Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
TREATMENT OF MUSCULAR DYSTROPHIES
Document Type and Number:
WIPO Patent Application WO/2019/110988
Kind Code:
A1
Abstract:
There is described a nucleic acid molecule comprising a nucleotide sequence encoding a functional dystrophin protein. Also described is a vector, a host cell and a pharmaceutical composition comprising the nucleic acid molecule; use of the nucleic acid molecule in therapy, such as in the treatment of a muscular dystrophy; and a method of treating muscular dystrophy, the method comprising administering a therapeutically effective amount of the nucleic acid molecule to a patient suffering from a muscular dystrophy.

Inventors:
DICKSON GEORGE (GB)
POPPLEWELL LINDA (GB)
Application Number:
PCT/GB2018/053521
Publication Date:
June 13, 2019
Filing Date:
December 05, 2018
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ROYAL HOLLOWAY & BEDFORD NEW COLLEGE (GB)
International Classes:
A61K38/17; A61K48/00; C12N15/86
Domestic Patent References:
WO2017191274A22017-11-09
WO2016177911A12016-11-10
Foreign References:
EP2960336A12015-12-30
JPH11318467A1999-11-24
Other References:
FOSTER H ET AL: "Genetic therapeutic approaches for Duchenne muscular dystrophy", HUMAN GENE THERAPY, LIEBERT, US, vol. 23, no. 7, 1 July 2012 (2012-07-01), pages 676 - 687, XP002688790, ISSN: 1043-0342, [retrieved on 20120530], DOI: 10.1089/HUM.2012.099
JARMIN SUSAN ET AL: "New developments in the use of gene therapy to treat Duchenne muscular dystrophy", EXPERT OPINION ON BIOLOGICAL THE, INFORMA HEALTHCARE, ASHLEY, LONDON; GB, vol. 14, no. 2, 1 February 2014 (2014-02-01), pages 209 - 230, XP009187623, ISSN: 1471-2598, DOI: 10.1517/14712598.2014.866087
Attorney, Agent or Firm:
WITHERS & ROGERS LLP (GB)
Download PDF:
Claims:
Claims

1. A nucleic acid molecule comprising a nucleotide sequence encoding a functional dystrophin protein, wherein the nucleotide sequence has at least 77% identity to the sequence of SEQ ID NO. 1.

2. The nucleic acid molecule of claim 1, wherein the nucleotide sequence has at least 80% identity to the sequence of SEQ ID NO. 1.

3. The nucleic acid molecule of claim 1 or 2, wherein the nucleotide sequence has at least 90% identity to the sequence of SEQ ID NO. 1.

4. The nucleic acid molecule of any preceding claim, wherein the nucleotide sequence has at least 95% identity to the sequence of SEQ ID NO. 1.

5. The nucleic acid molecule of any preceding claim, wherein the nucleotide sequence has at least 98% identity to the sequence of SEQ ID NO. 1.

6. The nucleic acid molecule of any preceding claim, wherein the nucleotide sequence has the sequence of SEQ ID NO. 1.

7. The nucleic acid molecule of any preceding claim, wherein the nucleotide sequence encodes a functional human dystrophin protein.

8. The nucleic acid molecule of any preceding claim, wherein the nucleotide sequence encodes a functional dystrophin protein having the amino acid sequence of SEQ ID NO. 2.

9. The nucleic acid molecule of any preceding claim, wherein the nucleotide sequence encoding the dystrophin protein is between 11,025 and 11,085 nucleotides in length.

10. A nucleic acid molecule comprising at least exons 53 to 79 of a nucleotide sequence encoding a functional dystrophin protein, wherein the nucleotide sequence has at least 77% identity to the sequence of SEQ ID NO. 1.

11. A vector comprising the nucleic acid molecule of any preceding claim.

12. The vector of claim 11, when dependent on one of claims 1 to 9, wherein the vector is comprised of three AAV vectors, each containing a portion of the nucleic acid molecule of any one of claims 1 to 9, wherein following transduction of a cell with the three AAV vectors, the nucleic acid molecule of any one of claims 1 to 9 is produced.

13. A vector according to claim 11, wherein the vector is a Puc57-human DMD intron 1 plasmid repair template as depicted in Figure 6 or a lentiviral human DMD intron 1 plasmid repair template as depicted in Figure 7.

14. A host cell comprising the nucleic acid molecule of any one of claims 1 to 10 or the vector of any one of claims 11 to 13.

15. A pharmaceutical composition comprising the nucleic acid molecule of any one of claims 1 to 10 or the vector of any one of claims 11 to 13 and one or more pharmaceutically acceptable excipients.

16. A method of treating muscular dystrophy, the method comprising administering a therapeutically effective amount of the nucleic acid molecule of any one of claims 1 to 10 or the vector of any one of claims 11 to 13 to a patient suffering from a muscular dystrophy.

17. The nucleic acid molecule of any one of claims 1 to 10 or the vector of any one of claims 11 to 13 for use in therapy.

18. The nucleic acid molecule of any one of claims 1 to 10 or the vector of any one of claims 11 to 13 for use in the treatment of a muscular dystrophy.

19. Use of the nucleic acid molecule of any one of claims 1 to 10 or the vector of any one of claims 11 to 13 in the manufacture of a medicament for treating a muscular dystrophy.

20. The method of claim 16, the nucleic acid molecule for use according to claim 18 or the use of claim 19, wherein the muscular dystrophy is selected from Duchenne muscular dystrophy (DMD), Becker muscular dystrophy (BMD) and cardiomyopathy. 21. The method of claim 16, the nucleic acid molecule for use according to claim 16 or the use of claim 19, wherein the muscular dystrophy is Duchenne muscular dystrophy (DMD).

22. A method for delivery of a nucleotide sequence encoding a functional dystrophin protein to a subj ect, which method comprises administering to the said subj ect the nucleic acid molecule of any one of claims 1 to 10 or the vector of any one of claims 11 to 13.

Description:
Treatment of Muscular Dystrophies

Field of the Invention

The present invention relates to a codon optimised dystrophin coding sequence. The invention also relates to vectors comprising the codon optimised dystrophin coding sequence, the use of the codon optimised dystrophin coding sequence in treating muscular dystrophies and methods of treating muscular dystrophies involving the codon optimised dystrophin coding sequence. Muscular dystrophies that can be treated include Duchenne muscular dystrophy (DMD).

Background to the Invention

Duchenne Muscular Dystrophy is an X-linked inherited condition with an incidence of 1 in 3000-5000 boys. The DMD gene encodes dystrophin, a molecular linker between the intracellular actin and extracellular matrix, crucial to correct muscle contractility and integrity. In the absence of this protein, eccentric contractions result in muscle damage as contractile force may not be dissipated correctly, in the short-term muscle can be regenerated by satellite cells. Continued cycles of contraction and regeneration, propagates muscle fibrosis, scarring and ultimately lipid invasion. Initially this manifests within the proximal skeletal muscles of the limbs, reducing the child’s mobility, before eventually progressing to respiratory and cardiac systems, requiring invasive support systems and ultimately causing death between the second and third decade of life.

Currently, the vast majority of gene and therapy approaches are focused upon the restoration of a shortened yet semi-functional dystrophin, producing a clinically lessoned, Becker muscular dystrophy phenotype by addressing specific patient mutations. Some of the current approaches include:

Truncated microdystrophin AAV vectors are in development for DMD gene therapy, but clearly key domains of the full-length protein have been removed and the microdystrophin may be sub-optimal in skeletal muscle, smooth muscle, heart, and CNS locations. Antisense oligonucleotide approaches aiming to mask an mRNA splice site and facilitate the skipping of an exon. The major target is exon 51. However, due to the significant number of exons in which a mutation can occur, the therapeutic applicability of each antisense oligonucleotide is relatively low.

- Multiplex CRISPR-mediated deletion across exons 45-55, thereby removing a major mutation hotspot. This is applicable to approximately 68% of patients.

- NHEJ-mediated genome editing approaches whereby the microinsertions and deletions (InDels) occurring as a byproduct of the NHEJ DNA repair pathway are utilized in 1 in 3 cases to restore the reading frame.

A major limitation of these approaches is only certain patient cohorts can benefit from such therapies.

In view of the limitations in the current approaches, it would be preferable to develop a treatment approach which is more universal in nature so that it could be applied to a larger proportion of patients. Further, it would be advantageous if the whole dystrophin protein could be restored rather than a truncated form of it.

Summary of the Invention

The inventors have designed an optimised cDNA sequence encoding full-length human dystrophin. This provides improved protein expression compared to the native wild-type human gene sequence.

