WO2008018795A1 - Methods and means for treating dna repeat instability associated genetic disorders - Google Patents

Methods and means for treating dna repeat instability associated genetic disorders Download PDF

Info

Publication number
WO2008018795A1
WO2008018795A1 PCT/NL2007/050399 NL2007050399W WO2008018795A1 WO 2008018795 A1 WO2008018795 A1 WO 2008018795A1 NL 2007050399 W NL2007050399 W NL 2007050399W WO 2008018795 A1 WO2008018795 A1 WO 2008018795A1
Authority
WO
WIPO (PCT)
Prior art keywords
oligonucleotide
repeat
nucleotides
sequence
complementary
Prior art date
Application number
PCT/NL2007/050399
Other languages
French (fr)
Inventor
Josephus Johannes De Kimpe
Gerard Johannes Platenburg
Derick Gert Wansink
Original Assignee
Prosensa Technologies B.V.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to AU2007282224A priority Critical patent/AU2007282224B2/en
Application filed by Prosensa Technologies B.V. filed Critical Prosensa Technologies B.V.
Priority to NZ574807A priority patent/NZ574807A/en
Priority to PL07808532T priority patent/PL2049664T3/en
Priority to CA2660523A priority patent/CA2660523C/en
Priority to CN2007800298485A priority patent/CN101501193B/en
Priority to US12/377,160 priority patent/US20100184833A1/en
Priority to DK07808532.1T priority patent/DK2049664T3/en
Priority to JP2009523740A priority patent/JP6047270B2/en
Priority to EP07808532A priority patent/EP2049664B1/en
Priority to ES07808532T priority patent/ES2373246T3/en
Priority to SI200730778T priority patent/SI2049664T1/en
Priority to AT07808532T priority patent/ATE524547T1/en
Publication of WO2008018795A1 publication Critical patent/WO2008018795A1/en
Priority to IL196921A priority patent/IL196921A/en
Priority to HK09109656.7A priority patent/HK1130510A1/en
Priority to US14/809,483 priority patent/US9890379B2/en
Priority to US15/855,848 priority patent/US10689646B2/en
Priority to US16/660,387 priority patent/US11274299B2/en
Priority to US17/591,681 priority patent/US20220267765A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • 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
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3212'-O-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/34Spatial arrangement of the modifications
    • C12N2310/346Spatial arrangement of the modifications having a combination of backbone and sugar modifications
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers

Abstract

The current invention provides for methods and medicaments that apply oligonucleotide molecules complementary onl y to a repetitive sequence in a human gene transcript, for the manufacture of a medicament for the diagnosis, treatment or prevention of a cis-element repeat instability associated genetic disorders in humans. The invention hence provides a method of treatment for cis-element repeat instability associated genetic disorders. The invention also pertains to modified oligonucleotides which can be applied in method of the invention to prevent the accumulation and/or translation of repeat expanded transcripts in cells.

