Open access peer-reviewed chapter

Duchenne Muscular Dystrophy (DMD) Treatment: Past and Present Perspectives

Written By

Nahla O. Mousa, Ahmed Osman, Nagia Fahmy, Ahmed Abdellatif and Waheed K. Zahra

Submitted: 28 February 2020 Reviewed: 07 May 2020 Published: 13 June 2020

DOI: 10.5772/intechopen.92765

From the Edited Volume

Muscular Dystrophy - Research Updates and Therapeutic Strategies

Edited by Gisela Gaina

Chapter metrics overview

884 Chapter Downloads

View Full Metrics

Abstract

Duchenne muscular dystrophy (DMD) is one of the fatal X-linked disorders that are characterized by progressive muscle weakness and occur due to mutation in the largest human gene known as the DMD gene which encodes dystrophin protein that is mandatory for keeping the muscles structurally and functionally intact. The disease always affects boys (1 from every ~5000), and in some cases the female carriers are symptomatic. The disease usually leads to impairment in cardiac and pulmonary functions leading to the death of the patients in very young ages. Understanding DMD through precise molecular diagnosis will aid in determining the suitable therapeutic approach for the cases like designing exon-skipping antisense oligonucleotides (AOs) or stem cell-based therapies in conjunction with gene editing techniques (CRISPR/Cas9). Such therapies can correct the genetic defect in the DMD gene and ameliorate the symptoms. In this chapter, we will illustrate the past and current strategies for DMD disease treatment.

Keywords

  • DMD
  • exon skipping
  • CRISPR
  • cardiosphere
  • utrophin

1. Introduction

Duchenne muscular dystrophy (DMD) is a fatal X-linked disorder characterized by skeletal muscle wasting that is resulted from mutations in the dystrophin gene [1]. The disease occurs at a frequency of about 1 in ~5000 newborn males, making it the most common severe neuromuscular disease in humans. Dystrophin is present in normal individuals from fetal life onwards in all skeletal, cardiac, and smooth muscles; the absence of dystrophin protein causes muscle weakness and protein degradation and ultimately causes cell death. Death usually occurs in the third decade of life as the result of respiratory or heart failure [2]. The precise diagnosis for DMD should contain a combination of genetic testing after muscle biopsy and clinical observation of muscle strength and function.

The main current medication so far is corticosteroids, which have been shown to increase muscle strength in many studies. Genetic therapy using mini-/microdystrophin vectors, suppression of premature termination codon, exon-skipping antisense oligonucleotides (AOs) which bind with RNA and exclude specific sites of RNA splicing producing a dystrophin that is smaller but functional, and such new emerging drugs are the pass to the new era towards DMD treatment. In the next section, we will review all available FDA-approved treatments and recent research trials aiming at ameliorating DMD symptoms.

Advertisement

2. Methods for treatment

2.1 Corticosteroids

Corticosteroids were the first line of treatment for DMD; it was first used by Drachman et al. in 1974 [3] when they had promising positive results in their study after using prednisone (anti-inflammatory glucocorticosteroid). Since then, many studies were carried out to test the therapeutic effect of such treatment since it was found to improve muscle performance.

Deflazacart (DFZ), an oxazolidine derivative of prednisone, was used by an Italian group [4] and other groups [5, 6, 7], and the drug demonstrated efficiency in disease treatment and preserved lung function. The exact mechanism of DFZ is not yet known; however, it might regulate some signaling cascades. It was found to activate calcineurin/NF-AT pathway [8]. Also, DFZ may act by decreasing necrosis and muscle inflammation and reducing the degree of muscle degeneration. It can also act through modulating dystrophin expression and inducing the myogenesis in addition to having positive effects on muscular tissue mass [9].

Despite the advantages of using steroids, they also had side effects like gaining weight, affecting bone mineral density, which leads to vertebral fractures and behavioral changes. Furthermore, high dosages are required to reach the target effect and to be active at the site inflammation. Also, the drug can be accumulated in other nontargeted areas [10, 11].

In one of their studies, Luhder et al. [12] tried to improve the therapeutic effect of the steroids through developing an 80 nm PEGylated nano-liposome that is conjugated with the steroid prodrug “methylprednisolone hemisuccinate.” The results of their study showed that such structure was selectively targeting the diaphragm in vivo (using mdx mouse model) when administered intravenously and the treatment reduced the infiltration with macrophages and serum levels of transforming growth factor beta. Most importantly, the study showed that long-term use of this formulation leads to enhanced mobility and increased muscle strength.

2.2 Exon skipping

Exon skipping is considered as one of the mutation-based treatments for Duchenne muscular dystrophy [13]. In DMD, some deletions in specific exons lead to the disruption of the reading frame of the dystrophin protein, and consequently such deletions lead to the production of truncated product missing a huge part of the protein (usually missing the rod domain and C-terminal domain).

However, sometimes, deleting additional exons may restore the reading frame and lead to the production of dystrophin protein missing only a portion of the central rod domain while the C-terminal domain remains intact, and hence the protein product in this case is lacking specific regions, but it is semi-functional and can induce Becker-like symptoms instead of the complete loss of the muscular function [14].