Therefore, in a first aspect, there is provided a nucleic acid molecule comprising a nucleotide sequence encoding a functional dystrophin protein, wherein the nucleotide sequence has at least 77% identity to the sequence of SEQ ID NO. 1.

The optimised cDNA sequence encoding full-length human dystrophin has been shown to produce about a 22-fold increase in protein expression compared to the native sequence. In a second aspect, there is provided a vector for expressing a dystrophin protein, the vector comprising the nucleic acid molecule described above. This means that the vector contains a nucleotide sequence encoding a functional dystrophin protein so that when this sequence is expressed, a functional dystrophin protein is produced by the cell in which the vector is contained.

In a further aspect, there is provided a pharmaceutical composition comprising a nucleic acid molecule or a vector as described above and one or more pharmaceutically acceptable excipients.

In additional aspects, there is provided the use of a nucleic acid molecule or a vector described above in therapy, for example, in the treatment of muscular dystrophies, and a method of treating muscular dystrophies comprising administering a therapeutically effective amount of a nucleic acid molecule or a vector as described above to a patient suffering from a muscular dystrophy.

Detailed Description of the Invention

As indicated above, there is provided a nucleic acid molecule comprising a nucleotide sequence encoding a functional dystrophin protein, wherein the nucleotide sequence has at least 77% identity to the sequence of SEQ ID NO. 1.

The sequence of SEQ ID NO. 1 is a codon optimised nucleotide sequence encoding the full-length human dystrophin protein. The dystrophin nucleotide sequence was optimised by taking into account the following desired parameters: to avoid where applicable cis- acting motifs including internal TATA boxes, chi-sites and ribosomal entry, AT or GC rich sequence stretches, RNA instability motifs, repeat sequences and RNA secondary structures, and cryptic splice donor and acceptor sites in higher eukaryotes. The cDNA sequence alterations have the effect of increasing translational efficiency, mRNA stability, gene transcription and consequently protein synthesis, thus enhancing the level of transgene product per unit of gene transferred. As a result, this nucleotide coding sequence has surprisingly been found to produce about a 22-fold increase in protein expression compared to the native gene sequence. This increase was not expected by the inventors when producing the sequence.

Codon optimisation of sequences has been known for some time. However, the results of this have been mixed. Codon optimising any particular sequence does not necessarily result in an increase in protein expression. Often, expression is the same and sometimes worse compared to the native sequence. Further, where there is an increase in expression, the degree of improvement can also vary significantly, with an increase of less than 10- fold being quite common. As a result, when codon optimising a sequence, there is no expectation that this will result in an increase in protein expression, and there is certainly no expectation that this will result in an increase in expression of more than 5-10 fold. Certainly, you would not expect to achieve an increase of about 22-fold as for the sequence disclosed herein.

Further, there are a number of algorithms which are used in the codon optimisation of sequences and these different algorithms produce different sequences as a result of the optimisation process. These different sequences generally produce different levels of protein expression. However, no one algorithm consistently produces better results than the others. As a result, it is not possible to predict which codon optimisation algorithm will provide the best results for any particular sequence.

With regard to dystrophin, whilst groups have previously tried codon optimising microdystrophin constructs, the results of codon optimisation of full-length dystrophin has not been reported. Therefore, the approach used by the inventors is unconventional compared to other research in this area. Further, in view of the fact that no results have been reported on the codon optimisation of full-length dystrophin, there was no expectation that this approach would be successful, let alone produce the surprising results that have been demonstrated by the inventors.

The DMD gene, encoding the dystrophin protein, is one of the longest human genes known, covering 2.3 megabases (0.08% of the human genome). 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 3686 amino acid residues. Mutations in the DMD gene cause a number of muscular conditions, including Duchenne muscular dystrophy (DMD), Becker muscular dystrophy (BMD) and cardiomyopathy.

Previous attempts to address the mutations in the dystrophin protein have focused on producing shorter, partially functional dystrophin variants which can reduce the severity of the muscular dystrophy. Despite many efforts, delivery of the full-length dystrophin gene has not occurred with any convincing expression levels. Mainly this has been the result of the large transgene, limited delivery processes and the native sequence being sub-optimal for expression. However, it is thought that the optimised dystrophin construct with increased expression may produce high enough levels of dystrophin to ameliorate the clinical phenotypes from a relatively low level of correction. As a result, existing strategies to repair the dystrophin gene/deliver the transgene could be employed enabling a more‘universal’ therapeutic strategy, thereby overcoming the current mutation specific constraints in many therapeutic strategies.

The nucleotide sequence has at least 77% sequence identity to the sequence of SEQ ID NO. 1. In some embodiments, the nucleotide sequence has at least 78% identity to the sequence of SEQ ID NO. 1. In various embodiments, the nucleotide sequence has at least 79% identity to the sequence of SEQ ID NO. 1. In further embodiments, the nucleotide sequence has at least 80% identity to the sequence of SEQ ID NO. 1. In some embodiments, the nucleotide sequence has at least 81% identity to the sequence of SEQ ID NO. 1. In other embodiments, the nucleotide sequence has at least 82% identity to the sequence of SEQ ID NO. 1. In particular embodiments, the nucleotide sequence has at least 83% identity to the sequence of SEQ ID NO. 1. In further embodiments, the nucleotide sequence has at least 84% identity to the sequence of SEQ ID NO. 1. In some embodiments, the nucleotide sequence has at least 85% identity to the sequence of SEQ ID NO. 1. In other embodiments, the nucleotide sequence has at least 86% identity to the sequence of SEQ ID NO. 1. In particular embodiments, the nucleotide sequence has at least 87% identity to the sequence of SEQ ID NO. 1. In further embodiments, the nucleotide sequence has at least 88% identity to the sequence of SEQ ID NO. 1. In some embodiments, the nucleotide sequence has at least 89% identity to the sequence of SEQ ID NO. 1. In further embodiments, the nucleotide sequence has at least 90% identity to the sequence of SEQ ID NO. 1. In some embodiments, the nucleotide sequence has at least 91% identity to the sequence of SEQ ID NO. 1. In other embodiments, the nucleotide sequence has at least 92% identity to the sequence of SEQ ID NO. 1. In particular embodiments, the nucleotide sequence has at least 93% identity to the sequence of SEQ ID NO. 1. In further embodiments, the nucleotide sequence has at least 94% identity to the sequence of SEQ ID NO. 1. In some embodiments, the nucleotide sequence has at least 95% identity to the sequence of SEQ ID NO. 1. In other embodiments, the nucleotide sequence has at least 96% identity to the sequence of SEQ ID NO. 1. In particular embodiments, the nucleotide sequence has at least 97% identity to the sequence of SEQ ID NO. 1. In further embodiments, the nucleotide sequence has at least 98% identity to the sequence of SEQ ID NO. 1. In some embodiments, the nucleotide sequence has at least 99% identity to the sequence of SEQ ID NO. 1. In other embodiments, the nucleotide sequence has at least 99.5% identity to the sequence of SEQ ID NO. 1. In particular embodiment, the nucleotide sequence has the sequence of SEQ ID NO. 1. In certain embodiments, the nucleotide sequence has the sequence of SEQ ID NO. 3.

The nucleotide sequence encodes a functional dystrophin protein. A functional dystrophin protein is one which can form part of a protein complex known as the costamere or the dystrophin-associated protein complex, which acts as a transmembrane platform that anchors the extracellular matrix (ECM) to the intracellular cytoskeleton. The dystrophin protein has four main functional domains: an actin-binding amino- terminal domain; a central rod domain; a cysteine-rich domain and a carboxyl-terminus. These bind to a number of structures/proteins to allow dystrophin to correctly carry out its function in the dystrophin-associated protein complex. For example, dystrophin binds to actin filaments, microtubules and a number of proteins which help to anchor the dystrophin at the sarcolemma. A skilled person would readily be able to identify whether a dystrophin protein is functional. For example, this could be done by using an assay involving immunohistochemical staining of treated muscle sections, staining for dystrophin, and looking for restoration of the dystrophin-associated protein complex at the sarcolemma through staining (see Counsell JR et al., Sci Rep. 7:44775 (2017); Koo T et al., Hum Gene Ther. 25(2):98-l08 (2014); Koo T et al., Hum Gene Ther. 22(11): 1379- 88 (2011); Le Guiner C et al., Nat Commun. 8: 16105 (2017); and Meng J et al., Sci Rep. 6: 19750 (2016)).