Description

Title: Methods and means for treating DNA repeat instability associated genetic disorders
Field of the invention
The current invention relates to the field of medicine, in particular to the treatment of genetic disorders associated with genes that have unstable repeats in their coding or non-coding sequences, most in particular unstable repeats in the human Huntington disease causing HD gene or the myotonic dystrophy type 1 causing DMPK gene.
Background of the invention
Instability of gene-specific microsatellite and minisatellite repetitive sequences, leading to increase in length of the repetitive sequences in the satellite, is associated with about 35 human genetic disorders. Instability of trinucleotide repeats is for instance found in genes causing X-linked spinal and bulbar muscular atrophy (SBMA), myotonic dystrophy type 1 (DMl), fragile X syndrome (FRAX genes A, E, F), Huntington's disease (HD) and several spinocerebellar ataxias (SCA gene family). Unstable repeats are found in coding regions of genes, such as the Huntington's disease gene, whereby the phenotype of the disorder is brought about by alteration of protein function and/or protein folding. Unstable repeat units are also found in untranslated regions, such as in myotonic dystrophy type 1 (DMl) in the 3' UTR or in intronic sequences such as in myotonic dystrophy type 2 (DM2). The normal number of repeats is around 5 to 37 for DMPK, but increases to premutation and full disease state two to ten fold or more, to 50, 100 and sometimes 1000 or more repeat units. For DM2/ZNF9 increases to 10,000 or more repeats have been reported. (Cleary and Pearson, Cytogenet. Genome Res. 100: 25-55, 2003).
The causative gene for Huntington's disease, HD, is located on chromosome 4. Huntington's disease is inherited in an autosomal dominant fashion. When the gene has more than 35 CAG trinucleotide repeats coding for a polyglutamine stretch, the number of repeats can expand in successive generations. Because of the progressive increase in length of the repeats, the disease tends to increase in severity and presents at an earlier age in successive generations, a process called anticipation. The product of the HD gene is the 348 kDa cytoplasmic protein huntingtin. Huntingtin has a characteristic sequence of fewer than 40 glutamine amino acid residues in the normal form; the mutated huntingtin causing the disease has more than 40 residues. The continuous expression of mutant huntingtin molecules in neuronal cells results in the formation of large protein deposits which eventually give rise to cell death, especially in the frontal lobes and the basal ganglia (mainly in the caudate nucleus). The severity of the disease is generally proportional to the number of extra residues.
DMl is the most common muscular dystrophy in adults and is an inherited, progressive, degenerative, multisystemic disorder of predominantly skeletal muscle, heart and brain. DMl is caused by expansion of an unstable trinucleotide (CTG)n repeat in the 3 ' untranslated region of the DMPK gene (myotonic dystrophy protein kinase) on human chromosome 19q (Brook et al, Cell, 1992). Type 2 myotonic dystrophy (DM2) is caused by a CCTG expansion in intron 1 of the ZNF9 gene, (Liquori et al, Science 2001). In the case of myotonic dystrophy type 1, the nuclear- cytoplasmic export of DMPK transcripts is blocked by the increased length of the repeats, which form hairpin-like secondary structures that accumulate in nuclear foci. DMPK transcripts bearing a long (CUG)n tract can form hairpin-like structures that bind proteins of the muscleblind family and subsequently aggregate in ribonuclear foci in the nucleus. These nuclear inclusions are thought to sequester muscleblind proteins, and potentially other factors, which then become limiting to the cell. In DM2, accumulation of ZNF9 RNA carrying the (CCUG)n expanded repeat form similar foci. Since muscleblind proteins are splicing factors, their depletion results in a dramatic rearrangement in splicing of other transcripts. Transcripts of many genes consequently become aberrantly spliced, for instance by inclusion of fetal exons, or exclusion of exons, resulting in non-functional proteins and impaired cell function.
The observations and new insights above have led to the understanding that unstable repeat diseases, such as myotonic dystrophy type 1, Huntington's disease and others can be treated by removing, either fully or at least in part, the aberrant transcript that causes the disease. For DMl, the aberrant transcript that accumulates in the nucleus could be down regulated or fully removed. Even relatively small reductions of the aberrant transcript could release substantial and possibly sufficient amounts of sequestered cellular factors and thereby help to restore normal RNA processing and cellular metabolism for DM (Kanadia et al., PNAS 2006). In the case of HD, a reduction in the accumulation of huntingtin protein deposits in the cells of an HD patient can ameliorate the symptoms of the disease.
A few attempts have been made to design methods of treatment and medicaments for unstable repeat disease myotonic dystrophy type 1 using antisense nucleic acids, RNA interference or ribozymes. (i) Langlois et al. (Molecular Therapy, Vol. 7 No. 5, 2003) designed a ribozyme capable of cleaving DMPK mRNA. The hammerhead ribozyme is provided with a stretch RNA complementary to the 3' UTR of DMPK just before the CUG repeat. In vivo, vector transcribed ribozyme was capable of cleaving and diminishing in transfected cells both the expanded CUG repeat containing mRNA as well as the normal mRNA species with 63 and 50 % respectively. Hence, also the normal transcript is gravely affected by this approach and the affected mRNA species with expanded repeats are not specifically targeted.
(ii) Another approach was taken by Langlois et al., (Journal Biological Chemistry, vol 280, no.17, 2005) using RNA interference. A lentivirus-delivered short- hairpin RNA (shRNA) was introduced in DMl myoblasts and demonstrated to down regulate nuclear retained mutant DMPK mRNAs. Four shRNA molecules were tested, two were complementary against coding regions of DMPK, one against a unique sequence in the 3' UTR and one negative control with an irrelevant sequence. The first two shRNAs were capable of down regulating the mutant DMPK transcript with the amplified repeat to about 50%, but even more effective in down regulating the cytoplasmic wildtype transcript to about 30% or less. Equivalent synthetic siRNA delivered by cationic lipids was ineffective. The shRNA directed at the 3' UTR sequence proved to be ineffective for both transcripts. Hence, also this approach is not targeted selectively to the expanded repeat mRNA species.
(iii) A third approach by Furling et al. (Gene Therapy, Vol.10, p795-802, 2003) used a recombinant retrovirus expressing a 149-bp long antisense RNA to inhibit DMPK mRNA levels in human DMl myoblasts. A retrovirus was designed to provide DMl cells with the 149 bp long antisense RNA complementary to a 39 bp-long (CUG) 13 repeat and a 110 bp region following the repeat to increase specificity. This method yielded a decrease in mutated (repeat expanded) DMPK transcript of 80%, compared to a 50% reduction in the wild type DMPK transcript and restoration of differentiation and functional characteristics in infected DMl myoblasts. Hence, also this approach is not targeted selectively to the expanded repeat mRNA species, it depends on a very long antisense RNA and can only be used in combination with recombinant viral delivery techniques.
Detailed description of the invention
The methods and techniques described above provide nucleid acid based methods that cause non-selective breakdown of both the affected repeat expanded allele and unaffected (normal) allele for genetic diseases that are associated with repeat instability and/or expansion. Moreover, the art employs sequences specific for the gene associated with the disease and does not provide a method that is applicable to several genetic disorders associated with repeat expansion. Finally, the art only teaches methods that involve use of recombinant DNA vector delivery systems, which need to be adapted for each oligonucleotide and target cell and which still need to be further optimised.
The current invention provides a solution for these problems by using a short single stranded nucleic acid molecule that comprises or consists of a sequence, which is complementary to the expanded repeat region only, i.e. it does not rely on hybridisation to unique sequences in exons or introns of the repeat containing gene. Furthermore, it is not essential that the employed nucleic acid (oligonucleotide) reduces transcipts by the RNAse H mediated breakdown mechanism. Without wishing to be bound by theory, the current invention may cause a decrease in transcript levels by alterations in posttranscriptional processing and/or splicing of the premature RNA. A decrease in transcript levels via alternative splicing and/or postranscriptional processing is thought to result in transcripts lacking the overly expanded or instable (tri)nucleotide repeat, but still possessing functional activities. The reduction of aberrant transcripts by altered RNA processing and/or splicing may prevent accumulation and/or translation of aberrant, repeat expanded transcripts in cells.
Without wishing to be bound by theory the method of the current invention is also thought to provide specificity for the affected transcript with the expanded repeat because the kinetics for hybridisation to the expanded repeat are more favourable. The likelihood that a repeat specific complementary nucleic acid oligonucleotide molecule will hybridise to a complementary stretch in an RNA or DNA molecule increases with the size of the repetitive stretch. RNA molecules and in particular RNA molecules comprising repetitive sequences are normally internally paired, forming a secondary structure comprising open loops and closed hairpin parts. Only the open parts are relatively accessible for complementary nucleic acids. The short repeat stretches of a wild type transcript not associated with disease is often only 5 to about 20-40 repeats and due to the secondary structure relatively inaccessible for base pairing with a complementary nucleic acid. In contrast, the repeat units of the expanded repeat and disease associated allele is normally at least 2 fold expanded but usually even more, 3, 5, 10 fold, up to 100 or even more than 1000 fold expansion for some unstable repeat disorders. This expansion increases the likelihood that part of the repeat is, at least temporarily, in an open loop structure and thereby more accessible to base pairing with a complementary nucleic acid molecule, relative to the wild type allele. So despite the fact that the oligonucleotide is complementary to a repeat sequence present in both wildtype and repeat-expanded transcripts and could theoretically hybridise to both transcripts, the current invention teaches that oligonucleotides complementary to the repetitive tracts preferably hybridise to the disease-associated or disease-causing transcripts and leave the function of normal transcripts relatively unaffected. This selectivity is beneficial for treating diseases associated with repeat instability irrespective of the mechanism of reduction of the aberrant transcript.
The invention thus provides a method for the treatment of unstable cw-element DNA repeat associated genetic disorders, by providing nucleic acid molecules that are complementary to and/or capable of hybridising to the repetitive sequences only. This method thereby preferentially targets the expanded repeat transcripts and leaves the transcripts of the normal, wild type allele relatively unaffected. This is advantageous since the normal allele can thereby provide for the normal function of the gene, which is at least desirable and, depending on the particular gene with unstable DNA repeats, may in many cases be essential for the cell and/or individual to be treated.
Furthermore, this approach is not limited to a particular unstable DNA repeat associated genetic disorder, but may be applied to any of the known unstable DNA repeat diseases, such as, but not limited to: coding regions repeat diseases having a polyglutamine (CAG) repeat: Huntington's disease, Haw River syndrome, Kennedy's disease/spinobulbar muscular atrophy, spino-cerebellar ataxia, or diseases having polyalanine (GCG) repeats such as: infantile spasm syndrome, deidocranial dysplasia, blepharophimosis/ptosis/epicanthus invensus syndrome, hand-foot-genital syndrome, synpolydactyly, oculopharyngeal muscular dystrophy, holoprosencephaly. Diseases with repeats in non-coding regions of genes to be treated according to the invention comprise the trinucleotide repeat disorders (mostly CTG and/or CAG and/or CCTG repeats): myotonic dystrophy type 1, myotonic dystrophy type 2, Friedreich's ataxia (mainly GAA), spino-cerebellar ataxia, autism. Furthermore, the method of the invention can be applied to fragile site associated repeat disorder comprising various fragile X-syndromes, Jacobsen syndrome and other unstable repetitive element disorders such as myoclonus epilepsy, facioscapulohumeral dystrophy and certain forms of diabetes mellitus type 2. Another advantage of the current invention is that the oligonucleotides specific for a repeat region may be administered directly to cells and it does not rely on vector-based delivery systems. The techniques described in the prior art, for instance those mentioned above for treatment of DMl and removal of DMPK transcripts from cells, require the use of vector based delivery systems to administer sufficient levels of oligonucleotides to the cell. The use of plasmid or viral vectors is yet less desirable for therapeutic purposes because of current strict safety regulations for therapeutic recombinant DNA vectors, the production of sufficient recombinant vectors for broad clinical application and the limited control and reversibility of an exaggerated (or nonspecific) response after application. Nevertheless, optimisation in future is likely in these areas and viral delivery of plasmids could yield an advantageous long lasting effect. The current inventors have surprisingly found that oligonucleotides that comprise or consist of a sequence that is complementary to repetitive sequences of expanded repeat transcripts, due to the expansion of their molecular target for hybridisation, have a much increased affinity and/or avidity for their target in comparison to oligonucleotides that are specific for unique sequences in a transcript. Because of this high affinity and avidity for the repeat expanded target transcript, lower amounts of oligonucleotide suffice to yield sufficient inhibition and/or reduction of the repeat expanded allele by RNase H degradation, RNA interference degradation or altered post-transcriptional processing (comprising but not limited to splicing or exon skipping) activities. The oligonucleotides of the current invention which are complementary to repetitive sequences only, may be produced synthetically and are potent enough to be effective when delivered directly to cells using commonly applied techniques for direct delivery of oligonucleotides to cells and/or tissues. Recombinant vector delivery systems may, when desired, be circumvented by using the method and the oligonucleotide molecules of the current invention.
In a first aspect, the current invention discloses and teaches the use of an oligonucleotide comprising or consisting of a sequence that is complementary only to a repetitive sequence in a human gene transcript for the manufacture of a medicament for the diagnosis, treatment or prevention of a cw-element repeat instability associated genetic disorders in humans. The invention hence provides a method of treatment for czs -element repeat instability associated genetic disorders.
In a second aspect, the invention also pertains to an oligonucleotide which can be used in the first aspect of the invention and/or applied in method of the invention to prevent the accumulation and/or translation of repeat expanded transcripts in cells.
An oligonucleotide of the invention may comprise a sequence that is complementary only to a repetitive sequence as defined below. Preferably, the repetitive sequence 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 more. In a most preferred embodiment, the oligonucleotide of the invention consists of a sequence that is complementary only to a repetitive sequence as defined below. For example, an oligonucleotide may comprise a sequence that is complementary only to a repetitive sequence as defined below and a targeting part, which is later on called a targeting ligand.
A repeat or repetitive element or repetitive sequence or repetitive stretch is herein defined as a repetition of at least 3, 4, 5, 10, 100, 1000 or more, of a repetitive unit or repetitive nucleotide unit or repeat nucleotide unit comprising a trinucleotide repetitive unit, or alternatively a 4, 5 or 6 nucleotide repetitive unit, in a transcribed gene sequence in the genome of a subject, including a human subject.
An oligonucleotide may be single stranded or double stranded. Double stranded means that the oligonucleotide is an heterodimer made of two complementary strands, such as in a siRNA. In a preferred embodiment, an oligonucleotide is single stranded. A single stranded oligonucleotide has several advantages compared to a double stranded siRNA oligonucleotide: (i) its synthesis is expected to be easier than two complementary siRNA strands; (ii) there is a wider range of chemical modifications possible to optimise more effective uptake in cells, a better (physiological) stability and to decrease potential generic adverse effects; and (iii) siRNAs have a higher potential for non-specific effects and exaggerated pharmacology (e.g. less control possible of effectiveness and selectivity by treatment schedule or dose) and (iv) siRNAs are less likely to act in the nucleus and cannot be directed against introns. Therefore, in a preferred embodiment of the first aspect, the invention relates to the use of a single stranded oligonucleotide comprising or consisting of a sequence that is complementary only to a repetitive sequence in a human gene transcript for the manufacture of a medicament for the diagnosis, treatment or prevention of a cw-element repeat instability associated genetic disorders in humans.
The oligonucleotide(s) preferably comprise at least 10 to about 50 consecutive nucleotides complementary to a repetitive element, more preferably 12 to 45 nucleotides, even more preferably 12 to 30, and most preferably 12 to 25 nucleotides complementary to a repetitive stretch, preferably having a trinucleotide repeat unit or a tetranucleotide repeat unit. The oligonucleotide may be complementary to and/or capable of hybridizing to a repetitive stretch in a coding region of a transcript, preferably a polyglutamine (CAG) or a polyalanine (GCG) coding tract. The oligonucleotide may also be complementary to and/or capable of hybridizing to a non- coding region for instance 5' or 3' untranslated regions, or intronic sequences present in precursor RNA molecules.
In a preferred embodiment the oligonucleotide to be used in the method of the invention comprises or consists of a sequence that is complementary to a repetitive element having as repetitive nucleotide unit a repetitive nucleotide unit selected from the group consisting of (CAG)n, (GCG)n, (CUG)n, (CGG)n (GAA)n, (GCC)n and (CCUG)n. and said oligonucleotide being a single or double stranded oligonucleotide. Preferably, the oligonucleotide is double stranded.
