Antisense oligonucleotides that bind to exon 51 of human dystrophin pre-mRNA

Information

  • Patent Grant
  • 11891603
  • Patent Number
    11,891,603
  • Date Filed
    Friday, September 3, 2021
    2 years ago
  • Date Issued
    Tuesday, February 6, 2024
    3 months ago
Abstract
The present invention relates to a therapeutic antisense oligonucleotide which binds to exon 51 of the human dystrophin pre-mRNA to induce exon skipping, and conjugates and compositions thereof for the treatment of DMD.
Description
SEQUENCE LISTING

The instant application contains a Sequence Listing which has been filed electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Feb. 15, 2020, is named 45532-746-301_SL.txt and is 92,718 bytes in size.


BACKGROUND OF THE INVENTION

Disruption of alternative splicing underlies many diseases, and modulation of splicing using antisense oligonucleotides can have therapeutic implications. Splice-switching antisense oligonucleotides (SSOs) are emerging treatments for neuromuscular diseases, with several SSOs currently undergoing clinical trials for conditions such as spinal muscular atrophy (SMA) and Duchenne muscular dystrophy (DMD), where antisense-mediated exon skipping can restore the open reading frame and allow the synthesis of partly or wholly functional proteins instead of non-functional ones.


Duchenne muscular dystrophy (DMD) is one of the most prevalent lethal genetic disorders in boys worldwide, with an incidence of approx. 1 in 3,600-9,337 live male births. DMD is caused by the absence of dystrophin protein due to mutations in the dystrophin (DMD) gene. The gene encoding the protein contains 79 exons spread out over more than 2 million nucleotides of DNA. Any exonic mutation that changes the reading frame of the exon, or introduces a stop codon, or is characterized by removal of an entire out of frame exon or exons or duplications of one or more exons has the potential to disrupt production of functional dystrophin, resulting in DMD. A less severe form of muscular dystrophy, Becker muscular dystrophy (BMD) has been found to arise where a mutation, typically a deletion of one or more exons, results in a correct reading frame along the entire dystrophin transcript, such that translation of mRNA into protein is not prematurely terminated. If the joining of the upstream and downstream exons in the processing of a mutated dystrophin pre-mRNA maintains the correct reading frame of the gene, the result is an mRNA coding for a protein with a short internal deletion that retains some activity resulting in a Becker Page 1 Header phenotype. Deletions of an exon or exons which do not alter the reading frame of a dystrophin protein give rise to a BMD phenotype, whereas an exon deletion that causes a frame-shift will give rise to DMD (MonAc0, Bertelson et al. 1988). In general, dystrophin mutations including point mutations and exon deletions that change the reading frame and thus interrupt proper protein translation result in DMD.


Currently one of the most promising therapeutic avenues is exon skipping using antisense oligonucleotides (AOs). Exon skipping can restore the reading frame by removing the mutant exon and/or its flanking exon(s) from the DMD pre-mRNA, enabling the production of truncated but partly-functional dystrophin protein. A majority of DMD patients harbour deletion mutations and 20% of these are amenable to exon 51 skipping.


In September 2016, the US Food and Drug Administration (FDA) conditionally approved the first DMD antisense drug, eteplirsen (Exondys 51), which was developed to exclude exon 51 from mutant DMD. Eteplirsen is an AO modified with a phosphorodiamidate morpholino oligomer (morpholino or PMO), an antisense chemistry that has been well-established in terms of its safety and effectiveness. However, eteplirsen remains controversial as there is only weak evidence supporting the effectiveness of the drug, both in terms of restoring dystrophin protein to therapeutically beneficial levels, and improving clinical outcomes. The FDA has previously rejected another drug candidate for DMD exon 51 skipping: the 2′-O-methyl-phosphorothioate-based AO ‘drisapersen’. Although therapeutics must ensure the highest possible benefit for the lowest amount of risk, no significant improvements in muscle function were demonstrated upon treatment with drisapersen, and its use led to concerns over safety.


Therefore, exon skipping therapies currently face a major challenge in that their observed efficacy in patients has been very low despite the fact that significant therapeutic effects have been demonstrated in many animal studies.


Exon skipping efficiency is largely dependent on the AO target sequence: however, there has been little debate or discussion that the sequences targeted by eteplirsen and drisapersen might not be the optimal choices for exon skipping therapy. Several groups have undertaken large-scale AO screening efforts to determine effective AO sequences computationally and empirically. However, the exon skipping effectiveness of designed AOs has not been evaluated both quantitatively and statistically. Although restoring dystrophin protein expression is necessary to improve dystrophic muscle function, the ability of AOs to rescue dystrophin protein expression has not been reported with sufficient Page 2 Header methods of quantification in previous AO screening studies. Other studies have highly relied on RT-PCR from primary DMD muscle cells. It is remarkable that the AO sequences of eteplirsen and drisapersen were determined only within this context.


Thus, the effectiveness of exon 51 skipping therapy could be improved by selecting more optimal AO sequences, and by performing more rigorous AO screening using a more reliable and direct biological measure—such as rescued dystrophin protein in DMD—for validating the best antisense oligonucleotides to be taken forward in a clinical trial.


It is an aim of one or more aspects of the present invention to address one or more such problems in the art.


SUMMARY OF THE INVENTION

According to a first aspect of the present invention, there is provided an antisense oligonucleotide capable of binding to exon 51 of human dystrophin pre-mRNA, wherein binding of the antisense oligonucleotide takes place entirely within the region between 0 and +89 of the pre-mRNA sequence, and wherein the antisense oligonucleotide comprises at least 27 bases.


According to a second aspect of the present invention, there is provided a conjugate comprising an antisense oligonucleotide according to the first aspect and a carrier, wherein the carrier is conjugated to the antisense oligonucleotide.


According to a third aspect of the present invention, there is provided a cell loaded with a conjugate of the second aspect.


According to a fourth aspect of the present invention, there is provided a pharmaceutical composition comprising an antisense oligonucleotide according to the first aspect, and/or a conjugate according to the second aspect, and a pharmaceutically acceptable excipient. According to a fifth aspect of the present invention, there is provided a method of treating a muscular disorder in a subject, comprising administering an effective amount of an antisense oligonucleotide capable of binding to exon 51 of human dystrophin pre-mRNA to a subject, wherein binding of the antisense oligonucleotide takes place entirely within the region between 0 and +89 of the pre-mRNA sequence, and wherein the antisense oligonucleotide comprises at least 27 bases.


According to a sixth aspect of the present invention, there is provided an antisense oligonucleotide capable of binding to exon 51 of human dystrophin pre-mRNA for use in Page 3 Header the treatment of a muscular disorder in a subject, wherein binding of the antisense oligonucleotide takes place entirely within the region between 0 and +89 of the pre-mRNA sequence, and wherein the antisense oligonucleotide comprises at least 27 bases.


According to a seventh aspect of the present invention, there is provided a method of increasing human dystrophin protein expression in a cell comprising contacting the cell with an effective amount of an antisense oligonucleotide capable of binding to exon 51 of human dystrophin pre-mRNA, wherein binding of the antisense oligonucleotide takes place entirely within the region between 0 and +89 of the pre-mRNA sequence, and wherein the antisense oligonucleotide comprises at least 27 bases.


DETAILED DESCRIPTION OF THE INVENTION

The inventors have developed a series of antisense oligonucleotides binding within the early region at 0 to +89 of exon 51 of the dystrophin pre-mRNA sequence and which have a longer than usual length of at least 27 bases, each having remarkable efficiency and effectiveness.


In order to produce the antisense oligonucleotides, the inventors performed a study which quantitatively evaluated the effectiveness of morpholino-based antisense oligonucleotides for exon 51 skipping using a systematic screening method involving in silico, in vitro, and in vivo tests.


The inventors carried out a combination screening using a computational analysis to predict exon skipping efficiency of designed antisense oligonucleotide sequences followed by in vitro tests of morpholino antisense oligonucleotides in immortalized DMD patient-derived muscle cell lines. This research revealed that the beginning of the human dystrophin exon 51 sequence is a very promising target region for inducing exon skipping, specifically the region of 0 to +89 of the sequence. This is notably different from the internal region targeted by the known eteplirsen and drisapersen antisense therapies.


The antisense oligonucleotides identified from this region were then optimised for the most effective restoration of dystrophin production in muscle cells. Various factors were investigated, including the length of the antisense oligonucleotides. Surprisingly, the inventors found that antisense oligonucleotides binding in this early region are more effective when they are longer than many of the known antisense oligonucleotide sequences against exon 51. Specifically, the inventors identified an upward trend correlating effectiveness with the length of the antisense oligonucleotide from 27 bases and Page 4 Header longer. The inventors have shown that just a few bases difference means the antisense oligonucleotide has a significantly different efficiency. As demonstrated herein, 30-mer antisense oligonucleotides work up to 1.5-fold better than a 25-mer of the same sequence (42% vs. 65% skipping efficiency). Without wishing to be bound by theory, this may be because longer sequences can be more specific to the target sequence and less likely to cause off-target effects.


It is demonstrated herein that the inventors' optimisation of these identified antisense oligonucleotide sequences has enabled efficiency in exon 51 skipping and in rescuing dystrophin protein to increase by up to more than 12-fold and 7-fold respectively compared to the industry standard ‘eteplirsen’ sequence. Furthermore, statistically significant in vivo exon 51 skipping by the most effective antisense oligonucleotide identified through these in vitro screenings was confirmed using transgenic mice harbouring the human DMD gene, which has never been shown for the eteplirsen or drisapersen antisense oligonucleotides. Accordingly, the antisense oligonucleotides described herein are shown to provide an effective therapy and treatment for muscular disorders, especially for the treatment of DMD. These antisense oligonucleotides are not only providing an alternative therapy into a field of medicine in which only one such drug has been approved for market. They also provide an improved option for treatment which is several times more effective at increasing dystrophin protein expression. This is expected to provide a viable option for treatment for those suffering from DMD and other muscular disorders with strong evidence to support the effectiveness of the therapy.


For the avoidance of doubt, and in order to clarify the way in which the present disclosure is to be interpreted, certain terms used in accordance with the present invention will now be defined further.


The invention includes any combination of the aspects and features described except where such a combination is clearly impermissible or expressly avoided.


It is noted that where aspects of the invention may refer methods or uses including an antisense oligonucleotide, this may also include a conjugate or pharmaceutical composition comprising an antisense oligonucleotide as defined herein.


The section headings used herein are for organisational purposes only and are not to be construed as limiting the subject matter described.


Antisense Oligonucleotide


The present invention relates to antisense oligonucleotides having a length of at least 27 bases that bind to exon 51 of human dystrophin pre-mRNA within the region of 0 to +89 which can be used to treat muscular disorders.


Suitably, ‘antisense oligonucleotides’ may be referred to herein as ‘AOs’ or ‘oligos’ or ‘oligomers’.


Suitably the antisense oligonucleotide induces skipping of exon 51 of the human dystrophin gene.


Suitably the antisense oligonucleotide increases skipping of exon 51 of the human dystrophin gene.


Suitably the antisense oligonucleotide allows expression of functional human dystrophin protein.


Suitably the antisense oligonucleotide increases expression of functional human dystrophin protein.


Suitably, the antisense oligonucleotide comprises at least 28 bases, suitably at least 29 bases, suitably at least 30 bases.


Suitably, the antisense oligonucleotide comprises between 27 and 30 bases. In one embodiment, the antisense oligonucleotide comprises 30 bases.


In one embodiment, the antisense oligonucleotide consists of 30 bases.


Suitably, the binding of the antisense oligonucleotide takes place entirely within the region between 0 and +88, 0 and +87, 0 and +86, 0 and +85, 0 and +84, 0 and +83, 0 and +82, 0 and +81, 0 and +80, 0 and +79, 0 and +78 of the pre-mRNA sequence.


In one embodiment, the binding of the antisense oligonucleotide takes place entirely within the region between 0 and +78 of the pre-mRNA sequence.


Suitably, the antisense oligonucleotide comprises at least 27 bases of one of the following sequences: SEQ ID NO.1 (Ac0), SEQ ID NO.2 (Ac5), SEQ ID NO.3 (Ac26), SEQ ID NO.4 (Ac30), or SEQ ID NO.5 (Ac48).


Suitably, the antisense oligonucleotide comprises at least 28 bases of one of the following sequences: SEQ ID NO.1 (Ac0), SEQ ID NO.2 (Ac5), SEQ ID NO.3 (Ac26), SEQ ID NO.4 (Ac30), or SEQ ID NO.5 (Ac48).


Suitably, the antisense oligonucleotide comprises at least 29 bases of one of the following sequences: SEQ ID NO.1 (Ac0), SEQ ID NO.2 (Ac5), SEQ ID NO.3 (Ac26), SEQ ID NO.4 (Ac30), or SEQ ID NO.5 (Ac48).


Suitably, the antisense oligonucleotide comprises at least 27 contiguous bases of one of the following sequences: SEQ ID NO.1 (Ac0), SEQ ID NO.2 (Ac5), SEQ ID NO.3 (Ac26), SEQ ID NO.4 (Ac30), or SEQ ID NO.5 (Ac48).


Suitably, the antisense oligonucleotide comprises at least 28 contiguous bases of one of the following sequences: SEQ ID NO.1 (Ac0), SEQ ID NO.2 (Ac5), SEQ ID NO.3 (Ac26), SEQ ID NO.4 (Ac30), or SEQ ID NO.5 (Ac48).


Suitably, the antisense oligonucleotide comprises at least 29 contiguous bases of one of the following sequences: SEQ ID NO.1 (Ac0), SEQ ID NO.2 (Ac5), SEQ ID NO.3 (Ac26), SEQ ID NO.4 (Ac30), or SEQ ID NO.5 (Ac48).


Suitably, the antisense oligonucleotide shares at least 70%, at least 80%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identity with one of the following sequences: SEQ ID NO.1 (Ac0), SEQ ID NO.2 (Ac5), SEQ ID NO.3 (Ac26), SEQ ID NO.4 (Ac30), or SEQ ID NO.5 (Ac48).


Suitably, the antisense oligonucleotide shares between 90% and 100% identity with one of the following sequences: SEQ ID NO.1 (Ac0), SEQ ID NO.2 (Ac5), SEQ ID NO.3 (Ac26), SEQ ID NO.4 (Ac30), or SEQ ID NO.5 (Ac48).


Suitably, the antisense oligonucleotide shares at least 70%, at least 80%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% homology with one of the following sequences: SEQ ID NO.1 (Ac0), SEQ ID NO.2 (Ac5), SEQ ID NO.3 (Ac26), SEQ ID NO.4 (Ac30), or SEQ ID NO.5 (Ac48).


Suitably, the antisense oligonucleotide shares between 90% and 100% homology with one of the following sequences: SEQ ID NO.1 (Ac0), SEQ ID NO.2 (Ac5), SEQ ID NO.3 (Ac26), SEQ ID NO.4 (Ac30), or SEQ ID NO.5 (Ac48).


Suitably, the antisense oligonucleotide may comprise a variant antisense oligonucleotide which differs from one of the following sequences: SEQ ID NO.1 (Ac0), SEQ ID NO.2 (Ac5), SEQ ID NO.3 (Ac26), SEQ ID NO.4 (Ac30), or SEQ ID NO.5 (Ac48) by 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 bases.


Suitably, the antisense oligonucleotide may comprise a variant antisense oligonucleotide which differs from one of the following sequences: SEQ ID NO.1 (Ac0), SEQ ID NO.2 (Ac5), SEQ ID NO.3 (Ac26), SEQ ID NO.4 (Ac30), or SEQ ID NO.5 (Ac48) by up to 3 bases. Suitably, the antisense oligonucleotide may comprise a variant antisense oligonucleotide which differs from one of the following sequences: SEQ ID NO.1 (Ac0), SEQ ID NO.2 (Ac5), SEQ ID NO.3 (Ac26), SEQ ID NO.4 (Ac30), or SEQ ID NO.5 (Ac48) by up to 2 bases. Suitably, the antisense oligonucleotide may comprise a variant antisense oligonucleotide which differs from one of the following sequences: SEQ ID NO.1 (Ac0), SEQ ID NO.2 (Ac5), SEQ ID NO.3 (Ac26), SEQ ID NO.4 (Ac30), or SEQ ID NO.5 (Ac48) by a single base. Suitably, the antisense oligonucleotide comprises one of the following sequences: SEQ ID NO.1 (Ac0), SEQ ID NO.2 (Ac5), SEQ ID NO.3 (Ac26), SEQ ID NO.4 (Ac30), or SEQ ID NO.5 (Ac48).


Suitably, the antisense oligonucleotide consists of one of the following sequences: SEQ ID NO.1 (Ac0), SEQ ID NO.2 (Ac5), SEQ ID NO.3 (Ac26), SEQ ID NO.4 (Ac30), or SEQ ID NO.5 (Ac48).


Suitably, the antisense oligonucleotide comprises SEQ ID NO.1 (Ac0) or SEQ ID NO.5 (Ac48).


In one embodiment, the antisense oligonucleotide comprises SEQ ID NO.1 (Ac0).


Suitably, the antisense oligonucleotide consists of SEQ ID NO.1 (Ac0) or SEQ ID NO.5 (Ac48).


In one embodiment, the antisense oligonucleotide consists of SEQ ID NO.1 (Ac0).


It will be appreciated that the invention may further include aspects directed towards each of the individual antisense oligonucleotide sequences listed in Table 3 i.e. an antisense oligonucleotide comprising or consisting of any of the sequences listed in Table 3. Furthermore, in accordance with the second aspect of the invention, a conjugate comprising an antisense oligonucleotide as listed in Table 3 is envisaged. Furthermore a pharmaceutical composition in accordance with the fourth aspect of the invention,


comprising an antisense oligonucleotide as listed in Table 3 or a conjugate thereof is envisaged. Furthermore a medical use in accordance with the fifth aspect of the invention, comprising an antisense oligonucleotide as listed in Table 3 for the treatment of a muscular disorder is envisaged. Furthermore a method of treatment in accordance with the sixth aspect comprising an antisense oligonucleotide as listed in Table 3 is envisaged. Furthermore a method of increasing human dystrophin protein expression in a cell in accordance with the seventh aspect comprising an antisense oligonucleotide as listed in Table 3 is envisaged.


Suitably, the antisense oligonucleotide is synthetic, and non-natural.


Suitably, the antisense oligonucleotide may be routinely made through the well-known technique of solid phase synthesis. Equipment for such synthesis is sold by several manufacturers including, for example, Applied Biosystems (Foster City, Calif.). One method for synthesising oligonucleotides on a modified solid support is described in U.S. Pat. No. 4,458,066. Any other means for such synthesis known in the art may additionally or alternatively be employed.


Suitably, the antisense oligonucleotide is an antisense oligonucleotide analogue.


Suitably, the term ‘oligonucleotide analogue’ and ‘nucleotide analogue’ refer to any modified synthetic analogues of oligonucleotides or nucleotides respectively that are known in the art.


Suitable examples of oligonucleotide analogues include peptide nucleic acids (PNAs), morpholino oligonucleotides, phosphorothioate oligonucleotides, phosphorodithioate oligonucleotides, alkylphosphonate oligonucleotides, acylphosphonate oligonucleotides and phosphoramidite oligonucleotides.


Suitably, the antisense oligonucleotide comprises morpholino subunits. Suitably therefore, the antisense oligonucleotide is a morpholino antisense oligonucleotide.


Suitably, the antisense oligonucleotide comprises morpholino subunits linked together by phosphorus-containing linkages. Suitably therefore, the antisense oligonucleotide is a phosphoramidate or phosphorodiamidate morpholino antisense oligonucleotide.


