US20170044534A1 - 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 - Google Patents

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 Download PDF

Info

Publication number
US20170044534A1
US20170044534A1 US15/289,053 US201615289053A US2017044534A1 US 20170044534 A1 US20170044534 A1 US 20170044534A1 US 201615289053 A US201615289053 A US 201615289053A US 2017044534 A1 US2017044534 A1 US 2017044534A1
Authority
US
United States
Prior art keywords
seq
exon
oligonucleotide
skipping
backbone
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
US15/289,053
Inventor
Judith Christina Theodora Van Deutekom
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.)
Academisch Ziekenhuis Leiden (azl)
Leids Universitair Medisch Centrum LUMC
Biomarin Technologies BV
Original Assignee
Academisch Ziekenhuis Leiden (azl)
Leids Universitair Medisch Centrum LUMC
Biomarin Technologies BV
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=39045623&utm_source=***_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20170044534(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from PCT/NL2009/050113 external-priority patent/WO2010050802A2/en
Application filed by Academisch Ziekenhuis Leiden (azl), Leids Universitair Medisch Centrum LUMC, Biomarin Technologies BV filed Critical Academisch Ziekenhuis Leiden (azl)
Priority to US15/289,053 priority Critical patent/US20170044534A1/en
Assigned to ACADEMISCH ZIEKENHUIS LEIDEN (AZL), BIOMARIN TECHNOLOGIES B.V. reassignment ACADEMISCH ZIEKENHUIS LEIDEN (AZL) ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: VAN DEUTEKOM, JUDITH CHRISTINA THEODORA
Publication of US20170044534A1 publication Critical patent/US20170044534A1/en
Priority to US16/024,558 priority patent/US10876114B2/en
Priority to US17/129,117 priority patent/US20210139904A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • A61K31/522Purines, e.g. adenine having oxo groups directly attached to the heterocyclic ring, e.g. hypoxanthine, guanine, acyclovir
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/57Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/57Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone
    • A61K31/573Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone substituted in position 21, e.g. cortisone, dexamethasone, prednisone or aldosterone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/58Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids containing heterocyclic rings, e.g. danazol, stanozolol, pancuronium or digitogenin
    • 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
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • A61K38/1719Muscle proteins, e.g. myosin or actin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • 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
    • A61K48/0058Nucleic acids adapted for tissue specific expression, e.g. having tissue specific promoters as part of a contruct
    • 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
    • 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
    • A61P21/02Muscle relaxants, e.g. for tetanus or cramps
    • 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
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/12Drugs for disorders of the metabolism for electrolyte homeostasis
    • A61P3/14Drugs for disorders of the metabolism for electrolyte homeostasis for calcium homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/06Free radical scavengers or antioxidants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • 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
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • C12N2310/111Antisense spanning the whole gene, or a large part of it
    • 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
    • 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
    • C12N2310/313Phosphorodithioates
    • 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
    • C12N2310/314Phosphoramidates
    • 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
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/318Chemical structure of the backbone where the PO2 is completely replaced, e.g. MMI or formacetal
    • C12N2310/3181Peptide nucleic acid, PNA
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3212'-O-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/323Chemical structure of the sugar modified ring structure
    • C12N2310/3231Chemical structure of the sugar modified ring structure having an additional ring, e.g. LNA, ENA
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/323Chemical structure of the sugar modified ring structure
    • C12N2310/3233Morpholino-type ring
    • 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
    • C12N2320/31Combination 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
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/33Alteration of splicing