The use of an oligonucleotide that comprises or consists of a sequence that is complementary to a polyglutamine (CAG)n tract in a transcript is particularly useful for the diagnosis, treatment and/or prevention of the human disorders Huntington's disease, several forms of spino-cerebellar ataxia or Haw River syndrome, X-linked spinal and bulbar muscular atrophy and/or dentatorubral-pallidoluysian atrophy caused by repeat expansions in the HD, HDL2/JPH3, SBMA/AR, SCA1/ATX1, SCA2/ATX2, SCA3/ATX3, SCA6/CACNAIA, SCA7, SCAl 7, AR or DRPLA human genes.
The use of an oligonucleotide that comprises or consists of a sequence that is complementary to a polyalanine (GCG)n tract in a transcript is particularly useful for the diagnosis, treatment and/or prevention of the human disorders: infantile spasm syndrome, deidocranial dysplasia, blepharophimosis, hand-foot-genital disease, synpolydactyly, oculopharyngeal muscular dystrophy and/or holoprosencephaly, which are caused by repeat expansions in the ARX, CBFAl, FOXL2, H0XA13, H0XD13, OPDM/PABP2, TCFBRl or ZIC2 human genes. The use of an oligonucleotide that comprises or consists of a sequence that is complementary to a (CUG)n repeat in a transcript and is particularly useful for the diagnosis, treatment and/or prevention of the human genetic disorder myotonic dystrophy type 1, spino-cerebrellar ataxia 8 and/or Huntington's disease-like 2 caused by repeat expansions in the DM1/DMPK, SCA8 or JPH3 genes respectively. Preferably, these genes are from human origin.
The use of an oligonucleotide that comprises or consists of a sequence that is complementary to a (CCUG)n repeat in a transcript is particularly useful for the diagnosis, treatment and/or prevention of the human genetic disorder myotonic dystrophy type 2, caused by repeat expansions in the DM2/ZNF9 gene. The use of an oligonucleotide that comprises or consists of a sequence that is complementary to a (CGG)n repeat in a transcript is particularly useful for the diagnosis, treatment and/or prevention of human fragile X syndromes, caused by repeat expansion in the FRAXA/FMRl, FRAXE/FMR2 and FRAXF/FAMl IA genes.
The use of an oligonucleotide that comprises or consists of a sequence that is complementary to a (CCG)n repeat in a transcript is particularly useful for the diagnosis, treatment and/or prevention of the human genetic disorder Jacobsen syndrome, caused by repeat expansion in the FRAl 1B/CBL2 gene.
The use of an oligonucleotide that comprises or consists of a sequence that is complementary to a (GAA)n repeat in a transcript is particularly useful for the diagnosis, treatment and/or prevention of the human genetic disorder Friedreich's ataxia.
The use of an oligonucleotide that comprises or consists of a sequence that is complementary to a (ATTCT)n repeat in an intron is particularly useful for the diagnosis, treatment and/or prevention of the human genetic disorder Spinocerebellar ataxia type 10 (SCAlO).
The repeat-complementary oligonucleotide to be used in the method of the invention may comprise or consist of RNA, DNA, Locked nucleic acid (LNA), peptide nucleic acid (PNA), morpholino phosphorodiamidates (PMO), ethylene bridged nucleic acid (ENA) or mixtures/hybrids thereof that comprise combinations of naturally occurring DNA and RNA nucleotides and synthetic, modified nucleotides. In such oligonucleotides, the phosphodiester backbone chemistry may further be replaced by other modifications, such as phosphorothioates or methylphosphonates. Other oligonucleotide modifications exist and new ones are likely to be developed and used in practice. However, all such oligonucleotides have the character of an oligomer with the ability of sequence specific binding to RNA. Therefore in a preferred embodiment, the oligonucleotide comprises or consists of RNA nucleotides, DNA nucleotides, locked nucleic acid (LNA) nucleotides, peptide nucleic acid (PNA) nucleotides, morpholino phosphorodiamidates, ethylene-bridged nucleic acid (ENA) nucleotides or mixtures thereof with or without phosphorothioate containing backbones.
Oligonucleotides containing at least in part naturally occurring DNA nucleotides are useful for inducing degradation of DNA-RNA hybrid molecules in the cell by RNase H activity (EC.3.1.26.4).
Naturally occurring RNA ribonucleotides or RNA-like synthetic ribonucleotides comprising oligonucleotides may be applied in the method of the invention to form double stranded RNA-RNA hybrids that act as enzyme-dependent antisense through the RNA interference or silencing (RNAi/siRNA) pathways, involving target RNA recognition through sense-antisense strand pairing followed by target RNA degradation by the RNA-induced silencing complex (RISC).
Alternatively or in addition, steric blocking antisense oligonucleotides (RNase-H independent antisense) interfere with gene expression or other precursor RNA or messenger RNA-dependent cellular processes, in particular but not limited to RNA splicing and exon skipping, by binding to a target sequence of RNA transcript and getting in the way of processes such as translation or blocking of splice donor or splice acceptor sites. Alteration of splicing and exon skipping techniques using modified antisense oligonucleotides are well documented, known to the skilled artisan and may for instance be found in US6,210,892, WO9426887, WO04/083446 and WO02/24906. Moreover, steric hindrance may inhibit the binding of proteins, nuclear factors and others and thereby contribute to the decrease in nuclear accumulation or ribonuclear foci in diseases like DMl .
The oligonucleotides of the invention, which may comprise synthetic or modified nucleotides, complementary to (expanded) repetitive sequences are useful for the method of the invention for reducing or inactivating repeat containing transcripts via the siRNA / RNA interference or silencing pathway. Single or double stranded oligonucleotides to be used in the method of the invention may comprise or consist of DNA nucleotides, RNA nucleotides, 2'-0 substituted ribonucleotides, including alkyl and methoxy ethyl substitutions, peptide nucleic acid (PNA), locked nucleic acid (LNA) and morpholino (PMO) antisense oligonucleotides and ethylene-bridged nucleotides (ENA) and combinations thereof, optionally chimeras with RNAse H dependent antisense. Integration of locked nucleic acids in the oligonucleotide changes the conformation of the helix after base pairing and increases the stability of the duplex. Integration of LNA bases into the oligonucleotide sequence can therefore be used to increase the ability of complementary oligonucleotides of the invention to be active in vitro and in vivo to increase RNA degradation inhibit accumulation of transcripts or increase exon skipping capabilities. Peptide nucleic acids (PNAs), an artificial DNA/RNA analog, in which the backbone is a pseudopeptide rather than a sugar, have the ability to form extremely stable complexes with complementary DNA oligomers, by increased binding and a higher melting temperature. Also PNAs are superior reagents in antisense and exon skipping applications of the invention. Most preferably, the oligonucleotides to be used in the method of this invention comprise, at least in part or fully, 2'-O-methoxy ethyl phosphorothioate RNA nucleotides or 2'-O-methyl phosphorothioate RNA nucleotides. Oligonucleotides comprising or consisting of a sequence that is complementary to a repetitive sequence selected from the group consisting of (CAG)n, (GCG)n, (CUG)n, (CGG)n, (CCG)n, (GAA)n, (GCQn and (CCUG)n having a length of 10 to 50, more preferably 12 to 35, most preferably 12 to 25 nucleotides, and comprising T-O- methoxyethyl phosphorothioate RNA nucleotides, 2'-O-methyl phosphorothioate RNA nucleotides, LNA nucleotides or PMO nucleotides are most preferred for use in the invention for the diagnosis, treatment of prevention of cw-element repeat instability genetic disorders.
Accordingly, in a preferred embodiment, an oligonucleotide of the invention and used in the invention comprises or consists of a sequence that is complementary to a repetitive sequence selected from the group consisting of (CAG)n, (GCG)n, (CUG)n, (CGG)n , (GAA)n, (GCC)n and (CCUG)n, has a length of 10 to 50 nucleotides and is further characterized by: a) comprising 2'-0-substituted RNA phosphorothioate nucleotides such as 2'-O-methyl or 2'-O-methoxy ethyl RNA phosphorothiote nucleotides, LNA nucleotides or PMO nucleotides. The nucleotides could be used in any combination and/or with DNA phosphorothioate or RNA nucleotides; and/or b) being a single stranded oligonucleotide.
Accordingly, in another preferred embodiment, an oligonucleotide of the invention and used in the invention comprises or consists of a sequence that is complementary to a repetitive sequence selected from the group consisting of (CAG)n, (GCG)n, (CUG)n, (CGG)n, (GAA)n, (GCC)n and (CCUG)n, has a length of 10 to 50 nucleotides and is further characterized by: c) comprising 2'-0-substituted RNA phosphorothioate nucleotides such as 2'-O-methyl or 2'-O-methoxy ethyl RNA phosphorothiote nucleotides, LNA nucleotides or PMO nucleotides. The nucleotides could be used in combination and/or with DNA phosphorothioate or RNA nucleotides; and/or d) being a double stranded oligonucleotide.
In case, the invention relates to a double stranded oligonucleotide with two complementary strands, the antisense strand, complementary only to a repetitive sequence in a human gene transcript, this double stranded oligonucleotide is preferably not the siRNA with antisense RNA strand (CUG)7 and sense RNA strand (GCA)7 applied to cultured monkey fibroblast (COS-7) or human neuroblastoma (SH-SY5Y) cell lines with or without transfection with a human Huntington gene exon 1 fused to GFP and as depicted in Wanzhao Liu et al (Wanzhao Liu et al, (2003), Proc. Japan Acad, 79: 293-298). More preferably, the invention does neither relate to the double stranded oligonucleotide siRNA (with antisense strand (CUG)7 and sense strand (GCA)v) nor to its use for the manufacture of a medicament for the treatment or prevention of Huntington disease, even more preferably for the treatment or prevention of Huntington disease gene exon 1 containing construct.
Although use of a single oligonucleotide may be sufficient for reducing the amount of repeat expanded transcripts, such as nuclear accumulated DMPK or ZNF9 transcripts or segments thereof or sufficient reduction of accumulation of repeat expanded HD protein, it is also within the scope of the invention to combine 2, 3, 4, 5 or more oligonucleotides. The oligonucleotide comprising or consisting of a sequence that is complementary to a repetitive part of a transcript may be advantageously combined with oligonucleotides that comprise or consist of sequences that are complementary to and/or capable of hybridizing with unique sequences in a repeat containing transcript. The method of the invention and the medicaments of the invention comprising repeat specific oligonucleotides may also be combined with any other treatment or medicament for cw-element repeat instability genetic disorders. For diagnostic purposes the oligonucleotide used in the method of the invention may be provided with a radioactive label or fluorescent label allowing detection of transcripts in samples, in cells in situ in vivo, ex vivo or in vitro. For myotonic dystrophy, labelled oligonucleotides may be used for diagnostic purposes, for visualisation of nuclear aggregates of DMPK or ZNF9 RNA transcript molecules with associated proteins. Fluorescent labels may comprise Cy3, Cy5, FITC, TRITC, Rhodamine, GFP and the like. Radioactive labels may comprise 3H, 35S, 32 33P, 125I. Enzymes and/or immunogenic haptens such as digoxigenin, bio tin and other molecular tags (HA, Myc, FLAG, VSV, lexA) may also be used. Accordingly, in a further aspect, the invention discloses an vitro or ex vivo detection and/or diagnostic method wherein a oligonucleotide as defined above is used.
The oligonucleotides for use according to the invention are suitable for direct administration to cells, tissues and/or organs in vivo of individuals affected by or at risk of developing a cw-element repeat instability disorder, and may be administered directly in vivo, ex vivo or in vitro. Alternatively, the oligonucleotides may be provided by a nucleic acid vector capable of conferring expression of the oligonucleotide in human cells, in order to allow a sustainable source of the oligonucleotides. Oligonucleotide molecules according to the invention may be provided to a cell, tissue, organ and/or subject to be treated in the form of an expression vector that is capable of conferring expression of the oligonucleotide in human cells. The vector is preferably introduced in the cell by a gene delivery vehicle. Preferred vehicles for delivery are viral vectors such as retroviral vectors, adeno-associated virus vectors (AAV), adenoviral vectors, Semliki Forest virus vectors (SFV), EBV vectors and the like. Also plasmids, artificial chromosomes, plasmids suitable for targeted homologous recombination and integration in the human genome of cells may be suitably applied for delivery of oligonucleotides. Preferred for the current invention are those vectors wherein transcription is driven from polIII promoters, and/or wherein transcripts are in the form fusions with Ul or U7 transcripts, which yield good results for delivering small transcripts. In a preferred embodiment, a concentration of oligonucleotide, which is ranged between about 0.1 nM and about 1 μM is used. More preferably, the concentration used is ranged between about 0.3 to about 400 nM, even more preferably between about 1 to about 200 nM. If several oligonucleotides are used, this concentration 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, the oligonucleotides to be used in the invention to prevent, treat or diagnose cw-element repeat instability disorders are synthetically produced and administered directly to cells, tissues, organs and/or patients in formulated form in pharmaceutically acceptable compositions. The delivery of the pharmaceutical compositions 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. An intrathecal or intraventricular administration (in the cerebrospinal fluid) is preferably realized by introducing a diffusion pump into the body of a subject. Several diffusion pumps are known to the skilled person.
Pharmaceutical compositions that are to be used to target the oligonucleotide molecules comprising or consisting of a sequence that is complementary to repetitive sequences may comprise various excipients such as diluents, fillers, preservatives, solubilisers and the like, which may for instance be found in Remington: The Science and Practice of Pharmacy, 20th Edition. Baltimore, MD: Lippincott Williams & Wilkins, 2000.
Particularly preferred for the method of the invention is the use of excipients that will aid in delivery of the oligonucleotides to the cells and into the cells, in particular excipients capable of forming complexes, vesicles and/or liposomes that deliver substances and/or oligonucleotide(s) complexed or trapped in the vesicles or liposomes through a cell membrane. Many of these substances are known in the art. Suitable substances comprise polyethylenimine (PEI), ExGen 500, synthetic amphiphils (SAINT-18), lipofectinTM, DOTAP and/or viral capsid proteins that are capable of self assembly into particles that can deliver oligonucleotides to cells. Lipofectin represents an example of liposomal transfection agents. It consists of two lipid components, a cationic lipid N-[I -(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 oligonucleotides across cell membranes into cells. In addition to these common nanoparticle materials, the cationic peptide protamine offers an alternative approach to formulate oligonucleotides 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 the oligonucleotides. The skilled person may select and adapt any of the above or other commercially available alternative excipients and delivery systems to package and deliver oligonucleotides for use in the current invention to deliver oligonucleotides for the treatment of cw-element repeat instability disorders in humans. In addition, the 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 oligonucleotide from vesicles, e.g. endosomes or lysosomes. Such targeting ligand would also encompass molecules facilitating the uptake of oligonucleotides into the brain through the blood brain barrier. Therefore, in a preferred embodiment, an oligonucleotide in a medicament is provided with at least an excipient and/or a targeting ligand for delivery and/or a delivery device of the oligonucleotide to cells and/or enhancing its intracellular delivery. Accordingly, the invention also encompasses a pharmaceutically acceptable composition comprising an oligonucleotide of the invention and further comprising at least one excipient and/or a targeting ligand for delivery and/or a delivery device of the oligonucleotide to the cell and/or enhancing its intracellular delivery.
The invention also pertains to a method for the reduction of repeat containing gene transcripts in a cell comprising the administration of a single strand or double stranded oligonucleotide molecule, preferably comprising 2'-0-substituted RNA phosphorothioate nucleotides such as 2'-O-methyl or 2'-O-methoxy ethyl RNA phosphorothioate nucleotides or LNA nucleotides or PMO nucleotides, and having a length of 10 to 50 nucleotides that are complementary to the repetitive sequence only. The nucleotides could be used in combination and/or with DNA phosphorothioate nucleotides. 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 combinations and/or items not specifically mentioned are not excluded. 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".
Figure legends Figure 1 : Northern blot of RNA isolated from myotubes transfected with different oligonucleotides or mock control. The myotubes were derived from immorto mouse myoblast cell lines containing a transgenic human DMPK genes with (CTG)n repeat expansion length of approximately 500 next to its normal mouse DMPK gene without (CTG) repeat. The blot shows human DMPK mRNA (top), mouse DMPK (mDMPK) mRNA (middle) and mouse GAPDH mRNA (bottom).
Figure 2: The human and mouse DMPK signals of figure 1 were quantified by phosphoimager analysis and normalized to the GAPDH signal. The results are expressed relative to the mock treatment (set to 100).
Figure 3 : Northern blot of total RNA isolated from murine myotubes containing a mouse-human chimaeric DMPK gene in which the 3' part of the mDMPK gene was replaced by the cognate segment of the human DMPK gene including a (CTG)πo- repeat. The blot was probed for DMPK mRNA (upper panel) and mouse GAPDH mRNA (bottom). Cells were transfected with antisense oligonucleotide PS58 or control.
Figure 4 shows the response of DM500 myotubes treated with various concentrations of oligonucleotide PS58. The expression of hDMPK was quantified via Northern blot analysis followed by phosphoimager analysis. The signal was normalised to the GAPDH signal and expressed relative to the response after mock treatment.
Figure 5 shows the Northern blot of total RNA of DM500 myotubes transfected with 20OnM PS58 at different time points: 2h, 4h, 8h and 48h before harvesting. Mock treatment was performed 48h before harvesting. Northern blots show human and mouse DMPK and mouse GAPDH mRNA. These were quantified by phosphoimager and the normalized DMPK signal was expressed relative to mock treatment. Figure 6 shows the Northern blot of total RNA of DM500 myotubes harvested 2d, 4d, 6d and 8d after transfection with 200 nM PS58 or mock control. Northern blot analysis and quantification was performed as before.
