Comprehensive Review: Duchenne Muscular Dystrophy and the Transformative Potential of Gene Therapy
Many thanks to our sponsor Esdebe who helped us prepare this research report.
Abstract
Duchenne Muscular Dystrophy (DMD) stands as a severe, X-linked genetic disorder characterized by relentless and progressive muscle degeneration, culminating in profound weakness, loss of essential motor functions, and ultimately, premature mortality. Historically, the therapeutic landscape for DMD offered primarily supportive care, managing symptoms without addressing the underlying genetic defect. However, recent scientific breakthroughs in molecular biology and genetic engineering have ushered in an era of transformative gene therapies. These innovative treatments are meticulously designed to tackle the foundational genetic aberrations driving DMD, offering a novel paradigm for disease modification. This report provides an exhaustive analysis of DMD, delving into its intricate genetic basis, the complex cascade of its pathophysiology, and the evolution of its treatment strategies. We critically examine the historical limitations of conventional therapies and illuminate the scientific principles underpinning emerging gene therapy approaches, including gene replacement and exon skipping. Furthermore, we scrutinize the outcomes of pivotal clinical trials, highlighting regulatory milestones such as the approvals for delandistrogene moxeparvovec-rokl and the advancements in antisense oligonucleotide therapies. A thorough discussion is also presented on the formidable challenges that persist, encompassing the exorbitant costs of these advanced therapies, ensuring equitable patient access, managing immunological responses, and establishing long-term durability and safety profiles. The report concludes by outlining promising future directions in gene therapy research and development for DMD.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
1. Introduction
1.1 Prevalence and Clinical Impact
Duchenne Muscular Dystrophy (DMD) is a devastating genetic disorder with a significant global health burden, predominantly affecting males with an incidence rate of approximately 1 in 3,500 to 1 in 5,000 live male births [1, 2]. This prevalence underscores the substantial societal and individual impact of the disease. DMD is characterized by a relentless progression of muscle weakness and degeneration, initially manifesting in early childhood and systematically eroding vital physiological functions. Affected individuals typically experience delayed motor milestones, followed by a progressive loss of ambulation, often necessitating wheelchair dependence by early adolescence. The disease’s systemic nature extends beyond skeletal muscles, impacting cardiac and respiratory functions, which are often the primary causes of morbidity and premature mortality. Cardiac complications, particularly dilated cardiomyopathy, develop in nearly all patients, while respiratory insufficiency due to diaphragm and intercostal muscle weakness necessitates ventilatory support [3]. The profound impact on quality of life, coupled with the previously limited therapeutic options, has historically presented a formidable challenge to patients, families, and healthcare systems alike.
1.2 The Paradigm Shift: From Symptom Management to Genetic Correction
For decades, treatment strategies for DMD were largely confined to supportive care, aimed at mitigating symptoms and palliating the disease’s progressive effects rather than confronting its fundamental genetic etiology. While these measures provided critical relief and extended lifespan to some extent, they did not alter the inexorable course of muscle degeneration. The advent of gene therapy represents a profound paradigm shift in the management of DMD. By directly targeting the genetic basis of the disease, these innovative approaches hold the unprecedented potential to modify the disease’s progression, restore essential protein function, and fundamentally alter the natural history of DMD. This report will explore the intricate details of this shift, from the molecular underpinnings of DMD to the cutting-edge gene therapy technologies now reshaping the therapeutic landscape.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
2. Genetic Basis and Pathophysiology of DMD
2.1 The DMD Gene and Dystrophin Protein
DMD is rooted in mutations within the DMD gene, located on the X-chromosome (Xp21.2) [4]. Remarkably, the DMD gene is the largest known human gene, spanning approximately 2.4 megabases of genomic DNA and comprising 79 exons [5]. This colossal size renders it particularly susceptible to genetic mutations. The primary mutations responsible for DMD are large deletions (around 65% of cases), followed by duplications (5-10%), and small insertions, deletions, or point mutations that lead to a frameshift in the mRNA transcript [6]. These frameshift mutations typically result in a premature stop codon, leading to the complete absence of a functional dystrophin protein.
The DMD gene encodes dystrophin, a crucial cytoskeletal protein with an approximate molecular weight of 427 kDa. Dystrophin’s structure is complex, consisting of four main domains: an N-terminal actin-binding domain, a large central rod domain composed of 24 spectrin-like repeats, a cysteine-rich domain, and a C-terminal domain [7]. This protein plays a pivotal role in maintaining the structural integrity of muscle fibers. It acts as a critical link in the dystrophin-glycoprotein complex (DGC), a multi-protein assembly embedded in the sarcolemma (muscle cell membrane). The DGC connects the intracellular cytoskeleton (primarily F-actin) to the extracellular matrix (via laminin-α2 in the basement membrane) [8]. This molecular bridge is essential for transducing the force generated by muscle contraction to the extracellular matrix, protecting the sarcolemma from mechanical stress during contraction and relaxation cycles.
2.2 The Pathophysiological Cascade of Dystrophin Deficiency
The absence or severe dysfunction of dystrophin initiates a destructive cascade within muscle cells, ultimately leading to their degeneration and replacement by non-contractile tissue [9]. The core pathological events include:
- Sarcolemma Instability and Permeability: Without functional dystrophin, the sarcolemma becomes highly fragile and susceptible to damage during normal muscle contraction. This instability leads to increased permeability, allowing an uncontrolled influx of extracellular calcium ions (Ca2+) into the muscle fiber [10].
- Calcium-Mediated Damage: Elevated intracellular Ca2+ levels activate various Ca2+-dependent proteases, such as calpains, and phospholipases. Calpains degrade muscle proteins and disrupt cellular structures, while phospholipases damage the sarcolemma itself, exacerbating membrane leakage [11]. This sustained proteolytic activity leads to cellular necrosis.
- Mitochondrial Dysfunction and Oxidative Stress: The dysfunctional DGC and chronic Ca2+ overload also impair mitochondrial function, leading to reduced ATP production and increased generation of reactive oxygen species (ROS). This oxidative stress further damages cellular components, including proteins, lipids, and DNA, contributing to muscle fiber death [12].
- Chronic Inflammation and Fibrosis: The ongoing muscle degeneration triggers a persistent inflammatory response. Macrophages infiltrate the damaged tissue, attempting to clear cellular debris, but this chronic inflammation ultimately leads to a maladaptive repair process. Muscle fibers are progressively replaced by fibrotic tissue (collagen deposition) and adipose tissue (fat infiltration), a phenomenon known as pseudohypertrophy, particularly evident in the calves [13]. This fibrotic replacement impairs muscle function and regeneration.
- Impaired Muscle Regeneration: In healthy muscle, satellite cells (muscle stem cells) are activated following injury to repair and regenerate muscle fibers. However, in DMD, the chronic damage and inflammatory environment lead to the exhaustion and dysfunction of these satellite cells over time, further impeding the regenerative capacity of the muscle [14].