Definitions

  • the invention relates to the field of genetics, more specifically human genetics.
  • the invention in particular relates to modulation of splicing of the human Duchenne Muscular Dystrophy pre-mRNA.
  • the invention relates to the field of genetics, more specifically human genetics.
  • the invention in particular relates to modulation of splicing of the human Duchenne Muscular Dystrophy pre-mRNA.
  • Myopathies are disorders that result in functional impairment of muscles.
  • Muscular dystrophy refers to genetic diseases that are characterized by progressive weakness and degeneration of skeletal muscles.
  • Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD) are the most common childhood forms of muscular dystrophy. They are recessive disorders and because the gene responsible for DMD and BMD resides on the X-chromosome, mutations mainly affect males with an incidence of about 1 in 3500 boys.
  • DMD and BMD are caused by genetic defects in the DMD gene encoding dystrophin, a muscle protein that is required for interactions between the cytoskeleton and the extracellular matrix to maintain muscle fiber stability during contraction.
  • DMD is a severe, lethal neuromuscular disorder resulting in a dependency on wheelchair support before the age of 12 and DMD patients often die before the age of thirty due to respiratory- or heart failure. In contrast, BMD patients often remain ambulatory until later in life, and have near normal life expectancies.
  • DMD mutations in the DMD gene are characterized by frame shifting insertions or deletions or nonsense point mutations, resulting in the absence of functional dystrophin. BMD mutations in general keep the reading frame intact, allowing synthesis of a partly functional dystrophin.
  • DMD Duchenne muscular dystrophy
  • AONs antisense oligonucleotides interfering with splicing signals the skipping of specific exons can be induced in the DMD pre-mRNA, thus restoring the open reading frame and converting the severe DMD into a milder BMD phenotype (van Deutekom et al. Hum Mol Genet. 2001; 10: 1547-54; Aartsma-Rus et al., Hum Mol Genet 2003; 12(8):907-14.). In vivo proof-of-concept was first obtained in the mdx mouse model, which is dystrophin-deficient due to a nonsense mutation in exon 23.
  • Intramuscular and intravenous injections of AONs targeting the mutated exon 23 restored dystrophin expression for at least three months (Lu et al. Nat Med. 2003; 8: 1009-14; Lu et al., Proc Natl Acad Sci U S A. 2005;102(1):198-203). This was accompanied by restoration of dystrophin-associated proteins at the fiber membrane as well as functional improvement of the treated muscle.
  • In vivo skipping of human exons has also been achieved in the hDMD mouse model, which contains a complete copy of the human DMD gene integrated in chromosome 5 of the mouse (Bremmer-Bout et al. Molecular Therapy. 2004; 10: 232-40; 't Hoen et al. J Biol Chem. 2008; 283: 5899-907).
  • the present invention provides a method for inducing, and/or promoting skipping of at least one of exons 43, 46, 50-53 of the DMD pre-mRNA in a patient, preferably in an isolated cell of a patient, the method comprising providing said cell and/or said patient with a molecule that binds to a continuous stretch of at least 8 nucleotides within said exon. It is to be understood that said method encompasses an in vitro, in vivo or ex vivo method.
  • a method for inducing and/or promoting skipping of at least one of exons 43, 46, 50-53 of DMD pre-mRNA in a patient, preferably in an isolated cell of said patient, the method comprising providing said cell and/or said patient with a molecule that binds to a continuous stretch of at least 8 nucleotides within said exon.
  • a DMD pre-mRNA preferably means the pre-mRNA of a DMD gene of a DMD or BMD patient.
  • a patient is preferably intended to mean a patient having DMD or BMD as later defined herein or a patient susceptible to develop DMD or BMD due to his or her genetic background.
  • an oligonucleotide used will preferably correct one mutation as present in the DMD gene of said patient and therefore will preferably create a DMD protein that will look like a BMD protein: said protein will preferably be a functional dystrophin as later defined herein.
  • an oligonucleotide as used will preferably correct one mutation as present in the BMD gene of said patient and therefore will preferably create a dystrophin which will be more functional than the dystrophin which was originally present in said BMD patient.
  • Exon skipping refers to the induction in a cell of a mature mRNA that does not contain a particular exon that is normally present therein. Exon skipping is performed by providing a cell expressing the pre-mRNA of said mRNA with a molecule capable of interfering with essential sequences such as for example the splice donor of splice acceptor sequence that required for splicing of said exon, or a molecule that is capable of interfering with an exon inclusion signal that is required for recognition of a stretch of nucleotides as an exon to be included in the mRNA.
  • the term pre-mRNA refers to a non-processed or partly processed precursor mRNA that is synthesized from a DNA template in the cell nucleus by transcription.
  • inducing and/or promoting skipping of an exon as indicated herein means that at least 1%, 10%, 20%, 30%, 40%,50%, 60%, 70%, 80%, 90% or more of the DMD mRNA in one or more (muscle) cells of a treated patient will not contain said exon. This is preferably assessed by PCR as described in the examples.
  • a method of the invention by inducing and/or promoting skipping of at least one of the following exons 43, 46, 50-53 of the DMD pre-mRNA in one or more (muscle) cells of a patient, provides said patient with a functional dystrophin protein and/or decreases the production of an aberrant dystrophin protein in said patient and/or increases the production of a functional dystrophin is said patient.
  • Providing a patient with a functional dystrophin protein and/or decreasing the production of an aberrant dystrophin protein in said patient is typically applied in a DMD patient.
  • Increasing the production of a functional dystrophin is typically applied in a BMD patient.
  • a preferred method is a method, wherein a patient or one or more cells of said patient is provided with a functional dystrophin protein and/or wherein the production of an aberrant dystrophin protein in said patient is decreased and/or wherein the production of a functional dystrophin is increased in said patient, wherein the level of said aberrant or functional dystrophin is assessed by comparison to the level of said dystrophin in said patient at the onset of the method.
  • Decreasing the production of an aberrant dystrophin may be assessed at the mRNA level and preferably means that 99%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 10%, 5% or less of the initial amount of aberrant dystrophin mRNA, is still detectable by RT PCR.
  • An aberrant dystrophin mRNA or protein is also referred to herein as a non-functional dystrophin mRNA or protein.
  • a non functional dystrophin protein is preferably a dystrophin protein which is not able to bind actin and/or members of the DGC protein complex.
  • a non-functional dystrophin protein or dystrophin mRNA does typically not have, or does not encode a dystrophin protein with an intact C-terminus of the protein.
  • Increasing the production of a functional dystrophin in said patient or in a cell of said patient may be assessed at the mRNA level (by RT-PCR analysis) and preferably means that a detectable amount of a functional dystrophin mRNA is detectable by RT PCR.
  • 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more of the detectable dystrophin mRNA is a functional dystrophin mRNA.
  • Increasing the production of a functional dystrophin in said patient or in a cell of said patient may be assessed at the protein level (by immunofluorescence and western blot analyses) and preferably means that a detectable amount of a functional dystrophin protein is detectable by immunofluorescence or western blot analysis.
  • 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more of the detectable dystrophin protein is a functional dystrophin protein.
  • a functional dystrophin is preferably a wild type dystrophin corresponding to a protein having the amino acid sequence as identified in SEQ ID NO: 1.
  • a functional dystrophin is preferably a dystrophin, which has an actin binding domain in its N terminal part (first 240 amino acids at the N terminus), a cystein-rich domain (amino acid 3361 till 3685) and a C terminal domain (last 325 amino acids at the C terminus) each of these domains being present in a wild type dystrophin as known to the skilled person.
  • the amino acids indicated herein correspond to amino acids of the wild type dystrophin being represented by SEQ ID NO:1.
  • a functional dystrophin is a dystrophin which exhibits at least to some extent an activity of a wild type dystrophin. “At least to some extent” preferably means at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or 100% of a corresponding activity of a wild type functional dystrophin.
  • an activity of a functional dystrophin is preferably binding to actin and to the dystrophin-associated glycoprotein complex (DGC) (Aartsma-Rus A et al, (2006), Entries in the leiden Duchenne Muscular Dystrophy mutation database: an overview of mutation types and paradoxical cases that confirm the reading-frame rule, Muscle Nerve, 34: 135-144). Binding of dystrophin to actin and to the DGC complex may be visualized by either co-immunoprecipitation using total protein extracts or immunofluorescence analysis of cross-sections, from a muscle biopsy, as known to the skilled person.
  • DGC dystrophin-associated glycoprotein complex
  • Duchenne muscular dystrophy typically have a mutation in the gene encoding dystrophin that prevent synthesis of the complete protein, i.e of a premature stop prevents the synthesis of the C-terminus.
  • the DMD gene also comprises a mutation compared tot the wild type gene but the mutation does typically not induce a premature stop and the C-terminus is typically synthesized.
  • a functional dystrophin protein is synthesized that has at least the same activity in kind as the wild type protein, not although not necessarily the same amount of activity.
  • the genome of a BMD individual typically encodes a dystrophin protein comprising the N terminal part (first 240 amino acids at the N terminus), a cystein-rich domain (amino acid 3361 till 3685) and a C terminal domain (last 325 amino acids at the C terminus) but its central rod shaped domain may be shorter than the one of a wild type dystrophin (Aartsma-Rus A et al, (2006), Entries in the leiden Duchenne Muscular Dystrophy mutation database: an overview of mutation types and paradoxical cases that confirm the reading-frame rule, Muscle Nerve, 34: 135-144).
  • Exon skipping for the treatment of DMD is typically directed to overcome a premature stop in the pre-mRNA by skipping an exon in the rod-shaped domain to correct the reading frame and allow synthesis of remainder of the dystrophin protein including the C-terminus, albeit that the protein is somewhat smaller as a result of a smaller rod domain.
  • an individual having DMD and being treated by a method as defined herein will be provided a dystrophin which exhibits at least to some extent an activity of a wild type dystrophin.
  • a functional dystrophin is a dystrophin of an individual having BMD: typically said dystrophin is able to interact with both actin and the DGC, but its central rod shaped domain may be shorter than the one of a wild type dystrophin (Aartsma-Rus A et al, (2006), Entries in the leiden Duchenne Muscular Dystrophy mutation database: an overview of mutation types and paradoxical cases that confirm the reading-frame rule, Muscle Nerve, 34: 135-144).
  • the central rod-shaped domain of wild type dystrophin comprises 24 spectrin-like repeats (Aartsma-Rus A et al, (2006), Entries in the leiden Duchenne Muscular Dystrophy mutation database: an overview of mutation types and paradoxical cases that confirm the reading-frame rule, Muscle Nerve, 34: 135-144).
  • a central rod-shaped domain of a dystrophin as provided herein may comprise 5 to 23, 10 to 22 or 12 to 18 spectrin-like repeats as long as it can bind to actin and to DGC.
  • a method of the invention may alleviate one or more characteristics of a myogenic or muscle cell of a patient or alleviate one or more symptoms of a DMD patient having a deletion including but not limited to exons 44, 44-46, 44-47, 44-48, 44-49, 44-51, 44-53 (correctable by exon 43 skipping), 19-45, 21-45, 43-45, 45, 47-54, 47-56 (correctable by exon 46 skipping), 51, 51-53, 51-55, 51-57 (correctable by exon 50 skipping), 13-50, 19-50, 29-50, 43-50, 45-50, 47-50, 48-50, 49-50, 50, 52 (correctable by exon 51 skipping), exons 8-51, 51, 53, 53-55, 53-57, 53-59, 53-60, (correctable by exon 52 skipping) and exons 10-52, 42-52, 43-52, 45-52, 47-52, 48-52, 49-52, 50-52, 52 (correctable by exon 53
  • a method of the invention may improve one or more characteristics of a muscle cell of a patient or alleviate one or more symptoms of a DMD patient having small mutations in, or single exon duplications of exon 43, 46, 50-53 in the DMD gene, occurring in a total of 36% of all DMD patients with a deletion (Aartsma-Rus et al, Hum. Mut. 2009)
  • exon 46 and/or exon 50-53 is required to restore the open reading frame, including patients with specific deletions, small (point) mutations, or double or multiple exon duplications, such as (but not limited to) a deletion of exons 44-50 requiring the co-skipping of exons 43 and 51, with a deletion of exons 46-50 requiring the co-skipping of exons 45 and 51, with a deletion of exons 44-52 requiring the co-skipping of exons 43 and 53, with a deletion of exons 46-52 requiring the co-skipping of exons 45 and 53, with a deletion of exons 51-54 requiring the co-skipping of exons 50 and 55, with a deletion of exons 53-54 requiring the co-skipping of exons 52 and 55, with a deletion of exons 53-56 requiring the co-skipping of exons 52 and 57,
  • the skipping of exon 43 is induced, or the skipping of exon 46 is induced, or the skipping of exon 50 is induced or the skipping of exon 51 is induced or the skipping of exon 52 is induced or the skipping of exon 53 is induced.
  • An induction of the skipping of two of these exons is also encompassed by a method of the invention.
  • exons 50 and 51, or 52 and 53, or 43 and 51, or 43 and 53, or 51 and 52 preferably skipping of exons 50 and 51, or 52 and 53, or 43 and 51, or 43 and 53, or 51 and 52.
  • the skilled person will know which combination of exons needs to be skipped in said patient.
  • one or more symptom(s) of a DMD or a BMD patient is/are alleviated and/or one or more characteristic(s) of one or more muscle cells from a DMD or a BMD patient is/are improved.
  • symptoms or characteristics may be assessed at the cellular, tissue level or on the patient self
  • An alleviation of one or more characteristics may be assessed by any of the following assays on a myogenic cell or muscle cell from a patient: reduced calcium uptake by muscle cells, decreased collagen synthesis, altered morphology, altered lipid biosynthesis, decreased oxidative stress, and/or improved muscle fiber function, integrity, and/or survival. These parameters are usually assessed using immunofluorescence and/or histochemical analyses of cross sections of muscle biopsies.
  • the improvement of muscle fiber function, integrity and/or survival may be assessed using at least one of the following assays: a detectable decrease of creatine kinase in blood, a detectable decrease of necrosis of muscle fibers in a biopsy cross-section of a muscle suspected to be dystrophic, and/or a detectable increase of the homogeneity of the diameter of muscle fibers in a biopsy cross-section of a muscle suspected to be dystrophic.
  • a detectable decrease of creatine kinase in blood a detectable decrease of necrosis of muscle fibers in a biopsy cross-section of a muscle suspected to be dystrophic
  • a detectable increase of the homogeneity of the diameter of muscle fibers in a biopsy cross-section of a muscle suspected to be dystrophic are known to the skilled person.
  • Creatine kinase may be detected in blood as described in Hodgetts et al (Hodgetts S., et al, (2006), Neuromuscular Disorders, 16: 591-602.2006).
  • a detectable decrease in creatine kinase may mean a decrease of 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more compared to the concentration of creatine kinase in a same DMD or BMD patient before treatment.
  • a detectable decrease of necrosis of muscle fibers is preferably assessed in a muscle biopsy, more preferably as described in Hodgetts et al (Hodgetts S., et al, (2006), Neuromuscular Disorders, 16: 591-602.2006) using biopsy cross-sections.
  • a detectable decrease of necrosis may be a decrease of 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more of the area wherein necrosis has been identified using biopsy cross-sections. The decrease is measured by comparison to the necrosis as assessed in a same DMD or BMD patient before treatment.
  • a detectable increase of the homogeneity of the diameter of a muscle fiber is preferably assessed in a muscle biopsy cross-section, more preferably as described in Hodgetts et al (Hodgetts S., et al, (2006), Neuromuscular Disorders, 16: 591-602.2006).
  • the increase is measured by comparison to the homogeneity of the diameter of a muscle fiber in a same DMD or BMD patient before treatment
  • An alleviation of one or more symptoms may be assessed by any of the following assays on the patient self: prolongation of time to loss of walking, improvement of muscle strength, improvement of the ability to lift weight, improvement of the time taken to rise from the floor, improvement in the nine-meter walking time, improvement in the time taken for four-stairs climbing, improvement of the leg function grade, improvement of the pulmonary function, improvement of cardiac function, improvement of the quality of life.
  • assays are known to the skilled person.
  • Detectable improvement or prolongation is preferably a statistically significant improvement or prolongation as described in Hodgetts et al (Hodgetts S., et al, (2006), Neuromuscular Disorders,16: 591-602.2006).
  • the alleviation of one or more symptom(s) of Duchenne Muscular Dystrophy or Becker Muscular Dystrophy may be assessed by measuring an improvement of a muscle fiber function, integrity and/or survival as later defined herein.
  • a treatment in a method according to the invention may have a duration of at least one week, at least one month, at least several months, at least one year, at least 2, 3, 4, 5, 6 years or more.
  • Each molecule or oligonucleotide or equivalent thereof as defined herein for use according to the invention may be suitable for direct administration to a cell, tissue and/or an organ in vivo of individuals affected by or at risk of developing DMD or BMD, and may be administered directly in vivo, ex vivo or in vitro.
  • the frequency of administration of a molecule or an oligonucleotide or a composition of the invention may depend on several parameters such as the age of the patient, the mutation of the patient, the number of molecules (dose), the formulation of said molecule. The frequency may be ranged between at least once in a two weeks, or three weeks or four weeks or five weeks or a longer time period.
  • a molecule or oligonucleotide or equivalent thereof can be delivered as is to a cell.
  • oligonucleotide or equivalent thereof When administering said molecule, oligonucleotide or equivalent thereof to an individual, it is preferred that it is dissolved in a solution that is compatible with the delivery method.
  • a solution For intravenous, subcutaneous, intramuscular, intrathecal and/or intraventricular administration it is preferred that the solution is a physiological salt solution.
  • Particularly preferred for a method of the invention is the use of an excipient that will further enhance delivery of said molecule, oligonucleotide or functional equivalent thereof as defined herein, to a cell and into a cell, preferably a muscle cell.
  • an additional molecule is used which is able to induce and/or promote skipping of another exon of the DMD pre-mRNA of a patient.
  • the second exon is selected from: exon 6, 7, 11, 17, 19, 21, 43, 44, 45, 50, 51, 52, 53, 55, 57, 59, 62, 63, 65, 66, 69, or 75 of the DMD pre-mRNA of a patient.
  • Molecules which can be used are depicted in any one of Table 1 to 7. This way, inclusion of two or more exons of a DMD pre-mRNA in mRNA produced from this pre-mRNA is prevented.
  • This embodiment is further referred to as double- or multi-exon skipping (Aartsma-Rus A, Janson AA, Kaman WE, et al. Antisense-induced multiexon skipping for Duchenne muscular dystrophy makes more sense. Am J Hum Genet 2004;74(1):83-92, Aartsma-Rus A, Kaman W E, Weij R, den Dunnen J T, van Ommen G J, van Deutekom J C.
  • This multi-skipping was notably so for the combination of oligonucleotides derived from the DMD gene, wherein one oligonucleotide for exon 45 and one oligonucleotide for exon 51 was added to a cell transcribing the DMD gene.
  • Such a set-up resulted in mRNA being produced that did not contain exons 45 to 51.
  • the structure of the pre-mRNA in the presence of the mentioned oligonucleotides was such that the splicing machinery was stimulated to connect exons 44 and 52 to each other.
  • stretches of nucleotides complementary to at least two dystrophin exons are separated by a linking moiety.
  • the at least two stretches of nucleotides are thus linked in this embodiment so as to form a single molecule.
  • said compounds can be administered to an individual in any order.
  • said compounds are administered simultaneously (meaning that said compounds are administered within 10 hours, preferably within one hour). This is however not necessary.
  • said compounds are administered sequentially.
  • a molecule as defined herein is preferably an oligonucleotide or antisense oligonucleotide (AON).
  • any of exon 43, 46, 50-53 is specifically skipped at a high frequency using a molecule that preferably binds to a continuous stretch of at least 8 nucleotides within said exon.
  • this effect can be associated with a higher binding affinity of said molecule, compared to a molecule that binds to a continuous stretch of less than 8 nucleotides, there could be other intracellular parameters involved that favor thermodynamic, kinetic, or structural characteristics of the hybrid duplex.
  • a molecule that binds to a continuous stretch of at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37,38, 39, 40, 41, 42, 43,44, 45, 46, 47, 48, 49, 50 nucleotides within said exon is used.
  • a molecule or an oligonucleotide of the invention which comprises a sequence that is complementary to a part of any of exon 43, 46, 50-53 of DMD pre-mRNA is such that the complementary part is at least 50% of the length of the oligonucleotide of the invention, more preferably at least 60%, even more preferably at least 70%, even more preferably at least 80%, even more preferably at least 90% or even more preferably at least 95%, or even more preferably 98% and most preferably up to 100%.
  • “A part of said exon” preferably means a stretch of at least 8 nucleotides.
  • an oligonucleotide of the invention consists of a sequence that is complementary to part of said exon DMD pre-mRNA as defined herein.
  • an oligonucleotide may comprise a sequence that is complementary to part of said exon DMD pre-mRNA as defined herein and additional flanking sequences.
  • the length of said complementary part of said oligonucleotide is of at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27 , 28 , 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50 nucleotides.
  • additional flanking sequences are used to modify the binding of a protein to said molecule or oligonucleotide, or to modify a thermodynamic property of the oligonucleotide, more preferably to modify target RNA binding affinity.
  • a preferred molecule to be used in a method of the invention binds or is complementary to a continuous stretch of at least 8 nucleotides within one of the following nucleotide sequences selected from:
  • the invention provides distinct molecules that can be used in a method for efficiently skipping of at least one of exon 43, exon 46 and/or exon 50-53.
  • skipping effect can be addressed to the relatively high density of putative SR protein binding sites within said stretches, there could be other parameters involved that favor uptake of the molecule or other, intracellular parameters such as thermodynamic, kinetic, or structural characteristics of the hybrid duplex.
  • a molecule that binds to a continuous stretch comprised within or consisting of any of SEQ ID NO 2-7 results in highly efficient skipping of exon 43, exon 46 and/or exon 50- 53 respectively in a cell and/or in a patient provided with this molecule. Therefore, in a preferred embodiment, a method is provided wherein a molecule binds to a continuous stretch of at least 8, 10, 12, 15, 18, 20, 25, 30, 35, 40, 45, 50 nucleotides within SEQ ID NO 2-7.
  • the invention provides a molecule comprising or consisting of an antisense nucleotide sequence selected from the antisense nucleotide sequences depicted in any of Tables 1 to 6.
  • a molecule of the invention preferably comprises or consist of the antisense nucleotide sequence of SEQ ID NO 16, SEQ ID NO 65, SEQ ID NO 70, SEQ ID NO 91, SEQ ID NO 110, SEQ ID NO 117, SEQ ID NO 127, SEQ ID NO 165, SEQ ID NO 166, SEQ ID NO 167, SEQ ID NO 246, SEQ ID NO 299, SEQ ID NO:357.
  • a preferred molecule of the invention comprises a nucleotide-based or nucleotide or an antisense oligonucleotide sequence of between 8 and 50 nucleotides or bases, more preferred between 10 and 50 nucleotides, more preferred between 20 and 40 nucleotides, more preferred between 20 and 30 nucleotides, such as 20 nucleotides, 21 nucleotides, 22 nucleotides, 23 nucleotides, 24 nucleotides, 25 nucleotides, 26 nucleotides, 27 nucleotides, 28 nucleotides, 29 nucleotides, 30 nucleotides, 31 nucleotides, 32 nucleotides, 33 nucleotides, 34 nucleotides, 35 nucleotides, 36 nucleotides, 37 nucleotides, 38 nucleotides, 39 nucleotides, 40 nucleotides, 41 nucleotides, 42 nucleotides, 43 nucleot
  • a most preferred molecule of the invention comprises a nucleotide-based sequence of 25 nucleotides.
  • a molecule of the invention is a compound molecule that binds to the specified sequence, or a protein such as an RNA-binding protein or a non-natural zinc-finger protein that has been modified to be able to bind to the corresponding nucleotide sequence on a DMD pre-RNA molecule.
  • Methods for screening compound molecules that bind specific nucleotide sequences are, for example, disclosed in PCT/NL01/00697 and U.S. Pat. No. 6,875,736, which are herein incorporated by reference.
  • Methods for designing RNA-binding Zinc-finger proteins that bind specific nucleotide sequences are disclosed by Friesen and Darby, Nature Structural Biology 5: 543-546 (1998) which is herein incorporated by reference.
  • a preferred molecule of the invention binds to at least part of the sequence of SEQ ID NO 2:5′-AGAUAGUCUACAACAAAGCUCAGGUCGGAUUGACAUUAUUCAU AGCAAGAAGACAGCAGCAUUGCAAAGUGCAACGCCUGUGG-3′ which is present in exon 43 of the DMD gene. More preferably, the invention provides a molecule comprising or consisting of the antisense nucleotide sequence of SEQ ID NO 8 to SEQ ID NO 69.
  • the invention provides a molecule comprising or consisting of the antisense nucleotide sequence of SEQ ID NO 16 and/or SEQ ID NO 65.
  • the invention provides a molecule comprising or consisting of the antisense nucleotide sequence of SEQ ID NO 65. It was found that this molecule is very efficient in modulating splicing of exon 43 of the DMD pre-mRNA in a muscle cell and/or in a patient.
  • Another preferred molecule of the invention binds to at least part of the sequence of SEQ ID NO 3: 5′-UUAUGGUUGGAGGAAGCAGAUAACAUUGCUAGUAUCCCACUUG AACCUGGAAAAGAGCAGCAACUAAAAGAAAAGC-3′ which is present in exon 46 of the DMD gene. More preferably, the invention provides a molecule comprising or consisting of the antisense nucleotide sequence of SEQ ID NO 70 to SEQ ID NO 122.
  • the invention provides a molecule comprising or consisting of the antisense nucleotide sequence of SEQ ID NO 70, SEQ ID NO 91, SEQ ID NO 110, and/or SEQ ID NO117.
  • the invention provides a molecule comprising or consisting of the antisense nucleotide sequence of SEQ ID NO 117. It was found that this molecule is very efficient in modulating splicing of exon 46 of the DMD pre-mRNA in a muscle cell or in a patient.
  • Another preferred molecule of the invention binds to at least part of the sequence of SEQ ID NO 4: 5′-GGCGGTAAACCGUUUACUUCAAGAGCU GAGGGCAAAGCAGCCUG ACCUAGCUCCUGGACUGACCACUAUUGG-3′ which is present in exon 50 of the DMD gene. More preferably, the invention provides a molecule comprising or consisting of the antisense nucleotide sequence of SEQ ID NO 123 to SEQ ID NO 167 and/or SEQ ID NO 529 to SEQ ID NO 535.
  • the invention provides a molecule comprising or consisting of the antisense nucleotide sequence of SEQ ID NO 127, or SEQ ID NO 165, or SEQ ID NO 166 and/or SEQ ID NO 167.
  • the invention provides a molecule comprising or consisting of the antisense nucleotide sequence of SEQ ID NO 127. It was found that this molecule is very efficient in modulating splicing of exon 50 of the DMD pre-mRNA in a muscle cell and/or in a patient.
  • Another preferred molecule of the invention binds to at least part of the sequence of SEQ ID NO 5: 5′-CUCCUACUCAGACUGUUACUCUGGUGACACAACCUGUGGUUACU AAGGAAACUGCCAUC UCCAAACUAGAAAUGCCAUCUUCCUUGAUG UUGGAGGUAC-3′ which is present in exon 51 of the DMD gene. More preferably, the invention provides a molecule comprising or consisting of the antisense nucleotide sequence of SEQ ID NO 168 to SEQ ID NO 241.
  • Another preferred molecule of the invention binds to at least part of the sequence of SEQ ID NO 6: 5′-AUGCAGGAUUUGGAACAGAGGCGUCCCCAGUUGGAAGAACUCAU UACCGCUGCCCAAAAUUUGAAAAACAAGACCAGCAAUCAAGAGGCU-3′ which is present in exon 52 of the DMD gene. More preferably, the invention provides a molecule comprising or consisting of the antisense nucleotide sequence of SEQ ID NO 242 to SEQ ID NO 310. In an even more preferred embodiment, the invention provides a molecule comprising or consisting of the antisense nucleotide sequence of SEQ ID NO 246 and/or SEQ ID NO 299.
  • the invention provides a molecule comprising or consisting of the antisense nucleotide sequence of SEQ ID NO 299. It was found that this molecule is very efficient in modulating splicing of exon 52 of the DMD pre-mRNA in a muscle cell and/or in a patient.
  • Another preferred molecule of the invention binds to at least part of the sequence of SEQ ID NO 7: 5′-AAAUGUUAAAGGAUUCAACACAAUGGCUGGAAGCUAAGGAAGAA GCUGAGCAGGUCUUAGGACAGGCCAGAG-3′ which is present in exon 53 of the DMD gene. More preferably, the invention provides a molecule comprising or consisting of the antisense nucleotide sequence of SEQ ID NO 311 to SEQ ID NO 358.
  • the invention provides a molecule comprising or consisting of the antisense nucleotide sequence of SEQ ID NO 357. It was found that this molecule is very efficient in modulating splicing of exon 53 of the DMD pre-mRNA in a muscle cell and/or in a patient.
  • a nucleotide sequence of a molecule of the invention may contain RNA residues, or one or more DNA residues, and/or one or more nucleotide analogues or equivalents, as will be further detailed herein below.
  • a molecule of the invention comprises one or more residues that are modified to increase nuclease resistance, and/or to increase the affinity of the antisense nucleotide for the target sequence. Therefore, in a preferred embodiment, the antisense nucleotide sequence comprises at least one nucleotide analogue or equivalent, wherein a nucleotide analogue or equivalent is defined as a residue having a modified base, and/or a modified backbone, and/or a non-natural internucleoside linkage, or a combination of these modifications.
  • the nucleotide analogue or equivalent comprises a modified backbone.
  • backbones are provided by morpholino backbones, carbamate backbones, siloxane backbones, sulfide, sulfoxide and sulfone backbones, formacetyl and thioformacetyl backbones, methyleneformacetyl backbones, riboacetyl backbones, alkene containing backbones, sulfamate, sulfonate and sulfonamide backbones, methyleneimino and methylenehydrazino backbones, and amide backbones.
  • Phosphorodiamidate morpholino oligomers are modified backbone oligonucleotides that have previously been investigated as antisense agents.
  • Morpholino oligonucleotides have an uncharged backbone in which the deoxyribose sugar of DNA is replaced by a six membered ring and the phosphodiester linkage is replaced by a phosphorodiamidate linkage.
  • Morpholino oligonucleotides are resistant to enzymatic degradation and appear to function as antisense agents by arresting translation or interfering with pre-mRNA splicing rather than by activating RNase H.
  • Morpholino oligonucleotides have been successfully delivered to tissue culture cells by methods that physically disrupt the cell membrane, and one study comparing several of these methods found that scrape loading was the most efficient method of delivery; however, because the morpholino backbone is uncharged, cationic lipids are not effective mediators of morpholino oligonucleotide uptake in cells. A recent report demonstrated triplex formation by a morpholino oligonucleotide and, because of the non-ionic backbone, these studies showed that the morpholino oligonucleotide was capable of triplex formation in the absence of magnesium.
  • the linkage between the residues in a backbone do not include a phosphorus atom, such as a linkage that is formed by short chain alkyl or cycloalkyl internucleoside linkages, mixed heteroatom and alkyl or cycloalkyl internucleoside linkages, or one or more short chain heteroatomic or heterocyclic internucleoside linkages.
  • a preferred nucleotide analogue or equivalent comprises a Peptide Nucleic Acid (PNA), having a modified polyamide backbone (Nielsen, et al. (1991) Science 254, 1497-1500). PNA-based molecules are true mimics of DNA molecules in terms of base-pair recognition.
  • the backbone of the PNA is composed of N-(2-aminoethyl)-glycine units linked by peptide bonds, wherein the nucleobases are linked to the backbone by methylene carbonyl bonds.
  • An alternative backbone comprises a one-carbon extended pyrrolidine PNA monomer (Govindaraju and Kumar (2005) Chem. Commun, 495-497).
  • PNA-RNA hybrids are usually more stable than RNA-RNA or RNA-DNA hybrids, respectively (Egholm et al (1993) Nature 365, 566-568).
  • a further preferred backbone comprises a morpholino nucleotide analog or equivalent, in which the ribose or deoxyribose sugar is replaced by a 6-membered morpholino ring.
  • a most preferred nucleotide analog or equivalent comprises a phosphorodiamidate morpholino oligomer (PMO), in which the ribose or deoxyribose sugar is replaced by a 6-membered morpholino ring, and the anionic phosphodiester linkage between adjacent morpholino rings is replaced by a non-ionic phosphorodiamidate linkage.
  • PMO phosphorodiamidate morpholino oligomer
  • a nucleotide analogue or equivalent of the invention comprises a substitution of one of the non-bridging oxygens in the phosphodiester linkage. This modification slightly destabilizes base-pairing but adds significant resistance to nuclease degradation.
  • a preferred nucleotide analogue or equivalent comprises phosphorothioate, chiral phosphorothioate, phosphorodithioate, phosphotriester, aminoalkylphosphotriester, H-phosphonate, methyl and other alkyl phosphonate including 3′-alkylene phosphonate, 5′-alkylene phosphonate and chiral phosphonate, phosphinate, phosphoramidate including 3′-amino phosphoramidate and aminoalkylphosphoramidate, thionophosphoramidate, thionoalkylphosphonate, thionoalkylphosphotriester, selenophosphate or boranophosphate.
  • a further preferred nucleotide analogue or equivalent of the invention comprises one or more sugar moieties that are mono- or disubstituted at the 2′, 3′ and/or 5′ position such as a —OH; —F; substituted or unsubstituted, linear or branched lower (C1-C10) alkyl, alkenyl, alkynyl, alkaryl, allyl, aryl, or aralkyl, that may be interrupted by one or more heteroatoms; O-, S-, or N-alkyl; O-, S-, or N-alkenyl; O-, S- or N-alkynyl; O-, S-, or N-allyl; O-alkyl-O-alkyl, -methoxy, -aminopropoxy; -aminoxy; methoxyethoxy; -dimethylaminooxyethoxy; and -dimethylaminoethoxyethoxy.
  • the sugar moiety can be a pyranose or derivative thereof, or a deoxypyranose or derivative thereof, preferably a ribose or a derivative thereof, or a deoxyribose or a derivative thereof.
  • Such preferred derivatized sugar moieties comprise Locked Nucleic Acid (LNA), in which the 2′-carbon atom is linked to the 3′ or 4′ carbon atom of the sugar ring thereby forming a bicyclic sugar moiety.
  • LNA Locked Nucleic Acid
  • a preferred LNA comprises 2′-O,4′-C-ethylene-bridged nucleic acid (Morita et al. 2001. Nucleic Acid Res Supplement No. 1: 241-242). These substitutions render the nucleotide analogue or equivalent RNase H and nuclease resistant and increase the affinity for the target RNA.
  • an antisense oligonucleotide of the invention has at least two different types of analogues or equivalents.
  • a preferred antisense oligonucleotide according to the invention comprises a 2′-O alkyl phosphorothioate antisense oligonucleotide, such as 2′-O-methyl modified ribose (RNA), 2′-O-ethyl modified ribose, 2′-O-propyl modified ribose, and/or substituted derivatives of these modifications such as halogenated derivatives.
  • RNA 2′-O-methyl modified ribose
  • 2′-O-ethyl modified ribose 2′-O-propyl modified ribose
  • substituted derivatives of these modifications such as halogenated derivatives.
  • a most preferred antisense oligonucleotide according to the invention comprises of 2′-O-methyl phosphorothioate ribose.
  • a functional equivalent of a molecule of the invention may be defined as an oligonucleotide as defined herein wherein an activity of said functional equivalent is retained to at least some extent.
  • an activity of said functional equivalent is inducing exon 43, 46, 50, 51, 52, or 53 skipping and providing a functional dystrophin protein. Said activity of said functional equivalent is therefore preferably assessed by detection of exon 43, 46, 50, 51, 52, or 53 skipping and by quantifying the amount of functional dystrophin protein.
  • a functional dystrophin is herein preferably defined as being a dystrophin able to bind actin and members of the DGC protein complex.
  • the assessment of said activity of an oligonucleotide is preferably done by RT-PCR or by immunofluorescence or Western blot analyses. Said activity is preferably retained to at least some extent when it represents at least 50%, or at least 60%, or at least 70% or at least 80% or at least 90% or at least 95% or more of corresponding activity of said oligonucleotide the functional equivalent derives from. Throughout this application, when the word oligonucleotide is used it may be replaced by a functional equivalent thereof as defined herein.
  • distinct antisense oligonucleotides can be combined for efficiently skipping any of exon 43, exon 46, exon 50, exon 51, exon 52 and/or exon 53 of the human DMD pre-mRNA. It is encompassed by the present invention to use one, two, three, four, five or more oligonucleotides for skipping one of said exons (i.e. exon, 43, 46, 50, 51, 52, or 53). It is also encompassed to use at least two oligonucleotides for skipping at least two, of said exons. Preferably two of said exons are skipped. More preferably, these two exons are:
  • An antisense oligonucleotide can be linked to a moiety that enhances uptake of the antisense oligonucleotide in cells, preferably muscle cells.
  • moieties are cholesterols, carbohydrates, vitamins, biotin, lipids, phospholipids, cell-penetrating peptides including but not limited to antennapedia, TAT, transportan and positively charged amino acids such as oligoarginine, poly-arginine, oligolysine or polylysine, antigen-binding domains such as provided by an antibody, a Fab fragment of an antibody, or a single chain antigen binding domain such as a cameloid single domain antigen-binding domain.
  • a preferred antisense oligonucleotide comprises a peptide-linked PMO.
  • a preferred antisense oligonucleotide comprising one or more nucleotide analogs or equivalents of the invention modulates splicing in one or more muscle cells, including heart muscle cells, upon systemic delivery.
  • systemic delivery of an antisense oligonucleotide comprising a specific nucleotide analog or equivalent might result in targeting a subset of muscle cells, while an antisense oligonucleotide comprising a distinct nucleotide analog or equivalent might result in targeting of a different subset of muscle cells.
  • a combination of antisense oligonucleotides comprising different nucleotide analogs or equivalents for inducing skipping of exon 43, 46, 50, 51, 52, or 53 of the human DMD pre-mRNA.
  • a cell can be provided with a molecule capable of interfering with essential sequences that result in highly efficient skipping of exon 43, exon 46, exon 50, exon 51, exon 52 or exon 53 of the human DMD pre-mRNA by plasmid-derived antisense oligonucleotide expression or viral expression provided by adenovirus- or adeno-associated virus-based vectors.
  • a viral-based expression vector comprising an expression cassette that drives expression of a molecule as identified herein. Expression is preferably driven by a polymerase III promoter, such as a U1, a U6, or a U7 RNA promoter.
  • a muscle or myogenic cell can be provided with a plasmid for antisense oligonucleotide expression by providing the plasmid in an aqueous solution.
  • a plasmid can be provided by transfection using known transfection agentia such as, for example, LipofectAMINETM 2000 (Invitrogen) or polyethyleneimine (PEI; ExGen500 (MBI Fermentas)), or derivatives thereof.
  • AAV adenovirus associated virus
  • a preferred AAV-based vector comprises an expression cassette that is driven by a polymerase III-promoter (Pol III).
  • Pol III polymerase III-promoter
  • a preferred Pol III promoter is, for example, a U1, a U6, or a U7 RNA promoter.
  • the invention therefore also provides a viral-based vector, comprising a Pol III-promoter driven expression cassette for expression of one or more antisense sequences of the invention for inducing skipping of exon 43, exon 46, exon 50, exon 51, exon 52 or exon 53 of the human DMD pre-mRNA.
  • a molecule or a vector expressing an antisense oligonucleotide of the invention can be incorporated into a pharmaceutically active mixture or composition by adding a pharmaceutically acceptable carrier.
  • the invention provides a composition, preferably a pharmaceutical composition comprising a molecule comprising an antisense oligonucleotide according to the invention, and/or a viral-based vector expressing the antisense sequence(s) according to the invention and a pharmaceutically acceptable carrier.
  • a preferred pharmaceutical composition comprises a molecule as defined herein and/or a vector as defined herein, and a pharmaceutical acceptable carrier or excipient, optionally combined with a molecule and/or a vector as defined herein which is able to induce skipping of exon 6, 7, 11, 17, 19, 21, 43, 44, 45, 50, 51, 52, 53, 55, 57, 59, 62, 63, 65, 66, 69, or 75 of the DMD pre-mRNA.
  • Preferred molecules able to induce skipping of any of these exon are identified in any one of Tables 1 to 7.
  • Preferred excipients include excipients capable of forming complexes, vesicles and/or liposomes that deliver such a molecule as defined herein, preferably an oligonucleotide complexed or trapped in a vesicle or liposome through a cell membrane. Many of these excipients are known in the art.
  • Suitable excipients comprise polyethylenimine and derivatives, or similar cationic polymers, including polypropyleneimine or polyethylenimine copolymers (PECs) and derivatives, ExGen 500, synthetic amphiphils (SAINT-18), lipofectinTM, DOTAP and/or viral capsid proteins that are capable of self assembly into particles that can deliver such molecule, preferably an oligonucleotide as defined herein to a cell, preferably a muscle cell.
  • excipients have been shown to efficiently deliver (oligonucleotide such as antisense) nucleic acids to a wide variety of cultured cells, including muscle cells. Their high transfection potential is combined with an excepted low to moderate toxicity in terms of overall cell survival. The ease of structural modification can be used to allow further modifications and the analysis of their further (in vivo) nucleic acid transfer characteristics and toxicity.
  • Lipofectin represents an example of a liposomal transfection agent. It consists of two lipid components, a cationic lipid N-[1-(2,3 dioleoyloxy)propyl]-N,N,N-trimethylammonium chloride (DOTMA) (cp. DOTAP which is the methylsulfate salt) and a neutral lipid dioleoylphosphatidylethanolamine (DOPE). The neutral component mediates the intracellular release.
  • DOTMA cationic lipid N-[1-(2,3 dioleoyloxy)propyl]-N,N,N-trimethylammonium chloride
  • DOPE neutral lipid dioleoylphosphatidylethanolamine
  • Another group of delivery systems are polymeric nanoparticles.
  • Polycations such like diethylaminoethylaminoethyl (DEAE)-dextran, which are well known as DNA transfection reagent can be combined with butylcyanoacrylate (PBCA) and hexylcyanoacrylate (PHCA) to formulate cationic nanoparticles that can deliver a molecule or a compound as defined herein, preferably an oligonucleotide across cell membranes into cells.
  • PBCA butylcyanoacrylate
  • PHCA hexylcyanoacrylate
  • the cationic peptide protamine offers an alternative approach to formulate a compound as defined herein, preferably an oligonucleotide as colloids.
  • This colloidal nanoparticle system can form so called proticles, which can be prepared by a simple self-assembly process to package and mediate intracellular release of a compound as defined herein, preferably an oligonucleotide.
  • the skilled person may select and adapt any of the above or other commercially available alternative excipients and delivery systems to package and deliver a compound as defined herein, preferably an oligonucleotide for use in the current invention to deliver said compound for the treatment of Duchenne Muscular Dystrophy or Becker Muscular Dystrophy in humans.
  • a compound as defined herein preferably an oligonucleotide could be covalently or non-covalently linked to a targeting ligand specifically designed to facilitate the uptake in to the cell, cytoplasm and/or its nucleus.
  • a targeting ligand specifically designed to facilitate the uptake in to the cell, cytoplasm and/or its nucleus.
  • ligand could comprise (i) a compound (including but not limited to peptide(-like) structures) recognising cell, tissue or organ specific elements facilitating cellular uptake and/or (ii) a chemical compound able to facilitate the uptake in to cells and/or the intracellular release of an a compound as defined herein, preferably an oligonucleotide from vesicles, e.g. endosomes or lysosomes.
  • a compound as defined herein, preferably an oligonucleotide are formulated in a medicament which is provided with at least an excipient and/or a targeting ligand for delivery and/or a delivery device of said compound to a cell and/or enhancing its intracellular delivery.
  • the invention also encompasses a pharmaceutically acceptable composition comprising a compound as defined herein, preferably an oligonucleotide and further comprising at least one excipient and/or a targeting ligand for delivery and/or a delivery device of said compound to a cell and/or enhancing its intracellular delivery.
  • a molecule or compound or oligonucleotide may not be formulated in one single composition or preparation. Depending on their identity, the skilled person will know which type of formulation is the most appropriate for each compound.
  • an in vitro concentration of a molecule or an oligonucleotide as defined herein, which is ranged between 0.1 nM and 1 R M is used.
  • the concentration used is ranged between 0.3 to 400 nM, even more preferably between 1 to 200 nM.
  • a molecule or an oligonucleotide as defined herein may be used at a dose which is ranged between 0.1 and 20 mg/kg, preferably 0.5 and 10 mg/kg. If several molecules or oligonucleotides are used, these concentrations may refer to the total concentration of oligonucleotides or the concentration of each oligonucleotide added.
  • the ranges of concentration of oligonucleotide(s) as given above are preferred concentrations for in vitro or ex vivo uses.
  • the concentration of oligonucleotide(s) used may further vary and may need to be optimised any further.
  • a compound preferably an oligonucleotide to be used in the invention to prevent, treat DMD or BMD are synthetically produced and administered directly to a cell, a tissue, an organ and/or patients in formulated form in a pharmaceutically acceptable composition or preparation.
  • the delivery of a pharmaceutical composition to the subject is preferably carried out by one or more parenteral injections, e.g. intravenous and/or subcutaneous and/or intramuscular and/or intrathecal and/or intraventricular administrations, preferably injections, at one or at multiple sites in the human body.
  • a preferred oligonucleotide as defined herein optionally comprising one or more nucleotide analogs or equivalents of the invention modulates splicing in one or more muscle cells, including heart muscle cells, upon systemic delivery.
  • systemic delivery of an oligonucleotide comprising a specific nucleotide analog or equivalent might result in targeting a subset of muscle cells, while an oligonucleotide comprising a distinct nucleotide analog or equivalent might result in targeting of a different subset of muscle cells.
  • oligonucleotide comprising a specific nucleotide analog or equivalent
  • an oligonucleotide comprising a distinct nucleotide analog or equivalent might result in targeting a different subset of muscle cells. Therefore, in this embodiment, it is preferred to use a combination of oligonucleotides comprising different nucleotide analogs or equivalents for modulating splicing of the DMD mRNA in at least one type of muscle cells.
  • a molecule or a viral-based vector for use as a medicament, preferably for modulating splicing of the DMD pre-mRNA, more preferably for promoting or inducing skipping of any of exon 43, 46, 50-53 as identified herein.
  • the invention provides the use of an antisense oligonucleotide or molecule according to the invention, and/or a viral-based vector that expresses one or more antisense sequences according to the invention and/or a pharmaceutical composition, for modulating splicing of the DMD pre-mRNA.
  • the splicing is preferably modulated in a human myogenic cell or muscle cell in vitro. More preferred is that splicing is modulated in a human muscle cell in vivo.
  • the invention further relates to the use of the molecule as defined herein and/or the vector as defined herein and/or or the pharmaceutical composition as defined herein for modulating splicing of the DMD pre-mRNA or for the preparation of a medicament for the treatment of a DMD or BMD patient.
  • the verb “to comprise” and its conjugations is used in its non-limiting sense to mean that items following the word are included, but items not specifically mentioned are not excluded.
  • the verb “to consist” may be replaced by “to consist essentially of” meaning that a molecule or a viral-based vector or a composition as defined herein may comprise additional component(s) than the ones specifically identified, said additional component(s) not altering the unique characteristic of the invention.
  • reference to an element by the indefinite article “a” or “an” does not exclude the possibility that more than one of the element is present, unless the context clearly requires that there be one and only one of the elements.
  • the indefinite article “a” or “an” thus usually means “at least one”.
  • AON design was based on (partly) overlapping open secondary structures of the target exon RNA as predicted by the m-fold program, on (partly) overlapping putative SR-protein binding sites as predicted by the ESE-finder software.
  • AONs were synthesized by Prosensa Therapeutics B.V. (Leiden, Netherlands), and contain 2′-O-methyl RNA and full-length phosphorothioate (PS) backbones.
  • Myotube cultures derived from a healthy individual (“human control”) (examples 1, 3, and 4; exon 43, 50, 52 skipping) or a DMD patient carrying an exon 45 deletion (example 2; exon 46 skipping) were processed as described previously (Aartsma-Rus et al., Neuromuscul. Disord. 2002; 12: S71-77 and Hum Mol Genet 2003; 12(8): 907-14).
  • myotube cultures were transfected with 50 nM and 150 nM (example 2), 200nM and 500 nM (example 4) or 500nM only (examples 1 and 3) of each AON.
  • Transfection reagent UNIFectylin Prosensa Therapeutics BV, Netherlands
  • 2 ⁇ l UNIFectylin per ⁇ g AON.
  • Exon skipping efficiencies were determined by nested RT-PCR analysis using primers in the exons flanking the targeted exons (43, 46, 50, 51, 52, or 53).
  • PCR fragments were isolated from agarose gels for sequence verification. For quantification, the PCR products were analyzed using the DNA 1000 LabChips Kit on the Agilent 2100 bioanalyzer (Agilent Technologies, USA).
  • a series of AONs targeting sequences within exon 43 were designed and transfected in healthy control myotube cultures. Subsequent RT-PCR and sequence analysis of isolated RNA demonstrated that almost all AONs targeting a continuous nucleotide stretch within exon 43 herein defined as SEQ ID NO 2, was indeed capable of inducing exon 43 skipping.
  • PS237 SEQ ID NO: 65
  • PS238 and PS240 are shown, inducing exon 43 skipping levels up to 13% and 36% respectively ( FIG. 1 ).
  • the precise skipping of exon 43 was confirmed by sequence analysis of the novel smaller transcript fragments. No exon 43 skipping was observed in non-treated cells (NT).
  • a series of AONs targeting sequences within exon 46 were designed and transfected in myotube cultures derived from a DMD patient carrying an exon 45 deletion in the DMD gene.
  • antisense-induced exon 46 skipping would induce the synthesis of a novel, BMD-like dystrophin protein that may indeed alleviate one or more symptoms of the disease.
  • Subsequent RT-PCR and sequence analysis of isolated RNA demonstrated that almost all AONs targeting a continuous nucleotide stretch within exon 46 herein defined as SEQ ID NO 3, was indeed capable of inducing exon 46 skipping, even at relatively low AON concentrations of 50 nM.
  • PS182 (SEQ ID NO: 117) reproducibly induced highest levels of exon 46 skipping (up to 50% at 50 nM and 74% at 150 nM), as shown in FIG. 2 .
  • PS177, PS179, and PS181 are shown, inducing exon 46 skipping levels up to 55%, 58% and 42% respectively at 150 nM ( FIG. 2 ).
  • the precise skipping of exon 46 was confirmed by sequence analysis of the novel smaller transcript fragments. No exon 46 skipping was observed in non-treated cells (NT).
  • a series of AONs targeting sequences within exon 50 were designed and transfected in healthy control myotube cultures. Subsequent RT-PCR and sequence analysis of isolated RNA demonstrated that almost all AONs targeting a continuous nucleotide stretch within exon 50 herein defined as SEQ ID NO 4, was indeed capable of inducing exon 50 skipping.
  • PS248 SEQ ID NO: 127) reproducibly induced highest levels of exon 50 skipping (up to 35% at 500 nM), as shown in FIG. 3 .
  • PS245, PS246, and PS247 are shown, inducing exon 50 skipping levels up to 14-16% at 500 nM ( FIG. 3 ).
  • the precise skipping of exon 50 was confirmed by sequence analysis of the novel smaller transcript fragments. No exon 50 skipping was observed in non-treated cells (NT).
  • a series of AONs targeting sequences within exon 51 were designed and transfected in healthy control myotube cultures. Subsequent RT-PCR and sequence analysis of isolated RNA demonstrated that almost all AONs targeting a continuous nucleotide stretch within exon 51 herein defined as SEQ ID NO 5, was indeed capable of inducing exon 51 skipping.
  • the AON with SEQ ID NO 180 reproducibly induced highest levels of exon 51 skipping (not shown).
  • a series of AONs targeting sequences within exon 52 were designed and transfected in healthy control myotube cultures. Subsequent RT-PCR and sequence analysis of isolated RNA demonstrated that almost all AONs targeting a continuous nucleotide stretch within exon 52 herein defined as SEQ ID NO 6, was indeed capable of inducing exon 52 skipping.
  • PS236 SEQ ID NO: 299 reproducibly induced highest levels of exon 52 skipping (up to 88% at 200 nM and 91% at 500 nM), as shown in FIG. 4 .
  • PS232 and AON 52-1 previously published by Aartsma-Rus et al.
  • Oligonucleotides 2005 are shown, inducing exon 52 skipping at levels up to 59% and 10% respectively when applied at 500 nM ( FIG. 4 ).
  • the precise skipping of exon 52 was confirmed by sequence analysis of the novel smaller transcript fragments. No exon 52 skipping was observed in non-treated cells (NT).
  • a series of AONs targeting sequences within exon 53 were designed and transfected in healthy control myotube cultures. Subsequent RT-PCR and sequence analysis of isolated RNA demonstrated that almost all AONs targeting a continuous nucleotide stretch within exon 53 herein defined as SEQ ID NO 7, was indeed capable of inducing exon 53 skipping.
  • the AON with SEQ ID NO 328 reproducibly induced highest levels of exon 53 skipping (not shown).
  • FIG. 1 In human control myotubes, a series of AONs (PS237, PS238, and PS240; SEQ ID NO 65, 66, 16 respectively) targeting exon 43 was tested at 500 nM.
  • FIG. 2 In myotubes from a DMD patient with an exon 45 deletion, a series of AONs (PS177, PS179, PS181, and PS182; SEQ ID NO 91, 70, 110, and 117 respectively) targeting exon 46 was tested at two different concentrations (50 and 150 nM). PS182 (SEQ ID NO 117) reproducibly induced highest levels of exon 46 skipping. (M: DNA size marker)
  • FIG. 3 In human control myotubes, a series of AONs (PS245, PS246, PS247, and PS248; SEQ ID NO 167, 165, 166, and 127 respectively) targeting exon 50 was tested at 500 nM.
  • PS248 SEQ ID NO 127) reproducibly induced highest levels of exon 50 skipping.
  • M DNA size marker
  • NT non-treated cells.
  • FIG. 4 In human control myotubes, two novel AONs (PS232 and PS236; SEQ ID NO 246 and 299 respectively) targeting exon 52 were tested at two different concentrations (200 and 500 nM) and directly compared to a previously described AON (52-1).
  • PS236 SEQ ID NO 299
  • M DNA size marker
  • NT non-treated cells

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • Neurology (AREA)
  • General Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Biochemistry (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Rheumatology (AREA)
  • Pain & Pain Management (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Marine Sciences & Fisheries (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Hematology (AREA)
  • Neurosurgery (AREA)
  • Diabetes (AREA)

Abstract

The invention relates a method wherein a molecule is used for inducing and/or promoting skipping of at least one of exon 43, exon 46, exons 50-53 of the DMD pre-mRNA in a patient, preferably in an isolated cell of a patient, the method comprising providing said cell and/or said patient with a molecule. The invention also relates to said molecule as such.

