 to h29AON#11) the hypothesized ERS. Gel mobility shift assays were performed (as described) to identify those AONs with highest affinity for the target RNA (FIG. 8). Subsequently, h29AON#1, #2, #4, #6, #9, #10, and #11 were transfected into human control myotube cultures using the PEI transfection reagent. RNA was isolated 24 hrs post-transfection, and cDNA was generated using an exon 31-specific reverse primer. PCR-amplification of the targeted region was performed using different primer combinations flanking exon 29. This RT-PCR and subsequent sequence analysis (FIGS. 8B and 8C) revealed that we were able to induce the skipping of exon 29 from the human dystrophin transcript. However, the AONs that facilitated this skipping were directed to sequences both within and outside the hypothesized ERS (h29AON#1, #2, #4, #6, #9, and #11). These results suggest that skipping of exon 29 occurs independent of whether or not exon 29 contains an ERS and that, therefore, the binding of the AONs to exon 29 more likely inactivated an exon inclusion signal rather than an ERS. This proof of ERS-independent exon skipping may extend the overall applicability of this therapy to exons without ERS's.
  • h29AON#1: 5′ TATCCTCTGAATGTCGCATC (SEQ ID NO:26)
    h29AON#2: 5′ GGTTATCCTCTGAATGTCGC (SEQ ID NO:27)
    h29AON#3: 5′ TCTGTTAGGGTCTGTGCC (SEQ ID NO:28)
    h29AON#4: 5′ CCATCTGTTAGGGTCTGTG (SEQ ID NO:29)
    h29AON#5: 5′ GTCTGTGCCAATATGCG (SEQ ID NO:30)
    h29AON#6: 5′ TCTGTGCCAATATGCGAATC (SEQ ID NO:31)
    h29AON#7: 5′ TGTCTCAAGTTCCTC (SEQ ID NO:32)
    h29AON#8: 5′ GAATTAAATGTCTCAAGTTC (SEQ ID NO:33)
    h29AON#9: 5′ TTAAATGTCTCAAGTTCC (SEQ ID NO:34)
    h29AON#10: 5′ GTAGTTCCCTCCAACG (SEQ ID NO:35)
    h29AON#11: 5′ CATGTAGTTCCCTCC (SEQ ID NO:36)
  • AON-Induced Exon 46 Skipping In Vivo in Murine Muscle Tissue.
  • Following the promising results in cultured muscle cells, we tested the different mouse dystrophin exon 46-specific AONs in vivo by injecting them, linked to polyethylenimine (PEI), into the gastrocnemius muscles of control mice. With mAON#4, #6, and #11, previously shown to be effective in mouse muscle cells in vitro, we were able to induce the skipping of exon 46 in muscle tissue in vivo as determined by both RT-PCR and sequence analysis (FIG. 9). The in vivo exon 46 skipping was dose-dependent with highest efficiencies (up to 10%) following injection of 20 μg per muscle per day for two subsequent days.
  • REFERENCES
    • Achsel et al., 1996, J. Biochem. 120, pp. 53-60.
    • Bruice T. W. and W. F. Lima, 1997, Biochemistry 36(16): pp. 5004-5019.
    • Brunak at al., 1991, J. Mol. Biol. 220, pp. 49-65.
    • Dunckley M. G. et al., 1998, Human molecular genetics 7, pp. 1083-1090.
    • Ginjaar et al., 2000, EJHG, vol. 8, pp. 793-796.
    • Mann et al., 2001, PNAS vol. 98, pp. 42-47.
    • Tanaka et al., 1994, Mol. Cell. Biol. 14, pp. 1347-1354.
    • Wilton S. D., et al., 1999, Neuromuscular disorders 9, pp. 330-338.
      Details and background on Duchenne Muscular Dystrophy and related diseases can be found on website WorldWideWeb.dmd.nl

Claims (24)

1. A method for directing splicing of a pre-mRNA in a cell capable of performing a splicing operation to reduce the production of an undesired protein in the cell, the method comprising:
contacting the pre-mRNA in the cell with an antisense-oligonucleotide capable of specifically inhibiting an exon inclusion signal of at least one exon in the pre-mRNA, wherein the antisense-oligonucleotide is directed against the interior of the at least one exon and contains between 14-40 nucleotides.
2. The method according to claim 1, wherein the mRNA encodes a functional protein.
3. The method according to claim 1, wherein the undesired protein comprises two or more domains, wherein at least one of the domains is encoded by the mRNA as a result of skipping of at least part of an exon in the pre-mRNA.
4. The method according to claim 1, wherein the contacting results in activation of a cryptic splice site in a contacted exon.
5. A method for at least in part decreasing the production of an aberrant protein in a cell, the cell comprising pre-mRNA comprising exons coding for the protein, the method comprising:
providing the cell with an antisense-oligonucleotide capable of specifically inhibiting an exon inclusion signal of at least one of the exons, wherein the antisense-oligonucleotide is directed against the interior of the at least one exon and contains between 14-40 nucleotides, the method further comprising allowing translation of mRNA produced from splicing of the pre-mRNA.
6. The method according to claim 1, wherein the exon inclusion signal comprises an exon recognition sequence.
7. The method according to claim 1, wherein the exon inclusion signal is present in an exon comprising a strong splice donor/acceptor pair.
8. The method according to claim 1, wherein the translation results in a mutant or normal dystrophin protein.
9. The method according to claim 8, wherein the mutant dystrophin protein is equivalent to a dystrophin protein of a Becker Muscular Dystrophy patient.
10. The method according to claim 9, wherein the antisense-oligonucleotide contains between 15-25 nucleotides.
11. The method according to claim 1, further comprising providing the cell with another antisense-oligonucleotide capable of inhibiting an exon inclusion signal present in another exon of the pre-mRNA.
12. A method for determining whether a nucleic acid, having complementarity to a part of an exon, is capable of specifically inhibiting an exon inclusion signal of the exon, the method comprising:
providing a cell having a pre-mRNA containing the exon, with the nucleic acid,
culturing the cell to allow the formation of an mRNA from the pre-mRNA, and
determining whether the exon is absent from the mRNA.
13. The method according to claim 12, further comprising determining in vitro the relative binding affinity of the nucleic acid to an RNA molecule comprising the exon.
14. A nucleic acid obtainable by the method according to claim 12.
15. A nucleic acid delivery vehicle comprising a nucleic acid according to claim 14, or the complement thereof.
16. A nucleic acid delivery vehicle capable of expressing the nucleic acid of claim 14.
17. A non-human animal provided with the nucleic acid of claim 14.
18. The non-human animal of claim 17, further comprising a nucleic acid encoding a human protein.
19. The non-human animal of claim 18, further comprising a silencing mutation in the gene encoding an animal homologue of the human protein.
20. The method according to claim 1, wherein the undesired protein in the wild-type has at least two functional domains generated from distinct parts of the primary amino acid sequence.
21. The method according to claim 1, wherein the undesired protein is an aberrant protein.
22. The method according to claim 21, wherein the aberrant protein is an oncoprotein or viral protein.
23. The method according to claim 1, wherein the undesired protein is involved in a genetic disease or genetic predisposition to disease.
24. The method according to claim 1, wherein the undesired protein is involved in breast cancer, colon cancer, tuberous sclerosis, neurofibromatosis, hemophilia A or congenital hypothyroidism.
US11/982,285 2000-09-21 2007-10-31 Induction of exon skipping in eukaryotic cells Abandoned US20080209581A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US11/982,285 US20080209581A1 (en) 2000-09-21 2007-10-31 Induction of exon skipping in eukaryotic cells
US14/331,934 US20140350076A1 (en) 2000-09-21 2014-07-15 Induction of exon skipping in eukaryotic cells
US14/712,753 US20150322434A1 (en) 2000-09-21 2015-05-14 Induction of exon skipping in eukaryotic cells
US14/839,200 US20150361424A1 (en) 2000-09-21 2015-08-28 Induction of exon skipping in eukaryotic cells

