EP3824086A1 - Antisense-oligonukleotide zur wiederherstellung aberranter spleissung von abca4 - Google Patents

Antisense-oligonukleotide zur wiederherstellung aberranter spleissung von abca4

Info

Publication number
EP3824086A1
EP3824086A1 EP19732376.9A EP19732376A EP3824086A1 EP 3824086 A1 EP3824086 A1 EP 3824086A1 EP 19732376 A EP19732376 A EP 19732376A EP 3824086 A1 EP3824086 A1 EP 3824086A1
Authority
EP
European Patent Office
Prior art keywords
abca4
seq
molecule
exon
nucleotides
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP19732376.9A
Other languages
English (en)
French (fr)
Inventor
Robert Wilhelmus Johanna COLLIN
Alejandro GARANTO IGLESIAS
Franciscus Peter Maria CREMERS
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Stichting Radboud Universitair Medisch Centrum
Original Assignee
Stichting Katholieke Universiteit
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
Application filed by Stichting Katholieke Universiteit filed Critical Stichting Katholieke Universiteit
Publication of EP3824086A1 publication Critical patent/EP3824086A1/de
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • 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
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/33Alteration of splicing

Definitions

  • Antisense oligonucleotides rescue aberrant splicing of ABCA4.
  • the invention relates to the fields of medicine and immunology.
  • it relates to novel antisense oligonucleotides that may be used in the treatment, prevention and/or delay of an ABCA4- associated condition.
  • ABCA4 Autosomal recessive mutations in ABCA4 cause Stargardt disease, a progressive disorder characterized by central vision loss and often leading to complete blindness.
  • a typical hallmark of Stargardt disease is the presence of many yellow spots (flecks) distributed throughout the fundus of the patients.
  • the ABCA4 gene is comprised of 50 exons and encodes a protein consisting of 2273 amino acids. This protein is expressed in the outer segments of cone and rod photoreceptor cells and plays an important role in the removal of waste products following phototransduction.
  • variants in ABCA4 can also lead to other subtypes of retinal disease ranging from bull’s eye maculopathy to autosomal recessive cone-rod dystrophy (arCRD; Cremers et al, 1998; Maugeri et al, 2000) and pan-retinal dystrophies (Cremers et al, 1998; Martinez-Mir et al, 1998), depending on the severity of the alleles.
  • arCRD autosomal recessive cone-rod dystrophy
  • pan-retinal dystrophies Cremers et al, 1998; Martinez-Mir et al, 1998), depending on the severity of the alleles.
  • B ⁇ a ⁇ e ⁇ c ABCA4 variants can be identified in approximately 80% of the cases with STGD1 (Allikmets et al, 1997; Fujinami et al, 2013; Lewis et al, 1999; Maugeri et al, 1999; Rivera et al, 2000; Schulz et al, 2017; Webster et al, 2001 ; Zernant et al, 201 1 ; Zernant et al, 2017), and 30% of cases with arCRD (Maugeri et al, 2000), after sequencing coding regions and flanking splice sites.
  • the invention provides for an ABCA4 exon 28 retention molecule that binds to and/or is complementary to a polynucleotide with the nucleotide sequence as shown in SEQ ID NO:4, preferably the molecule is complementary to a polynucleotide with SEQ ID NO:5, more preferably the molecule is complementary to a polynucleotide with SEQ ID NO:6.
  • the invention provides for an antisense oligonucleotide ABCA4 exon 28 retention molecule wherein the antisense oligonucleotide comprises or consists of a sequence selected from the group consisting of SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, and SEQ ID NO: 21 , preferably the antisense oligonucleotide comprises or consist of SEQ ID NO: 19.
  • the invention provide for a set of ABCA4 exon 28 retention molecules comprising at least two antisense oligonucleotide according to the invention, wherein preferably the set comprises or consists of at least two antisense oligonucleotides selected from the group consisting of SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, and SEQ ID NO: 21.
  • the invention provides for a viral vector expressing an ABCA4 exon 28 retention molecule as defined herein.
  • the invention provides for a ABCA4 exon 28 retention molecule as defined herein use as a medicament, preferably for use as a medicament for treating an ABCA4 related disease or a condition requiring modulating splicing of ABCA4 or for use in a method of treating an ABCA4 related disease or a condition requiring modulating splicing of ABCA4.
  • antisense oligonucleotides are substantially complementary (antisense) to their target, allowing them to bind to the corresponding pre-mRNA molecule, thereby, without being bound by theory, preventing the binding of proteins essential for splicing.
  • this lack of binding results in the skipping of the targeted exon, as the present inventors have previously shown for the c.4539+1 100A>G, c.4539+1 106C>T, c.4539+2001 G>A and c.4539+2028C>T mutations in ABCA4 (W02018/10901 1 ).
  • AONs can also be employed to force the inclusion of exons that due to mutations are absent in the mutant transcripts of the corresponding gene.
  • the present inventors have identified one ABCA4 variant, c.4253+43G>A, that is very common in the population, present in -0.4% of all individuals worldwide. Although this mutation is not fully penetrant (not all individuals with this mutation will get STGD1 ), it is one of the most common mutations in patients with STGD1 , in particularly those with a relatively late-onset.
  • the inventors have demonstrated that, in patients with this mutation, the amount of ABCA4 transcripts that contain exon 28 is lower compared to control individuals, indicating that this mutation results in a partial skipping of exon 28 from the ABCA4 mRNA. Occasionally, this process is accompanied by co- skipping of adjacent exons such as exon 27 or exon 29.
  • specific AONs are able to redirect the aberrant splicing of ABCA4 Vnat is caused by the intronic c.4253+43G>A mutation leading to the inclusion of exon 28.
  • exon retention is herein defined as inducing, producing or increasing production within a cell of a mature mRNA that does retain a particular exon that should be present in the mature mRNA without (aberrant) exon skipping. Exon retention is achieved by providing a cell expressing the pre-mRNA of said mature mRNA with an AON molecule capable of interfering with sequences such as, for example, alternative splice sites upstream or downstream of the regular splice sites.