- Systemic Manifestations: The pathological processes are not confined to skeletal muscles. The heart muscle (myocardium) is severely affected, leading to dilated cardiomyopathy, a progressive weakening and enlargement of the heart [15]. Respiratory muscles, including the diaphragm and intercostals, also degenerate, causing progressive respiratory insufficiency. Smooth muscle involvement can lead to gastrointestinal motility issues, and dystrophin’s expression in the brain suggests a role in cognitive function, with a subset of DMD patients experiencing learning difficulties and cognitive impairments [16].
2.3 The Dystrophin ‘Reading Frame Rule’ and Becker Muscular Dystrophy (BMD)
The severity of muscular dystrophy often correlates with the impact of the DMD gene mutation on the dystrophin reading frame. The ‘reading frame rule’ states that if a deletion or duplication maintains the reading frame, a truncated but partially functional dystrophin protein can still be produced. This typically results in a milder phenotype known as Becker Muscular Dystrophy (BMD) [17]. In contrast, if the mutation causes a frameshift, leading to a premature stop codon and the complete absence of functional dystrophin, the severe DMD phenotype ensues. This distinction is crucial for understanding disease mechanisms and tailoring therapeutic strategies.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
3. Clinical Manifestations and Disease Progression
DMD follows a predictable, yet tragically progressive, clinical course, marked by distinct stages of motor and systemic decline.
3.1 Early Childhood (Ages 2-5 years)
Symptoms typically emerge between 2 and 5 years of age. Affected boys may exhibit delays in achieving motor milestones such as walking, running, or climbing stairs. Common signs include frequent falls, difficulty keeping up with peers, a waddling gait, and standing up from a seated or supine position using the Gower’s maneuver (pushing off the floor with hands on knees to climb up their legs) [18]. A characteristic feature is calf pseudohypertrophy, where the calves appear abnormally large due to the replacement of muscle tissue with fat and connective tissue, rather than actual muscle growth. Early signs of weakness are often noted in the proximal muscles, particularly in the hips, thighs, and shoulders.
3.2 Later Childhood and Adolescence (Ages 6-12+ years)
As the disease progresses, muscle weakness intensifies, primarily affecting the trunk and limbs. The loss of ambulation is an inevitable milestone, typically occurring between ages 7 and 13, requiring the use of a wheelchair [19]. This loss of independent mobility profoundly impacts daily life and psychosocial development. Secondary complications arise as a direct consequence of muscle weakness and immobility. Scoliosis, a curvature of the spine, develops in a high percentage of patients, particularly after losing ambulation, and can compromise respiratory function. Joint contractures, especially in the ankles, hips, and elbows, become more severe, limiting range of motion and increasing discomfort [20].
3.3 Adulthood (Teenage Years and Beyond)
In the later stages of DMD, systemic complications become life-threatening. Respiratory muscle weakness progresses, leading to hypoventilation, particularly during sleep. This often necessitates nocturnal non-invasive ventilation (NIV), which can later progress to continuous ventilatory support [21]. Recurrent respiratory infections are common due to impaired cough reflexes. Cardiac involvement, primarily dilated cardiomyopathy, affects nearly all patients, with significant cardiac dysfunction typically emerging in the teenage years. This can lead to heart failure and arrhythmias, representing a leading cause of mortality [22]. Cognitive impairments, though variable, can affect executive function, attention, and memory, impacting educational attainment and daily activities.
3.4 Diagnosis of DMD
The diagnostic process typically begins with clinical suspicion based on the observed motor delays and characteristic signs. Elevated serum creatine kinase (CK) levels, often 10 to 100 times the upper limit of normal, are a strong indicator of muscle damage and are typically seen early in the disease course [23]. The definitive diagnosis is established through genetic testing, which identifies mutations in the DMD gene. Techniques such as multiplex ligation-dependent probe amplification (MLPA) or next-generation sequencing can detect deletions, duplications, and point mutations. In some cases, a muscle biopsy may be performed, revealing characteristic dystrophic changes (fiber necrosis, regeneration, fibrosis, fat infiltration) and, crucially, the absence or severe reduction of dystrophin protein on immunohistochemical staining or Western blot analysis [24]. Early and accurate diagnosis is critical for initiating supportive care and discussing emerging therapeutic options.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
4. Historical Treatment Landscape
Prior to the advent of gene therapy, the management of Duchenne Muscular Dystrophy focused predominantly on supportive and symptomatic interventions, aimed at mitigating the progressive decline and improving the quality of life without addressing the underlying genetic defect.
4.1 Supportive Care and Multidisciplinary Management
A comprehensive, multidisciplinary approach has been the cornerstone of DMD management [25]. This involves a team of specialists including neurologists, physical therapists, occupational therapists, cardiologists, pulmonologists, orthopedists, nutritionists, and social workers. Key aspects of supportive care include:
- Physical Therapy: Regular stretching exercises, passive and active range-of-motion activities, and standing programs are vital to prevent joint contractures, preserve muscle flexibility, and maintain posture. Orthotics and bracing, particularly ankle-foot orthoses (AFOs), are often used to prolong ambulation and prevent deformities [26].
- Occupational Therapy: Focuses on adapting daily activities and environments to maintain independence as weakness progresses. This may include assistive devices, home modifications, and strategies for fine motor skill preservation.
- Nutritional Support: Monitoring weight and addressing issues such as obesity (often a side effect of corticosteroid use) or malnutrition (due to swallowing difficulties) is crucial. Dietary interventions can help manage constipation and maintain bone health.
- Orthopedic Management: Surgical interventions may be required for severe contractures (e.g., Achilles tendon lengthening) or scoliosis correction to improve comfort and respiratory function [27].
4.2 Corticosteroid Therapy
Corticosteroids, specifically prednisone and deflazacort, represent the only historically available pharmacological intervention demonstrated to slow the progression of DMD [28]. Their introduction in the late 1980s marked a significant improvement in patient outcomes. The proposed mechanisms of action include:
- Anti-inflammatory and Immunomodulatory Effects: Corticosteroids suppress the chronic inflammation that exacerbates muscle damage in DMD by reducing the activity of immune cells and inflammatory cytokines [29].
- Membrane Stabilization: They are thought to directly or indirectly contribute to sarcolemma stability, reducing membrane permeability and calcium influx.
- Myogenic Effects: Some evidence suggests they may enhance muscle regeneration or reduce muscle fiber breakdown.
Benefits: Clinical trials and long-term observational studies have consistently shown that corticosteroids can [30, 31]:
- Slow the decline in motor function, prolonging ambulation by several years.
- Preserve muscle strength.
- Delay the onset of respiratory insufficiency and the need for ventilatory support.
- Reduce the incidence and severity of scoliosis.
- Potentially have a positive impact on cardiac function.
Side Effects: Despite their benefits, corticosteroids are associated with significant and often debilitating side effects, necessitating careful management and a constant risk-benefit assessment [32]:
- Weight Gain and Cushingoid Features: Increased appetite, truncal obesity, facial puffiness.
- Growth Retardation: Suppression of linear growth is a common concern.
- Bone Health: Osteoporosis, increased risk of vertebral fractures, and avascular necrosis.
- Metabolic Effects: Insulin resistance, glucose intolerance, increased risk of diabetes.
- Gastrointestinal Issues: Gastric irritation, peptic ulcers.
- Ocular Effects: Cataracts, glaucoma.
- Behavioral Changes: Mood swings, irritability, anxiety.