The main idea of exon skipping is using the “antisense oligonucleotide” molecules to induce the skipping of a specific exon (other than the already mutated one) and prevent it from being translated to restore the reading frame. As an example, patients with exon 45 deletion could be treated through the skipping of an additional exon 44. Eteplirsen (Exondys51™) based on phosphorodiamidite morpholino oligomer (PMD) is an FDA-approved antisense treatment to skip exon 51 for patients with mutation ▲49–50 [15]. Also, drisapersen (based on 2′-O-methyl phosphorothioate; 2′-OMePS-modified AOs) is one of the AOs that are designed to treat DMD patients with mutations that can be ameliorated by exon 51 skipping; however it was not approved by the FDA [16, 17].

Various modifications can take place to the sugar of the oligonucleotide or to the backbone of the oligo. This could include phosphorodiamidate morpholino, locked nucleic acid (LNA), or peptide-conjugated oligo. Regarding the morpholinos, the oligonucleotide backbone is replaced with the morpholino backbone which makes the oligonucleotide nontoxic and has high affinity to RNA molecules. The locked nucleic acids are oligonucleotides that have a modified ribose sugar where the 2′ oxygen is connected with the 4′ carbon atom which creates a locked ribose ring. Also, the LNAs are nontoxic with superior affinity to complementary targeted RNA sequences [18].

The main problem in developing such treatments based on the skipping is that it will only fit a small group of patients (a mutation-specific AO should be developed for each group of patients and will not be suitable for other patients); also some patients have deletions in critical parts of the protein, and hence skipping of other exons will not have a therapeutic impact (Table 1).

ChemistryRoute of administrationThe used modelTreatment strategyTreatment effectsReference
Phosphorodiamidate morpholino oligomers (Ex6A, Ex6B, Ex8A, and Ex8G)IntravenousNeonatal CXMDJExon 6–9 skippingDystrophin restoration across skeletal muscles (14% of healthy levels)
Reduction of fibrosis and/or necrosis area
[19]
Phosphorodiamidate morpholino oligomer (NS-065/NCNP-01)Endo-Porter reagentFibroblasts from patients with DMD involving deletion of exons 45–52 or exons 48–52 and injected with MYOD for myotube differentiationExon 53 skippingRestored dystrophin protein levels in the cells[20]
Phosphorodiamidate morpholino oligomerIntramuscular and intravenousmdx52 mouse modelExon 51 skippingOnly the protocol was mentioned[21]
Phosphorodiamidate morpholino oligomer (NS-065/NCNP-01)IntravenousPatients with DMDExon 53 skippingIncreased dystrophin/spectrin ratio in 7 of 10 patients in TA muscle biopsies[22]
Pip6a-PMO; PMOME23, sequence GGCCAAACCTCGGCTT-ACCTGAAATIntravenousCmah-/-mdx miceExon 23 skippingDystrophin restoration in the heart
Reduction in myocardial fibrosis
Reducing maximum pressure and arterial elastance
[23]
Inhibitor of CDC2-like kinase 1 (named TG693)Oral
Lipofectamine reagent
Male Jcl:TCR mice
Patient-derived myotubes
Exon 31 skippingIt induces exon skipping and restored dystrophin expression in patient-derived cells. And it modulated splicing in mouse skeletal muscle[24]
Morpholino AOs targeting DMD exon 51Endo-Porter transfection
Intramuscular
Immortalized DMD muscle cells
hDMD/Dmd null mice
Exon 51 skippingThe rescue of dystrophin protein expression[25]

Table 1.

Studies conducted on treatment of DMD using exon skipping (during 2017–2019).

2.3 Induced pluripotent stem cells along with genome editing technique

The sole cause of DMD is the presence of mutation that adversely affects the DMD gene. So, in order to permanently fix such mutations and treat this condition, patients could be provided with muscle cells harboring the normal copy of DMD gene. Since it is hard to get mature muscle fibers from a normal individual to be used as a source of healthy muscle cells with normal DMD gene, also the availability of such source of cells will not guarantee the process of grafting in the patient’s muscles since it could be subjected to rejection by the body and can initiate an aggressive immune response. Cell reprogramming and genome editing techniques efficiently aid in solving this puzzling dilemma [25]. The process of cell reprogramming paved the road towards developing normal muscle fibers by starting with patient-specialized adult cells followed by inducing the production of induced pluripotent stem cells (iPSCs) (using the Nobel prize-winning technology of reprogramming using specific transcription factors like Oct4, Sox2, Klf4, and L-Myc) [26]. Also, some microRNAs have the potential to reprogram the adult cells efficiently (like miR-302b, miR-372) [27].

After the reprogramming and the production of stem cells, gene editing technologies should be used to correct the mutation of the gene. CRISPR/Cas 9 is now a leading technology that is presently considered as an avenue for DMD treatment; the RNA-guided DNA endonuclease system allows the correction of the DMD segment which is essential for dystrophin restoration [28, 29].