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
EP00203283A EP1191097A1 (en) 2000-09-21 2000-09-21 Induction of exon skipping in eukaryotic cells
EP00203283.7 2000-09-21
PCT/NL2001/000697 WO2002024906A1 (en) 2000-09-21 2001-09-21 Induction of exon skipping in eukaryotic cells
US10/395,031 US7973015B2 (en) 2000-09-21 2003-03-21 Induction of exon skipping in eukaryotic cells
US11/982,285 US20080209581A1 (en) 2000-09-21 2007-10-31 Induction of exon skipping in eukaryotic cells

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US10/395,031 Continuation US7973015B2 (en) 2000-09-21 2003-03-21 Induction of exon skipping in eukaryotic cells
US10/395,031 Division US7973015B2 (en) 2000-09-21 2003-03-21 Induction of exon skipping in eukaryotic cells

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/383,897 Continuation US20090228998A1 (en) 2000-09-21 2009-03-30 Induction of exon skipping in eukaryotic cells

Publications (1)

Publication Number Publication Date
US20080209581A1 true US20080209581A1 (en) 2008-08-28

Family

ID=8172043

Family Applications (6)

Application Number Title Priority Date Filing Date
US10/395,031 Expired - Fee Related US7973015B2 (en) 2000-09-21 2003-03-21 Induction of exon skipping in eukaryotic cells
US11/982,285 Abandoned US20080209581A1 (en) 2000-09-21 2007-10-31 Induction of exon skipping in eukaryotic cells
US12/383,897 Abandoned US20090228998A1 (en) 2000-09-21 2009-03-30 Induction of exon skipping in eukaryotic cells
US14/331,934 Abandoned US20140350076A1 (en) 2000-09-21 2014-07-15 Induction of exon skipping in eukaryotic cells
US14/712,753 Abandoned US20150322434A1 (en) 2000-09-21 2015-05-14 Induction of exon skipping in eukaryotic cells
US14/839,200 Abandoned US20150361424A1 (en) 2000-09-21 2015-08-28 Induction of exon skipping in eukaryotic cells

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/395,031 Expired - Fee Related US7973015B2 (en) 2000-09-21 2003-03-21 Induction of exon skipping in eukaryotic cells

Family Applications After (4)

Application Number Title Priority Date Filing Date
US12/383,897 Abandoned US20090228998A1 (en) 2000-09-21 2009-03-30 Induction of exon skipping in eukaryotic cells
US14/331,934 Abandoned US20140350076A1 (en) 2000-09-21 2014-07-15 Induction of exon skipping in eukaryotic cells
US14/712,753 Abandoned US20150322434A1 (en) 2000-09-21 2015-05-14 Induction of exon skipping in eukaryotic cells
US14/839,200 Abandoned US20150361424A1 (en) 2000-09-21 2015-08-28 Induction of exon skipping in eukaryotic cells

Country Status (17)

Country Link
US (6) US7973015B2 (en)
EP (14) EP1191097A1 (en)
JP (7) JP4846965B2 (en)
AT (1) ATE409224T2 (en)
AU (4) AU2002211062C1 (en)
CA (1) CA2423044C (en)
CY (10) CY1109601T1 (en)
DE (1) DE60135936D1 (en)
DK (10) DK2594640T3 (en)
ES (11) ES2561292T3 (en)
HK (8) HK1184820A1 (en)
LT (3) LT2801618T (en)
NZ (1) NZ524853A (en)
PT (7) PT2940139T (en)
SI (1) SI2801618T1 (en)
TR (1) TR201810606T4 (en)
WO (1) WO2002024906A1 (en)

Cited By (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100130591A1 (en) * 2008-10-24 2010-05-27 Peter Sazani Multiple exon skipping compositions for dmd
US20110015258A1 (en) * 2004-06-28 2011-01-20 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
US20110046360A1 (en) * 2002-11-25 2011-02-24 Masafumi Matsuo ENA NUCLEIC ACID DRUGS MODIFYING SPLICING IN mRNA PRECURSOR
US8637483B2 (en) 2009-11-12 2014-01-28 The University Of Western Australia Antisense molecules and methods for treating pathologies
EP2796425A1 (en) 2013-04-24 2014-10-29 Corning Incorporated Delamination resistant pharmaceutical glass containers containing active pharmaceutical ingredients
US9217148B2 (en) 2013-03-14 2015-12-22 Sarepta Therapeutics, Inc. Exon skipping compositions for treating muscular dystrophy
US9506058B2 (en) 2013-03-15 2016-11-29 Sarepta Therapeutics, Inc. Compositions for treating muscular dystrophy
US9605019B2 (en) 2011-07-19 2017-03-28 Wave Life Sciences Ltd. Methods for the synthesis of functionalized nucleic acids
US9611471B2 (en) 2010-08-05 2017-04-04 Academisch Ziekenhuis Leiden Antisense oligonucleotide directed removal of proteolytic cleavage sites from proteins
US9617547B2 (en) 2012-07-13 2017-04-11 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant
US9695211B2 (en) 2008-12-02 2017-07-04 Wave Life Sciences Japan, Inc. Method for the synthesis of phosphorus atom modified nucleic acids
US9744183B2 (en) 2009-07-06 2017-08-29 Wave Life Sciences Ltd. Nucleic acid prodrugs and methods of use thereof
US9890379B2 (en) 2006-08-11 2018-02-13 Biomarin Technologies B.V. Treatment of genetic disorders associated with DNA repeat instability
US9926544B2 (en) 2014-01-24 2018-03-27 Am-Pharma B.V. Chimeric alkaline phosphatase-like proteins
US9982257B2 (en) 2012-07-13 2018-05-29 Wave Life Sciences Ltd. Chiral control
US10144933B2 (en) 2014-01-15 2018-12-04 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having immunity induction activity, and immunity induction activator
US10149905B2 (en) 2014-01-15 2018-12-11 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having antitumor effect and antitumor agent
US10160969B2 (en) 2014-01-16 2018-12-25 Wave Life Sciences Ltd. Chiral design
US10167309B2 (en) 2012-07-13 2019-01-01 Wave Life Sciences Ltd. Asymmetric auxiliary group
US10179912B2 (en) 2012-01-27 2019-01-15 Biomarin Technologies B.V. RNA modulating oligonucleotides with improved characteristics for the treatment of duchenne and becker muscular dystrophy
US10246707B2 (en) 2008-05-14 2019-04-02 Biomarin Technologies B.V. Method for efficient exon (44) skipping in duchenne muscular dystrophy and associated means
US10322173B2 (en) 2014-01-15 2019-06-18 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having anti-allergic activity, and anti-allergic agent
US10428019B2 (en) 2010-09-24 2019-10-01 Wave Life Sciences Ltd. Chiral auxiliaries
US10450568B2 (en) 2015-10-09 2019-10-22 Wave Life Sciences Ltd. Oligonucleotide compositions and methods thereof
US10570380B2 (en) 2014-01-24 2020-02-25 Am-Pharma B.V. Downstream processing of an alkaline phosphatase
US10876114B2 (en) 2007-10-26 2020-12-29 Biomarin Technologies B.V. Methods and means for efficient skipping of at least one of the following exons of the human Duchenne muscular dystrophy gene: 43, 46, 50-53
USRE48960E1 (en) 2004-06-28 2022-03-08 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof

Families Citing this family (98)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20020097241A (en) 2000-05-04 2002-12-31 에이브이아이 바이오파마 인코포레이티드 Splice-region antisense composition and method
EP1191097A1 (en) 2000-09-21 2002-03-27 Leids Universitair Medisch Centrum Induction of exon skipping in eukaryotic cells
ITRM20020253A1 (en) * 2002-05-08 2003-11-10 Univ Roma SNRNA CHEMICAL MOLECULES WITH ANTISENSE SEQUENCES FOR SPLICING JUNCTIONS OF THE DYSTROPHINE GENE AND THERAPEUTIC APPLICATIONS.
CA2524255C (en) 2003-03-21 2014-02-11 Academisch Ziekenhuis Leiden Modulation of exon recognition in pre-mrna by interfering with the secondary rna structure
CA2523672C (en) 2003-04-29 2012-07-17 Avi Biopharma, Inc. Compositions for enhancing transport of molecules into cells
US20050288246A1 (en) 2004-05-24 2005-12-29 Iversen Patrick L Peptide conjugated, inosine-substituted antisense oligomer compound and method
FR2874384B1 (en) * 2004-08-17 2010-07-30 Genethon ADENO-ASSOCIATED VIRAL VECTOR FOR PRODUCING EXON JUMP IN A GENE ENCODING A PROTEIN WITH DISPENSABLE DOMAINS
CA2596506C (en) 2005-02-09 2021-04-06 Avi Biopharma, Inc. Antisense composition and method for treating muscle atrophy
AU2006237727B2 (en) * 2005-04-22 2012-06-28 Academisch Ziekenhuis Leiden Modulation of exon recognition in pre-mRNA by interfering with the binding of SR proteins and by interfering with secondary RNA structure.
US8067571B2 (en) 2005-07-13 2011-11-29 Avi Biopharma, Inc. Antibacterial antisense oligonucleotide and method
KR101789603B1 (en) * 2005-11-10 2017-11-21 더 유니버시티 오브 노쓰 캐롤라이나 엣 채플 힐 Splice switching oligomers for tnf superfamily receptors and their use in treatment of disease
US7785834B2 (en) * 2005-11-10 2010-08-31 Ercole Biotech, Inc. Soluble TNF receptors and their use in treatment of disease
WO2007123391A1 (en) 2006-04-20 2007-11-01 Academisch Ziekenhuis Leiden Therapeutic intervention in a genetic disease in an individual by modifying expression of an aberrantly expressed gene.
EP1857548A1 (en) 2006-05-19 2007-11-21 Academisch Ziekenhuis Leiden Means and method for inducing exon-skipping
US20090264353A1 (en) * 2007-10-19 2009-10-22 Santaris Pharma A/S Splice Switching Oligomers for TNF Superfamily Receptors and their Use in Treatment of Disease
ES2694726T3 (en) * 2007-06-29 2018-12-26 Sarepta Therapeutics, Inc. Tissue specific peptide conjugates and methods
US20100016215A1 (en) * 2007-06-29 2010-01-21 Avi Biopharma, Inc. Compound and method for treating myotonic dystrophy
JP2010533170A (en) 2007-07-12 2010-10-21 プロセンサ テクノロジーズ ビー.ブイ. Molecules for targeting compounds to various selected organs, tissues or tumor cells
JP5706157B2 (en) 2007-07-12 2015-04-22 プロセンサ テクノロジーズ ビー.ブイ.Prosensa Technologies B.V. Molecules for targeting compounds to various selected organs or tissues
USRE48468E1 (en) 2007-10-26 2021-03-16 Biomarin Technologies B.V. Means and methods for counteracting muscle disorders
WO2009099326A1 (en) 2008-02-08 2009-08-13 Prosensa Holding Bv Methods and means for treating dna repeat instability associated genetic disorders
US8084601B2 (en) 2008-09-11 2011-12-27 Royal Holloway And Bedford New College Royal Holloway, University Of London Oligomers
US20120046342A1 (en) 2009-04-24 2012-02-23 Prosensa Technologies B.V. Oligonucleotide comprising an inosine for treating dmd
US20110269665A1 (en) 2009-06-26 2011-11-03 Avi Biopharma, Inc. Compound and method for treating myotonic dystrophy
US20120270930A1 (en) 2009-10-29 2012-10-25 Academisch Ziekenhuis Leiden H.O.D.N. Lumc Methods and compositions for dysferlin exon-skipping
JP6141018B2 (en) 2009-12-24 2017-06-07 バイオマリン テクノロジーズ ベー.フェー. Molecules for treating inflammatory disorders
TWI541024B (en) 2010-09-01 2016-07-11 日本新藥股份有限公司 Antisense nucleic acid
WO2012138223A2 (en) 2011-04-05 2012-10-11 Academisch Ziekenhuis Leiden H.O.D.N. Lumc Compounds and methods for altering activin receptor-like kinase signalling
US9161948B2 (en) 2011-05-05 2015-10-20 Sarepta Therapeutics, Inc. Peptide oligonucleotide conjugates
EP2750715B1 (en) 2011-08-30 2018-10-31 The Regents of The University of California Identification of small molecules that enhance therapeutic exon skipping
PL2753694T3 (en) 2011-09-05 2018-01-31 Stichting Katholieke Univ Antisense oligonucleotides for the treatment of leber congenital amaurosis
US20130085139A1 (en) 2011-10-04 2013-04-04 Royal Holloway And Bedford New College Oligomers
CA2861247C (en) * 2011-12-28 2021-11-16 Nippon Shinyaku Co., Ltd. Antisense nucleic acids
DE102012103041A1 (en) 2012-04-10 2013-10-10 Eberhard-Karls-Universität Tübingen Universitätsklinikum New isolated antisense-oligonucleotide comprising sequence that is hybridized to messenger RNA-splicing-sequence of mutation-bearing exons of pre-messenger RNA of titin-gene and induces skipping of exons, used to treat heart disease
AU2013285698A1 (en) * 2012-07-03 2015-02-19 Biomarin Technologies B.V. Oligonucleotide for the treatment of muscular dystrophy patients
ES2791778T3 (en) 2012-08-01 2020-11-05 Ikaika Therapeutics Llc Mitigation of tissue damage and fibrosis through anti-LTBP4 antibodies
RU2730681C2 (en) 2014-03-12 2020-08-24 Ниппон Синяку Ко., Лтд. Antisense nucleic acids
WO2015171918A2 (en) 2014-05-07 2015-11-12 Louisiana State University And Agricultural And Mechanical College Compositions and uses for treatment thereof
EP3167064A1 (en) 2014-07-10 2017-05-17 Stichting Katholieke Universiteit Antisense oligonucleotides for the treatment of usher syndrome type 2
EP3180034B1 (en) * 2014-08-11 2022-04-20 The Board of Regents of The University of Texas System Prevention of muscular dystrophy by crispr/cas9-mediated gene editing
WO2016037163A1 (en) 2014-09-07 2016-03-10 Selecta Biosciences, Inc. Methods and compositions for attenuating gene therapy anti-viral transfer vector immune responses
WO2016089866A1 (en) 2014-12-01 2016-06-09 President And Fellows Of Harvard College Rna-guided systems for in vivo gene editing
GB201504124D0 (en) 2015-03-11 2015-04-22 Proqr Therapeutics B V Oligonucleotides
MA41795A (en) 2015-03-18 2018-01-23 Sarepta Therapeutics Inc EXCLUSION OF AN EXON INDUCED BY ANTISENSE COMPOUNDS IN MYOSTATIN
US11020417B2 (en) 2015-06-04 2021-06-01 Sarepta Therapeutics, Inc Methods and compounds for treatment of lymphocyte-related diseases and conditions
RS60493B1 (en) 2015-09-15 2020-08-31 Nippon Shinyaku Co Ltd Antisense nucleic acid
JP2018530560A (en) 2015-10-09 2018-10-18 サレプタ セラピューティクス, インコーポレイテッド Compositions and methods for the treatment of Duchenne muscular dystrophy and related disorders
WO2017136435A1 (en) 2016-02-01 2017-08-10 The Usa, As Represented By The Secretary, Department Of Health And Human Services Office Of Technology Transfer National Institute Of Health Compounds for modulating fc-epsilon-ri-beta expression and uses thereof
WO2017186739A1 (en) 2016-04-25 2017-11-02 Proqr Therapeutics Ii B.V. Oligonucleotides to treat eye disease
JP2019525742A (en) 2016-06-30 2019-09-12 サレプタ セラピューティクス, インコーポレイテッド Exon skipping oligomer for muscular dystrophy
SG10201607303YA (en) 2016-09-01 2018-04-27 Agency Science Tech & Res Antisense oligonucleotides to induce exon skipping
GB201616202D0 (en) 2016-09-23 2016-11-09 Proqr Therapeutics Ii Bv Antisense oligonucleotides for the treatment of eye deisease
US11236333B2 (en) 2016-12-13 2022-02-01 Stichting Katholieke Universiteit Antisense oligonucleotides for the treatment of Stargardt disease
BR112019012647A2 (en) 2016-12-19 2019-11-19 Sarepta Therapeutics Inc exon jump oligomer conjugates for muscular dystrophy
WO2018118627A1 (en) 2016-12-19 2018-06-28 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
AU2017382773A1 (en) 2016-12-19 2019-08-01 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
MX2020000676A (en) 2017-07-18 2021-05-14 Csl Behring Gene Therapy Inc Compositions and methods for treating beta-hemoglobinopathies.
GB201711809D0 (en) * 2017-07-21 2017-09-06 Governors Of The Univ Of Alberta Antisense oligonucleotide
BR122020005073A2 (en) 2017-08-04 2020-10-13 Skyhawk Therapeutics, Inc. COMPOUND, PHARMACEUTICAL COMPOSITION AND USE OF THE COMPOUND
CN111201325A (en) * 2017-08-11 2020-05-26 新加坡科技研究局 Methods for screening for splice variants or events
EA201991450A1 (en) 2017-09-22 2019-12-30 Сарепта Терапьютикс, Инк. OLIGOMER CONJUGATES FOR EXONISM SKIP IN MUSCULAR DYSTROPHY
JP2020536060A (en) 2017-09-28 2020-12-10 サレプタ セラピューティクス, インコーポレイテッド Combination therapy to treat muscular dystrophy
WO2019075360A1 (en) 2017-10-13 2019-04-18 Selecta Biosciences, Inc. Methods and compositions for attenuating anti-viral transfer vector igm responses
GB201803010D0 (en) 2018-02-26 2018-04-11 Royal Holloway & Bedford New College Neurodegenerative disorders
US10765760B2 (en) 2018-05-29 2020-09-08 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
WO2020015959A1 (en) 2018-07-19 2020-01-23 Stichting Katholieke Universiteit Antisense oligonucleotides rescue aberrant splicing of abca4.
WO2020028864A1 (en) 2018-08-02 2020-02-06 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating facioscapulohumeral muscular dystrophy
US11168141B2 (en) 2018-08-02 2021-11-09 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating dystrophinopathies
CA3108282A1 (en) 2018-08-02 2020-02-06 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating dystrophinopathies
JP2022510673A (en) 2018-12-04 2022-01-27 スティッチング カソリーケ ウニベルシテイト Antisense oligonucleotide rescues abnormal splicing of ABCA4
EP3897745A1 (en) 2018-12-23 2021-10-27 CSL Behring LLC Haematopoietic stem cell-gene therapy for wiskott-aldrich syndrome
CA3123045A1 (en) 2018-12-23 2020-07-02 Csl Behring L.L.C. Donor t-cells with kill switch
WO2020148400A1 (en) 2019-01-16 2020-07-23 Stichting Katholieke Universiteit Antisense oligonucleotides for use in the treatment of crpc
AU2020214704A1 (en) 2019-01-28 2021-08-26 Proqr Therapeutics Ii B.V. Antisense oligonucleotides for the treatment of leber's congenital amaurosis
KR20210135241A (en) 2019-02-05 2021-11-12 스카이호크 테라퓨틱스, 인코포레이티드 Methods and compositions for controlling splicing
WO2020163544A1 (en) 2019-02-06 2020-08-13 Skyhawk Therapeutics, Inc. Methods and compositions for modulating splicing
WO2020201144A1 (en) 2019-04-02 2020-10-08 Proqr Therapeutics Ii B.V. Antisense oligonucleotides for immunotherapy
AU2020259856A1 (en) 2019-04-18 2021-11-18 Proqr Therapeutics Ii B.V. Antisense oligonucleotides for the treatment of usher syndrome
BR112021023594A2 (en) 2019-05-28 2022-02-08 Selecta Biosciences Inc Methods and compositions for attenuated antiviral transfer vector immune response
WO2020254249A1 (en) 2019-06-21 2020-12-24 Proqr Therapeutics Ii B.V. Delivery of nucleic acids for the treatment of auditory disorders
JP2022543474A (en) 2019-08-08 2022-10-12 ユーシーエル ビジネス リミテッド Antisense oligonucleotides rescue ABCA4 aberrant splicing
EP4051794A1 (en) 2019-10-31 2022-09-07 Stichting Radboud universitair medisch centrum Allele-specific silencing therapy for dfna9 using antisense oligonucleotides
JPWO2021172498A1 (en) 2020-02-28 2021-09-02
CA3166720A1 (en) 2020-03-04 2021-09-10 Jim SWILDENS Antisense oligonucleotides for use in the treatment of usher syndrome
WO2021263070A1 (en) 2020-06-26 2021-12-30 Csl Behring Llc Donor t-cells with kill switch
WO2022090256A1 (en) 2020-10-26 2022-05-05 Proqr Therapeutics Ii B.V. Antisense oligonucleotides for the treatment of stargardt disease
CN112430645A (en) * 2020-12-09 2021-03-02 北京华瑞康源生物科技发展有限公司 Relative quantitative method and kit for detecting human DMD gene copy number by multiple real-time fluorescence PCR method
AU2022271873A1 (en) 2021-05-10 2024-01-04 Entrada Therapeutics, Inc. Compositions and methods for intracellular therapeutics
WO2022271818A1 (en) 2021-06-23 2022-12-29 Entrada Therapeutics, Inc. Antisense compounds and methods for targeting cug repeats
EP4359526A1 (en) 2021-06-25 2024-05-01 Stichting Radboud universitair medisch centrum Allele-specific silencing therapy for dfna21 using antisense oligonucleotides
US11771776B2 (en) 2021-07-09 2023-10-03 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating dystrophinopathies
US11638761B2 (en) 2021-07-09 2023-05-02 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating Facioscapulohumeral muscular dystrophy
WO2023064367A1 (en) 2021-10-12 2023-04-20 Selecta Biosciences, Inc. Methods and compositions for attenuating anti-viral transfer vector igm responses
EP4215614A1 (en) 2022-01-24 2023-07-26 Dynacure Combination therapy for dystrophin-related diseases
WO2023172624A1 (en) 2022-03-09 2023-09-14 Selecta Biosciences, Inc. Immunosuppressants in combination with anti-igm agents and related dosing
DE102022124232A1 (en) 2022-09-21 2024-03-21 Carl von Ossietzky Universität Oldenburg, Körperschaft des öffentlichen Rechts Antisense oligonucleotides for the treatment of Joubert syndrome
WO2024074668A1 (en) 2022-10-06 2024-04-11 Stichting Radboud Universitair Medisch Centrum Antisense oligonucleotides for treatment of usher 2a. exons 30-31
WO2024074670A1 (en) 2022-10-06 2024-04-11 Stichting Radboud Universitair Medisch Centrum Antisense oligonucleotides for treatment of usher 2a. exon 68