  • An AON according to the invention is said to induce exon retention if the retention percentage as measured, preferably by real-time quantitative RT-PCR analysis, is at least 30%, or at least 35%, or at least 40%, or at least 45%, or at least 50%, or at least 55%, or at least 60%, or at least 65%, or at least 70%, or at least 75%, or at least 80%, or at least 85%, or at least 90%, or at least 95%, or 100% of mature mRNA.
  • the complementary regions are preferably designed such that, when combined, they are specific for the exon in the pre-mRNA. Such specificity may be created with various lengths of complementary regions, as this depends on the actual sequences in other (pre-)mRNA molecules in the system. The risk that the AON will also be able to hybridize to one or more other pre-mRNA molecules decreases with increasing size of the AON. It is clear that AONs comprising mismatches in the region of complementarity but that retain the capacity to hybridize and/or bind to the targeted region(s) in the pre-mRNA, can be used in the invention.
  • At least the complementary parts do not comprise such mismatches as AONs lacking mismatches in the complementary part typically have a higher efficiency and a higher specificity than AONs having such mismatches in one or more complementary regions. It is thought, that higher hybridization strengths, (i.e. increasing number of interactions with the opposing strand) are favorable in increasing the efficiency of the process of interfering with the splicing machinery of the system.
  • An AON according to the invention preferably does not contain a stretch of CpG, more preferably does not contain any CpG.
  • the presence of a CpG or a stretch of CpG in an oligonucleotide is usually associated with an increased immunogenicity of said oligonucleotide (Dorn and Kippenberger, 2008). This increased immunogenicity is undesired since it may induce damage of the tissue to be treated, i.e. the eye. Immunogenicity may be assessed in an animal model by assessing the presence of CD4+ and/or CD8+ cells and/or inflammatory mononucleocyte infiltration.
  • Immunogenicity may also be assessed in blood of an animal or of a human being treated with an AON according to the invention by detecting the presence of a neutralizing antibody and/or an antibody recognizing said AON using a standard immunoassay known to the skilled person.
  • An inflammatory reaction, type l-like interferon production, IL-12 production and/or an increase in immunogenicity may be assessed by detecting the presence or an increasing amount of a neutralizing antibody or an antibody recognizing said AON using a standard immunoassay.
  • the AON according to the invention furthermore preferably has acceptable RNA binding kinetics and/or thermodynamic properties.
  • RNA binding kinetics and/or thermodynamic properties are at least in part determined by the melting temperature of an oligonucleotide (Tm; calculated with the oligonucleotide properties calculator (www. unc. edu/-cail/biotool/oligo/index) for single stranded RNA using the basic Tm and the nearest neighbor model), and/or the free energy of the AON-target exon complex (using RNA structure version 4.5). If a Tm is too high, an AON is expected to be less specific. An acceptable Tm and free energy depend on the sequence of the AON. Therefore, it is difficult to give preferred ranges for each of these parameters.
  • Tm oligonucleotide
  • An acceptable Tm may be ranged between 35 and 70 °C and an acceptable free energy may be ranged between 15 and 45 kcal/mol.
  • the skilled person may therefore first choose an AON as a potential therapeutic compound as binding and/or being complementary to SEQ ID NO: 4, or a part thereof as defined later herein. The skilled person may check that said AON is able to bind to said sequences as earlier defined herein. Optionally in a second step, he may use the invention to further optimize said AON by checking for the absence of CpG and/or by optimizing its Tm and/or free energy of the AON-target complex.
  • He may try to design an AON wherein few, preferably, no CpG and/or wherein a more acceptable Tm and/or free energy are obtained by choosing a distinct sequence of ABCA4 (including SEQ ID NO: 4 or a part thereof) to which the AON is complementary.
  • a distinct sequence of ABCA4 including SEQ ID NO: 4 or a part thereof
  • the skilled person may improve any of these parameters by decreasing the length of the AON, and/or by choosing a distinct stretch within any of SEQ ID NO: 4 to which the AON is complementary and/or by altering the chemistry of the AON.
  • An AON for redirecting splicing according to the invention may comprise one of more RNA residue (ribonucleotide), or one or more DNA residue (deoxyribonucleotide), and/or one or more nucleotide analogues or equivalents, as will be further detailed herein below.
  • the terms“aberrant ABCA4 exon 28” of “aberrant exon 28” are considered to be synonymous, and considered to mean the presence of a mutation in exon 28 (SEQ ID NO:4) of the ABCA4 gene, resulting in an ABC4A mRNA with at least one substitution, deletion or insertion in exon 28 of ABCA4 (SEQ ID NO:4).
  • an exon retention molecule is preferably a nucleic acid molecule, preferably an oligonucleotide.
  • an exon retention molecule according to the invention is a nucleic acid molecule, preferably an oligonucleotide, which is complementary or substantially complementary to a nucleotide sequence as shown in in SEQ ID NO:4, preferably the molecule is complementary to a polynucleotide with SEQ ID NO:5, more preferably the molecule is complementary to a polynucleotide with SEQ ID NO:6 or a part thereof of ABCA4 .
  • a preferred ABCA4 exon 28 retention molecule is a nucleic acid molecule that comprises or consists of an antisense oligonucleotide that is complementary to the polynucleotide wherein the antisense oligonucleotide has a length from about 8 to about 100 nucleotides, more preferred 10 to about 40 nucleotides, more preferred from about 12 to about 30 nucleotides, more preferred from about 14 to about 28 nucleotides, nucleotides, most preferred about 20 to 24 nucleotides, such as 20 nucleotides, 21 nucleotides, 22 nucleotides, 23 nucleotides, 24 nucleotides or 25 nucleotides.
  • the invention provides a ABCA4 exon 28 retention molecule, wherein the antisense oligonucleotide comprises or consists of a sequence selected from the group consisting of SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, and SEQ ID NO: 21 , preferably the antisense oligonucleotide comprises or consist of SEQ ID NO: 19.
  • the antisense oligonucleotide comprises or consists of a sequence selected from the group consisting of SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, and SEQ ID NO: 21
  • the antisense oligonucleotide comprises or consist of SEQ ID NO: 19
