Dystrophinopathies cover a spectrum of X-linked muscle diseases caused by mutations in the dystrophin gene (OMIM 300377). Duchenne muscular dystrophy (DMD; OMIM 310200) is the most common form, which shows a severe phenotype clinically characterized by rapid progression in early childhood and loss of independent ambulation (LoA) by the age of 13 years; it affects 1 in 3600–6000 live male births[1]. The prevalence of DMD has been reported to be 1 in 4560 live male births in China[2]. Accordingly, about 1100 new cases are born every year in China. Milder allelic forms also exist, including intermediate muscular dystrophy (IMD) and Becker muscular dystrophy (BMD), which LoA typically at 13–16 years or over 16 years of age, respectively. Other types of dystrophinopathies occur at a lower incidence, including DMD and BMD manifesting in female carriers, isolated X-linked cardiomyopathy, and isolated quadriceps myopathy[3].
Pharmacological and care-based interventions (corticosteroid, rehabilitative, cardiac, orthopedic, respiratory, gastrointestinal, speech/swallowing, nutrition, and psychosocial interventions) can improve the muscle function, quality of life, and longevity of DMD patients[1]. Moreover, some therapeutic medicines have been used in clinical settings. In August 2014, ataluren, used to treat people with DMD who have a nonsense mutation in the dystrophin gene in ambulatory boys aged ≥ 5 years, received market authorization from the European Commission; it is currently sold under the trade name Translarna in the European Union. The outcome of a phase III trial of Ataluren (ACT DMD; ClinicalTrials.gov identifiers: NCT01826487) showed that the benefits observed in patients with a baseline 6 min walk distance of 300 m or more to less than 400 m supported the clinical benefit of ataluren versus a placebo in patients with nonsense mutation DMD, particularly when considering the totality of supporting evidence. The data also confirmed the clinical benefit of ataluren in terms of preserving muscle function[4]. However, the United States Food and Drug Administration (FDA) filed a Refuse to File letter on February 22, 2016, claiming that the results of the phase IIb study[5] and ACT DMD phase III trial[4] did not demonstrate adequate evidence of effectiveness. Two international multicenter clinical trials of ataluren continued to examine the long-term efficacy and safety of ataluren (ClinicalTrials.gov identifiers: NCT03179631 and NCT01247207); five hospitals, including our hospital in China, participated in these trials. In September 2016, the FDA conditionally approved the first phosphorodiamidate morpholino oligomer (morpholino)-based antisense oligonucleotide drug, eteplirsen, developed for DMD exon 51 skipping, based on the outcomes of a clinical trial[6]. Charleston et al.[7] examined the quantification of novel dystrophin production in patients with DMD after long-term treatment with eteplirsen; their results showed a sustained increase in dystrophin protein production and the presence of a novel in-frame mRNA gene product. Taken together, these results serve as evidence for the successful mechanism of eteplirsen and support the use of dystrophin as a pharmacodynamic marker, thus suggesting that the therapeutic approach is working as intended. Subsequent clinical trials on eteplirsen are active or recruiting (ClinicalTrials.gov identifiers: NCT03992430, NCT04179409, NCT03985878, and NCT03218995). Vyondys 53 (golodirsen) to treat patients with DMD amenable to exon 53 skipping was approved in the United States in December 2019, based on positive results from a phase I/II clinical trial[8]. Frank et al. reported on the safety, pharmacokinetics, exon 53 skipping, and dystrophin expression in golodirsen-treated patients with DMD amenable to exon 53 skipping (ClinicalTrials.gov identifiers: NCT02310906.). They found that golodirsen was well-tolerated. Muscle biopsies from golodirsen-treated patients have shown increased exon 53 skipping, dystrophin production, and correct dystrophin sarcolemmal localization[9]. In March 2020, another intravenous medicine, Viltepso (viltolarsen), received its first global approval in Japan for treating DMD patients, with confirmed deletion of the dystrophin gene that is amenable to exon 53 skipping. Viltolarsen is under regulatory review in the U.S., and clinical trials continue globally (e.g., U.S. and Canada)[10]. In February 2021, Casimersen was approved for medical use in the U.S. It is the first FDA-approved targeted treatment for people with a confirmed mutation of the DMD gene that is amenable to skipping exon 45. In addition, many promising therapeutic strategies are currently underway, providing hope for identifying definitive treatments for this currently incurable disease, as summarized below[11–14].
(1) Gene replacement with adeno-associated virus. Currently, two phase-III independent clinical trials of slightly different microdystrophins, viral vectors, and gene promoters are ongoing in patients with DMD (ClinicalTrials.gov, Identifier: NCT05096221; ClinicalTrials.gov, Identifier: NCT04281485; ClinicalTrials.gov, Identifier: NCT05096221).
(2) Mutation-specific therapeutic approaches for repairing the genetic defects or the related transcripts, such as exon-skipping agents (e.g., exon 51, exon 45, and exon 53 skipping agents) and stop codon readthrough agents as mentioned above.
(3) Genome editing. CRISPR-Cas9 technology can be used to induce double-stranded DNA breaks at specific places in the genome. These breaks activate DNA repair systems, which can lead either to homologous recombination, which corrects mutations or to non-homologous end joining, which leads to random insertions and/or deletions (that is, additional mutations).
(4) Symptomatic agents that target the major pathologic changes for alleviating symptoms, including agents used to prevent muscle damage, reduce inflammation, accelerate muscle repair, increase blood flow to muscles, and stop muscle fibrosis. Several phase III clinical trials are recruiting (ClinicalTrials.gov, Identifier: NCT04371666 and NCT04632940; ClinicalTrials.gov, Identifier: NCT04587908).
(5) Dystrophin surrogates. Upregulation or replacement of utrophin, a developmental paralog of dystrophin was the first surrogate to show promise as a DMD treatment. Dystrophin surrogates may have the greatest potential in combination with dystrophin correction or replacement strategies.
(6) Cell therapy. Dystrophin expression derived from human induced pluripotent stem cells (iPSCs) showed modest gains but led to functional improvement in the mdx mouse. Patient-derived iPSCs have yielded a theoretically unlimited supply of self-renewing, fetal stage, therapeutic myogenic stem cells, allowing for gene replacement or correction before engraftment. Moreover, because skeletal muscle is relatively inhospitable to tumor formation, it represents an attractive target tissue for PSC therapy.
As for BMD, clinical trials that aim to improve motor function and heart problems are currently in progress (ClinicalTrials.gov, Identifier: NCT05166109; ClinicalTrials.gov, Identifier: NCT03238235; ClinicalTrials.gov, Identifier: NCT05160415; and ClinicalTrials.gov, Identifier: NCT04386304).
Given these promising results, it becomes clear that the clinical trials for dystrophinopathy require international collaboration to establish sufficient patient enrollment for therapeutic significance to be demonstrated statistically. Based on this, we created a database for patients with dystrophinopathy at Children’s Hospital of Fudan University (referred to as the CHFU Database for Dystrophinopathy) in 2011. Over the past 10 years, 2097 patients with dystrophinopathy have been registered in our database. Thus, this database provides a general picture of the natural history and the current status of diagnosis and treatment of patients with dystrophinopathy in East China. Clearly, a database with a large number of patients with dystrophinopathy will be critical for international efforts to develop experimental therapies, facilitate trial design for new treatment strategies, and provide real-world data as external controls. Here, we describe the genetic diagnosis, clinical outcomes, and treatment status of the patients registered in our database.