In order to conduct a gene editing experiment with CRISPR/Cas9 system, two important elements should be provided: guide RNA (gRNA) specific for the target gene and Cas9 nuclease (Sp. Cas9 (from Streptococcus pyogenes; 4.10 kb) or Sp. Cas9 (Staphylococcus aureus; 3.16 kb)) or Cj. Cas9 (Campylobacter jejuni; 2.95 kb) that can cleave DNA strands where the guide RNA is bound and in the presence of three- to five-nucleotide proto-spacer adjacent motif (PAM) sequence to be digested. Upon the binding of the gRNA, Cas9 can induce a double-strand break which is then repaired by the cell through the nonhomologous end joining, and this will initiate a repair mechanism in which nucleotides will be added or deleted at the cleaved site which can consequently restore the reading frame of the DMD gene to the normal ORF. In some cases, single (or several) gRNA molecule could be designed to target splicing sites which can lead to the skipping of specific exon leading to the production of functional proteins. Additionally, base editing mediated by CRISPR/Cas9 could be obtained through Cas9 enzymes lacking the nuclease activity, so it can induce only a single-strand break. Such enzymes can catalyze base editing (A:T to G:C) through having a cytidine deaminase activity [30].

Ousterout et al. in their study used another editing protocol (zinc finger nuclease) to delete exon 51 from the transcript from patient-derived myoblasts [31]. Also, Young et al. carried out CRISPR/Cas9 experiment utilizing a single pair of guide RNAs to delete exons 45–55 in iPSC, and such deletion leads to the expression of stable dystrophin protein with improved membrane stability in derived skeletal myotubes and cardiomyocytes [32]. Another study by Duchene et al. utilized a single guide RNA to produce a hybrid exon which led to the production of functional dystrophin protein with completely normal structure [33]. The main advantage of this reprogramming protocol is that it allows performing an autologous grafting of the muscle cells to patients.

For the expression of the specific gRNA molecules inside the muscle cells, adeno-associated virus (AAV) vectors will be used. Sometimes, the expression of the gRNAs can lead to off-target effect due to the incorrect binding with another similar DNA sequence inside the host cell. In order to avoid this damaging effect, AAV vectors expressing multiple gRNA molecules could be used.

After the completion of the gene editing process, the edited cells would be treated with myogenic factors to convert the edited stem cells again to myoblasts for the myogenic differentiation (Table 2).

Plasmids (source of Cas9 and guide RNAs)Route of administrationThe used modelTreatment strategyReference
Adeno-associated viral vectors of serotype 9 carrying an intein-split Cas9
A pair of guide RNAs targeting sequences flanking exon 51 (AAV9-Cas9-gE51)
Intramuscular injectionDMDΔ52 pigsExcision of exon 51[34]
SaCas9 expression plasmid
Two gRNA expression cassettes driven by the human U6 pol. III promoter
(AAV8 and AAV9)
Locally in the TA musclesC57BL/10ScSn-Dmdmdx/JExcision of exon 23[35]
pSpCas9 expression plasmid
AAV TRISPR-sgRNA-CK8e-GFP plasmid contained three
sgRNAs driven by the U6, H1, or 7SK promoter and green florescent protein (GFP) driven by the CK8e regulatory cassette
Transfection reagent
Locally in the TA muscles
Human DMD-derived iPSCs
▲Exon 44 DMD mice
Excision of exons 43 and 45[36]
Streptococcus pyogenes Cas9
Single guide RNA (sgRNA-51)
(AAV9-Cas9 and AAV9-sgRNA-51)
Locally in the cranial tibialis muscles▲Exon 50 canine modelExcision of exon 51[37]
spCas9 and crDMDint2.1 and int2.6 gRNAsTransfection reagent (linear polyethylenimine derivative)Immortalized myoblasts from DMD patientExcision of duplicated exon 2[38]
Lenti-V2-Ugi-nCas9-AIDx or Lenti-V2-AIDx-nSaCas9 (KKH)-Ugi (2.5 μg) and pCDNA3 UgiTransfection reagent (lipid-based)▲51-iPSCs of a male DMD patientExcision of exon 50[39]
CRISPR-Cas9 variant (D10A Cas9 nickase (nCas9) or catalytically deficient D10A/H840A Cas9 (dCas9) from S. pyogenes) and a deaminase protein from various sources
sgRNA (gX20) under the control of the U6 promoter (pAAV-ITR-ABE-NT-sgRNA)
MicromanipulatorMouse zygote from DMD knockout mouseBase editing of exon 20[40]
Plasmids containing regulatory cassettes for expression of Cas9 or gRNAs flanked by AAV serotype 2 inverted terminal repeats
(ITRs)
Electroporation
Intramuscular
Fibroblasts isolated from male mdx4cv mice
Male mdx4cv mice
Excision of exons 52 and 53[41]
pX601-AAV CMV::NLS-SaCas9-NLS-3xHA-bGHpA;U6::BsaI-sgRNA (PX601)Transfection reagent (lipid-based)MyoblastsExcision of exons 47 and 48[33]
DMD iPSCExcision of exons 44–55[32]
The gRNA cassettes gI43, gI52, gI53, and gI54.2 (targeting different introns) controlled by human U6 RNA polymerase III promoter
Plasmids AU53_pAd
Shu.gI52.gI53.PGK.Cas9.SV40pA, plasmids pLV.gI52 and pLV.gI53
Transduction (by gelatin)DMD myoblasts Δ48–50 and Δ45–52Excision of exon 53[42]
pAAV-ITR-CjCas9-sgRNA, pAAV2/9 encoding for AAV2rep and AAV9cap, and helper plasmidIntramuscular in the TA muscleKnockout miceExcision of exon 23[43]
pSpCas9(BB)-2A-GFP (PX458)Transfection reagentDMD hiPSCs, hiPSC-derived cardiac muscle cellsExcision of exon 51, introns 47, 50, 54[44]
pSpCas9(BB)-2A-GFP (PX458)Intramuscular in the TA muscleMice ▲50Excision of exon 51[45]
Purified Cas9 protein and in vitro transcribed gRNAGold nanoparticlesPrimary myoblasts C57BL/10ScSn-Dmdmdx/J (mdx) miceExcision of exon 23[46]
pSpCas9(BB)-2A-GFP (PX458)NucleofectionInduced pluripotent stem cells (iPSCs)Deleting exons 3–9, 6–9, or 7–11[47]
Nuclease-expressing plasmids (TALENs, left and right; CRISPR, Cas9 and sgRNA)ElectroporationDMD fibroblasts were derived from a DMD patient lacking exon 44Excision of exon 45[48]
Cas9 mixed with 44C1, 44C2, 45C2, and 45C3 gRNAs produced via in vitro transcriptionElectroporationhDMD (Tg(DMD)72Thoen/J, 018900), C57BL/10 mdx (001801), and mdxD2 (D1.B10-Dmdmdx/J, 013141)Excision of exons 45–55[49]
AdV-Cas9-RFP
AdG-gRNA-Donor
Transfection reagent (lipid based)Skeletal muscle cell culture derived from C57BL/10ScSn-Dmd mdx/JExcision of exon 23[50]
Cpf1 gRNAs targeting the human DMD or the mouse Dmd locus (subcloned into pLbCpf1-2A-GFP and pAsCpf1-2A-GFP)NucleofectionDMD iPSC cellsExcision of exon 51[51]