  • the antisense oligonucleotide has a length of 8 to 100 nucleotides, preferably a length of 10 to 40 nucleotides, more preferably a length of 12 to 30 nucleotides, most preferably the retention molecule has a length of 18 to 24 nucleotides, such as 18, 19, 20, 21 , 22, 23 or 24 nucleotides.
  • the invention provides for an ABCA4 retention molecule comprising or preferably consisting of the AON SEQ ID NO: 19. It was found that this molecule was very efficient in redirecting the aberrant splicing of ABCA4 that is caused by the intronic c.4253+43G>A.
  • This preferred exon retention molecule of the invention comprising SEQ ID NO: 19, preferably comprises from about 8 to about 100 nucleotides, more preferred 10 to about 40 nucleotides, more preferred from about 12 to about 30 nucleotides, more preferred from about 14 to about 28 nucleotides, nucleotides, most preferred about 20 to 24 nucleotides, such as 20 nucleotides, 21 nucleotides, 22 nucleotides, 23 nucleotides, 24 nucleotides or 25 nucleotides
  • an AON for redirecting splicing according to the invention comprises one or more residues that are modified to increase nuclease resistance, and/or to increase the affinity of the antisense oligonucleotide for the target sequence. Therefore, in a preferred embodiment, the AON comprises at least one nucleotide analogue or equivalent, wherein a nucleotide analogue or equivalent is defined as a residue having a modified base, and/or a modified backbone, and/or a non-natural internucleoside linkage, or a combination of these modifications.
  • the nucleotide analogue or equivalent comprises a modified backbone.
  • backbones are provided by morpholino backbones, carbamate backbones, siloxane backbones, sulfide, sulfoxide and sulfone backbones, formacetyl and thioformacetyl backbones, methyleneformacetyl backbones, riboacetyl backbones, alkene containing backbones, sulfamate, sulfonate and sulfonamide backbones, methyleneimino and methylenehydrazino backbones, and amide backbones.
  • Phosphorodiamidate morpholino oligomers are modified backbone oligonucleotides that have previously been investigated as antisense agents.
  • Morpholino oligonucleotides have an uncharged backbone in which the deoxyribose sugar of DNA is replaced by a six membered ring and the phosphodiester linkage is replaced by a phosphorodiamidate linkage. Morpholino oligonucleotides are resistant to enzymatic degradation and appear to function as antisense agents by arresting translation or interfering with pre-mRNA splicing rather than by activating RNase H.
  • Morpholino oligonucleotides have been successfully delivered to tissue culture cells by methods that physically disrupt the cell membrane, and one study comparing several of these methods found that scrape loading was the most efficient method of delivery; however, because the morpholino backbone is uncharged, cationic lipids are not effective mediators of morpholino oligonucleotide uptake in cells.
  • a recent report demonstrated triplex formation by a morpholino oligonucleotide and, because of the non-ionic backbone, these studies showed that the morpholino oligonucleotide was capable of triplex formation in the absence of magnesium.
  • the linkage between the residues in a backbone do not include a phosphorus atom, such as a linkage that is formed by short chain alkyl or cycloalkyl internucleoside linkages, mixed heteroatom and alkyl or cycloalkyl internucleoside linkages, or one or more short chain heteroatomic or heterocyclic internucleoside linkages.
  • a further preferred backbone comprises a morpholino nucleotide analog or equivalent, in which the ribose or deoxyribose sugar is replaced by a 6-membered morpholino ring.
  • a most preferred nucleotide analog or equivalent comprises a phosphorodiamidate morpholino oligomer (PMO), in which the ribose or deoxyribose sugar is replaced by a 6-membered morpholino ring, and the anionic phosphodiester linkage between adjacent morpholino rings is replaced by a non-ionic phosphorodiamidate linkage.
  • PMO phosphorodiamidate morpholino oligomer
  • a nucleotide analogue or equivalent according to the invention comprises a substitution of one of the non-bridging oxygens in the phosphodiester linkage. This modification slightly destabilizes base-pairing but adds significant resistance to nuclease degradation.
  • a preferred nucleotide analogue or equivalent comprises phosphorothioate, chiral phosphorothioate, phosphorodithioate, phosphotriester, aminoalkylphosphotriester, H- phosphonate, methyl and other alkyl phosphonate including 3'-alkylene phosphonate, 5'-alkylene phosphonate and chiral phosphonate, phosphinate, phosphoramidate including 3'-amino phosphoramidate and aminoalkylphosphoramidate, thionophosphoramidate, thionoalkylphosphonate, thionoalkylphosphotriester, selenophosphate or boranophosphate.
  • a further preferred nucleotide analogue or equivalent according to the invention comprises one or more sugar moieties that are mono- or disubstituted at the 2', 3' and/or 5' position such as a -OH; - F; substituted or unsubstituted, linear or branched lower (CI-C10) alkyl, alkenyl, alkynyl, alkaryl, allyl, or aralkyl, that may be interrupted by one or more heteroatoms; 0-, S-, or N-alkyl; 0-, S-, or N-alkenyl; 0-, S-or N-alkynyl; 0-, S-, or N- allyl; O-alkyl-O-alkyl, -methoxy, -aminopropoxy; methoxyethoxy; dimethylaminooxyethoxy; and -dimethylaminoethoxyethoxy.
  • sugar moieties that are mono- or disubstituted
  • the sugar moiety can be a pyranose or derivative thereof, or a deoxypyranose or derivative thereof, preferably ribose or derivative thereof, or deoxyribose or derivative of.
  • a preferred derivatized sugar moiety comprises a Locked Nucleic Acid (LNA), in which the 2'-carbon atom is linked to the 3' or 4' carbon atom of the sugar ring thereby forming a bicyclic sugar moiety.
  • LNA Locked Nucleic Acid
  • a preferred LNA comprises 2'-0, 4'-C- ethylene-bridged nucleic acid (Morita et al., 2001 ). These substitutions render the nucleotide analogue or equivalent RNase H and nuclease resistant and increase the affinity for the target RNA.
  • a nucleotide analogue or equivalent according to the invention comprises one or more base modifications or substitutions.
  • Modified bases comprise synthetic and natural bases such as inosine, xanthine, hypoxanthine and other -aza, deaza, -hydroxy, -halo, -thio, thiol, -alkyl, -alkenyl, -alkynyl, thioalkyl derivatives of pyrimidine and purine bases that are or will be known in the art.
  • an AON has at least two different types of analogues or equivalents.
  • an ABCA4 exon 28 retention molecule comprises a 2'-0 alkyl phosphorothioate antisense oligonucleotide, such as 2'-0-methyl modified ribose (RNA), 2'-0-ethyl modified ribose, 2'-0-propyl modified ribose, and/or substituted derivatives of these modifications such as halogenated derivatives.