The terms ‘morpholino antisense oligonucleotide’ or ?MO’ (phosphoramidate or phosphorodiamidate morpholino oligonucleotide) refer to an antisense oligonucleotide Page 9 Header analog composed of morpholino subunit structures, where (i) the structures are linked together by phosphorus-containing linkages, suitably one to three atoms long, suitably two atoms long, and suitably uncharged or cationic, joining the morpholino nitrogen of one subunit to a 5′ exocyclic carbon of an adjacent subunit, and (ii) each morpholino ring bears a purine or pyrimidine base-pairing moiety effective to bind, by base specific hydrogen bonding, to a base in a polynucleotide.


Suitably, the antisense oligonucleotide comprises phosphorus-containing intersubunit linkages joining a morpholino nitrogen of one subunit to a 5′ exocyclic carbon of an adjacent subunit.


Suitably, the antisense oligonucleotide comprises phosphorus-containing intersubunit linkages in accordance with the following structure (I):




embedded image


wherein:


Y1 is 0, S, NH—, or —CH2-;


Z is 0 or S;


Pj is a purine or pyrimidine base-pairing moiety effective to bind, by base-specific hydrogen bonding, to a base in a polynucleotide; and X is fluoro, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted thioalkoxy, amino, optionally substituted alkylamino, or optionally substituted heterocyclyl.


Optionally, variations can be made to the intersubunit linkage as long as the variations do not interfere with binding or activity. For example, the oxygen attached to phosphorus may be substituted with sulfur (thiophosphorodiamidate). The 5′ oxygen may be substituted with amino or lower alkyl substituted amino. The pendant nitrogen attached to the phosphorus may be unsubstituted, monosubstituted, or disubstituted with (optionally substituted) lower alkyl.


Suitably, the synthesis, structures, and binding characteristics of morpholino oligonucleotides are detailed in U.S. Pat. Nos. 5,698,685, 5,217,866, 5,142,047, 5,034,506, 5,166,315, 5,521,063, and 5,506,337, and PCT Appn. No. PCT/US07/11435.


Binding of the Antisense Oligonucleotide


The present invention relates to an antisense oligonucleotide capable of binding within the region 0 to +89 of exon 51 of human dystrophin pre-mRNA.


By ‘capable of binding’ it is meant that the antisense oligonucleotide comprises a sequence with is able to bind to human dystrophin pre-mRNA in the region stated.


Suitably, the antisense oligonucleotide is complementary to a sequence of human dystrophin pre-mRNA in the region stated.


Suitably, the antisense oligonucleotide comprises a sequence which is complementary to a sequence of human dystrophin pre-mRNA in the region stated.


The antisense oligonucleotide and a sequence within the region 0 to +89 of exon 51 of human dystrophin pre-mRNA are complementary to each other when a sufficient number of corresponding positions in each molecule are occupied by nucleotides which can hydrogen bond with each other and thereby cause exon skipping, suitably exon skipping of exon 51. Thus, ‘hybridisable’ and ‘complementary’ are terms which are used to indicate a sufficient degree of complementarity or pairing such that stable and specific binding occurs between the antisense oligonucleotide and a sequence within region 0 to +89 of exon 51 of human dystrophin pre-mRNA. Suitably, the antisense oligonucleotide is sufficiently hybridisable and/or complementary to a sequence within region 0 to +89 of exon 51 of human dystrophin pre-mRNA to induce exon skipping, suitably exon skipping of exon 51. Suitably, the antisense oligonucleotide may not be 100% complementary to a sequence within region of 0 to +89 of exon 51 of human dystrophin pre-mRNA. However, suitably the antisense oligonucleotide is sufficiently complementary to avoid non-specific binding.


Suitably, the antisense oligonucleotide is at least 70%, at least 80%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% at least 99% complementary to a sequence within the region 0 to +89 of exon 51 of human dystrophin pre-mRNA.


It is understood that in order for the antisense oligonucleotide to be capable of binding, it does not require that the entire length of the antisense oligonucleotide binds to the human dystrophin pre-mRNA. It will be appreciated that a portion of the antisense oligonucleotide Page 11 Header may not bind to the human dystrophin pre-mRNA, for example the 5′ or the 3′ ends of the antisense oligonucleotide. However, in accordance with the first aspect, the parts of the antisense oligonucleotide which are bound to the human dystrophin pre-mRNA must fall within the region of 0 to +89 of exon 51.


Suitably, therefore, the antisense oligonucleotide is hybridisable to a sequence within the region of 0 to +89 of exon 51 of human dystrophin pre-mRNA. Suitably, the antisense oligonucleotide is sufficiently hybridisable to a sequence within the region of 0 to +89 of exon 51 of human dystrophin pre-mRNA to cause exon skipping of exon 51.


Human Dystrophin


The present invention relates to a therapeutic antisense oligonucleotide for use in the treatment of muscular disorders, particularly dystrophin disorders such as DMD.


Dystrophin is a rod-shaped cytoplasmic protein, and a vital part of the protein complex that connects the cytoskeleton of a muscle fibre to the surrounding extracellular matrix through the cell membrane.


Dystrophin protein contains multiple functional domains. The DMD gene, encoding the dystrophin protein, is one of the longest known human genes covering 2.3 megabases (0.08% of the human genome) at locus Xp21. The primary transcript in muscle measures about 2,100 kilobases and takes 16 hours to transcribe; the mature mRNA measures 14.0 kilobases. The 79-exon muscle transcript codes for a protein of 3685 amino acid residues. Dystrophin protein contains an actin binding domain and a central rod domain. This large central domain is formed by 24 spectrin-like triple-helical elements of about 109 amino acids, which have homology to alpha-actinin and spectrin. The repeats are typically interrupted by four proline-rich non-repeat segments, also referred to as hinge regions. Each repeat is encoded by two exons, typically interrupted by an intron between amino acids 47 and 48 in the first part of alpha-helix 2. The other intron is found at different positions in the repeat, usually scattered over helix 3. Dystrophin also contains a cysteine-rich domain including a cysteine-rich segment (i.e., 15 Cysteines in 280 amino acids).


In normal cases, the amino-terminus of dystrophin binds to F-actin and the carboxy-terminus binds to the dystrophin-associated protein complex (DAPC) at the sarcolemma. The DAPC includes the dystroglycans, sarcoglycans, integrins and caveolin, and mutations in any of these components cause autosomally inherited muscular dystrophies. Normal skeletal muscle tissue contains only small amounts of dystrophin (about 0.002% of total muscle protein), but its absence (or abnormal expression) leads to the development of a Page 12 Header severe and currently incurable symptoms most readily characterized by several aberrant intracellular signaling pathways that ultimately yield pronounced myofiber necrosis as well as progressive muscle weakness and fatigability. The DAPC is destabilized when dystrophin is absent, which results in diminished levels of the member proteins, and in turn leads to progressive fibre damage and membrane leakage. In various forms of muscular dystrophy, such as Duchenne's muscular dystrophy (DMD) and Becker's muscular dystrophy (BMD), muscle cells produce an altered and functionally defective form of dystrophin, or no dystrophin at all, mainly due to mutations in the gene sequence that lead to incorrect splicing. The predominant expression of the defective dystrophin protein, or the complete lack of dystrophin or a dystrophin-like protein, leads to rapid progression of muscle degeneration.


The mRNA encoding dystrophin in muscular dystrophy patients typically contains out-of-frame mutations (e.g. deletions, insertions or splice site mutations), resulting in frameshift or early termination of the translation process, so that in most muscle fibres no functional dystrophin is produced.


Suitably, the antisense oligonucleotide triggers exon skipping to restore the reading frame of the dystrophin mRNA. Suitably, the antisense oligonucleotide triggers exon skipping of exon 51 to restore the reading frame of the dystrophin mRNA. Suitably, restoration of the reading frame restores production of a partially functional dystrophin protein.


Suitably, the partially functional dystrophin is a truncated dystrophin protein.


Suitably, the truncated dystrophin protein is the same dystrophin protein produced in patients suffering from the less severe muscular disorder; BMD.


Muscular Disorder


The present invention relates to the use of therapeutic antisense oligonucleotides in the treatment of muscular disorders.


Suitably the muscular disorder is selected from any muscular disorder resulting from a genetic mutation.


Suitably the muscular disorder is selected from any muscular disorder resulting from a genetic mutation in a gene associated with muscle function.


Suitably the muscular disorder is selected from any muscular disorder resulting from a genetic mutation in the human dystrophin gene.


Suitably, the muscular disorder is selected from any muscular dystrophy disorder.


Suitably, the muscular disorder is selected from Duchenne muscular dystrophy, Becker muscular dystrophy, congenital muscular dystrophy, Distal muscular dystrophy, Emery-Dreifuss muscular dystrophy, Facioscapulohumeral muscular dystrophy, Limb-girdle muscular dystrophy, Myotonic muscular dystrophy, Oculopharyngeal Muscular dystrophy. Suitably, the muscular disorder is Duchenne Muscular Dystrophy (DMD) or Becker Muscular Dystrophy (BMD).


In one embodiment, the muscular disorder is DMD.


Carrier and Conjugate


The present invention also relates to a conjugate of the antisense oligonucleotide with a carrier.


Suitably, the carrier may comprise any molecule operable to transport the antisense oligonucleotide into a target cell, suitably into a muscle cell.


Suitable carriers may include; peptides, small molecule chemicals, polymers, nanoparticles, lipids, liposomes, exosomes or the like.


Suitably, the carrier is a peptide. The peptide may be selected from viral proteins such as VP22 (derived from herpes virus tegument protein), snake venom protein such as CyLOP-1 (derived from crotamin), cell adhesion glycoproteins such as pVEC (derived from murine vascular endothelial-cadherin protein), Penetratin (Antennapedia homeodomain), Tat (human immunodeficiency virus transactivating regulatory protein) or reverse Tat, for example.


Suitably, the peptide is a cell penetrating peptide.


Suitably, the peptide is an arginine-rich cell penetrating peptide.


The use of arginine-rich peptide carriers is particularly useful. Certain arginine based peptide carriers have been shown to be highly effective at delivery of antisense compounds into primary cells including muscle cells (Marshall, Oda et al. 2007; Jearawiriyapaisarn, Moulton et al. 2008; Wu, Moulton et al. 2008). Furthermore, compared to other peptides,


the arginine peptide carriers when conjugated to an antisense oligonucleotide, demonstrate an enhanced ability to alter splicing of several gene transcripts (Marshall, Oda et al. 2007). Suitably, the arginine-rich cell penetrating peptide may be selected from those carrier peptides described in WO2015075747, WO2013030569, WO2009147368, US20120289457, or US20160237426, for example.


In one embodiment, the arginine rich cell penetrating peptide is selected from those described in WO2013030569 or WO2009147368.


Suitably, the carrier has the capability of inducing cell penetration of the antisense oligonucleotide within at least 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% of cells of a given cell culture population. Suitably, the carrier has the capability of inducing cell penetration of the antisense oligonucleotide within at least 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% of muscle cells in a muscle cell culture.


Suitably, conjugation of the carrier to the antisense oligonucleotide may be at any position suitable for forming a covalent bond between the carrier and the antisense oligonucleotide or between the linker moiety and the antisense oligonucleotide. For example, conjugation of a carrier may be at the 3′ end of the antisense oligonucleotide. Alternatively, conjugation of a carrier to the antisense oligonucleotide may be at the 5′ end of the oligonucleotide. Alternatively, a carrier may be conjugated to the antisense oligonucleotide through any of the intersubunit linkages.


Suitably, the carrier is covalently coupled at its N-terminal or C-terminal residue to the 3′ or 5′ end of the antisense oligonucleotide.


Suitably, the carrier is coupled at its C-terminal residue to the 5′ end of the antisense oligonucleotide.


Optionally, where the antisense oligonucleotide comprises phosphorus-containing intersubunit linkages, and the carrier is a peptide, the peptide may be conjugated to the antisense oligonucleotide via a covalent bond to the phosphorous of the terminal linkage group.


Alternatively, when the carrier is a peptide, and the antisense oligonucleotide is a morpholino, the peptide may be conjugated to the nitrogen atom of the 3′ terminal morpholino group of the oligomer.


Optionally, the carrier may be conjugated to the antisense oligonucleotide via a linker moiety. Optionally, the linker moiety may comprise one or more of: an optionally substituted piperazinyl moiety, a beta alanine, glycine, proline, and/or a 6-aminohexanoic acid residue in any combination.


Alternatively, the carrier may be conjugated directly to the antisense oligonucleotide without a linker moiety.


Suitably the conjugate may further comprise a homing moiety.


Suitably, the homing moiety is selective for a selected mammalian tissue, i.e., the same tissue being targeted by the antisense oligonucleotide. Suitably, the homing moiety is selective for muscle tissue.


Suitably, the homing moiety is a homing peptide.


Suitable homing peptides are disclosed in ‘Effective Dystrophin Restoration by a Novel Muscle-Homing Peptide—Morpholino Conjugate in Dystrophin-Deficient mdx Mice’ Gao et Mol Ther. 2014 July; 22(7): 1333-1341, for example.


Suitably, the carrier peptide and the homing peptide may be formed as a chimeric fusion protein.


Suitably, the conjugate may comprise a chimeric peptide formed from a cell penetrating peptide and a muscle-specific homing peptide.


Optionally, the conjugate may be of the form: carrier peptide-homing peptide-antisense oligonucleotide or of the form: homing peptide-carrier peptide-antisense oligonucleotide. Suitably, the antisense oligonucleotide may be conjugated to a carrier that enhances the solubility of the antisense oligonucleotide. Suitably the solubility in an aqueous medium. Suitably, a carrier that enhances solubility may be conjugated to the antisense oligonucleotide in addition to a carrier operable to transport the antisense oligonucleotide. Suitably, the carrier that enhances solubility and the carrier that transports the antisense oligonucleotide may be formed as a chimeric fusion protein.


Suitable carriers that enhance the solubility of an antisense oligonucleotide are polymers, such as polyethylene glycol, or triethylene glycol.


Pharmaceutically Acceptable Excipient


The present invention further relates to a pharmaceutical composition comprising the antisense oligonucleotide of the invention or a conjugate thereof, further comprising one or more pharmaceutically acceptable excipients.


Suitably, the pharmaceutical composition is prepared in a manner known in the art (as described in Remingtons Pharmaceutical Sciences, Mack Publ. Co., Easton, Pa. (1985)), with pharmaceutically inert inorganic and/or organic excipients being used. The term ‘pharmaceutically acceptable’ refers to molecules and compositions that are physiologically tolerable and do not typically produce an allergic or similarly untoward reaction when administered to a patient.


Suitably, the pharmaceutical composition may be formulated as a pill, tablet, coated tablet, hard gelatin capsule, soft gelatin capsule and/or suppository, solution and/or syrup, injection solution, microcapsule, implant and/or rod, and the like.


In one embodiment, the pharmaceutical composition may be formulated as an injection solution.


Suitably, pharmaceutically acceptable excipients for preparing pills, tablets, coated tablets and hard gelatin capsules may be selected from any of: Lactose, corn starch and/or derivatives thereof, talc, stearic acid and/or its salts, etc.


Suitably, pharmaceutically acceptable excipients for preparing soft gelatin capsules and/or suppositories may be selected from fats, waxes, semisolid and liquid polyols, natural and/or hardened oils, etc.


Suitably, pharmaceutically acceptable excipients for preparing solutions and/or syrups may be selected from water, sucrose, invert sugar, glucose, polyols, etc.


Suitably, pharmaceutically acceptable excipients for preparing injection solutions may be selected from water, saline, alcohols, glycerol, polyols, vegetable oils, etc.


Suitably, pharmaceutically acceptable excipients for preparing microcapsules, implants and/or rods may be selected from mixed polymers such as glycolic acid and lactic acid or the like.


In addition, the pharmaceutical composition may comprise a liposome formulation which are described in N. Weiner, (Drug Develop Ind Pharm 15 (1989) 1523), “Liposome Dermatics” (Springer Verlag 1992) and Hayashi (Gene Therapy 3 (1996) 878).


Optionally, the pharmaceutical composition may comprise two or more different antisense oligonucleotides or conjugates thereof. Optionally, the pharmaceutical composition may further comprise one or more antisense oligonucleotides or conjugates thereof targeting different exons, suitably different exons of the human dystrophin pre-mRNA. Optionally, the one or more further antisense oligonucleotides or conjugates thereof may target exons adjacent to exon 51, for example, exon 50 or exon 52 of the human dystrophin pre-mRNA. Suitably, the one or more antisense oligonucleotides or conjugates thereof targeting different exons of the human dystrophin pre-mRNA are operable, together with the antisense oligonucleotide of the invention, to restore the reading frame of dystrophin mRNA.


Optionally, the pharmaceutical composition may further comprise one or more antisense oligonucleotides or conjugates thereof targeting different genes. For example, the one or more further antisense oligonucleotides or conjugates thereof may target myostatin. Such dual targeting is described in ‘Dual exon skipping in myostatin and dystrophin for Duchenne muscular dystrophy’ Kemaladewi et al. BMC Med Genomics. 2011 Apr. 20; 4:36.


Optionally, the one or more further antisense oligonucleotides may be joined together and/or joined to the antisense oligonucleotide of the first aspect.


Optionally, the antisense oligonucleotide and/or conjugate may be present in the pharmaceutical composition as a physiologically tolerated salt. Suitably, physiologically tolerated salts retain the desired biological activity of the antisense oligonucleotide and/or conjugate thereof and do not impart undesired toxicological effects. For antisense oligonucleotides, suitable examples of pharmaceutically acceptable salts include (a) salts formed with cations such as sodium, potassium, ammonium, magnesium, calcium, polyamines such as spermine and spermidine, etc.; (b) acid addition salts formed with inorganic acids, for example hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid and the like; (c) salts formed with organic acids such as, for example, acetic acid, oxalic acid, tartaric acid, succinic acid, maleic acid, fumaric acid, gluconic acid, citric acid, malic acid, ascorbic acid, benzoic acid, tannic acid, palmitic acid, alginic acid, polyglutamic acid, naphthalenesulfonic acid, methanesulfonic acid, p-toluenesulfonic acid, naphthalenedisulfonic acid, polygalacturonic acid, and the like; and (d) salts formed from elemental anions such as chlorine, bromine, and iodine.


Optionally, the pharmaceutical composition may comprise, in addition to at least one antisense oligonucleotide and/or conjugate, one or more different therapeutically active ingredients. The one or more therapeutically active ingredients may be selected from, for example: corticosteroids, utrophin-upregulators, TGF-beta inhibitors, and myostatin inhibitors.


Suitably, in addition to the active ingredients and excipients, a pharmaceutical composition may also comprise additives, such as fillers, extenders, disintegrants, binders, lubricants, wetting agents, stabilizing agents, emulsifiers, preservatives, sweeteners, dyes, flavorings or aromatizing agents, thickeners, diluents or buffering substances, and, in addition, solvents and/or solubilizing agents and/or agents for achieving a slow release effect, and also salts for altering the osmotic pressure, coating agents and/or antioxidants. Suitable additives may include Tris-HCl, acetate, phosphate, Tween 80, Polysorbate 80, ascorbic acid, sodium metabisulfite, Thimersol, benzyl alcohol, lactose, mannitol, or the like.


Administration


The present invention relates to a therapeutic antisense oligonucleotide and to a pharmaceutical composition comprising the therapeutic antisense oligonucleotide which are for administration to a subject.


Suitably, the antisense oligonucleotide and/or pharmaceutical composition may be for topical, enteral or parenteral administration.