Table 2.

Studies conducted on treatment of DMD using gene editing techniques (CRISPR/Cas9) (during 2017–2020).

2.4 Gene therapy

Gene therapy is one of the most appealing techniques that are used to deliver a normal copy of the DMD gene to express the fully functional dystrophin protein. This method implies injecting the patients with plasmids carrying normal dystrophin cDNA (~12 kb).

In 2002, the first phase 1 trial of DMD gene therapy took place using full-length dystrophin [52]. After that, adeno-associated viral vectors carrying mini forms of dystrophin cDNA were used for gene therapy, and this was better regarding the packaging size of the plasmids, and it is much easier to transfer/deliver mini forms of DMD gene [53, 54].

However, such therapeutic approach faced another problem which is the distribution of the plasmids across all affected muscular tissue that is spreading all over the body, and that is why microdystrophin plasmids and systemic AAV delivery were developed and improved to solve such problem. Evidence from many trials using animal models revealed that gene therapy can lead to long-term expression of functional protein [55, 56, 57].

In 2017, Le Guiner et al. studied the effect of the delivery of rAAV2/8 vector expressing a canine microdystrophin (cMD1) in golden retriever muscular dystrophy (GRMD) dogs in the absence of immunosuppression. Such treatment affected the deterioration of the muscular activity, and the gene expression was maintained over a long period [56]. Recently in 2020, Genthon and Sarepta contracted Yposkesi for manufacturing the AAV microdystrophin vector on a large scale.

2.5 Dystrophin-expressing chimeric cells

As previously mentioned, the absence of dystrophin is the main cause of DMD disease and the aggressive symptoms including muscle weakness and degeneration of muscle fibers. Such defect in the DMD gene can be edited using gene editing technology; however such technology can lead to off-target mutations which consequently can have damaging effects, and that is why more therapies had to be developed to address the disadvantages of such techniques.

Chimeric cell therapy is an alternative therapeutic approach that is usually done through the fusion of normal myoblasts and dystrophin-deficient myoblasts using polyethylene glycol (PEG). The success of this process could be tested using immunophenotyping (flow cytometry) and dystrophin immunostaining. This fusion will be followed by transplantation of chimeric cells in the defected muscle. The chimeric cells always show behavior like the donor cells regarding dystrophin expression and myogenic differentiation, and this dramatically improves the muscle function after being transplanted [58].

2.6 Cardiosphere-derived cells (CDCs)

Cardiosphere-derived cells are a type of cells that are retrieved from cardiac explants that can be cultured in vitro. Such cells have anti-inflammatory, antioxidant, and antibiotic activities. Several studies tested the ability of CDCs to alter the pathophysiology of DMD after the injection of these cells directly into the cardiac muscle.

Recently it was found that using CDCs greatly enhanced the phenotypic status of cardiac and skeletal muscles. The therapeutic effects of CDCs are usually attributed to the secretion of specific exosomes carrying specific genetic material to distal cells to exert its biological effect. Such CDCs along with their secreted exosomes can be injected intravenously, and it was found that they can greatly enhance the skeletal and cardiac muscle functions and boost the muscle generation process [59, 60].