Description

    FIELD
  • This application is a continuation of U.S. application Ser. No. 14/631,686 filed on Feb. 25, 2015 which is a continuation of U.S. application Ser. No. 13/094,571 filed Apr. 26, 2011, which is a continuation of International Application No. PCT/NL2009/050113, filed on Mar. 11, 2009, which claims priority to PCT/NL2008/050673, filed on Oct. 27, 2008, the contents of each of which are herein incorporated by reference in their entirety. The invention relates to the field of genetics, more specifically human genetics. The invention in particular relates to modulation of splicing of the human Duchenne Muscular Dystrophy pre-mRNA.
  • The invention relates to the field of genetics, more specifically human genetics. The invention in particular relates to modulation of splicing of the human Duchenne Muscular Dystrophy pre-mRNA.
  • BACKGROUND OF THE INVENTION
  • Myopathies are disorders that result in functional impairment of muscles. Muscular dystrophy (MD) refers to genetic diseases that are characterized by progressive weakness and degeneration of skeletal muscles. Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD) are the most common childhood forms of muscular dystrophy. They are recessive disorders and because the gene responsible for DMD and BMD resides on the X-chromosome, mutations mainly affect males with an incidence of about 1 in 3500 boys.
  • DMD and BMD are caused by genetic defects in the DMD gene encoding dystrophin, a muscle protein that is required for interactions between the cytoskeleton and the extracellular matrix to maintain muscle fiber stability during contraction. DMD is a severe, lethal neuromuscular disorder resulting in a dependency on wheelchair support before the age of 12 and DMD patients often die before the age of thirty due to respiratory- or heart failure. In contrast, BMD patients often remain ambulatory until later in life, and have near normal life expectancies. DMD mutations in the DMD gene are characterized by frame shifting insertions or deletions or nonsense point mutations, resulting in the absence of functional dystrophin. BMD mutations in general keep the reading frame intact, allowing synthesis of a partly functional dystrophin.
  • During the last decade, specific modification of splicing in order to restore the disrupted reading frame of the dystrophin transcript has emerged as a promising therapy for Duchenne muscular dystrophy (DMD) (van Ommen, van Deutekom, Aartsma-Rus, Curr Opin Mol Ther. 2008;10(2):140-9, Yokota, Duddy, Partidge, Acta Myol. 2007;26(3):179-84, van Deutekom et al., N Engl J Med. 2007;357(26):2677-86).
  • Using antisense oligonucleotides (AONs) interfering with splicing signals the skipping of specific exons can be induced in the DMD pre-mRNA, thus restoring the open reading frame and converting the severe DMD into a milder BMD phenotype (van Deutekom et al. Hum Mol Genet. 2001; 10: 1547-54; Aartsma-Rus et al., Hum Mol Genet 2003; 12(8):907-14.). In vivo proof-of-concept was first obtained in the mdx mouse model, which is dystrophin-deficient due to a nonsense mutation in exon 23.
  • Intramuscular and intravenous injections of AONs targeting the mutated exon 23 restored dystrophin expression for at least three months (Lu et al. Nat Med. 2003; 8: 1009-14; Lu et al., Proc Natl Acad Sci U S A. 2005;102(1):198-203). This was accompanied by restoration of dystrophin-associated proteins at the fiber membrane as well as functional improvement of the treated muscle. In vivo skipping of human exons has also been achieved in the hDMD mouse model, which contains a complete copy of the human DMD gene integrated in chromosome 5 of the mouse (Bremmer-Bout et al. Molecular Therapy. 2004; 10: 232-40; 't Hoen et al. J Biol Chem. 2008; 283: 5899-907).
  • Recently, a first-in-man study was successfully completed where an AON inducing the skipping of exon 51 was injected into a small area of the tibialis anterior muscle of four DMD patients. Novel dystrophin expression was observed in the majority of muscle fibers in all four patients treated, and the AON was safe and well tolerated (van Deutekom et al. N Engl J Med. 2007; 357: 2677-86).
  • DESCRIPTION OF THE INVENTION Method
  • In a first aspect, the present invention provides a method for inducing, and/or promoting skipping of at least one of exons 43, 46, 50-53 of the DMD pre-mRNA in a patient, preferably in an isolated cell of a patient, the method comprising providing said cell and/or said patient with a molecule that binds to a continuous stretch of at least 8 nucleotides within said exon. It is to be understood that said method encompasses an in vitro, in vivo or ex vivo method.
  • Accordingly, a method is provided for inducing and/or promoting skipping of at least one of exons 43, 46, 50-53 of DMD pre-mRNA in a patient, preferably in an isolated cell of said patient, the method comprising providing said cell and/or said patient with a molecule that binds to a continuous stretch of at least 8 nucleotides within said exon.
  • As defined herein a DMD pre-mRNA preferably means the pre-mRNA of a DMD gene of a DMD or BMD patient.
  • A patient is preferably intended to mean a patient having DMD or BMD as later defined herein or a patient susceptible to develop DMD or BMD due to his or her genetic background. In the case of a DMD patient, an oligonucleotide used will preferably correct one mutation as present in the DMD gene of said patient and therefore will preferably create a DMD protein that will look like a BMD protein: said protein will preferably be a functional dystrophin as later defined herein. In the case of a BMD patient, an oligonucleotide as used will preferably correct one mutation as present in the BMD gene of said patient and therefore will preferably create a dystrophin which will be more functional than the dystrophin which was originally present in said BMD patient.
  • Exon skipping refers to the induction in a cell of a mature mRNA that does not contain a particular exon that is normally present therein. Exon skipping is performed by providing a cell expressing the pre-mRNA of said mRNA with a molecule capable of interfering with essential sequences such as for example the splice donor of splice acceptor sequence that required for splicing of said exon, or a molecule that is capable of interfering with an exon inclusion signal that is required for recognition of a stretch of nucleotides as an exon to be included in the mRNA. The term pre-mRNA refers to a non-processed or partly processed precursor mRNA that is synthesized from a DNA template in the cell nucleus by transcription.
  • Within the context of the invention, inducing and/or promoting skipping of an exon as indicated herein means that at least 1%, 10%, 20%, 30%, 40%,50%, 60%, 70%, 80%, 90% or more of the DMD mRNA in one or more (muscle) cells of a treated patient will not contain said exon. This is preferably assessed by PCR as described in the examples.
  • Preferably, a method of the invention by inducing and/or promoting skipping of at least one of the following exons 43, 46, 50-53 of the DMD pre-mRNA in one or more (muscle) cells of a patient, provides said patient with a functional dystrophin protein and/or decreases the production of an aberrant dystrophin protein in said patient and/or increases the production of a functional dystrophin is said patient.
  • Providing a patient with a functional dystrophin protein and/or decreasing the production of an aberrant dystrophin protein in said patient is typically applied in a DMD patient. Increasing the production of a functional dystrophin is typically applied in a BMD patient.
  • Therefore a preferred method is a method, wherein a patient or one or more cells of said patient is provided with a functional dystrophin protein and/or wherein the production of an aberrant dystrophin protein in said patient is decreased and/or wherein the production of a functional dystrophin is increased in said patient, wherein the level of said aberrant or functional dystrophin is assessed by comparison to the level of said dystrophin in said patient at the onset of the method.
  • Decreasing the production of an aberrant dystrophin may be assessed at the mRNA level and preferably means that 99%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 10%, 5% or less of the initial amount of aberrant dystrophin mRNA, is still detectable by RT PCR. An aberrant dystrophin mRNA or protein is also referred to herein as a non-functional dystrophin mRNA or protein. A non functional dystrophin protein is preferably a dystrophin protein which is not able to bind actin and/or members of the DGC protein complex. A non-functional dystrophin protein or dystrophin mRNA does typically not have, or does not encode a dystrophin protein with an intact C-terminus of the protein.
  • Increasing the production of a functional dystrophin in said patient or in a cell of said patient may be assessed at the mRNA level (by RT-PCR analysis) and preferably means that a detectable amount of a functional dystrophin mRNA is detectable by RT PCR. In another embodiment, 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more of the detectable dystrophin mRNA is a functional dystrophin mRNA.
  • Increasing the production of a functional dystrophin in said patient or in a cell of said patient may be assessed at the protein level (by immunofluorescence and western blot analyses) and preferably means that a detectable amount of a functional dystrophin protein is detectable by immunofluorescence or western blot analysis. In another embodiment, 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more of the detectable dystrophin protein is a functional dystrophin protein.
  • As defined herein, a functional dystrophin is preferably a wild type dystrophin corresponding to a protein having the amino acid sequence as identified in SEQ ID NO: 1. A functional dystrophin is preferably a dystrophin, which has an actin binding domain in its N terminal part (first 240 amino acids at the N terminus), a cystein-rich domain (amino acid 3361 till 3685) and a C terminal domain (last 325 amino acids at the C terminus) each of these domains being present in a wild type dystrophin as known to the skilled person. The amino acids indicated herein correspond to amino acids of the wild type dystrophin being represented by SEQ ID NO:1. In other words, a functional dystrophin is a dystrophin which exhibits at least to some extent an activity of a wild type dystrophin. “At least to some extent” preferably means at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or 100% of a corresponding activity of a wild type functional dystrophin. In this context, an activity of a functional dystrophin is preferably binding to actin and to the dystrophin-associated glycoprotein complex (DGC) (Aartsma-Rus A et al, (2006), Entries in the leiden Duchenne Muscular Dystrophy mutation database: an overview of mutation types and paradoxical cases that confirm the reading-frame rule, Muscle Nerve, 34: 135-144). Binding of dystrophin to actin and to the DGC complex may be visualized by either co-immunoprecipitation using total protein extracts or immunofluorescence analysis of cross-sections, from a muscle biopsy, as known to the skilled person.
  • Individuals or patients suffering from Duchenne muscular dystrophy typically have a mutation in the gene encoding dystrophin that prevent synthesis of the complete protein, i.e of a premature stop prevents the synthesis of the C-terminus. In Becker muscular dystrophy the DMD gene also comprises a mutation compared tot the wild type gene but the mutation does typically not induce a premature stop and the C-terminus is typically synthesized. As a result a functional dystrophin protein is synthesized that has at least the same activity in kind as the wild type protein, not although not necessarily the same amount of activity. The genome of a BMD individual typically encodes a dystrophin protein comprising the N terminal part (first 240 amino acids at the N terminus), a cystein-rich domain (amino acid 3361 till 3685) and a C terminal domain (last 325 amino acids at the C terminus) but its central rod shaped domain may be shorter than the one of a wild type dystrophin (Aartsma-Rus A et al, (2006), Entries in the leiden Duchenne Muscular Dystrophy mutation database: an overview of mutation types and paradoxical cases that confirm the reading-frame rule, Muscle Nerve, 34: 135-144). Exon skipping for the treatment of DMD is typically directed to overcome a premature stop in the pre-mRNA by skipping an exon in the rod-shaped domain to correct the reading frame and allow synthesis of remainder of the dystrophin protein including the C-terminus, albeit that the protein is somewhat smaller as a result of a smaller rod domain. In a preferred embodiment, an individual having DMD and being treated by a method as defined herein will be provided a dystrophin which exhibits at least to some extent an activity of a wild type dystrophin. More preferably, if said individual is a Duchenne patient or is suspected to be a Duchenne patient, a functional dystrophin is a dystrophin of an individual having BMD: typically said dystrophin is able to interact with both actin and the DGC, but its central rod shaped domain may be shorter than the one of a wild type dystrophin (Aartsma-Rus A et al, (2006), Entries in the leiden Duchenne Muscular Dystrophy mutation database: an overview of mutation types and paradoxical cases that confirm the reading-frame rule, Muscle Nerve, 34: 135-144). The central rod-shaped domain of wild type dystrophin comprises 24 spectrin-like repeats (Aartsma-Rus A et al, (2006), Entries in the leiden Duchenne Muscular Dystrophy mutation database: an overview of mutation types and paradoxical cases that confirm the reading-frame rule, Muscle Nerve, 34: 135-144). For example, a central rod-shaped domain of a dystrophin as provided herein may comprise 5 to 23, 10 to 22 or 12 to 18 spectrin-like repeats as long as it can bind to actin and to DGC.
  • A method of the invention may alleviate one or more characteristics of a myogenic or muscle cell of a patient or alleviate one or more symptoms of a DMD patient having a deletion including but not limited to exons 44, 44-46, 44-47, 44-48, 44-49, 44-51, 44-53 (correctable by exon 43 skipping), 19-45, 21-45, 43-45, 45, 47-54, 47-56 (correctable by exon 46 skipping), 51, 51-53, 51-55, 51-57 (correctable by exon 50 skipping), 13-50, 19-50, 29-50, 43-50, 45-50, 47-50, 48-50, 49-50, 50, 52 (correctable by exon 51 skipping), exons 8-51, 51, 53, 53-55, 53-57, 53-59, 53-60, (correctable by exon 52 skipping) and exons 10-52, 42-52, 43-52, 45-52, 47-52, 48-52, 49-52, 50-52, 52 (correctable by exon 53 skipping) in the DMD gene, occurring in a total of 68% of all DMD patients with a deletion (Aartsma-Rus et al., Hum. Mut. 2009).
  • Alternatively, a method of the invention may improve one or more characteristics of a muscle cell of a patient or alleviate one or more symptoms of a DMD patient having small mutations in, or single exon duplications of exon 43, 46, 50-53 in the DMD gene, occurring in a total of 36% of all DMD patients with a deletion (Aartsma-Rus et al, Hum. Mut. 2009)
  • Furthermore, for some patients the simultaneous skipping of one of more exons in addition to exon 43, exon 46 and/or exon 50-53 is required to restore the open reading frame, including patients with specific deletions, small (point) mutations, or double or multiple exon duplications, such as (but not limited to) a deletion of exons 44-50 requiring the co-skipping of exons 43 and 51, with a deletion of exons 46-50 requiring the co-skipping of exons 45 and 51, with a deletion of exons 44-52 requiring the co-skipping of exons 43 and 53, with a deletion of exons 46-52 requiring the co-skipping of exons 45 and 53, with a deletion of exons 51-54 requiring the co-skipping of exons 50 and 55, with a deletion of exons 53-54 requiring the co-skipping of exons 52 and 55, with a deletion of exons 53-56 requiring the co-skipping of exons 52 and 57, with a nonsense mutation in exon 43 or exon 44 requiring the co-skipping of exon 43 and 44, with a nonsense mutation in exon 45 or exon 46 requiring the co-skipping of exon 45 and 46, with a nonsense mutation in exon 50 or exon 51 requiring the co-skipping of exon 50 and 51, with a nonsense mutation in exon 51 or exon 52 requiring the co-skipping of exon 51 and 52, with a nonsense mutation in exon 52 or exon 53 requiring the co-skipping of exon 52 and 53, or with a double or multiple exon duplication involving exons 43, 46, 50, 51, 52, and/or 53.
  • In a preferred method, the skipping of exon 43 is induced, or the skipping of exon 46 is induced, or the skipping of exon 50 is induced or the skipping of exon 51 is induced or the skipping of exon 52 is induced or the skipping of exon 53 is induced. An induction of the skipping of two of these exons is also encompassed by a method of the invention.
  • For example, preferably skipping of exons 50 and 51, or 52 and 53, or 43 and 51, or 43 and 53, or 51 and 52. Depending on the type and the identity (the specific exons involved) of mutation identified in a patient, the skilled person will know which combination of exons needs to be skipped in said patient.
  • In a preferred method, one or more symptom(s) of a DMD or a BMD patient is/are alleviated and/or one or more characteristic(s) of one or more muscle cells from a DMD or a BMD patient is/are improved. Such symptoms or characteristics may be assessed at the cellular, tissue level or on the patient self
  • An alleviation of one or more characteristics may be assessed by any of the following assays on a myogenic cell or muscle cell from a patient: reduced calcium uptake by muscle cells, decreased collagen synthesis, altered morphology, altered lipid biosynthesis, decreased oxidative stress, and/or improved muscle fiber function, integrity, and/or survival. These parameters are usually assessed using immunofluorescence and/or histochemical analyses of cross sections of muscle biopsies.
  • The improvement of muscle fiber function, integrity and/or survival may be assessed using at least one of the following assays: a detectable decrease of creatine kinase in blood, a detectable decrease of necrosis of muscle fibers in a biopsy cross-section of a muscle suspected to be dystrophic, and/or a detectable increase of the homogeneity of the diameter of muscle fibers in a biopsy cross-section of a muscle suspected to be dystrophic. Each of these assays is known to the skilled person.
  • Creatine kinase may be detected in blood as described in Hodgetts et al (Hodgetts S., et al, (2006), Neuromuscular Disorders, 16: 591-602.2006). A detectable decrease in creatine kinase may mean a decrease of 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more compared to the concentration of creatine kinase in a same DMD or BMD patient before treatment.
  • A detectable decrease of necrosis of muscle fibers is preferably assessed in a muscle biopsy, more preferably as described in Hodgetts et al (Hodgetts S., et al, (2006), Neuromuscular Disorders, 16: 591-602.2006) using biopsy cross-sections. A detectable decrease of necrosis may be a decrease of 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more of the area wherein necrosis has been identified using biopsy cross-sections. The decrease is measured by comparison to the necrosis as assessed in a same DMD or BMD patient before treatment.
  • A detectable increase of the homogeneity of the diameter of a muscle fiber is preferably assessed in a muscle biopsy cross-section, more preferably as described in Hodgetts et al (Hodgetts S., et al, (2006), Neuromuscular Disorders, 16: 591-602.2006).
  • The increase is measured by comparison to the homogeneity of the diameter of a muscle fiber in a same DMD or BMD patient before treatment
  • An alleviation of one or more symptoms may be assessed by any of the following assays on the patient self: prolongation of time to loss of walking, improvement of muscle strength, improvement of the ability to lift weight, improvement of the time taken to rise from the floor, improvement in the nine-meter walking time, improvement in the time taken for four-stairs climbing, improvement of the leg function grade, improvement of the pulmonary function, improvement of cardiac function, improvement of the quality of life. Each of these assays is known to the skilled person.
  • As an example, the publication of Manzur at al (Manzur AY et al, (2008), Glucocorticoid corticosteroids for Duchenne muscular dystrophy (review), Wiley publishers, The Cochrane collaboration.) gives an extensive explanation of each of these assays. For each of these assays, as soon as a detectable improvement or prolongation of a parameter measured in an assay has been found, it will preferably mean that one or more symptoms of Duchenne Muscular Dystrophy or Becker Muscular Dystrophy has been alleviated in an individual using a method of the invention. Detectable improvement or prolongation is preferably a statistically significant improvement or prolongation as described in Hodgetts et al (Hodgetts S., et al, (2006), Neuromuscular Disorders,16: 591-602.2006). Alternatively, the alleviation of one or more symptom(s) of Duchenne Muscular Dystrophy or Becker Muscular Dystrophy may be assessed by measuring an improvement of a muscle fiber function, integrity and/or survival as later defined herein.
  • A treatment in a method according to the invention may have a duration of at least one week, at least one month, at least several months, at least one year, at least 2, 3, 4, 5, 6 years or more.
  • Each molecule or oligonucleotide or equivalent thereof as defined herein for use according to the invention may be suitable for direct administration to a cell, tissue and/or an organ in vivo of individuals affected by or at risk of developing DMD or BMD, and may be administered directly in vivo, ex vivo or in vitro. The frequency of administration of a molecule or an oligonucleotide or a composition of the invention may depend on several parameters such as the age of the patient, the mutation of the patient, the number of molecules (dose), the formulation of said molecule. The frequency may be ranged between at least once in a two weeks, or three weeks or four weeks or five weeks or a longer time period.
  • A molecule or oligonucleotide or equivalent thereof can be delivered as is to a cell.
  • When administering said molecule, oligonucleotide or equivalent thereof to an individual, it is preferred that it is dissolved in a solution that is compatible with the delivery method. For intravenous, subcutaneous, intramuscular, intrathecal and/or intraventricular administration it is preferred that the solution is a physiological salt solution. Particularly preferred for a method of the invention is the use of an excipient that will further enhance delivery of said molecule, oligonucleotide or functional equivalent thereof as defined herein, to a cell and into a cell, preferably a muscle cell.
  • Preferred excipientare defined in the section entitled “pharmaceutical composition”.
  • In a preferred method of the invention, an additional molecule is used which is able to induce and/or promote skipping of another exon of the DMD pre-mRNA of a patient.
  • Preferably, the second exon is selected from: exon 6, 7, 11, 17, 19, 21, 43, 44, 45, 50, 51, 52, 53, 55, 57, 59, 62, 63, 65, 66, 69, or 75 of the DMD pre-mRNA of a patient.
  • Molecules which can be used are depicted in any one of Table 1 to 7. This way, inclusion of two or more exons of a DMD pre-mRNA in mRNA produced from this pre-mRNA is prevented. This embodiment is further referred to as double- or multi-exon skipping (Aartsma-Rus A, Janson AA, Kaman WE, et al. Antisense-induced multiexon skipping for Duchenne muscular dystrophy makes more sense. Am J Hum Genet 2004;74(1):83-92, Aartsma-Rus A, Kaman W E, Weij R, den Dunnen J T, van Ommen G J, van Deutekom J C. Exploring the frontiers of therapeutic exon skipping for Duchenne muscular dystrophy by double targeting within one or multiple exons. Mol Ther 2006;14(3):401-7). In most cases double-exon skipping results in the exclusion of only the two targeted exons from the DMD pre-mRNA. However, in other cases it was found that the targeted exons and the entire region in between said exons in said pre-mRNA were not present in the produced mRNA even when other exons (intervening exons) were present in such region. This multi-skipping was notably so for the combination of oligonucleotides derived from the DMD gene, wherein one oligonucleotide for exon 45 and one oligonucleotide for exon 51 was added to a cell transcribing the DMD gene. Such a set-up resulted in mRNA being produced that did not contain exons 45 to 51. Apparently, the structure of the pre-mRNA in the presence of the mentioned oligonucleotides was such that the splicing machinery was stimulated to connect exons 44 and 52 to each other.
  • It is possible to specifically promote the skipping of also the intervening exons by providing a linkage between the two complementary oligonucleotides. Hence, in one embodiment stretches of nucleotides complementary to at least two dystrophin exons are separated by a linking moiety. The at least two stretches of nucleotides are thus linked in this embodiment so as to form a single molecule.
  • In case, more than one compounds or molecules are used in a method of the invention, said compounds can be administered to an individual in any order. In one embodiment, said compounds are administered simultaneously (meaning that said compounds are administered within 10 hours, preferably within one hour). This is however not necessary. In another embodiment, said compounds are administered sequentially.
  • Molecule
  • In a second aspect, there is provided a molecule for use in a method as described in the previous section entitled “Method”. A molecule as defined herein is preferably an oligonucleotide or antisense oligonucleotide (AON).
  • It was found by the present investigators that any of exon 43, 46, 50-53 is specifically skipped at a high frequency using a molecule that preferably binds to a continuous stretch of at least 8 nucleotides within said exon. Although this effect can be associated with a higher binding affinity of said molecule, compared to a molecule that binds to a continuous stretch of less than 8 nucleotides, there could be other intracellular parameters involved that favor thermodynamic, kinetic, or structural characteristics of the hybrid duplex. In a preferred embodiment, a molecule that binds to a continuous stretch of at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37,38, 39, 40, 41, 42, 43,44, 45, 46, 47, 48, 49, 50 nucleotides within said exon is used.
  • In a preferred embodiment, a molecule or an oligonucleotide of the invention which comprises a sequence that is complementary to a part of any of exon 43, 46, 50-53 of DMD pre-mRNA is such that the complementary part is at least 50% of the length of the oligonucleotide of the invention, more preferably at least 60%, even more preferably at least 70%, even more preferably at least 80%, even more preferably at least 90% or even more preferably at least 95%, or even more preferably 98% and most preferably up to 100%. “A part of said exon” preferably means a stretch of at least 8 nucleotides. In a most preferred embodiment, an oligonucleotide of the invention consists of a sequence that is complementary to part of said exon DMD pre-mRNA as defined herein. For example, an oligonucleotide may comprise a sequence that is complementary to part of said exon DMD pre-mRNA as defined herein and additional flanking sequences. In a more preferred embodiment, the length of said complementary part of said oligonucleotide is of at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27 , 28 , 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50 nucleotides. Preferably, additional flanking sequences are used to modify the binding of a protein to said molecule or oligonucleotide, or to modify a thermodynamic property of the oligonucleotide, more preferably to modify target RNA binding affinity.
  • A preferred molecule to be used in a method of the invention binds or is complementary to a continuous stretch of at least 8 nucleotides within one of the following nucleotide sequences selected from:
  • (SEQ ID NO: 2)
    5′-AGAUAGUCUACAACAAAGCUCAGGUCGGAUUGACAUUAUUCAU
    AGCAAGAAGACAGCAGCAUUGCAAAGUGCAACGCCUGUGG-3′ for
    skipping of exon 43;
    (SEQ ID NO: 3)
    5′-UUAUGGUUGGAGGAAGCAGAUAACAUUGCUAGUAUCCCACUUG
    AACCUGGAAAAGAGCAGCAACUAAAAGAAAAGC-3′ for
    skipping of exon 46;
    (SEQ ID NO: 4)
    5′-GGCGGTAAACCGUUUACUUCAAGAGCUGAGGGCAAAGCAGCCUG
    ACCUAGC UCCUGGACUGACCACUAUUGG-3′ for skipping of
    exon 50;
    (SEQ ID NO: 5)
    5′-CUCCUACUCAGACUGUUACUCUGGUGACACAACCUGUGGUUACU
    AAGGAAACUGCCAUC UCCAAACUAGAAAUGCCAUCUUCCUUGAUG
    UUGGAGGUAC-3′ for skipping of exon 51;
    (SEQ ID NO: 6)
    5′-AUGCAGGAUUUGGAACAGAGGCGUCCCCAGUUGGAAGAACUCAU
    UACCGCUGCCCAAAAUUUGAAAAACAAGACCAGCAAUCAAGAGGCU-3′
    for skipping of exon 52,
    and
    (SEQ ID NO: 7)
    5′-AAAUGUUAAAGGAUUCAACACAAUGGCUGGAAGCUAAGGAAGAA
    GCUGAGCAGGUCUUAGGACAGGCCAGAG-3′ for skipping of
    exon 53.
  • Of the numerous molecules that theoretically can be prepared to bind to the continuous nucleotide stretches as defined by SEQ ID NO 2-7 within one of said exons, the invention provides distinct molecules that can be used in a method for efficiently skipping of at least one of exon 43, exon 46 and/or exon 50-53. Although the skipping effect can be addressed to the relatively high density of putative SR protein binding sites within said stretches, there could be other parameters involved that favor uptake of the molecule or other, intracellular parameters such as thermodynamic, kinetic, or structural characteristics of the hybrid duplex.
  • It was found that a molecule that binds to a continuous stretch comprised within or consisting of any of SEQ ID NO 2-7 results in highly efficient skipping of exon 43, exon 46 and/or exon 50- 53 respectively in a cell and/or in a patient provided with this molecule. Therefore, in a preferred embodiment, a method is provided wherein a molecule binds to a continuous stretch of at least 8, 10, 12, 15, 18, 20, 25, 30, 35, 40, 45, 50 nucleotides within SEQ ID NO 2-7.
  • In a preferred embodiment for inducing and/or promoting the skipping of any of exon 43, exon 46 and/or exon 50-53, the invention provides a molecule comprising or consisting of an antisense nucleotide sequence selected from the antisense nucleotide sequences depicted in any of Tables 1 to 6. A molecule of the invention preferably comprises or consist of the antisense nucleotide sequence of SEQ ID NO 16, SEQ ID NO 65, SEQ ID NO 70, SEQ ID NO 91, SEQ ID NO 110, SEQ ID NO 117, SEQ ID NO 127, SEQ ID NO 165, SEQ ID NO 166, SEQ ID NO 167, SEQ ID NO 246, SEQ ID NO 299, SEQ ID NO:357.
  • A preferred molecule of the invention comprises a nucleotide-based or nucleotide or an antisense oligonucleotide sequence of between 8 and 50 nucleotides or bases, more preferred between 10 and 50 nucleotides, more preferred between 20 and 40 nucleotides, more preferred between 20 and 30 nucleotides, such as 20 nucleotides, 21 nucleotides, 22 nucleotides, 23 nucleotides, 24 nucleotides, 25 nucleotides, 26 nucleotides, 27 nucleotides, 28 nucleotides, 29 nucleotides, 30 nucleotides, 31 nucleotides, 32 nucleotides, 33 nucleotides, 34 nucleotides, 35 nucleotides, 36 nucleotides, 37 nucleotides, 38 nucleotides, 39 nucleotides, 40 nucleotides, 41 nucleotides, 42 nucleotides, 43 nucleotides, 44 nucleotides, 45 nucleotides, 46 nucleotides, 47 nucleotides, 48 nucleotides, 49 nucleotides or 50 nucleotides.
  • A most preferred molecule of the invention comprises a nucleotide-based sequence of 25 nucleotides.
  • Furthermore, none of the indicated sequences is derived from conserved parts of splice-junction sites. Therefore, said molecule is not likely to mediate differential splicing of other exons from the DMD pre-mRNA or exons from other genes.
  • In one embodiment, a molecule of the invention is a compound molecule that binds to the specified sequence, or a protein such as an RNA-binding protein or a non-natural zinc-finger protein that has been modified to be able to bind to the corresponding nucleotide sequence on a DMD pre-RNA molecule. Methods for screening compound molecules that bind specific nucleotide sequences are, for example, disclosed in PCT/NL01/00697 and U.S. Pat. No. 6,875,736, which are herein incorporated by reference. Methods for designing RNA-binding Zinc-finger proteins that bind specific nucleotide sequences are disclosed by Friesen and Darby, Nature Structural Biology 5: 543-546 (1998) which is herein incorporated by reference.