Figure 7 shows a Northern blot of total RNA from MyoD -trans formed myoblasts treated with oligonucleotide PS58 (20 and 200 nM) or mock control. The myoblasts were derived from fibroblasts obtained from a congenital myotonic dystrophy type I patient bearing a hDMPK allele with a triplet repeat expansion length of approximately 1500 and a hDMPK allele with normal length of 11 repeats. The Northern blot was hybridized with a human DMPK (hDMPK) probe and GAPDH mRNA probe. The human DMPK signals were normalized to the GAPDH signal and expressed relative to mock control.
Figure 8 shows the RT-PCR analysis of DM500 myotubes transfected with 200 nM of oligonucleotide PS58, specific to the (CUG) repeat sequence only, oligonucleotide
PSl 13, specific to a sequence in exon 1, or mock control. RT-PCR analysis was performed with primers specific for hDMPK mRNA and three other gene transcripts with a naturally occurring (CUG) repeat in mice: Ptbpl mRNA with a (CUG)6,
Syndecan3 mRNA with a (CUG)6 and Taxilinbeta mRNA with a (CUG)9. The intensity of the signals were normalized to the actin signal and expressed relative to mock control.
Figure 9 shows FISH analysis of DM500 myoblasts transfected with 20OnM PS58 (B) or mock control (A). Fourty eight hours after the start of the treatment, the cells were washed and fixed and subsequently hybridized with fluorescently labeled oligonucleotide Cy3 -(CAG) 10-Cy3. The ribonuclear foci indicative of hDMPK (CUG)5oo mRNA aggregation in the nucleus were visualized using a Bio-Rad MRC 1024 confocal laser scanning microscope and LaserSharp2000 acquisition software.
Figure 10 shows the relative cell count for the presence of ribonuclear foci in the nucleus of DM500 myoblasts transfected with PS58 or mock control from the experiment depicted in Figure 9. Figure 11 shows the RT-PCR analysis of hDMPK mRNA in muscle of DM500 mice treated with PS58 or mock control. Shortly, PS58 (2nmol) was injected in the GPS complex of one-year-old DM500 mice and this procedure was repeated after 24h. After 15 days, M. plantaris and M. gastrocnemius were isolated and RT-PCR was performed on total RNA for hDMPK and mouse actin. The intensity of the hDMPK signal was normalized to the actin signal and the values expressed relative to mock control.
Figure 12 shows a Northern blot analysis of DM500 myotubes treated with different oligonucleotides (20OnM) or mock control. PS58, PS146 and PS147 carried a full 2'O- methyl phosphorothiate backbone, but differed in length, (CAG)7, (CUG)IO and
(CUG)5, respectively. PS142 has a complete phosphorothiate DNA backbone with a
(CAG)7 sequence. hDMPK and mDMPK signals were normalized to mouse GAPDH and expressed as percentage to mock control. Quantification is shown in the lower panel.
Examples Example 1.
Immortomyoblast cell lines were derived from DM500 or CTGl 10 mice using standard techniques known to the skilled person. DM500 mice were derived from mice obtained from de Gourdon group in Paris. CTGl 10 mice are described below and present at the group of Wieringa and Wansink in Nijmegen. Immortomyoblast cell lines DM500 or CTGI lO with variable (CTG)n repeat length in the DMPK gene were grown subconfluent and maintained in a 5% CO2 atmosphere at 33°C on 0.1% gelatin coated dishes. Myoblast cells were grown subconfluent in DMEM supplemented with 20% FCS, 50 μg/ml gentamycin and 20 units of γ-interferon/ml. Myotube formation was induced by growing myoblast cells on Matrigel (BD Biosciences) coated dishes and placing a confluent myoblast culture at 37 0C and in DMEM supplemented with 5% horse serum and 50 μg/ml gentamycin. After five days on this low serum media contracting myotubes arose in culture and were transfected with the desired oligonucleotides. For transfection NaCl (500 mM, filter sterile), oligonucleotide and transfection reagens PEI (ExGen 500, Fermentas) were added in this specific order and directly mixed. The oligonucleotide transfection solution contained a ratio of 5 μl ExGen500 per ug oligonucleotide which is according to the instructions (ExGen 500, Fermentas). After 15 minutes of incubation at room temperature the oligonucleotide transfection solution was added to the low serum medium with the cultured myotubes and gently mixed. The final oligonucleotide concentration was 20OnM. Mock control treatment is carried out with transfection solution without an oligonucleotide. After four hours of incubation at 37 0C, fresh medium was added to the culture (resulting in a dilution of approximately 2.3x) and incubation was extended overnight at 37 0C. The next day the medium containing the oligonucleotide was removed and fresh low serum medium was added to the myotubes which were kept in culture at 37 0C for another day. Fourty eight hours after the addition of oligonucelotide to the myotube culture (which is seven days after switching to low serum conditions to induced myotube formation), RNA was isolated with the "Total RNA mini kit" (Bio-Rad) and prepared for Northern blot and RT-PCR analysis. The Northern blot was hybridized with a radioactive human DMPK (hDMPK) probe and a mouse GAPDH probe. The probe used for DMPK is a human DMPK cDNA consisting of the DMPK open reading frame with full 3' UTR and 11 CTGs.
The human and mouse DMPK signal were quantified by phosphoimager analysis and normalized to the GAPDH signal. Primers that were used for the RT-PCR for hDMPK mRNA were situated in the 3 'untranslated part with the sequence 5'- GGGGGATCACAGACCATT-3 ' and 5 '-TCAATGCATCCAAAACGTGGA-S ' and for murine actin the primers were as followed: Actin sense 5'- GCTAYGAGCTGCCTGACGG-3' and Actin antisense 5'-
GAGGCCAGGATGGAGCC-3' . PCR products were run on an agarose gel and the signal was quantified using Labworks 4.0 (UVP Biolmaging systems, Cambridge, United Kingdom). The intensity of each band was normalized to the intensity of the corresponding actin band and expressed relative to mock control. Thirteen different oligonucleotides were tested (for an overview see Table 1) as described above on the immortomyoblast DM500 cell line containing transgenic human DMPK gene with (CTG)n repeat length of approximately 500 and a normal mouse DMPK gene without (CTG) repeat. Figure 1 shows the Northern blot of the isolated RNA from the oligonucleotide transfected myotubes visualized with the hDMPK probe and a GAPDH probe for loading control. Quantification of the human DMPK (with CTG repeat) and murine DMPK (without CTG repeat) signal on the Northerm blot is shown in Figure 2. The signal was normalized to murine GAPDH and expressed relative to mock control.
Table 2 indicates the level of hDMPK mRNA reduction that is caused by a specific oligonucleotide. The minus (-) stands for no reduction and the number of positive signs (+) stands for the relative level of hDMPK mRNA break-down. Clearly, oligonucleotide PS58, specifically targeted to the repeat sequence, is much more potent in reducing or altering hDMPK transcripts than the other oligonucleotides complementary to unique sequences in the hDMPK transcripts.
Figure 3 shows the effect of PS58 in murine immortomyotubes derived from CTGI lO mice, a knock-in mouse containing a DMPK gene with the 3' part of the human DMPK gene including a (CTG) repeat of approximately 110. Northern blot analysis showed that the DMPK transcript containing the (CTG)110 repeat was reduced by the treatment with oligonucleotide PS58 but not after mock treatment.
Example 2 (Figure 4)
The DM500 immortomyoblast cell line carrying a human DMPK gene with an approximate (CTG)500 repeat expansion was cultured, prepared and transfected as described above (see example 1). In this example, the transfection was carried out with PS58 at different concentrations. Eighty four hours after start of treatment, the myotubes were harvested and Northern blot analysis was performed on isolated RNA as described above (see example 1).
Figure 4 shows the quantification of the hDMPK mRNA signal preformed by phosphoimager analysis and normalized to the GAPDH signal at different concentrations. Under these conditions, a half maximal effect was observed at around 1 nM.
Example 3 (Figure 5 and 6)
The DM500 immortomyoblast cell line carrying a human DMPK gene with an approximate (CTG)500 repeat expansion was cultured, prepared and transfected as described above (see example 1). However, in this example the transfection with
20OnM PS58 was carried out at different time points. Usually DM500 myotubes were harvested seven days after switching to low serum conditions to induce myotube formation. The standard procedure (as in example 1 and 2) was to start treatment (trans fection) 48h (two days) before harvesting. Now, treatment with PS58 was started 2h - 48h (Figure 5) or 2d-8d (Figure 6) before harvesting. Northern blot analysis and quantification was performed as before.
Figure 5 shows that expanded hDMPK mRNA in DM500 myotubes was decreased rapidly within 2 h of treatment with oligonucleotide PS58 compared to mock control treatment.
Figure 6 shows a persistent decrease in expanded hDMPK mRNA in DM500 myotubes for at least 8 days. Please note that in the case of the 8d experiment, cells were transfected in the myoblast stage (approximately 60% confluent, 330C, high serum) and that they have received fresh medium on various occasions until harvesting (including a change to low serum at 37 0C, two days after trans fection). Example 2 and 3 are indicative of a highly efficient inhibitory intervention by an oligonucleotide directed solely to the repeat expansion. The magnitude of this effect might be influenced by the relative low levels of hDMPK expression in these model cell systems, which normally is also seen in humans.
Example 4 (Figure 7)
In this example, fibroblasts obtained from a human patient with congenital myotonic dystrophy type 1 (cDMl) were used. These patient cells carry one disease causing DMPK allele with a triplet repeat expansion length of 1500 and one normal DMPK allele with a repeat length of 11. The size of the (CTG)n expansion on both alleles was confirmed with PCR and Southern blotting.
The fibroblasts were grown sub confluent and maintained in a 5% CO2 atmosphere at 37°C on 0.1% gelatin coated dishes. Fibroblasts were grown subconfluent in DMEM supplemented with 10% FCS and 50 μg/ml gentamycin. Myotube formation was induced by growing fibroblasts cells on Matrigel (BD Biosciences) coated dishes and infecting the cells at 75% confluency with MyoD-expressing adenovirus (Ad5Fib50MyoD, Crucell, Leiden) (MOI=IOO) in DMEM supplemented with 2% HS and 50 μg/ml gentamycin for 2 hours. After the incubation period MyoD adenovirus was removed and DMEM supplemented with 10% FCS and 50 μg/ml gentamycin was added. The cells were maintained in this medium in a 5% CO2 atmosphere at 37°C until 100% confluency. At this point cells were placed in DMEM supplemented with 2% FCS and 50 μg/ml gentamycin. After five days on this low serum media cells were transfected with PS58 following the procedure according to the instructions (ExGen 500, Fermentas) and as described above. The final oligonucleotide concentration was 200 nM and 20 nM. Fourty eight hours after start of the treatment (which is seven days after switching to low serum conditions), RNA was isolated with the "Total RNA mini kit" (Bio-Rad) and prepared for Northern blot. The Northern blot was hybridized with a radioactive human DMPK (hDMPK) and mouse GAPDH mRNA probe. The human DMPK signals were quantified by phosphoimager analysis and normalized to the GAPDH signal and expressed relative to mock control. Figure 7 shows the Northern blot analysis of the MyoD -transformed myoblasts treated with oligonucleotide PS58 (20 and 200 nM). The results demonstrate an effective complete inhibition of the disease-causing hDMPK (CUG)1500 RNA transcript, while the smaller normal hDMPK (CUG)11 RNA transcript is only moderately affected at the two concentrations. Thus, oligonucleotides directed to the repeat region exhibit selectivity towards the larger repeat size (or disease causing expansion).
Example 5 (Figure 8)
In this example, the DM500 immortomyoblast cell line carrying a human DMPK gene with an approximate (CTG)500 repeat expansion was cultured, transfected and analysed as described before in example 1. The DM500 myotubes were treated 48h before harvesting with 200 nM of oligonucleotide PS58, specific to the (CUG) repeat sequence only, oligonucleotide PSl 13, specific to a sequence in exon 1, or mock control. RT-PCR analysis was performed on hDMPK mRNA expressed in this murine cell line (for primers see example 1) and on three other gene transcripts with a naturally occurring (CUG) repeat in mice, Ptbpl with a (CUG)6, Syndecan3 with a (CUG)6 and Taxilinbeta with a (CUG)9.
The PCR primers used were for Ptbpl : 5 '-TCTGTCCCTAATGTCCATGG-S ' and 5'- GCCATCTGCACAAGTGCGT-3'; for Syndecan3: 5 '-GCTGTTGCTGCCACCGCT- 3' and 5 '-GGCGCCTCGGGAGTGCTA-S '; and for Taxilinbeta: 5'- CTCAGCCCTGCTGCCTGT-3 ' and 5 '-CAGACCCATACGTGCTTATG-S '. The PCR products were run on an agarose gel and signals were quantified using the Labworks 4.0 program (UVP Biolmaging systems, Cambridge, United Kingdom). The intensity of each signal was normalized to the corresponding actin signal and expressed relative to mock control. Figure 8 shows the RT-PCR results with a maximal inhibition of hDMPK mRNA expression by PS58. The other gene transcripts carrying a naturally occurring small (CUG) repeat were not or only marginally affected by the oligonucleotide PS58, specific to the (CUG) repeat, compared to oligonucleotide PSl 13, which has no complementary sequence to these gene transcripts.
This example confirms the selectivity of an oligonucleotide, directed solely to the repeat region, towards the long repeat size (or disease causing expansion) compared to naturally occurring shorter repeat sizes.
Example 6 (Figure 9 en 10)
In this example, the DM500 immortomyoblast cell line carrying a human DMPK gene with an approximate (CTG)500 repeat expansion was cultured and transfected with PS58 (200 nM). Here, FISH analysis was carried out on the cells. Fourty eight hours after the start of the treatment, the cells were fixed with 4% formaldehyde, 5mM MgCh and Ix PBS for 30 minutes. Hybridization with fluorescently labeled oligonucleotide Cy3 -(CAG) 10-Cy3 was performed overnight at 37°C in a humid chamber. After hybridization the material was washed and mounted in mowiol and allowed to dry overnight. Nuclear inclusions (ribonuclear foci) were visualized using a Bio-Rad MRC 1024 confocal laser scanning microscope and LaserSharp2000 acquisition software. In total 50 cells were counted and scored for the presence of inclusions in the nuclei of these cells.
Literature indicates that DMPK mRNA containing a (CUG) expanded repeat accumulates and aggregates in the nucleus to form ribonuclear foci with regulatory nuclear proteins and transcription factors. Therefore, normal nuclear gene processing and cell function gets impaired.
Figure 9 shows a mock treated cell containing ribonuclear inclusions in the nucleus, while these are no longer present in the cell nucleus after treatment with PS58. Figure 10 shows that the percentage of nuclei containing ribonuclear foci seen under control conditions in DM500 myotubes is strongly decreased by the treatment with PS58. This result demonstrates that inhibition of hDMPK mRNA expression also inhibits the disease related triplet repeat (CUG) rich inclusions.
Example 7 (Figure 11) Here, the effect of PS58 was evaluated in vivo in DM500 mice containing hDMPK with a (CTG)n expansion of approximately 500 triplets. The DM500 mice were derived by somatic expansion from the DM300 mouse (e.g. see Gomes-Pereira M et al (2007) PLoS Genet. 2007 3(4): e52). A (CTG) triplet repeat expansion of approximately 500 was confirmed by southern blot and PCR analysis.
In short, PS58 was mixed with transfection agent ExGen 500 (Fermentas) according to the accompanying instructions for in vivo use. PS58 (2 nmol, in the transfection solution with Exgen 500) was injected (40μl) in the GPS complex of one-year-old DM500 mice and this procedure was repeated after 24h. As a control, DM500 mice were treated similarly with the transfection solution without PS58. After 15 days, the mice were sacrificed, muscles were isolated and total RNA was isolated from the tissues (using Trizol, Invitrogen). RT-PCR analysis was performed to detect hDMPK mRNA in the muscle similar as described above. The intensity of each band was performed using the Labworks 4.0 program (UVP Biolmaging systems, Cambridge, United Kingdom) and normalized to the intensity of the corresponding actin band. Primer location is indicated in the figure.
Figure 11 shows that in vivo treatment of DM500 mice with PS58 strongly reduced the presence of hDMPK mRNA containing a (CUG)n repeat expansion compared to mock treatment in the M. plantaris and M. gastrocnemius.
Example 8 (Figure 12)
In this example, different oligonucleotides (in length and backbone chemistry) but all with a sequence directed solely to the (CTG)n repeat expansion were compared. DM500 myotubes were cultured, transfected and analysed as described above in example 1. Northern blots were quantified by phosphoimager analysis and DMPK signals were normalized to GAPDH.
Here, the DM500 myotubes were treated with the following oligonucleotides (20OnM), all with a complete phosphorothioate backbone (see Table 3). Figure 12 shows that treatment of the DM500 myotubes results in a complete reduction of (CUG)n expanded hDMPK mRNA for all oligonucleotides tested. Under the present conditions, the maximal effect obtainable is independent of oligonucleotide length, backbone modification or potential mechanism of inhibition by the employed single stranded oligonucleotides. Example 9
Fibroblasts (GM 00305) from a male patient with Huntington's Disease were obtained from Coriell Cell Repository (Camden, New Jersey, US) and cultured according to the accompanying instructions and standard techniques known to the skilled person in the art. Huntington patients carry one healthy and one disease-causing allele of the Huntington gene resulting in the expression of both mRNAs with respectively a normal number and an expanded number of (CAG) repeats, respectively. The fibroblasts were transfected with a 21-mer 2'O-methyl phosphorothioate RNA antisense oligonucleotide PS57 with a (CUG)7 sequence, complementary to the (CAG) triplet repeat in Huntington mRNA. Transfection occurred at 100 or 200 nM in the presence of PEI as indicated by the manufacturer. Twenty four hours after transfection the cells were harvested and total RNA was isolated and analysed by RT-PCR. The Huntington transcript was determined using primers in downstream exon 64 (5 ' GAAAG TCAGT CCGGG TAGAA CTTC 3' and 5' CAGAT ACCCG CTCCA TAGCA A 3'). This method detects both types of Huntington mRNAs, the normal and mutant transcript with the additional (CAG) expansion. GAPDH mRNA (housekeeping gene) was also determined. The signals were quantified and the total amount of Huntington mRNA was normalised to the amount of GAPDH mRNA in the same sample. The results are expressed relative to a control treated (without oligonucleotide) sample from fibroblasts (which was to 100%).
In the samples from fibroblasts transfected with either 100 or 200 nM of PS57, significantly lower levels of total Huntington mRNA levels were observed of approximately 53% and 66% compared to the levels in control-treated cells, respectively.
Thus, PS57, an oligonucleotide directed only to the (CAG) repeat, induces a decrease in Huntington mRNA levels and these results are consistent with a selective inhibition of mutant over normal Huntington mRNA.
Table 1 : Overview oligonucleotides tested
Figure imgf000028_0001
Table 2. Reduction of hDMPK mRNA after oligo transfection:
Figure imgf000028_0002
(-) indicates no reduction, (+) indicates leve o re uction in hDMPK mRNA.
Table 3: Oligonucleotides used in example 9
Figure imgf000029_0001
all oligonucleotides full length phosphorothioate and substitution