In preferred embodiments, the nucleotide sequence encodes a functional human dystrophin protein. The sequences of appropriate dystrophin proteins are well known to those skilled in the art. For example, a number of dystrophin isoforms are known. Therefore, the nucleotide sequence may encode a dystrophin protein selected from isoform 1 (identifier: Pl 1532-2), isoform 2 (identifier: Pl 1532-3), isoform 3 (identifier: Pl 1532-4) and isoform 4 (identifier: Pl 1532-1). Preferably, the nucleotide sequence encodes isoform 4 (identifier: Pl 1532-1) of the human dystrophin protein. The amino acid sequence of the native human dystrophin protein can be found as SEQ ID NO. 2. Therefore, in some embodiments, the nucleotide sequence encodes a dystrophin protein having the amino acid sequence of SEQ ID NO. 2. Other dystrophin proteins that may be encoded by the nucleotide sequence include natural variants with mutations that do not affect the function of the dystrophin protein.

The human dystrophin protein is 3686 amino acids in length. Therefore, in some embodiments, the nucleotide sequence encodes a dystrophin protein having at least 3675 amino acids. In other embodiments, the nucleotide sequence encodes a dystrophin protein having at least 3680 amino acids. In further embodiments, the nucleotide sequence encodes a dystrophin protein having at least 3682 amino acids. In various embodiments, the nucleotide sequence encodes a dystrophin protein having at least 3683 amino acids. In certain embodiments, the nucleotide sequence encodes a dystrophin protein having at least 3684 amino acids. In particular embodiments, the nucleotide sequence encodes a dystrophin protein having at least 3685 amino acids.

In some embodiments, the nucleotide sequence encodes a dystrophin protein having at most 3697 amino acids. In other embodiments, the nucleotide sequence encodes a dystrophin protein having at most 3692 amino acids. In further embodiments, the nucleotide sequence encodes a dystrophin protein having at most 3690 amino acids. In various embodiments, the nucleotide sequence encodes a dystrophin protein having at most 3689 amino acids. In certain embodiments, the nucleotide sequence encodes a dystrophin protein having at most 3688 amino acids. In particular embodiments, the nucleotide sequence encodes a dystrophin protein having at most 3687 amino acids.

In particular embodiments, the nucleotide sequence encodes a dystrophin protein having about 3686 amino acids. In these embodiments, the nucleotide sequence can be said to encode a 'full-length' human dystrophin protein.

The nucleotide sequence encoding a dystrophin protein is preferably between 11,025 and 11,085 nucleotides in length. In some embodiments, the nucleotide sequence encoding a functional dystrophin protein is between 11,040 and 11,070 nucleotides in length. In other embodiments, the nucleotide sequence encoding a functional dystrophin protein is between 11,052 and 11,064 nucleotides in length. In particular embodiments, the nucleotide sequence encoding a functional dystrophin protein is about 11,058 nucleotides in length.

The nucleic acid molecule may comprise additional nucleotide sequences which encode further peptides/proteins or perform some further function, for example, aiding in the expression of the nucleotide sequence encoding the dystrophin protein. For example, the nucleic acid molecule may comprise a nucleotide sequence which encodes a green fluorescent protein (GFP) such that when the nucleic acid molecule is expressed, a dystrophin protein is produced which is tethered to the GFP.

Preferably, the nucleic acids described above are isolated.

It would be well with the capabilities of a skilled person to produce the nucleic acid molecules described above. This could be done, for example, using chemical synthesis of a given sequence with appropriate enzymatic ligation, where necessary.

The nucleic acid molecule can be any type of nucleic acid composed of nucleotides. The nucleic acid should be able to be expressed so that a protein is produced. Preferably, the nucleic acid is DNA or RNA. In some embodiments, the nucleic acid molecule is DNA, such as cDNA.

In a second aspect, there is provided a vector for expressing a dystrophin protein. The vector comprises the nucleic acid molecule described above. This means that the vector contains a nucleotide sequence encoding a functional dystrophin protein so that when this sequence is expressed, a functional dystrophin protein is produced by the cell in which the vector is contained.

In a therapeutic setting, the vector can take on a number of different forms depending on how the nucleic acid molecule is delivered to the cells of a patient suffering from a muscular dystrophy associated with a defective dystrophin protein. Various approaches are described in Chamberlain JR and Chamberlain JS ("Progress toward Gene Therapy for Duchenne Muscular Dystrophy", Mol Ther. 25(5): 1125-1131 (2017)). For example, the nucleic acid molecule may be delivered by a transposon system (e.g. see Ley D et al, Stem Cell Res. 13(3 Pt A):390-403 (2014)), an artificial chromosome (e.g. see Tedesco FS, Chromosome Res. 23(1): 135-41 (2015)), exploitation of the homology directed repair (HDR) DNA pathway (e.g. see Popplewell et al, Hum Gene Ther. (7):692-70l (2013)), a lentiviral vector (e.g. see Counsell JR et al, Sci Rep. 7(l):79 (2017)), or AAV vectors using a triple-transplicing approach (e.g. see Koo T et al., Hum Gene Ther. 25(2):98-l08 (2014)).

In the triple-transplicing approach, the dystrophin cDNA is split across three adeno- associated viral vectors, which associate together in a directional manner when co expressed due to corresponding splice acceptors and donors within each cassette. Alternatively, the sequence is split in three, each with a linked group 1 intron ribozymes, and each sequence expressed from an AAV vector leading to mRNAs which are spliced together in the correct order and orientation. Therefore, there is provided three AAV vectors, each containing a portion of the nucleic acid molecule described above, wherein following transduction of a cell with the three AAV vectors, the nucleic acid molecule is produced. In this context, the nucleic acid molecule may be produced as DNA or RNA (e.g. mRNA). The exploitation of the homology directed repair (HDR) DNA pathway is a genetic engineering approach which involves production of a targeted DNA lesion in the DMD intron mediated by a specialised endonuclease and the exploitation of the HDR DNA pathway to integrate full-length dystrophin encoded by an exogenous cDNA donor.

In some approaches, stem cells may be isolated from a patient suffering from a muscular dystrophy associated with a defective dystrophin protein and these stem cells modified to incorporate the nucleic acid molecule described above, before the stem cells are reintroduced into the patient (e.g. see Zhu P et al, Mol Ther Nucleic Acids. 7:31-41 (2017) and Meng J et al., Sci Rep. 6: 19750 (2016)). Alternatively, induced pluripotent stem cells may be used (e.g. see Gee P et al, Stem Cells Int. 2017:8765154 (2017)).

In certain embodiments in which the nucleic acid molecule is expressed by the vector (rather than being incorporated into the genetic material through a genetic engineering technique), the vector further comprises a promoter. The promoter causes expression of the nucleotide sequence encoding a functional dystrophin protein. Any appropriate promoter may be used, such as cytomegalovirus (CMV), Spc5. l2, muscle creatine kinase (MCK), dMCK, tMCK, desmin (Des), alpha- myosin heavy chain (a-MHC), myosin light chain 2 (MLC-2), cardiac troponin C (cTnC) and slow isoform of troponin I (TnIS). Preferably, the promoter is a muscle specific promoter such as Spc5. l2, muscle creatine kinase (MCK), dMCK, tMCK, desmin (Des), alpha-myosin heavy chain (a-MHC), myosin light chain 2 (MLC-2), cardiac troponin C (cTnC) and slow isoform of troponin I (TnIS).

In vectors which are designed to integrate the dystrophin coding sequence into the genetic material of a cell rather than simply express the dystrophin coding sequence, for example by exploiting the homology directed repair (HDR) DNA pathway, the vector may not contain the entire dystrophin coding sequence (although in some embodiments, it may). Instead, it may contain a fragment of the dystrophin coding sequence which is then integrated into the defective sequence to bring about correction of the dystrophin sequence. Importantly, the fragment of the dystrophin coding sequence must be long enough so that it replaces the part of the defective dystrophin sequence containing the disease causing mutation. In this approach, the fragment of the dystrophin coding sequence may be integrated after the first few exons of the naturally occurring sequence or even later in the dystrophin sequence. For example, the vector may contain exons 2- 79, or shorter variants such as 45-79 or even 53-79.