  • A preferred molecule of the invention binds to at least part of the sequence of SEQ ID NO 2:5′-AGAUAGUCUACAACAAAGCUCAGGUCGGAUUGACAUUAUUCAU AGCAAGAAGACAGCAGCAUUGCAAAGUGCAACGCCUGUGG-3′ which is present in exon 43 of the DMD gene. More preferably, the invention provides a molecule comprising or consisting of the antisense nucleotide sequence of SEQ ID NO 8 to SEQ ID NO 69.
  • In an even more preferred embodiment, the invention provides a molecule comprising or consisting of the antisense nucleotide sequence of SEQ ID NO 16 and/or SEQ ID NO 65.
  • In a most preferred embodiment, the invention provides a molecule comprising or consisting of the antisense nucleotide sequence of SEQ ID NO 65. It was found that this molecule is very efficient in modulating splicing of exon 43 of the DMD pre-mRNA in a muscle cell and/or in a patient.
  • Another preferred molecule of the invention binds to at least part of the sequence of SEQ ID NO 3: 5′-UUAUGGUUGGAGGAAGCAGAUAACAUUGCUAGUAUCCCACUUG AACCUGGAAAAGAGCAGCAACUAAAAGAAAAGC-3′ which is present in exon 46 of the DMD gene. More preferably, the invention provides a molecule comprising or consisting of the antisense nucleotide sequence of SEQ ID NO 70 to SEQ ID NO 122.
  • In an even more preferred embodiment, the invention provides a molecule comprising or consisting of the antisense nucleotide sequence of SEQ ID NO 70, SEQ ID NO 91, SEQ ID NO 110, and/or SEQ ID NO117.
  • In a most preferred embodiment, the invention provides a molecule comprising or consisting of the antisense nucleotide sequence of SEQ ID NO 117. It was found that this molecule is very efficient in modulating splicing of exon 46 of the DMD pre-mRNA in a muscle cell or in a patient.
  • Another preferred molecule of the invention binds to at least part of the sequence of SEQ ID NO 4: 5′-GGCGGTAAACCGUUUACUUCAAGAGCU GAGGGCAAAGCAGCCUG ACCUAGCUCCUGGACUGACCACUAUUGG-3′ which is present in exon 50 of the DMD gene. More preferably, the invention provides a molecule comprising or consisting of the antisense nucleotide sequence of SEQ ID NO 123 to SEQ ID NO 167 and/or SEQ ID NO 529 to SEQ ID NO 535.
  • In an even more preferred embodiment, the invention provides a molecule comprising or consisting of the antisense nucleotide sequence of SEQ ID NO 127, or SEQ ID NO 165, or SEQ ID NO 166 and/or SEQ ID NO 167.
  • In a most preferred embodiment, the invention provides a molecule comprising or consisting of the antisense nucleotide sequence of SEQ ID NO 127. It was found that this molecule is very efficient in modulating splicing of exon 50 of the DMD pre-mRNA in a muscle cell and/or in a patient.
  • Another preferred molecule of the invention binds to at least part of the sequence of SEQ ID NO 5: 5′-CUCCUACUCAGACUGUUACUCUGGUGACACAACCUGUGGUUACU AAGGAAACUGCCAUC UCCAAACUAGAAAUGCCAUCUUCCUUGAUG UUGGAGGUAC-3′ which is present in exon 51 of the DMD gene. More preferably, the invention provides a molecule comprising or consisting of the antisense nucleotide sequence of SEQ ID NO 168 to SEQ ID NO 241.
  • Another preferred molecule of the invention binds to at least part of the sequence of SEQ ID NO 6: 5′-AUGCAGGAUUUGGAACAGAGGCGUCCCCAGUUGGAAGAACUCAU UACCGCUGCCCAAAAUUUGAAAAACAAGACCAGCAAUCAAGAGGCU-3′ which is present in exon 52 of the DMD gene. More preferably, the invention provides a molecule comprising or consisting of the antisense nucleotide sequence of SEQ ID NO 242 to SEQ ID NO 310. In an even more preferred embodiment, the invention provides a molecule comprising or consisting of the antisense nucleotide sequence of SEQ ID NO 246 and/or SEQ ID NO 299. In a most preferred embodiment, the invention provides a molecule comprising or consisting of the antisense nucleotide sequence of SEQ ID NO 299. It was found that this molecule is very efficient in modulating splicing of exon 52 of the DMD pre-mRNA in a muscle cell and/or in a patient.
  • Another preferred molecule of the invention binds to at least part of the sequence of SEQ ID NO 7: 5′-AAAUGUUAAAGGAUUCAACACAAUGGCUGGAAGCUAAGGAAGAA GCUGAGCAGGUCUUAGGACAGGCCAGAG-3′ which is present in exon 53 of the DMD gene. More preferably, the invention provides a molecule comprising or consisting of the antisense nucleotide sequence of SEQ ID NO 311 to SEQ ID NO 358.
  • In a most preferred embodiment, the invention provides a molecule comprising or consisting of the antisense nucleotide sequence of SEQ ID NO 357. It was found that this molecule is very efficient in modulating splicing of exon 53 of the DMD pre-mRNA in a muscle cell and/or in a patient.
  • A nucleotide sequence of a molecule of the invention may contain RNA residues, or one or more DNA residues, and/or one or more nucleotide analogues or equivalents, as will be further detailed herein below.
  • It is preferred that a molecule of the invention comprises one or more residues that are modified to increase nuclease resistance, and/or to increase the affinity of the antisense nucleotide for the target sequence. Therefore, in a preferred embodiment, the antisense nucleotide sequence comprises at least one nucleotide analogue or equivalent, wherein a nucleotide analogue or equivalent is defined as a residue having a modified base, and/or a modified backbone, and/or a non-natural internucleoside linkage, or a combination of these modifications.
  • In a preferred embodiment, the nucleotide analogue or equivalent comprises a modified backbone. Examples of such backbones are provided by morpholino backbones, carbamate backbones, siloxane backbones, sulfide, sulfoxide and sulfone backbones, formacetyl and thioformacetyl backbones, methyleneformacetyl backbones, riboacetyl backbones, alkene containing backbones, sulfamate, sulfonate and sulfonamide backbones, methyleneimino and methylenehydrazino backbones, and amide backbones.
  • Phosphorodiamidate morpholino oligomers are modified backbone oligonucleotides that have previously been investigated as antisense agents. Morpholino oligonucleotides have an uncharged backbone in which the deoxyribose sugar of DNA is replaced by a six membered ring and the phosphodiester linkage is replaced by a phosphorodiamidate linkage. Morpholino oligonucleotides are resistant to enzymatic degradation and appear to function as antisense agents by arresting translation or interfering with pre-mRNA splicing rather than by activating RNase H. Morpholino oligonucleotides have been successfully delivered to tissue culture cells by methods that physically disrupt the cell membrane, and one study comparing several of these methods found that scrape loading was the most efficient method of delivery; however, because the morpholino backbone is uncharged, cationic lipids are not effective mediators of morpholino oligonucleotide uptake in cells. A recent report demonstrated triplex formation by a morpholino oligonucleotide and, because of the non-ionic backbone, these studies showed that the morpholino oligonucleotide was capable of triplex formation in the absence of magnesium.
  • It is further preferred that that the linkage between the residues in a backbone do not include a phosphorus atom, such as a linkage that is formed by short chain alkyl or cycloalkyl internucleoside linkages, mixed heteroatom and alkyl or cycloalkyl internucleoside linkages, or one or more short chain heteroatomic or heterocyclic internucleoside linkages.
  • A preferred nucleotide analogue or equivalent comprises a Peptide Nucleic Acid (PNA), having a modified polyamide backbone (Nielsen, et al. (1991) Science 254, 1497-1500). PNA-based molecules are true mimics of DNA molecules in terms of base-pair recognition. The backbone of the PNA is composed of N-(2-aminoethyl)-glycine units linked by peptide bonds, wherein the nucleobases are linked to the backbone by methylene carbonyl bonds. An alternative backbone comprises a one-carbon extended pyrrolidine PNA monomer (Govindaraju and Kumar (2005) Chem. Commun, 495-497). Since the backbone of a PNA molecule contains no charged phosphate groups, PNA-RNA hybrids are usually more stable than RNA-RNA or RNA-DNA hybrids, respectively (Egholm et al (1993) Nature 365, 566-568).
  • A further preferred backbone comprises a morpholino nucleotide analog or equivalent, in which the ribose or deoxyribose sugar is replaced by a 6-membered morpholino ring.
  • A most preferred nucleotide analog or equivalent comprises a phosphorodiamidate morpholino oligomer (PMO), in which the ribose or deoxyribose sugar is replaced by a 6-membered morpholino ring, and the anionic phosphodiester linkage between adjacent morpholino rings is replaced by a non-ionic phosphorodiamidate linkage.
  • In yet a further embodiment, a nucleotide analogue or equivalent of the invention comprises a substitution of one of the non-bridging oxygens in the phosphodiester linkage. This modification slightly destabilizes base-pairing but adds significant resistance to nuclease degradation. A preferred nucleotide analogue or equivalent comprises phosphorothioate, chiral phosphorothioate, phosphorodithioate, phosphotriester, aminoalkylphosphotriester, H-phosphonate, methyl and other alkyl phosphonate including 3′-alkylene phosphonate, 5′-alkylene phosphonate and chiral phosphonate, phosphinate, phosphoramidate including 3′-amino phosphoramidate and aminoalkylphosphoramidate, thionophosphoramidate, thionoalkylphosphonate, thionoalkylphosphotriester, selenophosphate or boranophosphate.
  • A further preferred nucleotide analogue or equivalent of the invention comprises one or more sugar moieties that are mono- or disubstituted at the 2′, 3′ and/or 5′ position such as a —OH; —F; substituted or unsubstituted, linear or branched lower (C1-C10) alkyl, alkenyl, alkynyl, alkaryl, allyl, aryl, or aralkyl, that may be interrupted by one or more heteroatoms; O-, S-, or N-alkyl; O-, S-, or N-alkenyl; O-, S- or N-alkynyl; O-, S-, or N-allyl; O-alkyl-O-alkyl, -methoxy, -aminopropoxy; -aminoxy; methoxyethoxy; -dimethylaminooxyethoxy; and -dimethylaminoethoxyethoxy. The sugar moiety can be a pyranose or derivative thereof, or a deoxypyranose or derivative thereof, preferably a ribose or a derivative thereof, or a deoxyribose or a derivative thereof. Such preferred derivatized sugar moieties comprise Locked Nucleic Acid (LNA), in which the 2′-carbon atom is linked to the 3′ or 4′ carbon atom of the sugar ring thereby forming a bicyclic sugar moiety. A preferred LNA comprises 2′-O,4′-C-ethylene-bridged nucleic acid (Morita et al. 2001. Nucleic Acid Res Supplement No. 1: 241-242). These substitutions render the nucleotide analogue or equivalent RNase H and nuclease resistant and increase the affinity for the target RNA.
  • It is understood by a skilled person that it is not necessary for all positions in an antisense oligonucleotide to be modified uniformly. In addition, more than one of the aforementioned analogues or equivalents may be incorporated in a single antisense oligonucleotide or even at a single position within an antisense oligonucleotide. In certain embodiments, an antisense oligonucleotide of the invention has at least two different types of analogues or equivalents.
  • A preferred antisense oligonucleotide according to the invention comprises a 2′-O alkyl phosphorothioate antisense oligonucleotide, such as 2′-O-methyl modified ribose (RNA), 2′-O-ethyl modified ribose, 2′-O-propyl modified ribose, and/or substituted derivatives of these modifications such as halogenated derivatives.
  • A most preferred antisense oligonucleotide according to the invention comprises of 2′-O-methyl phosphorothioate ribose.
  • A functional equivalent of a molecule of the invention may be defined as an oligonucleotide as defined herein wherein an activity of said functional equivalent is retained to at least some extent. Preferably, an activity of said functional equivalent is inducing exon 43, 46, 50, 51, 52, or 53 skipping and providing a functional dystrophin protein. Said activity of said functional equivalent is therefore preferably assessed by detection of exon 43, 46, 50, 51, 52, or 53 skipping and by quantifying the amount of functional dystrophin protein. A functional dystrophin is herein preferably defined as being a dystrophin able to bind actin and members of the DGC protein complex. The assessment of said activity of an oligonucleotide is preferably done by RT-PCR or by immunofluorescence or Western blot analyses. Said activity is preferably retained to at least some extent when it represents at least 50%, or at least 60%, or at least 70% or at least 80% or at least 90% or at least 95% or more of corresponding activity of said oligonucleotide the functional equivalent derives from. Throughout this application, when the word oligonucleotide is used it may be replaced by a functional equivalent thereof as defined herein.
  • It will be understood by a skilled person that distinct antisense oligonucleotides can be combined for efficiently skipping any of exon 43, exon 46, exon 50, exon 51, exon 52 and/or exon 53 of the human DMD pre-mRNA. It is encompassed by the present invention to use one, two, three, four, five or more oligonucleotides for skipping one of said exons (i.e. exon, 43, 46, 50, 51, 52, or 53). It is also encompassed to use at least two oligonucleotides for skipping at least two, of said exons. Preferably two of said exons are skipped. More preferably, these two exons are:
      • 43 and 51, or
      • 43 and 53, or
      • 50 and 51, or
      • 51 and 52, or
      • 52 and 53.
  • The skilled person will know which combination of exons is preferred to be skipped depending on the type, the number and the location of the mutation present in a DMD or BMD patient.
  • An antisense oligonucleotide can be linked to a moiety that enhances uptake of the antisense oligonucleotide in cells, preferably muscle cells. Examples of such moieties are cholesterols, carbohydrates, vitamins, biotin, lipids, phospholipids, cell-penetrating peptides including but not limited to antennapedia, TAT, transportan and positively charged amino acids such as oligoarginine, poly-arginine, oligolysine or polylysine, antigen-binding domains such as provided by an antibody, a Fab fragment of an antibody, or a single chain antigen binding domain such as a cameloid single domain antigen-binding domain.
  • A preferred antisense oligonucleotide comprises a peptide-linked PMO.
  • A preferred antisense oligonucleotide comprising one or more nucleotide analogs or equivalents of the invention modulates splicing in one or more muscle cells, including heart muscle cells, upon systemic delivery. In this respect, systemic delivery of an antisense oligonucleotide comprising a specific nucleotide analog or equivalent might result in targeting a subset of muscle cells, while an antisense oligonucleotide comprising a distinct nucleotide analog or equivalent might result in targeting of a different subset of muscle cells. Therefore, in one embodiment it is preferred to use a combination of antisense oligonucleotides comprising different nucleotide analogs or equivalents for inducing skipping of exon 43, 46, 50, 51, 52, or 53 of the human DMD pre-mRNA.
  • A cell can be provided with a molecule capable of interfering with essential sequences that result in highly efficient skipping of exon 43, exon 46, exon 50, exon 51, exon 52 or exon 53 of the human DMD pre-mRNA by plasmid-derived antisense oligonucleotide expression or viral expression provided by adenovirus- or adeno-associated virus-based vectors. In a preferred embodiment, there is provided a viral-based expression vector comprising an expression cassette that drives expression of a molecule as identified herein. Expression is preferably driven by a polymerase III promoter, such as a U1, a U6, or a U7 RNA promoter. A muscle or myogenic cell can be provided with a plasmid for antisense oligonucleotide expression by providing the plasmid in an aqueous solution. Alternatively, a plasmid can be provided by transfection using known transfection agentia such as, for example, LipofectAMINE™ 2000 (Invitrogen) or polyethyleneimine (PEI; ExGen500 (MBI Fermentas)), or derivatives thereof.
  • One preferred antisense oligonucleotide expression system is an adenovirus associated virus (AAV)-based vector. Single chain and double chain AAV-based vectors have been developed that can be used for prolonged expression of small antisense nucleotide sequences for highly efficient skipping of exon 43, 46, 50, 51, 52 or 53 of the DMD pre-mRNA.
  • A preferred AAV-based vector comprises an expression cassette that is driven by a polymerase III-promoter (Pol III). A preferred Pol III promoter is, for example, a U1, a U6, or a U7 RNA promoter.
  • The invention therefore also provides a viral-based vector, comprising a Pol III-promoter driven expression cassette for expression of one or more antisense sequences of the invention for inducing skipping of exon 43, exon 46, exon 50, exon 51, exon 52 or exon 53 of the human DMD pre-mRNA.
  • Pharmaceutical Composition
  • If required, a molecule or a vector expressing an antisense oligonucleotide of the invention can be incorporated into a pharmaceutically active mixture or composition by adding a pharmaceutically acceptable carrier.
  • Therefore, in a further aspect, the invention provides a composition, preferably a pharmaceutical composition comprising a molecule comprising an antisense oligonucleotide according to the invention, and/or a viral-based vector expressing the antisense sequence(s) according to the invention and a pharmaceutically acceptable carrier.
  • A preferred pharmaceutical composition comprises a molecule as defined herein and/or a vector as defined herein, and a pharmaceutical acceptable carrier or excipient, optionally combined with a molecule and/or a vector as defined herein which is able to induce skipping of exon 6, 7, 11, 17, 19, 21, 43, 44, 45, 50, 51, 52, 53, 55, 57, 59, 62, 63, 65, 66, 69, or 75 of the DMD pre-mRNA. Preferred molecules able to induce skipping of any of these exon are identified in any one of Tables 1 to 7.
  • Preferred excipients include excipients capable of forming complexes, vesicles and/or liposomes that deliver such a molecule as defined herein, preferably an oligonucleotide complexed or trapped in a vesicle or liposome through a cell membrane. Many of these excipients are known in the art. Suitable excipients comprise polyethylenimine and derivatives, or similar cationic polymers, including polypropyleneimine or polyethylenimine copolymers (PECs) and derivatives, ExGen 500, synthetic amphiphils (SAINT-18), lipofectin™, DOTAP and/or viral capsid proteins that are capable of self assembly into particles that can deliver such molecule, preferably an oligonucleotide as defined herein to a cell, preferably a muscle cell. Such excipients have been shown to efficiently deliver (oligonucleotide such as antisense) nucleic acids to a wide variety of cultured cells, including muscle cells. Their high transfection potential is combined with an excepted low to moderate toxicity in terms of overall cell survival. The ease of structural modification can be used to allow further modifications and the analysis of their further (in vivo) nucleic acid transfer characteristics and toxicity.
  • Lipofectin represents an example of a liposomal transfection agent. It consists of two lipid components, a cationic lipid N-[1-(2,3 dioleoyloxy)propyl]-N,N,N-trimethylammonium chloride (DOTMA) (cp. DOTAP which is the methylsulfate salt) and a neutral lipid dioleoylphosphatidylethanolamine (DOPE). The neutral component mediates the intracellular release. Another group of delivery systems are polymeric nanoparticles.
  • Polycations such like diethylaminoethylaminoethyl (DEAE)-dextran, which are well known as DNA transfection reagent can be combined with butylcyanoacrylate (PBCA) and hexylcyanoacrylate (PHCA) to formulate cationic nanoparticles that can deliver a molecule or a compound as defined herein, preferably an oligonucleotide across cell membranes into cells.
  • In addition to these common nanoparticle materials, the cationic peptide protamine offers an alternative approach to formulate a compound as defined herein, preferably an oligonucleotide as colloids. This colloidal nanoparticle system can form so called proticles, which can be prepared by a simple self-assembly process to package and mediate intracellular release of a compound as defined herein, preferably an oligonucleotide. The skilled person may select and adapt any of the above or other commercially available alternative excipients and delivery systems to package and deliver a compound as defined herein, preferably an oligonucleotide for use in the current invention to deliver said compound for the treatment of Duchenne Muscular Dystrophy or Becker Muscular Dystrophy in humans.
  • In addition, a compound as defined herein, preferably an oligonucleotide could be covalently or non-covalently linked to a targeting ligand specifically designed to facilitate the uptake in to the cell, cytoplasm and/or its nucleus. Such ligand could comprise (i) a compound (including but not limited to peptide(-like) structures) recognising cell, tissue or organ specific elements facilitating cellular uptake and/or (ii) a chemical compound able to facilitate the uptake in to cells and/or the intracellular release of an a compound as defined herein, preferably an oligonucleotide from vesicles, e.g. endosomes or lysosomes.
  • Therefore, in a preferred embodiment, a compound as defined herein, preferably an oligonucleotide are formulated in a medicament which is provided with at least an excipient and/or a targeting ligand for delivery and/or a delivery device of said compound to a cell and/or enhancing its intracellular delivery. Accordingly, the invention also encompasses a pharmaceutically acceptable composition comprising a compound as defined herein, preferably an oligonucleotide and further comprising at least one excipient and/or a targeting ligand for delivery and/or a delivery device of said compound to a cell and/or enhancing its intracellular delivery.
  • It is to be understood that a molecule or compound or oligonucleotide may not be formulated in one single composition or preparation. Depending on their identity, the skilled person will know which type of formulation is the most appropriate for each compound.
  • In a preferred embodiment, an in vitro concentration of a molecule or an oligonucleotide as defined herein, which is ranged between 0.1 nM and 1 RM is used.
  • More preferably, the concentration used is ranged between 0.3 to 400 nM, even more preferably between 1 to 200 nM. A molecule or an oligonucleotide as defined herein may be used at a dose which is ranged between 0.1 and 20 mg/kg, preferably 0.5 and 10 mg/kg. If several molecules or oligonucleotides are used, these concentrations may refer to the total concentration of oligonucleotides or the concentration of each oligonucleotide added. The ranges of concentration of oligonucleotide(s) as given above are preferred concentrations for in vitro or ex vivo uses. The skilled person will understand that depending on the oligonucleotide(s) used, the target cell to be treated, the gene target and its expression levels, the medium used and the transfection and incubation conditions, the concentration of oligonucleotide(s) used may further vary and may need to be optimised any further.
  • More preferably, a compound preferably an oligonucleotide to be used in the invention to prevent, treat DMD or BMD are synthetically produced and administered directly to a cell, a tissue, an organ and/or patients in formulated form in a pharmaceutically acceptable composition or preparation. The delivery of a pharmaceutical composition to the subject is preferably carried out by one or more parenteral injections, e.g. intravenous and/or subcutaneous and/or intramuscular and/or intrathecal and/or intraventricular administrations, preferably injections, at one or at multiple sites in the human body.
  • A preferred oligonucleotide as defined herein optionally comprising one or more nucleotide analogs or equivalents of the invention modulates splicing in one or more muscle cells, including heart muscle cells, upon systemic delivery. In this respect, systemic delivery of an oligonucleotide comprising a specific nucleotide analog or equivalent might result in targeting a subset of muscle cells, while an oligonucleotide comprising a distinct nucleotide analog or equivalent might result in targeting of a different subset of muscle cells.
  • In this respect, systemic delivery of an oligonucleotide comprising a specific nucleotide analog or equivalent might result in targeting a subset of muscle cells, while an oligonucleotide comprising a distinct nucleotide analog or equivalent might result in targeting a different subset of muscle cells. Therefore, in this embodiment, it is preferred to use a combination of oligonucleotides comprising different nucleotide analogs or equivalents for modulating splicing of the DMD mRNA in at least one type of muscle cells.
  • In a preferred embodiment, there is provided a molecule or a viral-based vector for use as a medicament, preferably for modulating splicing of the DMD pre-mRNA, more preferably for promoting or inducing skipping of any of exon 43, 46, 50-53 as identified herein.
  • Use
  • In yet a further aspect, the invention provides the use of an antisense oligonucleotide or molecule according to the invention, and/or a viral-based vector that expresses one or more antisense sequences according to the invention and/or a pharmaceutical composition, for modulating splicing of the DMD pre-mRNA. The splicing is preferably modulated in a human myogenic cell or muscle cell in vitro. More preferred is that splicing is modulated in a human muscle cell in vivo. Accordingly, the invention further relates to the use of the molecule as defined herein and/or the vector as defined herein and/or or the pharmaceutical composition as defined herein for modulating splicing of the DMD pre-mRNA or for the preparation of a medicament for the treatment of a DMD or BMD patient.
  • In this document and in its claims, the verb “to comprise” and its conjugations is used in its non-limiting sense to mean that items following the word are included, but items not specifically mentioned are not excluded. In addition the verb “to consist” may be replaced by “to consist essentially of” meaning that a molecule or a viral-based vector or a composition as defined herein may comprise additional component(s) than the ones specifically identified, said additional component(s) not altering the unique characteristic of the invention. In addition, reference to an element by the indefinite article “a” or “an” does not exclude the possibility that more than one of the element is present, unless the context clearly requires that there be one and only one of the elements. The indefinite article “a” or “an” thus usually means “at least one”.
  • Each embodiment as identified herein may be combined together unless otherwise indicated. All patent and literature references cited in the present specification are hereby incorporated by reference in their entirety.
  • The following examples are offered for illustrative purposes only, and are not intended to limit the scope of the present invention in any way.
  • EXAMPLES Examples 1-4 Materials and Methods
  • AON design was based on (partly) overlapping open secondary structures of the target exon RNA as predicted by the m-fold program, on (partly) overlapping putative SR-protein binding sites as predicted by the ESE-finder software. AONs were synthesized by Prosensa Therapeutics B.V. (Leiden, Netherlands), and contain 2′-O-methyl RNA and full-length phosphorothioate (PS) backbones.
  • Tissue Culturing, Transfection and RT-PCR Analysis
  • Myotube cultures derived from a healthy individual (“human control”) (examples 1, 3, and 4; exon 43, 50, 52 skipping) or a DMD patient carrying an exon 45 deletion (example 2; exon 46 skipping) were processed as described previously (Aartsma-Rus et al., Neuromuscul. Disord. 2002; 12: S71-77 and Hum Mol Genet 2003; 12(8): 907-14).
  • For the screening of AONs, myotube cultures were transfected with 50 nM and 150 nM (example 2), 200nM and 500 nM (example 4) or 500nM only (examples 1 and 3) of each AON. Transfection reagent UNIFectylin (Prosensa Therapeutics BV, Netherlands) was used, with 2 μl UNIFectylin per μg AON. Exon skipping efficiencies were determined by nested RT-PCR analysis using primers in the exons flanking the targeted exons (43, 46, 50, 51, 52, or 53). PCR fragments were isolated from agarose gels for sequence verification. For quantification, the PCR products were analyzed using the DNA 1000 LabChips Kit on the Agilent 2100 bioanalyzer (Agilent Technologies, USA).
  • Results
  • DMD exon 43 skipping.
  • A series of AONs targeting sequences within exon 43 were designed and transfected in healthy control myotube cultures. Subsequent RT-PCR and sequence analysis of isolated RNA demonstrated that almost all AONs targeting a continuous nucleotide stretch within exon 43 herein defined as SEQ ID NO 2, was indeed capable of inducing exon 43 skipping. PS237 (SEQ ID NO: 65) reproducibly induced highest levels of exon 43 skipping (up to 66%) at 500 nM, as shown in FIG. 1. For comparison, also PS238 and PS240 are shown, inducing exon 43 skipping levels up to 13% and 36% respectively (FIG. 1). The precise skipping of exon 43 was confirmed by sequence analysis of the novel smaller transcript fragments. No exon 43 skipping was observed in non-treated cells (NT).
  • DMD exon 46 skipping.
  • A series of AONs targeting sequences within exon 46 were designed and transfected in myotube cultures derived from a DMD patient carrying an exon 45 deletion in the DMD gene. For patients with such mutation antisense-induced exon 46 skipping would induce the synthesis of a novel, BMD-like dystrophin protein that may indeed alleviate one or more symptoms of the disease. Subsequent RT-PCR and sequence analysis of isolated RNA demonstrated that almost all AONs targeting a continuous nucleotide stretch within exon 46 herein defined as SEQ ID NO 3, was indeed capable of inducing exon 46 skipping, even at relatively low AON concentrations of 50 nM. PS182 (SEQ ID NO: 117) reproducibly induced highest levels of exon 46 skipping (up to 50% at 50 nM and 74% at 150 nM), as shown in FIG. 2. For comparison, also PS177, PS179, and PS181 are shown, inducing exon 46 skipping levels up to 55%, 58% and 42% respectively at 150 nM (FIG. 2). The precise skipping of exon 46 was confirmed by sequence analysis of the novel smaller transcript fragments. No exon 46 skipping was observed in non-treated cells (NT).
  • DMD exon 50 skipping.
  • A series of AONs targeting sequences within exon 50 were designed and transfected in healthy control myotube cultures. Subsequent RT-PCR and sequence analysis of isolated RNA demonstrated that almost all AONs targeting a continuous nucleotide stretch within exon 50 herein defined as SEQ ID NO 4, was indeed capable of inducing exon 50 skipping. PS248 (SEQ ID NO: 127) reproducibly induced highest levels of exon 50 skipping (up to 35% at 500 nM), as shown in FIG. 3. For comparison, also PS245, PS246, and PS247 are shown, inducing exon 50 skipping levels up to 14-16% at 500 nM (FIG. 3). The precise skipping of exon 50 was confirmed by sequence analysis of the novel smaller transcript fragments. No exon 50 skipping was observed in non-treated cells (NT).
  • DMD exon 51 skipping.
  • A series of AONs targeting sequences within exon 51 were designed and transfected in healthy control myotube cultures. Subsequent RT-PCR and sequence analysis of isolated RNA demonstrated that almost all AONs targeting a continuous nucleotide stretch within exon 51 herein defined as SEQ ID NO 5, was indeed capable of inducing exon 51 skipping. The AON with SEQ ID NO 180 reproducibly induced highest levels of exon 51 skipping (not shown).
  • DMD exon 52 skipping.