Suitably, the antisense oligonucleotide and/or pharmaceutical composition may be for administration orally, transdermally, intravenously, intrathecally, intramuscularly, subcutaneously, nasally, transmucosally or the like.


In one embodiment, the antisense oligonucleotide and/or pharmaceutical composition is for intramuscular administration.


In one embodiment, the antisense oligonucleotide and/or pharmaceutical composition is for intramuscular administration by injection.


An ‘effective amount’ or ‘therapeutically effective amount’ refers to an amount of the antisense oligonucleotide, administered to a subject, either as a single dose or as part of a Page 19 Header series of doses, which is effective to produce a desired physiological response or therapeutic effect in the subject.


Suitably, the desired physiological response includes increased expression of a relatively functional or biologically active form of the dystrophin protein, suitably in muscle tissues or cells that contain a defective dystrophin protein or no dystrophin.


Suitably, the desired therapeutic effects include improvements in the symptoms or pathology of a muscular disorder, reducing the progression of symptoms or pathology of a muscular disorder, and slowing the onset of symptoms or pathology of a muscular disorder. Examples of such symptoms include fatigue, mental retardation, muscle weakness, difficulty with motor skills (e.g., running, hopping, jumping), frequent falls, and difficulty walking.


Suitably, the antisense oligonucleotide or conjugate thereof are administered at a dose in the range from about 0.0001 to about 100 mg per kilogram of body weight per day.


Suitably, the antisense oligonucleotide or conjugate thereof are administered daily, once every 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 days, once every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 weeks, or once every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 months.


Suitably, the dose and frequency of administration may be decided by a physician, as needed, to maintain the desired expression of a functional dystrophin protein.


Suitably, the antisense oligonucleotide or conjugate thereof may be administered as two, three, four, five, six or more sub-doses separately at appropriate intervals throughout the day, optionally, in unit dosage forms.


Subject


The present invention also relates to treatment of a muscular disorder by administering a therapeutically effective amount of the antisense oligonucleotide or conjugate thereof to a subject in need thereof.


Suitably the subject has a muscular disorder, as defined above.


Suitably, the subject is mammalian. Suitably the subject is human.


Suitably the subject may be male or female. However, suitably the subject is male.


Suitably, the subject is any age. However, suitably the subject is between the ages of 1 month old to 50 years old, suitably between the ages of 1 years old and 30 years old, suitably between the ages of 2 years old to 27 years old, suitably between the ages of 4 years old to 25 years old


Increased Exon Skipping and Dystrophin Expression


The present invention relates to a therapeutic antisense oligonucleotide for use in the treatment of muscular disorder by inducing exon skipping in the human dystrophin pre-mRNA to restore functional dystrophin protein expression.


Suitably, a ‘functional’ dystrophin protein refers to a dystrophin protein having sufficient biological activity to reduce the progressive degradation of muscle tissue that is otherwise characteristic of muscular dystrophy when compared to the defective form of dystrophin protein that is present in subjects with a muscular disorder such as DMD.


Suitably, a functional dystrophin protein may have about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% of the in vitro or in vivo biological activity of wild-type dystrophin. Suitably, a functional dystrophin protein has at least 10% to 20% of the in vitro or in vivo biological activity of wild-type dystrophin.


Suitably, the activity of dystrophin in muscle cultures in vitro can be measured according to myotube size, myofibril organization, contractile activity, and spontaneous clustering of acetylcholine receptors (see, e.g., Brown et al., Journal of Cell Science. 112:209-216, 1999).


Animal models are also valuable resources for studying the pathogenesis of disease, and provide a means to test dystrophin-related activity. Two of the most widely used animal models for DMD research are the mdx mouse and the golden retriever muscular dystrophy (GRMD) dog, both of which are dystrophin negative (see, e.g., Collins & Morgan, Int J Exp Pathol 84: 165-172, 2003). These and other animal models can be used to measure the functional activity of various dystrophin proteins.


Suitably, ‘exon skipping’ refers to the process by which an entire exon, or a portion thereof, is removed from a given pre-processed RNA (pre-mRNA), and is thereby excluded from being present in the mature RNA that is translated into a protein.


Suitably, the portion of the protein that is otherwise encoded by the skipped exon is not present in the expressed form of the protein.


Suitably therefore, exon skipping creates a truncated, though still functional, form of the protein as defined above.


Suitably, the exon being skipped is an exon from the human dystrophin gene, which may contain a mutation or other alteration in its sequence that otherwise causes aberrant splicing.


Suitably, the exon being skipped is exon 51 of the dystrophin gene.


Suitably, the antisense oligonucleotide is operable to induce exon skipping in dystrophin pre-mRNA.


Suitably, the antisense oligonucleotide is operable to induce exon skipping of exon 51 in dystrophin pre-mRNA.


Suitably, the antisense oligonucleotide is operable to increase expression of a functional form of a dystrophin protein in muscle tissue, and is operable to increase muscle function in muscle tissue.


Suitably, the antisense oligonucleotide is operable to increase muscle function by at least about 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% compared to muscle function in subjects with a muscular disorder such as DMD that have not received the antisense oligonucleotide.


Suitably, the antisense oligonucleotide is operable to increase the percentage of muscle fibres that express a functional dystrophin protein in about at least 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% of muscle fibres compared to subjects with a muscular disorder such as DMD that have not received the antisense oligonucleotide.


Suitably, the antisense oligonucleotide is operable to induce expression of a functional form of a dystrophin protein to a level of at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 25, 40, 45, or 50% of the expression of dystrophin protein in wild type cells and/or subjects. Suitably, the antisense oligonucleotide is operable to induce expression of a functional form of a dystrophin protein to a level of at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, or 20% of the expression of dystrophin protein in wild type cells and/or subjects.


Suitably, the antisense oligonucleotide is operable to induce expression of a functional form of a dystrophin protein to a level of at least 10, 15, or 20% of the expression of dystrophin protein in wild type cells and/or subjects.


Suitably, the antisense oligonucleotide is operable to induce exon 51 skipping in the dystrophin pre-mRNA to a level of at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100%.


Suitably, the antisense oligonucleotide is operable to induce exon 51 skipping in the dystrophin pre-mRNA to a level of at least 60%, 70%, 80%, 90%, or 100%.


Suitably, the antisense oligonucleotide is operable to induce exon 51 skipping in the dystrophin pre-mRNA to a level of between 60% to 80%.


An ‘increased’ or ‘enhanced’ amount may include an increase that is 1.1, 1.2, 1.5, 2, 2.5, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50 or more times the amount produced when no antisense oligonucleotide compound (the absence of an agent) or a control compound is administered under the same circumstances.


Suitably, an ‘increased’ or ‘enhanced’ amount is a statistically significant amount.





BRIEF DESCRIPTION OF THE DRAWINGS

Certain embodiments of the present invention will now be described with reference to the following figures and tables in which:



FIG. 1 Shows in vitro screening of antisense oligonucleotides (AOs) and analog AOs of eteplirsen (aEte) and drisapersen (aDri) at 10 μM, in immortalized clonal exon 52-deleted DMD skeletal muscle cells (KM571). Differentiated myotubes were harvested at day 5 following transfection (A) Efficiency of exon 51 skipping as measured by one-step RT-PCR. Representative images are shown. M, 100 bp marker; blank, no RNA template. (B) Efficiency in inducing truncated dystrophin protein as measured by quantitative Western blotting with the anti-dystrophin C-terminal antibody. Rescued dystrophin protein levels are calculated using calibration curves with healthy 8220 cells. Data represent mean±SD from 3-4 independent experiments. ** p<0.01 vs aEte, t p<0.05 and tt p<0.01 vs aDri, § § p<0.01 vs all of AOs in (A) and vs Ac0 in (B).



FIG. 2 Shows a time-course analysis of dystrophin exon 51 skipping and protein in an exon 52-deleted DMD-KM571 cell line transfected with Ac0, Ac48, and analog AOs of Page 23 Header eteplirsen and drisapersen at 5 μM. Samples were collected at days 2 and 11 post-transfection (A) RT-PCR analysis of exon 51 skipping. M, 100 bp marker; R, replicate number; blank, no RNA templates. (B) Quantification of induced dystrophin protein by Western blotting with the anti-dystrophin C-terminal antibody. Representative replicates from 3 independent experiments are shown.



FIG. 3 Shows the dose-dependent effects of Ac0, Ac48, and analogs AOs of eteplirsen and drisapersen in immortalized DMD skeletal muscle cells as measured by one-step RT-PCR and quantitative Western blotting. DMD skeletal muscle cells were transfected with AOs at 1, 3, and 10 μM and harvested at day 5 post-transfection. (A) and (B) show exon 51 skipping efficiency and expression levels of rescued dystrophin protein, respectively, in DMD muscle cells with exon 52 deletion mutation (ID KM 571). Efficacy of skipping exon 51 and rescuing dystrophin protein expression is shown in (C) and (D), respectively, in DMD muscle cells harboring exons 48-50 deletion mutation (ID 6594). Data represent mean±SD from 3-7 independent experiments in the KM571 cell line and from 3-4 independent experiments in the 6594 cell line. * p<0.05, ** p<0.01 vs aEte; t p<0.05 and tt p<0.01 vs Ac48; § p<0.05, § § p<0.01 vs aDri in the same concentration, NS, no significance vs Ac0 at the next dose; ns, no significance vs Ac0 at 10 μM. (E) Dose-responsiveness to the AOs analysed by regression model. Statistical validity of regression equations in skipping and producing dystrophin protein was p<0.008 and p<0.014, respectively. Plots indicate values of exon skipping or dystrophin protein levels predicted in the regression analysis. The regression slope and 95% confidence interval (CI) are shown in individual AOs.



FIG. 4 Shows Immunocytochemistry in immortalized DMD patient-derived skeletal muscle cells with exon 52 (ID KM571) and exons 48-50 deletion mutations (ID 6594). Cells at day 5 post-transfection with 10 μM Ac0, Ac48, and analog eteplirsen (aEte) were stained with anti-dystrophin C-terminal antibody. Grey lines indicate dystrophin-positive myotubes. White dots indicate nuclei counter-stained with DAPI. * indicates representative false-positive myotubes due to their contraction or detachment from the culture plate. Representative images are shown from 3 independent experiments. Scale bar: 100 μm.



FIG. 5 Shows Length optimization of the Ac0 morpholino antisense oligonucleotide. Immortalized DMD muscle cells were transfected with Ac0 morpholinos composed of 25-, 26-, 27-, 28-, 29-, and 30-mer lengths. A representative image and quantification of exon 51 skipping induced by Ac0 morpholinos at 1 μM (A and B) and 3 μM (C and D) in DMD


muscle cells with exon 52 deletion (KM571) are shown as represented by RT-PCR. (E-H) indicate the results in immortalized DMD cells with exons 48-50 deletion. The data are shown from 3 independent experiments.



FIG. 6 shows Exon 51 skipping efficiency induced by Ac0, Ac48, analog AOs of eteplirsen (aEte) and drisapersen (aDri) in primary DMD and healthy skeletal muscle cells. Differentiated myotubes were transfected with Ac0, Ac48, and analog eteplirsen and drisapersen at 10 μM, and then harvested 3 days later. Exon 51 skipping efficiency as represented by one-step RT-PCR was shown in primary DMD cells with the deletion mutation of exons 45-50 (ID 4546) (A and B) or exons 49-50 (ID 4555) (C and D), and primary healthy muscle cells (E and F). Data represent mean±SD from at least triplicate wells in each condition. M, 100 bp marker. * p<0.05 and ** p<0.01 vs Ac48, tt p<0.01 vs aEte, § § <0.01 vs aDri.



FIG. 7 shows In vivo efficacy of 30-mer Ac0 antisense morpholino oligonucleotide in the hDMD/Dmd-null mouse model. Exon skipping efficacy was analysed by RT-PCR with tibialis anterior muscles 2 weeks after the intramuscular injection of Ac0 morpholino or analog eteplirsen, aEte (50 μg in 30 μL saline). (A) Densitometry analysis of exon 51 skipping as represented by a microchip-based capillary electrophoresis system. (B) Averaged percentage of exon 51 skipping efficiency (mean±SE). N=7 in each group. M, marker; NT, non-treated muscle, UM, upper marker dye; LM, lower marker dye.



FIG. 8 shows In vivo efficacy of 30-mer Ac48 antisense morpholino oligonucleotide in the hDMD/Dmd-null mouse model. Exon skipping efficacy was analysed by RT-PCR with tibialis anterior muscles 2 weeks after the intramuscular injection of Ac48 morpholino or analog drisapersen aDri (50 μg in 30 μL saline). (A) Densitometry analysis of exon 51 skipping as represented by a microchip-based capillary electrophoresis system. (B) Averaged percentage of exon 51 skipping efficiency (mean±SE). N=7 in each group. M, marker; NT, non-treated muscle, UM, upper marker dye; LM, lower marker dye.





Throughout the description and claims of this specification, the words “comprise” and “contain” and variations of them mean “including but not limited to”, and they are not intended to (and do not) exclude other moieties, additives, components, integers or steps. Throughout the description and claims of this specification, the singular encompasses the plural unless the context otherwise requires. In particular, where the indefinite article is used, the specification is to be understood as contemplating plurality as well as singularity, unless the context requires otherwise.


Features, integers, characteristics, compounds, chemical moieties or groups described in conjunction with a particular aspect, embodiment or example of the invention are to be understood to be applicable to any other aspect, embodiment or example described herein unless incompatible therewith. All of the features disclosed in this specification (including any accompanying claims, abstract and drawings), and/or all of the steps of any method or 27 process so disclosed, may be combined in any combination, except combinations where at least some of such features and/or steps are mutually exclusive.


The invention is not restricted to the details of any foregoing embodiments. The invention extends to any novel one, or any novel combination, of the features disclosed in this specification (including any accompanying claims, abstract and drawings), or to any novel one, or any novel combination, of the steps of any method or process so disclosed. The reader's attention is directed to all papers and documents which are filed concurrently with or previous to this specification in connection with this application and which are open to public inspection with this specification, and the contents of all such papers and documents are incorporated herein by reference.


EXAMPLES

Materials and Methods


1.1 Design and in Silico Screening of AOs.


413 30-mer and 25-mer AOs targeting exon 51 were designed and analysed using the AO predictive algorithm we recently developed (see Table 3). Table 3 shows in the columns from left to right; the exon number, the distance from acceptor splice site, the AO sequence (5′ to 3′), the predicted skipping %, and the ranking within the screen. The left hand AOs are 30mers and the right hand AOs are 25-mers. Based on predicted exon skipping efficiencies, 8 AOs spaced at least 4 bases apart were selected for in vitro screening (Table 2). Target sequence specificities of selected AOs, eteplirsen, and drisapersen were analysed using The University of California, Santa Cruz Genome Browser (http://genome.ucsc.edu/index.html), confirming that the AO sequences theoretically do not bind any non-target RNA sequences with 100% identity.


1.2 Antisense Morpholinos.


All antisense sequences, including analog AOs of eteplirsen and drisapersen, were synthesized with the morpholino chemistry by Gene Tools (Philomath, Oreg.).


1.3 Cells.


Immortalized human skeletal muscle cells derived from three healthy subjects (IDs 8220, CHQ, and KM155) and two DMD patients harbouring deletion mutations of exon 52 (ID KM571) and exons 48-50 (ID 6594) in the DMD gene, respectively, were generated by transduction with human telomerase-expressing and cyclin-dependent kinase 4-expressing vectors in the Institute of Myology human cell immortalization platform, as previously described.33 The three immortalized healthy muscle cell lines were characterized and the clonal line 8220, which showed the highest dystrophin expression was selected as a positive control to prevent overestimation of rescued dystrophin expression in immortalized DMD cells. Primary skeletal muscle cells derived from DMD patients with deletion mutations of ex45-50 (ID 4546) and ex49-50 (ID 4555) and a healthy subject were prepared by the BioBank of Skeletal Muscle, Nerve Tissue, DNA and cell lines.


1.4 AO Transfection.


To mimic as closely as possible the in vivo effects of AO-mediated exon skipping therapy, mature, differentiated myotubes expressing sufficient levels of DMD mRNA were used for in vitro screening. Cells were cultured in proliferation conditions with growth medium (GM): DMEM/F12 with skeletal muscle supplement mix (Promocell, Heidelberg, Germany), 20% fetal bovine serum (Life Technologies, Waltham, Mass.), and antibiotics (50 units penicillin and 50 μg/ml streptomycin, Life Technologies, Waltham, Mass.). Immortalized and primary DMD skeletal muscle cells were seeded at 1.7×104/cm2 and 2.2×104/cm2, respectively, in collagen type I-coated 12 or 24-well culture plates. Two days after seeding, at approximately 80-90% confluence, GM was replaced with differentiation medium (DM): DMEM/F12 with 2% horse serum (GE Healthcare, Chicago, Ill.), lx ITS solution (Sigma, St. Louis, Mo.), and antibiotics. After three days in DM, cells were transfected with AO at 1, 3, 5 or 10 μM containing 6 μM of Endo-porter transfection reagent (Gene Tools, Philomath, Oreg.) (concentrated AOs at 1 mM were incubated at 65° C. for 10 min just before diluting with DM). Two days following AO transfection, AO-containing DM was replaced with regular DM. Cells were harvested at day 2, 5, or 11 after AO transfection (day 5, 8 or 14 following differentiation).


1.5 Mice.


Animal studies were approved by the Animal Care and Use Committee at the University of Alberta, Children's National Medical Center, and National Center of Neurology and Page 27 Header Psychiatry (NCNP). Male and female Dmd exon 52-deficient mdx5242 and wild-type mice (Jackson Laboratory, Bar Harbor, Me.) with a C57BL/6J background were prepared at age 4-8 weeks. Dmd mutation in affected mice was confirmed by genotyping with PCR. A transgenic mouse model harboring the human DMD gene and lacking the mouse Dmd gene (hDMD/Dmd-null mouse) was generated by cross-breeding male hDMD mice (Jackson Laboratory, Bar Harbor, Me.) with female Dmd-null mice.


1.6 Intramuscular Injection.


Mouse version morpholinos of Ac0, Ac48, eteplirsen or drisapersen at 5 or 20 μg in 40 μL saline were intramuscularly injected into tibialis anterior (TA) muscle under inhalation anesthesia with isoflurane as previously described.43 Fifty-μg of Ac0 morpholino and analog eteplirsen in 30 μL saline was injected into TA muscles of hDMD/Dmd-null mice. All muscle samples were harvested 2 weeks after intramuscular injection.


1.7 Exon Skipping Analysis by RT-PCR.


Total RNA was extracted with Trizol (Invitrogen, Waltham, Mass.) as previously described. RT-PCR to detect dystrophin mRNA was performed with the SuperScript III One-Step RT-PCR System (Invitrogen, Waltham, Mass.) and 0.2 μM of forward and reverse primers (see Table 1) for 200 ng and 320 ng of total RNA in immortalized and primary skeletal muscle cells, respectively. Primers were designed using Primer3Plus software and their specificity was confirmed in healthy human skeletal muscle cells (line 8220). The RT-PCR conditions were as follows: 50° C. for 5 minutes; 94° C. for 2 minutes; 35 cycles at 94° C. for 15 seconds, 60° C. for 30 seconds, and 68° C. for 35 seconds; and 68° C. for 5 minutes. PCR products were separated on a 1.5% agarose gel and visualized by SYBR Safe DNA Gel Stain (Invitrogen, Waltham, Mass.). Using ImageJ software (NIH) or the MCE-202 MultiNA system (Shimadzu, Kyoto, Japan), the efficiency of exon 51 skipping was calculated using the following formula:







exon





51

-

skipped





transcript






intensity
/

(

native
+
intermediate
+

exon





51

-

skipped





transcript





intensities


)


×
100


(
%
)




Ex





51

skippedtranscript


Native
+

Ex





51

skippedtranscript



×

d


(
%
)







Unknown top bands above the native band, possibly coming from unexpected splicing events, were excluded from quantification of skipping efficiency. The sequences of the PCR products were confirmed with Big Dye Terminator v3.1 (Applied Biosystems, Waltham, Mass.). GAPDH or 18S ribosomal RNA was used as an internal control.