Therefore, in some embodiments, there is provided a nucleic acid molecule comprising at least exons 53 to 79 of a nucleotide sequence encoding a functional dystrophin protein, wherein the nucleotide sequence has at least 77% identity to the sequence of SEQ ID NO. 1. Further, there may be provided a nucleic acid molecule comprising at least exons 45 to 79 of a nucleotide sequence encoding a functional dystrophin protein, wherein the nucleotide sequence has at least 77% identity to the sequence of SEQ ID NO. 1. In addition, there may be provided a nucleic acid molecule comprising at least exons 10 to 79 of a nucleotide sequence encoding a functional dystrophin protein, wherein the nucleotide sequence has at least 77% identity to the sequence of SEQ ID NO. 1. In various embodiments, the nucleic acid molecule comprises at least exons 9 to 79 of the nucleotide sequence encoding a functional dystrophin protein. In other embodiments, the nucleic acid molecule comprises at least exons 8 to 79 of the nucleotide sequence encoding a functional dystrophin protein. In certain embodiments, the nucleic acid molecule comprises at least exons 7 to 79 of the nucleotide sequence encoding a functional dystrophin protein. In some embodiments, the nucleic acid molecule comprises at least exons 6 to 79 of the nucleotide sequence encoding a functional dystrophin protein. In various embodiments, the nucleic acid molecule comprises at least exons 5 to 79 of the nucleotide sequence encoding a functional dystrophin protein. In other embodiments, the nucleic acid molecule comprises at least exons 4 to 79 of the nucleotide sequence encoding a functional dystrophin protein. In certain embodiments, the nucleic acid molecule comprises at least exons 3 to 79 of the nucleotide sequence encoding a functional dystrophin protein. In some embodiments, the nucleic acid molecule comprises at least exons 2 to 79 of the nucleotide sequence encoding a functional dystrophin protein. The nucleotide sequence described in this paragraph may have at least 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or 100% identity to the sequence of SEQ ID NO. 1 as described with respect to the full sequence above. This means that the nucleic acid molecule comprises at least exons 10 to 79 of a nucleotide sequence encoding a functional dystrophin protein, wherein the nucleotide sequence has at least 78%, 79%, 80%, etc. identity to the sequence of SEQ ID NO. 1. This also applies to the rest of the statements above, e.g. the nucleic acid molecule may comprise at least exons 53 to 79, at least exons 45 to 79, at least exons 9 to 79, at least exons 8 to 79, at least exons 7 to 79, etc. of the nucleotide sequence encoding a functional dystrophin protein. Also provided is a vector comprising the nucleic acid molecule described above.

In some embodiments, the vector described above is a Puc57-human DMD intron 1 plasmid repair template as depicted in Figure 6. In various embodiments, the vector described above is a lentiviral human DMD intron 1 plasmid repair template as depicted in Figure 7.

The invention also provides a host cell comprising any one of the nucleic acid molecules or vectors described above. Preferably, the vector is capable of expressing the dystrophin nucleotide sequence in the host. The host may be any suitable host.

As used herein, the term "host" refers to cells which harbour a nucleic acid molecule or a vector, as well as cells that are suitable for use in expressing a recombinant gene or protein. It is not intended that the present invention be limited to any particular type of cell. Indeed, it is contemplated that any suitable cell will find use in the present invention as a host. A host cell may be in the form of a single cell or a population of similar or different cells, for example in the form of a culture (such as a liquid culture or a culture on a solid substrate). In some embodiments, the host cell may be a stem cell. This may be an autologous human stem cell or an induced pluripotent stem cell.

The host cell may permit the expression of the nucleic acid molecule. Thus, the host cell may be, for example, a bacterial, a yeast, an insect or a mammalian cell. Suitable mammalian cells may be from a human, a non-human primate, a rodent, especially a mouse, or may be canine, feline, ovine or porcine. Where the mammalian cell is a human cell, such as a stem cell, it is preferably isolated. In one aspect, the invention provides a pharmaceutical composition comprising a nucleic acid molecule or a vector of the invention and one or more pharmaceutically acceptable excipients. The one or more excipients include carriers, diluents and/or other medicinal agents, pharmaceutical agents or adjuvants, etc.

The invention also provides a method of treating a muscular dystrophy, the method comprising administering a therapeutically effective amount of a nucleic acid molecule or a vector as described above to a patient suffering from the muscular dystrophy. Preferably, the patient is human.

The muscular dystrophy is associated with a mutation in the DMD gene. The muscular dystrophy may be selected from Duchenne muscular dystrophy (DMD), Becker muscular dystrophy (BMD) and cardiomyopathy.

When the muscular dystrophy is“treated” in the above method, this means that one or more symptoms of the muscular dystrophy are ameliorated. It does not mean that the symptoms of the muscular dystrophy are completely remedied so that they are no longer present in the patient, although in some methods, this may be the case. The method of treating results in one or more of the symptoms of the muscular dystrophy being less severe than before treatment.

A "therapeutically effective amount" refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result, such as raising the level of (functional) dystrophin in a subject (so as to lead to dystrophin production at a level sufficient to ameliorate the symptoms of the muscular dystrophy).

Further, the invention provides the nucleic acid molecule encoding a functional dystrophin protein as described above, or a vector as described above for use in therapy, for example, in the treatment of a muscular dystrophy. In addition, the invention provides the use of the nucleic acid molecule encoding a functional dystrophin protein as described above or a vector as described above in the manufacture of a medicament for treating a muscular dystrophy.

The invention also provides a method for delivery of a nucleotide sequence encoding a functional dystrophin protein to a subject, which method comprises administering to the said subject a nucleic acid molecule encoding a functional dystrophin protein as described above or a vector as described above.

In the description above, the term“identity” is used to refer to the similarity of two sequences. For the purpose of this invention, it is defined here that in order to determine the percent identity of two nucleotide sequences, the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in the sequence of a first nucleic acid for optimal alignment with a second nucleic acid sequence). The nucleotide residues at nucleotide positions are then compared. When a position in the first sequence is occupied by the same nucleotide residue as the corresponding position in the second sequence, then the molecules are identical at that position. The percent identity between the two sequences is a function of the number of identical positions shared by the sequences (i.e., % identity = number of identical positions/total number of positions (i.e. overlapping positions) x 100). Preferably, the two sequences are the same length. A sequence comparison is typically carried out over the entire length of the two sequences being compared.

The skilled person will be aware of the fact that several different computer programs are available to determine the identity between two sequences. For instance, a comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm. In a preferred embodiment, the percent identity between two nucleic acid sequences is determined using the sequence alignment software Emboss Stretcher (www.ebi.ac.uk/Tools/psa/emboss_stretcher) using the following pairwise alignment options: Matrix: DNAfull; Gap open: 16; Gap extend: 4; and Output format: Pair. An alternative option is to use Clone Manager 9 (Sci-Ed software - www.scied.com) using global DNA alignment; parameters: both strands; scoring matrix: linear (mismatch 2, OpenGap 4, ExtGap 1).

Alternatively, the percent identity between two nucleic acid sequences can be determined using the Needleman and Wunsch (1970) algorithm which has been incorporated into the GAP program in the Accelrys GCG software package (available at http://www.accelrys.com/products/gcg/), using either a Blosum 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of 1, 2, 3, 4, 5, or 6. A further method to assess the percent identity between two nucleic acid sequences can be to use the BLAST sequence comparison tool available on the National Center for Biotechnology Information (NCBI) website (www.blast.ncbi.nlm.nih.gov), for example using BLASTn for nucleotide sequences using the default parameters.

All patent and literature references cited in the present specification are hereby incorporated by reference in their entirety.

Brief Description of the Drawings

The invention will now be described in detail by way of example only with reference to the figures which are as follows:

Figure 1 : Visual demonstration that sequence optimisation of cDNA improves recombinant protein expression as demonstrated by using constructs encoding native and optimised, full-length dystrophin tethered to eGFP. Hek293T cells transfected with native and sequence optimised full-length dystrophin-eGFP constructs, Nat-hDys and Opt-HuDys (SO) respectively. Transient transfections were undertaken at 4pg with a 5: 1 viafect reagentDNA ratio. Subsequent GFP expression was imaged at sequential time points of 24, 48 and 72 hours with the Zeiss microscope at lOx magnification and 30ms BF and 800ms GFP channel exposures. A noticeable accumulation of green fluorescence can be seen in sequential time points for both constructs; in addition a difference in fluorescence intensity is seen between native and optimised dystrophin encoding constructs. Figure 2: Optimisation of cDNA sequence increases recombinant dystrophin expression. Plasmids expressing native (Nat-hDys) and sequence-optimised dystrophin (Opt-HuDys- SO) cDNAs from the CMV promoter were transfected into HEK293 cells (n=5). After 72h cultures were harvested and processed for Western blotting (A) 50pg total protein lysate was analysed with antibodies to dystrophin (6C5) and alpha-tubulin. Dystrophin bands were then quantified relative to the alpha-tubulin loading controls and mean intensity ratios plotted (B: mean+SEM: ***8 p<0.00l (unpaired t-test). Sequence optimisation produced a 57-fold increase in expression.