  • RNA 2'-0-methyl modified ribose
  • RNA 2'-0-ethyl modified ribose
  • 2'-0-propyl modified ribose substituted derivatives of these modifications such as halogenated derivatives.
  • the invention comprises a set of ABCA4 exon 28 retention molecules comprising at least two ABCA4 exon 28 retention molecules according to the invention.
  • set comprises or consists of at least two, at least three, at least four antisense oligonucleotides selected from the group consisting of SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, and SEQ ID NO: 21.
  • such set may comprise or consist of:
  • antisense oligonucleotide SEQ ID NO: 19 and antisense oligonucleotide SEQ ID NO: 21 , or
  • antisense oligonucleotide SEQ ID NO: 20 antisense oligonucleotide SEQ ID NO: 21 .
  • Stargardt disease (STGD1 ) usually has an autosomal recessive inheritance caused by biallelic ABCA4 variants. It is thus entirely plausible that patients can carry several ABCA4 mutations thereby requiring a combination of AONs for the treatment of the disease. Accordingly, the invention provides for a composition comprising one or more of the AONs of the invention and optionally, one or more AONs targeting other mutations in ABCA4. For example, the invention provides for a composition comprising an AON of the present invention and an AON described in W02018/10901 1.
  • An AON for redirecting splicing according to the invention may be indirectly administrated using suitable means known in the art. It may for example be provided to an individual or a cell, tissue or organ of said individual as such, as a so-called‘naked’ AON. It may also be administered in the form of an expression vector wherein the expression vector encodes an RNA transcript comprising the sequence of said AON according to the invention.
  • the expression vector is preferably introduced into a cell, tissue, organ or individual via a gene delivery vehicle.
  • a viral-based expression vector comprising an expression cassette or a transcription cassette that drives expression or transcription of an AON for redirecting splicing according to the invention. Accordingly, the invention provides for a viral vector expressing an antisense oligonucleotide according to the invention when placed under conditions conducive to expression of the molecule.
  • a cell can be provided with an AON for redirecting splicing according to the invention by plasmid- derived antisense oligonucleotide expression or viral expression provided by adenovirus- or adeno- associated virus-based vectors. Expression may be driven by an RNA polymerase II promoter (Pol II) such as a U7 RNA promoter or an RNA polymerase III (Pol III) promoter, such as a U6 RNA promoter.
  • RNA polymerase II promoter such as a U7 RNA promoter or an RNA polymerase III (Pol III) promoter, such as a U6 RNA promoter.
  • a preferred delivery vehicle is a viral vector such as an adeno-associated virus vector (AAV), or a retroviral vector such as a lentivirus vector and the like.
  • plasmids artificial chromosomes, plasmids usable for targeted homologous recombination and integration in the human genome of cells may be suitably applied for delivery of an AON for redirecting splicing according to the invention.
  • Preferred for the invention are those vectors wherein transcription is driven from Poll II promoters, and/or wherein transcripts are in the form fusions with U1 or U7 transcripts, which yield good results for delivering small transcripts. It is within the skill of the artisan to design suitable transcripts.
  • Pollll driven transcripts preferably, in the form of a fusion transcript with an U1 or U7 transcript. Such fusions may be generated as previously described (Gorman et al., 1998).
  • the invention accordingly provides for a viral-based vector, comprising a Pol II or a Pol III promoter driven expression cassette for expression of an AON for redirecting splicing according to the invention.
  • a protein shell may also be named a capsid protein shell.
  • AAV vector may have one or preferably all wild type AAV genes deleted, but may still comprise functional ITR nucleic acid sequences. Functional ITR sequences are necessary for the replication, rescue and packaging of AAV virions.
  • the ITR sequences may be wild type sequences or may have at least 80%, 85%, 90%, 95, or 100% sequence identity with wild type sequences or may be altered by for example in insertion, mutation, deletion or substitution of nucleotides, as long as they remain functional.
  • functionality refers to the ability to direct packaging of the genome into the capsid shell and then allow for expression in the host cell to be infected or target cell.
  • a capsid protein shell may be of a different serotype than the AAV vector genome ITR.
  • An AAV vector according to present the invention may thus be composed of a capsid protein shell, i.e. the icosahedral capsid, which comprises capsid proteins (VP1 , VP2, and/or VP3) of one AAV serotype, e.g. AAV serotype 2, whereas the ITRs sequences contained in that AAV5 vector may be any of the AAV serotypes described above, including an AAV2 vector.
  • An “AAV2 vector” thus comprises a capsid protein shell of AAV serotype 2
  • e.g. an“AAV5 vector” comprises a capsid protein shell of AAV serotype 5, whereby either may encapsidate any AAV vector genome ITR according to the invention.
  • a recombinant AAV vector according to the invention comprises a capsid protein shell of AAV serotype 2, 5, 8 or AAV serotype 9 wherein the AAV genome or ITRs present in said AAV vector are derived from AAV serotype 2, 5, 8 or AAV serotype 9; such AAV vector is referred to as an AAV2/2, AAV 2/5, AAV2/8, AAV2/9, AAV5/2, AAV5/5, AAV5/8, AAV 5/9, AAV8/2, AAV 8/5, AAV8/8, AAV8/9, AAV9/2, AAV9/5, AAV9/8, or an AAV9/9 vector.
  • a recombinant AAV vector according to the invention comprises a capsid protein shell of AAV serotype 2 and the AAV genome or ITRs present in said vector are derived from AAV serotype 5; such vector is referred to as an AAV 2/5 vector.
  • a recombinant AAV vector according to the invention comprises a capsid protein shell of AAV serotype 2 and the AAV genome or ITRs present in said vector are derived from AAV serotype 9; such vector is referred to as an AAV 2/9 vector.
  • a recombinant AAV vector according to the invention comprises a capsid protein shell of AAV serotype 2 and the AAV genome or ITRs present in said vector are derived from AAV serotype 2; such vector is referred to as an AAV 2/2 vector.
  • a nucleic acid molecule encoding an AON for redirecting splicing according to the invention represented by a nucleic acid sequence of choice is preferably inserted between the AAV genome or ITR sequences as identified above, for example an expression construct comprising an expression regulatory element operably linked to a coding sequence and a 3’ termination sequence.