2.7 Stop codon read-through therapy

In some of the mutations affecting the DMD gene, a premature stop codon is produced that can significantly disturb the reading frame and gives a truncated abnormal protein that cannot maintain the structural and functional properties of the muscle fibers.

A class of antibiotics called aminoglycosides was found to bind to rRNA at their decoding sites, preventing the stop codons from being read by binding to the A site (acceptor site) in the ribosomes and forcing the cell to prevent reading the stop codon, hence leading to the production of fully functional proteins.

PTC124 (ataluren; trade name, Translarna™) is one of the drugs with the read-through properties that are used for the treatment of DMD. Clinical trials showed that this drug when administered orally induced the expression of the dystrophin protein.

However, this treatment can only be used in ~15% of the cases who have a stop signal resulted from point mutation in the DMD gene. Also, it must be administered in increasing doses; beside it has many side effects such as renal toxicity. That is why there is a need to develop other alternatives with other structures to be safer with chronic usage.

2.8 Utrophin modulation

Recently, DMD symptoms were found to be managed after the administration of utrophin protein expression enhancers (utrophin is a dystrophin homolog; 395 KDa in size) to DMD patients delays the need of wheelchair and efficiently substitutes non-functional dystrophin.

Like dystrophin, the utrophin is present in the sarcolemma in the first developmental stages, and then replacement with dystrophin took place during muscular maturation. However, utrophin was found to be present in the myotendinous junction in adults. Interestingly, the expression of utrophin becomes elevated as a normal repair mechanism to compensate the absence of functional dystrophin in regenerated muscles.

SMTC-1100 is one of the chemical molecules that showed a great potential to increase the expression of DMD transcript and protein as well. This drug can be administered orally, if it was found to be safe and well tolerated in volunteers. However further studies are still required to detect if high dosage of this drug is safe or not.

Recently, ASA or adenylo-succinic acid improved the status of the TA muscles in mdx mice after administration of this compound in the drinking water. This molecule regulated the expression of the utrophin protein and hence greatly reduced the damaged area [61].

Another study group designed AAV vector carrying mini forms of the utrophin protein (μUtro). Their results showed that expression of this functional copy of utrophin protein (dystrophin analogue) after administration of the utrophin vector in DMD animal models completely reduced the muscle necrosis and regeneration [62].

Advertisement

3. Conclusion

Many medications have been used for DMD treatment and for preventing further deterioration of the cases. Corticosteroids were the first line of effective therapy of DMD; however, it does not modify the genetic mutations of the gene and does not affect the expression levels of dystrophin protein. Consequently, other treatments were developed including read-through stop codon, gene therapies, and exon skipping AOs which modulate and upregulate the levels of functional dystrophin transcript and protein in the muscles. Genome editing technology is also a powerful tool that can treat DMD permanently through the correction of the mutated sequence of DMD gene through the administration of sequence-specific guide RNA strands to bind selectively in the sequence to be edited. Also upregulating utrophin can help in the management of the cases. In addition, dystrophin-expressing chimeric cells and cardiosphere-derived cells are two emerging techniques that have the potential to treat DMD. Other medications will be developed to treat all DMD patients with different mutations with minimum side effects and maximum improvement in the status of the muscular system.