Claims

Claims
1. Use of a single stranded oligonucleotide comprising or consisting of a sequence that is complementary only to a repetitive sequence in a gene transcript for the manufacture of a medicament for the treatment or prevention of human cw-element repeat instability associated genetic disorders.
2. The use according to claim 1 wherein the repetitive element is present in a coding sequence of the gene transcript.
3. The use according to claim 1 wherein the repetitive element is present in a non-coding sequence of the gene transcript.
4. The use according to any of the preceding claims wherein the oligonucleotide comprises or consists of a sequence that is complementary to a repetitive element selected from the group consisting of (CAG)n, (GCG)n, (CUG)n, (CGG)n, (CCG)n, (GAA)n, (GCC)n and (CCUG)n.
5. Use according to claim 2, wherein the oligonucleotide comprises or consists of a sequence that is complementary to a polyglutamine (CAG)n tract and wherein the repeat instability disorder is Huntington's disease, spinocerebellar ataxias, Haw River syndrome, X-linked spinal and bulbar muscular atrophy and/or dentatorubral-pallidoluysian atrophy.
6. Use according to claim 2, wherein the oligonucleotide comprises or consists of a sequence that is complementary to a polyalanine (GCG)n tract and wherein the repeat instability disorder is selected from the group consisting of infantile spasm syndrome, deidocranial dysplasia, blepharophimosis, hand-foot-genital disease, synpolydactyly, oculopharyngeal muscular dystrophy and/or holoprosencephaly.
7. Use according to claim 3, wherein the oligonucleotide comprises or consists of a sequence that is complementary to a (CUG)n repeat and wherein the repeat instability disorder is myotonic dystrophy type 1, spino-cerebrellar ataxia 8 and/or Huntington's disease-like 2.
8. Use according to claim 3 wherein the oligonucleotide comprises or consists of a sequence that is complementary to a (CCUG)n repeat and wherein the repeat instability disorder is myotonic dystrophy type 2.
9. Use according to claim 3, wherein the oligonucleotide comprises or consists of a sequence that is complementary to a (CGG)n repeat and wherein the repeat instability disorder is fragile X syndrome.
10. Use according to claim 3, wherein the oligonucleotide comprises or consists of a sequence that is complementary to a (GAA)n repeat and wherein the repeat instability disorder is Friedreich's ataxia.
11. Use according to any of the preceding claims wherein the oligonucleotide has a length of about 10 to about 50 nucleotides, preferably 12 to 30 nucleotides.
12. Use according any of the preceding claims wherein the oligonucleotide is comprised of RNA nucleotides, DNA nucleotides, locked nucleic acid (LNA) nucleotides, peptide nucleic acid (PNA) nucleotides, morpholino phosphorodiamidates, ethylene-bridged nucleic acid (ENA) nucleotides or mixtures thereof with or without a phosphorothioate containing backbone.
13. Use according to claim 12, wherein the oligonucleotide comprises T-O- substituted RNA phosphorothioate nucleotides.
14. Use according to any of the preceding claims wherein the oligonucleotide in the medicament is provided by a nucleic acid vector capable of conferring expression of the oligonucleotide.
15. Use according to any of the preceding claims, wherein the oligonucleotide in the medicament is provided with at least an excipient and/or targeting ligand for delivery of the oligonucleotide to cells and/or enhancing its intracellular delivery.
16. A single stranded oligonucleotide comprising or consisting of a sequence that is complementary to a repetitive sequence selected from the group consisting of (CAG)n, (GCG)n, (CUG)n, (CGG)n, (GAA)n, (GCC)n and (CCUG)n, having a length of 10 to 50 nucleotides.
17. An oligonucleotide according to claim 16, further comprising 2'-O- substituted RNA phosphorothioate nucleotides, DNA phosphorothioate nucleotides, locked nuclear acid (LNA) nucleotides or morpholino nucleotides and/or combinations thereof.
18. An oligonucleotide as defined in any one of claims 1 to 17, which is provided with a radioactive label or fluorescent label.
19. Pharmaceutically acceptable composition comprising an oligonucleotide as defined in any of claims 1 to 18.
20. Pharmaceutically acceptable composition of claim 19, further comprising at least one excipient and/or targeting ligand for delivery of the oligonucleotide to the cell and/or enhancing its intracellular delivery.
21. Nucleic acid vector, preferably a viral vector, capable of conferring expression of an oligonucleotide as defined in any of claims 1 to 18 in human cells.
22. Method for the reduction of repeat containing gene transcripts in a cell comprising the administration of an oligonucleotide as defined in any one of claims 1 to 18.
23. In vitro or ex vivo detection and/or diagnostic method wherein the oligonucleotide according to any of claims 17-18 is used.
PCT/NL2007/050399 2006-08-11 2007-08-10 Methods and means for treating dna repeat instability associated genetic disorders WO2008018795A1 (en)

Priority Applications (18)

Application Number Priority Date Filing Date Title
JP2009523740A JP6047270B2 (en) 2006-08-11 2007-08-10 Methods and means for treating genetic disorders associated with instability of DNA repeats
NZ574807A NZ574807A (en) 2006-08-11 2007-08-10 Methods and means for treating dna repeat instability associated genetic disorders
PL07808532T PL2049664T3 (en) 2006-08-11 2007-08-10 Single stranded oligonucleotides complementary to repetitive elements for treating DNA repeat instability associated genetic disorders
CA2660523A CA2660523C (en) 2006-08-11 2007-08-10 Methods and means for treating dna repeat instability associated genetic disorders
CN2007800298485A CN101501193B (en) 2006-08-11 2007-08-10 Methods and means for treating DNA repeat instability associated genetic disorders
US12/377,160 US20100184833A1 (en) 2006-08-11 2007-08-10 Methods and means for treating dna repeat instability associated genetic disorders
ES07808532T ES2373246T3 (en) 2006-08-11 2007-08-10 COMPLEMENTARY MONOCATENARY OLIGONUCLEOTIDES OF REPETITIVE ELEMENTS FOR THE TREATMENT OF GENETIC DISORDERS ASSOCIATED WITH THE INSTABILITY OF DNA REPETITIONS.
EP07808532A EP2049664B1 (en) 2006-08-11 2007-08-10 Single stranded oligonucleotides complementary to repetitive elements for treating DNA repeat instability associated genetic disorders
DK07808532.1T DK2049664T3 (en) 2006-08-11 2007-08-10 Single-stranded oligonucleotides, complementary to repetitive elements, for the treatment of DNA repetitive instability-associated disorders
AU2007282224A AU2007282224B2 (en) 2006-08-11 2007-08-10 Methods and means for treating DNA repeat instability associated genetic disorders
SI200730778T SI2049664T1 (en) 2006-08-11 2007-08-10 Single stranded oligonucleotides complementary to repetitive elements for treating DNA repeat instability associated genetic disorders
AT07808532T ATE524547T1 (en) 2006-08-11 2007-08-10 SINGLE STRANDED OLIGONUCLEOTIDES COMPLEMENTARY TO REPETITIVE ELEMENTS FOR THE TREATMENT OF DNA REPEATS-INSTABILITY-ASSOCIATED DISEASES
IL196921A IL196921A (en) 2006-08-11 2009-02-05 Single stranded oligonucleotides complementary to repetitive elements for treating dna repeat instability associated genetic disorders
HK09109656.7A HK1130510A1 (en) 2006-08-11 2009-10-20 Single stranded oligonucleotides complementary to repetitive elements for treating dna repeat instability associated genetic disorders
US14/809,483 US9890379B2 (en) 2006-08-11 2015-07-27 Treatment of genetic disorders associated with DNA repeat instability
US15/855,848 US10689646B2 (en) 2006-08-11 2017-12-27 Treatment of genetic disorders associated with DNA repeat instability
US16/660,387 US11274299B2 (en) 2006-08-11 2019-10-22 Methods and means for treating DNA repeat instability associated genetic disorders
US17/591,681 US20220267765A1 (en) 2006-08-11 2022-02-03 Treatment of genetic disorders associated with dna repeat instability

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
EP06118809.0 2006-08-11
EP06118809 2006-08-11
EP06119247 2006-08-21
EP06119247.2 2006-08-21

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US12/377,160 A-371-Of-International US20100184833A1 (en) 2006-08-11 2007-08-10 Methods and means for treating dna repeat instability associated genetic disorders
US14/809,483 Division US9890379B2 (en) 2006-08-11 2015-07-27 Treatment of genetic disorders associated with DNA repeat instability

Publications (1)

Publication Number Publication Date
WO2008018795A1 true WO2008018795A1 (en) 2008-02-14

Family

ID=38668759

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/NL2007/050399 WO2008018795A1 (en) 2006-08-11 2007-08-10 Methods and means for treating dna repeat instability associated genetic disorders

Country Status (16)

Country Link
US (5) US20100184833A1 (en)
EP (1) EP2049664B1 (en)
JP (2) JP6047270B2 (en)
AT (1) ATE524547T1 (en)
AU (1) AU2007282224B2 (en)
CA (1) CA2660523C (en)
CY (1) CY1111975T1 (en)
DK (1) DK2049664T3 (en)
ES (1) ES2373246T3 (en)
HK (1) HK1130510A1 (en)
IL (1) IL196921A (en)
NZ (1) NZ574807A (en)
PL (1) PL2049664T3 (en)
PT (1) PT2049664E (en)
SI (1) SI2049664T1 (en)
WO (1) WO2008018795A1 (en)

Cited By (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009099326A1 (en) * 2008-02-08 2009-08-13 Prosensa Holding Bv Methods and means for treating dna repeat instability associated genetic disorders
WO2010014592A1 (en) 2008-07-29 2010-02-04 The Board Of Regents Of The University Of Texas Sytem Selective inhibition of polyglutamine protein expression
WO2010115993A1 (en) * 2009-04-10 2010-10-14 Association Institut De Myologie Tricyclo-dna antisense oligonucleotides, compositions, and methods for the treatment of disease
WO2011097641A1 (en) * 2010-02-08 2011-08-11 Isis Pharmaceuticals, Inc. Methods and compositions useful in treatment of diseases or conditions related to repeat expansion
WO2011150408A2 (en) 2010-05-28 2011-12-01 Avi Biopharma, Inc. Oligonucleotide analogues having modified intersubunit linkages and/or terminal groups
WO2012012443A2 (en) 2010-07-19 2012-01-26 Bennett C Frank Modulation of dystrophia myotonica-protein kinase (dmpk) expression
WO2012018257A1 (en) * 2010-08-05 2012-02-09 Academisch Ziekenhuis Leiden H.O.D.N. Lumc Antisense oligonucleotide directed removal of proteolytic cleavage sites from proteins
WO2012138289A1 (en) 2011-04-08 2012-10-11 Zain-Luqman Rula Diagnosis and treatment of friedreich's ataxia
WO2012150960A1 (en) 2011-05-05 2012-11-08 Avi Biopharma, Inc. Peptide oligonucleotide conjugates
US8415465B2 (en) 2006-01-26 2013-04-09 Isis Pharmaceuticals, Inc. Compositions and their uses directed to huntingtin
WO2013142087A1 (en) 2012-03-20 2013-09-26 Sarepta Therapeutics, Inc. Boronic acid conjugates of oligonucleotide analogues
WO2013162363A1 (en) * 2012-04-23 2013-10-31 Prosensa Technologies B.V. RNA modulating oligonucleotides with improved characteristics for the treatment of neuromuscular disorders
US8609065B2 (en) 2007-07-12 2013-12-17 Prosensa Technologies B.V. Molecules for targeting compounds to various selected organs, tissues or tumor cells
US8906873B2 (en) 2009-09-11 2014-12-09 Isis Pharmaceuticals, Inc. Modulation of huntingtin expression
WO2015053624A3 (en) * 2013-10-07 2015-08-27 Academisch Ziekenhuis Leiden H.O.D.N. Lumc Antisense oligonucleotide directed removal of proteolytic cleavage sites, the hchwa-d mutation, and trinucleotide repeat expansions
US9139828B2 (en) 2008-05-14 2015-09-22 Prosensa Technologies B.V. Method for efficient exon (44) skipping in duchenne muscular dystrophy and associated means
US9249243B2 (en) 2005-07-13 2016-02-02 Sarepta Therapeutics, Inc. Antibacterial antisense oligonucleotide and method
US9278987B2 (en) 2011-11-18 2016-03-08 Sarepta Therapeutics, Inc. Functionally-modified oligonucleotides and subunits thereof
WO2016088797A1 (en) * 2014-12-02 2016-06-09 国立大学法人東京医科歯科大学 Nucleic acid reducing als-causing protein toxicity
US9499818B2 (en) 2007-10-26 2016-11-22 BioMarin Technologies, B.V. 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
EP3034083A3 (en) * 2006-09-21 2016-12-21 University of Rochester Antisense oligonucleotides for use in treating myotonic dystrophy
WO2017060317A1 (en) * 2015-10-05 2017-04-13 Proqr Therapeutics Ii B.V. Use of single-stranded antisense oligonucleotide in prevention or treatment of genetic diseases involving a trinucleotide repeat expansion
EP3033425A4 (en) * 2013-08-16 2017-07-26 Rana Therapeutics, Inc. Compositions and methods for modulating expression of frataxin
EP2812342B1 (en) 2012-02-08 2017-11-15 Ionis Pharmaceuticals, Inc. Modulation of rna by repeat targeting
US9890379B2 (en) 2006-08-11 2018-02-13 Biomarin Technologies B.V. Treatment of genetic disorders associated with DNA repeat instability
US9944926B2 (en) 2011-11-30 2018-04-17 Sarepta Therapeutics, Inc. Induced exon inclusion in spinal muscle atrophy
US9976138B2 (en) 2011-08-29 2018-05-22 Ionis Pharmaceuticals, Inc. Methods and compounds useful in conditions related to repeat expansion
US10017763B2 (en) 2010-09-03 2018-07-10 Sarepta Therapeutics, Inc. dsRNA molecules comprising oligonucleotide analogs having modified intersubunit linkages and/or terminal groups
US10041074B2 (en) 2013-08-16 2018-08-07 Translate Bio Ma, Inc. Euchromatic region targeting methods for modulating gene expression
US10066228B2 (en) 2011-11-30 2018-09-04 Sarepta Therapeutics, Inc. Oligonucleotides for treating expanded repeat diseases
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
US10202599B2 (en) 2011-08-11 2019-02-12 Ionis Pharmaceuticals, Inc. Selective antisense compounds and uses thereof
US10435430B2 (en) 2013-07-31 2019-10-08 Ionis Pharmaceuticals, Inc. Methods and compounds useful in conditions related to repeat expansion
US10533171B2 (en) 2009-04-24 2020-01-14 Biomarin Technologies B.V. Oligonucleotide comprising an inosine for treating DMD
US10563198B2 (en) 2015-03-11 2020-02-18 Wings Therapeutics, Inc. Oligonucleotides matching COL7A1 exon 73 for epidermolysis bullosa therapy
US10758558B2 (en) 2015-02-13 2020-09-01 Translate Bio Ma, Inc. Hybrid oligonucleotides and uses thereof
US10905709B2 (en) 2015-08-28 2021-02-02 Sarepta Therapeutics, Inc. Modified antisense oligomers for exon inclusion in spinal muscular atrophy
US10954519B2 (en) 2013-08-09 2021-03-23 lonis Pharmaceuticals, Inc. Compounds and methods for modulation of dystrophia myotonica-protein kinase (DMPK) expression
US11020417B2 (en) 2015-06-04 2021-06-01 Sarepta Therapeutics, Inc Methods and compounds for treatment of lymphocyte-related diseases and conditions
WO2021230286A1 (en) 2020-05-12 2021-11-18 田辺三菱製薬株式会社 Compound, method and pharmaceutical composition for regulating expression of ataxin 3
US11603530B2 (en) 2020-03-30 2023-03-14 Neubase Therapeutics, Inc. Modified peptide nucleic acid compositions
US11633498B2 (en) 2021-07-09 2023-04-25 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating myotonic dystrophy
US11679161B2 (en) 2021-07-09 2023-06-20 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating facioscapulohumeral muscular dystrophy
US11787869B2 (en) 2018-08-02 2023-10-17 Dyne Therapeutics, Inc. Methods of using muscle targeting complexes to deliver an oligonucleotide to a subject having facioscapulohumeral muscular dystrophy or a disease associated with muscle weakness
US11833221B2 (en) 2021-09-01 2023-12-05 Ionis Pharmaceuticals, Inc. Oligomeric compounds for reducing DMPK expression
US11911484B2 (en) 2018-08-02 2024-02-27 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating myotonic dystrophy
US11931421B2 (en) 2022-04-15 2024-03-19 Dyne Therapeutics, Inc. Muscle targeting complexes and formulations for treating myotonic dystrophy
US11981897B2 (en) 2021-02-10 2024-05-14 Ionis Pharmaceuticals, Inc. Compounds and methods for modulation of dystrophia myotonica-protein kinase (DMPK) expression