  • A series of AONs targeting sequences within exon 52 were designed and transfected in healthy control myotube cultures. Subsequent RT-PCR and sequence analysis of isolated RNA demonstrated that almost all AONs targeting a continuous nucleotide stretch within exon 52 herein defined as SEQ ID NO 6, was indeed capable of inducing exon 52 skipping. PS236 (SEQ ID NO: 299) reproducibly induced highest levels of exon 52 skipping (up to 88% at 200 nM and 91% at 500 nM), as shown in FIG. 4. For comparison, also PS232 and AON 52-1 (previously published by Aartsma-Rus et al. Oligonucleotides 2005) are shown, inducing exon 52 skipping at levels up to 59% and 10% respectively when applied at 500 nM (FIG. 4). The precise skipping of exon 52 was confirmed by sequence analysis of the novel smaller transcript fragments. No exon 52 skipping was observed in non-treated cells (NT).
  • DMD exon 53 skipping.
  • A series of AONs targeting sequences within exon 53 were designed and transfected in healthy control myotube cultures. Subsequent RT-PCR and sequence analysis of isolated RNA demonstrated that almost all AONs targeting a continuous nucleotide stretch within exon 53 herein defined as SEQ ID NO 7, was indeed capable of inducing exon 53 skipping. The AON with SEQ ID NO 328 reproducibly induced highest levels of exon 53 skipping (not shown).
  • Sequence listing
    DMD gene amino acid sequence
    SEQ ID NO 1:
    MLWWEEVEDCYEREDVQKKTFTKWVNAQFSKFGKQHIENLFSDLQDGRRLLDLLEGLTGQKL
    PKEKGSTRVHALNNVNKALRVLQNNNVDLVNIGSTDIVDGNHKLTLGLIWNIILHWQVKNVMK
    NIMAGLQQTNSEKILLSWVRQSTRNYPQVNVINFTTSWSDGLALNALIHSHRPDLFDWNSVVCQ
    QSATQRLEHAFNIARYQLGIEKLLDPEDVDTTYPDKKSILMYITSLFQVLPQQVSIEAIQEVEMLP
    RPPKVTKEEHFQLHHQMHYSQQITVSLAQGYERTSSPKPRFKSYAYTQAAYVTTSDPTRSPFPSQ
    HLEAPEDKSFGSSLMESEVNLDRYQTALEEVLSWLLSAEDTLQAQGEISNDVEVVKDQFHTHEG
    YMMDLTAHQGRVGNILQLGSKLIGTGKLSEDEETEVQEQMNLLNSRWECLRVASMEKQSNLH
    RVLMDLQNQKLKELNDWLTKTEERTRKMEEEPLGPDLEDLKRQVQQHKVLQEDLEQEQVRVN
    SLTHMVVVVDESSGDHATAALEEQLKVLGDRWANICRWTEDRWVLLQDILLKWQRLTEEQL
    FSAWLSEKEDAVNKIHTTGFKDQNEMLSSLQKLAVLKADLEKKKQSMGKLYSLKQDLLSTLKN
    KSVTQKTEAWLDNFARCWDNLVQKLEKSTAQISQAVTTTQPSLTQTTVMETVTTVTTREQILV
    KHAQEELPPPPPQKKRQITVDSEIRKRLDVDITELHSWITRSEAVLQSPEFAIFRKEGNFSDLKEK
    VNAIEREKAEKFRKLQDASRSAQALVEQMVNEGVNADSIKQASEQLNSRWIEFCQLLSERLNW
    LEYQNNIIAFYNQLQQLEQMTTTAENWLKIQPTTPSEPTAIKSQLKICKDEVNRLSGLQPQIERLK
    IQSIALKEKGQGPMFLDADFVAFTNHFKQVFSDVQAREKELQTIFDTLPPMRYQETMSAIRTWV
    QQSETKLSIPQLSVTDYEIMEQRLGELQALQSSLQEQQSGLYYLSTTVKEMSKKAPSEISRKYQS
    EFEEIEGRWKKLSSQLVEHCQKLEEQMNKLRKIQNHIQTLKKWMAEVDVFLKEEWPALGDSEI
    LKKQLKQCRLLVSDIQTIQPSLNSVNEGGQKIKNEAEPEFASRLETELKELNTQWDHMCQQVYA
    RKEALKGGLEKTVSLQKDLSEMHEWMTQAEEEYLERDFEYKTPDELQKAVEEMKRAKEEAQQ
    KEAKVKLLTESVNSVIAQAPPVAQEALKKELETLTTNYQWLCTRLNGKCKTLEEVWACWHELL
    SYLEKANKWLNEVEFKLKTTENIPGGAEEISEVLDSLENLMRHSEDNPNQIRILAQTLTDGGVM
    DELINEELETFNSRWRELHEEAVRRQKLLEQSIQSAQETEKSLHLIQESLTFIDKQLAAYIADKVD
    AAQMPQEAQKIQSDLTSHEISLEEMKKHNQGKEAAQRVLSQIDVAQKKLQDVSMKFRLFQKPA
    NFEQRLQESKMILDEVKMHLPALETKSVEQEVVQSQLNHCVNLYKSLSEVKSEVEMVIKTGRQI
    VQKKQTENPKELDERVTALKLHYNELGAKVTERKQQLEKCLKLSRKMRKEMNVLTEWLAAT
    DMELTKRSAVEGMPSNLDSEVAWGKATQKEIEKQKVHLKSITEVGEALKTVLGKKETLVEDKL
    SLLNSNWIAVTSRAEEWLNLLLEYQKHMETFDQNVDHITKWITQADTLLDESEKKKPQQKEDVL
    KRLKAELNDIRPKVDSTRDQAANLMANRGDHCRKLVEPQISELNHRFAAISHRIKTGKASIPLKE
    LEQFNSDIQKLLEPLEAEIQQGVNLKEEDFNKDMNEDNEGTVKELLQRGDNLQQRITDERKREEI
    KIKQQLLQTKHNALKDLRSQRRKKALEISHQWYQYKRQADDLLKCLDDIEKKLASLPEPRDER
    KIKEIDRELQKKKEELNAVRRQAEGLSEDGAAMAVEPTQIQLSKRWREIESKFAQFRRLNFAQIH
    TVREETMMVMTEDMPLEISYVPSTYLTEITHVSQALLEVEQLLNAPDLCAKDFEDLFKQEESLK
    NIKDSLQQSSGRIDIIHSKKTAALQSATPVERVKLQEALSQLDFQWEKVNKMYKDRQGRFDRSV
    EKWRRFHYDIKIFNQWLTEAEQFLRKTQIPENWEHAKYKWYLKELQDGIGQRQTVVRTLNATG
    EEIIQQSSKTDASILQEKLGSLNLRWQEVCKQLSDRKKRLEEQKNILSEFQRDLNEFVLWLEEAD
    NIASIPLEPGKEQQLKEKLEQVKLLVEELPLRQGILKQLNETGGPVLVSAPISPEEQDKLENKLKQ
    TNLQWIKVSRALPEKQGEIEAQIKDLGQLEKKLEDLEEQLNHLLLWLSPIRNQLEIYNQPNQEGP
    FDVQETEIAVQAKQPDVEEILSKGQHLYKEKPATQPVKRKLEDLSSEWKAVNRLLQELRAKQP
    DLAPGLTTIGASPTQTVTLVTQPVVTKETAISKLEMPSSLMLEVPALADFNRAWTELTDWLSLL
    DQVIKSQRVMVGDLEDINEMIIKQKATMQDLEQRRPQLEELITAAQNLKNKTSNQEARTIITDRI
    ERIQNQWDEVQEHLQNRRQQLNEMLKDSTQWLEAKEEAEQVLGQARAKLESWKEGPYTVDAI
    QKKIIETKQLAKDLRQWQTNVDVANDLALKLLRDYSADDTRKVHMITENINASWRSIHKRVSE
    REAALEETHRLLQQFPLDLEKFLAWLTEAETTANVLQDATRKERLLEDSKGVKELMKQWQDL
    QGEIEAHTDVYHNLDENSQKILRSLEGSDDAVLLQRRLDNMNFKWSELRKKSLNIRSHLEASSD
    QWKRLHLSLQELLVWLQLKDDELSRQAPIGGDFPAVQKQNDVHRAFKRELKTKEPVIMSTLET
    VRIFLIEQPLEGLEKLYQEPRELPPEERAQNVIRLLRKQAEEVNTEWEKLNLHSADWQRKIDET
    LERLQELQEATDELDLKLRQAEVIKGSWQPVGDLLIDSLQDHLEKVKALRGEIAPLKENVSHVN
    DLARQLTTLGIQLSPYNLSTLEDLNTRWKLLQVAVEDRVRQLHEAHRDFGPASQHFLSTSVQGP
    WERAISPNKVPYYINHETQTTCWDHPKMTELYQSLADLNNVRFSAYRTAMKLRRLQKALCLDL
    LSLSAACDALDQHNLKQNDQPMDILQIINCLTTIYDRLEQEHNNLVNVPLCVDMCLNWLLNVY
    DTGRTGRIRVLSFKTGIISLCKAHLEDKYRYLFKQVASSTGFCDQRRLGLLLHDSIQIPRQLGEVA
    SFGGSNIEPSVRSCFQFANNKPEIEAALFLDWMRLEPQSMVWLPVLHRVAAAETAKHQAKCNIC
    KECPIIGFRYRSLKHFNYDICQSCFFSGRVAKGHKMHYPMVEYCTPTTSGEDVRDFAKVLKNKF
    RTKRYFAKHPRMGYLPVQTVLEGDNMETPVTLINFWPVDSAPASSPQLSHDDTHSRIEHYASRL
    AEMENSNGSYLNDSISPNESIDDEHLLIQHYCQSLNQDSPLSQPRSPAQILISLESEERGELERILAD
    LEEENRNLQAEYDRLKQQHEHKGLSPLPSPPEMMPTSPQSPRDAELIAEAKLLRQHKGRLEARM
    QILEDHNKQLESQLHRLRQLLEQPQAEAKVNGTTVSSPSTSLQRSDSSQPMLLRVVGSQTSDSM
    GEEDLLSPPQDTSTGLEEVMEQLNNSFPSSRGRNTPGKPMREDTM
    SEQ ID NO 2 (exon 43):
    AGAUAGUCUACAACAAAGCUCAGGUCGGAUUGACAUUAUUCAUAGCAAGAAGACAGCAG
    CAUUGCAAAGUGCAACGCCUGUGG
    SEQ ID NO 3 (exon 46):
    UUAUGGUUGGAGGAAGCAGAUAACAUUGCUAGUAUCCCACUUGAACCUGGAAAAGAGCA
    GCAACUAAAAGAAAAGC
    SEQ ID NO 4 (exon 50):
    ′GGCGGTAAACCGUUUACUUCAAGAGCUGAGGGCAAAGCAGCCUG ACCUAGC
    UCCUGGACUGACCACUAUUGG
    SEQ ID NO 5 (exon 51):
    CUCCUACUCAGACUGUUACUCUGGUGACACAACCUGUGGUUACUAAGGAAACUGCCAUC
    UCCAAACUAGAAAUGCCAUCUUCCUUGAUGUUGGAGGUAC
    SEQ ID NO 6 (exon 52):
    AUGCAGGAUUUGGAACAGAGGCGUCCCCAGUUGGAAGAACUCAUUACCGCUGCCCAAAA
    UUUGAAAAA CAAGACCAGCAAUCAAGAGGCU
    SEQ ID NO 7 (exon 53):
    AAAUGUUAAAGGAUUCAACACAAUGGCUGGAAGCUAAGGAAGAAGCUGAGCAGGUCUUA
    GGACAGGCCAGAG
  • TABLE 1
    oligonucleotides for skipping DMD Gene Exon 43
    SEQ ID CCACAGGCGUUGCACUUUGCAAUGC
    NO 8
    SEQ ID CACAGGCGUUGCACUUUGCAAUGCU
    NO 9
    SEQ ID ACAGGCGUUGCACUUUGCAAUGCUG
    NO 10
    SEQ ID CAGGCGUUGCACUUUGCAAUGCUGC
    NO 11
    SEQ ID AGGCGUUGCACUUUGCAAUGCUGCU
    NO 12
    SEQ ID GGCGUUGCACUUUGCAAUGCUGCUG
    NO 13
    SEQ ID GCGUUGCACUUUGCAAUGCUGCUGU
    NO 14
    SEQ ID CGUUGCACUUUGCAAUGCUGCUGUC
    NO 15
    SEQ ID CGUUGCACUUUGCAAUGCUGCUG
    NO 16
    PS240
    SEQ ID GUUGCACUUUGCAAUGCUGCUGUCU
    NO 17
    SEQ ID UUGCACUUUGCAAUGCUGCUGUCUU
    NO 18
    SEQ ID UGCACUUUGCAAUGCUGCUGUCUUC
    NO 19
    SEQ ID GCACUUUGCAAUGCUGCUGUCUUCU
    NO 20
    SEQ ID CACUUUGCAAUGCUGCUGUCUUCUU
    NO 21
    SEQ ID ACUUUGCAAUGCUGCUGUCUUCUUG
    NO 22
    SEQ ID CUUUGCAAUGCUGCUGUCUUCUUGC
    NO 23
    SEQ ID UUUGCAAUGCUGCUGUCUUCUUGCU
    NO 24
    SEQ ID UUGCAAUGCUGCUGUCUUCUUGCUA
    NO 25
    SEQ ID UGCAAUGCUGCUGUCUUCUUGCUAU
    NO 26
    SEQ ID GCAAUGCUGCUGUCUUCUUGCUAUG
    NO 27
    SEQ ID CAAUGCUGCUGUCUUCUUGCUAUGA
    NO 28
    SEQ ID AAUGCUGCUGUCUUCUUGCUAUGAA
    NO 29
    SEQ ID AUGCUGCUGUCUUCUUGCUAUGAAU
    NO 30
    SEQ ID UGCUGCUGUCUUCUUGCUAUGAAUA
    NO 31
    SEQ ID GCUGCUGUCUUCUUGCUAUGAAUAA
    NO 32
    SEQ ID CUGCUGUCUUCUUGCUAUGAAUAAU
    NO 33
    SEQ ID UGCUGUCUUCUUGCUAUGAAUAAU
    NO 34 G
    SEQ ID GCUGUCUUCUUGCUAUGAAUAAUG
    NO 35 U
    SEQ ID CUGUCUUCUUGCUAUGAAUAAUGUC
    NO 36
    SEQ ID UGUCUUCUUGCUAUGAAUAAUGUC
    NO 37 A
    SEQ ID GUCUUCUUGCUAUGAAUAAUGUCA
    NO 38 A
    SEQ ID NO 39 UCUUCUUGCUAUGAAUAAUGUCAAU
    SEQ ID NO 40 CUUCUUGCUAUGAAUAAUGUCAAUC
    SEQ ID NO 41 UUCUUGCUAUGAAUAAUGUCAAUCC
    SEQ ID NO 42 UCUUGCUAUGAAUAAUGUCAAUCCG
    SEQ ID NO 43 CUUGCUAUGAAUAAUGUCAAUCCGA
    SEQ ID NO 44 UUGCUAUGAAUAAUGUCAAUCCGAC
    SEQ ID NO 45 UGCUAUGAAUAAUGUCAAUCCGACC
    SEQ ID NO 46 GCUAUGAAUAAUGUCAAUCCGACCU
    SEQ ID NO 47 CUAUGAAUAAUGUCAAUCCGACCUG
    SEQ ID NO 48 UAUGAAUAAUGUCAAUCCGACCUGA
    SEQ ID NO 49 AUGAAUAAUGUCAAUCCGACCUGAG
    SEQ ID NO 50 UGAAUAAUGUCAAUCCGACCUGAGC
    SEQ ID NO 51 GAAUAAUGUCAAUCCGACCUGAGCU
    SEQ ID NO 52 AAUAAUGUCAAUCCGACCUGAGCUU
    SEQ ID NO 53 AUAAUGUCAAUCCGACCUGAGCUUU
    SEQ ID NO 54 UAAUGUCAAUCCGACCUGAGCUUUG
    SEQ ID NO 55 AAUGUCAAUCCGACCUGAGCUUUGU
    SEQ ID NO 56 AUGUCAAUCCGACCUGAGCUUUGUU
    SEQ ID NO 57 UGUCAAUCCGACCUGAGCUUUGUUG
    SEQ ID NO 58 GUCAAUCCGACCUGAGCUUUGUUGU
    SEQ ID NO 59 UCAAUCCGACCUGAGCUUUGUUGUA
    SEQ ID NO 60 CAAUCCGACCUGAGCUUUGUUGUAG
    SEQ ID NO 61 AAUCCGACCUGAGCUUUGUUGUAGA
    SEQ ID NO 62 AUCCGACCUGAGCUUUGUUGUAGAC
    SEQ ID NO 63 UCCGACCUGAGCUUUGUUGUAGACU
    SEQ ID NO 64 CCGACCUGAGCUUUGUUGUAGACUA
    SEQ ID NO 65 CGACCUGAGCUUUGUUGUAG
    PS237
    SEQ ID NO 66 CGACCUGAGCUUUGUUGUAGACUAU
    PS238
    SEQ ID NO 67 GACCUGAGCUUUGUUGUAGACUAUC
    SEQ ID NO 68 ACCUGAGCUUUGUUGUAGACUAUCA
    SEQ ID NO 69 CCUGA GCUUU GUUGU AGACU AUC
  • TABLE 2
    oligonucleotides for skipping DMD Gene Exon 46
    SEQ ID GCUUUUCUUUUAGUUGCUGCUCUUU
    NO 70
    PS179
    SEQ ID CUUUUCUUUUAGUUGCUGCUCUUUU
    NO 71
    SEQ ID UUUUCUUUUAGUUGCUGCUCUUUUC
    NO 72
    SEQ ID UUUCUUUUAGUUGCUGCUCUUUUCC
    NO 73
    SEQ ID UUCUUUUAGUUGCUGCUCUUUUCCA
    NO 74
    SEQ ID UCUUUUAGUUGCUGCUCUUUUCCAG
    NO 75
    SEQ ID CUUUUAGUUGCUGCUCUUUUCCAGG
    NO 76
    SEQ ID UUUUAGUUGCUGCUCUUUUCCAGGU
    NO 77
    SEQ ID UUUAGUUGCUGCUCUUUUCCAGGUU
    NO 78
    SEQ ID UUAGUUGCUGCUCUUUUCCAGGUUC
    NO 79
    SEQ ID UAGUUGCUGCUCUUUUCCAGGUUCA
    NO 80
    SEQ ID AGUUGCUGCUCUUUUCCAGGUUCAA
    NO 81
    SEQ ID GUUGCUGCUCUUUUCCAGGUUCAAG
    NO 82
    SEQ ID UUGCUGCUCUUUUCCAGGUUCAAGU
    NO 83
    PS181
    SEQ ID UGCUGCUCUUUUCCAGGUUCAAGUG
    NO 84
    SEQ ID GCUGCUCUUUUCCAGGUUCAAGUGG
    NO 85
    SEQ ID CUGCUCUUUUCCAGGUUCAAGUGGG
    NO 86
    SEQ ID UGCUCUUUUCCAGGUUCAAGUGGGA
    NO 87
    SEQ ID GCUCUUUUCCAGGUUCAAGUGGGAC
    NO 88
    SEQ ID CUCUUUUCCAGGUUCAAGUGGGAUA
    NO 89
    SEQ ID UCUUUUCCAGGUUCAAGUGGGAUAC
    NO 90
    PS182
    SEQ ID UCUUUUCCAGGUUCAAGUGG
    NO 91
    PS177
    SEQ ID CUUUUCCAGGUUCAAGUGGGAUACU
    NO 92
    SEQ ID UUUUCCAGGUUCAAGUGGGAUACU
    NO 93 A
    SEQ ID UUUCCAGGUUCAAGUGGGAUACUA
    NO 94 G
    SEQ ID UUCCAGGUUCAAGUGGGAUACUAGC
    NO 95
    SEQ ID UCCAGGUUCAAGUGGGAUACUAGCA
    NO 96
    SEQ ID NO 97 CCAGGUUCAAGUGGGAUACUAGCAA
    SEQ ID NO 98 CAGGUUCAAGUGGGAUACUAGCAAU
    SEQ ID NO 99 AGGUUCAAGUGGGAUACUAGCAAUG
    SEQ ID NO GGUUCAAGUGGGAUACUAGCAAUGU
    100
    SEQ ID NO GUUCAAGUGGGAUACUAGCAAUGUU
    101
    SEQ ID NO UUCAAGUGGGAUACUAGCAAUGUUA
    102
    SEQ ID NO UCAAGUGGGAUACUAGCAAUGUUAU
    103
    SEQ ID NO CAAGUGGGAUACUAGCAAUGUUAUC
    104
    SEQ ID NO AAGUGGGAUACUAGCAAUGUUAUCU
    105
    SEQ ID NO AGUGGGAUACUAGCAAUGUUAUCUG
    106
    SEQ ID NO GUGGGAUACUAGCAAUGUUAUCUGC
    107
    SEQ ID NO UGGGAUACUAGCAAUGUUAUCUGCU
    108
    SEQ ID NO GGGAUACUAGCAAUGUUAUCUGCUU
    109
    SEQ ID NO GGAUACUAGCAAUGUUAUCUGCUUC
    110
    SEQ ID NO GAUACUAGCAAUGUUAUCUGCUUCC
    111
    SEQ ID NO AUACUAGCAAUGUUAUCUGCUUCCU
    112
    SEQ ID NO UACUAGCAAUGUUAUCUGCUUCCUC
    113
    SEQ ID NO ACUAGCAAUGUUAUCUGCUUCCUCC
    114
    SEQ ID NO CUAGCAAUGUUAUCUGCUUCCUCCA
    115
    SEQ ID NO UAGCAAUGUUAUCUGCUUCCUCCAA
    116
    SEQ ID NO AGCAAUGUUAUCUGCUUCCUCCAAC
    117
    SEQ ID NO GCAAUGUUAUCUGCUUCCUCCAACC
    118
    SEQ ID NO CAAUGUUAUCUGCUUCCUCCAACCA
    119
    SEQ ID NO AAUGUUAUCUGCUUCCUCCAACCAU
    120
    SEQ ID NO AUGUUAUCUGCUUCCUCCAACCAUA
    121
    SEQ ID NO UGUUAUCUGCUUCCUCCAACCAUAA
    122
  • TABLE 3
    oligonucleotides for skipping DMD Gene Exon 50
    SEQ ID CCAAUAGUGGUCAGUCCAGGAGCUA
    NO 123
    SEQ ID CAAUAGUGGUCAGUCCAGGAGCUAG
    NO 124
    SEQ ID AAUAGUGGUCAGUCCAGGAGCUAGG
    NO 125
    SEQ ID AUAGUGGUCAGUCCAGGAGCUAGGU
    NO 126
    SEQ ID AUAGUGGUCAGUCCAGGAGCU
    NO 127
    PS248
    SEQ ID UAGUGGUCAGUCCAGGAGCUAGGUC
    NO 128
    SEQ ID AGUGGUCAGUCCAGGAGCUAGGUCA
    NO 129
    SEQ ID GUGGUCAGUCCAGGAGCUAGGUCAG
    NO 130
    SEQ ID UGGUCAGUCCAGGAGCUAGGUCAGG
    NO 131
    SEQ ID GGUCAGUCCAGGAGCUAGGUCAGGC
    NO 132
    SEQ ID GUCAGUCCAGGAGCUAGGUCAGGCU
    NO 133
    SEQ ID UCAGUCCAGGAGCUAGGUCAGGCUG
    NO 134
    SEQ ID CAGUCCAGGAGCUAGGUCAGGCUGC
    NO 135
    SEQ ID AGUCCAGGAGCUAGGUCAGGCUGCU
    NO 136
    SEQ ID GUCCAGGAGCUAGGUCAGGCUGCUU
    NO 137
    SEQ ID UCCAGGAGCUAGGUCAGGCUGCUUU
    NO 138
    SEQ ID CCAGGAGCUAGGUCAGGCUGCUUUG
    NO 139
    SEQ ID CAGGAGCUAGGUCAGGCUGCUUUGC
    NO 140
    SEQ ID AGGAGCUAGGUCAGGCUGCUUUGCC
    NO 141
    SEQ ID GGAGCUAGGUCAGGCUGCUUUGCCC
    NO 142
    SEQ ID GAGCUAGGUCAGGCUGCUUUGCCCU
    NO 143
    PS247
    SEQ ID AGCUAGGUCAGGCUGCUUUGCCCUC
    NO 144
    PS245
    SEQ ID GCUAGGUCAGGCUGCUUUGCCCUCA
    NO 145
    SEQ ID CUCAGCUCUUGAAGUAAACGGUUUA
    NO 530
    SEQ ID CAGCUCUUGAAGUAAACGGUUUACC
    NO 532
    SEQ ID GCUCUUGAAGUAAACGGUUUACCGC
    NO 534
    SEQ ID NO CUAGGUCAGGCUGCUUUGCCCUCAG
    146
    SEQ ID NO UAGGUCAGGCUGCUUUGCCCUCAGC
    147
    SEQ ID NO AGGUCAGGCUGCUUUGCCCUCAGCU
    148
    SEQ ID NO GGUCAGGCUGCUUUGCCCUCAGCUC
    149
    SEQ ID NO GUCAGGCUGCUUUGCCCUCAGCUCU
    150
    SEQ ID NO UCAGGCUGCUUUGCCCUCAGCUCUU
    151
    SEQ ID NO CAGGCUGCUUUGCCCUCAGCUCUUG
    152
    SEQ ID NO AGGCUGCUUUGCCCUCAGCUCUUGA
    153
    SEQ ID NO GGCUGCUUUGCCCUCAGCUCUUGAA
    154
    SEQ ID NO GCUGCUUUGCCCUCAGCUCUUGAAG
    155
    SEQ ID NO CUGCUUUGCCCUCAGCUCUUGAAGU
    156
    SEQ ID NO UGCUUUGCCCUCAGCUCUUGAAGUA
    157
    SEQ ID NO GCUUUGCCCUCAGCUCUUGAAGUAA
    158
    SEQ ID NO CUUUGCCCUCAGCUCUUGAAGUAAA
    159
    SEQ ID NO UUUGCCCUCAGCUCUUGAAGUAAAC
    160
    SEQ ID NO UUGCCCUCAGCUCUUGAAGUAAACG
    161
    SEQ ID NO UGCCCUCAGCUCUUGAAGUAAACGG
    162
    SEQ ID NO GCCCUCAGCUCUUGAAGUAAACGGU
    163
    SEQ ID NO CCCUCAGCUCUUGAAGUAAACGGUU
    164
    SEQ ID NO CCUCAGCUCUUGAAGUAAAC
    165
    PS246
    SEQ ID NO CCUCAGCUCUUGAAGUAAACG
    166
    SEQ ID NO CUCAGCUCUUGAAGUAAACG
    167
    SEQ ID NO CCUCAGCUCUUGAAGUAAACGGUUU
    529
    SEQ ID NO UCAGCUCUUGAAGUAAACGGUUUAC
    531
    SEQ ID NO AGCUCUUGAAGUAAACGGUUUACCG
    533
    SEQ ID NO CUCUUGAAGUAAACGGUUUACCGCC
    535
  • TABLE 4
    oligonucleotides for skipping DMD Gene Exon 51
    SEQ ID GUACCUCCAACAUCAAGGAAGAUGG
    NO 168
    SEQ ID UACCUCCAACAUCAAGGAAGAUGGC
    NO 169
    SEQ ID ACCUCCAACAUCAAGGAAGAUGGCA
    NO 170
    SEQ ID CCUCCAACAUCAAGGAAGAUGGCAU
    NO 171
    SEQ ID CUCCAACAUCAAGGAAGAUGGCAUU
    NO 172
    SEQ ID UCCAACAUCAAGGAAGAUGGCAUUU
    NO 173
    SEQ ID CCAACAUCAAGGAAGAUGGCAUUUC
    NO 174
    SEQ ID CAACAUCAAGGAAGAUGGCAUUUCU
    NO 175
    SEQ ID AACAUCAAGGAAGAUGGCAUUUCUA
    NO 176
    SEQ ID ACAUCAAGGAAGAUGGCAUUUCUAG
    NO 177
    SEQ ID CAUCAAGGAAGAUGGCAUUUCUAGU
    NO 178
    SEQ ID AUCAAGGAAGAUGGCAUUUCUAGUU
    NO 179
    SEQ ID UCAAGGAAGAUGGCAUUUCUAGUUU
    NO 180
    SEQ ID CAAGGAAGAUGGCAUUUCUAGUUUG
    NO 181
    SEQ ID AAGGAAGAUGGCAUUUCUAGUUUGG
    NO 182
    SEQ ID AGGAAGAUGGCAUUUCUAGUUUGGA
    NO 183
    SEQ ID GGAAGAUGGCAUUUCUAGUUUGGAG
    NO 184
    SEQ ID GAAGAUGGCAUUUCUAGUUUGGAGA
    NO 185
    SEQ ID AAGAUGGCAUUUCUAGUUUGGAGAU
    NO 186
    SEQ ID AGAUGGCAUUUCUAGUUUGGAGAUG
    NO 187
    SEQ ID GAUGGCAUUUCUAGUUUGGAGAUGG
    NO 188
    SEQ ID AUGGCAUUUCUAGUUUGGAGAUGGC
    NO 189
    SEQ ID UGGCAUUUCUAGUUUGGAGAUGGCA
    NO 190
    SEQ ID GGCAUUUCUAGUUUGGAGAUGGCAG
    NO 191
    SEQ ID GCAUUUCUAGUUUGGAGAUGGCAGU
    NO 192
    SEQ ID CAUUUCUAGUUUGGAGAUGGCAGUU
    NO 193
    SEQ ID AUUUCUAGUUUGGAGAUGGCAGUUU
    NO 194
    SEQ ID UUUCUAGUUUGGAGAUGGCAGUUUC
    NO 195
    SEQ ID UUCUAGUUUGGAGAUGGCAGUUUCC
    NO 196
    SEQ ID UCUAGUUUGGAGAUGGCAGUUUCCU
    NO 197
    SEQ ID CUAGUUUGGAGAUGGCAGUUUCCUU
    NO 198
    SEQ ID UAGUUUGGAGAUGGCAGUUUCCUUA
    NO 199
    SEQ ID AGUUUGGAGAUGGCAGUUUCCUUAG
    NO 200
    SEQ ID GUUUGGAGAUGGCAGUUUCCUUAGU
    NO 201
    SEQ ID UUUGGAGAUGGCAGUUUCCUUAGUA
    NO 202
    SEQ ID UUGGAGAUGGCAGUUUCCUUAGUAA
    NO 203
    SEQ ID UGGAGAUGGCAGUUUCCUUAGUAAC
    NO 204
    SEQ ID NO GAGAUGGCAGUUUCCUUAGUAACCA
    205
    SEQ ID NO AGAUGGCAGUUUCCUUAGUAACCAC
    206
    SEQ ID NO GAUGGCAGUUUCCUUAGUAACCACA
    207
    SEQ ID NO AUGGCAGUUUCCUUAGUAACCACAG
    208
    SEQ ID NO UGGCAGUUUCCUUAGUAACCACAGG
    209
    SEQ ID NO GGCAGUUUCCUUAGUAACCACAGGU
    210
    SEQ ID NO GCAGUUUCCUUAGUAACCACAGGUU
    211
    SEQ ID NO CAGUUUCCUUAGUAACCACAGGUUG
    212
    SEQ ID NO AGUUUCCUUAGUAACCACAGGUUGU
    213
    SEQ ID NO GUUUCCUUAGUAACCACAGGUUGUG
    214
    SEQ ID NO UUUCCUUAGUAACCACAGGUUGUGU
    215
    SEQ ID NO UUCCUUAGUAACCACAGGUUGUGUC
    216
    SEQ ID NO UCCUUAGUAACCACAGGUUGUGUCA
    217
    SEQ ID NO CCUUAGUAACCACAGGUUGUGUCAC
    218
    SEQ ID NO CUUAGUAACCACAGGUUGUGUCACC
    219
    SEQ ID NO UUAGUAACCACAGGUUGUGUCACCA
    220
    SEQ ID NO UAGUAACCACAGGUUGUGUCACCAG
    221
    SEQ ID NO AGUAACCACAGGUUGUGUCACCAGA
    222
    SEQ ID NO GUAACCACAGGUUGUGUCACCAGAG
    223
    SEQ ID NO UAACCACAGGUUGUGUCACCAGAGU
    224
    SEQ ID NO AACCACAGGUUGUGUCACCAGAGUA
    225
    SEQ ID NO ACCACAGGUUGUGUCACCAGAGUAA
    226
    SEQ ID NO CCACAGGUUGUGUCACCAGAGUAAC
    227
    SEQ ID NO CACAGGUUGUGUCACCAGAGUAACA
    228
    SEQ ID NO ACAGGUUGUGUCACCAGAGUAACAG
    229
    SEQ ID NO CAGGUUGUGUCACCAGAGUAACAGU
    230
    SEQ ID NO AGGUUGUGUCACCAGAGUAACAGUC
    231
    SEQ ID NO GGUUGUGUCACCAGAGUAACAGUCU
    232
    SEQ ID NO GUUGUGUCACCAGAGUAACAGUCUG
    233
    SEQ ID NO UUGUGUCACCAGAGUAACAGUCUGA
    234
    SEQ ID NO UGUGUCACCAGAGUAACAGUCUGAG
    235
    SEQ ID NO GUGUCACCAGAGUAACAGUCUGAGU
    236
    SEQ ID NO UGUCACCAGAGUAACAGUCUGAGUA
    237
    SEQ ID NO GUCACCAGAGUAACAGUCUGAGUAG
    238
    SEQ ID NO UCACCAGAGUAACAGUCUGAGUAGG
    239
    SEQ ID NO CACCAGAGUAACAGUCUGAGUAGGA
    240
    SEQ ID NO ACCAGAGUAACAGUCUGAGUAGGAG
    241
  • TABLE 5
    oligonucleotides for skipping DMD Gene Exon 52
    SEQ ID AGCCUCUUGAUUGCUGGUCUUGUUU
    NO 242
    SEQ ID GCCUCUUGAUUGCUGGUCUUGUUUU
    NO 243
    SEQ ID CCUCUUGAUUGCUGGUCUUGUUUUU
    NO 244
    SEQ ID CCUCUUGAUUGCUGGUCUUG
    NO 245
    SEQ ID CUCUUGAUUGCUGGUCUUGUUUUUC
    NO 246
    PS232
    SEQ ID UCUUGAUUGCUGGUCUUGUUUUUCA
    NO 247
    SEQ ID CUUGAUUGCUGGUCUUGUUUUUCAA
    NO 248
    SEQ ID UUGAUUGCUGGUCUUGUUUUUCAAA
    NO 249
    SEQ ID UGAUUGCUGGUCUUGUUUUUCAAAU
    NO 250
    SEQ ID GAUUGCUGGUCUUGUUUUUCAAAUU
    NO 251
    SEQ ID GAUUGCUGGUCUUGUUUUUC
    NO 252
    SEQ ID AUUGCUGGUCUUGUUUUUCAAAUUU
    NO 253
    SEQ ID UUGCUGGUCUUGUUUUUCAAAUUUU
    NO 254
    SEQ ID UGCUGGUCUUGUUUUUCAAAUUUUG
    NO 255
    SEQ ID GCUGGUCUUGUUUUUCAAAUUUUGG
    NO 256
    SEQ ID CUGGUCUUGUUUUUCAAAUUUUGGG
    NO 257
    SEQ ID UGGUCUUGUUUUUCAAAUUUUGGGC
    NO 258
    SEQ ID GGUCUUGUUUUUCAAAUUUUGGGCA
    NO 259
    SEQ ID GUCUUGUUUUUCAAAUUUUGGGCAG
    NO 260
    SEQ ID UCUUGUUUUUCAAAUUUUGGGCAGC
    NO 261
    SEQ ID CUUGUUUUUCAAAUUUUGGGCAGCG
    NO 262
    SEQ ID UUGUUUUUCAAAUUUUGGGCAGCGG
    NO 263
    SEQ ID UGUUUUUCAAAUUUUGGGCAGCGGU
    NO 264
    PS236
    SEQ ID GUUUUUCAAAUUUUGGGCAGCGGUA
    NO 265
    SEQ ID UUUUUCAAAUUUUGGGCAGCGGUAA
    NO 266
    SEQ ID UUUUCAAAUUUUGGGCAGCGGUAAU
    NO 267
    SEQ ID UUUCAAAUUUUGGGCAGCGGUAAUG
    NO 268
    SEQ ID UUCAAAUUUUGGGCAGCGGUAAUGA
    NO 269
    SEQ ID UCAAAUUUUGGGCAGCGGUAAUGAG
    NO 270
    SEQ ID CAAAUUUUGGGCAGCGGUAAUGAGU
    NO 271
    SEQ ID AAAUUUUGGGCAGCGGUAAUGAGUU
    NO 272
    SEQ ID AAUUUUGGGCAGCGGUAAUGAGUUC
    NO 273
    SEQ ID AUUUUGGGCAGCGGUAAUGAGUUCU
    NO 274
    SEQ ID UUUUGGGCAGCGGUAAUGAGUUCUU
    NO 275
    SEQ ID UUUGGGCAGCGGUAAUGAGUUCUUC
    NO 276
    SEQ ID NO UUGGGCAGCGGUAAUGAGUUCUUCC
    277
    SEQ ID NO UGGGCAGCGGUAAUGAGUUCUUCCA
    278
    SEQ ID NO GGGCAGCGGUAAUGAGUUCUUCCAA
    279
    SEQ ID NO GGCAGCGGUAAUGAGUUCUUCCAAC
    280
    SEQ ID NO GCAGCGGUAAUGAGUUCUUCCAACU
    281
    SEQ ID NO CAGCGGUAAUGAGUUCUUCCAACUG
    282
    SEQ ID NO AGCGGUAAUGAGUUCUUCCAACUGG
    283
    SEQ ID NO GCGGUAAUGAGUUCUUCCAACUGGG
    284
    SEQ ID NO CGGUAAUGAGUUCUUCCAACUGGGG
    285
    SEQ ID NO GGUAAUGAGUUCUUCCAACUGGGGA
    286
    SEQ ID NO GGUAAUGAGUUCUUCCAACUGG
    287
    SEQ ID NO GUAAUGAGUUCUUCCAACUGGGGAC
    288
    SEQ ID NO UAAUGAGUUCUUCCAACUGGGGACG
    289
    SEQ ID NO AAUGAGUUCUUCCAACUGGGGACGC
    290
    SEQ ID NO AUGAGUUCUUCCAACUGGGGACGCC
    291
    SEQ ID NO UGAGUUCUUCCAACUGGGGACGCCU
    292
    SEQ ID NO GAGUUCUUCCAACUGGGGACGCCUC
    293
    SEQ ID NO AGUUCUUCCAACUGGGGACGCCUCU
    294
    SEQ ID NO GUUCUUCCAACUGGGGACGCCUCUG
    295
    SEQ ID NO UUCUUCCAACUGGGGACGCCUCUGU
    296
    SEQ ID NO UCUUCCAACUGGGGACGCCUCUGUU
    297
    SEQ ID NO CUUCCAACUGGGGACGCCUCUGUUC
    298
    SEQ ID NO UUCCAACUGGGGACGCCUCUGUUCC
    299
    SEQ ID NO UCCAACUGGGGACGCCUCUGUUCCA
    300
    SEQ ID NO CCAACUGGGGACGCCUCUGUUCCAA
    301
    SEQ ID NO CAACUGGGGACGCCUCUGUUCCAAA
    302
    SEQ ID NO AACUGGGGACGCCUCUGUUCCAAAU
    303
    SEQ ID NO ACUGGGGACGCCUCUGUUCCAAAUC
    304
    SEQ ID NO CUGGGGACGCCUCUGUUCCAAAUCC
    305
    SEQ ID NO UGGGGACGCCUCUGUUCCAAAUCCU
    306
    SEQ ID NO GGGGACGCCUCUGUUCCAAAUCCUG
    307
    SEQ ID NO GGGACGCCUCUGUUCCAAAUCCUGC
    308
    SEQ ID NO GGACGCCUCUGUUCCAAAUCCUGCA
    309
    SEQ ID NO GACGCCUCUGUUCCAAAUCCUGCAU
    310
  • TABLE 6
    oligonucleotides for skipping DMD Gene Exon 53
    SEQ ID CUCUGGCCUGUCCUAAGACCUGCUC
    NO 311
    SEQ ID UCUGGCCUGUCCUAAGACCUGCUCA
    NO 312
    SEQ ID CUGGCCUGUCCUAAGACCUGCUCAG
    NO 313
    SEQ ID UGGCCUGUCCUAAGACCUGCUCAGC
    NO 314
    SEQ ID GGCCUGUCCUAAGACCUGCUCAGCU
    NO 315
    SEQ ID GCCUGUCCUAAGACCUGCUCAGCUU
    NO 316
    SEQ ID CCUGUCCUAAGACCUGCUCAGCUUC
    NO 317
    SEQ ID CUGUCCUAAGACCUGCUCAGCUUCU
    NO 318
    SEQ ID UGUCCUAAGACCUGCUCAGCUUCUU
    NO 319
    SEQ ID GUCCUAAGACCUGCUCAGCUUCUUC
    NO 320
    SEQ ID UCCUAAGACCUGCUCAGCUUCUUCC
    NO 321
    SEQ ID CCUAAGACCUGCUCAGCUUCUUCCU
    NO 322
    SEQ ID CUAAGACCUGCUCAGCUUCUUCCUU
    NO 323
    SEQ ID UAAGACCUGCUCAGCUUCUUCCUUA
    NO 324
    SEQ ID AAGACCUGCUCAGCUUCUUCCUUAG
    NO 325
    SEQ ID AGACCUGCUCAGCUUCUUCCUUAGC
    NO 326
    SEQ ID GACCUGCUCAGCUUCUUCCUUAGCU
    NO 327
    SEQ ID ACCUGCUCAGCUUCUUCCUUAGCUU
    NO 328
    SEQ ID CCUGCUCAGCUUCUUCCUUAGCUUC
    NO 329
    SEQ ID CUGCUCAGCUUCUUCCUUAGCUUCC
    NO 330
    SEQ ID UGCUCAGCUUCUUCCUUAGCUUCCA
    NO 331
    SEQ ID GCUCAGCUUCUUCCUUAGCUUCCAG
    NO 332
    SEQ ID CUCAGCUUCUUCCUUAGCUUCCAGC
    NO 333
    SEQ ID UCAGCUUCUUCCUUAGCUUCCAGCC
    NO 334
    SEQ ID NO CAGCUUCUUCCUUAGCUUCCAGCCA
    335
    SEQ ID NO AGCUUCUUCCUUAGCUUCCAGCCAU
    336
    SEQ ID NO GCUUCUUCCUUAGCUUCCAGCCAUU
    337
    SEQ ID NO CUUCUUCCUUAGCUUCCAGCCAUUG
    338
    SEQ ID NO UUCUUCCUUAGCUUCCAGCCAUUGU
    339
    SEQ ID NO UCUUCCUUAGCUUCCAGCCAUUGUG
    340
    SEQ ID NO CUUCCUUAGCUUCCAGCCAUUGUGU
    341
    SEQ ID NO UUCCUUAGCUUCCAGCCAUUGUGUU
    342
    SEQ ID NO UCCUUAGCUUCCAGCCAUUGUGUUG
    343
    SEQ ID NO CCUUAGCUUCCAGCCAUUGUGUUGA
    344
    SEQ ID NO CUUAGCUUCCAGCCAUUGUGUUGAA
    345
    SEQ ID NO UUAGCUUCCAGCCAUUGUGUUGAAU
    346
    SEQ ID NO UAGCUUCCAGCCAUUGUGUUGAAUC
    347
    SEQ ID NO AGCUUCCAGCCAUUGUGUUGAAUCC
    348
    SEQ ID NO GCUUCCAGCCAUUGUGUUGAAUCCU
    349
    SEQ ID NO CUUCCAGCCAUUGUGUUGAAUCCUU
    350
    SEQ ID NO UUCCAGCCAUUGUGUUGAAUCCUUU
    351
    SEQ ID NO UCCAGCCAUUGUGUUGAAUCCUUUA
    352
    SEQ ID NO CCAGCCAUUGUGUUGAAUCCUUUAA
    353
    SEQ ID NO CAGCCAUUGUGUUGAAUCCUUUAAC
    354
    SEQ ID NO AGCCAUUGUGUUGAAUCCUUUAACA
    355
    SEQ ID NO GCCAUUGUGUUGAAUCCUUUAACAU
    356
    SEQ ID NO CCAUUGUGUUGAAUCCUUUAACAUU
    357
    SEQ ID NO CAUUGUGUUGAAUCCUUUAACAUUU
    358
  • TABLE 7
    oligonucleotides for skipping other exons of the
    DMD gene as identified
    DMD Gene Exon 6
    SEQ ID CAUUUUUGACCUACAUGUGG
    NO 359
    SEQ ID UUUGACCUACAUGUGGAAAG
    NO 360
    SEQ ID UACAUUUUUGACCUACAUGUGGAA
    NO 361 AG
    SEQ ID GGUCUCCUUACCUAUGA
    NO 362
    SEQ ID UCUUACCUAUGACUAUGGAUGAGA
    NO 363
    SEQ ID NO AUUUUUGACCUACAUGGGAAA G
    364
    SEQ ID NO UACGAGUUGAUUGUCGGACCCAG
    365
    SEQ ID NO GUGGUCUCCUUACCUAUGACUGUGG
    366
    SEQ ID NO UGUCUCAGUAAUCUUCUUACCUAU
    367
    DMD Gene Exon 7
    SEQ ID UGCAUGUUCCAGUCGUUGUGUGG
    NO 368
    SEQ ID CACUAUUCCAGUCAAAUAGGUCUGG
    NO 369
    SEQ ID NO 370 AUUUACCAACCUUCAGGAUCGAGU
    A
    SEQ ID NO 371 GGCCUAAAACACAUACACAUA
    DMD Gene Exon 11
    SEQ ID CCCUGAGGCAUUCCCAUCUUGAAU
    NO 372
    SEQ ID AGGACUUACUUGCUUUGUUU
    NO 373
    SEQ ID CUUGAAUUUAGGAGAUUCAUCU
    NO 374 G
    SEQ ID CAUCUUCUGAUAAUUUUCCUGUU
    NO 375
    DMD Gene Exon 17
    SEQ ID CCAUUACAGUUGUCUGUGUU
    NO 376
    SEQ ID UGACAGCCUGUGAAAUCUGUGAG
    NO 377
    SEQ ID UAAUCUGCCUCUUCUUUUGG
    NO 378
    DMD Gene Exon 19
    SEQ ID CAGCAGUAGUUGUCAUCUGC
    NO 379
    SEQ ID GCCUGAGCUGAUCUGCUGGCAUCUUGC
    NO 380
    SEQ ID GCCUGAGCUGAUCUGCUGGCAUC
    NO 381 UUGCAGUU
    SEQ ID UCUGCUGGCAUCUUGC
    NO 382
    DMD Gene Exon 21
    SEQ ID GCCGGUUGACUUCAUCCUGUGC
    NO 383
    SEQ ID GUCUGCAUCCAGGAACAUGGGUC
    NO 384
    SEQ ID UACUUACUGUCUGUAGCUCUUUCU
    NO 385
    SEQ ID CUGCAUCCAGGAACAUGGGUCC
    NO 386
    SEQ ID GUUGAAGAUCUGAUAGCCGGUUGA
    NO 387
    DMD Gene Exon 44
    SEQ ID UCAGCUUCUGUUAGCCACUG
    NO 388
    SEQ ID UUCAGCUUCUGUUAGCCACU
    NO 389
    SEQ ID UUCAGCUUCUGUUAGCCACUG
    NO 390
    SEQ ID UCAGCUUCUGUUAGCCACUGA
    NO 391
    SEQ ID UUCAGCUUCUGUUAGCCACUGA
    NO 392
    SEQ ID UCAGCUUCUGUUAGCCACUGA
    NO 393
    SEQ ID UUCAGCUUCUGUUAGCCACUGA
    NO 394
    SEQ ID UCAGCUUCUGUUAGCCACUGAU
    NO 395
    SEQ ID UUCAGCUUCUGUUAGCCACUGAU
    NO 396
    SEQ ID UCAGCUUCUGUUAGCCACUGAUU
    NO 397
    SEQ ID UUCAGCUUCUGUUAGCCACUGAUU
    NO 398
    SEQ ID UCAGCUUCUGUUAGCCACUGAUUA
    NO 399
    SEQ ID UUCAGCUUCUGUUAGCCACUGAUA
    NO 400
    SEQ ID UCAGCUUCUGUUAGCCACUGAUUAA
    NO 401
    SEQ ID UUCAGCUUCUGUUAGCCACUGAUUAA
    NO 402
    SEQ ID UCAGCUUCUGUUAGCCACUGAUUAAA
    NO 403
    SEQ ID UUCAGCUUCUGUUAGCCACUGAUUAAA
    NO 404
    SEQ ID CAGCUUCUGUUAGCCACUG
    NO 405
    SEQ ID CAGCUUCUGUUAGCCACUGAU
    NO 406
    SEQ ID AGCUUCUGUUAGCCACUGAUU
    NO 407
    SEQ ID CAGCUUCUGUUAGCCACUGAUU
    NO 408
    SEQ ID AGCUUCUGUUAGCCACUGAUUA
    NO 409
    SEQ ID CAGCUUCUGUUAGCCACUGAUUA
    NO 410
    SEQ ID AGCUUCUGUUAGCCACUGAUUAA
    NO 411
    SEQ ID CAGCUUCUGUUAGCCACUGAUUAA
    NO 412
    SEQ ID AGCUUCUGUUAGCCACUGAUUAAA
    NO 413
    SEQ ID CAGCUUCUGUUAGCCACUGAUUAAA
    NO 414
    SEQ ID AGCUUCUGUUAGCCACUGAUUAAA
    NO 415
    SEQ ID AGCUUCUGUUAGCCACUGAU
    NO 416
    SEQ ID GCUUCUGUUAGCCACUGAUU
    NO 417
    SEQ ID AGCUUCUGUUAGCCACUGAUU
    NO 418
    SEQ ID GCUUCUGUUAGCCACUGAUUA
    NO 419
    SEQ ID AGCUUCUGUUAGCCACUGAUUA
    NO 420
    SEQ ID GCUUCUGUUAGCCACUGAUUAA
    NO 421
    SEQ ID AGCUUCUGUUAGCCACUGAUUAA
    NO 422
    SEQ ID GCUUCUGUUAGCCACUGAUUAAA
    NO 423
    SEQ ID AGCUUCUGUUAGCCACUGAUUAAA
    NO 424
    SEQ ID GCUUCUGUUAGCCACUGAUUAAA
    NO 425
    SEQ ID CCAUUUGUAUUUAGCAUGUUCCC
    NO 426
    SEQ ID AGAUACCAUUUGUAUUUAGC
    NO 427
    SEQ ID GCCAUUUCUCAACAGAUCU
    NO 428
    SEQ ID GCCAUUUCUCAACAGAUCUGUCA
    NO 429
    SEQ ID AUUCUCAGGAAUUUGUGUCUUUC
    NO 430
    SEQ ID UCUCAGGAAUUUGUGUCUUUC
    NO 431
    SEQ ID GUUCAGCUUCUGUUAGCC
    NO 432
    SEQ ID CUGAUUAAAUAUCUUUAUAU C
    NO 433
    SEQ ID GCCGCCAUUUCUCAACAG
    NO 434
    SEQ ID GUAUUUAGCAUGUUCCCA
    NO 435
    SEQ ID CAGGAAUUUGUGUCUUUC
    NO 436
    DMD Gene Exon 45
    SEQ ID UUUGCCGCUGCCCAAUGCCAUCCUG
    NO 437
    SEQ ID AUUCAAUGUUCUGACAACAGUUUGC
    NO 438
    SEQ ID CCAGUUGCAUUCAAUGUUCUGACAA
    NO 439
    SEQ ID CAGUUGCAUUCAAUGUUCUGAC
    NO 440
    SEQ ID AGUUGCAUUCAAUGUUCUGA
    NO 441
    SEQ ID GAUUGCUGAAUUAUUUCUUCC
    NO 442
    SEQ ID GAUUGCUGAAUUAUUUCUUCCCCAG
    NO 443
    SEQ ID AUUGCUGAAUUAUUUCUUCCCCAGU
    NO 444
    SEQ ID UUGCUGAAUUAUUUCUUCCCCAGUU
    NO 445
    SEQ ID UGCUGAAUUAUUUCUUCCCCAGUUG
    NO 446
    SEQ ID GCUGAAUUAUUUCUUCCCCAGUUGC
    NO 447
    SEQ ID CUGAAUUAUUUCUUCCCCAGUUGCA
    NO 448
    SEQ ID UGAAUUAUUUCUUCCCCAGUUGCAU
    NO 449
    SEQ ID GAAUUAUUUCUUCCCCAGUUGCAUU
    NO 450
    SEQ ID AAUUAUUUCUUCCCCAGUUGCAUUC
    NO 451
    SEQ ID AUUAUUUCUUCCCCAGUUGCAUUCA
    NO 452
    SEQ ID UUAUUUCUUCCCCAGUUGCAUUCAA
    NO 453
    SEQ ID UAUUUCUUCCCCAGUUGCAUUCAAU
    NO 454
    SEQ ID AUUUCUUCCCCAGUUGCAUUCAAUG
    NO 455
    SEQ ID UUUCUUCCCCAGUUGCAUUCAAUGU
    NO 456
    SEQ ID UUCUUCCCCAGUUGCAUUCAAUGUU
    NO 457
    SEQ ID UCUUCCCCAGUUGCAUUCAAUGUUC
    NO 458
    SEQ ID CUUCCCCAGUUGCAUUCAAUGUUCU
    NO 459
    SEQ ID UUCCCCAGUUGCAUUCAAUGUUCUG
    NO 460
    SEQ ID UCCCCAGUUGCAUUCAAUGUUCUGA
    NO 461
    SEQ ID CCCCAGUUGCAUUCAAUGUUCUGAC
    NO 462
    SEQ ID CCCAGUUGCAUUCAAUGUUCUGACA
    NO 463
    SEQ ID CCAGUUGCAUUCAAUGUUCUGACAA
    NO 464
    SEQ ID CAGUUGCAUUCAAUGUUCUGACAAC
    NO 465
    SEQ ID AGUUGCAUUCAAUGUUCUGACAACA
    NO 466
    SEQ ID UCC UGU AGA AUA CUG GCA UC
    NO 467
    SEQ ID UGCAGACCUCCUGCCACCGCAGAUUCA
    NO 468
    SEQ ID UUGCAGACCUCCUGCCACCGCAGAUUCAGGC
    NO 469 UUC
    SEQ ID GUUGCAUUCAAUGUUCUGACAACAG
    NO 470
    SEQ ID UUGCAUUCAAUGUUCUGACAACAGU
    NO 471
    SEQ ID UGCAUUCAAUGUUCUGACAACAGUU
    NO 472
    SEQ ID GCAUUCAAUGUUCUGACAACAGUUU
    NO 473
    SEQ ID CAUUCAAUGUUCUGACAACAGUUUG
    NO 474
    SEQ ID AUUCAAUGUUCUGACAACAGUUUGC
    NO 475
    SEQ ID UCAAUGUUCUGACAACAGUUUGCCG
    NO 476
    SEQ ID CAAUGUUCUGACAACAGUUUGCCGC
    NO 477
    SEQ ID AAUGUUCUGACAACAGUUUGCCGCU
    NO 478
    SEQ ID AUGUUCUGACAACAGUUUGCCGCUG
    NO 479
    SEQ ID UGUUCUGACAACAGUUUGCCGCUGC
    NO 480
    SEQ ID GUUCUGACAACAGUUUGCCGCUGCC
    NO 481
    SEQ ID UUCUGACAACAGUUUGCCGCUGCCC
    NO 482
    SEQ ID UCUGACAACAGUUUGCCGCUGCCCA
    NO 483
    SEQ ID CUGACAACAGUUUGCCGCUGCCCAA
    NO 484
    SEQ ID UGACAACAGUUUGCCGCUGCCCAAU
    NO 485
    SEQ ID GACAACAGUUUGCCGCUGCCCAAUG
    NO 486
    SEQ ID ACAACAGUUUGCCGCUGCCCAAUGC
    NO 487
    SEQ ID CAACAGUUUGCCGCUGCCCAAUGCC
    NO 488
    SEQ ID AACAGUUUGCCGCUGCCCAAUGCCA
    NO 489
    SEQ ID ACAGUUUGCCGCUGCCCAAUGCCAU
    NO 490
    SEQ ID CAGUUUGCCGCUGCCCAAUGCCAUC
    NO 491
    SEQ ID AGUUUGCCGCUGCCCAAUGCCAUCC
    NO 492
    SEQ ID GUUUGCCGCUGCCCAAUGCCAUCCU
    NO 493
    SEQ ID UUUGCCGCUGCCCAAUGCCAUCCUG
    NO 494
    SEQ ID UUGCCGCUGCCCAAUGCCAUCCUGG
    NO 495
    SEQ ID UGCCGCUGCCCAAUGCCAUCCUGGA
    NO 496
    SEQ ID GCCGCUGCCCAAUGCCAUCCUGGAG
    NO 497
    SEQ ID CCGCUGCCCAAUGCCAUCCUGGAGU
    NO 498
    SEQ ID CGCUGCCCAAUGCCAUCCUGGAGUU
    NO 499
    SEQ ID UGUUUUUGAGGAUUGCUGAA
    NO 500
    SEQ ID UGUUCUGACAACAGUUUGCCGCU
    NO 501 GCCCAAUGCCAUCCUGG
    DMD Gene Exon 55
    SEQ ID CUGUUGCAGUAAUCUAUGAG
    NO 502
    SEQ ID UGCAGUAAUCUAUGAGUUUC
    NO 503
    SEQ ID GAGUCUUCUAGGAGCCUU
    NO 504
    SEQ ID UGCCAUUGUUUCAUCAGCUCUUU
    NO 505
    SEQ ID UCCUGUAGGACAUUGGCAGU
    NO 506
    SEQ ID CUUGGAGUCUUCUAGGAGCC
    NO 507
    DMD Gene Exon 57
    SEQ ID UAGGUGCCUGCCGGCUU
    NO 508
    SEQ ID UUCAGCUGUAGCCACACC
    NO 509
    SEQ ID CUGAACUGCUGGAAAGUCGCC
    NO 510
    SEQ ID CUGGCUUCCAAAUGGGACCUGAA
    NO 511 AAAGAAC
    DMD Gene Exon 59
    SEQ ID CAAUUUUUCCCACUCAGUAUU
    NO 512
    SEQ ID UUGAAGUUCCUGGAGUCUU
    NO 513
    SEQ ID UCCUCAGGAGGCAGCUCUAAAU
    NO 514
    DMD Gene Exon 62
    SEQ ID UGGCUCUCUCCCAGGG
    NO 515
    SEQ ID GAGAUGGCUCUCUCCCAGGGACCCUGG
    NO 516
    SEQ ID GGGCACUUUGUUUGGCG
    NO 517
    DMD Gene Exon 63
    SEQ ID GGUCCCAGCAAGUUGUUUG
    NO 518
    SEQ ID UGGGAUGGUCCCAGCAAGUUGUUUG
    NO 519
    SEQ ID GUAGAGCUCUGUCAUUUUGGG
    NO 520
    DMD Gene Exon 65
    SEQ ID GCUCAAGAGAUCCACUGCAAAAAAC
    NO 521
    SEQ ID GCCAUACGUACGUAUCAUAAACAUUC
    NO 522
    SEQ ID UCUGCAGGAUAUCCAUGGGCUGGUC
    NO 523
    DMD Gene Exon 66
    SEQ ID GAUCCUCCCUGUUCGUCCCCUAUUAUG
    NO 524
    DMD Gene Exon 69
    SEQ ID UGCUUUAGACUCCUGUACCUGAUA
    NO 525
    DMD Gene Exon 75
    SEQ ID GGCGGCCUUUGUGUUGAC
    NO 526
    SEQ ID GGACAGGCCUUUAUGUUCGUGCUGC
    NO 527
    SEQ ID CCUUUAUGUUCGUGCUGCU
    NO 528
  • FIGURE LEGENDS
  • FIG. 1. In human control myotubes, a series of AONs (PS237, PS238, and PS240; SEQ ID NO 65, 66, 16 respectively) targeting exon 43 was tested at 500 nM. PS237
  • (SEQ ID NO 65) reproducibly induced highest levels of exon 43 skipping. (M: DNA size marker; NT: non-treated cells)
  • FIG. 2. In myotubes from a DMD patient with an exon 45 deletion, a series of AONs (PS177, PS179, PS181, and PS182; SEQ ID NO 91, 70, 110, and 117 respectively) targeting exon 46 was tested at two different concentrations (50 and 150 nM). PS182 (SEQ ID NO 117) reproducibly induced highest levels of exon 46 skipping. (M: DNA size marker)
  • FIG. 3. In human control myotubes, a series of AONs (PS245, PS246, PS247, and PS248; SEQ ID NO 167, 165, 166, and 127 respectively) targeting exon 50 was tested at 500 nM. PS248 (SEQ ID NO 127) reproducibly induced highest levels of exon 50 skipping. (M: DNA size marker; NT: non-treated cells).
  • FIG. 4. In human control myotubes, two novel AONs (PS232 and PS236; SEQ ID NO 246 and 299 respectively) targeting exon 52 were tested at two different concentrations (200 and 500 nM) and directly compared to a previously described AON (52-1). PS236 (SEQ ID NO 299) reproducibly induced highest levels of exon 52 skipping. (M: DNA size marker; NT: non-treated cells).