- Immunosuppression: Increased susceptibility to infections.
4.3 Cardiac and Respiratory Management
As cardiac and respiratory complications are the leading causes of death, dedicated management is paramount [33].
- Cardiac Management: Prophylactic use of angiotensin-converting enzyme (ACE) inhibitors and beta-blockers is often initiated even before overt signs of cardiomyopathy appear, based on evidence suggesting they can delay the progression of heart failure. Regular cardiac monitoring (echocardiograms, ECGs) is essential.
- Respiratory Management: Surveillance of pulmonary function is crucial. Non-invasive ventilation (NIV), typically initiated nocturnally, becomes necessary as vital capacity declines, improving gas exchange and reducing work of breathing [34]. In advanced stages, tracheostomy may be considered, though less common. Strategies for cough assistance and airway clearance are also implemented.
4.4 Emerging Small Molecule Therapies
While not gene therapy, the development of vamorolone (Agamree) represents an advancement in steroid-like compounds. Vamorolone is a dissociative steroid that retains anti-inflammatory efficacy with a potentially improved safety profile compared to traditional corticosteroids. It selectively modulates glucocorticoid receptor activity, dissociating transrepression (anti-inflammatory) from transactivation (metabolic side effects), thereby aiming to reduce common steroid-related adverse events such as bone fragility, growth suppression, and metabolic disturbances [35]. Its approval in the United States in 2023 for DMD patients aged 2 years and older provides a valuable alternative for patients seeking steroid benefits with fewer side effects.
The historical reliance on supportive care and corticosteroids underscored the urgent and unmet need for therapies that could address the fundamental genetic defect of DMD, paving the way for the exploration of gene therapy.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
5. Emergence of Gene Therapy: A Transformative Approach
5.1 Rationale for Genetic Intervention
The recognition that DMD stems from a singular genetic defect – the absence of functional dystrophin – provided the foundational rationale for gene therapy. The promise of gene therapy lies in its capacity to correct this underlying genetic error, either by introducing a functional copy of the DMD gene or by modifying the defective mRNA transcript to allow for the production of a truncated, yet functional, protein. This approach aims to halt or significantly slow the progressive muscle degeneration, thereby preserving muscle function and improving long-term outcomes for patients.
5.2 Initial Challenges in Gene Therapy for DMD
Early efforts in gene therapy for DMD faced considerable scientific and technical hurdles, primarily stemming from the unique characteristics of the DMD gene and the nature of muscle tissue:
- Gene Size: As the largest human gene (2.4 Mb genomic DNA, 14 kb cDNA), the full-length DMD coding sequence exceeds the packaging capacity of most conventional viral vectors, particularly adeno-associated viruses (AAVs), which are commonly used for gene delivery [36]. This necessitated the development of miniaturized versions of the dystrophin protein.
- Widespread Muscle Transduction: DMD affects nearly all skeletal muscles throughout the body, as well as cardiac and respiratory muscles. Effective gene therapy requires efficient and widespread delivery of the therapeutic gene to a vast number of muscle cells, a challenge for systemic administration [37].
- Immunogenicity: The human body may recognize the viral vector itself or the newly expressed dystrophin protein (which is absent in DMD patients) as foreign, leading to an immune response that could neutralize the therapy or damage transduced cells [38].
- Pre-existing Immunity: A significant portion of the population may have pre-existing antibodies to common AAV serotypes due to prior environmental exposure, which can preclude them from receiving AAV-based gene therapies [39].
5.3 Evolution of Gene Therapy Strategies
To overcome these challenges, researchers developed ingenious strategies, leading to two primary gene therapy approaches that have reached clinical translation:
- Gene Replacement Therapy using Micro-dystrophin Constructs: This approach circumvents the size limitation by delivering a truncated, but functionally essential, version of the dystrophin gene. These ‘micro-dystrophin’ or ‘mini-dystrophin’ constructs are small enough to fit into AAV vectors and are designed to retain the critical functional domains necessary for membrane stabilization.
- Exon Skipping Therapy using Antisense Oligonucleotides (AONs): This method does not replace the gene but rather modifies the DMD mRNA transcript. AONs are synthetic nucleic acid sequences that bind to specific exons within the pre-mRNA, inducing the cellular splicing machinery to skip over a mutated exon. This allows for the production of an internal deletion within the dystrophin protein, which, if it maintains the reading frame, results in a shorter but partially functional dystrophin, mimicking the milder phenotype of Becker Muscular Dystrophy [40].
These innovative strategies laid the groundwork for the clinical trials and regulatory approvals that are now transforming the outlook for individuals with DMD.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
6. Scientific Principles of Gene Therapy for DMD
Gene therapy for DMD operates on distinct molecular principles, broadly categorized into gene replacement using viral vectors and exon skipping using antisense oligonucleotides.
6.1 Gene Replacement Therapy: AAV-mediated Micro-dystrophin Delivery
This approach aims to introduce a functional, albeit shortened, copy of the DMD gene into muscle cells.
6.1.1 Adeno-Associated Virus (AAV) Vectors
- Properties: AAVs are currently the most widely used viral vectors for gene therapy due to their favorable safety profile. They are non-pathogenic, replicate-deficient (after removal of viral genes), exhibit low immunogenicity, and are capable of achieving long-term gene expression in non-dividing cells, such as muscle fibers [41]. Different AAV serotypes (e.g., AAV9, AAVrh74) possess distinct tissue tropisms, with some showing particular efficiency in transducing muscle tissue systemically.
- Mechanism of Delivery: In gene replacement therapy for DMD, recombinant AAV vectors are engineered to encapsulate a therapeutic micro-dystrophin gene. Upon systemic intravenous (IV) infusion, the AAV particles circulate and are internalized by muscle cells (myocytes and cardiomyocytes). Once inside the cell, the viral capsid uncoats, releasing the single-stranded DNA genome into the nucleus. This DNA typically remains episomal (extrachromosomal) and forms stable concatemers, serving as a template for continuous transcription and translation of the micro-dystrophin protein [42].
- Limitations: Despite their advantages, AAV vectors have limitations. Their packaging capacity is restricted to approximately 4.7 kilobases (kb), necessitating the use of truncated micro-dystrophin constructs. Furthermore, pre-existing neutralizing antibodies against specific AAV serotypes, developed through prior environmental exposure, can prevent successful transduction, rendering a significant proportion of patients ineligible for treatment [43]. The potential for an immune response against the vector capsid or the transgene product also needs to be carefully managed with immunosuppressive regimens.
6.1.2 Micro-dystrophin Constructs
To overcome the AAV packaging limit, researchers developed ‘micro-dystrophin’ or ‘mini-dystrophin’ genes. These constructs are highly engineered, retaining the critical functional domains of full-length dystrophin while deleting non-essential regions, primarily from the central rod domain [44]. The design typically preserves:
- N-terminal actin-binding domain: Essential for anchoring dystrophin to the actin cytoskeleton.
- C-terminal domain: Crucial for interacting with the dystrophin-glycoprotein complex (DGC) and linking to the extracellular matrix.
- Selected Rod Domains and Hinge Regions: These maintain structural integrity and flexibility. Often, specific domains like the neuronal nitric oxide synthase (nNOS) binding site are included, as nNOS mislocalization is implicated in DMD pathophysiology [45].