Families Citing this family (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK2351844T3 (en) 2003-04-29 2014-09-22 Sarepta Therapeutics Inc Preparations for enhancing transport and antisense efficiency of nucleic acid analog in cells
US20050288246A1 (en) 2004-05-24 2005-12-29 Iversen Patrick L Peptide conjugated, inosine-substituted antisense oligomer compound and method
US20100016215A1 (en) * 2007-06-29 2010-01-21 Avi Biopharma, Inc. Compound and method for treating myotonic dystrophy
US20110269665A1 (en) 2009-06-26 2011-11-03 Avi Biopharma, Inc. Compound and method for treating myotonic dystrophy
JP2014513946A (en) * 2011-04-22 2014-06-19 プロセンサ テクノロジーズ ビー.ブイ. Novel compounds for the treatment, delay and / or prevention of human genetic diseases such as myotonic dystrophy type 1 (DM1)
US9161948B2 (en) 2011-05-05 2015-10-20 Sarepta Therapeutics, Inc. Peptide oligonucleotide conjugates
JP6902869B2 (en) 2014-03-19 2021-07-14 アイオーニス ファーマシューティカルズ, インコーポレーテッドIonis Pharmaceuticals,Inc. Composition for regulating the expression of ataxin 2
WO2015143245A1 (en) * 2014-03-19 2015-09-24 Isis Pharmaceuticals, Inc. Methods for modulating ataxin 2 expression
MA45328A (en) 2016-04-01 2019-02-06 Avidity Biosciences Llc NUCLEIC ACID-POLYPEPTIDE COMPOSITIONS AND USES THEREOF
JP7236707B2 (en) 2016-08-12 2023-03-10 ザ・ホスピタル・フォー・シック・チルドレン Methods of treating diseases associated with repeat instability
HUE059843T2 (en) 2016-12-19 2023-01-28 Sarepta Therapeutics Inc Exon skipping oligomer conjugates for muscular dystrophy
JP2020505330A (en) 2017-01-06 2020-02-20 アビディティー バイオサイエンシーズ エルエルシー Nucleic acid polypeptide compositions and methods for inducing exon skipping
WO2018152523A1 (en) * 2017-02-20 2018-08-23 Northwestern University Use of trinucleotide repeat rnas to treat cancer
WO2018161032A1 (en) 2017-03-03 2018-09-07 The Regents Of The University Of California RNA TARGETING OF MUTATIONS VIA SUPPRESSOR tRNAs AND DEAMINASES
GB201711809D0 (en) 2017-07-21 2017-09-06 Governors Of The Univ Of Alberta Antisense oligonucleotide
WO2019113393A1 (en) 2017-12-06 2019-06-13 Avidity Biosciences Llc Compositions and methods of treating muscle atrophy and myotonic dystrophy
EP4257128A3 (en) * 2018-04-11 2023-11-15 Design Therapeutics, Inc. Methods and compounds for the treatment of genetic disease
AR115847A1 (en) 2018-07-25 2021-03-03 Ionis Pharmaceuticals Inc COMPOUNDS AND METHODS TO REDUCE THE EXPRESSION OF ATXN2
TW202039573A (en) 2018-12-21 2020-11-01 美商亞維代堤生物科學公司 Anti-transferrin receptor antibodies and uses thereof
AU2020321466A1 (en) 2019-07-26 2022-02-17 Proqr Therapeutics Ii B.V. Ophthalmic compositions comprising viscosifying polymers and nucleic acids
CA3159944A1 (en) 2019-12-02 2021-06-10 David HUSS Therapeutic editing
AU2021237465A1 (en) 2020-03-19 2022-10-13 Avidity Biosciences, Inc. Compositions and methods of treating Facioscapulohumeral muscular dystrophy
JP2023527638A (en) 2020-03-27 2023-06-30 アビディティー バイオサイエンシーズ,インク. Compositions and methods for treating facioscapulohumeral muscular dystrophy
EP4247392A1 (en) * 2020-06-03 2023-09-27 Takeda Pharmaceuticals U.S.A., Inc. Methods for the treatment of nucleotide repeat expansion disorders associated with msh3 activity
US11912779B2 (en) 2021-09-16 2024-02-27 Avidity Biosciences, Inc. Compositions and methods of treating facioscapulohumeral muscular dystrophy

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994026887A1 (en) 1993-05-11 1994-11-24 The University Of North Carolina At Chapel Hill Antisense oligonucleotides which combat aberrant splicing and methods of using the same
US6210892B1 (en) 1998-10-07 2001-04-03 Isis Pharmaceuticals, Inc. Alteration of cellular behavior by antisense modulation of mRNA processing
WO2002024906A1 (en) 2000-09-21 2002-03-28 Academisch Ziekenhuis Leiden Induction of exon skipping in eukaryotic cells
WO2003013437A2 (en) * 2001-08-07 2003-02-20 University Of Delaware Compositions and methods for the prevention and treatment of huntington's disease
WO2004083446A2 (en) 2003-03-21 2004-09-30 Academisch Ziekenhuis Leiden Modulation of exon recognition in pre-mrna by interfering with the secondary rna structure
CA2526893A1 (en) * 2003-05-14 2004-11-25 Japan Science And Technology Agency Inhibition of the expression of huntingtin gene
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)

Family Cites Families (144)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5034506A (en) 1985-03-15 1991-07-23 Anti-Gene Development Group Uncharged morpholino-based polymers having achiral intersubunit linkages
US5541308A (en) * 1986-11-24 1996-07-30 Gen-Probe Incorporated Nucleic acid probes for detection and/or quantitation of non-viral organisms
US5766847A (en) 1988-10-11 1998-06-16 Max-Planck-Gesellschaft Zur Forderung Der Wissenschaften E.V. Process for analyzing length polymorphisms in DNA regions
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
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.
JPH07501204A (en) 1991-06-28 1995-02-09 マサチューセッツ インスティテュート オブ テクノロジー Topical oligonucleotide therapy
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
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
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
EP0758403B1 (en) 1994-05-05 1998-06-24 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
US6300060B1 (en) 1995-11-09 2001-10-09 Dana-Farber Cancer Institute, Inc. Method for predicting the risk of prostate cancer morbidity and mortality
SK109198A3 (en) 1996-02-14 1999-06-11 Isis Pharmaceuticals Inc Sugar-modified gapped oligonucleotides
US6251589B1 (en) 1996-07-18 2001-06-26 Srl, Inc. Method for diagnosing spinocerebellar ataxia type 2 and primers therefor
WO1998018920A1 (en) 1996-10-30 1998-05-07 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
AU7265298A (en) 1997-04-29 1998-11-24 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
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
US6924355B2 (en) * 1998-09-01 2005-08-02 Genentech, Inc. PRO1343 polypeptides
US5945290A (en) 1998-09-18 1999-08-31 Isis Pharmaceuticals, Inc. Antisense modulation of RhoA expression
CA2343934A1 (en) 1998-09-25 2000-04-06 The Children's Medical Center Corporation Short peptides which selectively modulate the activity of protein kinases
US6172216B1 (en) * 1998-10-07 2001-01-09 Isis Pharmaceuticals Inc. Antisense modulation of BCL-X expression
US6399575B1 (en) * 1998-11-10 2002-06-04 Auburn University Methods and compositions for targeting compounds to the central nervous system
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
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
WO2000078813A2 (en) 1999-06-18 2000-12-28 Emory University Huntington disease cellular model: stably transfected pc12 cells expressing mutant huntingtin
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
WO2001079283A1 (en) 2000-04-13 2001-10-25 University Of British Columbia 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
CA2407309C (en) 2000-04-28 2011-08-02 Xiao Xiao Dna sequences encoding dystrophin minigenes and methods of use thereof
CA2407942A1 (en) 2000-05-01 2001-11-08 Hybridon, Inc. Modulation of oligonucleotide cpg-mediated immune stimulation by positional modification of nucleosides
US20020013287A1 (en) * 2000-05-09 2002-01-31 Reliable Biopharmaceuticals, Inc. St Louis Missouri Polymeric compounds useful as prodrugs
CA2410758A1 (en) * 2000-05-31 2001-12-06 Genset S.A. Use of acrp30 globular head to promote increases in muscle mass and muscle differentiation
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
US6727355B2 (en) 2000-08-25 2004-04-27 Jcr Pharmaceuticals Co., Ltd. Pharmaceutical composition for treatment of Duchenne muscular dystrophy
AU2001296600A1 (en) 2000-10-06 2002-04-15 Regents Of The University Of Michigan Mini-dystrophin nucleic acid and peptide sequences
AU2002248224B2 (en) 2000-10-27 2006-11-09 Baylor College Of Medicine Methods and compositions for the identification and treatment of neurodegenerative disorders
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
CA2414782C (en) * 2001-05-11 2012-10-09 Regents Of The University Of Minnesota Intron associated with myotonic dystrophy type 2 and methods of use
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)
EP1627061B1 (en) 2001-05-18 2009-08-12 Sirna Therapeutics, Inc. RNA INTERFERENCE MEDIATED INHIBITION OF GENE EXPRESSION USING CHEMICALLY MODIFIED 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
DK1406667T3 (en) 2001-07-06 2008-06-16 Topigen Pharmaceuticals Inc Methods to increase in vivo efficacy of oligonucleotides and inhibit mammalian inflammation
WO2003014145A2 (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
AU2002363253A1 (en) 2001-11-01 2003-05-12 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
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)
WO2003074551A2 (en) 2002-03-01 2003-09-12 The Administrators Of The Tulane Educational Fund Conjugates of therapeutic or cytotoxic 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.
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
EP1585560A4 (en) 2002-07-26 2011-03-16 Mirus Bio Corp 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
CA2493297A1 (en) 2002-08-12 2004-02-19 Universite De Sherbrooke Methods to reprogram splice site selection in pre-messenger rnas
GB0219143D0 (en) 2002-08-16 2002-09-25 Univ Leicester Modified tailed oligonucleotides
WO2004037854A1 (en) 2002-10-23 2004-05-06 Centre For Research And Technology Hellas/Institute Of Agrobiotechnology In.A Prion protein-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
EP2314691A3 (en) * 2002-11-14 2012-01-18 Dharmacon, Inc. Fuctional and hyperfunctional siRNA
CA2796924C (en) 2002-11-25 2016-12-13 Nonprofit Organization Translational Research Organization Of Duchenne Muscular Dystrophy Ena nucleic acid pharmaceuticals capable of modifying splicing of mrna precursors
GB0228079D0 (en) 2002-12-02 2003-01-08 Laxdale Ltd Huntington's Disease
CA2518475C (en) * 2003-03-07 2014-12-23 Alnylam Pharmaceuticals, Inc. Irna agents comprising asymmetrical modifications
US7514551B2 (en) * 2003-04-03 2009-04-07 Enzo Life Sciences, Inc. Multisignal labeling reagents, and processes and uses therefor
EP1635870A2 (en) 2003-06-02 2006-03-22 Wyeth Use of myostatin (gdf8) inhibitors in conjunction with corticosteroids for treating neuromuscular disorders
DK1495769T3 (en) 2003-07-11 2008-06-23 Lbr Medbiotech B V Mannose-6-phosphate receptor-mediated gene transfer to muscle cells
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
JP2007508030A (en) 2003-10-14 2007-04-05 カーネル・バイオファーマ・インコーポレイテッド Two-phase PNA conjugate for delivering PNA across the blood brain barrier
US20050191636A1 (en) 2004-03-01 2005-09-01 Biocept, Inc. Detection of STRP, such as fragile X syndrome
WO2005086768A2 (en) 2004-03-11 2005-09-22 Albert Einstein College Of Medicine Of Yeshiva University 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
CA2567810A1 (en) 2004-05-27 2005-12-08 Acceleron Pharma Inc. Cerberus/coco derivatives and uses thereof
US7807816B2 (en) * 2004-06-28 2010-10-05 University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
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
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
US20120122801A1 (en) 2005-01-05 2012-05-17 Prosensa B.V. Mannose-6-phosphate receptor mediated gene transfer into muscle cells
US20060148740A1 (en) * 2005-01-05 2006-07-06 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
US20090312532A1 (en) 2005-04-22 2009-12-17 Van Deutekom Judith Christina Modulation of exon recognition in pre-mrna by interfering with the binding of sr proteins and by interfering with secodary rna structure
US7906617B2 (en) * 2005-12-15 2011-03-15 E. I. Du Pont De Nemours And Company Polyethylene binding peptides and methods of use
EP2428227B1 (en) 2006-01-26 2016-06-22 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
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
AU2007282224B2 (en) 2006-08-11 2013-08-29 Vico Therapeutics B.V. Methods and means for treating DNA repeat instability associated genetic disorders
WO2009008727A2 (en) 2007-07-12 2009-01-15 Prosensa Technologies B.V. Molecules for targeting compounds to various selected organs or tissues
AU2008273094B2 (en) 2007-07-12 2013-05-09 Prosensa Technologies B.V. Molecules for targeting compounds to various selected organs, tissues or tumor cells
AU2008317566B2 (en) 2007-10-26 2014-05-01 Academisch Ziekenhuis Leiden Means and methods for counteracting muscle disorders
NZ587178A (en) 2008-02-08 2011-11-25 Prosensa Holding Bv Methods and means for treating dna repeat instability associated genetic disorders
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
WO2010006237A2 (en) 2008-07-11 2010-01-14 Alnylam Pharmaceuticals, Inc. Phosphorothioate oligonucleotides non-nucleosidic phosphorothiotes as delivery agents for irna agents
EP2317847B1 (en) 2008-07-29 2019-04-17 The Board of Regents of The University of Texas System Selective inhibition of polyglutamine protein expression
US20110294870A1 (en) 2008-12-04 2011-12-01 Opko Curna, Llc Treatment of tumor suppressor gene related diseases by inhibition of natural antisense transcript to the gene
US20120046342A1 (en) 2009-04-24 2012-02-23 Prosensa Technologies B.V. Oligonucleotide comprising an inosine for treating dmd
SG176716A1 (en) 2009-06-08 2012-01-30 Miragen Therapeutics CHEMICAL MODIFICATION MOTIFS FOR miRNA INHIBITORS AND MIMETICS
EP2516647B1 (en) 2009-12-24 2016-12-14 BioMarin Technologies B.V. Molecule for treating an inflammatory disorder
US20130059902A1 (en) 2010-02-08 2013-03-07 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
EP3633038A3 (en) 2010-07-19 2020-07-29 Ionis Pharmaceuticals, Inc. Modulation of dystrophia myotonica-protein kinase (dmpk) expression
EP2605794B1 (en) 2010-08-20 2016-07-20 Replicor Inc. Oligonucleotide chelate complexes
EP2673361B1 (en) 2011-02-08 2016-04-13 Ionis Pharmaceuticals, Inc. Oligomeric compounds comprising bicyclic nucleotides and uses thereof
JP2014513946A (en) 2011-04-22 2014-06-19 プロセンサ テクノロジーズ ビー.ブイ. Novel compounds for the treatment, delay and / or prevention of human genetic diseases such as myotonic dystrophy type 1 (DM1)
KR102183273B1 (en) 2011-05-05 2020-11-27 사렙타 쎄러퓨틱스, 인코퍼레이티드 Peptide Oligonucleotide conjugates
ES2832531T3 (en) 2011-11-30 2021-06-10 Sarepta Therapeutics Inc Oligonucleotides for the treatment of repeat expansion diseases
BR112014018427B1 (en) 2012-01-27 2021-11-03 Biomarin Technologies B.V. RNA MODULATOR OLIGONUCLEOTIDES WITH IMPROVED FEATURES FOR THE TREATMENT OF DUCHENNE AND BECKER'S MUSCULAR DYSTROPHY
EP3330278A1 (en) 2012-02-08 2018-06-06 Ionis Pharmaceuticals, Inc. Modulation of rna by repeat targeting
CN104271741A (en) 2012-04-23 2015-01-07 普罗森萨科技有限公司 RNA modulating oligonucleotides with improved characteristics for the treatment of neuromuscular disorders
JP6460983B2 (en) 2012-07-03 2019-01-30 バイオマリン テクノロジーズ ベー.フェー. Oligonucleotides for the treatment of patients with muscular dystrophy