  • AAV helper functions generally refers to the corresponding AAV functions required for AAV replication and packaging supplied to the AAV vector in trans.
  • AAV helper functions complement the AAV functions which are missing in the AAV vector, but they lack AAV ITRs (which are provided by the AAV vector genome).
  • AAV helper functions include the two major ORFs of AAV, namely the rep coding region and the cap coding region or functional substantially identical sequences thereof.
  • AAV helper virus provides additional functions required for AAV replication and packaging.
  • Suitable AAV helper viruses include adenoviruses, herpes simplex viruses (such as HSV types 1 and 2) and vaccinia viruses.
  • the additional functions provided by the helper virus can also be introduced into the host cell via vectors, as described in US 6,531 ,456 incorporated herein by reference.
  • an AAV genome as present in a recombinant AAV vector according to the invention does not comprise any nucleotide sequences encoding viral proteins, such as the rep (replication) or cap (capsid) genes of AAV.
  • An AAV genome may further comprise a marker or reporter gene, such as a gene for example encoding an antibiotic resistance gene, a fluorescent protein (e.g. gfp) or a gene encoding a chemically, enzymatically or otherwise detectable and/or selectable product (e.g. lacZ, aph, etc.) known in the art.
  • an AAV vector according to the invention is constructed and produced according to the method according to ISITA et al., 2016 which is herein incorporated by reference.
  • a preferred AAV vector according to the invention is an AAV vector, preferably an AAV2/5, AAV2/8, AAV2/9 or AAV2/2 vector, expressing an AON for redirecting splicing according to the invention that is an AON that comprises, or preferably consists of, a sequence that is:
  • nucleotide sequence consisting of SEQ ID NO:4 preferably the molecule is complementary to a polynucleotide with SEQ ID NO:5, more preferably the molecule is complementary to a polynucleotide with SEQ ID NO:6;
  • a polynucleotide with a nucleotide sequence selected from the group consisting of SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, and SEQ ID NO: 21.
  • An AON for redirecting splicing according to the invention can be delivered as such as a ‘naked’ AON to an individual, a cell, tissue or organ of said individual.
  • the molecule is dissolved in a solution that is compatible with the delivery method.
  • Retina cells can be provided with a plasmid for antisense oligonucleotide expression by providing the plasmid in an aqueous solution.
  • a preferred delivery method for an AON for redirecting splicing or a plasmid for expression of such AON is a viral vector or are nanoparticles.
  • viral vectors or nanoparticles are delivered to retina or other relevant cells.
  • Such delivery to retina cells or other relevant cells may be in vivo, in vitro or ex vivo; see e.g., ISo et al, 2016, which is herein incorporated by reference.
  • a plasmid can be provided by transfection using known transfection agents.
  • the solution is a physiological salt solution.
  • an excipient or transfection agents that will aid in delivery of each of the constituents as defined herein to a cell and/or into a cell, preferably a retina cell.
  • excipients or transfection agents capable of forming complexes, nanoparticles, micelles, vesicles and/or liposomes that deliver each constituent as defined herein, complexed or trapped in a vesicle or liposome through a cell membrane. Many of these excipients are known in the art.
  • Suitable excipients or transfection agentia comprise polyethylenimine (PEI; ExGen500 (MBI Fermentas)), LipofectAMINETM 2000 (Invitrogen) or derivatives thereof, or similar cationic polymers, including polypropyleneimine or polyethylenimine copolymers (PECs) and derivatives, synthetic amphiphils (SAINT-18), lipofectinTM, DOTAP and/or viral capsid proteins that are capable of self-assembly into particles that can deliver each constitutent as defined herein to a cell, preferably a retina cell.
  • Such excipients have been shown to efficiently deliver an oligonucleotide such as AONs to a wide variety of cultured cells, including retina cells. Their high transfection potential is combined with an excepted low to moderate toxicity in terms of overall cell survival. The ease of structural modification can be used to allow further modifications and the analysis of their further (in vivo) nucleic acid transfer characteristics and toxicity.
  • Polycations such as 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 each constituent as defined herein, preferably an AON according to the invention, across cell membranes into cells.
  • PBCA butylcyanoacrylate
  • PHCA hexylcyanoacrylate
  • the cationic peptide protamine offers an alternative approach to formulate an oligonucleotide with 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 an oligonucleotide.
  • the skilled person may select and adapt any of the above or other commercially available alternative excipients and delivery systems to package and deliver an exon retention molecule for use in the current invention to deliver it for the prevention, treatment or delay of ABCA4-related disease or condition.
  • Prevention, treatment or delay of an ABCA4- related disease or condition is herein preferably defined as preventing, halting, ceasing the progression of, or reversing partial or complete visual impairment or blindness that is caused by a genetic defect in the ABCA4 gene.
  • an AON for redirecting splicing according to the invention could be covalently or non- covalently linked to a targeting ligand specifically designed to facilitate the uptake into the cell, cytoplasm and/or its nucleus.
  • a targeting ligand specifically designed to facilitate the uptake into the cell, cytoplasm and/or its nucleus.
  • ligand could comprise (i) a compound (including but not limited to peptide(-like) structures) recognizing 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.
  • an AON for redirecting splicing according to the invention is formulated in a composition or a medicament or a composition, which is provided with at least an excipient and/or a targeting ligand for delivery and/or a delivery device thereof to a cell and/or enhancing its intracellular delivery.
  • compositions may not be suitably formulated in one single combination or composition or preparation.
  • the skilled person will know which type of formulation is the most appropriate for each constituent as defined herein.