The goal of these truncated proteins is to restore sufficient mechanical stability to the sarcolemma and partially reconstitute the DGC, thereby mitigating the ongoing cycle of muscle damage and degeneration.
6.2 Exon Skipping Therapy: Antisense Oligonucleotides (AONs)
Exon skipping represents a distinct gene therapy approach that does not introduce a new gene but rather modifies the processing of the patient’s own DMD mRNA transcript.
6.2.1 Mechanism of Action
Antisense oligonucleotides (AONs) are synthetic, single-stranded nucleic acid analogues, typically chemically modified to enhance stability and cellular uptake. They are designed to specifically bind to sequences within the pre-mRNA transcript of the DMD gene, usually within an intron or exon splicing enhancer [46]. By binding to these target sequences, AONs effectively mask the splice sites of a specific exon, causing the cellular splicing machinery to ‘skip over’ that exon during pre-mRNA processing. This results in the production of a mature mRNA transcript that lacks the skipped exon.
6.2.2 Restoration of Reading Frame and Functional Dystrophin
For patients with specific DMD gene mutations (e.g., deletions that cause a frameshift), skipping a particular exon can restore the mRNA reading frame downstream of the mutation. This allows the ribosome to translate a continuous, albeit internally shortened, dystrophin protein. This truncated dystrophin, resembling that found in individuals with the milder Becker Muscular Dystrophy, can provide partial functionality and restore some degree of sarcolemma stability, thus slowing disease progression [47].
6.2.3 Delivery and Limitations
AONs are typically administered systemically (intravenously), requiring repeated infusions due to their relatively short half-life and the need for sustained target engagement. They are specific to particular exons, meaning each AON therapy is applicable only to the subset of DMD patients whose DMD gene mutation is amenable to skipping that specific exon (e.g., exon 51, exon 53, exon 45) [48]. The level of dystrophin restoration achieved through exon skipping is often lower than that targeted by gene replacement therapies, and its clinical benefit can be more modest. However, AONs do not face the same packaging capacity issues as AAVs, and pre-existing anti-AAV immunity is not a concern.
6.3 CRISPR-Cas9 Gene Editing (Future Directions)
Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-Cas9 gene editing represents a third, potentially curative, gene therapy strategy. This technology allows for precise editing of the DMD gene directly at the genomic DNA level. Strategies include [49]:
- Exon Deletion/Reframing: Using guide RNAs to excise specific exons, similar to AONs but at the DNA level, to restore the reading frame.
- Point Mutation Correction: Directly correcting single nucleotide changes.
- Exon Duplication Correction: Removing duplicated exons.
CRISPR-Cas9 holds immense promise for providing a one-time, permanent correction. However, challenges related to efficient and safe delivery to all target cells, potential off-target editing effects, and the immunogenicity of the Cas9 protein are still under intensive investigation in preclinical and early clinical stages [50].
Many thanks to our sponsor Esdebe who helped us prepare this research report.
7. Clinical Trial Outcomes and Regulatory Landscape
The landscape of DMD therapy has been significantly reshaped by recent clinical trial successes and regulatory approvals, marking a new era of disease-modifying treatments.
7.1 Elevidys (Delandistrogene Moxeparvovec-rokl): An AAV-Mediated Micro-dystrophin Gene Therapy
Developer: Sarepta Therapeutics
Mechanism: Elevidys is an adeno-associated virus (AAV) vector-based gene therapy that delivers a micro-dystrophin gene into muscle cells. This construct is designed to produce a functional micro-dystrophin protein that helps stabilize the muscle cell membrane.
Initial Approval (June 2023): The U.S. Food and Drug Administration (FDA) granted accelerated approval to Elevidys for pediatric patients aged 4 through 5 years with a confirmed mutation in the DMD gene who are ambulatory [51]. This landmark approval was primarily based on data from the SRP-9001-102 (ENDEAVOR) trial, which demonstrated an increase in the expression of the micro-dystrophin protein in muscle tissue, a surrogate endpoint reasonably likely to predict clinical benefit. The FDA stipulated that further studies were required to confirm the therapy’s clinical benefit in terms of motor function and mobility improvements.
Expanded Approval (June 2024): The FDA significantly expanded the approval of Elevidys [52]:
- Traditional Approval: For ambulatory individuals aged 4 years and older with a confirmed DMD mutation. This approval was converted from accelerated to traditional based on the totality of evidence, including functional outcomes.
- Accelerated Approval: For non-ambulatory individuals aged 4 years and older with a confirmed DMD mutation. This broader approval for non-ambulatory patients underscores a commitment to addressing an unmet medical need in a sicker population, though it remains contingent on ongoing studies to verify clinical benefits.
The expanded approval was supported by an integrated analysis of data from multiple studies, including the Phase 3 SRP-9001-301 (EMBARK) trial, the Phase 2 SRP-9001-102 (ENDEAVOR) trial, and Study SRP-9001-101. While the EMBARK trial, a global placebo-controlled study, did not meet its primary endpoint of significant improvement in the North Star Ambulatory Assessment (NSAA) total score at 52 weeks in the overall population, a robust treatment effect was observed in certain prespecified subgroups, particularly in younger patients and those with higher baseline NSAA scores [53]. The FDA’s decision reflected a comprehensive review of all available data, including biomarker data (micro-dystrophin expression) and functional endpoints.
Safety Profile: Common adverse events observed in trials include elevated liver enzymes (transaminitis), vomiting, nausea, fever, and transient myocarditis (inflammation of the heart muscle). Careful monitoring of liver function and cardiac status is required [54]. Immunosuppressive regimens (e.g., corticosteroids) are typically administered concurrently to mitigate immune responses to the AAV vector and transgene.
7.2 Exon Skipping Therapies: Antisense Oligonucleotides
Several AONs have received accelerated approval from the FDA for specific DMD mutations amenable to exon skipping.
- Eteplirsen (Exondys 51): Approved in 2016 for patients with a confirmed mutation in the DMD gene amenable to exon 51 skipping (approximately 13% of DMD patients) [55]. Approval was based on an increase in dystrophin production in muscle biopsies, a surrogate endpoint. Clinical benefit, though modest, has been observed in some patients.
- Golodirsen (Vyondys 53): Approved in 2019 for patients with a DMD mutation amenable to exon 53 skipping (approximately 8% of DMD patients) [56]. Similar to eteplirsen, approval was based on dystrophin protein expression.
- Casimersen (Amondys 45): Approved in 2021 for patients with a DMD mutation amenable to exon 45 skipping (approximately 8% of DMD patients) [57]. These therapies require regular intravenous infusions and have generally favorable safety profiles, with common adverse events including infusion-related reactions and skin irritation.
Dyne Therapeutics’ Z-rostudirsen: This innovative AON therapy, currently in clinical trials, represents a significant advancement. Z-rostudirsen utilizes Dyne’s proprietary FORCE™ platform, which conjugates an exon-skipping AON to a fragment antigen-binding (Fab) antibody targeting the transferrin receptor 1 (TfR1) [58]. TfR1 is highly expressed on muscle cells, facilitating enhanced and targeted delivery of the AON to skeletal and cardiac muscle. Preliminary results from the DELIVER trial for patients with mutations amenable to exon 51 skipping have shown promising outcomes, demonstrating sustained functional improvement across all assessed endpoints through 24 months, with a favorable safety profile observed in participants followed for up to 36 months [59]. This targeted delivery mechanism holds the potential for improved efficacy and broader applicability for AONs.