Citations (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5034506A (en) * 1985-03-15 1991-07-23 Anti-Gene Development Group Uncharged morpholino-based polymers having achiral intersubunit linkages
US5418139A (en) * 1993-02-10 1995-05-23 University Of Iowa Research Foundation Method for screening for cardiomyopathy
US5541308A (en) * 1986-11-24 1996-07-30 Gen-Probe Incorporated Nucleic acid probes for detection and/or quantitation of non-viral organisms
US5593974A (en) * 1991-06-28 1997-01-14 Massachusetts Institute Of Technology Localized oligonucleotide therapy
US5608046A (en) * 1990-07-27 1997-03-04 Isis Pharmaceuticals, Inc. Conjugated 4'-desmethyl nucleoside analog compounds
US5627263A (en) * 1993-11-24 1997-05-06 La Jolla Cancer Research Foundation Integrin-binding peptides
US5658764A (en) * 1992-01-28 1997-08-19 North Shore University Hospital Research Corp. Method and kits for detection of fragile X specific, GC-rich DNA sequences
US5741645A (en) * 1993-06-29 1998-04-21 Regents Of The University Of Minnesota Gene sequence for spinocerebellar ataxia type 1 and method for diagnosis
US5766847A (en) * 1988-10-11 1998-06-16 Max-Planck-Gesellschaft Zur Forderung Der Wissenschaften E.V. Process for analyzing length polymorphisms in DNA regions
US5853995A (en) * 1997-01-07 1998-12-29 Research Development Foundation Large scale genotyping of diseases and a diagnostic test for spinocerebellar ataxia type 6
US5869252A (en) * 1992-03-31 1999-02-09 Abbott Laboratories Method of multiplex ligase chain reaction
US5916808A (en) * 1993-05-11 1999-06-29 The University Of North Carolina At Chapel Hill Antisense oligonucleotides which combat aberrant splicing and methods of using the same
US5962332A (en) * 1994-03-17 1999-10-05 University Of Massachusetts Detection of trinucleotide repeats by in situ hybridization
US5968909A (en) * 1995-08-04 1999-10-19 Hybridon, Inc. Method of modulating gene expression with reduced immunostimulatory response
US6124100A (en) * 1998-02-26 2000-09-26 Samsung Fine Chemicals Co. Ltd. Diagnostic method and kit for neuropsychiatric diseases using trinucleotide repeats sequence
US6130207A (en) * 1997-11-05 2000-10-10 South Alabama Medical Science Foundation Cell-specific molecule and method for importing DNA into a nucleus
US6133031A (en) * 1999-08-19 2000-10-17 Isis Pharmaceuticals Inc. Antisense inhibition of focal adhesion kinase expression
US6172208B1 (en) * 1992-07-06 2001-01-09 Genzyme Corporation Oligonucleotides modified with conjugate groups
US6172216B1 (en) * 1998-10-07 2001-01-09 Isis Pharmaceuticals Inc. Antisense modulation of BCL-X expression
US6210892B1 (en) * 1998-10-07 2001-04-03 Isis Pharmaceuticals, Inc. Alteration of cellular behavior by antisense modulation of mRNA processing
US6251589B1 (en) * 1996-07-18 2001-06-26 Srl, Inc. Method for diagnosing spinocerebellar ataxia type 2 and primers therefor
US6280938B1 (en) * 1997-08-19 2001-08-28 Regents Of The University Of Minnesota SCA7 gene and method of use
US6300060B1 (en) * 1995-11-09 2001-10-09 Dana-Farber Cancer Institute, Inc. Method for predicting the risk of prostate cancer morbidity and mortality
US6322978B1 (en) * 1998-04-20 2001-11-27 Joslin Diabetes Center, Inc. Repeat polymorphism in the frataxin gene and uses therefore
US6329501B1 (en) * 1997-05-29 2001-12-11 Auburn University Methods and compositions for targeting compounds to muscle
US20010056077A1 (en) * 1999-05-21 2001-12-27 Jcr Pharmaceuticals Co., Ltd Pharmaceutical composition for treatment of duchenne muscular dystrophy
US6355481B1 (en) * 1999-06-18 2002-03-12 Emory University Hybridoma cell line and monoclonal antibody for huntingtin protein
US6355690B1 (en) * 1998-01-26 2002-03-12 Niigata University Remedy for CAG repeat expansion diseases
US6369038B1 (en) * 1991-04-25 2002-04-09 Genset Closed antisense and sense oligonucleotides and their applications
US20020049173A1 (en) * 1999-03-26 2002-04-25 Bennett C. Frank Alteration of cellular behavior by antisense modulation of mRNA processing
US6379698B1 (en) * 1999-04-06 2002-04-30 Isis Pharmaceuticals, Inc. Fusogenic lipids and vesicles
US20020055481A1 (en) * 2000-08-25 2002-05-09 Jcr Pharmaceuticals Co., Ltd. Pharmaceutical composition for treatment of Duchenne muscular dystrophy
US6399575B1 (en) * 1998-11-10 2002-06-04 Auburn University Methods and compositions for targeting compounds to the central nervous system
US20020165150A1 (en) * 1998-09-25 2002-11-07 Children's Medical Center Corporation Tissue remodeling
US6514755B1 (en) * 1998-08-18 2003-02-04 Regents Of The University Of Minnesota SCA7 gene and methods of use
US20030073215A1 (en) * 1997-03-31 2003-04-17 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030124523A1 (en) * 2000-06-22 2003-07-03 Asselbergs Fredericus Alphonsus Maria Organic compounds
US6653467B1 (en) * 2000-04-26 2003-11-25 Jcr Pharmaceutical Co., Ltd. Medicament for treatment of Duchenne muscular dystrophy
US20030234845A1 (en) * 2002-06-21 2003-12-25 Kazuyoshi Tominaga Ink jet head and ink jet recording apparatus
US20030236214A1 (en) * 1999-06-09 2003-12-25 Wolff Jon A. Charge reversal of polyion complexes and treatment of peripheral occlusive disease
US6670461B1 (en) * 1997-09-12 2003-12-30 Exiqon A/S Oligonucleotide analogues
US20040132684A1 (en) * 2000-05-09 2004-07-08 Sampath Umashanker Polymeric nucleoside prodrugs
US6794192B2 (en) * 2000-06-29 2004-09-21 Pfizer Inc. Target
US20040226056A1 (en) * 1998-12-22 2004-11-11 Myriad Genetics, Incorporated Compositions and methods for treating neurological disorders and diseases
US7189530B2 (en) * 1998-09-01 2007-03-13 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US7250404B2 (en) * 1989-03-21 2007-07-31 Vical Incorporated Lipid-mediated polynucleotide administration to deliver a biologically active peptide and to induce a cellular immune response
US7534879B2 (en) * 2003-03-21 2009-05-19 Academisch Ziekenhuis Leiden Modulation of exon recognition in pre-mRNA by interfering with the secondary RNA structure