  • the invention provides a composition or a preparation which is in the form of a kit of parts comprising an AON for redirecting splicing according to the invention and a further adjunct compound as later defined herein.
  • an AON for redirecting splicing according to the invention or a vector, preferably a viral vector, according to the invention, expressing an AON for redirecting splicing according to the invention can be incorporated into a pharmaceutically active mixture by adding a pharmaceutically acceptable carrier.
  • the invention also provides for a composition, preferably a pharmaceutical composition, comprising an AON for redirecting splicing according to the invention, or a viral vector according to the invention and a pharmaceutically acceptable excipient.
  • a composition may comprise a single AON for redirecting splicing or viral vector according to the invention, but may also comprise multiple, distinct AONs for redirecting splicing or viral vectors according to the invention.
  • Such a pharmaceutical composition may comprise any pharmaceutically acceptable excipient, including a carrier, filler, preservative, adjuvant, solubilizer and/or diluent.
  • Such pharmaceutically acceptable carrier, filler, preservative, adjuvant, solubilizer and/or diluent may for instance be found in Remington, 2000. Each feature of said composition has earlier been defined herein.
  • EP2425 814 discloses an oil in water emulsion especially adapted for intraocular (intravitreal) administration of peptide or nucleic acid drugs. This emulsion is less dense than the vitreous fluid, so that the emulsion floats on top of the vitreous, avoiding that the injected drug impairs vision.
  • the concentration or dose defined herein may refer to the total concentration or dose of all oligonucleotides used or the concentration or dose of each exon rentention molecule used or added. Therefore, in an embodiment, there is provided a composition wherein each or the total amount of AONs for redirecting splicing according to the invention used is dosed in an amount ranged from 0.01 and 20 mg/kg, preferably from 0.05 and 20 mg/kg per eye.
  • a suitable intravitreal dose comprises between 0.05 mg and 5mg, preferably between 0.1 and 1 mg of total exon 28 retention molecule per eye, such as about per eye: 0.1 , 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9 or 1.0 mg.
  • the invention further provides for an AON for redirecting splicing according to the invention, or a viral vector according to the invention, or a (pharmaceutical) composition according to the invention for use as a medicament, preferably as a medicament for the treatment of an ABCA4- related disease or condition requiring modulating splicing of ABCA4 and for use as a medicament for the prevention, treatment or delay of an ABCA4- related disease or condition.
  • An AON for redirecting splicing according to the invention, or a viral vector according to the invention, or a (pharmaceutical) composition according to the invention for use as a medicament, preferably as a medicament for the treatment of an ABCA4- related disease or condition requiring modulating splicing of ABCA4 and for use as a medicament for the prevention, treatment or delay of an ABCA4- related disease or condition.
  • the invention further provides for the use of an AON for redirecting splicing according to the invention, a vector according to the invention or a (pharmaceutical) composition according to the invention for treating an ABCA4- related disease or condition requiring modulating splicing of ABCA4.
  • AON for redirecting splicing according to the invention
  • vector according to the invention or a (pharmaceutical) composition according to the invention for treating an ABCA4- related disease or condition requiring modulating splicing of ABCA4.
  • the invention further provides for, a method of treatment of an ABCA4- related disease or condition requiring modulating splicing of ABCA4, comprising said method comprising contacting a cell of said individual with an AON for redirecting splicing according to the invention, a vector according to the invention or a (pharmaceutical) composition according to the invention.
  • a method of treatment of an ABCA4- related disease or condition requiring modulating splicing of ABCA4 comprising said method comprising contacting a cell of said individual with an AON for redirecting splicing according to the invention, a vector according to the invention or a (pharmaceutical) composition according to the invention.
  • the invention further provides for the use of an AON for redirecting splicing according to the invention, a vector according to the invention or a (pharmaceutical) composition according to the invention for the preparation of a medicament for the treatment of an ABCA4- related disease or condition requiring modulating splicing of ABCA4.
  • An AON for redirecting splicing according to the invention a vector according to the invention or a (pharmaceutical) composition according to the invention for the preparation of a medicament for the treatment of an ABCA4- related disease or condition requiring modulating splicing of ABCA4.
  • the invention further provides for the use of an AON for redirecting splicing according to the invention, a vector according to the invention or a (pharmaceutical) composition according to the invention for the preparation of a medicament.
  • the invention further provides for the use of an AON for redirecting splicing according to the invention, a vector according to the invention or a (pharmaceutical) composition according to the invention or a method of treatment according to the invention wherein the ABCA4- related disease or condition is Stargardt disease.
  • Treatment in a use or in a method according to the invention is preferably at least once, and preferably lasts at least one week, one month, several months, one year, 2, 3, 4, 5, 6 years or longer, such as life-long.
  • Each AON for redirecting splicing according to the invention or equivalent thereof as defined herein for use according to the invention may be suitable for direct administration to a cell, tissue and/or an organ in vivo of individuals already affected or at risk of developing an ABCA4- related disease or condition, and may be administered directly in vivo, ex vivo or in vitro.
  • the frequency of administration of an AON, composition, compound or adjunct compound according to the invention may depend on several parameters such as the severity of the disease, the age of the patient, the mutation of the patient, the number of AON for redirecting splicing according to the invention (i.e. dose), the formulation of the AON, composition, compound or adjunct compound according to the invention, the route of administration and so forth.