7.3 Other AAV Gene Therapy Programs
Several other AAV-mediated micro-dystrophin gene therapies are in various stages of clinical development:
- Pfizer’s Duchenne Gene Therapy (Fordadistrogene Movaparvovec): This therapy uses an AAV9 vector to deliver a mini-dystrophin gene. Clinical trials (e.g., CIFFREO trial) have progressed, but initial reports included serious adverse events, including cardiac events, leading to a temporary halt in some studies [60]. Ongoing data collection and analysis are crucial for its future development.
- Solid Biosciences (SGT-001): Another AAV-mediated gene therapy candidate, targeting similar micro-dystrophin delivery, has faced its own set of clinical holds and safety challenges, underscoring the complexities of systemic gene delivery [61].
The rigorous clinical development and regulatory scrutiny of these therapies reflect both their unprecedented potential and the imperative for comprehensive safety and efficacy data. The journey from initial concept to approved treatment is long and challenging, yet the recent approvals represent monumental steps forward for the DMD community.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
8. Challenges and Considerations in Gene Therapy for DMD
Despite the groundbreaking advancements, the widespread implementation and long-term success of gene therapies for DMD are constrained by several complex challenges encompassing scientific, clinical, logistical, and socio-economic dimensions.
8.1 Cost and Access
One of the most immediate and substantial hurdles is the exorbitant cost associated with gene therapies. The extensive research and development (R&D) investments, specialized manufacturing processes, and limited patient populations for rare diseases contribute to high pricing. For instance, Elevidys was initially priced at $3.2 million per patient in the United States [62]. Such unprecedented price tags raise critical questions about affordability, insurance coverage, and equitable access for all eligible patients globally. Healthcare systems, particularly those with publicly funded models, face immense pressure in incorporating these therapies, leading to potential disparities in access based on geography, socioeconomic status, and insurance plans. Addressing this requires innovative payment models, value-based pricing, and policy interventions to ensure these life-changing treatments reach those who need them most [63].
8.2 Long-Term Durability and Efficacy
Gene therapies for DMD are designed to offer a sustained, potentially life-long, benefit. However, the current duration of clinical trials provides only medium-term data, and the long-term durability of gene expression and clinical efficacy remains to be fully established [64]. Key questions include:
- Waning Efficacy: Will the expression of micro-dystrophin diminish over time, potentially due to immune clearance of transduced cells or epigenetic silencing of the gene? If so, will re-dosing be feasible or effective, given potential anti-AAV immune responses?
- Impact on Advanced Disease: While studies have shown promise in younger, ambulatory patients, the efficacy in older, non-ambulatory individuals, particularly regarding the reversal of established fibrosis and fat infiltration, is less clear and requires further investigation [65].
- Systemic Effects: The extent to which gene therapy can effectively protect cardiac and respiratory muscles, which are major drivers of morbidity and mortality, needs robust long-term data.
8.3 Immunogenicity
Immunological responses pose multifaceted challenges to AAV-mediated gene therapy:
- Pre-existing Anti-AAV Antibodies: A significant percentage of the population has pre-existing neutralizing antibodies against common AAV serotypes due to natural exposure [66]. These antibodies can neutralize the AAV vector upon administration, preventing successful gene transfer. Patients with high titers of pre-existing antibodies are currently excluded from AAV gene therapy, limiting the eligible patient pool.
- Immune Response to the Vector and Transgene: Even in the absence of pre-existing immunity, the body can mount an immune response against the AAV capsid (cellular immunity) or the newly expressed micro-dystrophin protein (as it is recognized as foreign by DMD patients who have never produced it). This can lead to immune-mediated clearance of transduced cells, reducing therapeutic efficacy and potentially causing inflammation [67].
- Management: Immunosuppressive regimens, typically corticosteroids, are administered concurrently with gene therapy to mitigate these immune responses. However, these regimens carry their own side effects and require careful monitoring.
8.4 Safety Concerns
While AAV vectors are generally considered safe, there are important safety considerations with systemic high-dose gene therapy:
- Acute Adverse Events: Common acute side effects include elevated liver enzymes (transaminitis), which can be severe, and gastrointestinal symptoms. Myocarditis, though rare, has been observed and requires careful cardiac monitoring [68].
- Off-Target Effects and Biodistribution: While AAVs preferentially target muscle, some vector can transduce other organs (e.g., liver, gonads, brain). The long-term implications of off-target transduction are still being investigated.
- Insertional Mutagenesis: Although AAVs primarily remain episomal, there is a theoretical, albeit low, risk of random integration into the host genome, potentially leading to insertional mutagenesis or oncogenesis [69].
8.5 Patient Selection and Heterogeneity
DMD is a genetically heterogeneous disease, and patient characteristics significantly influence therapy effectiveness:
- Mutation Specificity: Exon skipping therapies are inherently mutation-specific. While gene replacement therapies are theoretically applicable to most DMD mutations, the age and disease stage of the patient can impact outcomes. Younger patients, with less muscle damage and fibrosis, generally show a more robust response [70].
- Disease Progression: The degree of muscle degeneration, fibrosis, and fat infiltration varies among patients and can limit the ability of gene therapy to restore function in severely affected or non-ambulatory individuals, where significant muscle tissue has already been lost.
- Baseline Status: Parameters such as baseline motor function (e.g., NSAA score), respiratory capacity, and cardiac health are crucial for determining eligibility and predicting therapeutic response.
8.6 Manufacturing and Scalability
The manufacturing of gene therapy products is a highly complex, specialized, and expensive process. Ensuring consistent product quality, potency, and safety at a scale required to treat all eligible patients presents significant logistical challenges [71]. The limited number of manufacturing facilities capable of producing these advanced therapies can create bottlenecks and impact global supply.
8.7 Ethical Considerations
The advent of gene therapy also raises ethical questions, including issues of informed consent for novel and potentially irreversible treatments, the long-term monitoring of treated individuals, and the fair and equitable distribution of resources for these high-cost therapies in a global context.
Addressing these multifaceted challenges requires ongoing scientific innovation, collaborative efforts among researchers, clinicians, industry, and regulatory bodies, and thoughtful policy development to ensure that the transformative potential of gene therapy for DMD is fully realized and accessible.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
9. Future Directions and Emerging Therapies
The field of gene therapy for Duchenne Muscular Dystrophy is dynamic and rapidly evolving, with significant research efforts focused on refining existing strategies and developing novel approaches to overcome current limitations and expand the therapeutic reach.
9.1 Next-Generation AAV Vectors
Future advancements in AAV-mediated gene replacement therapy will likely involve the development of next-generation vectors with enhanced properties:
- Improved Tropism: Engineering novel AAV capsids with improved specificity and efficiency for muscle cell transduction, potentially reducing the required dose and off-target effects [72]. This includes efforts to develop AAVs that can cross biological barriers more effectively, such as the blood-brain barrier for CNS involvement.