References

  1. 1. Hoffman EP, Brown RH Jr, Kunkel LM. Dystrophin: The protein product of the Duchenne muscular dystrophy locus. Cell. 1987;51(6):919-928
  2. 2. Gao QQ , McNally EM. The dystrophin complex: Structure, function, and implications for therapy. Comprehensive Physiology. 2015;5(3):1223-1239
  3. 3. Drachman DB, Toyka KV, Myer E. Prednisone in Duchenne muscular dystrophy. Lancet. 1974;2(7894):1409-1412
  4. 4. Angelini C, Pegoraro E, Turella E, Intino MT, Pini A, Costa C. Deflazacort in Duchenne dystrophy: Study of long-term effect. Muscle & Nerve. 1994;17(4):386-391
  5. 5. Bonifati MD, Ruzza G, Bonometto P, Berardinelli A, Gorni K, Orcesi S, et al. A multicenter, double-blind, randomized trial of deflazacort versus prednisone in Duchenne muscular dystrophy. Muscle & Nerve. 2000;23(9):1344-1347
  6. 6. Biggar WD, Harris VA, Eliasoph L, Alman B. Long-term benefits of deflazacort treatment for boys with Duchenne muscular dystrophy in their second decade. Neuromuscular Disorders. 2006;16(4):249-255
  7. 7. Biggar WD, Politano L, Harris VA, Passamano L, Vajsar J, Alman B, et al. Deflazacort in Duchenne muscular dystrophy: A comparison of two different protocols. Neuromuscular Disorders. 2004;14(8-9):476-482
  8. 8. St-Pierre SJ, Chakkalakal JV, Kolodziejczyk SM, Knudson JC, Jasmin BJ, Megeney LA. Glucocorticoid treatment alleviates dystrophic myofiber pathology by activation of the calcineurin/NF-AT pathway. The FASEB Journal. 2004;18(15):1937-1939
  9. 9. Jensen L, Petersson SJ, Illum NO, Laugaard-Jacobsen HC, Thelle T, Jorgensen LH, et al. Muscular response to the first three months of deflazacort treatment in boys with Duchenne muscular dystrophy. Journal of Musculoskeletal & Neuronal Interactions. 2017;17(2):8-18
  10. 10. McAdam LC, Mayo AL, Alman BA, Biggar WD. The Canadian experience with long-term deflazacort treatment in Duchenne muscular dystrophy. Acta Myologica. 2012;31(1):16-20
  11. 11. Lippuner K, Casez JP, Horber FF, Jaeger P. Effects of deflazacort versus prednisone on bone mass, body composition, and lipid profile: A randomized, double blind study in kidney transplant patients. The Journal of Clinical Endocrinology and Metabolism. 1998;83(11):3795-3802
  12. 12. Luhder F, Reichardt HM. Novel drug delivery systems tailored for improved administration of glucocorticoids. International Journal of Molecular Sciences. 2017;18(9):1836
  13. 13. Echevarria L, Aupy P, Goyenvalle A. Exon-skipping advances for Duchenne muscular dystrophy. Human Molecular Genetics. 2018;27(R2):R163-RR72
  14. 14. Aartsma-Rus A, Fokkema I, Verschuuren J, Ginjaar I, van Deutekom J, van Ommen GJ, et al. Theoretic applicability of antisense-mediated exon skipping for Duchenne muscular dystrophy mutations. Human Mutation. 2009;30(3):293-299
  15. 15. Charleston JS, Schnell FJ, Dworzak J, Donoghue C, Lewis S, Chen L, et al. Eteplirsen treatment for Duchenne muscular dystrophy: Exon skipping and dystrophin production. Neurology. 2018;90(24):e2146-e2e54
  16. 16. Goemans N, Mercuri E, Belousova E, Komaki H, Dubrovsky A, McDonald CM, et al. A randomized placebo-controlled phase 3 trial of an antisense oligonucleotide, drisapersen, in Duchenne muscular dystrophy. Neuromuscular Disorders. 2018;28(1):4-15
  17. 17. McDonald CM, Wong B, Flanigan KM, Wilson R, de Kimpe S, Lourbakos A, et al. Placebo-controlled phase 2 trial of drisapersen for Duchenne muscular dystrophy. Annals of Clinical Translational Neurology. 2018;5(8):913-926
  18. 18. Aartsma-Rus A, Kaman WE, Bremmer-Bout M, Janson AA, den Dunnen JT, van Ommen GJ, et al. Comparative analysis of antisense oligonucleotide analogs for targeted DMD exon 46 skipping in muscle cells. Gene Therapy. 2004;11(18):1391-1398
  19. 19. Lim KRQ , Echigoya Y, Nagata T, Kuraoka M, Kobayashi M, Aoki Y, et al. Efficacy of multi-exon skipping treatment in Duchenne muscular dystrophy dog model neonates. Molecular Therapy. 2019;27(1):76-86
  20. 20. Watanabe N, Nagata T, Satou Y, Masuda S, Saito T, Kitagawa H, et al. NS-065/NCNP-01: An antisense oligonucleotide for potential treatment of exon 53 skipping in Duchenne muscular dystrophy. Molecular Therapy—Nucleic Acids. 2018;13:442-449
  21. 21. Miyatake S, Mizobe Y, Takizawa H, Hara Y, Yokota T, Takeda S, et al. Exon skipping therapy using phosphorodiamidate morpholino oligomers in the mdx52 mouse model of Duchenne muscular dystrophy. Methods in Molecular Biology. 2018;1687:123-141
  22. 22. Komaki H, Nagata T, Saito T, Masuda S, Takeshita E, Sasaki M, et al. Systemic administration of the antisense oligonucleotide NS-065/NCNP-01 for skipping of exon 53 in patients with Duchenne muscular dystrophy. Science Translational Medicine. 2018;10(437)