Figure 3 : Optimisation of cDNA sequence increases recombinant dystrophin expression. Plasmids expressing native (Nat-hDys) and sequence-optimised dystrophin (Opt-HuDys- SO) cDNAs from the CMV promoter were transfected into HEK293 cells (n=5). After 72h cultures were harvested and processed for Western blotting (A) 50pg total protein lysate was analysed with antibodies to dystrophin (ManExlOl lc) and alpha-tubulin. Dystrophin bands were then quantified relative to the alpha-tubulin loading controls and mean intensity ratios plotted (B: mean+SEM: *** p<0.00l (unpaired t-test). Sequence optimisation produced a 22-fold increase in expression.

Figure 4: Optimisation of cDNA sequence increases recombinant dystrophin expression. Plasmids expressing native (Nat-hDys) and sequence-optimised dystrophin (Opt-HuDys- SO) cDNAs from the Spc5 l2 promoter were transfected into HEK293 cells (n=5). After 72h cultures were harvested and processed for Western blotting (A) 50pg total protein lysate was analysed with antibodies to dystrophin (6C5) and alpha-tubulin. Dystrophin bands were then quantified relative to the alpha-tubulin loading controls and mean intensity ratios plotted (B: mean+SEM: ***8 p<0.00l (unpaired t-test). Sequence optimisation produced a 15-fold increase in expression.

Figure 5: Optimisation of cDNA sequence increases recombinant dystrophin expression. Plasmids expressing native (Nat-hDys) and sequence-optimised dystrophin (HuDys-CO) cDNAs from the Spc5 l2 promoter were transfected into HEK293 cells (n=5). After 72h cultures were harvested and processed for Western blotting (A) 50pg total protein lysate was analysed with antibodies to dystrophin (ManExlOl lc) and alpha-tubulin. Dystrophin bands were then quantified relative to the alpha-tubulin loading controls, and mean intensity ratios plotted (B: mean+SEM: **** p<0.000l (unpaired t-test). Sequence optimisation produced a 17-fold increase in expression.

Figure 6: A linear schematic of the Puc57-human DMD intron 1 plasmid repair template. A linear schematic of Puc57-hINTl-RT empty vector. The Fsel and Notl sites flanking the Cyan fluorescent marker, serve as the directional cloning sites for the optimised full- length dystrophin. Upstream of this cloning site is a lkb left arm of homology, synthetic beta globin splice acceptor and Exons 2-6 of optimised DMD cDNA. Downstream of this cloning sequence is the WPRE and poly A to enhance expression. Followed thereafter by a floxed zeocin cassette, to facilitated selection and a lkb right arm of homology. Where possible these constituents are flanked by unique restriction sites that are annotated, ensuring that the repair template is amenable to changing constituents if required.

Figure 7: A schematic of the Lentiviral human DMD intron 1 plasmid repair template. A lentiviral vector encoding the human DMD Intron 1 repair template; notably, the constituents of the human DMD intron 1 plasmid run in reverse orientation to the transcription of the lentiviral plasmid; this is to prevent aberrant splicing from occurring that may be detrimental to viral mRNA transcription and subsequently viral production. This includes the F sel and Notl cloning sites downstream of optimised DMD cDNA exons 2-6, CFP protein and Woodchuck hepatitis virus regulatory element (WPRE). In addition the upstream and downstream lkb arms of homology isogenic to the genomic sequence adjacent to the guide sites in intron 1 and the floxed zeocin cassette are also present.

Examples

This work was carried out to assess the expression of recombinant dystrophin protein resulting from both native and sequence optimised cDNA encoding full-length dystrophin.

Materials & Methods

Materials and Methods for characterisation of expression differences of full-length native and sequence optimised dystrophin: Viafect Transient Transfection

Materials

• Viafect Transient Transfection Reagent (Promega)

· Serum Free Dulbecco’s Modified Eagle’s Medium (DMEM) (Gibco)

• Hek293T Cells (ATCC)

• Six Well Plate (Coming)

• Sterile Eppendorfs (Coming) Method

Hek293T cells were seeded at a density of 5 x 10 5 at Day 0 to attain 70-80% confluency 24 hours post seeding. The growth media was carefully changed 1 hour prior to transient transfection. During this time, master mix was produced comprising: 20pl of viafect reagent (Promega) and 4pg of native or sequence optimised dystrophin made to a 200m1 volume per well using serum free DMEM (Gibco). Importantly, the transfection reagent (pl):DNA mass (pg) was maintained at a 5: 1 ratio, when the mastermix was produced it was adjusted to include an extra half a well to account for pipetting error and all transient transfections were undertaken in a six well plate (Corning). In the production of a mastermix, a calculated volume of serum free DMEM at room temperature was pipetted into a sterile eppendorf. Then 4pg of DNA was added and the DNA-DMEM suspension agitated. This was incubated at room temperature for 5 minutes as per the manufacture’s protocol. Then a defined volume of Viafect transfection reagent was added drop-wise with continual agitation of the suspension and incubated at room temperature for 15 minutes. Post incubation, the transient transfection mixture was added to the well in a dropwise circular motion to ensure maximum cell coverage. Notably, this was also performed alongside a‘Mock’ condition in which cells were incubated in the presence of Viafect and DMEM in the absence of DNA, as a control. Due to the non-toxic nature of Viafect transfection reagent a media change was not necessitated post transfection. Cells were incubated for 72 hours prior to harvesting for total protein lysate. Protein Extraction and Quantification:

Materials

• Sterile PBS: 1 PBS Tablet (Gibco) dissolved in 500ml of ddH20. This was either autoclaved or filtered with a 0.22mM filter, with Class II Lamina flow hood.

• PAPBNI Buffer: NaCl 0.15M, HEPES 0.05M, NP-40.1%, Sodium Deoxycholate (SOC) 0.5%, SDS 0.10%, EDTA 0.01M, Protease Inhibitor tablet 1 in 50ml (Roche). This was aliquoted into 5mls, and stored at -20.

• Cells Scrappers (Invitrogen)

• Eppendorfs

• Benchtop Microcentrifuge

• DC Assay Protein kit: Reagent A, S and B (BioRad).

• 2pg BSA Standard (Invitrogen)

• 96 Well plate (Corning)

• 96 Well plate reader (Genbank)

Protein Extraction:

Post 72 hour incubation, growth media was aspirated, cells were washed with 500m1 ice cold PBS and IOOmI PABPN1 RIPA Buffer added. The cells were then scrapped down to the bottom of the well, with the plate held at a 45° angle, before being incubated at 4°C on ice for 5 minutes. The samples were then transferred to pre-chilled and labelled eppendorfs, prior to being vortexed every 30 seconds for a further 15 minutes. The resultant protein lysates were then centrifuged at 13,000 rpm for 15 minutes, in a benchtop microcentrifuge to allow cell debris to pellet. The supernatant was then transferred to a fresh pre-chilled and labelled 0.5ml screw top tube, and stored at -20°C.

Protein Quantification:

A defined dilution series of BSA in the protein extraction PABPN1 Buffer is prepared providing concentrations ranging from 0- 2pg of BSA respectively. As shown below: These protein standards were loaded at a volume of 0.5m1, alongside extracted protein samples at a volume of 5m1 in a 96 well plate. All samples were loaded in triplicate to assert the accuracy/ ensure reliability of the resulting absorption readings obtained.

In the fume hood: Reagent A +S (lml of A to 20m1 of S) was added at volume of 25m1 to each of the samples from a low to high concentration wherever possible; this being a precautionary measure to prevent contamination. Subsequently, 200m1 of Reagent B is applied and the resultant mixture is agitated and incubated for 15 minutes at room temperature. A colorimetric analysis is undertaken at 750nm using the (Gen) 96 well plate reader. The absorption readings were then, used to calculate an average protein concentration of the three samples from the standard curve.