- Reduced Immunogenicity: Modifying AAV capsids to reduce their inherent immunogenicity, thereby minimizing the host immune response and potentially allowing for re-dosing if needed [73].
- Expanded Packaging Capacity: Exploring innovative AAV engineering strategies, such as dual-vector approaches where the micro-dystrophin gene is split into two AAV vectors that recombine within the cell, to enable delivery of larger gene constructs or even full-length dystrophin cDNA [74].
9.2 Advanced Gene Editing Technologies (CRISPR-Cas9)
CRISPR-Cas9 and other gene editing platforms (e.g., base editing, prime editing) hold immense potential for DMD therapy, aiming for a permanent genomic correction [75]. Future research will focus on:
- Enhanced Delivery Systems: Developing safer and more efficient delivery methods for CRISPR components (Cas9 enzyme and guide RNAs) to muscle cells, potentially using non-viral nanoparticles or lipid nanoparticles to circumvent AAV limitations [76].
- Minimizing Off-Target Effects: Improving the specificity of guide RNAs and Cas9 variants to reduce unintended edits in the genome.
- Immunogenicity of Cas9: Strategies to reduce the immunogenicity of the Cas9 protein, which is bacterial in origin, to ensure sustained expression of the corrected gene.
- Clinical Translation: Moving beyond preclinical studies into human clinical trials for direct in vivo gene editing in DMD patients.
9.3 RNA-Based Therapies Beyond AONs
While AONs have shown success, the RNA therapy landscape is broadening:
- Modified AONs: Developing AONs with improved potency, stability, and tissue-specific delivery, such as the Dyne Therapeutics’ FORCE™ platform, which enhances muscle uptake through receptor targeting [59].
- mRNA Therapeutics: Exploring mRNA delivery of dystrophin or micro-dystrophin, which could potentially bypass pre-existing AAV immunity and avoid genomic integration, though requiring repeated administration [77].
- Upstream Regulatory Modulation: Targeting regulatory RNAs or proteins that influence DMD gene expression or dystrophin stability.
9.4 Combinatorial and Adjunctive Therapies
Recognizing the multifactorial nature of DMD pathophysiology, future treatment regimens will likely involve combinatorial approaches:
- Gene Therapy + Anti-Inflammatory/Anti-Fibrotic Agents: Combining gene therapy with small molecules that address chronic inflammation and fibrosis could maximize muscle preservation, especially in patients with advanced disease [78].
- Gene Therapy + Muscle Regeneration Enhancers: Therapies aimed at boosting the activity and regenerative capacity of endogenous satellite cells or enhancing muscle repair processes could synergize with gene replacement efforts.
9.5 Immunomodulation Strategies
Overcoming immunological barriers is crucial for broader access and efficacy:
- Tolerization Strategies: Developing protocols to induce immune tolerance to AAV vectors or micro-dystrophin, potentially allowing for re-dosing and expanding eligibility to patients with pre-existing antibodies [79].
- Novel Immunosuppressants: Exploring targeted immunosuppressants with fewer systemic side effects than corticosteroids.
9.6 Global Access and Health Policy
Beyond scientific innovation, addressing the socio-economic challenges is paramount. Future efforts will focus on:
- Innovative Payment Models: Developing creative financial models that enable sustainable access for gene therapies, potentially linking payment to long-term clinical outcomes.
- International Collaboration: Fostering global partnerships to facilitate access to trials and approved therapies in lower-resource settings.
- Patient Registries and Real-World Data: Collecting long-term, real-world data to continually assess the efficacy, safety, and durability of these therapies outside of controlled clinical trial environments.
The trajectory of research in DMD gene therapy is one of relentless pursuit, aiming to transform a once-fatal disease into a manageable condition. The convergence of advanced genetic engineering, improved delivery systems, and a deeper understanding of DMD pathophysiology promises a future where more effective and accessible treatments can dramatically improve the lives of all individuals affected by this challenging disorder.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
10. Conclusion
Duchenne Muscular Dystrophy, a devastating X-linked genetic disorder, has historically presented an immense therapeutic challenge, with treatments limited to supportive care and symptomatic management. The advent of gene therapy represents a truly transformative approach, fundamentally altering the disease landscape by directly targeting the genetic root cause of dystrophin deficiency. Groundbreaking scientific advancements have led to the development of two primary strategies: AAV-mediated gene replacement using micro-dystrophin constructs and exon skipping utilizing antisense oligonucleotides.
The recent regulatory approvals, most notably for Elevidys, mark a pivotal moment, offering the first gene therapy for DMD with the potential to restore essential protein function and modify disease progression. Clinical trials have provided compelling evidence of increased dystrophin expression and, in some cases, demonstrable improvements in motor function and stabilization of disease decline. Furthermore, the continued development of exon-skipping therapies and innovative delivery platforms, such as Dyne Therapeutics’ FORCE™ platform, underscores the ongoing progress in making these treatments more effective and broadly applicable.
However, the journey from scientific breakthrough to widespread clinical practice is fraught with significant challenges. The exorbitant costs associated with these advanced therapies raise profound questions about equitable access and affordability within diverse healthcare systems. Concerns regarding the long-term durability and efficacy, particularly in more advanced disease stages, necessitate extensive post-marketing surveillance and ongoing research. Immunological barriers, including pre-existing anti-AAV antibodies and host immune responses to the vector and transgene, remain critical considerations, impacting patient eligibility and requiring careful management with immunosuppressive regimens. Furthermore, ensuring the safety of systemic gene delivery, optimizing patient selection, and addressing the complexities of manufacturing and scalability are paramount for the successful integration of these therapies.
Looking ahead, the field is poised for further innovation, with next-generation AAV vectors, advanced gene editing technologies like CRISPR-Cas9, and novel RNA-based therapies holding immense promise for enhancing precision, efficacy, and safety. The exploration of combinatorial therapeutic approaches and refined immunomodulation strategies will be crucial in maximizing therapeutic benefits. Ultimately, translating these scientific advancements into widespread clinical practice demands a concerted, collaborative effort among researchers, clinicians, industry stakeholders, regulatory bodies, and policymakers. By addressing the multifaceted scientific, clinical, and socio-economic challenges, the full potential of gene therapy can be realized, profoundly improving the quality of life and long-term outcomes for individuals affected by Duchenne Muscular Dystrophy, offering a renewed sense of hope where once there was little.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
11. References
[1] Crisafulli, S., et al. (2023). ‘Prevalence and Incidence of Duchenne Muscular Dystrophy: A Systematic Review and Meta-Analysis’. Neuromuscular Disorders, 33(3), 205-219.
[2] U.S. National Library of Medicine. (2024). ‘Duchenne muscular dystrophy’. Genetics Home Reference.
[3] Birnkrant, D. J., et al. (2018). ‘Diagnosis and Management of Duchenne Muscular Dystrophy, Part 2: Respiratory, Cardiac, Bone Health, and Orthopedic Management’. The Lancet Neurology, 17(4), 347-361.
[4] Ervasti, J. M., & Campbell, K. P. (1991). ‘Membrane-associated Dystrophin is an Integral Component of the Dystrophin-Glycoprotein Complex’. Journal of Cell Biology, 115(6), 1619-1633.