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994026887A1 (en) 1993-05-11 1994-11-24 The University Of North Carolina At Chapel Hill Antisense oligonucleotides which combat aberrant splicing and methods of using the same
US6210892B1 (en) 1998-10-07 2001-04-03 Isis Pharmaceuticals, Inc. Alteration of cellular behavior by antisense modulation of mRNA processing
WO2002024906A1 (en) 2000-09-21 2002-03-28 Academisch Ziekenhuis Leiden Induction of exon skipping in eukaryotic cells
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)
WO2003013437A2 (en) * 2001-08-07 2003-02-20 University Of Delaware Compositions and methods for the prevention and treatment of huntington's disease
US20030109476A1 (en) * 2001-08-07 2003-06-12 Kmiec Eric B. Compositions and methods for the prevention and treatment of Huntington's disease
WO2004083446A2 (en) 2003-03-21 2004-09-30 Academisch Ziekenhuis Leiden Modulation of exon recognition in pre-mrna by interfering with the secondary rna structure
CA2526893A1 (en) * 2003-05-14 2004-11-25 Japan Science And Technology Agency Inhibition of the expression of huntingtin gene
WO2004101787A1 (en) * 2003-05-14 2004-11-25 Japan Science And Technology Agency Inhibition of the expression of huntington gene

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
BURNETT RYAN ET AL: "DNA sequence-specific polyamides alleviate transcription inhibition associated with long GAA.TTC repeats in Friedreich's ataxia.", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA 1 AUG 2006, vol. 103, no. 31, 1 August 2006 (2006-08-01), pages 11497 - 11502, XP002458876, ISSN: 0027-8424 *
CAPLEN N J ET AL: "Rescue of polyglutamine-mediated cytotoxicity by double-stranded RNA-mediated RNA interference", HUMAN MOLECULAR GENETICS, OXFORD UNIVERSITY PRESS, SURREY, GB, vol. 11, no. 2, 15 January 2002 (2002-01-15), pages 175 - 184, XP002380705, ISSN: 0964-6906 *
FURLING D ET AL: "Viral vector producing antisense RNA restores myotonic dystrophy myoblast functions.", GENE THERAPY, vol. 10, no. 9, May 2003 (2003-05-01), pages 795 - 802, XP002423924, ISSN: 0969-7128 *
HASHOLT LIS ET AL: "Antisense downregulation of mutant huntingtin in a cell model.", THE JOURNAL OF GENE MEDICINE JUN 2003, vol. 5, no. 6, June 2003 (2003-06-01), pages 528 - 538, XP002423925, ISSN: 1099-498X *
LANGLOIS MARC-ANDRÉ ET AL: "Hammerhead ribozyme-mediated destruction of nuclear foci in myotonic dystrophy myoblasts.", MOLECULAR THERAPY : THE JOURNAL OF THE AMERICAN SOCIETY OF GENE THERAPY MAY 2003, vol. 7, no. 5 Pt 1, May 2003 (2003-05-01), pages 670 - 680, XP002423926, ISSN: 1525-0016 *
LIU W ET AL: "Specific inhibition of Huntington's disease gene expression by siRNAs in cultures cells", PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES, TOKYO, JP, vol. 79B, no. 10, December 2003 (2003-12-01), pages 293 - 298, XP002983007, ISSN: 0386-2208 *
YEN L ET AL: "SEQUENCE-SPECIFIC CLEAVAGE OF HUNTINGTIN MRNA BY CATALYTIC DNA", ANNALS OF NEUROLOGY, BOSTON, US, vol. 46, no. 3, September 1999 (1999-09-01), pages 366 - 373, XP008031730, ISSN: 0364-5134 *