Western Blotting

Materials

• NuPage 10X Reducing Agent (ThermoFischer)

• NuPage 4x Loading Dye Sample (ThermoFischer)

• NuPage 3-8% Tris Acetate precast gradient gels (ThermoFischer)

• NuPage Antioxidant (ThermoFischer)

· Prestained HiMark Ladder (Life technologies)

• NuPage 3-8% Tris Acetate Running Buffer (ThermoFischer)

• NuPage 20X Transfer Buffer (ThermoFischer)

• Absolute Methanol (VWR)

• I-Cell Blot Tank (Thermo Fischer)

· 0.45mM Nitrocellulose membrane (GE Healthcare)

Ponceau Stain (ThermoFischer)

Filter paper

Marvel Milk Powder

Tween 20 Detergent (Sigma)

ECL solution 1 and 2 (Promega)

Amersham Hyperfilm l8cm x 24cm (GE Heatlhcare) • Odyssey SA (Licor)

Antibodies

Primary Antibodies

Secondary Antibodies

Sample Preparation

Samples of total protein lysate were produced in a 4x master mix, this was to allow repetitions with antibodies if required. Typically a 4x 40ul master stock would contain 200pg total protein. Samples were then prepared in a l.5ml screw top tube with: 4pl Reducing Agent, 10m1 of Loading Sample Dye and the remaining volume is supplemented with ddH20. The samples were prepared alongside a positive control, either dystrophin extracted from muscle or from a previous positive transfection. Then denatured by heating to 70°C for 10 minutes.

Gel Preparation and Electrophoresis

A 3-8% Tris-Acetate precast gradient gel (ThermoFischer) was used to resolve the full length dystrophin protein. In preparation of the gel, the comb was removed and wells washed with ddH20. In addition, a white adhesive strip sealing the foot of the gel was removed. Gels were then placed in the I-Cell Blot tanks vertically. Then a 10m1 aliquot of the 4x master stock of each protein sample was loaded alongside, a pre-stained Hi-Mark ladder (Life Technologies). The surrounding tank was filled approximately lcm from the top, with IX 3-8% Tris- Acetate buffer and 500m1 of antioxidant was applied immediately prior to the initiation of electrophoresis. The gel was run for approximately 1 hour and 15 minutes at 150V, in accordance with the Nupage technical guide. The blue loading dye reaching the ‘foot’ of the gel and the ladder separation were parameters by which sufficient separation was assessed. During this time blotting pads were soaked in lx transfer buffer: supplemented with 10% methanol and lml antioxidant. Filter paper and 0.45mM nitrocellulose membranes were cut to correct size for the transfer.

Electro-transfer to Nitrocellulose Membrane.

Upon suitable separation of the ladder and by extension the proteins; the Nupage Electro- transfer cassette was prepared in accordance with the protocol (see NuPage Technical guide 2013).

Once pre-soaked blotting pads were applied to the bottom of the electro-transfer cassette, the 3-8% Tris acetate gels cases were‘cracked’ open to liberate the polyacrylamide gels. The top of the gel, above the top band of the Hi-Mark ladder was removed and disregarded. The remainder of the gel was floated, using the buffer, to be situated above filter paper, lifted out of the transfer buffer and placed to the transfer cassette. Once performed, the 0.45pm nitrocellulose membrane was submersed in buffer and placed on top. This was then rolled across the surface of the gel using a plastic roller to ensure tight contact throughout. A filter paper and a blotting pad placed on top. The whole cassette was kept wet during this time. If a second gel was present then the process was repeated. When completed this was placed in the I-Cell tank. The top of the electro-transfer cassette was refilled with IX transfer buffer and the surrounding area filled with cold ddH20. The proteins were then transferred for 2 hours at 30V.

Post-Transfer Checks and Blocking

Following the two hour transfer, the membrane was stained with IX Ponceau. This stains all proteins across the lanes and is used to ensure that the transfer was complete and successful. The stain was then washed off with 0.1% PBS-T, washing at 5 minute intervals until no stain was visible. The nitrocellulose membrane was then blocked with 5% Marvel milk in 0.1% PBS-T for 1 hour at room temperature, to prevent non-specific binding. Once the membrane was blocked, the membrane was cut in half between the 55 and 7lkDa HiMark Ladder bands. The top piece of 0.45mM nitrocellulose was incubated at 4° overnight in a 1 in 100 diliution of 6C5 or MannExlO-l lc and the bottom a 1 in 2500 dilution of Rabbit Anti tubulin.

Visualisation of the Nitrocellulose Membrane Using The Odyssey

Following overnight incubation with primary antibodies, 4 washes in 0.1% PBS-T was undertaken for 5 minutes. The secondary antibodies, which are conjugated to a fluorescent label were diluted to 1 in 10,000. The nitrocellulose membrane was then incubated for a further hour, prior to the repetition of 4 washes for 5 minutes in 0.1% PBS-T. This was then scanned at 700nm and 800nm channels respectively, using the Odyssey SA machine (Li-Cor). Dystrophin would be present within the 800nm channel and the a-tubulin present within the 700nm channel.

Quantification of Dystrophin

Software

• ImageStudios Version 4 (Li-Cor)

Methods

The nitrocellulose membrane was then visualised with Image Studios Version 4. Bands were automatically identified using the software, and adjusted to be tight to the band in question in individual 800nm and 700nm channels. User defined noise values were subtracted away from the band in question and used to attain intensity values, for both dystrophin (800nm) and a-tubulin (700nm). The values were normalised to tubulin:

Dystrophin Fluorescence Intensity

- - - Normalised Values a Tubulin Fluorescence Intensity This was performed for each lane and the native and optimised dystrophin (n=5) and a mean value was attained. Once mean values were attained the Sequence optimised dys/tub ratio was normalised to the native dys/tub, to establish a fold difference in expression.

Results

Initial Assessment of dystrophin expression from native and optimised, GFP tethered dystrophin constructs:

Initially, a visual indication of whether sequence optimisation improves transcription and subsequently synthesis of recombinant dystrophin protein was sought. In pursuit of this plasmids containing native and optimised full-length dystrophin cDNA (SEQ ID NO: 1) directly tethered to eGFP were transiently transfected into Hek293T culture. Plasmids were driven by the Cytomegalovirus (CMV) promoter, a strong viral promoter to ensure higher levels of protein expression. The direct tethering of eGFP to dystrophin enables fluorescence to be used as an indicator of dystrophin expression; particularly, as the stoichiometry of eGFP: dystrophin is equivalent in the resultant fusion protein.

Both full-length native and optimised dystrophin plasmids were transfected at a 4pg dose to Hek293T using a 5: 1 transfection reagentDNA ratio. Cultures were then subject to microscopy imaging at 24, 48 and 72 hours post transfection; the latest time point in this series, being reflective of the time taken for dystrophin protein to accumulate, to optimal levels for detection by western blotting.

There was an apparent difference in green fluorescence, observed between native and optimised cultures post-transfection, across all time points examined. Initially, the number of GFP positive cells in the culture, indicated successful transfection of both native and optimised constructs. Moreover, the intensity of fluorescence appears to increase from 24-72 hour time points irrespective of optimisation; likely attributable to the accumulation of dystrophin-eGFP transcript and subsequent protein synthesis. However, there was a striking increase in fluorescence intensity, observed in the optimised dystrophin culture relative to the native, at all time points examined (Figure 1). This was suggested to be due to the sequence optimisation of the plasmid. The proposed implication being that the optimised construct enhances transcriptional efficiency and subsequently dystrophin-eGFP protein synthesis.

Quantifying the difference in expression of native and optimised dystrophin constructs driven by a CMV promoter:

The increased fluorescent intensity observed, prompted direct assessments of dystrophin protein expression for both CMV driven Nat-hDys and Opt-HuDys (SO) constructs. It was resolved that constructs without the GFP tag should be used in this examination. Transient transfections of plasmids were repeated at 4pg utilising the 5: 1 viafect transfection reagentDNA ratio previously described. Cultures were incubated for 72 hours post transfection and lysed for total protein; this being in line with optimal accumulation of dystrophin protein. The samples were subsequently quantified and screened with two dystrophin antibodies: The 6c5 antibody, which binds to the carboxyl C -terminus of the dystrophin protein and the MannExlOl lc, which binds to a dystrophin protein epitope, encoded between exons 10 and 11. This was performed prior to subsequent visualisation and quantification against an alpha-tubulin loading control using the Odyssey Licor system.

Importantly both constructs Nat-hDys and Opt-HuDys (SO), expressed a 427kDa protein as determined by a HiMark Nupage Ladder, consistent with full-length dystrophin. This was detected with both the 6C5 and the MannExlOl lc antibodies. Congruent with the previous investigation, the sequence optimised construct yielded a larger area band with increased intensity relative to the native. This trend was retained across 5 samples that were transiently transfected (Figure 2 and Figure 3). During the quantification, dystrophin fluorescence in the 800 channel was divided by tubulin in the 700 channel and a mean of Opt-HuDys-SO ratios attained were normalised to that of Nat-Dys. This form of analysis indicated a striking 57-fold difference with 6C5 antibody and a 22-fold difference with the MannExlOlc antibody. This was deemed statistically significant in both cases giving a p=<0.00l by an unpaired t-test.