TABLE 1





Name
Sequence (5′→3′)
Purpose







Ex49/50_94-10_hDMD_Fwd
CAGCCAGTGAAGAGGAAGTTAG
Immortal KM571 DMD cells with ex52


SEQ ID NO. 16

del.





Ex53 80-99 hDMD_Rv
CCAGCCATTGTGTTGAATCC
Primary DMD and healthy cells


SEQ ID NO.-17

hDMDIDmd-null mice





Ex47 60-79 hDMD Fwd
AGGACCCGTGCTTGTAAGTG
Immortal 6594 DMD cells with ex48-50


SEQ ID NO. 18

del.





Ex52_83-105_hDMD_Rv
GATTGTTCTAGCCTCTTGATTGC
Primary 4555 DMD cells with ex49-50


SEQ ID NO. 19

del.





Ex43/44_167-12_hDMD_Fwd
GACAAGGGCGATTTGACAG



SEQ ID NO. 20







Ex52_83-105_hDMD_Rv
GATTGTTCTAGCCTCTTGATTGC
Primary 4546 DMD cells with ex45-50


SEQ ID NO. 19

del.





Ex49/50 94-10 hDMD Fwd
CAGCCAGTGAAGAGGAAGTTAG
Primary healthy cells


SEQ ID NO. 16







Ex52 83-105 hDMD Rv
GATTGTTCTAGCCTCTTGATTGC
hDMD/Dmd-null mice


SEQ ID NO. 19







mDmd_ex49_83-102_Fwd
CAAGCACTCAGCCAGTGAAG



SEQ ID NO. 21







deletion mDmd_ex53_81-100_Rv
TCCAGCCATTGTGTTGAATC
mdx52 mice with ex52


SEQ ID NO. 22







hGAPDH_662-81_Fwd
TCCCTGAGCTGAACGGGAAG



SEQ ID NO. 23







control hGAPDH_860-79_Rv
GGAGGAGTGGGTGTCGCTGT
Internal


SEQ ID NO. 24







h18S_760-82_Fwd
TCGATGCTCTTAGCTGAGTGTCC



SEQ ID NO. 25







control h18S_1039-58_Rv
TGATCGTCTTCGAACCTCCG
Internal


SEQ ID NO. 26









1.8 Western Blotting


Cells were harvested with RIPA buffer (Thermo Scientific, Waltham, Mass.) containing cOmplete, Mini, EDTA-free protease inhibitor cocktail (Roche, Basel, Switzerland), and then homogenized by passing through a 21-gauge needle 10 times. The supernatants as loading samples were prepared by centrifugation at 14,000 g for 15 min at 4° C. Protein from muscle tissues were prepared as previously described. Protein concentrations were adjusted using the Bradford assay with supernatants diluted 100 times with distilled water. Proteins in a sample buffer containing 10% SDS, 70 mM Tris-HCl, pH 6.8, 5 mM EDTA, 20% glycerol, 0.004% bromophenol blue and 5% 2-mercaptoethanol were heated at 70° C. for 10 min. Western blotting was then done as previously described. 32,43,44 Twelve-μg and thirty-μg from cells and tissues, respectively, were used for sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Blots were incubated with a rabbit polyclonal antibody against dystrophin C-terminal (1:2500, ab15278, Abcam, Cambridge, United Kingdom) in the blocking solution or DYS1 antibody against dystrophin rod domain (1:400, Leica Biosystems, Buffalo Grove, Ill.) for 1 hour at room temperature. The primary antibody was reacted with HRP-conjugated anti-rabbit or mouse IgG H+L antibody (1:10,000, Bio-Rad, Hercules, Calif.). Expression levels of dystrophin protein induced by AOs were quantified using calibration curves (R2=0.93-0.99) from dystrophin protein of healthy 8220 skeletal muscle cells diluted with protein from non-treated DMD cells, or wild-type mouse using ImageJ (NIH). Alpha-tubulin was detected on the same membrane as a loading control. Myosin heavy chain (MyHC) on post-transferred gels was stained with Coomassie Brilliant Blue (Bio-Rad, Hercules, Calif.) as a loading control/differentiation marker.


1.9 Immunocytochemistry.


Cells were fixed with 4% paraformaldehyde for 5 min at room temperature. After washing with PBS containing 0.01% Triton X-100, cells were blocked with 10% goat serum (Life Technologies, Waltham, Mass.) in PBS with 0.05% Triton X-100 for 20 min and then incubated with anti-dystrophin C-terminal (ab15278) or rod-domain (DYS1) antibody at 1:50 dilution in blocking solution overnight at 4° C. Dystrophin signals were detected with Alexa 488- or 594-conjugated secondary antibody (1:500). Desmin (1:80, Abcam, Cambridge, United Kingdom) and MyHC-fast type (1:30, Leica Biosystems, Buffalo Grove, Ill.) were detected to confirm myogenic differentiation of cells. Cells were stored in SlowFade Gold Antifade Mountant with DAPI (Invitrogen, Waltham, Mass.) at 4° C. until analysed.


1.10 Immunohistochemistry.


Dystrophin-positive muscle fibers on cryosections from TA muscles of non-treated and treated mdx52 mice were detected with the ab15278 antibody as previously described. Signal intensity of dystrophin in the treated mice was compared with that in wild-type using neutral density filters (Eclipse TE 2000-U, Nikon, Tokyo, Japan).


1.11 Statistical Analysis.


For determining the significance of efficiencies in exon skipping and dystrophin protein rescue, data sets were prepared from at least three independent experiments in immortalized cells, triplicate wells in primary cells, and 3-7 mice. The statistical analysis between AO-treated groups was performed by one-way ANOVA followed by a post hoc Tukey-Kramer multiple comparison test. Simple linear regression analysis was performed for dose-responsiveness to AOs. Statistical significance was set at p<0.05 for all analyses.


2. Results


2.1 in Silico Screening of AOs for Exon 51 Skipping.


We designed total 413 AOs: 204 and 209 AOs with 30-mer and 25-mer lengths, respectively, which cover all possible target sites in DMD exon 51 (see Table 3). Our exon skipping efficiency algorithm (In Silico Screening Based on Predictive Algorithms as a Design Tool for Exon Skipping Oligonucleotides in Duchenne Muscular Dystrophy’ Echigoya et al. PLOS ONE March 2015) predicted that the highest efficiency for exon 51 skipping was 80.5% for 30-mer AOs, and 41.2% for 25-mer AOs in the initial 5′ site of exon 51. In silico screening indicated a very low exon skipping efficiency for the 30-base region targeted by eteplirsen (23.7%), which was ranked 92nd in all 413 AO candidates tested. It is noted that the drisapersen target site is completely encompassed by that of the 30-mer eteplirsen.


2.2 Characterization of Immortalized Clonal Healthy and DMD Skeletal Muscle Cell Lines.


Significant issues in preclinical testing with primary DMD muscle cells include low purity of muscle cells and insufficient amounts of mutant dystrophin mRNA, which present problems when trying to test AO efficacy. To overcome these hurdles, we generated immortalized clonal skeletal muscle cells from 3 healthy subjects and 2 DMD patients with exon 52 (ex52) and ex48-50 deletion (del.) mutations (IDs KM571 and 6594, respectively). All immortalized skeletal muscle cell lines tested expressed easily detectable dystrophin mRNA from day 3 Page 31 Header after induction of differentiation. To avoid overestimation of dystrophin protein levels induced by AOs in DMD cells, we selected a cell line (ID 8220) with the highest level of dystrophin protein among three immortalized healthy skeletal muscle cell lines as determined by Western blotting to serve as a positive control. Dystrophin protein expression in the 8220 cell line was also confirmed by immunocytochemistry.


2.3 In Vitro Screening of Exon 51 Skipping AOs.


Based on the in silico screening results, we selected eight 30-mer AOs, including both high-ranking and low-ranking sequences, spaced at least 4 bases apart from each other for in vitro screening (Table 2). In the present study, all tested AOs, including eteplirsen and drisapersen sequences, were synthesized using the morpholino chemistry that has been demonstrated to be well-tolerated in patients enrolled in clinical trials. Here, we termed control morpholino oligonucleotides having the same sequences as eteplirsen and drisapersen (produced by Gene Tools) as “analog eteplirsen” and “analog drisapersen”. In RT-PCR, 5 of our morpholino AOs (Ac0, Ac5, Ac26, Ac30 and Ac48) at 10 μM showed significantly higher skipping efficiency compared to analog eteplirsen and drisapersen in immortalized DMD skeletal muscle cells harboring ex52 del. (FIG. 1A). Of the tested AOs, Ac0 in particular had the highest skipping efficiency, reaching up to 72%, which was 4 and 25 times more efficient than analogs of eteplirsen and drisapersen, respectively. In Western blotting, Ac0 also induced the highest levels of dystrophin protein, reaching up to 16% of levels in the healthy control cell line, followed by Ac48 at 13% (FIG. 1B). Interestingly, the two AOs, Ac0 and Ac48, with the highest skipping efficiency when tested were not those predicted to be the best from the algorithm.














TABLE 2








Distance
Predicted



Name
Oligo sequence (5′ to 3′)
Length (mer)
from Ac
Skip %
Ranking




















hEx51_Ac9
CCACAGGTTGTGTCACCAGAGTAACAGTCT
30
9
80.5
 1


SEQ NO. 13










hEx51_Ac0
GTGTCACCAGAGTAACAGTCTGAGTAGGAG
30
0
80.1
 2


SEQ ID No. 1










hEx51_Ac5
AGGTTGTGTCACCAGAGTAACAGTCTGAGT
30
5
73.0
 4


SEQ ID NO. 2










hEx51_Ac26
GGCAGTTTCCTTAGTAACCACAGGTTGTGT
30
26
66.3
12


SEQ ID NO. 3










hEx51_Ac30
AGATGGCAGTTTCCTTAGTAACCACAGGTT
30
30
55.5
25


SEQ ID NO. 4










Eteplirsen
CTCCAACATCAAGGAAGATGGCATTTC TAG
30
65
23.7
67


SEQ ID NO. 6










hEx51_Ac48
ATGGCATTTCTAGTTTGGAGATGGCAGTTT
30
48
10.6
128 


SEQ ID NO. 5










hEx51_Ac141
TTATAACTTGATCAAGCAGAGAAAGCCAGT
30
141
 1.8
142 


SEQ ID NO. 14










hEx51_Ac207
atacCTTCTGCTTGATGATCATCTCGTTGA
30
207
NA
NA


SEQ ED NO. 15










Drisapersen
TCAAGGAAGATGGCATTTCT
20
67
NA
NA


SEQ ID NO. 7










hEx51_Ac0-29mer
TGTCACCAGAGTAACAGTCTGAGTAGGAG
29
0
NA
NA


SEQ ID NO. 8










hEx51_Ac0-28mer
GTCACCAGAGTAACAGTCTGAGTAGGAG
28
0
NA
NA


SEQ ID NO. 9










hEx51_Ac0-27mer
TCACCAGAGTAACAGTCTGAGTAGGAG
27
0
NA
NA


SEQ ID NO. 10










hEx51_Ac0-26mer
CACCAGAGTAACAGTCTGAGTAGGAG
26
0
NA
NA


SEQ ID NO. 11










hEx51_Ac0-25mer
ACCAGAGTAACAGTCTGAGTAGGAG
25
0
33.3
 10a


SEQ ID NO. 12










Ac, acceptor splice site.


Uncapitalized nucleotides indicate intronic sequence.


athe ranking in 25-mer AOs.






2.4 Time-Course Analysis with Ac0, Ac48, and Analog AOs of Eteplirsen and Drisapersen.


The persistent effects of Ac0, Ac48, and analogs of eteplirsen and drisapersen at 5 μM were examined in ex52 del. KM571 cells. The superiority of the oligonucleotides Ac0 and Ac48 of the present invention, with respect to exon skipping efficiency and dystrophin protein rescue, was observed at days 2 and 11 post-transfection compared to analog AOs of eteplirsen and drisapersen (FIG. 2).


2.5 Dose-Dependent Effects of Ac0, Ac48, and Analog Eteplirsen and Drisapersen.


RT-PCR showed that Ac0 at the highest concentration of 10 μM induced up to 74% and 64% exon 51 skipping in DMD KM571 (ex52 del.) and 6594 cells (ex48-50 del.), respectively, which were significantly higher than analogs of eteplirsen and drisapersen (FIG. 3). At the lowest concentration (1 μM), Ac0 showed 12 and 10 times higher exon skipping efficiency compared to analog eteplirsen in KM571 and 6594 cells, respectively. Interestingly, even a concentration of 1 μM Ac0 induced higher levels of exon 51 skipping than 10 μM analog eteplirsen (24% efficiency vs 15% in KM571 and 24% efficiency vs 21% in 6594, respectively). Quantitative Western blotting revealed that 10 μM Ac0 rescued dystrophin protein expression in DMD cell lines up to 21% of healthy cell line levels (FIG. 3A to D). Even at 1 μM, the relative ratio of Ac0 to analog eteplirsen represented 7.1 and 3.3 times higher efficiency in producing dystrophin protein in KM571 and 6594 cell lines, respectively. Ac0 at 1 μM enabled the production of rescued dystrophin protein at higher or comparable levels than analog eteplirsen at 10 μM (10% vs 6% in KM571 and 11% vs 10% in 6594, respectively), confirming that Ac0 is more than 10-fold effective at producing Page 33 Header dystrophin protein compared to analog eteplirsen concentration-wise. Analog drisapersen did not work effectively for either of exon skipping or dystrophin production in the DMD muscle cell lines. The exon skipping response to Ac0 and Ac48 occurred in a dose-dependent manner that was greater than both analog eteplirsen, and analog drisapersen (FIGS. 3A and C). The dose-responsiveness of Ac0 with respect to dystrophin protein production was also higher than the control analogs in both DMD cell lines (FIG. 3E).


2.6 Immunocytochemical Assessment of Dystrophin Protein Rescue.


Immunocytochemistry revealed that Ac0 and Ac48 at 10 μM yielded more dystrophin-positive myotubes and displayed stronger signal intensity in DMD skeletal muscle cell lines harboring ex52 and ex48-50 del. mutations compared to analog eteplirsen (FIG. 4).


2.7 Length Optimization of Ac0 Morpholino.


In silico and in vitro screening revealed that the initial 5′ region of exon 51 between 0 and +89 is an important region for influencing exon 51 skipping. To optimize the sequence length of Ac0 targeting this region, we compared the skipping efficiencies of Ac0 morpholinos of different lengths (25- to 30-mer), in which nucleotides at the 5′ site were systematically removed one at a time (see Table 2). In vitro testing in immortalized DMD muscle cells treated with 1 μM of these AOs showed that 25-30-mer Ac0 morpholinos produced efficient exon skipping (>20%) (FIG. 5), an effect that was not observed analog eteplirsen, and analog drisapersen at the same dose (FIG. 3). However, the efficiency of exon skipping increased as the length of the AO was increased. The statistically significant effectiveness of 30-mer Ac0 was confirmed at 1 and 3 μM doses compared to the shorter Ac0 morpholinos in both cell lines, even those AOs that are only 1 or 2 bases shorter.


2.8 Effect of Ac0, Ac48, and Analog Eteplirsen and Drisapersen on Primary DMD Patient-Derived Skeletal Muscle Cells.


We also tested the AOs in primary DMD skeletal muscle cells with exons 45-50 (ID 4546) or exons 49-50 del. mutations (ID 4555) to validate if the superior efficacy of 30-mer Ac0 is consistent for other muscle cell types and deletion mutation patterns. RT-PCR showed that Ac0 achieved significantly higher exon skipping efficiency in both primary DMD muscle cells compared to analog eteplirsen, or analog drisapersen (FIG. 6A to D): up to 5 and 7 times higher efficiency were observed compared to analog eteplirsen and drisapersen, respectively. A significant efficiency of Ac0-mediated exon 51 skipping was also confirmed Page 34 Header in primary healthy skeletal muscle cells (FIGS. 6E and F). Interestingly, with increasing exon 51 skipping efficiency, spontaneous exon 52 skipping, which does not disrupt the reading frame, was observed in primary healthy and DMD muscle cells, and an immortalized DMD muscle cell line with ex48-50 del. (6594).


2.10 In Vivo Efficacy of Ac0 Morpholino and Analog Eteplirsen in hDMD/Dmd-Null Mice.


A major hurdle in the development of exon skipping therapy is that human-specific AOs cannot always be tested in an appropriate animal model. This limits the evaluation of in vivo effects of AOs designed for patients. Here, we developed a new mouse model that has the full-length human DMD gene but lacks the entire mouse Dmd gene (hDMD/Dmd-null) to test the in vivo efficacy of human AOs. This mouse model was employed to avoid the cross-reaction between human sequences and mouse sequences (note that conventional mdx mice still have the mouse dystrophin mRNA, which can cross-react with human-targeting AOs), and obtained by cross-breeding between hDMD mice34 and Dmd-null mice35. Ac0, Ac48, analog eteplirsen or analog drisapersen was injected into TA muscles of these mice, and the effectiveness of in vivo exon 51 skipping was analysed 2 weeks after the injection. The result showed significantly greater exon skipping efficiency in mice treated with Ac0 compared to analog eteplirsen (FIG. 7). Visible exon 51-skipped bands were found in Ac48-treated mice, with an average exon skipping efficiency of 1.11% (±0.46%, SE). On the other hand, no quantifiable exon 51-skipped bands were observed in mice treated with analog drisapersen (FIG. 8).