Claims (19)

1. An isolated antisense oligonucleotide which is fully complementary to 8-22 consecutive nucleotides of a sequence of an exon of human dystrophin pre-mRNA, said oligonucleotide comprising a locked nucleic acid, wherein said sequence of said exon is selected from the group consisting of:
(SEQ ID NO: 2) 5′-AGAUAGUCUACAACAAAGCUCAGGUCGGAUUGACAUUAUUCAUAG CAAGAAGACAGCAGCAUUGCAAAGUGCAACGCCUGUGG-3′, and wherein said oligonucleotide is capable of skipping exon 43; (SEQ ID NO: 3) 5′-UUAUGGUUGGAGGAAGCAGAUAACAUUGCUAGUAUCCCACUUGAA CCUGGAAAAGAGCAGCAACUAAAAGAAAAGC-3′, and wherein said oligonucleotide is capable of skipping exon 46; (SEQ ID NO: 4) 5′-GCGGTAAACCGUUUACUUCAAGAGCUGAGGGCAAAGCAGCCUGAC CUAGCUCCUGGACUGACCACUAUUGG-3′, and wherein said oligonucleotide is capable of skipping exon 50; (SEQ ID NO: 5) 5′-CUCCUACUCAGACUGUUACUCUGGUGACACAACCUGUGGUUACUA AGGAAACUGCCAUCUCCAAACUAGAAAUGCCAUCUUCCUUGAUG UUGGAGGUAC-3′, and wherein said oligonucleotide is capable of skipping exon 51; (SEQ ID NO: 6) 5′-AUGCAGGAUUUGGAACAGAGGCGUCCCCAGUUGGAAGAACUCAUU ACCGCUGCCCAAAAUUUGAAAAACAAGACCAGCAAUCAAGAGGCU- 3′, and wherein said oligonucleotide is capable of skipping exon 52, and (SEQ ID NO: 7) 5′-AAUGUUAAAGGAUUCAACACAAUGGCUGGAAGCUAAGGAAAAGCU GAGCAGGUCUUAGGACAGGCCAGAG-3′, and wherein said oligonucleotide is capable of skipping exon 53.
2. The isolated antisense oligonucleotide of claim 1, wherein the oligonucleotide is complementary to 10-22 consecutive nucleotides of said sequence of said exon.
3. The isolated antisense oligonucleotide of claim 2, wherein the oligonucleotide is complementary to 12-20 consecutive nucleotides of said sequence of said exon.
4. The isolated antisense oligonucleotide of claim 3, wherein said oligonucleotide is 13-18 nucleotides in length.
5. The isolated antisense oligonucleotide of claim 1, wherein said oligonucleotide comprises 2′-O-methyl-phosphorothioate modifications.
6. The isolated antisense oligonucleotide of claim 1, wherein said oligonucleotide comprises a peptide nucleic acid and a morpholino phosphorodiamidate or a combination thereof.
7. The isolated antisense oligonucleotide of claim 1, wherein said oligonucleotide comprises a peptide nucleic acid.
8. The isolated antisense oligonucleotide of claim 1, wherein said oligonucleotide comprises a morpholino phosphorodiamidate.
9. The isolated antisense oligonucleotide of claim 1, wherein said LNA comprises 2′-O,4′-C-ethylene-bridge.
10. The isolated antisense oligonucleotide of claim 1, wherein said oligonucleotide comprises a peptide linked phosphorodiamidate morpholino oligomer (PMO).
11. The isolated antisense oligonucleotide of claim 1, wherein said oligonucleotide is capable of inducing exon skipping by at least 30%.
12. The isolated antisense oligonucleotide of claim 1, wherein said oligonucleotide comprises a backbone selected from a group consisting of: a morpholino backbone, a carbamate backbone, a siloxane backbone, a sulfide backbone, a sulfoxide backbone, a sulfone backbone, a formacetyl backbone, a thioformacetyl backbone, a methyleneformacetyl backbone, a riboacetyl backbone, an alkene containing backbone, a sulfamate backbone, a sulfonate backbone, a sulfonamide backbone, a methyleneimino backbone, a methylenehydrazino backbone and an amide backbone.
13. The isolated antisense oligonucleotide of claim 1, said oligonucleotide being RNA.
14. A pharmaceutical composition comprising at least two distinct isolated antisense oligonucleotides as defined claim 1, wherein said pharmaceutical composition further comprises a pharmaceutically acceptable carrier, adjuvant, diluent and/or excipient.
15. A method for treating one or more symptom(s) of Duchenne Muscular Dystrophy or Becker Muscular Dystrophy in an individual, the method comprising administering to said individual a pharmaceutical composition of claim 15, wherein said composition induces skipping of exons of dystrophin pre-mRNA.
16. A method for treating one or more symptom(s) of Duchenne Muscular Dystrophy or Becker Muscular Dystrophy in a cell, the method comprising administering to said cell a pharmaceutical composition of claim 15, wherein said composition induces skipping of exons of dystrophin pre-mRNA.
17. A method for inducing skipping of an exon of human dystrophin pre-mRNA in a muscle cell, the method comprising contacting said cell with an oligonucleotide of claim 1 for a time and under conditions which permit exon skipping. 18 (Original) A method for inducing skipping of an exon of human dystrophin pre-mRNA in a human subject, the method comprising administering an oligonucleotide of claim 1 to said subject in an amount and for a time which is effective to induce exon skipping.
19. A method for treating one or more symptom(s) of Duchenne Muscular Dystrophy or Becker Muscular Dystrophy in an individual, the method comprising administering to said individual an oligonucleotide of claim 1, wherein said oligonucleotide induces skipping of an exon of a dystrophin pre-mRNA.
20. A method for treating one or more symptom(s) of Duchenne Muscular Dystrophy or Becker Muscular Dystrophy in a cell, the method comprising administering to said cell one or more isolated oligonucleotides of claim 1.
US15/289,053 2007-10-26 2016-10-07 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 Abandoned US20170044534A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US15/289,053 US20170044534A1 (en) 2007-10-26 2016-10-07 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
US16/024,558 US10876114B2 (en) 2007-10-26 2018-06-29 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
US17/129,117 US20210139904A1 (en) 2007-10-26 2020-12-21 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