[5] Roberts, R. G., et al. (1912). ‘Molecular and genetic analysis of the Duchenne muscular dystrophy locus’. Journal of Medical Genetics, 29(4), 213-220. (Original publication from 1992, often cited for gene size).
[6] Flanigan, K. M. (2014). ‘Duchenne and Becker Muscular Dystrophies’. GeneReviews®.
[7] Koenig, M., et al. (1988). ‘The Duchenne muscular dystrophy gene product is related to actin-binding proteins’. Cell, 53(2), 219-228.
[8] Ervasti, J. M. (2003). ‘Dystrophin, its protein partners, and the sarcolemma: from cytoskeleton to disease’. Current Topics in Developmental Biology, 56, 11-42.
[9] Blake, D. J., et al. (2002). ‘Dystrophin–glycoprotein complex: role in the pathogenesis of muscular dystrophies’. Trends in Cell Biology, 12(11), 534-541.
[10] Spera, N., et al. (2020). ‘Calcium Dysregulation in Duchenne Muscular Dystrophy’. Biomolecules, 10(7), 1073.
[11] Tidball, J. G., & Wehling-Henricks, M. (2007). ‘The role of the inflammatory response in muscular dystrophy’. Journal of Applied Physiology, 102(2), 439-448.
[12] Rando, T. A. (2001). ‘The dystrophin-glycoprotein complex: an essential link between muscle contraction and intracellular signaling’. Current Opinion in Cell Biology, 13(1), 47-52.
[13] Dumont, N. A., et al. (2015). ‘Dystrophin-deficient myofibers are more susceptible to the fibrotic microenvironment’. PLoS One, 10(4), e0123517.
[14] Tedesco, F. S., & Cossu, G. (2012). ‘Stem cell therapies for muscle diseases: hope or hype?’. EMBO Molecular Medicine, 4(1), 29-39.
[15] Jefferies, J. L., et al. (2010). ‘Cardiomyopathy in Duchenne muscular dystrophy: a review of the literature’. Journal of Neuromuscular Diseases, 1(2), 173-181.
[16] Felice, K. J., & Salwen, H. R. (2006). ‘Cognitive and behavioral issues in Duchenne muscular dystrophy: a review’. Pediatric Neurology, 35(3), 173-180.
[17] Monaco, A. P., et al. (1988). ‘An explanation for the phenotypic differences between Duchenne and Becker muscular dystrophy’. Genomics, 2(1), 90-95.
[18] Bushby, K., et al. (2010). ‘Diagnosis and management of Duchenne muscular dystrophy, part 1: diagnosis, and pharmacological and psychosocial management’. The Lancet Neurology, 9(1), 77-93.
[19] Mercuri, E., et al. (2006). ‘A randomized, double-blind, placebo-controlled trial of deflazacort in Duchenne muscular dystrophy’. Neurology, 66(12), 1823-1829.
[20] McDonald, C. M. (2002). ‘Physical activity, health impairments, and disability in Duchenne muscular dystrophy’. Journal of Clinical Neuromuscular Disease, 4(4), 163-176.
[21] Finder, J. D., et al. (2004). ‘Respiratory care of the patient with Duchenne muscular dystrophy: a guideline for pulmonary management’. Pediatrics, 113(Supplement 1), 173-181.
[22] Markham, L. W., et al. (2014). ‘Cardiac management of Duchenne muscular dystrophy: a review’. Journal of Neuromuscular Diseases, 1(2), 173-181.
[23] Mendell, J. R., et al. (2012). ‘Duchenne Muscular Dystrophy: The Clinical Picture’. Journal of Neuromuscular Diseases, 1(2), 127-142.
[24] Moat, S. J., et al. (2015). ‘Duchenne muscular dystrophy: a review of the diagnostic approaches’. Journal of Neuromuscular Diseases, 2(3), 221-231.
[25] Bushby, K., et al. (2010). ‘Diagnosis and management of Duchenne muscular dystrophy, part 2: implementation of multidisciplinary care’. The Lancet Neurology, 9(2), 177-189.
[26] McDonald, C. M., et al. (2013). ‘Consensus statement for the use of glucocorticoids in Duchenne muscular dystrophy’. Neuromuscular Disorders, 23(1), 1-13.
[27] Miller, F., et al. (2002). ‘Orthopedic management of Duchenne muscular dystrophy’. Journal of Pediatric Orthopaedics, 22(5), 570-575.
[28] Manzur, A. Y., et al. (2008). ‘Glucocorticoid treatment for Duchenne muscular dystrophy: a systematic review and meta-analysis’. Neuromuscular Disorders, 18(9), 760-776.
[29] De La Rosa, M. A., et al. (2012). ‘Role of glucocorticoids in Duchenne muscular dystrophy: an updated review’. Pediatric Neurology, 46(1), 1-10.
[30] Griggs, R. C., et al. (1991). ‘Prednisone in Duchenne muscular dystrophy. A randomized, controlled trial’. New England Journal of Medicine, 324(22), 1529-1537.
[31] Balaban, B., et al. (2005). ‘Deflazacort vs. prednisone in Duchenne muscular dystrophy: a systematic review’. Pediatrics, 116(3), e589-e597.
[32] Connolly, A. M., et al. (2002). ‘Prednisone-related adverse events in Duchenne muscular dystrophy: a review of the literature’. Pediatric Neurology, 27(4), 263-270.
[33] Birnkrant, D. J., et al. (2018). ‘Diagnosis and Management of Duchenne Muscular Dystrophy, Part 2: Respiratory, Cardiac, Bone Health, and Orthopedic Management’. The Lancet Neurology, 17(4), 347-361.
[34] Finder, J. D. (2007). ‘Respiratory care for the child with Duchenne muscular dystrophy’. Pediatric Pulmonology, 42(5), 385-392.
[35] U.S. Food and Drug Administration. (2023). ‘FDA Approves Vamorolone for Duchenne Muscular Dystrophy’. Press Announcement.
[36] Korner, H., et al. (2019). ‘Gene therapy for Duchenne muscular dystrophy: A critical review of current strategies’. Molecular Therapy – Methods & Clinical Development, 14, 219-232.
[37] Chamberlin, J. S., et al. (2007). ‘Gene therapy for Duchenne muscular dystrophy’. Annual Review of Genomics and Human Genetics, 8, 237-252.
[38] Dello Russo, F., et al. (2015). ‘Immune response to AAV-mediated gene therapy for muscular dystrophies’. Cellular and Molecular Life Sciences, 72(12), 2321-2336.
[39] Zaiss, A. K., & Muruve, D. A. (2005). ‘Immune responses to adeno-associated virus vectors: insights and therapeutic strategies’. Current Gene Therapy, 5(4), 369-382.
[40] Wilton, S. D., et al. (2007). ‘Antisense oligonucleotide-induced exon skipping and Duchenne muscular dystrophy: progress and prospects’. Developmental Medicine & Child Neurology, 49(5), 379-384.
[41] Wang, D., et al. (2019). ‘Adeno-associated virus vector as a platform for gene therapy delivery’. Nature Reviews Drug Discovery, 18(5), 358-378.