Family Cites Families (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH07501204A (en) 1991-06-28 1995-02-09 マサチューセッツ インスティテュート オブ テクノロジー Topical oligonucleotide therapy
US5624803A (en) * 1993-10-14 1997-04-29 The Regents Of The University Of California In vivo oligonucleotide generator, and methods of testing the binding affinity of triplex forming oligonucleotides derived therefrom
US5854223A (en) 1995-10-06 1998-12-29 The Trustees Of Columbia University In The City Of New York S-DC28 as an antirestenosis agent after balloon injury
US7034009B2 (en) * 1995-10-26 2006-04-25 Sirna Therapeutics, Inc. Enzymatic nucleic acid-mediated treatment of ocular diseases or conditions related to levels of vascular endothelial growth factor receptor (VEGF-R)
TR199801581T2 (en) 1996-02-14 1998-10-21 Isis Pharmaceuticals, Inc. �ekerle de�i�tirilmi� bo�luklu oligon�kleotid’ler.
JP4627369B2 (en) * 1998-06-10 2011-02-09 バイオグノスティック ゲゼルシャフト フュア バイオモレキュラー ダイアグノスティック ミット ベシュレンクテル ハフツング How to stimulate the immune system
JP2000125448A (en) 1998-10-14 2000-04-28 Yazaki Corp Electrical junction box
JP2000256547A (en) 1999-03-10 2000-09-19 Sumitomo Dow Ltd Resin composition for heat-resistant card
EP1133993A1 (en) 2000-03-10 2001-09-19 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Substances for the treatment of spinal muscular atrophy
JP4836366B2 (en) * 2000-08-25 2011-12-14 雅文 松尾 Duchenne muscular dystrophy treatment
EP1191097A1 (en) * 2000-09-21 2002-03-27 Leids Universitair Medisch Centrum Induction of exon skipping in eukaryotic cells
EP4272748A3 (en) * 2004-06-28 2024-03-27 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
EP2203173B1 (en) * 2007-10-26 2015-12-23 Academisch Ziekenhuis Leiden Means and methods for counteracting muscle disorders
US8084601B2 (en) * 2008-09-11 2011-12-27 Royal Holloway And Bedford New College Royal Holloway, University Of London Oligomers
JP6141018B2 (en) * 2009-12-24 2017-06-07 バイオマリン テクノロジーズ ベー.フェー. Molecules for treating inflammatory disorders
CN104203289B (en) * 2012-01-27 2020-11-03 比奥马林技术公司 RNA-regulatory oligonucleotides with improved properties for the treatment of duchenne muscular dystrophy and becker muscular dystrophy
JP5794194B2 (en) 2012-04-19 2015-10-14 東京エレクトロン株式会社 Substrate processing equipment