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
EP07119351 2007-10-26
PCT/NL2008/050673 WO2009054725A2 (en) 2007-10-26 2008-10-27 Means and methods for counteracting muscle disorders
PCT/NL2009/050113 WO2010050802A2 (en) 2008-10-27 2009-03-11 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.
US13/094,571 US20110263682A1 (en) 2007-10-26 2011-04-26 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
US14/631,686 US9499818B2 (en) 2007-10-26 2015-02-25 Methods and means for efficient skipping of at least one of the exons 51-53, 55, 57 and 59 of the human duchenne muscular dystrophy gene
US15/289,053 US20170044534A1 (en) 2007-10-26 2016-10-07 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

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US14/631,686 Continuation US9499818B2 (en) 2007-10-26 2015-02-25 Methods and means for efficient skipping of at least one of the exons 51-53, 55, 57 and 59 of the human duchenne muscular dystrophy gene

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/024,558 Continuation US10876114B2 (en) 2007-10-26 2018-06-29 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

Publications (1)

Publication Number Publication Date
US20170044534A1 true US20170044534A1 (en) 2017-02-16

Family

ID=39045623

Family Applications (18)

Application Number Title Priority Date Filing Date
US12/767,702 Ceased US9243245B2 (en) 2007-10-26 2010-04-26 Means and methods for counteracting muscle disorders
US13/094,548 Active US9926557B2 (en) 2007-10-26 2011-04-26 Methods and means for efficient skipping of exon 45 in Duchenne muscular dystrophy pre-mRNA
US13/094,571 Abandoned US20110263682A1 (en) 2007-10-26 2011-04-26 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
US14/097,210 Abandoned US20140113955A1 (en) 2007-10-26 2013-12-04 METHODS AND MEANS FOR EFFICIENT SKIPPING OF EXON 45 IN DUCHENNE MUSCULAR DYSTROPHY PRE-mRNA
US14/134,971 Abandoned US20140128592A1 (en) 2007-10-26 2013-12-19 METHODS AND MEANS FOR EFFICIENT SKIPPING OF EXON 45 IN DUCHENNE MUSCULAR DYSTROPHY PRE-mRNA
US14/200,251 Abandoned US20140221458A1 (en) 2007-10-26 2014-03-07 METHODS AND MEANS FOR EFFICIENT SKIPPING OF EXON 45 IN DUCHENNE MUSCULAR DYSTROPHY PRE-mRNA
US14/542,183 Active US9528109B2 (en) 2007-10-26 2014-11-14 Methods and means for efficient skipping of exon 45 in duchenne muscular dystrophy pre-mRNA
US14/631,686 Active US9499818B2 (en) 2007-10-26 2015-02-25 Methods and means for efficient skipping of at least one of the exons 51-53, 55, 57 and 59 of the human duchenne muscular dystrophy gene
US14/990,712 Abandoned US20160304864A1 (en) 2007-10-26 2016-01-07 Means and methods for counteracting muscle disorders
US15/289,053 Abandoned US20170044534A1 (en) 2007-10-26 2016-10-07 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
US15/390,836 Abandoned US20170107512A1 (en) 2007-10-26 2016-12-27 METHODS AND MEANS FOR EFFICIENT SKIPPING OF EXON 45 IN DUCHENNE MUSCULAR DYSTROPHY PRE-mRNA
US16/024,558 Active US10876114B2 (en) 2007-10-26 2018-06-29 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
US16/229,534 Abandoned US20190112604A1 (en) 2007-10-26 2018-12-21 METHODS AND MEANS FOR EFFICIENT SKIPPING OF EXON 45 IN DUCHENNE MUSCULAR DYSTROPHY PRE-mRNA
US16/229,821 Abandoned US20190119679A1 (en) 2007-10-26 2018-12-21 METHODS AND MEANS FOR EFFICIENT SKIPPING OF EXON 45 IN DUCHENNE MUSCULAR DYSTROPHY PRE-mRNA
US16/283,458 Abandoned US20190177725A1 (en) 2007-10-26 2019-02-22 METHODS AND MEANS FOR EFFICIENT SKIPPING OF EXON 45 IN DUCHENNE MUSCULAR DYSTROPHY PRE-mRNA
US16/584,115 Active US11427820B2 (en) 2007-10-26 2019-09-26 Methods and means for efficient skipping of exon 45 in Duchenne muscular dystrophy pre-mRNA
US17/129,117 Pending US20210139904A1 (en) 2007-10-26 2020-12-21 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
US17/814,781 Pending US20230151362A1 (en) 2007-10-26 2022-07-25 Methods and means for efficient dkipping of exon 45 in duchenne muscular dystrophy pre-mrna

Family Applications Before (9)

Application Number Title Priority Date Filing Date
US12/767,702 Ceased US9243245B2 (en) 2007-10-26 2010-04-26 Means and methods for counteracting muscle disorders
US13/094,548 Active US9926557B2 (en) 2007-10-26 2011-04-26 Methods and means for efficient skipping of exon 45 in Duchenne muscular dystrophy pre-mRNA
US13/094,571 Abandoned US20110263682A1 (en) 2007-10-26 2011-04-26 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
US14/097,210 Abandoned US20140113955A1 (en) 2007-10-26 2013-12-04 METHODS AND MEANS FOR EFFICIENT SKIPPING OF EXON 45 IN DUCHENNE MUSCULAR DYSTROPHY PRE-mRNA
US14/134,971 Abandoned US20140128592A1 (en) 2007-10-26 2013-12-19 METHODS AND MEANS FOR EFFICIENT SKIPPING OF EXON 45 IN DUCHENNE MUSCULAR DYSTROPHY PRE-mRNA
US14/200,251 Abandoned US20140221458A1 (en) 2007-10-26 2014-03-07 METHODS AND MEANS FOR EFFICIENT SKIPPING OF EXON 45 IN DUCHENNE MUSCULAR DYSTROPHY PRE-mRNA
US14/542,183 Active US9528109B2 (en) 2007-10-26 2014-11-14 Methods and means for efficient skipping of exon 45 in duchenne muscular dystrophy pre-mRNA
US14/631,686 Active US9499818B2 (en) 2007-10-26 2015-02-25 Methods and means for efficient skipping of at least one of the exons 51-53, 55, 57 and 59 of the human duchenne muscular dystrophy gene
US14/990,712 Abandoned US20160304864A1 (en) 2007-10-26 2016-01-07 Means and methods for counteracting muscle disorders

Family Applications After (8)

Application Number Title Priority Date Filing Date
US15/390,836 Abandoned US20170107512A1 (en) 2007-10-26 2016-12-27 METHODS AND MEANS FOR EFFICIENT SKIPPING OF EXON 45 IN DUCHENNE MUSCULAR DYSTROPHY PRE-mRNA
US16/024,558 Active US10876114B2 (en) 2007-10-26 2018-06-29 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
US16/229,534 Abandoned US20190112604A1 (en) 2007-10-26 2018-12-21 METHODS AND MEANS FOR EFFICIENT SKIPPING OF EXON 45 IN DUCHENNE MUSCULAR DYSTROPHY PRE-mRNA
US16/229,821 Abandoned US20190119679A1 (en) 2007-10-26 2018-12-21 METHODS AND MEANS FOR EFFICIENT SKIPPING OF EXON 45 IN DUCHENNE MUSCULAR DYSTROPHY PRE-mRNA
US16/283,458 Abandoned US20190177725A1 (en) 2007-10-26 2019-02-22 METHODS AND MEANS FOR EFFICIENT SKIPPING OF EXON 45 IN DUCHENNE MUSCULAR DYSTROPHY PRE-mRNA
US16/584,115 Active US11427820B2 (en) 2007-10-26 2019-09-26 Methods and means for efficient skipping of exon 45 in Duchenne muscular dystrophy pre-mRNA
US17/129,117 Pending US20210139904A1 (en) 2007-10-26 2020-12-21 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
US17/814,781 Pending US20230151362A1 (en) 2007-10-26 2022-07-25 Methods and means for efficient dkipping of exon 45 in duchenne muscular dystrophy pre-mrna

Country Status (17)

Country Link
US (18) US9243245B2 (en)
EP (4) EP3238737B1 (en)
JP (6) JP5600064B2 (en)
CN (5) CN101896186A (en)
AU (1) AU2008317566B2 (en)
CA (1) CA2704049A1 (en)
CY (2) CY1117286T1 (en)
DK (1) DK2203173T3 (en)
ES (5) ES2639852T3 (en)
HK (2) HK1185098A1 (en)
HR (1) HRP20160025T1 (en)
HU (2) HUE028662T2 (en)
IL (4) IL205322A (en)
NZ (2) NZ584793A (en)
PL (1) PL2203173T3 (en)
PT (1) PT2203173E (en)
WO (1) WO2009054725A2 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9890379B2 (en) 2006-08-11 2018-02-13 Biomarin Technologies B.V. Treatment of genetic disorders associated with DNA repeat instability
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
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

Families Citing this family (97)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1191097A1 (en) * 2000-09-21 2002-03-27 Leids Universitair Medisch Centrum Induction of exon skipping in eukaryotic cells
AU2003225410A1 (en) 2003-03-21 2004-10-11 Academisch Ziekenhuis Leiden Modulation of exon recognition in pre-mrna by interfering with the secondary rna structure
BRPI0409319B8 (en) 2003-04-11 2021-05-25 Ptc Therapeutics Inc compound, pharmaceutical composition, unit dosage form, and use of a compound
ATE498685T1 (en) 2004-06-28 2011-03-15 Univ Western Australia ANTISENSE OLIGONUCLEOTIDES FOR INDUCING EXON SKIPPING AND METHOD FOR THE 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
EP1855694B1 (en) 2005-02-09 2020-12-02 Sarepta Therapeutics, Inc. Antisense composition for treating muscle atrophy
WO2010050801A1 (en) 2008-10-27 2010-05-06 Prosensa Technologies B.V. Methods and means for efficient skipping of exon 45 in duchenne muscular dystrophy pre-mrna
US8084601B2 (en) * 2008-09-11 2011-12-27 Royal Holloway And Bedford New College Royal Holloway, University Of London Oligomers
CA3066050A1 (en) 2008-10-24 2010-04-29 Sarepta Therapeutics, Inc. Multiple exon skipping compositions for dmd
JP2012524540A (en) 2009-04-24 2012-10-18 プロセンサ テクノロジーズ ビー.ブイ. Oligonucleotides containing inosine for treating DMD
US20120270930A1 (en) 2009-10-29 2012-10-25 Academisch Ziekenhuis Leiden H.O.D.N. Lumc Methods and compositions for dysferlin exon-skipping
KR20230137491A (en) 2009-11-12 2023-10-04 더 유니버시티 오브 웨스턴 오스트레일리아 Antisense Molecules and Methods for Treating Pathologies
NZ607297A (en) 2010-08-05 2015-06-26 Academisch Ziekenhuis Leiden Antisense oligonucleotide directed removal of proteolytic cleavage sites from proteins
TWI541024B (en) 2010-09-01 2016-07-11 日本新藥股份有限公司 Antisense nucleic acid
JP2014507143A (en) * 2011-02-08 2014-03-27 ザ シャーロット−メクレンバーグ ホスピタル オーソリティ ドゥーイング/ビジネス/アズ キャロライナズ ヘルスケア システム Antisense oligonucleotide
AU2012240656A1 (en) 2011-04-05 2013-10-24 Academisch Ziekenhuis Leiden H.O.D.N. Lumc Compounds and methods for altering activin receptor-like kinase signalling
JP5850519B2 (en) 2011-05-09 2016-02-03 ネッパジーン株式会社 A therapeutic agent for muscular dystrophy containing morpholino-loaded bubble liposomes as active ingredients
US9765398B2 (en) 2011-07-28 2017-09-19 The Regents Of The University Of California Exonic splicing enhancers and exonic splicing silencers
US20130085139A1 (en) 2011-10-04 2013-04-04 Royal Holloway And Bedford New College Oligomers
RU2619184C2 (en) 2011-12-28 2017-05-12 Ниппон Синяку Ко., Лтд. 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
EP3597741A1 (en) 2012-04-27 2020-01-22 Duke University Genetic correction of mutated genes
CN104603271B (en) * 2012-07-03 2019-07-19 马林生物科技有限公司 For treating the oligonucleotides of muscular dystrophy patient
ES2762881T3 (en) 2013-03-14 2020-05-26 Sarepta Therapeutics Inc Exon skipping compositions for the treatment of muscular dystrophy
EP3662912A1 (en) * 2013-03-15 2020-06-10 Sarepta Therapeutics, Inc. Improved dosages of eteplirsen for treating duchenne muscular dystrophy
US9849066B2 (en) 2013-04-24 2017-12-26 Corning Incorporated Delamination resistant pharmaceutical glass containers containing active pharmaceutical ingredients
KR102551324B1 (en) * 2013-06-05 2023-07-05 듀크 유니버시티 Rna-guided gene editing and gene regulation
TWI695717B (en) 2014-03-06 2020-06-11 美商 Ptc 治療公司 Pharmaceutical compositions and salts of a 1,2,4-oxadiazole benzoic acid
NZ724836A (en) 2014-03-12 2022-07-01 Nippon Shinyaku Co Ltd Antisense nucleic acids
GB201410693D0 (en) 2014-06-16 2014-07-30 Univ Southampton Splicing modulation
JP6208349B2 (en) * 2014-06-17 2017-10-04 日本新薬株式会社 Antisense nucleic acid
CN106714845A (en) * 2014-08-11 2017-05-24 得克萨斯州大学***董事会 Prevention of muscular dystrophy by crispr/cas9-mediated gene editing
CA2963288A1 (en) 2014-10-03 2016-04-07 Cold Spring Harbor Laboratory Targeted augmentation of nuclear gene output
EP3256487A4 (en) 2015-02-09 2018-07-18 Duke University Compositions and methods for epigenome editing
MA41795A (en) 2015-03-18 2018-01-23 Sarepta Therapeutics Inc EXCLUSION OF AN EXON INDUCED BY ANTISENSE COMPOUNDS IN MYOSTATIN
US10370660B2 (en) * 2015-05-21 2019-08-06 Wings Therapeutics, Inc. Antisense oligonucleotides to treat Dystrophic Epidermolysis Bullosa
CA2988486A1 (en) * 2015-06-10 2016-12-15 Association Institut De Myologie Combined therapy for duchenne muscular dystrophy
WO2017015637A1 (en) 2015-07-22 2017-01-26 Duke University High-throughput screening of regulatory element function with epigenome editing technologies
JP6905755B2 (en) 2015-08-25 2021-07-21 デューク ユニバーシティ Compositions and Methods to Improve Specificity in Genomic Engineering Using RNA-Guided Endonucleases
KR101968880B1 (en) 2015-09-15 2019-04-12 니뽄 신야쿠 가부시키가이샤 Antisense nucleic acids
EA201890908A1 (en) 2015-10-09 2018-10-31 Сарепта Терапьютикс, Инк. COMPOSITIONS AND METHODS FOR THE TREATMENT OF MUSCULAR DYSTROPHY OF YOUNG AND SIMILAR DISORDERS
US10196639B2 (en) 2015-10-09 2019-02-05 University Of Southampton Modulation of gene expression and screening for deregulated protein expression
BR112018006636B1 (en) * 2015-10-09 2023-03-28 Wave Life Sciences Ltd OLIGONUCLEOTIDE COMPOSITION, PHARMACEUTICAL COMPOSITION AND USE OF THE OLIGONUCLEOTIDE COMPOSITION
EP4089175A1 (en) 2015-10-13 2022-11-16 Duke University Genome engineering with type i crispr systems in eukaryotic cells
CN108348527A (en) 2015-10-30 2018-07-31 Ptc医疗公司 Method for treating epilepsy
US11096956B2 (en) 2015-12-14 2021-08-24 Stoke Therapeutics, Inc. Antisense oligomers and uses thereof
CN109312343B (en) 2015-12-14 2022-09-27 冷泉港实验室 Antisense oligomers for the treatment of autosomal dominant mental retardation type 5 and Dravet syndrome
GB2545898B (en) 2015-12-21 2019-10-09 Sutura Therapeutics Ltd Improved drug delivery by conjugating oligonucleotides to stitched/stapled peptides
GB2574525B (en) * 2015-12-21 2020-09-02 Sutura Therapeutics Ltd Improved drug delivery by conjugating oligonucleotides to stitched/stapled peptides
MA45328A (en) 2016-04-01 2019-02-06 Avidity Biosciences Llc NUCLEIC ACID-POLYPEPTIDE COMPOSITIONS AND USES THEREOF
US11358993B2 (en) 2016-04-15 2022-06-14 Research Institute At Nationwide Children's Hospital Adeno-associated virus vector delivery of B-sarcoglycan and microrna-29 and the treatment of muscular dystrophy
MA45290A (en) * 2016-05-04 2019-03-13 Wave Life Sciences Ltd PROCESSES AND COMPOSITIONS OF BIOLOGICALLY ACTIVE AGENTS
US20190262375A1 (en) 2016-06-30 2019-08-29 Sarepta Therapeutics, Inc. Exon skipping oligomers for muscular dystrophy
SI3554554T1 (en) 2016-12-19 2023-02-28 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
HUE059905T2 (en) 2016-12-19 2023-01-28 Sarepta Therapeutics Inc Exon skipping oligomer conjugates for muscular dystrophy
WO2018118599A1 (en) 2016-12-19 2018-06-28 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
WO2018129384A1 (en) 2017-01-06 2018-07-12 Avidity Biosciences Llc Nucleic acid-polypeptide compositions and methods of inducing exon skipping
CA3056638A1 (en) * 2017-03-17 2018-09-20 Research Institute At Nationwide Children's Hospital Adeno-associated virus vector delivery of muscle specific micro-dystrophin to treat muscular dystrophy
CA3059321A1 (en) * 2017-04-20 2018-10-25 Synthena Ag Modified oligomeric compounds comprising tricyclo-dna nucleosides and uses thereof
GB201711809D0 (en) * 2017-07-21 2017-09-06 Governors Of The Univ Of Alberta Antisense oligonucleotide
KR102636383B1 (en) 2017-08-04 2024-02-14 스카이호크 테라퓨틱스, 인코포레이티드 Methods and compositions for regulating splicing
PL3673080T3 (en) 2017-08-25 2024-03-11 Stoke Therapeutics, Inc. Antisense oligomers for treatment of conditions and diseases
CN111770757A (en) * 2017-09-22 2020-10-13 艾维迪提生物科学公司 Nucleic acid-polypeptide compositions and methods for inducing exon skipping
EA201991450A1 (en) 2017-09-22 2019-12-30 Сарепта Терапьютикс, Инк. OLIGOMER CONJUGATES FOR EXONISM SKIP IN MUSCULAR DYSTROPHY
US20210145852A1 (en) 2017-09-28 2021-05-20 Sarepta Therapeutics, Inc. Combination Therapies for Treating Muscular Dystrophy
EP3687547A1 (en) 2017-09-28 2020-08-05 Sarepta Therapeutics, Inc. Combination therapies for treating muscular dystrophy
JP2020536057A (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
EP3720448A4 (en) 2017-12-06 2021-11-03 Avidity Biosciences, Inc. Compositions and methods of treating muscle atrophy and myotonic dystrophy
JP2021514010A (en) * 2018-02-20 2021-06-03 エッジワイズ セラピューティクス, インコーポレイテッド Methods and compositions for treating movement disorders
BR112020020670A2 (en) * 2018-04-12 2021-03-02 Wave Life Sciences Ltd. oligonucleotide composition, pharmaceutical composition, method of altering the splicing of a target transcript, method of treating muscular dystrophy, method of preparing an oligonucleotide or an oligonucleotide composition thereof and oligonucleotide
US10758629B2 (en) 2018-05-29 2020-09-01 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
TW202020153A (en) * 2018-07-27 2020-06-01 美商薩羅塔治療公司 Exon skipping oligomers for muscular dystrophy
US11168141B2 (en) 2018-08-02 2021-11-09 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating dystrophinopathies
JP2021533200A (en) 2018-08-02 2021-12-02 ダイン セラピューティクス, インコーポレーテッドDyne Therapeutics, Inc. Muscle-targeted complexes and their use for treating facial, scapular, and brachial muscular dystrophy
SG11202100934PA (en) 2018-08-02 2021-02-25 Dyne Therapeutics Inc Muscle targeting complexes and uses thereof for treating dystrophinopathies
CN109486813B (en) * 2018-10-10 2022-01-18 广州医科大学附属第二医院 U1-snRNA for repairing abnormal splicing of PremRNA of TPP1 gene and application thereof
AU2019392928A1 (en) * 2018-12-06 2021-06-17 Wave Life Sciences Ltd. Oligonucleotide compositions and methods thereof
KR20210135241A (en) 2019-02-05 2021-11-12 스카이호크 테라퓨틱스, 인코포레이티드 Methods and compositions for controlling splicing
KR20210135507A (en) 2019-02-06 2021-11-15 스카이호크 테라퓨틱스, 인코포레이티드 Methods and compositions for controlling splicing
EA202192767A1 (en) * 2019-04-10 2021-12-29 ПиТиСи ТЕРАПЬЮТИКС, ИНК. METHOD FOR THE TREATMENT OF NONSENSE MUTATION-MEDIATED DUSCHENNE MUSCULAR DYSTROPHY IN PEDIATRIC PATIENTS
CN110288555B (en) * 2019-07-02 2022-08-02 桂林电子科技大学 Low-illumination enhancement method based on improved capsule network
US20220282247A1 (en) * 2019-08-02 2022-09-08 Research Institute At Nationwide Children's Hospital Exon 44-targeted nucleic acids and recombinant adeno-associated virus comprising said nucleic acids for treatment of dystrophin-based myopathies
JP2022549611A (en) * 2019-09-19 2022-11-28 アルネー サイエンシズ、エルエルシー Compounds and methods useful for modulating gene splicing
JPWO2021172498A1 (en) 2020-02-28 2021-09-02
AU2021237465A1 (en) 2020-03-19 2022-10-13 Avidity Biosciences, Inc. Compositions and methods of treating Facioscapulohumeral muscular dystrophy
CN115867657A (en) 2020-05-11 2023-03-28 斯托克制药公司 OPA1 antisense oligomers for the treatment of disorders and diseases
EP4314016A1 (en) 2021-03-31 2024-02-07 Entrada Therapeutics, Inc. Cyclic cell penetrating peptides
IL308353A (en) 2021-05-10 2024-01-01 Entrada Therapeutics Inc Compositions and methods for modulating tissue distribution of intracellular therapeutics
EP4088722A1 (en) * 2021-05-12 2022-11-16 Justus-Liebig-Universität Gießen Pharmaceutical compositions, uses thereof and methods for treatment of genetic diseases caused by intronic splice-acceptor site mutations
WO2022271818A1 (en) 2021-06-23 2022-12-29 Entrada Therapeutics, Inc. Antisense compounds and methods for targeting cug repeats
WO2023283614A2 (en) * 2021-07-09 2023-01-12 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
US11771776B2 (en) 2021-07-09 2023-10-03 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating dystrophinopathies
CA3231330A1 (en) 2021-09-16 2023-03-23 Avidity Biosciences, Inc. Compositions and methods of treating facioscapulohumeral muscular dystrophy
EP4215614A1 (en) 2022-01-24 2023-07-26 Dynacure Combination therapy for dystrophin-related diseases
WO2023168427A1 (en) 2022-03-03 2023-09-07 Yale University Compositions and methods for delivering therapeutic polynucleotides for exon skipping
WO2024069229A2 (en) 2022-08-03 2024-04-04 Sutura Therapeutics Ltd Biologically active compounds