Additionally, it should be emphasised that on examination of the western blot an alternative banding pattern between the two antibodies was observed. The 6C5 antibody appeared to produce a characteristic laddering pattern in the Opt-HuDys (SO) construct that was not observed in the case of the Nat-hDys construct. The additional banding seen had moderate intensity comparable to the full-length band. In contrast the MannexlOl lc antibody resulted in a singular sharp band for both Nat-hDys and Opt-HuDys (SO) constructs, with few additional bands at low intensity. In the case of the latter, this is likely the result of increasing the brightness to visualise the Nat-hDys bands.

Quantifying the difference in expression of native and optimised dystrophin constructs driven by a Spc512 promoter:

In the first series of investigations, CMV driven constructs were used. However, in the context of translational application, a muscle specific promoter would be advantageous in providing expression localised to muscle (Counsell JR et al, Sci Rep. 7:44775 (2017) and Meng, J. et al., Scientific Reports, 6(1), p.19750 (2016)). In examination of this, Nat- Dys and Opt-Dys-SO constructs driven by the muscle specific Spc5 l2 promoter, were transiently transfected at a 4pg dose and protein harvested at 72 hours. Protein samples were prepared, subject to western blot and quantified in a manner consistent with the CMV based experiments.

The Opt-Dys-SO construct once again resulted in a protein band of a greater area with an increased fluorescent intensity, relative to Nat-Dys with 6C5 and ManExlOl lc antibodies (Figure 4) and (Figure 5). In this instance a 15-fold difference and a 17-fold difference were determined respectively. Notably, the difference whilst less prominent than observed with the CMV driven constructs, is still present. Finally, the laddering effect previously observed with the 6C5 antibody, is retained even with the use of an alternative promoter. This finding was somewhat unexpected due to the use of the Spc5 l2 muscle- specific promoter, but was attributed to the Hek293T cell culture enabling ‘leaky expression’.

Design of an Exogenous repair template:

The exogenous repair template was designed with numerous features to make it optimal for this investigation. Firstly, it included a backbone sequence amenable to the cloning of multiple dystrophin cDNA variants, including the full-length optimised sequence. Secondly, a splice acceptor was appended at the 5’ end, in place of a promoter. Moreover, the presence of the 5’ splice acceptor would enable the endogenous Dp427m promoter and indeed other full-length promoters to splice to the exogenous repair template. As a consequence, the resultant protein will have the correct spatial and temporal expression patterns. It is hypothesised that this will ameliorate a range of pathogenic disease causing mutations across the DMD gene.

To attain a backbone sequence amenable to the cloning of a variety of dystrophin cDNA transgenes, all unique restriction endonucleases were identified across full-length Opt- HuDys (SO). This served to identify two unique restriction sites. At the 5’ end this was Fsel, present 30 nucleotides into exon 6 and at the 3’ end Notl situated at the terminus of exon 79. Thus a sequence was constructed with: DMD cDNA of exons 2-6 upstream of an Fsel site, an intervening cyan fluorescent marker sequence and finally, a Notl site. It was determined that this sequence would enable directional cloning of the sequence optimised dystrophin variant.

In construction of an Exon 2-6 DMD cDNA block, consensus sequences of exons 1-6 of the Dp427m isoform, were aligned against full-length sequence optimised dystrophin cDNA. In this manner exons 1-6 of sequence optimised dystrophin cDNA, was identified. Exon 1, determined as the first 31 nucleotides of the sequence, was subsequently removed (Koenig et al., Cell , 50(3), pp.509-17 (1987)). It was anticipated that these nucleotides would be reconstituted by the endogenous Dp427m promoter, if successful splicing to a delivered dystrophin transgene occurred. To this purpose, exon 2 was flanked at the 5’ end with a human b-globin synthetic splice acceptor, and other regulatory sequences to facilitate splicing, including: a polypyrimidine tract and synthetic branch points (Seth et al, The Journal of biological chemistry , 283(15), pp.10058-67 (2008); Popplewell et al., Human gene therapy , 24(7), pp.692-701 (2013)). These sequences were modified to include a silent mutation, to generate a Hpal restriction site and identified as a strong splice acceptor by human splice finder (Desmet et al., Nucleic acids research , 37(9), p.e67 (2009)). Thus this splice sequence was selected to facilitate the splicing of the endogenous promoter to the integrated dystrophin transgene. Moreover, the inclusion of the Hpal restriction site would enable this sequence to be replaced with a native sequence with ease should this be required.

The 3’ end of this cDNA block, downstream of the Notl site, was also flanked by a sequence indicated to improve transcription in lentiviral vectors; this being a mutated Woodchuck Hepatitis Virus Post-transcriptional regulatory element (mWPRE) fused to a polyA (Ranzani et al, Nat Methods. 10(2): 155-61 (2013)).

Between these Fsel and Notl sites, a Cyan fluorescent protein (CFP) was encoded. The sequence was modified to remove the initiating methionine and append the first two nucleotides of DMD exon 2. This was undertaken to retain the open reading frame of partial Dys-CFP fusion protein and reduce background fluorescence that may arise from aberrant firing of the methionine. This was anticipated to provide the benefit of a visual blue-fluorescent output, which could be used to indicate Dys-CFP transgene integration and track subsequent enrichment processes. It was anticipated that this would streamline the development of integration methods, in Hek239T and patient myoblast cultures. Moreover, the intervening CFP sequence could be used as a spacer within the DMD intron 1 repair template. The presence of a 49lbp band, removed upon Fsel and Notl double digest could be used to indicate successful cleavage of the repair template and facilitate sub-cloning of dystrophin cDNA variants.

Due to the HDR pathway occurring with relatively low efficiency, an antibiotic selection cassette, SV40-Zeocin-PolyA, was placed downstream of the WPRE and PolyA sequences. This encodes the She Ble protein, which when expressed renders the zeocin antibiotic inactive (Hockemeyer et al., Cell stem cell , 3(3), pp.346-353 (2008)). This sequence was modified to include a silent point mutation to remove a Fsel site present. This ensured the unique core Fsel and Notl sites required for directional sub-cloning of dystrophin variants was retained.

The zeocin selection cassette was resolved upon, as during immortalisation process of patient myoblasts, harbouring the deletion of exons 45-52 (D45-52); they were rendered resistant to puromycin and neomycin antibiotics (Mamchaoui K. et al, Skeletal muscle , 1, p.34 (2011)). Importantly, the cassette was floxed with LoxP sites that can conditionally remove intermediate sequences, in the presence of Cre-recombinase. Inclusion of the sequences of the LoxP sites was necessitated as post-enrichment of cells with successful integration; continued expression of the She Ble protein is undesirable. This is due to concerns that if cellular material is used to treat patients, in an ex-vivo engraftment manner they may acquire antibiotic resistance, which would have implications on their endogenous flora (Marie et al., The Journal of Gene Medicine, 12(4), pp.323-332 (2010)).

Once the sequences of all constituents were obtained, they were flanked with lkb arms of homology. These were derived from human DMD Intron 1 consensus sequence from NCBI. The lkb arms initiated 6 nucleotides upstream of guide 3 and downstream of guide 4; these being the most upstream and downstream of the CRISPR guides identified. The arms of homology were designed in this manner, as a single guide with the highest efficacy had not yet been identified. As a direct consequence of this, no guide sequences were encoded within the exogenous repair template. Thus circumventing the risk that the repair template may be cleaved or indeed dystrophin variants re-targeted upon integration into the genome, by the Cas9 system.

Once a full sequence of the human DMD intron 1 repair template was compiled, it was assessed in parallel with dystrophin variants and the ISceit-Lentiviral vector for common for non-cutters. The list of common non-cutters were examined for compatibility in a double digest setting and used to flank all components of the exogenous repair template. This sequence was used and the repair template in a Puc57 backbone was synthesised (Figure 6). Finally, the sequence was inserted in reverse orientation into the ISciet lentiviral backbone; this was performed to maintain the viral mRNA structure and prevent aberrant splicing or termination occurring (Figure 7).