SEQUENCES
















GTGTCACCAGAGTAACAGTCTGAGTAGGAG
Ac0 (SEQ ID NO. 1)





AGGTTGTGTCACCAGAGTAACAGTCTGAGT
Ac5 (SEQ ID NO. 2)





GGCAGTTTCCTTAGTAACCACAGGTTGTGT
Ac26 (SEQ ID NO. 3)





AGATGGCAGTTTCCTTAGTAACCACAGGTT
Ac30 (SEQ ID NO. 4)





ATGGCATTTCTAGTTTGGAGATGGCAGTTT
Ac48 (SEQ ID NO. 5)





CTCCAACATCAAGGAAGATGGCATTTCTAG
Eteplirsen (SEQ ID NO. 6)





TCAAGGAAGATGGCATTTCT
Drisapersen (SEQ ID NO. 7)





TGTCACCAGAGTAACAGTCTGAGTAGGAG
hEx51_Ac0-29mer (SEQ ID NO. 8)





GTCACCAGAGTAACAGTCTGAGTAGGAG
hEx51_Ac0-28mer (SEQ ID NO. 9)





TCACCAGAGTAACAGTCTGAGTAGGAG
hEx51_Ac0-27mer (SEQ ID NO. 10)





CACCAGAGTAACAGTCTGAGTAGGAG
hEx51_Ac0-26mer (SEQ ID NO. 11)





ACCAGAGTAACAGTCTGAGTAGGAG
hEx51_Ac0-25mer (SEQ ID NO. 12)





CCACAGGTTGTGTCACCAGAGTAACAGTCT
Ac9 (SEQ ID NO. 13)





TTATAACTTGATCAAGCAGAGAAAGCCAGT
Ac141 (SEQ ID NO. 14)





atacCTTCTGCTTGATGATCATCTCGTTGA
Ac207 (SEQ ID NO. 15)





CAGCCAGTGAAGAGGAAGTTAG
Ex49/50_94-10_hDMD_Fwd (SEQ ID NO. 16)





CCAGCCATTGTGTTGAATCC
Ex53_80-99_hDMD_Rv (SEQ ID NO. 17)





AGGACCCGTGCTTGTAAGTG
Ex47_60-79_hDMD_Fwd (SEQ ID NO. 18)





GATTGTTCTAGCCTCTTGATTGC
Ex52_83-105_hDMD_Rv (SEQ ID NO. 19)





GACAAGGGCGATTTGACAG
Ex43/44_167-12_hDMD_Fwd (SEQ ID NO. 20)





CAAGCACTCAGCCAGTGAAG
mDmd_ex49_83-102_Fwd (SEQ ID NO 21)





TCCAGCCATTGTGTTGAATC
deletionmDmd_ex53_81-100_Rv (SEQ ID NO. 22)





TCCCTGAGCTGAACGGGAAG
hGAPDH_662-81_Fwd (SEQ ID NO. 23)





TCCAGCCATTGTGTTGAATC
controlhGAPDH_860-79_Rv (SEQ ID NO. 24)





TCGATGCTCTTAGCTGAGTGTCC
h18S_760-82_Fwd (SEQ ID NO. 25)





TGATCGTCTTCGAACCTCCG
control h18S_1039-58_Rv (SEQ ID NO. 26)
