Patent Citations (54)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5034506A (en) * 1985-03-15 1991-07-23 Anti-Gene Development Group Uncharged morpholino-based polymers having achiral intersubunit linkages
US5541308A (en) * 1986-11-24 1996-07-30 Gen-Probe Incorporated Nucleic acid probes for detection and/or quantitation of non-viral organisms
US5766847A (en) * 1988-10-11 1998-06-16 Max-Planck-Gesellschaft Zur Forderung Der Wissenschaften E.V. Process for analyzing length polymorphisms in DNA regions
US7250404B2 (en) * 1989-03-21 2007-07-31 Vical Incorporated Lipid-mediated polynucleotide administration to deliver a biologically active peptide and to induce a cellular immune response
US5608046A (en) * 1990-07-27 1997-03-04 Isis Pharmaceuticals, Inc. Conjugated 4'-desmethyl nucleoside analog compounds
US6369038B1 (en) * 1991-04-25 2002-04-09 Genset Closed antisense and sense oligonucleotides and their applications
US5593974A (en) * 1991-06-28 1997-01-14 Massachusetts Institute Of Technology Localized oligonucleotide therapy
US5658764A (en) * 1992-01-28 1997-08-19 North Shore University Hospital Research Corp. Method and kits for detection of fragile X specific, GC-rich DNA sequences
US5869252A (en) * 1992-03-31 1999-02-09 Abbott Laboratories Method of multiplex ligase chain reaction
US6172208B1 (en) * 1992-07-06 2001-01-09 Genzyme Corporation Oligonucleotides modified with conjugate groups
US5418139A (en) * 1993-02-10 1995-05-23 University Of Iowa Research Foundation Method for screening for cardiomyopathy
US5976879A (en) * 1993-05-11 1999-11-02 The University Of North Carolina At Chapel Hill Antisense oligonucleotides which combat aberrant splicing and methods of using the same
US5916808A (en) * 1993-05-11 1999-06-29 The University Of North Carolina At Chapel Hill Antisense oligonucleotides which combat aberrant splicing and methods of using the same
US5741645A (en) * 1993-06-29 1998-04-21 Regents Of The University Of Minnesota Gene sequence for spinocerebellar ataxia type 1 and method for diagnosis
US5627263A (en) * 1993-11-24 1997-05-06 La Jolla Cancer Research Foundation Integrin-binding peptides
US5962332A (en) * 1994-03-17 1999-10-05 University Of Massachusetts Detection of trinucleotide repeats by in situ hybridization
US5968909A (en) * 1995-08-04 1999-10-19 Hybridon, Inc. Method of modulating gene expression with reduced immunostimulatory response
US6300060B1 (en) * 1995-11-09 2001-10-09 Dana-Farber Cancer Institute, Inc. Method for predicting the risk of prostate cancer morbidity and mortality
US6251589B1 (en) * 1996-07-18 2001-06-26 Srl, Inc. Method for diagnosing spinocerebellar ataxia type 2 and primers therefor
US5853995A (en) * 1997-01-07 1998-12-29 Research Development Foundation Large scale genotyping of diseases and a diagnostic test for spinocerebellar ataxia type 6
US20030073215A1 (en) * 1997-03-31 2003-04-17 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030082763A1 (en) * 1997-03-31 2003-05-01 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030082766A1 (en) * 1997-03-31 2003-05-01 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US6329501B1 (en) * 1997-05-29 2001-12-11 Auburn University Methods and compositions for targeting compounds to muscle
US7118893B2 (en) * 1997-08-19 2006-10-10 Regents Of The University Of Minnesota SCA7 gene and methods of use
US6280938B1 (en) * 1997-08-19 2001-08-28 Regents Of The University Of Minnesota SCA7 gene and method of use
US6670461B1 (en) * 1997-09-12 2003-12-30 Exiqon A/S Oligonucleotide analogues
US6130207A (en) * 1997-11-05 2000-10-10 South Alabama Medical Science Foundation Cell-specific molecule and method for importing DNA into a nucleus
US6355690B1 (en) * 1998-01-26 2002-03-12 Niigata University Remedy for CAG repeat expansion diseases
US6124100A (en) * 1998-02-26 2000-09-26 Samsung Fine Chemicals Co. Ltd. Diagnostic method and kit for neuropsychiatric diseases using trinucleotide repeats sequence
US6322978B1 (en) * 1998-04-20 2001-11-27 Joslin Diabetes Center, Inc. Repeat polymorphism in the frataxin gene and uses therefore
US6514755B1 (en) * 1998-08-18 2003-02-04 Regents Of The University Of Minnesota SCA7 gene and methods of use
US7189530B2 (en) * 1998-09-01 2007-03-13 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20020165150A1 (en) * 1998-09-25 2002-11-07 Children's Medical Center Corporation Tissue remodeling
US6210892B1 (en) * 1998-10-07 2001-04-03 Isis Pharmaceuticals, Inc. Alteration of cellular behavior by antisense modulation of mRNA processing
US6172216B1 (en) * 1998-10-07 2001-01-09 Isis Pharmaceuticals Inc. Antisense modulation of BCL-X expression
US6399575B1 (en) * 1998-11-10 2002-06-04 Auburn University Methods and compositions for targeting compounds to the central nervous system
US20040226056A1 (en) * 1998-12-22 2004-11-11 Myriad Genetics, Incorporated Compositions and methods for treating neurological disorders and diseases
US20020049173A1 (en) * 1999-03-26 2002-04-25 Bennett C. Frank Alteration of cellular behavior by antisense modulation of mRNA processing
US6379698B1 (en) * 1999-04-06 2002-04-30 Isis Pharmaceuticals, Inc. Fusogenic lipids and vesicles
US20010056077A1 (en) * 1999-05-21 2001-12-27 Jcr Pharmaceuticals Co., Ltd Pharmaceutical composition for treatment of duchenne muscular dystrophy
US6653466B2 (en) * 1999-05-21 2003-11-25 Jcr Pharmaceuticals Co., Ltd. Pharmaceutical composition for treatment of duchenne muscular dystrophy
US20030236214A1 (en) * 1999-06-09 2003-12-25 Wolff Jon A. Charge reversal of polyion complexes and treatment of peripheral occlusive disease
US6355481B1 (en) * 1999-06-18 2002-03-12 Emory University Hybridoma cell line and monoclonal antibody for huntingtin protein
US6133031A (en) * 1999-08-19 2000-10-17 Isis Pharmaceuticals Inc. Antisense inhibition of focal adhesion kinase expression
US6653467B1 (en) * 2000-04-26 2003-11-25 Jcr Pharmaceutical Co., Ltd. Medicament for treatment of Duchenne muscular dystrophy
US20040132684A1 (en) * 2000-05-09 2004-07-08 Sampath Umashanker Polymeric nucleoside prodrugs
US20100081627A1 (en) * 2000-05-09 2010-04-01 Reliable Biopharmaceutical, Inc. Polymeric nucleoside prodrugs
US20030124523A1 (en) * 2000-06-22 2003-07-03 Asselbergs Fredericus Alphonsus Maria Organic compounds
US6794192B2 (en) * 2000-06-29 2004-09-21 Pfizer Inc. Target
US20020055481A1 (en) * 2000-08-25 2002-05-09 Jcr Pharmaceuticals Co., Ltd. Pharmaceutical composition for treatment of Duchenne muscular dystrophy
US6727355B2 (en) * 2000-08-25 2004-04-27 Jcr Pharmaceuticals Co., Ltd. Pharmaceutical composition for treatment of Duchenne muscular dystrophy
US20030234845A1 (en) * 2002-06-21 2003-12-25 Kazuyoshi Tominaga Ink jet head and ink jet recording apparatus
US7534879B2 (en) * 2003-03-21 2009-05-19 Academisch Ziekenhuis Leiden Modulation of exon recognition in pre-mRNA by interfering with the secondary RNA structure

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Habara et al (J Med Genet 2009;46:542-547) *
Jou et al (HUMAN MUTATION 5:86-93 (1995)) *
Sertic et al (Coll. Atropol. 21: 151-156, 1997) *