  • the frequency of administration may vary between daily, weekly, at least once in two weeks, or three weeks or four weeks or five weeks or a longer time period.
  • Dose ranges of an AON, composition, compound or adjunct compound according to the invention are preferably designed on the basis of rising dose studies in clinical trials (in vivo use) for which rigorous protocol requirements exist.
  • An AON according to the invention may be used at a dose which is ranged from 0.01 and 20 mg/kg, preferably from 0.05 and 20 mg/kg.
  • a suitable intravitreal dose would be between 0.05 mg and 5mg, preferably between 0.1 and 1 mg per eye, such as about per eye: 0.1 , 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9 or 1.0 mg.
  • a concentration of an oligonucleotide as defined herein which is ranged from 0.1 nM and 1 mM is used. Preferably, this range is for in vitro use in a cellular model such as retina cells or retinal tissue. More preferably, the concentration used is ranged from 1 to 400 nM, even more preferably from 10 to 200 nM, even more preferably from 50 to 100 nM. If multiple distinct AONs are used, this concentration or dose may refer to the total concentration or dose of the AONs or the concentration or the dose of each AON added.
  • a viral vector preferably an AAV vector as described earlier herein, as delivery vehicle for a molecule according to the invention, is administered in a dose ranging from 1 x1 09 — 1x10 17 virus particles per injection, more preferably from 1x10 1 °— 1x10 12 virus particles per injection.
  • concentration or dose of AONs as depicted above are preferred concentrations or doses for in vivo, in vitro or ex vivo uses.
  • concentration or dose of AONs used may further vary and may need to be optimized any further.
  • An AON for redirecting splicing according to the invention, or a viral vector according to the invention, or a composition according to the invention for use according to the invention may be administered to a cell, tissue and/or an organ in vivo of individuals already affected or at risk of developing a ABCA4- related disease or condition, and may be administered in vivo, ex vivo or in vitro.
  • An AON for redirecting splicing according to the invention, or a viral vector according to the invention, or a composition according to the invention may be directly or indirectly administered to a cell, tissue and/or an organ in vivo of an individual already affected by or at risk of developing a ABCA4- related disease or condition, and may be administered directly or indirectly in vivo, ex vivo or in vitro.
  • the invention further provides for a method for modulating splicing of ABCA4 in a cell comprising contacting the cell, preferably a retina cell, with an AON for redirecting splicing according to the invention, or a viral vector according to the invention, or a (pharmaceutical) composition according to the invention.
  • the features of this aspect are preferably those defined earlier herein.
  • Contacting the cell with an AON for redirecting splicing according to the invention, or a viral vector according to the invention, or a composition according to the invention may be performed by any method known by the person skilled in the art. Use of the methods for delivery of AONs for redirecting splicing, viral vectors and compositions as described earlier herein is included. Contacting may be directly or indirectly and may be in vivo, ex vivo or in vitro.
  • the word "about” or “approximately” when used in association with a numerical value preferably means that the value may be the given value (of 10) more or less 5% of the value.
  • the sequence information as provided herein should not be so narrowly construed as to require inclusion of erroneously identified bases. The skilled person is capable of identifying such erroneously identified bases and knows how to correct for such errors. In case of sequence errors, the sequence of the polypeptide obtainable by expression of the gene present in SEQ ID NO: 1 containing the nucleic acid sequence coding for the polypeptide should prevail.
  • Figure 1 Schematic representation of the location of the c.4253+43G>A variant and the designed AONs.
  • FIG. 2 RT-PCR analysis of AONs in WT and MUT midigenes in HEK293T cells transfected with Wild-type (WT) or c.4253+43G>A mutant (MUT) constructs.
  • Sense oligonucleotide (SON) was used as a control.
  • Figure 3 Semi-quantification of the transcripts identified by RT-PCR. All ABCA4 transcripts detected in the gel were semi-quantified using FIJI software.
  • Figure 4 RT-PCR analysis using control and patient-derived photoreceptor progenitor cells (PPCs).
  • a midigene was created including part of the 26 intron, the complete exon 28, and part of exon 29.
  • This genomic region was cloned into a pCI-Neo-Rhodopsin vector using the Gateway System.
  • the resulting vector (coined pCI-Neo-Rho-ABCA4-26-29 wild-type, SEQ ID NO:22) was used to introduce the c. c.4243+43G>A mutation by site-directed mutagenesis (new vector was coined pCI- Neo-Rho-ABCA4-26-29 c.4243+43G>A SEQ ID NO:23).
  • the control and mutated vectors were validated by Sanger sequencing.
  • minigenes were then transfected in HEK293T cells, which were harvested 48 h post-transfection and were subjected to RT-PCR analysis in order to detect the splicing defect.
  • these midigene constructs can be transfected in eukaryotic immortalized cells (such as Human Embryonic Kidney cells HEK293T, which were used in this study), and the splice pattern can be studied via RT-PCR analysis. If the mutant construct shows an aberrant splicing pattern, co-transfection of midigenes together with AONs allows to determine the efficacy of the AONs to redirect splicing.
  • HEK293T cells were transfected with either the wild-type (WT) or c.4253+43G>A mutant (MUT) midigene construct. Twenty-four hours later, transfected cells were subdivided in wells to decrease transfection variability. AON was delivered at a final concentration of 0,5 uM. Forty-eight hours post- AON-transfection, cells were harvested and subjected to RNA analysis by RT-PCR. For the RT- PCR, total RNA was isolated by using the NucleoSpin RNA Clean-up Kit (catalog no., 740955-50; Macherey-Nagel, Ddren, Germany) according to the manufacturer's protocol.
  • RT-PCR ABCA4 was amplified from exon 26 to 29 using the primers represented by SEQ ID NOs: 24 and 25.
  • RHO was included as transfection control and ACTB as loading control (primers used SEQ ID NOs:26-29).
  • SON sense oligonucleotide
  • All tested AONs were capable of redirecting splicing ( Figure 2).
  • iPSCs induced pluripotent stem cells
  • PPCs photoreceptor precursor cells