TABLE 3







51
9
CCACAGGTTGTGTCACCAGAGTAACAGTCT
80.49
1
18
GTAACCACAGGTTGTGTCACCAGAG
41.20
1





51
0
GTGTCACCAGAGTAACAGTCTGAGTAGGAG
80.11
2
16
AACCACAGGTTGTGTCACCAGAGTA
39.94
2





51
10
ACCACAGGTTGTGTCACCAGAGTAACAGTC
79.98
3
12
ACAGGTTGTGTCACCAGAGTAACAG
38.08
3





51
5
AGGTTGTGTCACCAGAGTAACAGTCTGAGT
72.97
4
14
CCACAGGTTGTGTCACCAGAGTAAC
37.52
4





51
8
CACAGGTTGTGTCACCAGAGTAACAGTCTG
72.01
5
15
ACCACAGGTTGTGTCACCAGAGTAA
37.23
5





51
1
TGTGTCACCAGAGTAACAGTCTGAGTAGGA
71.94
6
31
GGCAGTTTCCTTAGTAACCACAGGT
37.18
6





51
2
TTGTGTCACCAGAGTAACAGTCTGAGTAGG
71.51
7
13
CACAGGTTGTGTCACCAGAGTAACA
36.66
7





51
11
AACCACAGGTTGTGTCACCAGAGTAACAGT
70.65
8
10
AGGTTGTGTCACCAGAGTAACAGTC
35.56
8





51
6
CAGGTTGTGTCACCAGAGTAACAGTCTGAG
68.18
9
11
CAGGTTGTGTCACCAGAGTAACAGT
33.75
9





51
7
ACAGGTTGTGTCACCAGAGTAACAGTCTGA
68.14
10
0
ACCAGAGTAACAGTCTGAGTAGGAG
33.34
10





51
4
GGTTGTGTCACCAGAGTAACAGTCTGAGTA
66.65
11
9
GGTTGTGTCACCAGAGTAACAGTCT
33.10
11





51
26
GGCAGTTTCCTTAGTAACCACAGGTTGTGT
66.32
12
17
TAACCACAGGTTGTGTCACCAGAGT
32.95
12





51
18
CCTTAGTAACCACAGGTTGTGTCACCAGAG
65.25
13
32
TGGCAGTTTCCTTAGTAACCACAGG
32.77
13





51
19
TCCTTAGTAACCACAGGTTGTGTCACCAGA
64.81
14
30
GCAGTTTCCTTAGTAACCACAGGTT
31.61
14





51
27
TGGCAGTTTCCTTAGTAACCACAGGTTGTG
64.09
15
19
AGTAACCACAGGTTGTGTCACCAGA
30.95
15





51
12
TAACCACAGGTTGTGTCACCAGAGTAACAG
64.08
16
23
CCTTAGTAACCACAGGTTGTGTCAC
30.66
16





51
13
GTAACCACAGGTTGTGTCACCAGAGTAACA
63.65
17
5
GTGTCACCAGAGTAACAGTCTGAGT
30.54
17





51
25
GCAGTTTCCTTAGTAACCACAGGTTGTGTC
61.81
18
1
CACCAGAGTAACAGTCTGAGTAGGA
30.26
18





51
29
GATGGCAGTTTCCTTAGTAACCACAGGTTG
61.44
19
6
TGTGTCACCAGAGTAACAGTCTGAG
29.52
19





51
14
AGTAACCACAGGTTGTGTCACCAGAGTAAC
57.56
20
24
TCCTTAGTAACCACAGGTTGTGTCA
28.97
20





51
23
AGTTTCCTTAGTAACCACAGGTTGTGTCAC
57.29
21
2
TCACCAGAGTAACAGTCTGAGTAGG
28.83
21





51
3
GTTGTGTCACCAGAGTAACAGTCTGAGTAG
56.65
22
3
GTCACCAGAGTAACAGTCTGAGTAG
26.60
22





51
17
CTTAGTAACCACAGGTTGTGTCACCAGAGT
56.39
23
25
TTCCTTAGTAACCACAGGTTGTGTC
26.23
23





51
24
CAGTTTCCTTAGTAACCACAGGTTGTGTCA
56.16
24
27
GTTTCCTTAGTAACCACAGGTTGTG
26.11
24





51
30
AGATGGCAGTTTCCTTAGTAACCACAGGTT
55.46
25
29
CAGTTTCCTTAGTAACCACAGGTTG
24.24
25





51
20
TTCCTTAGTAACCACAGGTTGTGTCACCAG
53.39
26
34
GATGGCAGTTTCCTTAGTAACCACA
22.08
26





51
16
TTAGTAACCACAGGTTGTGTCACCAGAGTA
53.04
27
8
GTTGTGTCACCAGAGTAACAGTCTG
21.25
27





51
15
TAGTAACCACAGGTTGTGTCACCAGAGTAA
52.28
28
7
TTGTGTCACCAGAGTAACAGTCTGA
21.05
28





51
22
GTTTCCTTAGTAACCACAGGTTGTGTCACC
51.90
29
4
TGTCACCAGAGTAACAGTCTGAGTA
20.02
29





51
28
ATGGCAGTTTCCTTAGTAACCACAGGTTGT
46.50
30
26
TTTCCTTAGTAACCACAGGTTGTGT
19.62
30





51
21
TTTCCTTAGTAACCACAGGTTGTGTCACCA
45.73
31
22
CTTAGTAACCACAGGTTGTGTCACC
18.61
31





51
31
GAGATGGCAGTTTCCTTAGTAACCACAGGT
43.71
32
33
ATGGCAGTTTCCTTAGTAACCACAG
18.23
32





51
32
GGAGATGGCAGTTTCCTTAGTAACCACAGG
38.58
33
108
TCTGTCCAAGCCCGGTTGAAATCTG
18.06
33





51
98
CCAAGCCCGGTTGAAATCTGCCAGAGCAGG
36.79
34
87
TCTGCCAGAGCAGGTACCTCCAACA
18.04
34





51
77
CAGAGCAGGTACCTCCAACATCAAGGAAGA
36.22
35
98
CCCGGTTGAAATCTGCCAGAGCAGG
17.52
35





51
46
GGCATTTCTAGTTTGGAGATGGCAGTTTCC
35.05
36
28
AGTTTCCTTAGTAACCACAGGTTGT
16.78
36





51
102
CTGTCCAAGCCCGGTTGAAATCTGCCAGAG
33.96
37
35
AGATGGCAGTTTCCTTAGTAACCAC
16.49
37





51
103
TCTGTCCAAGCCCGGTTGAAATCTGCCAGA
33.85
38
20
TAGTAACCACAGGTTGTGTCACCAG
16.42
38





51
78
CCAGAGCAGGTACCTCCAACATCAAGGAAG
32.83
39
83
CCAGAGCAGGTACCTCCAACATCAA
15.89
39





51
100
GTCCAAGCCCGGTTGAAATCTGCCAGAGCA
32.12
40
86
CTGCCAGAGCAGGTACCTCCAACAT
15.09
40





51
101
TGTCCAAGCCCGGTTGAAATCTGCCAGAGC
31.85
41
82
CAGAGCAGGTACCTCCAACATCAAG
14.95
41





51
53
GGAAGATGGCATTTCTAGTTTGGAGATGGC
31.58
42
84
GCCAGAGCAGGTACCTCCAACATCA
14.91
42





51
99
TCCAAGCCCGGTTGAAATCTGCCAGAGCAG
31.36
43
85
TGCCAGAGCAGGTACCTCCAACATC
14.66
43





51
106
AGTTCTGTCCAAGCCCGGTTGAAATCTGCC
31.20
44
110
GTTCTGTCCAAGCCCGGTTGAAATC
13.87
44





51
33
TGGAGATGGCAGTTTCCTTAGTAACCACAG
30.45
45
107
CTGTCCAAGCCCGGTTGAAATCTGC
12.76
45





51
105
GTTCTGTCCAAGCCCGGTTGAAATCTGCCA
29.76
46
109
TTCTGTCCAAGCCCGGTTGAAATCT
12.71
46





51
104
TTCTGTCCAAGCCCGGTTGAAATCTGCCAG
29.45
47
99
GCCCGGTTGAAATCTGCCAGAGCAG
12.07
47





51
117
GCCAGTCGGTAAGTTCTGTCCAAGCCCGGT
28.99
48
81
AGAGCAGGTACCTCCAACATCAAGG
11.09
48





51
87
TGAAATCTGCCAGAGCAGGTACCTCCAACA
27.92
49
36
GAGATGGCAGTTTCCTTAGTAACCA
10.96
49





51
37
AGTTTGGAGATGGCAGTTTCCTTAGTAACC
27.37
50
51
GGCATTTCTAGTTTGGAGATGGCAG
10.71
50





51
97
CAAGCCCGGTTGAAATCTGCCAGAGCAGGT
27.25
51
111
AGTTCTGTCCAAGCCCGGTTGAAAT
10.66
51





51
40
TCTAGTTTGGAGATGGCAGTTTCCTTAGTA
27.24
52
21
TTAGTAACCACAGGTTGTGTCACCA
10.43
52





51
76
AGAGCAGGTACCTCCAACATCAAGGAAGAT
27.06
53
106
TGTCCAAGCCCGGTTGAAATCTGCC
10.38
53





51
81
CTGCCAGAGCAGGTACCTCCAACATCAAGG
26.57
54
112
AAGTTCTGTCCAAGCCCGGTTGAAA
9.48
54





51
95
AGCCCGGTTGAAATCTGCCAGAGCAGGTAC
26.37
55
115
GGTAAGTTCTGTCCAAGCCCGGTTG
9.33
55





51
86
GAAATCTGCCAGAGCAGGTACCTCCAACAT
25.98
56
50
GCATTTCTAGTTTGGAGATGGCAGT
9.05
56





51
80
TGCCAGAGCAGGTACCTCCAACATCAAGGA
25.94
57
101
AAGCCCGGTTGAAATCTGCCAGAGC
8.92
57





51
96
AAGCCCGGTTGAAATCTGCCAGAGCAGGTA
25.55
58
103
CCAAGCCCGGTTGAAATCTGCCAGA
8.24
58





51
79
GCCAGAGCAGGTACCTCCAACATCAAGGAA
25.55
59
113
TAAGTTCTGTCCAAGCCCGGTTGAA
7.96
59





51
108
TAAGTTCTGTCCAAGCCCGGTTGAAATCTG
25.54
60
105
GTCCAAGCCCGGTTGAAATCTGCCA
7.95
60





51
90
GGTTGAAATCTGCCAGAGCAGGTACCTCCA
25.44
61
53
ATGGCATTTCTAGTTTGGAGATGGC
7.67
61





51
50
AGATGGCATTTCTAGTTTGGAGATGGCAGT
25.21
62
100
AGCCCGGTTGAAATCTGCCAGAGCA
7.33
62





51
89
GTTGAAATCTGCCAGAGCAGGTACCTCCAA
24.68
63
116
CGGTAAGTTCTGTCCAAGCCCGGTT
7.17
63





51
94
GCCCGGTTGAAATCTGCCAGAGCAGGTACC
23.96
64
97
CCGGTTGAAATCTGCCAGAGCAGGT
6.96
64





51
88
TTGAAATCTGCCAGAGCAGGTACCTCCAAC
23.90
65
117
TCGGTAAGTTCTGTCCAAGCCCGGT
6.90
65





51
34
TTGGAGATGGCAGTTTCCTTAGTAACCACA
23.77
66
102
CAAGCCCGGTTGAAATCTGCCAGAG
6.69
66





51
65
CTCCAACATCAAGGAAGATGGCATTTCTAG
23.66
67
114
GTAAGTTCTGTCCAAGCCCGGTTGA
6.65
67





51
35
TTTGGAGATGGCAGTTTCCTTAGTAACCAC
23.53
68
104
TCCAAGCCCGGTTGAAATCTGCCAG
6.63
68





51
91
CGGTTGAAATCTGCCAGAGCAGGTACCTCC
23.52
69
91
GAAATCTGCCAGAGCAGGTACCTCC
6.24
69





51
45
GCATTTCTAGTTTGGAGATGGCAGTTTCCT
23.40
70
37
GGAGATGGCAGTTTCCTTAGTAACC
6.17
70





51
57
TCAAGGAAGATGGCATTTCTAGTTTGGAGA
23.34
71
88
ATCTGCCAGAGCAGGTACCTCCAAC
6.14
71





51
118
AGCCAGTCGGTAAGTTCTGTCCAAGCCCGG
23.29
72
118
GTCGGTAAGTTCTGTCCAAGCCCGG
5.97
72





51
82
TCTGCCAGAGCAGGTACCTCCAACATCAAG
22.86
73
95
GGTTGAAATCTGCCAGAGCAGGTAC
5.25
73





51
93
CCCGGTTGAAATCTGCCAGAGCAGGTACCT
22.81
74
57
GAAGATGGCATTTCTAGTTTGGAGA
5.02
74





51
107
AAGTTCTGTCCAAGCCCGGTTGAAATCTGC
22.77
75
46
TTCTAGTTTGGAGATGGCAGTTTCC
5.00
75





51
116
CCAGTCGGTAAGTTCTGTCCAAGCCCGGTT
22.52
76
47
TTTCTAGTTTGGAGATGGCAGTTTC
4.87
76





51
109
GTAAGTTCTGTCCAAGCCCGGTTGAAATCT
22.51
77
89
AATCTGCCAGAGCAGGTACCTCCAA
4.60
77





51
110
GGTAAGTTCTGTCCAAGCCCGGTTGAAATC
22.46
78
58
GGAAGATGGCATTTCTAGTTTGGAG
4.30
78





51
92
CCGGTTGAAATCTGCCAGAGCAGGTACCTC
22.07
79
96
CGGTTGAAATCTGCCAGAGCAGGTA
3.79
79





51
36
GTTTGGAGATGGCAGTTTCCTTAGTAACCA
21.97
80
131
GCAGAGAAAGCCAGTCGGTAAGTTC
3.63
80





51
56
CAAGGAAGATGGCATTTCTAGTTTGGAGAT
21.85
81
80
GAGCAGGTACCTCCAACATCAAGGA
3.51
81





51
75
GAGCAGGTACCTCCAACATCAAGGAAGATG
21.81
82
120
CAGTCGGTAAGTTCTGTCCAAGCCC
3.35
82





51
39
CTAGTTTGGAGATGGCAGTTTCCTTAGTAA
21.62
83
128
GAGAAAGCCAGTCGGTAAGTTCTGT
3.12
83





51
64
TCCAACATCAAGGAAGATGGCATTTCTAGT
21.18
84
119
AGTCGGTAAGTTCTGTCCAAGCCCG
3.09
84





51
115
CAGTCGGTAAGTTCTGTCCAAGCCCGGTTG
20.90
85
92
TGAAATCTGCCAGAGCAGGTACCTC
2.10
85





51
52
GAAGATGGCATTTCTAGTTTGGAGATGGCA
20.77
86
40
TTTGGAGATGGCAGTTTCCTTAGTA
1.87
86





51
63
CCAACATCAAGGAAGATGGCATTTCTAGTT
20.61
87
90
AAATCTGCCAGAGCAGGTACCTCCA
1.74
87





51
111
CGGTAAGTTCTGTCCAAGCCCGGTTGAAAT
20.60
88
129
AGAGAAAGCCAGTCGGTAAGTTCTG
1.57
88





51
41
TTCTAGTTTGGAGATGGCAGTTTCCTTAGT
20.19
89
121
CCAGTCGGTAAGTTCTGTCCAAGCC
1.52
89





51
84
AATCTGCCAGAGCAGGTACCTCCAACATCA
19.86
90
122
GCCAGTCGGTAAGTTCTGTCCAAGC
0.99
90





51
113
GTCGGTAAGTTCTGTCCAAGCCCGGTTGAA
19.80
91
38
TGGAGATGGCAGTITCCITAGTAAC
0.44
91





51
114
AGTCGGTAAGTTCTGTCCAAGCCCGGTTGA
19.75
92
45
TCTAGTTTGGAGATGGCAGTTTCCT
0.40
92





51
73
GCAGGTACCTCCAACATCAAGGAAGATGGC
19.30
93
78
GCAGGTACCTCCAACATCAAGGAAG
0.10
93





51
38
TAGTTTGGAGATGGCAGTTTCCTTAGTAAC
19.21
94
48
ATTTCTAGTTTGGAGATGGCAGTTT
−0.21
94





51
119
AAGCCAGTCGGTAAGTTCTGTCCAAGCCCG
19.04
95
125
AAAGCCAGTCGGTAAGTTCTGTCCA
−0.23
95





51
67
ACCTCCAACATCAAGGAAGATGGCATTTCT
19.03
96
126
GAAAGCCAGTCGGTAAGTTCTGTCC
−0.58
96





51
83
ATCTGCCAGAGCAGGTACCTCCAACATCAA
18.98
97
39
TTGGAGATGGCAGTTTCCTTAGTAA
−0.64
97





51
58
ATCAAGGAAGATGGCATTTCTAGTTTGGAG
18.74
98
134
CAAGCAGAGAAAGCCAGTCGGTAAG
−0.80
98





51
112
TCGGTAAGTTCTGTCCAAGCCCGGTTGAAA
18.59
99
132
AGCAGAGAAAGCCAGTCGGTAAGTT
−0.92
99





51
54
AGGAAGATGGCATTTCTAGTTTGGAGATGG
18.20
100
137
GATCAAGCAGAGAAAGCCAGTCGGT
−0.94
100





51
85
AAATCTGCCAGAGCAGGTACCTCCAACATC
17.93
101
133
AAGCAGAGAAAGCCAGTCGGTAAGT
−1.00
101





51
66
CCTCCAACATCAAGGAAGATGGCATTTCTA
17.65
102
138
TGATCAAGCAGAGAAAGCCAGTCGG
−1.03
102





51
49
GATGGCATTTCTAGTTTGGAGATGGCAGTT
17.50
103
127
AGAAAGCCAGTCGGTAAGTTCTGTC
−1.07
103





51
42
TTTCTAGTTTGGAGATGGCAGTTTCCTTAG
17.40
104
136
ATCAAGCAGAGAAAGCCAGTCGGTA
−1.36
104





51
44
CATTTCTAGTTTGGAGATGGCAGTTTCCTT
16.99
105
72
ACCTCCAACATCAAGGAAGATGGCA
−1.50
105





51
51
AAGATGGCATTTCTAGTTTGGAGATGGCAG
16.77
106
79
AGCAGGTACCTCCAACATCAAGGAA
−1.50
106





51
55
AAGGAAGATGGCATTTCTAGTTTGGAGATG
16.50
107
130
CAGAGAAAGCCAGTCGGTAAGTTCT
−1.79
107





51
68
TACCTCCAACATCAAGGAAGATGGCATTTC
16.36
108
94
GTTGAAATCTGCCAGAGCAGGTACC
−1.88
108





51
121
GAAAGCCAGTCGGTAAGTTCTGTCCAAGCC
16.27
109
59
AGGAAGATGGCATTTCTAGTTTGGA
−2.14
109





51
120
AAAGCCAGTCGGTAAGTTCTGTCCAAGCCC
16.02
110
56
AAGATGGCATTTCTAGTTTGGAGAT
−2.16
110





51
74
AGCAGGTACCTCCAACATCAAGGAAGATGG
15.45
111
93
TTGAAATCTGCCAGAGCAGGTACCT
−2.19
111





51
122
AGAAAGCCAGTCGGTAAGTTCTGTCCAAGC
15.40
112
140
CTTGATCAAGCAGAGAAAGCCAGTC
−2.58
112





51
59
CATCAAGGAAGATGGCATTTCTAGTTTGGA
15.35
113
70
CTCCAACATCAAGGAAGATGGCATT
−2.88
113





51
123
GAGAAAGCCAGTCGGTAAGTTCTGTCCAAG
15.01
114
71
CCTCCAACATCAAGGAAGATGGCAT
−2.97
114





51
69
GTACCTCCAACATCAAGGAAGATGGCATTT
14.79
115
52
TGGCATTTCTAGTTTGGAGATGGCA
−3.00
115





51
60
ACATCAAGGAAGATGGCATTTCTAGTTTGG
14.56
116
63
ATCAAGGAAGATGGCATTTCTAGTT
−3.06
116





51
72
CAGGTACCTCCAACATCAAGGAAGATGGCA
13.59
117
42
AGTTTGGAGATGGCAGTTTCCTTAG
−3.16
117





51
124
AGAGAAAGCCAGTCGGTAAGTTCTGTCCAA
12.92
118
54
GATGGCATTTCTAGTTTGGAGATGG
−3.16
118





51
70
GGTACCTCCAACATCAAGGAAGATGGCATT
12.45
119
49
CATTTCTAGTTTGGAGATGGCAGTT
−3.37
119





51
71
AGGTACCTCCAACATCAAGGAAGATGGCAT
12.24
120
135
TCAAGCAGAGAAAGCCAGTCGGTAA
−3.39
120





51
62
CAACATCAAGGAAGATGGCATTTCTAGTTT
12.18
121
123
AGCCAGTCGGTAAGTTCTGTCCAAG
−3.44
121





51
61
AACATCAAGGAAGATGGCATTTCTAGTTTG
12.09
122
141
ACTTGATCAAGCAGAGAAAGCCAGT
−3.99
122





51
126
GCAGAGAAAGCCAGTCGGTAAGTTCTGTCC
12.04
123
68
CCAACATCAAGGAAGATGGCATTTC
−4.03
123





51
125
CAGAGAAAGCCAGTCGGTAAGTTCTGTCCA
11.49
124
124
AAGCCAGTCGGTAAGTTCTGTCCAA
−4.20
124





51
43
ATTTCTAGTTTGGAGATGGCAGTTTCCTTA
11.13
125
60
AAGGAAGATGGCATTTCTAGTTTGG
−4.22
125





51
47
TGGCATTTCTAGTTTGGAGATGGCAGTTTC
11.09
126
55
AGATGGCATTTCTAGTTTGGAGATG
−4.42
126





51
129
CAAGCAGAGAAAGCCAGTCGGTAAGTTCTG
10.80
127
142
AACTTGATCAAGCAGAGAAAGCCAG
−4.53
127





51
48
ATGGCATTTCTAGTTTGGAGATGGCAGTTT
10.61
128
73
TACCTCCAACATCAAGGAAGATGGC
−4.70
128





51
130
TCAAGCAGAGAAAGCCAGTCGGTAAGTTCT
10.43
129
139
TTGATCAAGCAGAGAAAGCCAGTCG
−4.97
129





51
128
AAGCAGAGAAAGCCAGTCGGTAAGTTCTGT
9.84
130
41
GTTTGGAGATGGCAGTTTCCTTAGT
−5.02
130





51
131
ATCAAGCAGAGAAAGCCAGTCGGTAAGTTC
8.53
131
74
GTACCTCCAACATCAAGGAAGATGG
−5.30
131





51
127
AGCAGAGAAAGCCAGTCGGTAAGTTCTGTC
8.26
132
64
CATCAAGGAAGATGGCATTTCTAGT
−5.36
132





51
136
ACTTGATCAAGCAGAGAAAGCCAGTCGGTA
6.43
133
65
ACATCAAGGAAGATGGCATTTCTAG
−5.43
133





51
137
AACTTGATCAAGCAGAGAAAGCCAGTCGGT
5.84
134
44
CTAGTTTGGAGATGGCAGTTTCCTT
−6.16
134





51
138
TAACTTGATCAAGCAGAGAAAGCCAGTCGG
5.26
135
61
CAAGGAAGATGGCATTTCTAGTTTG
−6.27
135





51
132
GATCAAGCAGAGAAAGCCAGTCGGTAAGTT
4.60
136
172
GTCACCCACCATCACCCTCTGTGAT
−7.68
136





51
140
TATAACTTGATCAAGCAGAGAAAGCCAGTC
4.36
137
69
TCCAACATCAAGGAAGATGGCATTT
−7.84
137





51
133
TGATCAAGCAGAGAAAGCCAGTCGGTAAGT
4.31
138
143
TAACTTGATCAAGCAGAGAAAGCCA
−8.29
138





51
134
TTGATCAAGCAGAGAAAGCCAGTCGGTAAG
3.75
139
62
TCAAGGAAGATGGCATTTCTAGTTT
−8.30
139





51
139
ATAACTTGATCAAGCAGAGAAAGCCAGTCG
2.70
140
77
CAGGTACCTCCAACATCAAGGAAGA
−8.53
140





51
135
CTTGATCAAGCAGAGAAAGCCAGTCGGTAA
2.28
141
67
CAACATCAAGGAAGATGGCATTTCT
−8.81
141





51
141
TTATAACTTGATCAAGCAGAGAAAGCCAGT
1.76
142
173
GGTCACCCACCATCACCCTCTGTGA
−8.87
142





51
142
TTTATAACTTGATCAAGCAGAGAAAGCCAG
−0.17
143
144
ATAACTTGATCAAGCAGAGAAAGCC
−8.98
143





51
146
TGATTTTATAACTTGATCAAGCAGAGAAAG
−3.06
144
75
GGTACCTCCAACATCAAGGAAGATG
−9.18
144





51
145
GATTTTATAACTTGATCAAGCAGAGAAAGC
−3.17
145
174
AGGTCACCCACCATCACCCTCTGTG
−9.52
145





51
156
TCACCCTCTGTGATTTTATAACTTGATCAA
−3.49
146
66
AACATCAAGGAAGATGGCATTTCTA
−9.58
146





51
167
GTCACCCACCATCACCCTCTGTGATTTTAT
−3.97
147
43
TAGTTTGGAGATGGCAGTTTCCTTA
−9.61
147





51
143
TTTTATAACTTGATCAAGCAGAGAAAGCCA
−4.01
148
146
TTATAACTTGATCAAGCAGAGAAAG
−9.74
148





51
174
CCTCAAGGTCACCCACCATCACCCTCTGTG
−4.37
149
171
TCACCCACCATCACCCTCTGTGATT
−10.00
149





51
144
ATTTTATAACTTGATCAAGCAGAGAAAGCC
−4.66
150
170
CACCCACCATCACCCTCTGTGATTT
−10.19
150





51
155
CACCCTCTGTGATTTTATAACTTGATCAAG
−5.42
151
147
TTTATAACTTGATCAAGCAGAGAAA
−10.27
151





51
168
GGTCACCCACCATCACCCTCTGTGATTTTA
−5.53
152
176
CAAGGTCACCCACCATCACCCTCTG
−11.06
152





51
175
TCCTCAAGGTCACCCACCATCACCCTCTGT
−5.64
153
145
TATAACTTGATCAAGCAGAGAAAGC
−11.20
153





51
157
ATCACCCTCTGTGATTTTATAACTTGATCA
−5.75
154
168
CCCACCATCACCCTCTGTGATTTTA
−11.23
154





51
173
CTCAAGGTCACCCACCATCACCCTCTGTGA
−6.14
155
169
ACCCACCATCACCCTCTGTGATTTT
−11.29
155





51
176
ATCCTCAAGGTCACCCACCATCACCCTCTG
−6.36
156
177
TCAAGGTCACCCACCATCACCCTCT
−11.45
156





51
172
TCAAGGTCACCCACCATCACCCTCTGTGAT
−6.44
157
181
ATCCTCAAGGTCACCCACCATCACC
−11.50
157





51
166
TCACCCACCATCACCCTCTGTGATTTTATA
−6.45
158
175
AAGGTCACCCACCATCACCCTCTGT
−11.55
158





51
163
CCCACCATCACCCTCTGTGATTTTATAACT
−6.75
159
180
TCCTCAAGGTCACCCACCATCACCC
−11.59
159





51
161
CACCATCACCCTCTGTGATTTTATAACTTG
−6.93
160
179
CCTCAAGGTCACCCACCATCACCCT
−11.76
160





51
147
GTGATTTTATAACTTGATCAAGCAGAGAAA
−7.04
161
183
ATATCCTCAAGGTCACCCACCATCA
−11.76
161





51
164
ACCCACCATCACCCTCTGTGATTTTATAAC
−7.04
162
178
CTCAAGGTCACCCACCATCACCCTC
−11.79
162





51
171
CAAGGTCACCCACCATCACCCTCTGTGATT
−7.07
163
76
AGGTACCTCCAACATCAAGGAAGAT
−12.00
163





51
169
AGGTCACCCACCATCACCCTCTGTGATTTT
−7.08
164
182
TATCCTCAAGGTCACCCACCATCAC
−12.19
164





51
153
CCCTCTGTGATTTTATAACTTGATCAAGCA
−7.09
165
156
CTCTGTGATTTTATAACTTGATCAA
−12.23
165





51
165
CACCCACCATCACCCTCTGTGATTTTATAA
−7.12
166
167
CCACCATCACCCTCTGTGATTTTAT
−12.52
166





51
178
ATATCCTCAAGGTCACCCACCATCACCCTC
−7.12
167
157
CCTCTGTGATTTTATAACTTGATCA
−12.99
167





51
160
ACCATCACCCTCTGTGATTTTATAACTTGA
−7.13
168
184
GATATCCTCAAGGTCACCCACCATC
−13.02
168





51
177
TATCCTCAAGGTCACCCACCATCACCCTCT
−7.18
169
148
TTTTATAACTTGATCAAGCAGAGAA
−13.20
169





51
154
ACCCTCTGTGATTTTATAACTTGATCAAGC
−7.53
170
190
CTCGTTGATATCCTCAAGGTCACCC
−13.32
170





51
170
AAGGTCACCCACCATCACCCTCTGTGATTT
−7.89
171
189
TCGTTGATATCCTCAAGGTCACCCA
−13.68
171





51
162
CCACCATCACCCTCTGTGATTTTATAACTT
−8.37
172
185
TGATATCCTCAAGGTCACCCACCAT
−13.68
172





51
159
CCATCACCCTCTGTGATTTTATAACTTGAT
−8.43
173
188
CGTTGATATCCTCAAGGTCACCCAC
−13.80
173





51
158
CATCACCCTCTGTGATTTTATAACTTGATC
−8.95
174
149
ATTTTATAACTTGATCAAGCAGAGA
−13.86
174





51
179
GATATCCTCAAGGTCACCCACCATCACCCT
−9.14
175
191
TCTCGTTGATATCCTCAAGGTCACC
−13.88
175





51
152
CCTCTGTGATTTTATAACTTGATCAAGCAG
−9.35
176
160
CACCCTCTGTGATTTTATAACTTGA
−13.94
176





51
149
CTGTGATTTTATAACTTGATCAAGCAGAGA
−9.51
177
187
GTTGATATCCTCAAGGTCACCCACC
−14.07
177





51
180
TGATATCCTCAAGGTCACCCACCATCACCC
−9.82
178
155
TCTGTGATTTTATAACTTGATCAAG
−14.17
178





51
181
TTGATATCCTCAAGGTCACCCACCATCACC
−9.89
179
158
CCCTCTGTGATTTTATAACTTGATC
−14.23
179





51
148
TGTGATTTTATAACTTGATCAAGCAGAGAA
−10.04
180
192
ATCTCGTTGATATCCTCAAGGTCAC
−14.36
180





51
182
GTTGATATCCTCAAGGTCACCCACCATCAC
−10.09
181
186
TTGATATCCTCAAGGTCACCCACCA
−14.42
181





51
183
CGTTGATATCCTCAAGGTCACCCACCATCA
−10.50
182
193
CATCTCGTTGATATCCTCAAGGTCA
−14.55
182





51
184
TCGTTGATATCCTCAAGGTCACCCACCATC
−10.67
183
161
TCACCCTCTGTGATTTTATAACTTG
−14.60
183





51
185
CTCGTTGATATCCTCAAGGTCACCCACCAT
−10.79
184
166
CACCATCACCCTCTGTGATITTATA
−14.64
184





51
186
TCTCGTTGATATCCTCAAGGTCACCCACCA
−11.32
185
194
TCATCTCGTTGATATCCTCAAGGTC
−14.75
185





51
188
CATCTCGTTGATATCCTCAAGGTCACCCAC
−11.44
186
196
GATCATCTCGTTGATATCCTCAAGG
−14.84
186





51
189
TCATCTCGTTGATATCCTCAAGGTCACCCA
−11.81
187
150
GATTTTATAACTTGATCAAGCAGAG
−14.89
187





51
187
ATCTCGTTGATATCCTCAAGGTCACCCACC
−11.82
188
197
TGATCATCTCGTTGATATCCTCAAG
−14.91
188





51
190
ATCATCTCGTTGATATCCTCAAGGTCACCC
−12.10
189
199
GATGATCATCTCGTTGATATCCTCA
−14.93
189





51
191
GATCATCTCGTTGATATCCTCAAGGTCACC
−12.59
190
198
ATGATCATCTCGTTGATATCCTCAA
−14.94
190





51
151
CTCTGTGATTTTATAACTTGATCAAGCAGA
−12.71
191
200
TGATGATCATCTCGTTGATATCCTC
−14.98
191





51
192
TGATCATCTCGTTGATATCCTCAAGGTCAC
−13.26
192
201
TTGATGATCATCTCGTTGATATCCT
−15.00
192





51
150
TCTGTGATTTTATAACTTGATCAAGCAGAG
−13.47
193
195
ATCATCTCGTTGATATCCTCAAGGT
−15.11
193





51
193
ATGATCATCTCGTTGATATCCTCAAGGTCA
−13.85
194
202
CTTGATGATCATCTCGTTGATATCC
−15.16
194





51
194
GATGATCATCTCGTTGATATCCTCAAGGTC
−13.95
195
159
ACCCTCTGTGATTTTATAACTTGAT
−15.57
195





51
195
TGATGATCATCTCGTTGATATCCTCAAGGT
−14.35
196
204
TGCTTGATGATCATCrCGTTGATAT
−15.61
196





51
196
TTGATGATCATCTCGTTGATATCCTCAAGG
−14.51
197
203
GCTTGATGATCATCTCGTTGATATC
−15.64
197





51
198
GCTTGATGATCATCTCGTTGATATCCTCAA
−14.58
198
164
CCATCACCCrCTGTGATTTTATAAC
−15.79
198





51
197
CTTGATGATCATCTCGTTGATATCCTCAAG
−14.70
199
205
CTGCTTGATGATCATCTCGTTGATA
−16.24
199





51
199
TGCTTGATGATCATCTCGTTGATATCCTCA
−14.77
200
165
ACCATCACCCTCTGTGATTTTATAA
−16.41
200





51
200
CTGCTTGATGATCATCTCGTTGATATCCTC
−15.02
201
163
CATCACCCTCTGTGATTTTATAACT
−16.49
201





51
201
TCTGCTTGATGATCATCTCGTTGATATCCT
−15.29
202
206
TCTGCTTGATGATCATCTCGTTGAT
−16.57
202





51
202
TTCTGCTTGATGATCATCTCGTTGATATCC
−15.67
203
207
TTCTGCTTGATGATCATCTCGTTGA
−16.92
203





51
203
CTTCTGCTTGATGATCATCTCGTTGATATC
−16.24
204
208
CTTCTGCTTGATGATCATCTCGTTG
−17.32
204





51




154
CTGTGATTTTATAACTTGATCAAGC
−17.42
205





51




162
ATCACCCTCTGTGATTTTATAACTT
−17.53
206





51




152
GTGATTTTATAACTTGATCAAGCAG
−17.91
207





51




151
TGATTTTATAACTTGATCAAGCAGA
−17.98
208





51




153
TGTGATTTTATAACTTGATCAAGCA
−19.57
209








Claims
  • 1. A modified synthetic antisense oligonucleotide analogue consisting of SEQ ID NO:1 (Ac0), SEQ ID NO:2 (Ac5), SEQ ID NO:3 (Ac26), SEQ ID NO:4 (Ac30), or SEQ ID NO:5 (Ac48).
  • 2. The modified synthetic antisense oligonucleotide analogue of claim 1, wherein the modified synthetic antisense oligonucleotide analogue is selected from a peptide nucleic acid (PNA), a morpholino oligonucleotide, a phosphorothioate oligonucleotide, a phosphorodithioate oligonucleotide, an alkylphosphonate oligonucleotide, an acylphosphonate oligonucleotide, and a phosphoramidite oligonucleotide.
  • 3. A conjugate comprising the modified synthetic antisense oligonucleotide analogue of claim 1 and a carrier, wherein the carrier is conjugated to the antisense oligonucleotide analogue.
  • 4. The conjugate of claim 3, wherein the carrier is operable to transport the modified synthetic antisense oligonucleotide analogue into a target cell.
  • 5. The conjugate of claim 3, wherein the carrier is selected from a peptide, a small molecule chemical, a polymer, a nanoparticle, a lipid, a liposome, and an exosome.
  • 6. The conjugate of claim 3, wherein the carrier is a cell penetrating peptide.
  • 7. The conjugate of claim 3, wherein the carrier is an arginine-rich cell penetrating peptide.
  • 8. A method of treating a muscular disorder in a subject in need thereof, the method comprising administering the modified synthetic antisense oligonucleotide analogue of claim 1 to said subject.
  • 9. The method of claim 8, wherein the muscular disorder is a disorder associated with a genetic mutation in a gene associated with muscle function.
  • 10. The method of claim 8, wherein the muscular disorder is Duchenne muscular dystrophy or Becker muscular dystrophy.
Priority Claims (1)
Number Date Country Kind
1711809 Jul 2017 GB national
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a continuation of U.S. patent application Ser. No. 16/632,827, filed on Jan. 21, 2020, which is a U.S. National Phase Entry of International Application No. PCT/CA2018/050881, filed on Jul. 20, 2018, which claims the benefit of foreign priority to Application No. GB 1711809.2, filed Jul. 21, 2017, each of which is incorporated herein by reference in its entirety.