Cited By (85)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8624019B2 (en) 2002-11-25 2014-01-07 Masafumi Matsuo ENA nucleic acid drugs modifying splicing in mRNA precursor
US9657049B2 (en) 2002-11-25 2017-05-23 Masafumi Matsuo ENA nucleic acid pharmaceuticals capable of modifying splicing of mRNA precursors
US20110046360A1 (en) * 2002-11-25 2011-02-24 Masafumi Matsuo ENA NUCLEIC ACID DRUGS MODIFYING SPLICING IN mRNA PRECURSOR
US9657050B2 (en) 2002-11-25 2017-05-23 Masafumi Matsuo ENA nucleic acid pharmaceuticals capable of modifying splicing of mRNA precursors
US9243026B2 (en) 2002-11-25 2016-01-26 Daiichi Sankyo Company, Limited ENA nucleic acid pharmaceuticals capable of modifying splicing of mRNA precursors
US10781451B2 (en) 2004-06-28 2020-09-22 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
US10227590B2 (en) 2004-06-28 2019-03-12 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
US8476423B2 (en) 2004-06-28 2013-07-02 The University of Western Austrailia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
US8486907B2 (en) 2004-06-28 2013-07-16 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
US8524880B2 (en) 2004-06-28 2013-09-03 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
US8455634B2 (en) 2004-06-28 2013-06-04 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
US9605262B2 (en) 2004-06-28 2017-03-28 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
USRE47691E1 (en) 2004-06-28 2019-11-05 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
USRE47751E1 (en) 2004-06-28 2019-12-03 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
US10266827B2 (en) 2004-06-28 2019-04-23 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
US9018368B2 (en) 2004-06-28 2015-04-28 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
US9024007B2 (en) 2004-06-28 2015-05-05 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
US9035040B2 (en) 2004-06-28 2015-05-19 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
US9175286B2 (en) 2004-06-28 2015-11-03 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
US8455636B2 (en) 2004-06-28 2013-06-04 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
US8450474B2 (en) 2004-06-28 2013-05-28 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
USRE47769E1 (en) 2004-06-28 2019-12-17 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
US8455635B2 (en) 2004-06-28 2013-06-04 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
US9249416B2 (en) 2004-06-28 2016-02-02 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
US9422555B2 (en) 2004-06-28 2016-08-23 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
US10995337B2 (en) 2004-06-28 2021-05-04 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
US9441229B2 (en) 2004-06-28 2016-09-13 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
US20110015258A1 (en) * 2004-06-28 2011-01-20 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
US10421966B2 (en) 2004-06-28 2019-09-24 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
US9447415B2 (en) 2004-06-28 2016-09-20 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
US9994851B2 (en) 2004-06-28 2018-06-12 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
USRE48960E1 (en) 2004-06-28 2022-03-08 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
US10968450B2 (en) 2004-06-28 2021-04-06 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
US9890379B2 (en) 2006-08-11 2018-02-13 Biomarin Technologies B.V. Treatment of genetic disorders associated with DNA repeat instability
US11274299B2 (en) 2006-08-11 2022-03-15 Vico Therapeutics B.V. Methods and means for treating DNA repeat instability associated genetic disorders
US10689646B2 (en) 2006-08-11 2020-06-23 Biomarin Technologies B.V. Treatment of genetic disorders associated with DNA repeat instability
US11427820B2 (en) 2007-10-26 2022-08-30 Biomarin Technologies B.V. Methods and means for efficient skipping of exon 45 in Duchenne muscular dystrophy pre-mRNA
US10876114B2 (en) 2007-10-26 2020-12-29 Biomarin Technologies B.V. Methods and means for efficient skipping of at least one of the following exons of the human Duchenne muscular dystrophy gene: 43, 46, 50-53
US10246707B2 (en) 2008-05-14 2019-04-02 Biomarin Technologies B.V. Method for efficient exon (44) skipping in duchenne muscular dystrophy and associated means
US9434948B2 (en) 2008-10-24 2016-09-06 Sarepta Therapeutics, Inc. Multiple exon skipping compositions for DMD
US9447416B2 (en) 2008-10-24 2016-09-20 Sarepta Therapeutics, Inc. Multiple exon skipping compositions for DMD
US9453225B2 (en) 2008-10-24 2016-09-27 Sarepta Therapeutics, Inc. Multiple exon skipping compositions for DMD
US20100130591A1 (en) * 2008-10-24 2010-05-27 Peter Sazani Multiple exon skipping compositions for dmd
US9447417B2 (en) 2008-10-24 2016-09-20 Sarepta Therapeutics, Inc. Multiple exon skipping compositions for DMD
US8865883B2 (en) 2008-10-24 2014-10-21 Sarepta Therapeutics, Inc. Multiple exon skipping compositions for DMD
US9234198B1 (en) 2008-10-24 2016-01-12 Sarepta Therapeutics, Inc. Multiple exon skipping compositions for DMD
US8871918B2 (en) 2008-10-24 2014-10-28 Sarepta Therapeutics, Inc. Multiple exon skipping compositions for DMD
US9695211B2 (en) 2008-12-02 2017-07-04 Wave Life Sciences Japan, Inc. Method for the synthesis of phosphorus atom modified nucleic acids
US10329318B2 (en) 2008-12-02 2019-06-25 Wave Life Sciences Ltd. Method for the synthesis of phosphorus atom modified nucleic acids
US9744183B2 (en) 2009-07-06 2017-08-29 Wave Life Sciences Ltd. Nucleic acid prodrugs and methods of use thereof
US10307434B2 (en) 2009-07-06 2019-06-04 Wave Life Sciences Ltd. Nucleic acid prodrugs and methods of use thereof
US10781450B2 (en) 2009-11-12 2020-09-22 Sarepta Therapeutics, Inc. Antisense molecules and methods for treating pathologies
US11447776B2 (en) 2009-11-12 2022-09-20 The University Of Western Australia Antisense molecules and methods for treating pathologies
US9228187B2 (en) 2009-11-12 2016-01-05 The University Of Western Australia Antisense molecules and methods for treating pathologies
US10287586B2 (en) 2009-11-12 2019-05-14 The University Of Western Australia Antisense molecules and methods for treating pathologies
US8637483B2 (en) 2009-11-12 2014-01-28 The University Of Western Australia Antisense molecules and methods for treating pathologies
US9758783B2 (en) 2009-11-12 2017-09-12 The University Of Western Australia Antisense molecules and methods for treating pathologies
US10590421B2 (en) 2010-08-05 2020-03-17 Academisch Ziekenhuis Leiden H.O.D.N. Lumc Antisense oligonucleotide directed removal of proteolytic cleavage sites, the HCHWA-D mutation, and trinucleotide repeat expansions
US9611471B2 (en) 2010-08-05 2017-04-04 Academisch Ziekenhuis Leiden Antisense oligonucleotide directed removal of proteolytic cleavage sites from proteins
US10364432B2 (en) 2010-08-05 2019-07-30 Academisch Ziekenhuis Leiden H.O.D.N. Lumc Antisense oligonucleotide directed removal of proteolytic cleavage sites from proteins
US10428019B2 (en) 2010-09-24 2019-10-01 Wave Life Sciences Ltd. Chiral auxiliaries
US10280192B2 (en) 2011-07-19 2019-05-07 Wave Life Sciences Ltd. Methods for the synthesis of functionalized nucleic acids
US9605019B2 (en) 2011-07-19 2017-03-28 Wave Life Sciences Ltd. Methods for the synthesis of functionalized nucleic acids
US10913946B2 (en) 2012-01-27 2021-02-09 Biomarin Technologies B.V. RNA modulating oligonucleotides with improved characteristics for the treatment of Duchenne and Becker muscular dystrophy
US10179912B2 (en) 2012-01-27 2019-01-15 Biomarin Technologies B.V. RNA modulating oligonucleotides with improved characteristics for the treatment of duchenne and becker muscular dystrophy
US10590413B2 (en) 2012-07-13 2020-03-17 Wave Life Sciences Ltd. Chiral control
US9982257B2 (en) 2012-07-13 2018-05-29 Wave Life Sciences Ltd. Chiral control
US9617547B2 (en) 2012-07-13 2017-04-11 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant
US10167309B2 (en) 2012-07-13 2019-01-01 Wave Life Sciences Ltd. Asymmetric auxiliary group
US9217148B2 (en) 2013-03-14 2015-12-22 Sarepta Therapeutics, Inc. Exon skipping compositions for treating muscular dystrophy
US11932851B2 (en) 2013-03-14 2024-03-19 Sarepta Therapeutics, Inc. Exon skipping compositions for treating muscular dystrophy
US10907154B2 (en) 2013-03-14 2021-02-02 Sarepta Therapeutics, Inc. Exon skipping compositions for treating muscular dystrophy
US9506058B2 (en) 2013-03-15 2016-11-29 Sarepta Therapeutics, Inc. Compositions for treating muscular dystrophy
US10364431B2 (en) 2013-03-15 2019-07-30 Sarepta Therapeutics, Inc. Compositions for treating muscular dystrophy
US10337003B2 (en) 2013-03-15 2019-07-02 Sarepta Therapeutics, Inc. Compositions for treating muscular dystrophy
EP2796425A1 (en) 2013-04-24 2014-10-29 Corning Incorporated Delamination resistant pharmaceutical glass containers containing active pharmaceutical ingredients
US10144933B2 (en) 2014-01-15 2018-12-04 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having immunity induction activity, and immunity induction activator
US10149905B2 (en) 2014-01-15 2018-12-11 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having antitumor effect and antitumor agent
US10322173B2 (en) 2014-01-15 2019-06-18 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having anti-allergic activity, and anti-allergic agent
US10160969B2 (en) 2014-01-16 2018-12-25 Wave Life Sciences Ltd. Chiral design
US10570380B2 (en) 2014-01-24 2020-02-25 Am-Pharma B.V. Downstream processing of an alkaline phosphatase
US9926544B2 (en) 2014-01-24 2018-03-27 Am-Pharma B.V. Chimeric alkaline phosphatase-like proteins
US10822597B2 (en) 2014-01-24 2020-11-03 Am-Pharma B.V. Chimeric alkaline phosphatase-like proteins
US11746340B2 (en) 2014-01-24 2023-09-05 Am-Pharma B.V. Chimeric alkaline phosphatase-like proteins
US10450568B2 (en) 2015-10-09 2019-10-22 Wave Life Sciences Ltd. Oligonucleotide compositions and methods thereof