  23. 23. Blain AM, Greally E, McClorey G, Manzano R, Betts CA, Godfrey C, et al. Peptide-conjugated phosphodiamidate oligomer-mediated exon skipping has benefits for cardiac function in mdx and Cmah-/-mdx mouse models of Duchenne muscular dystrophy. PLOS One. 2018;13(6):e0198897
  24. 24. Sako Y, Ninomiya K, Okuno Y, Toyomoto M, Nishida A, Koike Y, et al. Development of an orally available inhibitor of CLK1 for skipping a mutated dystrophin exon in Duchenne muscular dystrophy. Scientific Reports. 2017;7:46126
  25. 25. Echigoya Y, Lim KRQ , Trieu N, Bao B, Miskew Nichols B, Vila MC, et al. Quantitative antisense screening and optimization for exon 51 skipping in Duchenne muscular dystrophy. Molecular Therapy. 2017;25(11):2561-2572
  26. 26. Danisovic L, Culenova M, Csobonyeiova M. Induced pluripotent stem cells for Duchenne muscular dystrophy modeling and therapy. Cell. 2018;7(12):253
  27. 27. Miyoshi N, Ishii H, Nagano H, Haraguchi N, Dewi DL, Kano Y, et al. Reprogramming of mouse and human cells to pluripotency using mature microRNAs. Cell Stem Cell. 2011;8(6):633-638
  28. 28. Duchene B, Iyombe-Engembe JP, Rousseau J, Tremblay JP, Ouellet DL. From gRNA identification to the restoration of dystrophin expression: A dystrophin gene correction strategy for Duchenne muscular dystrophy mutations using the CRISPR-induced deletion method. Methods in Molecular Biology. 2018;1687:267-283
  29. 29. Min YL, Bassel-Duby R, Olson EN. CRISPR correction of Duchenne muscular dystrophy. Annual Review of Medicine. 2019;70:239-255
  30. 30. Adli M. The CRISPR tool kit for genome editing and beyond. Nature Communications. 2018;9(1):1911
  31. 31. Ousterout DG, Kabadi AM, Thakore PI, Perez-Pinera P, Brown MT, Majoros WH, et al. Correction of dystrophin expression in cells from Duchenne muscular dystrophy patients through genomic excision of exon 51 by zinc finger nucleases. Molecular Therapy. 2015;23(3):523-532
  32. 32. Young CS, Hicks MR, Ermolova NV, Nakano H, Jan M, Younesi S, et al. A single CRISPR-Cas9 deletion strategy that targets the majority of DMD patients restores dystrophin function in hiPSC-derived muscle cells. Cell Stem Cell. 2016;18(4):533-540
  33. 33. Duchene BL, Cherif K, Iyombe-Engembe JP, Guyon A, Rousseau J, Ouellet DL, et al. CRISPR-induced deletion with SaCas9 restores dystrophin expression in dystrophic models in vitro and in vivo. Molecular Therapy. 2018;26(11):2604-2616
  34. 34. Moretti A, Fonteyne L, Giesert F, Hoppmann P, Meier AB, Bozoglu T, et al. Somatic gene editing ameliorates skeletal and cardiac muscle failure in pig and human models of Duchenne muscular dystrophy. Nature Medicine. 2020;26(2):207-214
  35. 35. Nelson CE, Wu Y, Gemberling MP, Oliver ML, Waller MA, Bohning JD, et al. Long-term evaluation of AAV-CRISPR genome editing for Duchenne muscular dystrophy. Nature Medicine. 2019;25(3):427-432
  36. 36. Min YL, Li H, Rodriguez-Caycedo C, Mireault AA, Huang J, Shelton JM, et al. CRISPR-Cas9 corrects Duchenne muscular dystrophy exon 44 deletion mutations in mice and human cells. Science Advances. 2019;5(3):eaav4324
  37. 37. Amoasii L, Hildyard JCW, Li H, Sanchez-Ortiz E, Mireault A, Caballero D, et al. Gene editing restores dystrophin expression in a canine model of Duchenne muscular dystrophy. Science. 2018;362(6410):86-91
  38. 38. Lattanzi A, Duguez S, Moiani A, Izmiryan A, Barbon E, Martin S, et al. Correction of the exon 2 duplication in DMD myoblasts by a single CRISPR/Cas9 system. Molecular Therapy--Nucleic Acids. 2017;7:11-19
  39. 39. Yuan J, Ma Y, Huang T, Chen Y, Peng Y, Li B, et al. Genetic modulation of RNA splicing with a CRISPR-guided cytidine deaminase. Molecular Cell. 2018;72(2):380-394.e7
  40. 40. Ryu SM, Koo T, Kim K, Lim K, Baek G, Kim ST, et al. Adenine base editing in mouse embryos and an adult mouse model of Duchenne muscular dystrophy. Nature Biotechnology. 2018;36(6):536-539
  41. 41. Bengtsson NE, Hall JK, Odom GL, Phelps MP, Andrus CR, Hawkins RD, et al. Muscle-specific CRISPR/Cas9 dystrophin gene editing ameliorates pathophysiology in a mouse model for Duchenne muscular dystrophy. Nature Communications. 2017;8:14454
  42. 42. Maggio I, Liu J, Janssen JM, Chen X, Goncalves MA. Adenoviral vectors encoding CRISPR/Cas9 multiplexes rescue dystrophin synthesis in unselected populations of DMD muscle cells. Scientific Reports. 2016;6:37051
  43. 43. Koo T, Lu-Nguyen NB, Malerba A, Kim E, Kim D, Cappellari O, et al. Functional rescue of dystrophin deficiency in mice caused by frameshift mutations using Campylobacter jejuni Cas9. Molecular Therapy. 2018;26(6):1529-1538