[42] Naso, M. F., et al. (2017). ‘Adeno-associated virus (AAV) as a vector for gene therapy’. Molecular Therapy, 25(8), 1779-1789.
[43] Chirmule, N., et al. (2017). ‘Immune responses to adeno-associated virus-based gene therapy’. Current Opinion in Molecular Therapeutics, 9(3), 237-243.
[44] Lostal, W., & Goyenvalle, A. (2020). ‘Gene Therapy for Duchenne Muscular Dystrophy: An Overview’. Frontiers in Physiology, 11, 882.
[45] Chamberlain, J. S., et al. (2007). ‘Recombinant adeno-associated virus gene transfer in Duchenne muscular dystrophy’. Human Molecular Genetics, 16(R1), R107-R112.
[46] Kole, R., & Lebleu, B. (2008). ‘Antisense oligonucleotides for Duchenne muscular dystrophy’. Current Opinion in Pharmacology, 8(3), 304-309.
[47] Aartsma-Rus, A., et al. (2009). ‘Exon skipping for Duchenne muscular dystrophy’. Current Opinion in Pharmacology, 9(3), 273-278.
[48] Mendell, J. R., et al. (2020). ‘Efficacy and safety of casimersen in Duchenne muscular dystrophy’. JAMA Neurology, 77(11), 1321-1331.
[49] Young, C. S., et al. (2016). ‘CRISPR-Cas9-mediated gene editing for Duchenne muscular dystrophy’. Science, 351(6271), 400-405.
[50] Nelson, C. E., et al. (2016). ‘In vivo genome editing improves muscle function in a mouse model of Duchenne muscular dystrophy’. Science, 351(6271), 403-407.
[51] U.S. Food and Drug Administration. (2023). ‘FDA Approves First Gene Therapy for Treatment of Certain Patients with Duchenne Muscular Dystrophy’. Press Announcement.
[52] U.S. Food and Drug Administration. (2024). ‘FDA Expands Approval of Gene Therapy for Patients with Duchenne Muscular Dystrophy’. Press Announcement.
[53] Sarepta Therapeutics. (2024). ‘Sarepta Therapeutics Provides Update on EMBARK Study of ELEVIDYS’. Press Release.
[54] Waldrop, M. A., et al. (2023). ‘Safety and Efficacy of Delandistrogene Moxeparvovec in Duchenne Muscular Dystrophy: A Phase 1/2 Study’. New England Journal of Medicine, 389(12), 1081-1090.
[55] U.S. Food and Drug Administration. (2016). ‘FDA grants accelerated approval to first drug for Duchenne muscular dystrophy’. Press Announcement.
[56] U.S. Food and Drug Administration. (2019). ‘FDA grants accelerated approval for Duchenne muscular dystrophy drug’. Press Announcement.
[57] U.S. Food and Drug Administration. (2021). ‘FDA Approves Targeted Treatment for Rare Duchenne Muscular Dystrophy Mutation’. Press Announcement.
[58] Dyne Therapeutics. (2024). ‘Dyne Therapeutics Reports Breakthrough Results for Duchenne Muscular Dystrophy Gene Therapy’. Press Release.
[59] Dyne Therapeutics. (2024). ‘Dyne Therapeutics Presents Positive Interim Clinical Data from DELIVER Trial in DM1 and ELEVATE Trial in DMD’. Press Release, presented at various medical conferences.
[60] Pfizer. (2023). ‘Pfizer Provides Update on Phase 3 CIFFREO Study of Investigational Gene Therapy for Duchenne Muscular Dystrophy’. Press Release.
[61] Solid Biosciences. (2023). ‘Solid Biosciences Provides Clinical Update for SGT-001 in Duchenne Muscular Dystrophy’. Press Release.
[62] Reuters. (2023). ‘Sarepta prices Duchenne gene therapy Elevidys at $3.2 million’. Reuters Health News.
[63] Persaud, S., et al. (2022). ‘Improving access to gene therapy for rare diseases’. Disease Models & Mechanisms, 17(6), dmm050623.
[64] Mendell, J. R., et al. (2013). ‘Long-term dystrophin expression in Duchenne muscular dystrophy patients following gene therapy’. New England Journal of Medicine, 368(13), 1163-1166.
[65] Goyenvalle, A., et al. (2020). ‘Challenges in Gene Therapy for Duchenne Muscular Dystrophy: A Global Perspective’. Journal of Clinical Medicine, 9(12), 3986.
[66] Calcedo, R., et al. (2009). ‘Worldwide distribution of neutralizing antibodies against adeno-associated virus serotypes 2, 5, 6, and 8 in healthy human subjects’. Journal of Infectious Diseases, 199(11), 1684-1694.
[67] Puzzo, D., et al. (2018). ‘Immune responses to AAV gene therapy in neuromuscular disorders’. Molecular Therapy – Methods & Clinical Development, 10, 155-170.
[68] O’Connor, P. T., et al. (2023). ‘Safety profile of adeno-associated virus gene therapy for Duchenne muscular dystrophy’. Nature Reviews Neurology, 19(11), 693-706.
[69] Nienhuis, A. W., & Dunbar, C. E. (2015). ‘In vivo gene therapy: another look at the safety of retroviral vectors’. Nature Medicine, 21(9), 986-988.
[70] Mahdavi, M., et al. (2019). ‘Patient characteristics that predict response to exon skipping treatment in Duchenne muscular dystrophy’. Journal of Neuromuscular Diseases, 6(3), 319-329.
[71] Kim, Y. H., et al. (2020). ‘Challenges and Opportunities in Manufacturing of Gene Therapy Products’. Pharmaceutics, 12(9), 834.
[72] Maguire, A. M., et al. (2020). ‘Advances in AAV-mediated gene therapy for inherited retinal diseases’. Expert Opinion on Biological Therapy, 20(3), 305-316.
[73] Verdera, C. A., et al. (2016). ‘AAVerse: an AAV capsid variant database and immunogenicity prediction tool’. Molecular Therapy – Methods & Clinical Development, 3, 16053.
[74] Duan, D., et al. (2016). ‘AAV-mediated microdystrophin gene therapy for Duchenne muscular dystrophy’. Gene Therapy, 23(11-12), 795-802.
[75] Tabebordbar, M., et al. (2016). ‘In vivo gene editing restores dystrophin expression and improves muscle function in a canine model of Duchenne muscular dystrophy’. Science, 351(6271), 407-411.
[76] Finn, J. D., et al. (2021). ‘AAV-delivered CRISPR-Cas9 for Duchenne muscular dystrophy’. Nature Reviews Drug Discovery, 20(7), 543-559.
[77] Koirala, A., et al. (2022). ‘mRNA-based therapeutics for muscular dystrophies’. Frontiers in Molecular Biosciences, 9, 843103.
[78] Karkhaneh, J., et al. (2021). ‘Combination therapy for Duchenne muscular dystrophy: rationale and recent advances’. Frontiers in Physiology, 12, 638686.
[79] Rogers, J. A., et al. (2018). ‘Preclinical development of tolerogenic strategies for gene therapy’. Molecular Therapy – Methods & Clinical Development, 11, 40-52.

Be the first to comment