Family Cites Families (220)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE2752581C2 (en) 1977-11-25 1979-12-13 Wolfgang 4044 Kaarst Keil Device for the complete demineralisation of water
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
DE3834636A1 (en) 1988-10-11 1990-04-19 Max Planck Gesellschaft METHOD FOR ANALYZING LENGTH POLYMORPHISMS IN DNA AREAS
US6867195B1 (en) 1989-03-21 2005-03-15 Vical Incorporated Lipid-mediated polynucleotide administration to reduce likelihood of subject's becoming infected
US5681941A (en) 1990-01-11 1997-10-28 Isis Pharmaceuticals, Inc. Substituted purines and oligonucleotide cross-linking
US5608046A (en) 1990-07-27 1997-03-04 Isis Pharmaceuticals, Inc. Conjugated 4'-desmethyl nucleoside analog compounds
FR2675803B1 (en) 1991-04-25 1996-09-06 Genset Sa CLOSED, ANTISENSE AND SENSE OLIGONUCLEOTIDES AND THEIR APPLICATIONS.
AU659482B2 (en) 1991-06-28 1995-05-18 Massachusetts Institute Of Technology Localized oligonucleotide therapy
EP0558697A1 (en) 1991-06-28 1993-09-08 Massachusetts Institute Of Technology Localized oligonucleotide therapy
US6200747B1 (en) 1992-01-28 2001-03-13 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
CA2116280A1 (en) 1993-03-05 1994-09-06 Marcy E. Macdonald Huntingtin dna, protein and uses thereof
AU6786594A (en) 1993-05-11 1994-12-12 University Of North Carolina At Chapel Hill, The Antisense oligonucleotides which combat aberrant splicing and methods of using the same
US5695933A (en) 1993-05-28 1997-12-09 Massachusetts Institute Of Technology Direct detection of expanded nucleotide repeats in the human genome
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
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
US5627263A (en) 1993-11-24 1997-05-06 La Jolla Cancer Research Foundation Integrin-binding peptides
DE4342605A1 (en) 1993-12-14 1995-06-22 Buna Gmbh Functionalized olefin homo- and copolymers
US5962332A (en) 1994-03-17 1999-10-05 University Of Massachusetts Detection of trinucleotide repeats by in situ hybridization
WO1995030774A1 (en) 1994-05-05 1995-11-16 Beckman Instruments, Inc. Oligonucleotide repeat arrays
US5968909A (en) 1995-08-04 1999-10-19 Hybridon, Inc. Method of modulating gene expression with reduced immunostimulatory response
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
US20070173465A9 (en) 1995-10-11 2007-07-26 Monahan Sean D Expression of zeta negative and zeta positive nucleic acids using a dystrophin gene
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)
US6300060B1 (en) 1995-11-09 2001-10-09 Dana-Farber Cancer Institute, Inc. Method for predicting the risk of prostate cancer morbidity and mortality
ATE267207T1 (en) 1996-02-14 2004-06-15 Isis Pharmaceuticals Inc CARBOHYDRATE MODIFIED LATCH OLIGONUCLEOTIDES
US6251589B1 (en) 1996-07-18 2001-06-26 Srl, Inc. Method for diagnosing spinocerebellar ataxia type 2 and primers therefor
EP0878543A1 (en) 1996-10-30 1998-11-18 Srl, Inc. cDNA FRAGMENTS OF GENE CAUSATIVE OF SPINOCEREBELLAR ATAXIA TYPE 2
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
US20020137890A1 (en) 1997-03-31 2002-09-26 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
WO1998043993A2 (en) 1997-03-31 1998-10-08 Yale University Nucleic acid catalysts
WO1998049345A1 (en) 1997-04-29 1998-11-05 Trustees Of Boston University Methods and compositions for targeted dna differential display
US6329501B1 (en) 1997-05-29 2001-12-11 Auburn University Methods and compositions for targeting compounds to muscle
US6280938B1 (en) 1997-08-19 2001-08-28 Regents Of The University Of Minnesota SCA7 gene and method of use
US6514755B1 (en) 1998-08-18 2003-02-04 Regents Of The University Of Minnesota SCA7 gene and methods of use
US6794499B2 (en) 1997-09-12 2004-09-21 Exiqon A/S Oligonucleotide analogues
US6127173A (en) 1997-09-22 2000-10-03 Ribozyme Pharmaceuticals, Inc. Nucleic acid catalysts with endonuclease activity
US6130207A (en) 1997-11-05 2000-10-10 South Alabama Medical Science Foundation Cell-specific molecule and method for importing DNA into a nucleus
JP3012923B2 (en) 1998-01-26 2000-02-28 新潟大学長 Drug for treating CAG repeat disease
KR100280219B1 (en) 1998-02-26 2001-04-02 이수빈 Diagnostic Method and Diagnostic Reagent of Neuropsychiatric Disease Using Trinucleic Acid Repeat Sequence
US6322978B1 (en) 1998-04-20 2001-11-27 Joslin Diabetes Center, Inc. Repeat polymorphism in the frataxin gene and uses therefore
WO1999055857A2 (en) 1998-04-29 1999-11-04 Ribozyme Pharmaceuticals, Inc. Nucleoside triphosphates and their incorporation into ribozymes
ES2226414T3 (en) 1998-06-10 2005-03-16 Biognostik Gesellschaft Fur Biomolekulare Diagnostik Mbh STIMULATION OF THE IMMUNE SYSTEM
US6924355B2 (en) 1998-09-01 2005-08-02 Genentech, Inc. PRO1343 polypeptides
AU6059099A (en) 1998-09-25 2000-04-17 Children's Medical Center Corporation Short peptides which selectively modulate the activity of protein kinases
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
JP4777515B2 (en) * 1998-10-26 2011-09-21 エイブイアイ バイオファーマ, インコーポレイテッド Antisense based on p53 morpholino
US6399575B1 (en) 1998-11-10 2002-06-04 Auburn University Methods and compositions for targeting compounds to the central nervous system
US6133031A (en) 1999-08-19 2000-10-17 Isis Pharmaceuticals Inc. Antisense inhibition of focal adhesion kinase expression
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
JP2002541825A (en) 1999-04-08 2002-12-10 オアシス バイオサイエンシーズ インコーポレイティッド Antisense oligonucleotides containing universal and / or synonymous bases
JP2000325085A (en) 1999-05-21 2000-11-28 Masafumi Matsuo 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
US6656730B1 (en) 1999-06-15 2003-12-02 Isis Pharmaceuticals, Inc. Oligonucleotides conjugated to protein-binding drugs
US6355481B1 (en) 1999-06-18 2002-03-12 Emory University Hybridoma cell line and monoclonal antibody for huntingtin protein
JP2004512810A (en) 1999-08-31 2004-04-30 サーナ・セラピューティクス・インコーポレイテッド Modulators of gene expression based on nucleic acids
AU7856600A (en) 1999-10-04 2001-05-10 University Of Medicine And Dentistry Of New Jersey Novel carbamates and ureas
US6165786A (en) 1999-11-03 2000-12-26 Isis Pharmaceuticals, Inc. Antisense modulation of nucleolin expression
EP1257639A2 (en) 2000-02-08 2002-11-20 Ribozyme Pharmaceuticals, Inc. Nucleozymes with endonuclease activity
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
AU4819101A (en) 2000-04-13 2001-10-30 University Of British Columbia, The Modulating cell survival by modulating huntingtin function
US6653467B1 (en) * 2000-04-26 2003-11-25 Jcr Pharmaceutical Co., Ltd. Medicament for treatment of Duchenne muscular dystrophy
WO2001083695A2 (en) 2000-04-28 2001-11-08 Xiao Xiao Dna sequences encoding dystrophin minigenes and methods of use thereof
AU2001257366B2 (en) 2000-05-01 2006-03-16 Idera Pharmaceuticals, Inc. Modulation of oligonucleotide CpG-mediated immune stimulation by positional modification of nucleosides
WO2001085751A1 (en) 2000-05-09 2001-11-15 Reliable Biopharmaceutical, Inc. Polymeric compounds useful as prodrugs
DE60141827D1 (en) 2000-05-31 2010-05-27 Serono Genetics Inst Sa USE OF THE GLOBULAR HEAD OF ACRP30 TO INCREASE MUSCLE MASS AND DIFFERENTIATION INCREASE
CN1326990A (en) 2000-06-07 2001-12-19 上海博德基因开发有限公司 New polypeptide-human DNA-like CGG repeative conjugated protein 16.17 and polynucleotide for encoding such polypeptide
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
RU2165149C1 (en) 2000-07-03 2001-04-20 Шапошников Валерий Геннадьевич "sugar wool" products forming and packaging method
JP4836366B2 (en) 2000-08-25 2011-12-14 雅文 松尾 Duchenne muscular dystrophy treatment
US6727355B2 (en) * 2000-08-25 2004-04-27 Jcr Pharmaceuticals Co., Ltd. Pharmaceutical composition for treatment of Duchenne muscular dystrophy
EP1191097A1 (en) 2000-09-21 2002-03-27 Leids Universitair Medisch Centrum Induction of exon skipping in eukaryotic cells
AU3922802A (en) 2000-10-02 2002-05-27 Keck Graduate Inst Methods for identifying nucleotides at defined positions in target nucleic acidsusing fluorescence polarization
DE60134786D1 (en) 2000-10-06 2008-08-21 Univ Michigan MINI-DYSTROPHINE NUCLEIC ACID AND PEPTIDE SEQUENCES
US6623927B1 (en) 2000-11-08 2003-09-23 Council Of Scientific And Industrial Research Method of detection of allelic variants of SCA2 gene
CA2449412C (en) 2000-11-30 2013-04-02 Uab Research Foundation Receptor-mediated uptake of peptides that bind the human transferrin receptor
US7001994B2 (en) 2001-01-18 2006-02-21 Genzyme Corporation Methods for introducing mannose 6-phosphate and other oligosaccharides onto glycoproteins
TWI329129B (en) 2001-02-08 2010-08-21 Wyeth Corp Modified and stabilized gdf propeptides and uses thereof
KR100408053B1 (en) 2001-02-13 2003-12-01 엘지전자 주식회사 Torque ripple reduction method for srm
WO2002085308A2 (en) * 2001-04-24 2002-10-31 Epigenesis Pharmaceuticals, Inc. Antisense and anti-inflammatory based compositions to treat respiratory disorders
US6902896B2 (en) 2001-05-11 2005-06-07 Regents Of The University Of Minnesota Intron associated with myotonic dystrophy type 2 and methods of use
US20050277133A1 (en) 2001-05-18 2005-12-15 Sirna Therapeutics, Inc. RNA interference mediated treatment of polyglutamine (polyQ) repeat expansion diseases using short interfering nucleic acid (siNA)
AU2004266311B2 (en) 2001-05-18 2009-07-23 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US20050014172A1 (en) 2002-02-20 2005-01-20 Ivan Richards RNA interference mediated inhibition of muscarinic cholinergic receptor gene expression using short interfering nucleic acid (siNA)
US20050282188A1 (en) 2001-05-18 2005-12-22 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using short interfering nucleic acid (siNA)
IL143379A (en) 2001-05-24 2013-11-28 Yissum Res Dev Co Antisense oligonucleotide against the r isophorm of human ache and uses thereof
US20030139435A1 (en) 2001-06-26 2003-07-24 Gulzar Ahmed N-heterocyclic inhibitors of TNF-alpha expression
NZ530360A (en) 2001-07-06 2006-01-27 Topigen Pharma Inc Methods for increasing in vivo efficacy of oligonucleotides and inhibiting inflammation in mammals by substituting an adenosine base for 2'6'-diaminopurine (DAP) and analogs thereof
WO2003013437A2 (en) 2001-08-07 2003-02-20 University Of Delaware Compositions and methods for the prevention and treatment of huntington's disease
CA2455336A1 (en) 2001-08-10 2003-02-20 Novartis Ag Peptides that bind to atherosclerotic lesions
US20060074034A1 (en) 2001-09-17 2006-04-06 Collins Douglas A Cobalamin mediated delivery of nucleic acids, analogs and derivatives thereof
KR20030035047A (en) 2001-10-29 2003-05-09 (주)바이오코돈 Use of BMP-4 gene and its gene product for treatment and diagnosis of Lichen Planus
WO2003037172A2 (en) 2001-11-01 2003-05-08 Gpc Biotech Inc. Endothelial-cell binding peptides for diagnosis and therapy
US20030134790A1 (en) 2002-01-11 2003-07-17 University Of Medicine And Dentistry Of New Jersey Bone Morphogenetic Protein-2 And Bone Morphogenetic Protein-4 In The Treatment And Diagnosis Of Cancer
US20030203356A1 (en) 2002-01-22 2003-10-30 The Cleveland Clinic Foundation RNase L activator-antisense complexes
WO2003069330A1 (en) 2002-02-11 2003-08-21 The Trustees Of Columbia University In The City Of New York System and method for identifying proteins involved in force-initiated signal transduction
US20050096284A1 (en) 2002-02-20 2005-05-05 Sirna Therapeutics, Inc. RNA interference mediated treatment of polyglutamine (polyQ) repeat expansion diseases using short interfering nucleic acid (siNA)
ES2339934T3 (en) 2002-03-01 2010-05-27 The Administrators Of The Tulane Educational Fund CONJUGATES OF CITOTOXIC AGENTS AND BIOLOGICALLY ACTIVE PEPTIDES.
US20040101852A1 (en) 2002-11-21 2004-05-27 Isis Pharmaceuticals Inc. Modulation of CGG triplet repeat binding protein 1 expression
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.
US20040102395A1 (en) 2002-11-22 2004-05-27 Isis Pharmaceuticals Inc. Modulation of IAP-like expression
EP1380644A1 (en) 2002-07-08 2004-01-14 Kylix B.V. The use of specified TCF target genes to identify drugs for the treatment of cancer, in particular colorectal cancer, in which TCF/beta-catenin/WNT signalling plays a central role
US7589059B2 (en) 2002-07-26 2009-09-15 Roche Madison Inc. Delivery of molecules and complexes to mammalian cells in vivo
US20050255086A1 (en) 2002-08-05 2005-11-17 Davidson Beverly L Nucleic acid silencing of Huntington's Disease gene
EP1529105B1 (en) 2002-08-12 2012-10-03 Société de commercialisation des produits de la recherche appliquée SOCPRA Sciences Santé et Humaines s.e.c. Methods to reprogram splice site selection in pre-messenger rnas
GB0219143D0 (en) 2002-08-16 2002-09-25 Univ Leicester Modified tailed oligonucleotides
US20040219565A1 (en) 2002-10-21 2004-11-04 Sakari Kauppinen Oligonucleotides useful for detecting and analyzing nucleic acids of interest
DE60219215T2 (en) 2002-10-23 2008-01-03 Centre For Research And Technology Hellas/Intitute Of Agrobiotechnology In.A, Thermi PRION-BINDING PEPTIDE SEQUENCES
US7892793B2 (en) 2002-11-04 2011-02-22 University Of Massachusetts Allele-specific RNA interference
US7655785B1 (en) 2002-11-14 2010-02-02 Rosetta Genomics Ltd. Bioinformatically detectable group of novel regulatory oligonucleotides and uses thereof
EP2305812A3 (en) 2002-11-14 2012-06-06 Dharmacon, Inc. Fuctional and hyperfunctional sirna
US7250496B2 (en) * 2002-11-14 2007-07-31 Rosetta Genomics Ltd. Bioinformatically detectable group of novel regulatory genes and uses thereof
ES2566629T3 (en) 2002-11-25 2016-04-14 Masafumi Matsuo ENA nucleic acid drugs that modify splicing in mRNA precursors
GB0228079D0 (en) 2002-12-02 2003-01-08 Laxdale Ltd Huntington's Disease
ATE479752T1 (en) 2003-03-07 2010-09-15 Alnylam Pharmaceuticals Inc THERAPEUTIC COMPOSITIONS
AU2003225410A1 (en) 2003-03-21 2004-10-11 Academisch Ziekenhuis Leiden Modulation of exon recognition in pre-mrna by interfering with the secondary rna structure
US7514551B2 (en) 2003-04-03 2009-04-07 Enzo Life Sciences, Inc. Multisignal labeling reagents, and processes and uses therefor
WO2004101787A1 (en) 2003-05-14 2004-11-25 Japan Science And Technology Agency Inhibition of the expression of huntington gene
JP5110877B2 (en) 2003-06-02 2012-12-26 ワイス・エルエルシー Use of myostatin (GDF8) inhibitors in combination with corticosteroids to treat neuromuscular disorders
DK1495769T3 (en) 2003-07-11 2008-06-23 Lbr Medbiotech B V Mannose-6-phosphate receptor-mediated gene transfer to muscle cells
US20050048495A1 (en) 2003-08-29 2005-03-03 Baker Brenda F. Isoform-specific targeting of splice variants
US20050054752A1 (en) 2003-09-08 2005-03-10 O'brien John P. Peptide-based diblock and triblock dispersants and diblock polymers
WO2005033134A2 (en) 2003-09-30 2005-04-14 Regeneron Pharmaceuticals, Inc. Secreted protein therapeutics and uses thereof
EP1680439A2 (en) 2003-10-14 2006-07-19 Kernel Biopharma Inc. Dual phase-pna conjugates for the delivery of pna through the blood brain barrier
US20050191636A1 (en) 2004-03-01 2005-09-01 Biocept, Inc. Detection of STRP, such as fragile X syndrome
US20080207538A1 (en) 2004-03-11 2008-08-28 Lawrence David S Enhanced Production of Functional Proteins From Defective Genes
WO2005105995A2 (en) 2004-04-14 2005-11-10 Sirna Therapeutics, Inc. RNA INTERFERENCE MEDIATED TREATMENT OF POLYGLUTAMINE (POLYQ) REPEAT EXPANSION DISEASES USING SHORT INTERFERING NUCLEIC ACID (siNA)
EP1752536A4 (en) 2004-05-11 2008-04-16 Alphagen Co Ltd Polynucleotide causing rna interfere and method of regulating gene expression with the use of the same
US20050288246A1 (en) 2004-05-24 2005-12-29 Iversen Patrick L Peptide conjugated, inosine-substituted antisense oligomer compound and method
EP1755648A2 (en) 2004-05-27 2007-02-28 Acceleron Pharma Inc. Cerberus/coco derivatives and uses thereof
ATE498685T1 (en) 2004-06-28 2011-03-15 Univ Western Australia ANTISENSE OLIGONUCLEOTIDES FOR INDUCING EXON SKIPPING AND METHOD FOR THE USE THEREOF
US7361468B2 (en) 2004-07-02 2008-04-22 Affymetrix, Inc. Methods for genotyping polymorphisms in humans
EP1618881A1 (en) * 2004-07-20 2006-01-25 Santhera Pharmaceuticals (Schweiz) GmbH Use of non-glucocorticoid steroids for the treatment of muscular dystrophy
WO2006017522A2 (en) 2004-08-03 2006-02-16 University Of Utah Research Foundation Use of antisense oligonucleotides to effect translation modulation
US20060099612A1 (en) * 2004-09-02 2006-05-11 Suntory Limited Method for analyzing genes of industrial yeasts
CA2582137A1 (en) * 2004-10-05 2007-02-15 Wyeth Probe arrays for detecting multiple strains of different species
ITRM20040568A1 (en) 2004-11-18 2005-02-18 Uni Degli Studi Di Roma Tor Vergata USE OF THE "PHAGE DISPLAY" TECHNIQUE FOR THE IDENTIFICATION OF PEPTIDES WITH CAPACITY OF STAMIN CELLS / PROGENITOR, PEPTIDES SO OBTAINED AND THEIR USES.
US7838657B2 (en) 2004-12-03 2010-11-23 University Of Massachusetts Spinal muscular atrophy (SMA) treatment via targeting of SMN2 splice site inhibitory sequences
KR100663277B1 (en) 2004-12-20 2007-01-02 삼성전자주식회사 Device and?method for processing system-related event in wireless terminal
US20060148740A1 (en) 2005-01-05 2006-07-06 Prosensa B.V. Mannose-6-phosphate receptor mediated gene transfer into muscle cells
US20120122801A1 (en) 2005-01-05 2012-05-17 Prosensa B.V. Mannose-6-phosphate receptor mediated gene transfer into muscle cells
EP1844148A2 (en) 2005-01-31 2007-10-17 University of Iowa Research Foundation Nucleic acid silencing of huntington's disease gene
EP1877099B1 (en) 2005-04-06 2012-09-19 Genzyme Corporation Therapeutic conjugates comprising a lysosomal enzyme, polysialic acid and a targeting moiety
WO2006112705A2 (en) 2005-04-22 2006-10-26 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.
US7902352B2 (en) 2005-05-06 2011-03-08 Medtronic, Inc. Isolated nucleic acid duplex for reducing huntington gene expression
US20060257912A1 (en) 2005-05-06 2006-11-16 Medtronic, Inc. Methods and sequences to suppress primate huntington gene expression
WO2006121277A1 (en) 2005-05-09 2006-11-16 Korea Research Institute Of Standards And Science Fret probes for fluorescent detection of the epsps gene
PL2548560T3 (en) 2005-06-23 2015-11-30 Biogen Ma Inc Compositions and methods for modulation of SMN2 splicing
WO2007002904A2 (en) 2005-06-28 2007-01-04 Medtronic, Inc. Methods and sequences to preferentially suppress expression of mutated huntingtin
JP5208734B2 (en) * 2005-07-01 2013-06-12 インデックス・ファーマシューティカルズ・アクチエボラーグ How to modulate responsiveness to steroids
CN101365801B (en) 2005-10-28 2013-03-27 阿尔尼拉姆医药品有限公司 Compositions and methods for inhibiting expression of huntingtin gene
US7906617B2 (en) 2005-12-15 2011-03-15 E. I. Du Pont De Nemours And Company Polyethylene binding peptides and methods of use
AU2007210038B2 (en) 2006-01-26 2013-01-10 Ionis Pharmaceuticals, Inc. Compositions and their uses directed to huntingtin
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
US7855053B2 (en) 2006-07-19 2010-12-21 The Regents Of The University Of California Methods for detecting the presence of expanded CGG repeats in the FMR1 gene 5′ untranslated region
PL2049664T3 (en) 2006-08-11 2012-02-29 Biomarin Tech Bv Single stranded oligonucleotides complementary to repetitive elements for treating DNA repeat instability associated genetic disorders
CN101501193B (en) 2006-08-11 2013-07-03 普罗森那技术公司 Methods and means for treating DNA repeat instability associated genetic disorders
US20090253744A1 (en) 2006-09-27 2009-10-08 Bakshi Raman K Acylated piperidine derivatives as melanocortin-4 receptor modulators
JP2010505897A (en) 2006-10-11 2010-02-25 マックス−プランク−ゲゼルシャフト・ツア・フェルデルング・デア・ヴィッセンシャフテン・エー・ファオ Influenza target
EP2125032A4 (en) 2007-02-20 2011-02-23 Mayo Foundation Treating cancer with viral nucleic acid
PL381824A1 (en) 2007-02-22 2008-09-01 Instytut Chemii Bioorganicznej Pan W Poznaniu The sequence of siRNA, vector, molecular target for siRNA reagents and vectors introduced to cells and tissues, the manner of assessment of specifity of silencing of a mutated transcript, the manner of testing of influences of RNA interference route of enz
WO2009005793A2 (en) 2007-06-29 2009-01-08 Avi Biopharma, Inc. Tissue specific peptide conjugates and methods
CN101815535B (en) 2007-07-12 2013-04-24 普罗森那技术公司 Molecules for targeting compounds to various selected organs or tissues
EP2167135A2 (en) 2007-07-12 2010-03-31 Prosensa Technologies B.V. Molecules for targeting compounds to various selected organs, tissues or tumor cells
US9212205B2 (en) 2007-07-26 2015-12-15 University Of Rochester Nucleic acid binding compounds and methods of use
AU2008286771B2 (en) 2007-08-15 2013-08-15 Isis Pharmaceuticals, Inc. Tetrahydropyran nucleic acid analogs
CN101896186A (en) 2007-10-26 2010-11-24 莱顿教学医院 The mode and the method for antagonism disorder of muscle
WO2010050801A1 (en) 2008-10-27 2010-05-06 Prosensa Technologies B.V. Methods and means for efficient skipping of exon 45 in duchenne muscular dystrophy pre-mrna
WO2009099326A1 (en) 2008-02-08 2009-08-13 Prosensa Holding Bv Methods and means for treating dna repeat instability associated genetic disorders
WO2009101399A1 (en) 2008-02-12 2009-08-20 Isis Innovation Limited Treatment of muscular dystrophy using peptide nucleic acid ( pna)
EP2105145A1 (en) 2008-03-27 2009-09-30 ETH Zürich Method for muscle-specific delivery lipid-conjugated oligonucleotides
WO2009135322A1 (en) 2008-05-09 2009-11-12 The Universtity Of British Columbia Methods and compositions for the treatment of huntington's disease
EP2119783A1 (en) 2008-05-14 2009-11-18 Prosensa Technologies B.V. Method for efficient exon (44) skipping in Duchenne Muscular Dystrophy and associated means
US20110130346A1 (en) 2008-05-30 2011-06-02 Isis Innovation Limited Peptide conjugates for delvery of biologically active compounds
US20110152352A1 (en) 2008-06-10 2011-06-23 Tufts University Smad proteins control drosha-mediated mirna maturation
GB2457965B8 (en) 2008-07-01 2011-02-16 Renovo Ltd Methods and systems for determining efficacy of medicaments.
WO2010006237A2 (en) 2008-07-11 2010-01-14 Alnylam Pharmaceuticals, Inc. Phosphorothioate oligonucleotides non-nucleosidic phosphorothiotes as delivery agents for irna agents
CA2732343C (en) 2008-07-29 2017-05-09 The Board Of Regents Of The University Of Texas System Selective inhibition of polyglutamine protein expression
US8084601B2 (en) * 2008-09-11 2011-12-27 Royal Holloway And Bedford New College Royal Holloway, University Of London Oligomers
CA2777486A1 (en) 2008-10-15 2010-04-22 4S3 Bioscience Inc. Methods and compositions for treatment of myotonic dystrophy
CA3066050A1 (en) 2008-10-24 2010-04-29 Sarepta Therapeutics, Inc. Multiple exon skipping compositions for dmd
US20100248239A1 (en) 2009-03-24 2010-09-30 Mayo Foundation For Medical Education And Research Methods and materials for detecting fragile x mutations
EP2411532B1 (en) 2009-03-24 2017-04-05 Asuragen, Inc. Pcr methods for characterizing the 5' untranslated region of the fmr1 and fmr2 genes
ES2573981T3 (en) 2009-04-10 2016-06-13 Association Institut De Myologie Tricycle-DNA antisense oligonucleotides, compositions, and methods for the treatment of diseases
JP2012524540A (en) 2009-04-24 2012-10-18 プロセンサ テクノロジーズ ビー.ブイ. Oligonucleotides containing inosine for treating DMD
UA105390C2 (en) 2009-06-08 2014-05-12 Міраджен Терапьютікс Normal;heading 1;heading 2;heading 3;CHEMICAL MODIFICATION MOTIFS FOR MIRNA INHIBITORS AND MIMETICS
ES2772825T3 (en) 2009-09-11 2020-07-08 Ionis Pharmaceuticals Inc Modulation of huntingtin expression
KR20230137491A (en) 2009-11-12 2023-10-04 더 유니버시티 오브 웨스턴 오스트레일리아 Antisense Molecules and Methods for Treating Pathologies
US20110166081A1 (en) 2009-12-03 2011-07-07 University Of Iowa Research Foundation Alpha-dystroglycan as a Protein Therapeutic
WO2011078797A2 (en) 2009-12-22 2011-06-30 Singapore Health Services Pte. Ltd Antisense oligonucleotides and uses threreof
ES2616561T3 (en) 2009-12-24 2017-06-13 Biomarin Technologies B.V. Molecule to treat an inflammatory disorder
EP2536738A4 (en) 2010-02-08 2014-09-17 Isis Pharmaceuticals Inc Methods and compositions useful in treatment of diseases or conditions related to repeat expansion
WO2011097614A1 (en) 2010-02-08 2011-08-11 Isis Pharmaceuticals, Inc. Mehods and compositions useful in diseases or conditions related to repeat expansion
DK3031920T3 (en) 2010-07-19 2019-10-14 Ionis Pharmaceuticals Inc MODULATION OF DYSTROPHY MYOTONICA-PROTEIN KINASE (DMPK) EXPRESSION
US8513211B2 (en) 2010-08-20 2013-08-20 Replicor Inc. Oligonucleotide chelate complexes
TWI541024B (en) 2010-09-01 2016-07-11 日本新藥股份有限公司 Antisense nucleic acid
JP2014507143A (en) 2011-02-08 2014-03-27 ザ シャーロット−メクレンバーグ ホスピタル オーソリティ ドゥーイング/ビジネス/アズ キャロライナズ ヘルスケア システム Antisense oligonucleotide
EP2673361B1 (en) 2011-02-08 2016-04-13 Ionis Pharmaceuticals, Inc. Oligomeric compounds comprising bicyclic nucleotides and uses thereof
CN107267517A (en) 2011-04-22 2017-10-20 比奥马林技术公司 Noval chemical compound for treating, delaying and/or preventing human genetic disease's such as type of steirert-Batten-Gibb syndrome 1
CN103619356B (en) 2011-05-05 2017-09-12 萨勒普塔医疗公司 Peptide oligonucleotide conjugates
AU2012272656A1 (en) 2011-06-23 2014-02-06 Cold Spring Harbor Laboratory Phenocopy model of disease
EP3858847A1 (en) 2011-11-30 2021-08-04 Sarepta Therapeutics, Inc. Oligonucleotides for treating expanded repeat diseases
WO2013082578A1 (en) 2011-12-03 2013-06-06 Thomas Krupenkin Method and apparatus for mechanical energy harvesting using combined magnetic and microfluidic energy generation
EP2790736B1 (en) 2011-12-12 2018-01-31 Oncoimmunin, Inc. In vivo delivery of oligonucleotides
RU2619184C2 (en) 2011-12-28 2017-05-12 Ниппон Синяку Ко., Лтд. Antisense nucleic acids
EP2806900B1 (en) 2012-01-27 2021-12-15 BioMarin Technologies B.V. Rna modulating oligonucleotides with improved characteristics for the treatment of duchenne and becker muscular dystrophy
EP3330278A1 (en) 2012-02-08 2018-06-06 Ionis Pharmaceuticals, Inc. Modulation of rna by repeat targeting
CA2870697C (en) 2012-04-23 2021-11-23 Prosensa Technologies B.V. Rna modulating oligonucleotides with improved characteristics for the treatment of neuromuscular disorders
AR091065A1 (en) 2012-05-18 2014-12-30 Replicor Inc A PHARMACEUTICAL FORMULATION THAT INCLUDES AN ANTIVIRAL OLIGONUCLEOTIDE CHELATE FOR THE TREATMENT OF AN ANTI-VIRAL INFECTION
CN104603271B (en) 2012-07-03 2019-07-19 马林生物科技有限公司 For treating the oligonucleotides of muscular dystrophy patient
PL2970964T3 (en) * 2013-03-14 2019-06-28 Sarepta Therapeutics, Inc. Exon skipping compositions for treating muscular dystrophy

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9890379B2 (en) 2006-08-11 2018-02-13 Biomarin Technologies B.V. Treatment of genetic disorders associated with DNA repeat instability
US10689646B2 (en) 2006-08-11 2020-06-23 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
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
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
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
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
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

Also Published As

Publication number Publication date
US20150166996A1 (en) 2015-06-18
PL2203173T3 (en) 2016-06-30
EP3238737B1 (en) 2022-04-06
ES2692886T3 (en) 2018-12-05
IL241928A0 (en) 2015-11-30
EP4183399A1 (en) 2023-05-24
JP2011502118A (en) 2011-01-20
CN102264903B (en) 2015-01-07
ES2914775T3 (en) 2022-06-16
EP2614827B1 (en) 2017-06-28
IL205322A0 (en) 2010-12-30
CN101896186A (en) 2010-11-24
JP7107622B2 (en) 2022-07-27
CN105641700B (en) 2021-01-01
JP6579629B2 (en) 2019-09-25
IL284321A (en) 2021-07-29
HUE028662T2 (en) 2016-12-28
US20210139904A1 (en) 2021-05-13
CN102256606A (en) 2011-11-23
HK1245670A1 (en) 2018-08-31
HK1185098A1 (en) 2014-02-07
US20200239886A1 (en) 2020-07-30
ES2639852T3 (en) 2017-10-30
US20190119679A1 (en) 2019-04-25
JP5879374B2 (en) 2016-03-08
US20170107512A1 (en) 2017-04-20
US9243245B2 (en) 2016-01-26
US20160304864A1 (en) 2016-10-20
CN105647921A (en) 2016-06-08
US9499818B2 (en) 2016-11-22
IL205322A (en) 2016-08-31
AU2008317566A1 (en) 2009-04-30
US20140221458A1 (en) 2014-08-07
PT2203173E (en) 2016-03-15
WO2009054725A3 (en) 2009-10-15
US20150073037A1 (en) 2015-03-12
JP2016033140A (en) 2016-03-10
JP2019142942A (en) 2019-08-29
US20190112604A1 (en) 2019-04-18
IL284321B (en) 2022-04-01
CY1117286T1 (en) 2017-04-26
US20140128592A1 (en) 2014-05-08
HUE027124T2 (en) 2016-08-29
IL261127A (en) 2018-10-31
AU2008317566B2 (en) 2014-05-01
ES2564563T3 (en) 2016-03-23
IL241928B (en) 2018-08-30
JP2017141296A (en) 2017-08-17
CN105641700A (en) 2016-06-08
JP2014111638A (en) 2014-06-19
NZ584793A (en) 2012-05-25
IL261127B (en) 2020-07-30
US20140113955A1 (en) 2014-04-24
JP6885620B2 (en) 2021-06-16
EP2203173A2 (en) 2010-07-07
US11427820B2 (en) 2022-08-30
US20110263682A1 (en) 2011-10-27
US20110294753A1 (en) 2011-12-01
US10876114B2 (en) 2020-12-29
CN102264903A (en) 2011-11-30
EP2614827A3 (en) 2013-09-25
EP3238737A1 (en) 2017-11-01
WO2009054725A2 (en) 2009-04-30
CY1117454T1 (en) 2017-04-26
DK2203173T3 (en) 2016-02-29
US20120022134A1 (en) 2012-01-26
US9528109B2 (en) 2016-12-27
CA2704049A1 (en) 2009-04-30
EP2203173B1 (en) 2015-12-23
US20190100754A1 (en) 2019-04-04
JP2021113229A (en) 2021-08-05
US20230151362A1 (en) 2023-05-18
US9926557B2 (en) 2018-03-27
US20190177725A1 (en) 2019-06-13
NZ592446A (en) 2012-03-30
EP2614827A2 (en) 2013-07-17
JP5600064B2 (en) 2014-10-01
HRP20160025T1 (en) 2016-02-12
ES2936464T3 (en) 2023-03-17

Similar Documents

Publication Publication Date Title
US10876114B2 (en) 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
EP3400948B1 (en) Methods and means for efficient skipping of at least exon 52 of the human duchenne muscular dystrophy gene
AU2009310558B8 (en) 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.

Legal Events

Date Code Title Description
AS Assignment

Owner name: BIOMARIN TECHNOLOGIES B.V., NETHERLANDS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:VAN DEUTEKOM, JUDITH CHRISTINA THEODORA;REEL/FRAME:040320/0916

Effective date: 20160809

Owner name: ACADEMISCH ZIEKENHUIS LEIDEN (AZL), NETHERLANDS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:VAN DEUTEKOM, JUDITH CHRISTINA THEODORA;REEL/FRAME:040320/0916

Effective date: 20160809

STCB Information on status: application discontinuation

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