  44. 44. Long C, Li H, Tiburcy M, Rodriguez-Caycedo C, Kyrychenko V, Zhou H, et al. Correction of diverse muscular dystrophy mutations in human engineered heart muscle by single-site genome editing. Science Advances. 2018;4(1):eaap9004
  45. 45. Amoasii L, Long C, Li H, Mireault AA, Shelton JM, Sanchez-Ortiz E, et al. Single-cut genome editing restores dystrophin expression in a new mouse model of muscular dystrophy. Science Translational Medicine. 2017;9(418):eaan8081
  46. 46. Lee K, Conboy M, Park HM, Jiang F, Kim HJ, Dewitt MA, et al. Nanoparticle delivery of Cas9 ribonucleoprotein and donor DNA in vivo induces homology-directed DNA repair. Nature Biomedical Engineering. 2017;1:889-901
  47. 47. Kyrychenko V, Kyrychenko S, Tiburcy M, Shelton JM, Long C, Schneider JW, et al. Functional correction of dystrophin actin binding domain mutations by genome editing. JCI Insight. 2017;2(18):e95918
  48. 48. Li HL, Fujimoto N, Sasakawa N, Shirai S, Ohkame T, Sakuma T, et al. Precise correction of the dystrophin gene in Duchenne muscular dystrophy patient induced pluripotent stem cells by TALEN and CRISPR-Cas9. Stem Cell Reports. 2015;4(1):143-154
  49. 49. Young CS, Mokhonova E, Quinonez M, Pyle AD, Spencer MJ. Creation of a novel humanized dystrophic mouse model of Duchenne muscular dystrophy and application of a CRISPR/Cas9 gene editing therapy. Journal of Neuromuscular Diseases. 2017;4(2):139-145
  50. 50. Zhu P, Wu F, Mosenson J, Zhang H, He TC, Wu WS. CRISPR/Cas9-mediated genome editing corrects dystrophin mutation in skeletal muscle stem cells in a mouse model of muscle dystrophy. Molecular Therapy--Nucleic Acids. 2017;7:31-41
  51. 51. Zhang Y, Long C, Li H, McAnally JR, Baskin KK, Shelton JM, et al. CRISPR-Cpf1 correction of muscular dystrophy mutations in human cardiomyocytes and mice. Science Advances. 2017;3(4):e1602814
  52. 52. DelloRusso C, Scott JM, Hartigan-O’Connor D, Salvatori G, Barjot C, Robinson AS, et al. Functional correction of adult mdx mouse muscle using gutted adenoviral vectors expressing full-length dystrophin. Proceedings of the National Academy of Sciences of the United States of America. 2002;99(20):12979-12984
  53. 53. Sakamoto M, Yuasa K, Yoshimura M, Yokota T, Ikemoto T, Suzuki M, et al. Micro-dystrophin cDNA ameliorates dystrophic phenotypes when introduced into mdx mice as a transgene. Biochemical and Biophysical Research Communications. 2002;293(4):1265-1272
  54. 54. Harper SQ , Hauser MA, DelloRusso C, Duan D, Crawford RW, Phelps SF, et al. Modular flexibility of dystrophin: Implications for gene therapy of Duchenne muscular dystrophy. Nature Medicine. 2002;8(3):253-261
  55. 55. Koo T, Okada T, Athanasopoulos T, Foster H, Takeda S, Dickson G. Long-term functional adeno-associated virus-microdystrophin expression in the dystrophic CXMDj dog. The Journal of Gene Medicine. 2011;13(9):497-506
  56. 56. Le Guiner C, Servais L, Montus M, Larcher T, Fraysse B, Moullec S, et al. Long-term microdystrophin gene therapy is effective in a canine model of Duchenne muscular dystrophy. Nature Communications. 2017;8:16105
  57. 57. Yoshimura M, Sakamoto M, Ikemoto M, Mochizuki Y, Yuasa K, Miyagoe-Suzuki Y, et al. AAV vector-mediated microdystrophin expression in a relatively small percentage of mdx myofibers improved the mdx phenotype. Molecular Therapy. 2004;10(5):821-828
  58. 58. Siemionow M, Cwykiel J, Heydemann A, Garcia-Martinez J, Siemionow K, Szilagyi E. Creation of dystrophin expressing chimeric cells of myoblast origin as a novel stem cell based therapy for Duchenne muscular dystrophy. Stem Cell Reviews and Reports. 2018;14(2):189-199
  59. 59. Rogers RG, Fournier M, Sanchez L, Ibrahim AG, Aminzadeh MA, Lewis MI, et al. Disease-modifying bioactivity of intravenous cardiosphere-derived cells and exosomes in mdx mice. JCI Insight. 2019;4(7):e125754
  60. 60. Aminzadeh MA, Rogers RG, Fournier M, Tobin RE, Guan X, Childers MK, et al. Exosome-mediated benefits of cell therapy in mouse and human models of Duchenne muscular dystrophy. Stem Cell Reports. 2018;10(3):942-955
  61. 61. Timpani CA, Goodman CA, Stathis CG, White JD, Mamchaoui K, Butler-Browne G, et al. Adenylosuccinic acid therapy ameliorates murine Duchenne muscular dystrophy. Scientific Reports. 2020;10(1):1125
  62. 62. Song Y, Morales L, Malik AS, Mead AF, Greer CD, Mitchell MA, et al. Non-immunogenic utrophin gene therapy for the treatment of muscular dystrophy animal models. Nature Medicine. 2019;25(10):1505-1511

Written By

Nahla O. Mousa, Ahmed Osman, Nagia Fahmy, Ahmed Abdellatif and Waheed K. Zahra

Submitted: 28 February 2020 Reviewed: 07 May 2020 Published: 13 June 2020