US Referenced Citations (155)
Number Name Date Kind
4458066 Caruthers et al. Jul 1984 A
4694778 Learn et al. Sep 1987 A
5034506 Summerton et al. Jul 1991 A
5142047 Summerton et al. Aug 1992 A
5166315 Summerton et al. Nov 1992 A
5185444 Summerton et al. Feb 1993 A
5217866 Summerton et al. Jun 1993 A
5334711 Sproat et al. Aug 1994 A
5506337 Summerton et al. Apr 1996 A
5521063 Summerton et al. May 1996 A
5627053 Usman et al. May 1997 A
5698685 Summerton et al. Dec 1997 A
5716824 Beigelman et al. Feb 1998 A
5736557 Hofheinz et al. Apr 1998 A
5889136 Scaringe et al. Mar 1999 A
6008400 Scaringe et al. Dec 1999 A
6111086 Scaringe Aug 2000 A
6194551 Idusogie et al. Feb 2001 B1
6528624 Idusogie et al. Mar 2003 B1
6538124 Idusogie et al. Mar 2003 B1
6821783 Comely et al. Nov 2004 B1
6884869 Senter et al. Apr 2005 B2
7364731 Idusogie et al. Apr 2008 B2
7452987 Giese et al. Nov 2008 B2
7498258 Hiatt Mar 2009 B2
7498298 Doronina et al. Mar 2009 B2
7736647 Boumsell et al. Jun 2010 B2
7791195 Kim Sep 2010 B2
7833992 Vargeese et al. Nov 2010 B2
7850975 Mullis Dec 2010 B2
7893245 Giese et al. Feb 2011 B2
7923547 Mcswiggen et al. Apr 2011 B2
7943762 Weller et al. May 2011 B2
8084582 Dahiyat et al. Dec 2011 B2
8084598 Bentwich Dec 2011 B1
8090542 Khvorova et al. Jan 2012 B2
8202979 Mcswiggen et al. Jun 2012 B2
8248352 Kim et al. Aug 2012 B2
8273866 Mcswiggen et al. Sep 2012 B2
8288352 Doronina et al. Oct 2012 B2
8324370 Giese et al. Dec 2012 B2
8324371 Popplewell et al. Dec 2012 B2
8361979 Aartsma-Rus et al. Jan 2013 B2
8461325 Popplewell et al. Jun 2013 B2
8501703 Bennett et al. Aug 2013 B2
8501930 Rozema et al. Aug 2013 B2
8591910 Mullis Nov 2013 B2
8604184 Mullis et al. Dec 2013 B2
8609105 Senter et al. Dec 2013 B2
8618277 Beigelman et al. Dec 2013 B2
8637483 Wilton Jan 2014 B2
8648185 Mcswigen et al. Feb 2014 B2
8697688 Howard et al. Apr 2014 B2
8835402 Kole et al. Sep 2014 B2
8865883 Sazani Oct 2014 B2
8895722 Iversen et al. Nov 2014 B2
8933215 Giese et al. Jan 2015 B2
8936910 Mitsch et al. Jan 2015 B2
8969526 Baehner et al. Mar 2015 B2
9078911 Lu Jul 2015 B2
9089614 Lin et al. Jul 2015 B2
9096877 Johnson et al. Aug 2015 B2
9139828 Platenburg et al. Sep 2015 B2
9175286 Wilton et al. Nov 2015 B2
9181551 Mcswiggen et al. Nov 2015 B2
9222092 Giese et al. Dec 2015 B2
9228187 Wilton et al. Jan 2016 B2
9243251 Popplewell et al. Jan 2016 B2
9243252 Popplewell et al. Jan 2016 B2
9249416 Wilton et al. Feb 2016 B2
9260471 Cancilla et al. Feb 2016 B2
9416361 Iversen et al. Aug 2016 B2
9434948 Sazani et al. Sep 2016 B2
9441229 Wilton et al. Sep 2016 B2
9447415 Wilton et al. Sep 2016 B2
9447417 Sazani et al. Sep 2016 B2
9481905 Chen et al. Nov 2016 B2
9499818 Van Deutekom Nov 2016 B2
9528109 De Kimpe et al. Dec 2016 B2
9598496 Kurosawa et al. Mar 2017 B2
9605019 Verdine et al. Mar 2017 B2
9657294 Beigelman et al. May 2017 B2
9695211 Wada et al. Jul 2017 B2
9695423 Giese et al. Jul 2017 B2
9732344 Beigelman et al. Aug 2017 B2
9765338 Bennett et al. Sep 2017 B2
9771588 Mcswiggen et al. Sep 2017 B2
9796974 Rajeev et al. Oct 2017 B2
9890379 De Kimpe et al. Feb 2018 B2
9926557 De Kimpe et al. Mar 2018 B2
9982257 Butler et al. May 2018 B2
10000754 Beigelman et al. Jun 2018 B2
10704060 Gersbach et al. Jul 2020 B2
11142767 Yokota et al. Oct 2021 B2
11179472 Levin et al. Nov 2021 B2
11311627 Levin et al. Apr 2022 B1
11400163 Levin et al. Aug 2022 B2
20020142980 Thompson et al. Oct 2002 A1
20080311557 Elsemore et al. Dec 2008 A1
20090092985 Cardozo et al. Apr 2009 A1
20110081362 Elledge et al. Apr 2011 A1
20110263686 Wilton et al. Oct 2011 A1
20110294753 De Kimpe et al. Dec 2011 A1
20110301218 Bozzoni et al. Dec 2011 A1
20120065169 Hanson et al. Mar 2012 A1
20120094299 Ranum et al. Apr 2012 A1
20120172415 Voit et al. Jul 2012 A1
20120270925 Wilton et al. Oct 2012 A1
20120289457 Hanson Nov 2012 A1
20130024919 Wetter et al. Jan 2013 A1
20130028919 Howard et al. Jan 2013 A1
20130172238 Mitsch et al. Jul 2013 A1
20130309256 Lyon et al. Nov 2013 A1
20140127239 Howard May 2014 A1
20140194610 Verdine et al. Jul 2014 A1
20140246970 Park et al. Sep 2014 A1
20140254851 Chen Sep 2014 A1
20140286970 Jeffrey et al. Sep 2014 A1
20140294851 Nguyen Oct 2014 A1
20140296321 Iversen Oct 2014 A1
20140315862 Kaye Oct 2014 A1
20150037360 Smith Feb 2015 A1
20150105539 Miao et al. Apr 2015 A1
20150105540 Miao et al. Apr 2015 A1
20150110791 Zhang et al. Apr 2015 A1
20150211006 Butler et al. Jul 2015 A1
20150361428 Bestwick Dec 2015 A1
20160002637 Sazani et al. Jan 2016 A1
20160053262 Platenburg et al. Feb 2016 A1
20160102135 Escobar-Cabrera Apr 2016 A1
20160193355 Qin et al. Jul 2016 A1
20160201089 Gersbach et al. Jul 2016 A1
20160237426 Hanson Aug 2016 A1
20160298111 Bestwick et al. Oct 2016 A1
20160304864 De Kimpe et al. Oct 2016 A1
20160304877 Swayze et al. Oct 2016 A1
20160367687 Manoharan et al. Dec 2016 A1
20170067048 Wakayama et al. Mar 2017 A1
20170107512 De Kimpe et al. Apr 2017 A1
20170204410 Watanabe et al. Jul 2017 A1
20170204414 Van Deutekom et al. Jul 2017 A1
20170281795 Geall et al. Oct 2017 A1
20170342416 Mcswiggen et al. Nov 2017 A1
20180016574 Bestwick et al. Jan 2018 A1
20180044675 Watanabe et al. Feb 2018 A1
20180112214 De Kimpe et al. Apr 2018 A1
20180127758 Bennett May 2018 A1
20180163209 Bennett et al. Jun 2018 A1
20180305689 Sætrom et al. Oct 2018 A1
20180369400 Levin et al. Dec 2018 A1
20190240346 Sugo et al. Aug 2019 A1
20200282074 Levin et al. Sep 2020 A1
20220096647 Geall et al. Mar 2022 A1
20220235354 Darimont et al. Jul 2022 A1
20220313833 Levin et al. Oct 2022 A1
Foreign Referenced Citations (88)
Number Date Country
2013204438 May 2013 AU
102625840 Aug 2012 CN
103003430 Mar 2013 CN
106459955 Feb 2017 CN
0336675 Oct 1989 EP
0334656 Mar 1994 EP
1579015 Sep 2005 EP
1068241 Oct 2007 EP
2119783 Nov 2009 EP
2049664 Sep 2011 EP
2386636 Nov 2011 EP
2278004 Oct 2012 EP
2602322 Jun 2013 EP
2796425 Oct 2014 EP
2344637 Dec 2014 EP
1423406 Nov 2015 EP
3031920 Jun 2016 EP
2421971 Jul 2016 EP
2287306 Oct 2016 EP
3118311 Jan 2017 EP
3030658 Mar 2017 EP
2287305 Nov 2017 EP
2486141 Jan 2018 EP
2902406 Jan 2018 EP
2595664 Oct 2018 EP
WO-9104753 Apr 1991 WO
WO-9207065 Apr 1992 WO
WO-9315187 Aug 1993 WO
WO-9726270 Jul 1997 WO
WO-9734631 Sep 1997 WO
WO-9813526 Apr 1998 WO
WO-0149698 Jul 2001 WO
WO-03071872 Sep 2003 WO
WO-2004009851 Jan 2004 WO
WO-2004083446 Sep 2004 WO
WO-2006000057 Jan 2006 WO
WO-2006021724 Mar 2006 WO
WO-2006112705 Oct 2006 WO
WO-2008036127 Mar 2008 WO
WO-2009005793 Jan 2009 WO
WO-2009026933 Mar 2009 WO
WO-2009054725 Apr 2009 WO
WO-2009099942 Aug 2009 WO
WO-2009099991 Aug 2009 WO
WO-2009144481 Dec 2009 WO
WO-2009147368 Dec 2009 WO
WO-2010048586 Apr 2010 WO
WO-2010050801 May 2010 WO
WO-2010050802 May 2010 WO
WO-2011057350 May 2011 WO
WO-2011078797 Jun 2011 WO
WO-2011150408 Dec 2011 WO
WO-2012109296 Aug 2012 WO
WO-2012138487 Oct 2012 WO
WO-2013030569 Mar 2013 WO
WO-2013112053 Aug 2013 WO
WO-2013166155 Nov 2013 WO
WO-2014007620 Jan 2014 WO
WO-2014080251 May 2014 WO
WO-2014140277 Sep 2014 WO
WO-2014140317 Sep 2014 WO
WO-2014145090 Sep 2014 WO
WO-2014177042 Nov 2014 WO
WO-2014197748 Dec 2014 WO
WO-2014197854 Dec 2014 WO
WO-2015021457 Feb 2015 WO
WO-2015038426 Mar 2015 WO
WO-2015048792 Apr 2015 WO
WO-2015057699 Apr 2015 WO
WO-2015069587 May 2015 WO
WO-2015075747 May 2015 WO
WO-2015107425 Jul 2015 WO
WO-2016187425 Nov 2016 WO
WO-2016207240 Dec 2016 WO
WO-2017059131 Apr 2017 WO
WO-2017109494 Jun 2017 WO
WO-2017148879 Sep 2017 WO
WO-2017173408 Oct 2017 WO
WO-2017192679 Nov 2017 WO
WO-2017221883 Dec 2017 WO
WO-2018002812 Jan 2018 WO
WO-2018118599 Jun 2018 WO
WO-2018129384 Jul 2018 WO
WO-2019014772 Jan 2019 WO
WO-2019060775 Mar 2019 WO
WO-2020132584 Jun 2020 WO
WO-2020219820 Oct 2020 WO
WO-2021003573 Jan 2021 WO
Non-Patent Literature Citations (176)
Entry
El-Sayed et al. (“Delivery of macromolecules using arginine-rich cell-penetrating peptides: ways to overcome endosomal entrapment.” The AAPS journal 11.1 (2009): 13-22).
Aartsma-Rus et al. Guidelines for antisense oligonucleotide design and insight into splice-modulating mechanisms. Mol Ther 17(3):548-53 (2009).
Aartsma-Rus et al. Targeted exon skipping as a potential gene correction therapy for Duchenne muscular dystrophy. Neuromuscul Disord. 12 Suppl 1:S71-7 (2002).
Abramova et al. Novel oligonucleotide analogues based on morpholino nucleoside subunits-antisense technologies: new chemical possibilities. Indian Journal of Chemistry 48B:1721-1726 (2009).
Agarwal et al. A Pictet-Spengler ligation for protein chemical modification. PNAS 110(1):46-51 (2013).
Albarran et al. Efficient intracellular delivery of a pro-apoptotic peptide with a pH-responsive carrier. React Funct Polym 71:261-265 (2011).
Arechavala-Gomeza et al. Comparative analysis of antisense oligonucleotide sequences for targeted skipping of exon 51 during dystrophin pre-mRNA splicing in human muscle. Hum Gene Ther. 18(9):798-810 (2007).
Axup et al. Synthesis of site-specific antibody-drug conjugates using unnatural amino acids. PNAS 109(40):16101-16106 (2012).
Baumer et al. Antibody-mediated delivery of anti-KRAS-siRNA in vivo overcomes therapy resistance in colon cancer. Clin Can Res 21(6):1383-1394 (2015).
Beduneau et al. Design of targeted lipid nanocapsules by conjugation of whole antibodies and antibody Fab′ fragments. Biomaterials 28(33):4978-4990 (2007).
Beigelman et al. Chemical modification of hammerhead ribozymes. Catalytic activity and nuclease resistance. J Biol Chem 270:25702-25708 (1995).
Bell et al. Epidermal Growth Factor Receptor Mutations and Gene Amplification in Non-Small-Cell Lung Cancer: Molecular Analysis of the IDEAL/INTACT Gefitinib Trials. J Clin Oncol 23(31):8081-8092 (2005).
Bird et al. Single-chain antigen-binding proteins. Science 242:423-442 (1988).
Blaney et al. Traceless solid-phase organic synthesis. Chem. Rev. 102:2607-2024 (2002.
Brown et al. Dystrophic phenotype induced in vitro by antibody blockade of muscle alpha-dystroglycan-laminin interaction. J Cell Sci 112:209-216 (1999).
Bulmus et al. A new pH-responsive and glutathione-reactive, endosomal membrane-disruptive polymeric carrier for intracellular delivery of biomolecular drugs. J Controlled Release 93:105-120 (2003).
Burke et al. siRNA-mediated knockdown of P450 oxidoreductase in rats: a tool to reduce metabolism by CYPs and increase exposure of high clearance compounds. Pharm. Res. 31(12):3445-3460 (2014).
Burlina et al. Chemical engineering of RNase resistant and catalytically active hammerhead ribozymes. Bioorg Med Chem 5:1999-2010 (1997).
Casi et al. Site-specific traceless coupling of potent cytotoxic drugs to recombinant antibodies for pharmacodelivery. J Am Chem Soc 134(13):5887-5892 (2012).
Castaneda et al. Acid-cleavable thiomaleamic acid linker for homogeneous antibody-drug conjugation, Chem. Commun. 49:8187-8189 (2013).
Chen et al. Strand-specific 5′-O-methylation of siRNA duplexes controls guide strand selection and targeting specificity. RNA 14:263-274 (2008).
Clackson et al. Making antibody fragments using phage display libraries. Nature 352(6336):624-628 (1991).
Colberre-Garapin et al. A new dominant hybrid selective marker for higher eukaryotic cells. J Mol Biol 150:1-14 (1981).
Cole et al. The EBV-hybridoma technique and its application to human lung cancer. In, Monoclonal Antibodies and Cancer Therapy (vol. 27, UCLA Symposia on Molecular and Cellular Biology, New Series) (eds. R.A. Reisfeld and S.Sell), New York: Alan R. Liss, Inc. pp. 77-96 (1985).
Collins et al. Duchenne's muscular dystrophy: animal models used to investigate pathogenesis and develop therapeutic strategies. Int J Exp Pathol 84:165-172 (2003).
Crouse et al. Expression and amplification of engineered mouse dihydrofolate reductase minigenes. Mol Cell Biol 3(2):257-266 (1983).
Cuellar et al. Systematic evaluation of antibody-mediated siRNA delivery using an industrial platform of THIOMAB-siRNA conjugates. Nucleic Acids Res 43(2):1189-1203 (2015).
Dawson et al. Modulation of Reactivity in Native Chemical Ligation through the Use of Thiol Additives. J. Am. Chem. Soc. 119:4325-4329 (1997).
Dawson et al. Synthesis of proteins by native chemical ligation. Science 266(5186):776-779 (1994).
De Angelis et al. Chimeric snRNA molecules carrying antisense sequences against the splice junctions of exon 51 of the dystrophin pre-mRNA induce exon skipping and restoration of a dystrophin synthesis in Delta 48-50 DMD cells. PNAS USA 99:9456-9461 (2002).
Debinski et al. Monovalent immunotoxin containing truncated form of Pseudomonas exotoxin as potent antitumor agent. Cancer Research 52(19):5379-5385 (1992).
Deleavey et al. Designing chemically modified oligonucleotides for targeted gene silencing. Chem Biol. 19(8):937-954 (2012).
Dietel et al. A 2015 update on predictive molecular pathology and its role in targeted cancer therapy: a review focussing on clinical relevance. Cancer Gene Ther 22(9):417-430 (2015).
Dimasi et al. Development of a trispecific antibody designed to simultaneously and efficiently target three different antigens on tumor cells. Mol Pharm 12(9):3490-3501 (2015).
Domingo et al. Transferrin receptor as a target for antibody—drug conjugates. Methods in Enzymology 112:238-247 (1985).
Duncan et al. A polymer-Triton X-100 conjugate capable of pH-dependent red blood cell lysis: a model system illustrating the possibility of drug delivery within acidic intracellular compartments. J Drug Target 2:341-347 (1994).
Earnshaw et al. Modified oligoribonucleotides as site-specific probes of RNA structure and function. Biopolymers (Nucleic Acid Sciences) 48:39-55 (1998).
Echigoya et al. In Silico Screening Based on Predictive Algorithms as a Design Tool for Exon Skipping Oligonucleotides in Duchenne Muscular Dystrophy. PLoS One 10(3):e0120058 (2015).
El-Sayed et al. Delivery of macromolecules using arginine-rich cell-penetrating peptides: ways to overcome endosomal entrapment. The AAPS journal 11(1):13-22 (2009).
El-Sayed et al. Rational design of composition and activity correlations for pH-responsive and glutathione-reactive polymer therapeutics. J Control Release 104:417-427 (2005).
Feener et al. Alternative splicing of human dystrophin mRNA generates isoforms at the carboxy terminus. Nature 338:509-511 (Apr. 6, 1989).
Flanary et al. Antigen delivery with poly(propylacrylic acid) conjugation enhanced MHC-1 presentation and T-cell activation. Bioconjugate Chem. 20:241-248 (2009).
Gao et al. Effective Dystrophin Restoration by a Novel Muscle-Homing Peptide—Morpholino Conjugate in Dystrophin-Deficient mdx Mice. Mol Ther. 22(7):1333-1341 (2014).
Gaziova et al. Chemically defined polyethylene glycol siRNA conjugates with enhanced gene silencing effect. Bioorg Med Chem 22(7):2320-2326 (2014).
Goldspiel et al. Human gene therapy. Clin Pharm 12:488-505 (1993).
Griffey et al. 2′-0-aminopropyl ribonucleotides: a zwitterionic modification that enhances the exonuclease resistance and biological activity of antisense oligonucleotides, J. Med. Chem. 39(26):5100-5109 (1997).
Hackeng et al. Protein synthesis by native chemical ligation: Expanded scope by using straightforward methodology. PNAS USA 96:10068-10073 (1999).
Hanes et al. In vitro selection and evolution of functional proteins by using ribosome display. PNAS USA 94:4937-4942 (1997).
Hayash et al. Skin-specific in vivo gene and oligonucleotides transfer into fetal rats as novel model of tissue-specific overexpression of transgene or knock-out by antisense oligonucleotides. Gene Therapy 3:878-885 (1996).
Hejesen et al. A traceless aryl-triazene linker for DNA-directed chemistry. Org Biomol Chem 11(15):2493-2497 (2013).
Henry et al. pH-responsive poly(styrene-alt-maleic anhydride) alkylamide copolymers for intracellular drug delivery. Biomacromolecules 7:2407-2414 (2006).
Hitachi et al. Role of microRNAs in skeletal muscle hypertrophy. Front Physiol 16(4):408 (2014).
Hoffman et al. Restoring Dystrophin Expression in Duchenne Muscular Dystrophy Muscle: Progress in Exon Skipping and Stop Codon Read Through. Am J Pathol 179(1):12-22 (2011).