Also Published As

Publication number Publication date
EP2594641A1 (en) 2013-05-22
LT2284264T (en) 2017-03-10
CA2423044A1 (en) 2002-03-28
PT1619249E (en) 2009-01-07
EP2602322A1 (en) 2013-06-12
JP2004509622A (en) 2004-04-02
DK2602322T3 (en) 2016-04-18
EP2636740B1 (en) 2016-11-09
EP2801618B1 (en) 2017-04-19
JP2017200952A (en) 2017-11-09
EP2636740A1 (en) 2013-09-11
DK2801618T3 (en) 2017-07-24
JP2014065733A (en) 2014-04-17
CY1117852T1 (en) 2017-05-17
ES2567417T3 (en) 2016-04-22
JP6126983B2 (en) 2017-05-10
ES2629747T3 (en) 2017-08-14
PT2636741E (en) 2016-06-09
EP2594642B1 (en) 2015-12-30
HK1203554A1 (en) 2015-10-30
EP2940139B1 (en) 2018-07-04
ES2315788T5 (en) 2020-09-17
EP3382021A1 (en) 2018-10-03
PT2801618T (en) 2017-07-25
EP2602322B1 (en) 2016-03-02
LT2940139T (en) 2018-09-10
AU1106202A (en) 2002-04-02
EP2594642A1 (en) 2013-05-22
HK1184818A1 (en) 2014-01-30
EP1320597A2 (en) 2003-06-25
US20150361424A1 (en) 2015-12-17
EP1619249B1 (en) 2008-09-24
EP2801618A1 (en) 2014-11-12
ES2561293T3 (en) 2016-02-25
CY1117521T1 (en) 2017-04-26
CY1117522T1 (en) 2017-04-26
AU2011201325A1 (en) 2011-04-14
EP2594641B1 (en) 2015-12-30
TR201810606T4 (en) 2018-08-27
CA2423044C (en) 2016-08-23
DK2636742T3 (en) 2017-07-24
PT2594640E (en) 2016-03-11
JP6425775B2 (en) 2018-11-21
EP2636742A1 (en) 2013-09-11
AU2007234488A1 (en) 2007-12-06
JP4846965B2 (en) 2011-12-28
DK1619249T3 (en) 2009-02-02
US7973015B2 (en) 2011-07-05
EP2284264B1 (en) 2016-12-14
HK1216906A1 (en) 2016-12-09
ATE409224T2 (en) 2008-10-15
JP2016104795A (en) 2016-06-09
US20150322434A1 (en) 2015-11-12
EP2636741B1 (en) 2016-04-27
US20090228998A1 (en) 2009-09-10
AU2011201325B2 (en) 2011-12-22
HK1188250A1 (en) 2014-04-25
DE60135936D1 (en) 2008-11-06
DK1619249T4 (en) 2020-04-06
EP2594640B1 (en) 2015-12-30
CY1109601T1 (en) 2014-08-13
NZ524853A (en) 2004-11-26
CY1117318T1 (en) 2017-04-26
WO2002024906A9 (en) 2003-01-23
CY1121139T1 (en) 2020-05-29
PT2940139T (en) 2018-10-15
EP2940139A1 (en) 2015-11-04
CY1119026T1 (en) 2018-01-10
EP1191097A1 (en) 2002-03-27
ES2561292T3 (en) 2016-02-25
HK1188249A1 (en) 2014-04-25
CY1119088T1 (en) 2018-01-10
DK2594640T3 (en) 2016-03-21
ES2315788T3 (en) 2009-04-01
SI2801618T1 (en) 2017-07-31
DK2594642T3 (en) 2016-03-21
AU2007234488B2 (en) 2011-02-17
US20140350076A1 (en) 2014-11-27
DK2284264T3 (en) 2017-03-13
DK2636741T3 (en) 2016-07-04
US20030235845A1 (en) 2003-12-25
JP6511124B2 (en) 2019-05-15
LT2801618T (en) 2017-09-11
CY1118972T1 (en) 2018-01-10
JP6250078B2 (en) 2017-12-20
EP2594640A1 (en) 2013-05-22
ES2561294T3 (en) 2016-02-25
CY1117887T1 (en) 2017-05-17
ES2690049T3 (en) 2018-11-19
EP1619249A1 (en) 2006-01-25
HK1184820A1 (en) 2014-01-30
HK1184819A1 (en) 2014-01-30
DK2940139T3 (en) 2018-10-08
ES2628349T3 (en) 2017-08-02
WO2002024906A1 (en) 2002-03-28
DK2594641T3 (en) 2016-03-21
PT2284264T (en) 2017-03-06
EP2284264A1 (en) 2011-02-16
JP2019073555A (en) 2019-05-16
ES2610568T3 (en) 2017-04-28
PT2636742T (en) 2017-06-02
ES2581285T3 (en) 2016-09-05
EP2636741A1 (en) 2013-09-11
HK1184821A1 (en) 2014-01-30
AU2002211062C1 (en) 2011-02-03
JP2018052981A (en) 2018-04-05
EP1619249B2 (en) 2020-01-08
JP2011200235A (en) 2011-10-13
ES2609421T3 (en) 2017-04-20
AU2002211062B2 (en) 2007-11-29
EP2636742B1 (en) 2017-04-19

Similar Documents

Publication Publication Date Title
US7973015B2 (en) Induction of exon skipping in eukaryotic cells
AU2002211062A1 (en) Induction of exon skipping in eukaryotic cells
AU2012200761B2 (en) Induction of exon skipping in eukaryotic cells

Legal Events

Date Code Title Description
AS Assignment

Owner name: ACADEMISCH ZIEKENHUIS LEIDEN,NETHERLANDS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:VAN OMMEN, GARRIT-JAN B.;VAN DEUTEKOM, JUDITH C.T.;DEN DUNNEN, JOHANNES T.;REEL/FRAME:020124/0946

Effective date: 20030414

AS Assignment

Owner name: ACADEMISCH ZIEKENHUIS LEIDEN H.O.D.N. LUMC, NETHER

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:VAN OMMEN, GARRIT-JAN B.;VAN DEUTEKOM, JUDITH C.;DEN DUNNEN, JOHANNES T.;REEL/FRAME:023733/0740;SIGNING DATES FROM 20091201 TO 20100105

Owner name: ACADEMISCH ZIEKENHUIS LEIDEN H.O.D.N. LUMC,NETHERL

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:VAN OMMEN, GARRIT-JAN B.;VAN DEUTEKOM, JUDITH C.;DEN DUNNEN, JOHANNES T.;SIGNING DATES FROM 20091201 TO 20100105;REEL/FRAME:023733/0740

STCB Information on status: application discontinuation

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