Sub-cloning the optimised full-length dystrophin cDNA into the Puc57 Intron 1 exogenous repair template:

Following the synthesis of the Intron 1 exogenous repair template, focus was shifted to the sub-cloning of the optimised full-length dystrophin from Exon 6 to exon 79 between the Fsel and Notl endonuclease restriction sites. To this purpose, the optimised full-length dystrophin was subject to a series of diagnostic digests to assert the identity of the construct. Once assured the construct digested in a manner consistent with that anticipated; both the destination vector Puc57-hINTl-RT and the optimised full-length dystrophin plasmids were subject to double digest with Fsel and Notl restriction endonucleases. In the case of the destination vector the double digest served to remove the CFP marker and leave a 6638bps backbone. Whereas in the case of the optimised full- length dystrophin it enabled the cDNA insert of interest to be liberated. Samples were resolved by electrophoresis and subjected to overnight ligation, post gel purification. Subsequent ligation mixtures of insert and destination vector and a vector control, containing only digested backbone, were then subjected to standard heat shock transformation. The E.coli suspension was allowed to recover and plated onto ampicillin plates for an overnight incubation at 30°C. The vector control yielded no colonies, indicating that no self-ligation had occurred, likely attributable to the incompatible DNA termini resulting from the double digest. In contrast, the ligation mixtures yielded a high number of single colonies that could be picked and characterised. These results taken together serve to indicate that the optimised full-length dystrophin fragment from exon 6-79 was likely ligated into the destination vector backbone.

In an attempt to assess whether ligation was successful a colony PCR was undertaken. The primer pairs were designed for the colony PCR, so the forward primer was present within the Exon 2-6 region of the Puc57-hINTl-RT backbone upstream of the Fsel cleavage site; whereas the reverse primer would only be present if the optimised full- length dystrophin insert was ligated. Thus the resultant amplicon was only anticipated to occur in instances where the optimised full-length dystrophin transgene had been successfully inserted into the Puc57-hINTl-RT destination vector. Amplification of the Spc5 l2-HuDys (SO) plasmid was used as a positive control, the Puc57-hINTl-RT destination vector pre-digest and ligation served as a negative control.

The first 4 colonies from the colony PCR series were selected, grown as a starter culture, mini-prepped and subject to restriction digest. This was to further confirm presence and identity of the optimised full-length dystrophin transgene insert. Initially, the Fsel and Notl double digest were performed on putative Puc57-hINTl-HuDys-RT constructs, alongside the parental Spc5 l2-HuDys SO plasmid, which provided the insert. All four colonies gave the anticipated digest profile, alongside the Spc5 l2-HuDys (SO) control plasmid.

Additionally, a diagnostic digest using the Seal restriction endonuclease was undertaken. This produces distinct banding patterns for the parental Spc5 l2-HuDys (SO) and the Pu57-hINT-HuDys-RT. Once again the four colonies gave the anticipated digest profiles, consistent with the optimised full-length dystrophin being inserted into the Puc57-hINT- RT. The above screens indicate a repair template carrying the optimised full-length dystrophin exons 2-79 and an independent selection cassette, all flanked by arms of homology was produced.

Discussion

Sequence Optimised Dystrophin cDNA:

Sequence optimisation of full-length dystrophin cDNA was shown to enhance protein expression relative to native controls. This was established by microscopy imaging and western blotting. Both studies utilised plasmid constructs under the control of a CMV promoter, with the first investigation using dystrophin variants directly tethered to eGFP. Importantly, in the case of full-length dystrophin, a 22-fold difference in protein expression was observed between native and optimised constructs. This large increase in protein expression from optimised cDNA from both experiments was striking.

The investigation was then extended to include optimised full-length dystrophin cDNA under the control of the Spc5 l2 muscle restrictive promoter (Li et al., Nat Biotechnol. 17(3):241 -5 (1999); Athanasopoulos et al., Methods Mol Biol. 709:21-37 (2011)). This was performed to examine whether the expression of Spc5 l2 driven constructs could be compared in Hek293T cell culture. Importantly, the Spc5l2 promoter enabled full-length dystrophin expression in Hek293T cell culture; this is speculated to be due to this cell line facilitating leaky expression of this promoter. Interestingly, native and optimised full- length dystrophin cDNA produced a varying fold difference in protein expression, when constructs were driven by the CMV and Spc5 l2; being 22-fold and 15-17 fold respectively. This effect is attributed to two main parameters: the first being the relative strengths of the viral CMV and the muscle restrictive Spc5 l2 promoters. Secondly, it was speculated that the expression of the full-length dystrophin cDNA may be reduced with the Spc5 l2 promoter, due to its restrictive expression pattern.

The observations from both CMV and Spc5l2 studies together, could hold important implications for clinical translation. Numerous studies have provided an insight into the amount of dystrophin protein expression required relative to wild-type endogenous levels to ameliorate dystrophic pathology. Variable estimates have arisen, likely attributable to the nature of investigation from which estimates were derived and how dystrophin levels were quantified. Dystrophin expression of: 30% in BMD patients, 15% following antisense therapy in mice and finally, 20% in transgenic mice, were all shown to confer therapeutic benefit. Whilst estimates of therapeutic dystrophin expression vary between 15-30% among these investigations; they agree a uniform dystrophin expression across the majority of myo fibres, as opposed to a sporadic distribution provides greater functional improvement. In addition, they indicate that the level of dystrophin correction required to be therapeutic, will be influenced by the muscle pathology and disease progression of the patient seeking treatment. The striking increase in protein expression observed, with the use of sequence optimised cDNA encoding full-length dystrophin, and relative to its native counter-part could facilitate the attainment of such expression thresholds. By extension of this it is speculated they could greatly improve clinical outcomes and functional improvements observed in clinical trials.

The striking fold differences in protein expression observed between native and optimised constructs necessitate the examination of the potential effects of supra-physiological levels of dystrophin proteins. Importantly, others have demonstrated that overexpression of full-length dystrophin, of up to 50-fold higher than endogenous levels, was well tolerated (Chamberlain, Soc Gen Physiol Ser. 52: 19-29 (1997); Phelps SF, Hum Mol Genet. 4(8): 1251-8 (1995); Wells DJ, Hum Mol Genet. 4(8): 1245-50 (1995)). This level was shown to ameliorate dystrophic pathology with the absence of any detrimental effects. Design of an exogenous repair template:

The improved protein expression resulting from sequence optimised full-length dystrophin cDNA served to influence exogenous repair template design. This prompted the generation of a backbone that would enable the directional sub-cloning of sequence optimised full-length dystrophin cDNA, allowing the exogenous repair template to be trialled in integration experiments.

The exogenous repair template was designed to facilitate a Homology Directed Repair (HDR) outcome, at the human DMD intron 1 locus. It was designed with lkb arms of homology isogenic to the human genome, directly upstream and downstream of the region of homology identified. This length of isogenic sequence is deemed optimal for successful exploitation of HDR pathways. Extension of homology arms beyond this size only result in marginal increases of transgene integration. In addition, the repair template also includes a floxed zeocin cassette, to facilitate positive selection processes (Mulsant et al, Somat Cell Mol Genet. l4(3):243-52 (1988); Seth et al, The Journal of biological chemistry , 283(15), pp.10058-67 (2008)). This would enable enrichment of corrected cells, which is important due to the low efficiency of the HDR process.

Importantly, as scientists’ understanding of the DNA damage response (DDR) continues to evolve, so too does the manner in which DNA repair pathways are exploited to facilitate the integration of genetic material. Recent investigations have used NHEJ- DNA repair pathways to introduce genetic material (Maresca et al, Genome Res. 23(3):539-46 (2013); Suzuki et al., Nature. 540(7631): 144-149 (2016)). This strategy is reliant upon genomic target sites of the endonuclease TALEN or CRISPR, being encoded in reverse orientation directly adjacent to the transgene for which integration is desirable. The resultant in-situ cleavage of genome and exogenous repair template, facilitates the integration of the transgene independently of the HDR pathway (Suzuki et al, Nature. 540(7631): 144-149 (2016)). Importantly, the exogenous repair template in this investigation was designed so components were flanked with endonuclease restriction sites. Thus it could be easily customised to facilitate exploration of such strategies with dystrophin cDNA. Conclusions

A novel exogenous repair template was designed with restriction sites enabling the sub- cloning of full-length sequence optimised dystrophin cDNA. This design was founded upon the demonstration that sequence optimisation enhanced recombinant dystrophin protein expression. It was designed with lKb arms of homology isogenic to sequences upstream and downstream of the CRISPR MIT guide designs, identified within human DMD intron 1. Furthermore, it encodes a floxed zeocin cassette to facilitate positive selection during HDR investigations.

SEQUENCES

SEQ ID NO. 1 is a codon optimised full-length dystrophin cDNA including a 3bp“stop” codon (nucleotides 11,059-11,061). SEQ ID NO. 2 is the amino acid sequence of the human native dystrophin protein.

SEQ ID NO. 3 is a codon optimised full-length dystrophin cDNA (nucleotides 7-11,064) including a 6bp optimised Kozak sequence (nucleotides 1-6) and a 3bp“stop” codon (nucleotides 11,065-11,067).