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Molecular Biology (AREA)
  • Organic Chemistry (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biophysics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
EP19732376.9A 2018-07-19 2019-06-25 Antisense-oligonukleotide zur wiederherstellung aberranter spleissung von abca4 Pending EP3824086A1 (de)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP18184432 2018-07-19
PCT/EP2019/066895 WO2020015959A1 (en) 2018-07-19 2019-06-25 Antisense oligonucleotides rescue aberrant splicing of abca4.

Publications (1)

Publication Number Publication Date
EP3824086A1 true EP3824086A1 (de) 2021-05-26

Family

ID=63012884

Family Applications (1)

Application Number Title Priority Date Filing Date
EP19732376.9A Pending EP3824086A1 (de) 2018-07-19 2019-06-25 Antisense-oligonukleotide zur wiederherstellung aberranter spleissung von abca4

Country Status (2)

Country Link
EP (1) EP3824086A1 (de)
WO (1) WO2020015959A1 (de)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022090256A1 (en) 2020-10-26 2022-05-05 Proqr Therapeutics Ii B.V. Antisense oligonucleotides for the treatment of stargardt disease

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5139941A (en) 1985-10-31 1992-08-18 University Of Florida Research Foundation, Inc. AAV transduction vectors
US5952221A (en) 1996-03-06 1999-09-14 Avigen, Inc. Adeno-associated virus vectors comprising a first and second nucleic acid sequence
EP1191097A1 (de) 2000-09-21 2002-03-27 Leids Universitair Medisch Centrum Induktion von "exon-skipping" in eukaryotischen Zellen
DK2425814T3 (da) 2010-09-03 2013-09-08 Novagali Pharma Sa Vand-i-olieemulsion til behandling af en øjensygdom
WO2013081755A1 (en) * 2011-11-29 2013-06-06 Life Technologies Corporation Methods and compositions for multiplex pcr
JP2016522674A (ja) * 2012-05-16 2016-08-04 ラナ セラピューティクス インコーポレイテッド 遺伝子発現を調節するための組成物及び方法
EP3019610B1 (de) * 2013-07-08 2020-05-06 INSERM (Institut National de la Santé et de la Recherche Médicale) Verfahren zur durchführung von oligonucleotidvermitteltem antisense-exon-skipping in der netzhaut einer person, die darauf angewiesen ist
WO2016201272A1 (en) * 2015-06-12 2016-12-15 King Abdulaziz City For Science And Technology Method of diagnosing patients with conditions caused by mendelian mutations
EP3377116A4 (de) * 2015-11-19 2019-07-10 The Trustees of The University of Pennsylvania Zusammensetzungen und verfahren zur korrektur von erblicher augenerkrankung
JP2020500524A (ja) 2016-12-13 2020-01-16 スティヒティング カソリーケ ユニベルシタイトStichting Katholieke Universiteit スターガルト病の治療のためのアンチセンスオリゴヌクレオチド

Also Published As

Publication number Publication date
WO2020015959A1 (en) 2020-01-23

Similar Documents

Publication Publication Date Title
US11279933B2 (en) Antisense oligonucleotides for the treatment of leber congenital amaurosis
US11414662B2 (en) Antisense oligonucleotides for the treatment of usher syndrome type 2
US11479771B2 (en) Antisense oligonucleotides for the treatment of eye disease
US20220204972A1 (en) Antisense oligonucleotides for the treatment of Stargardt disease
US20220049250A1 (en) Antisense Oligonucleotides Rescue Aberrant Splicing of ABCA4
WO2020015959A1 (en) Antisense oligonucleotides rescue aberrant splicing of abca4.
US11739324B2 (en) Antisense oligonucleotides rescue aberrant splicing of ABCA4
US20220290154A1 (en) Antisense oligonucleotides rescue aberrant splicing of ABCA4
EA043834B1 (ru) Антисмысловые олигонуклеотиды для лечения болезни штаргардта

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20210126

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: STICHTING RADBOUD UNIVERSITAIR MEDISCH CENTRUM

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)