Hu et al. Site-specific Antibody-polymer Conjugates for siRNA Delivery. J Am Chem Soc 135(37):13885-13891 (2013).
Huang et al. Mechanisms of resistance to EGFR tyrosine kinase inhibitors. Acta Pharma Sinica B 5(5):390-401 (2015).
Hudson et al. Cellular delivery of hammerhead ribozymes conjugated to a transferrin receptor antibody. Int J Pharmaceuticals 182(1):49-58 (1999).
Huse et al. Generation of a large combinatorial library of the immunoglobulin repertoire in phage lambda. Science 246(4935):1275-1281 (1989).
Huston et al. Protein engineering of antibody binding sites: recovery of specific activity in an anti-digoxin single-chain Fv analogue produced in Escherichia coli. PNAS USA 85(16):5879-5883 (1988).
Ishikawa et al. Preparation of monomeric Fab′—horseradish peroxidase conjugate using thiol groups in the hinge and its evaluation in enzyme immunoassay and immunohistochemical staining. Ann N Y Acad Sci. 420:74-89 (1983).
Iversen et al. Optimized siRNA-PEG conjugates for extended blood circulation and reduced urine excretion in mice. Theranostics 3(3):201-209 (2013).
Jancik et al. Clinical relevance of KRAS in human cancers. J Biomed Biotechnol 2010:150960 (13 pgs.) (2010).
Jearawiriyapaisarn et al. Sustained Dystrophin Expression Induced by Peptide-conjugated Morpholino Oligomers in the Muscles of mdx Mice. Mol Ther. 16(9): 1624-1629 (2008).
Jones et al. Poly(2-alkylacrylic acid) polymers deliver molecules to the cytosol by pH-sensitive disruption of endosomal vesicles. Biochem J 372:65-75 (2003).
Karpeisky et al. Highly efficient synthesis of 2′-O-amino nucleosides and their incorporation in hammerhead ribozymes. Tetrahedron Lett 39:1131-1134 (1998).
Kemaladewi et al. Dual exon skipping in myostatin and dystrophin for Duchenne muscular dystrophy. BMC Med Genomics. 4:36 (2011).
Khormaee et al. Endosomolytic anionic polymer for the cytoplasmic delivery of siRNAs in localized in vivo applications. Adv Funct Mater 23:565-574 (2013).
Kim et al. PEG conjugated VEGF siRNA for anti-angiogenic gene therapy. J Cont Rel 116:123-129 (2006).
Kohler et al. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature 256:495-497 (1975).
Koizumi. ENA oligonucleotides as therapeutics. Curr Opin Mol Ther 8(2):144-149 (2006).
Kontermann et al. Bispecific antibodies. Drug Discov Today 20(7):838-847 (2015).
Kozbor et al. The production of monoclonal antibodies from human lymphocytes. Immunology Today 4:72-79 (1983).
Kutmeier et al. Assembly of humanized antibody genes from synthetic oligonucleotides using a single-round PCR. BioTechniques 17:242 (1994).
Lee et al. Antisense PMO cocktails effectively skip dystrophin exons 45-55 in myotubes transdifferentiated from DMD patient fibroblasts. PLoS One 13(5):e0197084 (2018).
Leigh et al. The Human Plasma Proteome: History, Character, and Diagnostic Prospects. Mol Cell Proteomics 1:845-867 (2002).
Levin. Targeting Therapeutic Oligonucleotides. N Engl J Med 376:86-88 (2017).
Loakes. Survey and summary: The applications of universal DNA base analogues. Nucleic Acids Research 29:2437-2447 (2001).
Loh et al. A Survey of siRNA Nanoscal Delivery Patents. 11 Nanotechnology Law & Bus. (pp. 29-37) (2014).
Lowy et al., Isolation of transforming DNA: Cloning the hamster aprt gene. Cell 22:817-823 (1980).
Lyon et al. Self-hydrolyzing maleimides improve the stability and pharmacological properties of antibody-drug conjugates. Nat. Biotechnol. 32(10):1059-1062 (2014).
Marshall et al. Arginine-rich cell-penetrating peptides facilitate delivery of antisense oligomers into murine leukocytes and alter pre-mRNA splicing. J Immunol Method 325(1-2):114-26 (2007).
Martinez et al. Single-stranded antisense siRNAs guide target RNA cleavage in RNAi. Cell 110(5):563-574 (2002).
McEnaney et al. Antibody-recruiting molecules: an emerging paradigm for engaging immune function in treating human disease. ACS Chem Biol. 7(7):1139-1151 (2012).
Mei et al. FBXO32 Targets c-Myc for Proteasomal Degradation and Inhibits c-Myc Activity. J Biol Chem 290:16202-16214 (2015).
Miyata et al. Polymer nanotechnology for nucleic acid delivery. Drug Delivery System 31(1):44-53 (2016) (English Abstract).
Monaco et al. An explanation for the phenotypic differences between patients bearing partial deletions of the DMD locus. Genomics 2(1):90-5 (1988).
Morgan et al. Human gene therapy. Ann Rev Biochem 62:191-217 (1993).
Morrison et al. Chimeric human antibody molecules: mouse antigen-binding domains with human constant region domains. PNAS USA 81(21):6851-6855 (1984).
Mulligan et al. Selection for animal cells that express the Escherichia coli gene coding for xanthine-guanine phosphoribosyltransferase. PNAS USA 78(4):2072-2076 (1981).
Mulligan. The basic science of gene therapy. Science 260(5110):926-932 (1993).
Naisbitt et al. Disposition of amodiaquine and related antimalarial agents in human neutrophils: implications for drug design. J Pharmacol Exp Ther 280:884-893 (1997).
Neuberger et al. Recombinant antibodies possessing novel effector functions. Nature 312(5995):604-608 (1984).
Normand-Sdiqui et al. Oligonucleotide delivery: Uptake of rat transferrin receptor antibody (OX / 26) conjugates into an in vitro immortalised cell line model of the blood, brain barrier. Int J Pharmaceuticals 163:63-71 (1998).
Obika et al. Synthesis of 2′-0,4′-C-methyleneuridine and -cytidine. Novel bicyclic nucleosides having a fixed C3′-endo sugar puckering. Tetrahedron Lett. 38(50):8735-8738 (1997).
O'Hare et al. Transformation of mouse fibroblasts to methotrexate resistance by a recombinant plasmid expressing a prokaryotic dihydrofolate reductase. PNAS USA 78:1527-1531 (1981).
PCT/CA2018/050881 International Search Report and Written Opinion dated Oct. 4, 2018.
PCT/US2018/012672 International Search Report and Written Opinion dated May 24, 2018.
PCT/US2018/012672 Invitation to Pay Additional Fees dated Mar. 20, 2018.
PCT/US2018/052289 International Search Report and Written Opinion dated Jan. 11, 2019.
Perrault et al. Mixed deoxyribo- and ribo-oligonucleotides with catalytic activity. Nature 344:565-568 (1990).
Pieken et al. Kinetic characterization of ribonuclease-resistant 2′-modified hammerhead ribozymes. Science 253:314-317 (1991).
Rozema et al. Dynamic PolyConjugates for targeted in vivo delivery of siRNA to hepatocytes. PNAS USA 104(32):12982-12987 (2007).
Santerre et al. Expression of prokaryotic genes for hygromycin B and G418 resistance as dominant-selection markers in mouse L cells. Gene 30(1-3):147-156 (1984).
Schnyder et al. Targeting of skeletal muscle in vitro using biotinylated immunoliposomes. Biochem J 377(Pt.1):61-67 (2004).
Schwarz et al. Evidence that siRNAs function as guides, not primers, in the Drosophila and human RNAi pathways. Molecular Cell 10:537-548 (2002).
Sekyere et al. Examination of the distribution of the transferrin homologue, melanotransferrin (tumour antigen p97), in mouse and human. Biochimica et Biophysica Acta 1722(2):131-142 (2005).
Singh et al. Recent developments in oligonucleotide conjugation. Chem Soc Rev 39(6):2054-2070 (2010).
Skerra et al. Assembly of a functional Immunoglobulin Fv fragment in Escherichia coli. Science 240(4855):1038-1041 (1988).
Strop et al. Location matters: site of conjugation modulates stability and pharmacokinetics of antibody drug conjugates. Chem Biol 20(2):161-167 (2013).
Sugo et al. Development of antibody-siRNA conjugate targeted to cardiac and skeletal muscles. J Control release 237:1-13 (2016).
Summerton, et al. Morpholino antisense oligomers: design, preparation, and properties. Antisense Nucleic Acid Drug Dev. Jun. 1997;7(3):187-95.
Suñé-Pou et al. Targeting Splicing in the Treatment of Human Disease. Genes 8:E87 (2017).
Suriano et al. Beta-catenin (CTNNB1) gene amplification: a new mechanism of protein overexpression in cancer. Genes Chromosomes Cancer 42(3):238-246 (2005).
Szybalska et al. Genetics of human cell line. IV. DNA-mediated heritable transformation of a biochemical trait. PNAS USA 48:2026-2034 (1962).
Takeda et al. Construction of chimaeric processed immunoglobulin genes containing mouse variable and human constant region sequences. Nature 314(6010):452-454 (1985).
Talasila et al. EGFR Wild-type Amplification and Activation Promote Invasion and Development of Glioblastoma Independent of Angiogenesis. Acta Neuropathol. 125(5):683-698 (2013).
Tolstoshev. Gene Therapy, Concepts, Current Trials and Future Directions. Ann. Rev. Pharmacol. Toxicol. 32:573-596 (1993).
U.S. Appl. No. 16/128,450 Miscellaneous Communication re: Third Party Submission dated Jul. 1, 2019.
U.S. Appl. No. 16/128,450 Office Action dated Apr. 19, 2019.
U.S. Appl. No. 16/128,450 Office Action dated Apr. 30, 2020.
U.S. Appl. No. 16/128,450 Office Action dated Dec. 16, 2020.
U.S. Appl. No. 16/128,450 Office Action dated Sep. 19, 2019.
U.S. Appl. No. 16/129,696 Miscellaneous Communication re: Third Party Submission dated Jul. 3, 2019.
U.S. Appl. No. 16/129,696 Office Action dated Apr. 13, 2020.
U.S. Appl. No. 16/129,696 Office Action dated Apr. 17, 2019.
U.S. Appl. No. 16/129,696 Office Action dated Sep. 19, 2019.
U.S. Appl. No. 16/632,827 Office Action dated Feb. 26, 2021.
Usman et al. Exploiting the chemical synthesis of RNA. Trends Biochem Sci 17:334-339 (1992).
Valtorta et al. KRAS gene amplification in colorectal cancer and impact on response to EGFR-targeted therapy. Int J Cancer 133:1259-1266 (2013).
Van Deutekom et al. Antisense-induced exon skipping restores dystrophin expression in DMD patient derived muscle cells. Hum Mol Genet. 10(15):1547-54 (2001).
Van Deutekom et al. Local dystrophin restoration with antisense oligonucleotide PRO051. N Engl J Med 357(26):2677-2686 (2007).
Van Vliet et al. Assessment of the feasibility of exon 45-55 multiexon skipping for duchenne muscular dystrophy. BMC Medical Genetics 9:105 (2008).
Verma et al. Modified oligonucleotides: synthesis and strategy for users. Annu Rev Biochem 67:99-134 (1998).
Walker et al. Improved cellular delivery of antisense oligonucleotides using transferrin receptor antibody-oligonucleotide conjugates. Pharmaceutical research 12(10):1548-1553 (1995).
Ward et al. Binding activities of a repertoire of single immunoglobulin variable domains secreted from Escherichia coli. Nature 341(6242):544-546 (1989).
Watts et al. Chemically modified siRNA: tools and applications. Drug Discov Today 13(19-20):842-855 (2008).
Weiner. Liposomes as a Drug Delivery System. Drug Develop Ind Pharm 15:1523 (1989).
Wigler et al. Transfer of purified herpes virus thymidine kinase gene to cultured mouse cells. Cell 11:223-232 (1977).
Wigler et al. Transformation of mammalian cells with an amplifiable dominant-acting gene. PNAS USA 77:3567-3570 (1980).
Winkler. Oligonucleotide conjugates for therapeutic applications. Ther Del 4(7):791-809 (2013).
Wong et al. Co-injection of a targeted, reversibly masked endosomolytic polymer dramatically improves the efficacy of cholesterol-conjugated small interfering RNAs in vivo. Nucleic Acid Ther 22(6):380-390 (2012).
Wu et al. Building complex glycopeptides: Development of a cysteine-free native chemical ligation protocol. Angew. Chem. Int. Ed. 45:4116-4125 (2006).
Wu et al. Cell-penetrating peptides as transporters for morpholino oligomers: effects of amino acid composition on intracellular delivery and cytotoxicity. Nucleic Acids Res 35(15):5182-5191 (2007).
Wu et al. Delivery systems for gene therapy. Biotherapy 3:87-95 (1991).
Wu et al. Effective rescue of dystrophin improves cardiac function in dystrophin-deficient mice by a modified morpholino oligomer. PNAS USA 105(39):14814-14819 (2008).
Wu et al. Site-specific chemical modification of recombinant proteins produced in mammalian cells by using the genetically encoded aldehyde tag. PNAS USA 106(9):3000-3005 (2009).
Xu et al. Delivery systems for siRNA drug development in cancer therapy. Asian Journal of Pharmaceutical Sciences 10(1):1-12 (2015).
Yessine et al. Characterization of the membrane-destabilizing properties of different pH-sensitive methacrylic acid copolymers. Biochimica et Biophysica Acta 1613:28-38 (2003).
Yuan et al. Development of siRNA payloads to target KRAS-mutant cancer. Cancer Discov 4(10):1182-1197 (2014).
Zhang et al. A remote arene-binding site on prostate specific membrane antigen revealed by antibody-recruiting small molecules. J Am Chem Soc. 132(36):12711-12716 (2010).
Gooding et al. Oligonucleotide conjugates—Candidates for gene silencing therapeutics. Eur J Pharm Biopharm. 107:321-40 (2016).
U.S. Appl. No. 17/463,473 Office Action dated Dec. 13, 2021.
U.S. Appl. No. 17/463,484 Office Action dated Jan. 4, 2022.
Aartsma-Rus et al., Antisense-induced multiexon skipping for Duchenne muscular dystrophy makes more sense. American Journal of Human Genetics 74(1):83-92 (2004).
Alegre et al. Effect of a Single Amino Acid Mutation on the Activating and Immunosuppressive Properties of a “Humanized” OKT3 Monoclonal Antibody. J Immunol 148:3461-3468 (1992).
Brain and Development 42:117-123 (2010).
Darimont et al. Aug. 2005 Abstract: A novel Antibody-Oligonucleotide Conjugate (AOC) platform enables efficient regulation of muscle targets in mice. Journal Of Cachexia, Sarcopenia And Muscle 8:999-1080 (2017).
Echigoya et al. Quantitative Antisense Screening and Optimization for Exon 51 Skipping in Duchenne Muscular Dystrophy. Mol Ther 25(11):2561-2572 (2017).
Ferreiro et al. Asymptomatic Becker muscular dystrophy in a family with a multiexon deletion. Muscle Nerve 39:239-243 (2009).
Idusogie, et al. Mapping of the C1q binding site on rituxan, a chimeric antibody with a human IgG1 Fc. J Immunol. Apr. 15, 2000;164(8):4178-84.
Kaneko et al. Optimizing Therapeutic Antibody Function: Progress With Fc Domain Engineering. Biodrugs 25(1):1-11 (2011).
Lazar et al. Engineered antibody Fc variants with enhanced effector function. PNAS USA 103(11):4005-10 (2006).
Lee et al. Direct Reprogramming of Human DMD Fibroblasts into Myotubes for In Vitro Evaluation of Antisense-Mediated Exon Skipping and Exons 45-55 Skipping Accompanied by Rescue of Dystrophin Expression. Methods Mol Biol. 1828:141-150 (2018).
Moore et al. Engineered Fc variant antibodies with enhanced ability to recruit complement and mediate effector functions. mAbs 2(2):181-189 (2010).
Nakamura et al. Follow-up of three patients with a large in-frame deletion of exons 45-55 in the Duchenne muscular dystrophy (DMD) gene. J. Clin. Neurosci. 15:757-763 (2008).
Natsume et al. Engineered Antibodies of IgG1/IgG3 Mixed Isotype With Enhanced Cytotoxic Activities. Cancer Res 68(10):3863-72 (2008).
PCT/US2020/029731 International Invitation to Pay Additional Fees dated Aug. 3, 2020.
PCT/US2020/029731 International Search Report and Written Opinion dated Oct. 6, 2020.
Shields et al. High Resolution Mapping of the Binding Site on Human IgG1 for FcγRI, FcγRII, FcγRIII, and FcRn and Design of IgG1 Variants with Improved Binding to the FcγR. J Biol Chem 276(9):6591-6604 (2001).
Stavenhagen et al. Enhancing the potency of therapeutic monoclonal antibodies via Fc optimization. Adv Enzyme Regul. 48:152-64 (2008).
Stavenhagen et al. Fc Optimization of Therapeutic Antibodies Enhances Their Ability to Kill Tumor Cells In vitro and Controls Tumor Expansion In vivo via Low-Affinity Activating Fcγ Receptors. Cancer Res 67(18):8882-91 (2007).
Suzuki et al. Endogenous Multiple Exon Skipping and Back-Splicing at the DMD Mutation Hotspot. Int J Mol Sci. 17(10):1722 (2016).
U.S. Appl. No. 16/129,696 Office Action dated Dec. 14, 2020.
U.S. Appl. No. 16/129,696 Office Action dated May 26, 2021.
U.S. Appl. No. 16/649,572 Miscellaneous Communication re: Third Party Submission dated Mar. 19, 2021.
U.S. Appl. No. 16/649,572 Office Action dated Feb. 22, 2023.
Xia et al. Intravenous siRNA of brain cancer with receptor targeting and avidin-biotin technology. Pharm Res 24(12):2309-16 (2007).
Related Publications (1)
Number Date Country
20210395741 A1 Dec 2021 US
Continuations (1)
Number Date Country
Parent 16632827 US
Child 17466833 US