Cited By (126)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9249243B2 (en) 2005-07-13 2016-02-02 Sarepta Therapeutics, Inc. Antibacterial antisense oligonucleotide and method
US10738307B2 (en) 2006-01-26 2020-08-11 Ionis Pharmaceuticals, Inc. Compositions and their uses directed to huntingtin
US9353372B2 (en) 2006-01-26 2016-05-31 Ionis Pharmaceuticals, Inc. Compositions and their uses directed to huntingtin
US9057066B2 (en) 2006-01-26 2015-06-16 Isis Pharmaceuticals, Inc. Compositions and their uses directed to huntingtin
US8952145B2 (en) 2006-01-26 2015-02-10 Isis Pharmaceuticals, Inc. Compositions and their uses directed to huntingtin
US8415465B2 (en) 2006-01-26 2013-04-09 Isis Pharmaceuticals, Inc. Compositions and their uses directed to huntingtin
US11274299B2 (en) 2006-08-11 2022-03-15 Vico Therapeutics B.V. Methods and means for treating DNA repeat instability associated genetic disorders
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
EP3034083A3 (en) * 2006-09-21 2016-12-21 University of Rochester Antisense oligonucleotides for use in treating myotonic dystrophy
US8609065B2 (en) 2007-07-12 2013-12-17 Prosensa Technologies B.V. Molecules for targeting compounds to various selected organs, tissues or tumor cells
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
US9926557B2 (en) 2007-10-26 2018-03-27 Biomarin Technologies B.V. Methods and means for efficient skipping of exon 45 in Duchenne muscular dystrophy pre-mRNA
US9499818B2 (en) 2007-10-26 2016-11-22 BioMarin Technologies, B.V. 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
WO2009099326A1 (en) * 2008-02-08 2009-08-13 Prosensa Holding Bv Methods and means for treating dna repeat instability associated genetic disorders
JP2011510678A (en) * 2008-02-08 2011-04-07 プロセンサ ホールディング ビーブイ Methods and means for treating DNA repeat instability related genetic disorders
US9139828B2 (en) 2008-05-14 2015-09-22 Prosensa Technologies B.V. Method for efficient exon (44) skipping in duchenne muscular dystrophy and associated means
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
US9340785B2 (en) 2008-07-29 2016-05-17 The Board Of Regents Of The University Of Texas System Selective inhibition of polyglutamine protein expression
EP2317847A1 (en) * 2008-07-29 2011-05-11 The Board of Regents of The University of Texas System Selective inhibition of polyglutamine protein expression
JP2015171368A (en) * 2008-07-29 2015-10-01 ザ ボード オブ リージェンツ オブ ザ ユニバーシティー オブ テキサス システム selective inhibition of polyglutamine protein expression
JP2011529686A (en) * 2008-07-29 2011-12-15 ザ ボード オブ リージェンツ オブ ザ ユニバーシティー オブ テキサス システム Selective inhibition of polyglutamine protein expression
WO2010014592A1 (en) 2008-07-29 2010-02-04 The Board Of Regents Of The University Of Texas Sytem Selective inhibition of polyglutamine protein expression
AU2009276763B2 (en) * 2008-07-29 2015-07-16 The Board Of Regents Of The University Of Texas Sytem Selective inhibition of polyglutamine protein expression
EP2317847A4 (en) * 2008-07-29 2014-10-15 Univ Texas Selective inhibition of polyglutamine protein expression
US8901095B2 (en) 2008-07-29 2014-12-02 The Board Of Regents Of The University Of Texas System Selective inhibition of polyglutamine protein expression
WO2010115993A1 (en) * 2009-04-10 2010-10-14 Association Institut De Myologie Tricyclo-dna antisense oligonucleotides, compositions, and methods for the treatment of disease
JP2019055990A (en) * 2009-04-10 2019-04-11 アソシアシオン・アンスティテュ・ドゥ・ミオロジーAssociation Institut De Myologie Tricyclo-dna antisense oligonucleotides, compositions and methods for treating disease
US10465191B2 (en) 2009-04-10 2019-11-05 Association Institut De Myologie Tricyclo-DNA antisense oligonucleotides, compositions, and methods for the treatment of disease
JP2016027823A (en) * 2009-04-10 2016-02-25 アソシアシオン・アンスティテュ・ドゥ・ミオロジーAssociation Institut De Myologie Tricyclo-dna antisense oligonucleotides, compositions, and methods for the treatment of disease
JP2012523225A (en) * 2009-04-10 2012-10-04 アソシアシオン・アンスティテュ・ドゥ・ミオロジー Tricyclo-DNA antisense oligonucleotides, compositions and methods for treatment of disease
US11034956B2 (en) 2009-04-24 2021-06-15 Biomarin Technologies B.V. Oligonucleotide comprising an inosine for treating DMD
US10533171B2 (en) 2009-04-24 2020-01-14 Biomarin Technologies B.V. Oligonucleotide comprising an inosine for treating DMD
US11634714B2 (en) 2009-04-24 2023-04-25 Biomarin Technologies B.V. Oligonucleotide comprising an inosine for treating DMD
US10202603B2 (en) 2009-09-11 2019-02-12 Ionis Pharmaceuticals, Inc. Modulation of huntingtin expression
US9273315B2 (en) 2009-09-11 2016-03-01 Ionis Pharmaceuticals, Inc. Modulation of huntingtin expression
US8906873B2 (en) 2009-09-11 2014-12-09 Isis Pharmaceuticals, Inc. Modulation of huntingtin expression
US11421231B2 (en) 2009-09-11 2022-08-23 Ionis Pharmaceuticals, Inc. Modulation of Huntington expression
US10619158B2 (en) 2009-09-11 2020-04-14 Ionis Pharmaceuticals, Inc. Modulation of huntingtin expression
US10837016B2 (en) 2009-09-11 2020-11-17 Ionis Pharmaceuticals, Inc. Modulation of huntingtin expression
WO2011097641A1 (en) * 2010-02-08 2011-08-11 Isis Pharmaceuticals, Inc. Methods and compositions useful in treatment of diseases or conditions related to repeat expansion
WO2011150408A2 (en) 2010-05-28 2011-12-01 Avi Biopharma, Inc. Oligonucleotide analogues having modified intersubunit linkages and/or terminal groups
US8779128B2 (en) 2010-05-28 2014-07-15 Sarepta Therapeutics, Inc. Oligonucleotide analogues having modified intersubunit linkages and/or terminal groups
US10202602B2 (en) 2010-05-28 2019-02-12 Sarepta Therapeutics, Inc. Oligonucleotide analogues having modified intersubunit linkages and/or terminal groups
US10760078B2 (en) 2010-05-28 2020-09-01 Sarepta Therapeutics, Inc. Oligonucleotide analogues having modified intersubunit linkages and/or terminal groups
US9469664B2 (en) 2010-05-28 2016-10-18 Sarepta Therapeutics, Inc. Oligonucleotide analogues having modified intersubunit linkages and/or terminal groups
AU2011282243B2 (en) * 2010-07-19 2016-09-22 Ionis Pharmaceuticals, Inc. Modulation of nuclear-retained RNA
EP3031920B1 (en) 2010-07-19 2019-08-21 Ionis Pharmaceuticals, Inc. Modulation of dystrophia myotonica-protein kinase (dmpk) expression
WO2012012443A2 (en) 2010-07-19 2012-01-26 Bennett C Frank Modulation of dystrophia myotonica-protein kinase (dmpk) expression
EP3633038A3 (en) * 2010-07-19 2020-07-29 Ionis Pharmaceuticals, Inc. Modulation of dystrophia myotonica-protein kinase (dmpk) expression
EP3031920A1 (en) * 2010-07-19 2016-06-15 Ionis Pharmaceuticals, Inc. Modulation of dystrophia myotonica-protein kinase (dmpk) expression
US9765338B2 (en) 2010-07-19 2017-09-19 Ionis Pharmaceuticals, Inc. Modulation of dystrophia myotonica-protein kinase (DMPK) expression
EP2595664A2 (en) * 2010-07-19 2013-05-29 Isis Pharmaceuticals, Inc. Modulation of nuclear-retained rna
EP2595663A2 (en) * 2010-07-19 2013-05-29 Isis Pharmaceuticals, Inc. Modulation of dystrophia myotonica-protein kinase (dmpk) expression
EP3489360A3 (en) * 2010-07-19 2019-08-28 Ionis Pharmaceuticals, Inc. Modulation of nuclear-retained rna
AU2016269447C1 (en) * 2010-07-19 2019-06-20 Ionis Pharmaceuticals, Inc. Modulation of nuclear-retained rna
EP2595663A4 (en) * 2010-07-19 2014-03-05 Isis Pharmaceuticals Inc Modulation of dystrophia myotonica-protein kinase (dmpk) expression
AU2016269447B2 (en) * 2010-07-19 2018-12-13 Ionis Pharmaceuticals, Inc. Modulation of nuclear-retained rna
KR101900770B1 (en) * 2010-07-19 2018-09-20 아이오니스 파마수티컬즈, 인코포레이티드 Modulation of dystrophia myotonica-protein kinase (dmpk) expression
EP2595664A4 (en) * 2010-07-19 2014-09-17 Isis Pharmaceuticals Inc Modulation of nuclear-retained rna
US9611471B2 (en) 2010-08-05 2017-04-04 Academisch Ziekenhuis Leiden Antisense oligonucleotide directed removal of proteolytic cleavage sites from proteins
US10364432B2 (en) 2010-08-05 2019-07-30 Academisch Ziekenhuis Leiden H.O.D.N. Lumc Antisense oligonucleotide directed removal of proteolytic cleavage sites from proteins
US20140039037A1 (en) * 2010-08-05 2014-02-06 Academisch Ziekenhuis Leiden H.O.D.N. Lumc Antisense oligonucleotide directed removal of proteolytic cleavage sites, the hchwa-d mutation, and trinucleotide repeat expansions
WO2012018257A1 (en) * 2010-08-05 2012-02-09 Academisch Ziekenhuis Leiden H.O.D.N. Lumc Antisense oligonucleotide directed removal of proteolytic cleavage sites from proteins
US10590421B2 (en) 2010-08-05 2020-03-17 Academisch Ziekenhuis Leiden H.O.D.N. Lumc Antisense oligonucleotide directed removal of proteolytic cleavage sites, the HCHWA-D mutation, and trinucleotide repeat expansions
US20130198877A1 (en) * 2010-08-05 2013-08-01 Academisch Ziekenhuis Leiden H.O.D.N. Lumc Antisense oligonucleotide directed removal of proteolytic cleavage sites from proteins
US10017763B2 (en) 2010-09-03 2018-07-10 Sarepta Therapeutics, Inc. dsRNA molecules comprising oligonucleotide analogs having modified intersubunit linkages and/or terminal groups
US11072793B2 (en) 2010-09-03 2021-07-27 Sarepta Therapeutics, Inc. DsRNA molecules comprising oligonucleotide analogs having modified intersubunit linkages and/or terminal groups
US9476043B2 (en) 2011-04-08 2016-10-25 Rula Zain-Luqman Diagnosis and treatment of friedreich's ataxia
WO2012138289A1 (en) 2011-04-08 2012-10-11 Zain-Luqman Rula Diagnosis and treatment of friedreich's ataxia
EP2707486A1 (en) * 2011-04-08 2014-03-19 Zain-Luqman, Rula Diagnosis and treatment of friedreich's ataxia
EP2707486A4 (en) * 2011-04-08 2015-02-18 Zain Luqman Rula Diagnosis and treatment of friedreich's ataxia
US10443053B2 (en) * 2011-04-08 2019-10-15 Rula Zain-Luqman Diagnosis and treatment of Friedreich's ataxia
WO2012150960A1 (en) 2011-05-05 2012-11-08 Avi Biopharma, Inc. Peptide oligonucleotide conjugates
US11732261B2 (en) 2011-08-11 2023-08-22 Ionis Pharmaceuticals, Inc. Selective antisense compounds and uses thereof
US10202599B2 (en) 2011-08-11 2019-02-12 Ionis Pharmaceuticals, Inc. Selective antisense compounds and uses thereof
US9976138B2 (en) 2011-08-29 2018-05-22 Ionis Pharmaceuticals, Inc. Methods and compounds useful in conditions related to repeat expansion
US9790499B2 (en) 2011-11-18 2017-10-17 Sarepta Therapeutics, Inc. Functionally-modified oligonucleotides and subunits thereof
US10344281B2 (en) 2011-11-18 2019-07-09 Sarepta Therapeutics, Inc. Functionally-modified oligonucleotides and subunits thereof
US9278987B2 (en) 2011-11-18 2016-03-08 Sarepta Therapeutics, Inc. Functionally-modified oligonucleotides and subunits thereof
US11208655B2 (en) 2011-11-18 2021-12-28 Sarepta Therapeutics, Inc. Functionally-modified oligonucleotides and subunits thereof
US11674139B2 (en) 2011-11-30 2023-06-13 Sarepta Therapeutics, Inc. Oligonucleotides for treating expanded repeat diseases
US10577605B2 (en) 2011-11-30 2020-03-03 Sarepta Therapeutics, Inc. Induced exon inclusion in spinal muscle atrophy
US9944926B2 (en) 2011-11-30 2018-04-17 Sarepta Therapeutics, Inc. Induced exon inclusion in spinal muscle atrophy
US10066228B2 (en) 2011-11-30 2018-09-04 Sarepta Therapeutics, Inc. Oligonucleotides for treating expanded repeat diseases
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
EP2812342B1 (en) 2012-02-08 2017-11-15 Ionis Pharmaceuticals, Inc. Modulation of rna by repeat targeting
WO2013142087A1 (en) 2012-03-20 2013-09-26 Sarepta Therapeutics, Inc. Boronic acid conjugates of oligonucleotide analogues
AU2013253074C1 (en) * 2012-04-23 2018-06-14 Biomarin Technologies B.V. RNA modulating oligonucleotides with improved characteristics for the treatment of neuromuscular disorders
AU2013253074B2 (en) * 2012-04-23 2018-02-22 Biomarin Technologies B.V. RNA modulating oligonucleotides with improved characteristics for the treatment of neuromuscular disorders
EP3604536A1 (en) * 2012-04-23 2020-02-05 BioMarin Technologies B.V. Rna modulating oligonucleotides with improved characteristics for the treatment of neuromuscular disorders
WO2013162363A1 (en) * 2012-04-23 2013-10-31 Prosensa Technologies B.V. RNA modulating oligonucleotides with improved characteristics for the treatment of neuromuscular disorders
US11345915B2 (en) 2012-04-23 2022-05-31 Vico Therapeutics B.V. RNA modulating oligonucleotides with improved characteristics for the treatment of neuromuscular disorders
US9745576B2 (en) 2012-04-23 2017-08-29 Biomarin Technologies B.V. RNA modulating oligonucleotides with improved characteristics for the treatment of neuromuscular disorders
EP3228712A1 (en) * 2012-04-23 2017-10-11 BioMarin Technologies B.V. Rna modulating oligonucleotides with improved characteristics for the treatment of neuromuscular disorders
US10435430B2 (en) 2013-07-31 2019-10-08 Ionis Pharmaceuticals, Inc. Methods and compounds useful in conditions related to repeat expansion
US10954519B2 (en) 2013-08-09 2021-03-23 lonis Pharmaceuticals, Inc. Compounds and methods for modulation of dystrophia myotonica-protein kinase (DMPK) expression
US10041074B2 (en) 2013-08-16 2018-08-07 Translate Bio Ma, Inc. Euchromatic region targeting methods for modulating gene expression
EP3033425A4 (en) * 2013-08-16 2017-07-26 Rana Therapeutics, Inc. Compositions and methods for modulating expression of frataxin
WO2015053624A3 (en) * 2013-10-07 2015-08-27 Academisch Ziekenhuis Leiden H.O.D.N. Lumc Antisense oligonucleotide directed removal of proteolytic cleavage sites, the hchwa-d mutation, and trinucleotide repeat expansions
WO2016088797A1 (en) * 2014-12-02 2016-06-09 国立大学法人東京医科歯科大学 Nucleic acid reducing als-causing protein toxicity
US10758558B2 (en) 2015-02-13 2020-09-01 Translate Bio Ma, Inc. Hybrid oligonucleotides and uses thereof
US11352626B2 (en) 2015-03-11 2022-06-07 Wings Therapeutics, Inc. Oligonucleotides matching COL7A1 exon 73 for epidermolysis bullosa therapy
US10563198B2 (en) 2015-03-11 2020-02-18 Wings Therapeutics, Inc. Oligonucleotides matching COL7A1 exon 73 for epidermolysis bullosa therapy
US11020417B2 (en) 2015-06-04 2021-06-01 Sarepta Therapeutics, Inc Methods and compounds for treatment of lymphocyte-related diseases and conditions
US10905709B2 (en) 2015-08-28 2021-02-02 Sarepta Therapeutics, Inc. Modified antisense oligomers for exon inclusion in spinal muscular atrophy
US20180282724A1 (en) * 2015-10-05 2018-10-04 Proqr Therapeutics Ii B.V. Use of single-stranded antisense oligonucleotide in prevention or treatment of genetic diseases involving a trinucleotide repeat expansion
CN108291225A (en) * 2015-10-05 2018-07-17 ProQR治疗上市公司Ⅱ The purposes being related in the genetic disease of trinucleotide repeat disease is being prevented or treated to single stranded antisense oligonucleotides
US10760076B2 (en) * 2015-10-05 2020-09-01 Proqr Therapeutics Ii B.V. Use of single-stranded antisense oligonucleotide in prevention or treatment of genetic diseases involving a trinucleotide repeat expansion
WO2017060317A1 (en) * 2015-10-05 2017-04-13 Proqr Therapeutics Ii B.V. Use of single-stranded antisense oligonucleotide in prevention or treatment of genetic diseases involving a trinucleotide repeat expansion
CN108291225B (en) * 2015-10-05 2022-07-12 ProQR治疗上市公司Ⅱ Use of single-stranded antisense oligonucleotides for preventing or treating genetic diseases involving repeated amplification of trinucleotide
AU2016335032B2 (en) * 2015-10-05 2022-04-14 Proqr Therapeutics Ii B.V. Use of single-stranded antisense oligonucleotide in prevention or treatment of genetic diseases involving a trinucleotide repeat expansion
US11787869B2 (en) 2018-08-02 2023-10-17 Dyne Therapeutics, Inc. Methods of using muscle targeting complexes to deliver an oligonucleotide to a subject having facioscapulohumeral muscular dystrophy or a disease associated with muscle weakness
US11795234B2 (en) 2018-08-02 2023-10-24 Dyne Therapeutics, Inc. Methods of producing muscle-targeting complexes comprising an anti-transferrin receptor antibody linked to an oligonucleotide
US11795233B2 (en) 2018-08-02 2023-10-24 Dyne Therapeutics, Inc. Muscle-targeting complex comprising an anti-transferrin receptor antibody linked to an oligonucleotide
US11911484B2 (en) 2018-08-02 2024-02-27 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating myotonic dystrophy
US11603530B2 (en) 2020-03-30 2023-03-14 Neubase Therapeutics, Inc. Modified peptide nucleic acid compositions
WO2021230286A1 (en) 2020-05-12 2021-11-18 田辺三菱製薬株式会社 Compound, method and pharmaceutical composition for regulating expression of ataxin 3
US11981897B2 (en) 2021-02-10 2024-05-14 Ionis Pharmaceuticals, Inc. Compounds and methods for modulation of dystrophia myotonica-protein kinase (DMPK) expression
US11633498B2 (en) 2021-07-09 2023-04-25 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating myotonic dystrophy
US11679161B2 (en) 2021-07-09 2023-06-20 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating facioscapulohumeral muscular dystrophy
US11844843B2 (en) 2021-07-09 2023-12-19 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating facioscapulohumeral muscular dystrophy
US11833221B2 (en) 2021-09-01 2023-12-05 Ionis Pharmaceuticals, Inc. Oligomeric compounds for reducing DMPK expression
US11931421B2 (en) 2022-04-15 2024-03-19 Dyne Therapeutics, Inc. Muscle targeting complexes and formulations for treating myotonic dystrophy

Also Published As

Publication number Publication date
PL2049664T3 (en) 2012-02-29
US10689646B2 (en) 2020-06-23
HK1130510A1 (en) 2009-12-31
ATE524547T1 (en) 2011-09-15
JP5934408B2 (en) 2016-06-15
DK2049664T3 (en) 2012-01-02
US20160053254A1 (en) 2016-02-25
ES2373246T3 (en) 2012-02-01
IL196921A0 (en) 2011-08-01
JP2010500023A (en) 2010-01-07
EP2049664A1 (en) 2009-04-22
CA2660523C (en) 2019-03-19
CA2660523A1 (en) 2008-02-14
AU2007282224B2 (en) 2013-08-29
US20180112214A1 (en) 2018-04-26
US11274299B2 (en) 2022-03-15
PT2049664E (en) 2012-01-03
AU2007282224A1 (en) 2008-02-14
US20100184833A1 (en) 2010-07-22
US20200140857A1 (en) 2020-05-07
IL196921A (en) 2013-11-28
NZ574807A (en) 2011-01-28
SI2049664T1 (en) 2012-04-30
JP2015157813A (en) 2015-09-03
CY1111975T1 (en) 2015-11-04
EP2049664B1 (en) 2011-09-14
US9890379B2 (en) 2018-02-13
JP6047270B2 (en) 2016-12-21
US20220267765A1 (en) 2022-08-25

Similar Documents

Publication Publication Date Title
US11274299B2 (en) Methods and means for treating DNA repeat instability associated genetic disorders
US8263760B2 (en) Methods and means for treating DNA repeat instability associated genetic disorders
US11781135B2 (en) Methods for treating Alzheimer's disease
JP6265940B2 (en) Selective inhibition of polyglutamine protein expression
EP2344637A1 (en) Methods and means for efficient skipping of exon 45 in duchenne muscular dystrophy pre-mrna
JP2012147790A (en) Means and method for inducing exon-skipping
WO2011097388A1 (en) Selective inhibition of polyglutamine protein expression
CN101501193B (en) Methods and means for treating DNA repeat instability associated genetic disorders
CA3020487A1 (en) Treatment of idiopathic pulmonary fibrosis using rna complexes that target connective tissue growth factor
JP2021533762A (en) Modified oligonucleotides that target SNPs
JP2023543494A (en) Treatment of optic atrophy

Legal Events

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

Ref document number: 200780029848.5

Country of ref document: CN

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

Ref document number: 07808532

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 196921

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2009523740

Country of ref document: JP

Ref document number: 2007808532

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2660523

Country of ref document: CA

Ref document number: 574807

Country of ref document: NZ

Ref document number: 554/KOLNP/2009

Country of ref document: IN

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2007282224

Country of ref document: AU

ENP Entry into the national phase

Ref document number: 2007282224

Country of ref document: AU

Date of ref document: 20070810

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: RU

WWE Wipo information: entry into national phase

Ref document number: 12377160

Country of ref document: US