Acta Scientific Medical Sciences

Review Article Volume 8 Issue 9

A Review on Duchenne Muscular Dystrophy - Market Analysis, Existing Products, Clinical Trials, and Preclinical Tissue Engineering Treatments

Carol Jarod and Tawil Bill*

Department of Bioengineering, University of California, Los Angeles (UCLA), USA

*Corresponding Author: Tawil Bill, Department of Bioengineering, University of California, Los Angeles (UCLA), USA.

Received: July 03, 2024; Published: August 28, 2024

Citation: Carol Jarod and Tawil Bill. “A Review on Duchenne Muscular Dystrophy - Market Analysis, Existing Products, Clinical Trials, and Preclinical Tissue Engineering Treatments”. Acta Scientific Medical Sciences 8.9 (2024): 125-141.

Abstract

Duchenne Muscular Dystrophy (DMD) is a severe genetic disorder characterized by progressive muscle degeneration and weakness, primarily affecting young males. Despite extensive research, no cure currently exists for DMD. With a large and expanding DMD treatment market, we expect new therapeutics to take over existing gold standards of therapy and transform patients’ prognoses. Existing therapies such as glucocorticoids, physical therapy, cardiac management, and respiratory therapy focus on symptom management and slowing disease progression. Still, there remains a high demand for long-term, effective treatments. Emerging DMD therapies have shown promising results, including gene therapy, exon skipping, and cell transplantation. Additionally, preclinical research in tissue engineering offers the potential for integrating therapeutic cells and bioactive molecules into DMD treatment. However, this field still has many challenges. This review provides an in-depth examination of DMD, including disease pathology, market size and trends, current therapeutic options, and products in clinical trials. We also explore innovative tissue engineering treatments in preclinical stages.

Keywords: Duchenne Muscular Dystrophy; Dystrophin; Satellite Cells; Gene Transfer; Exon Skipping; Cell Transplantation; Tissue Engineering

Abbreviations

Duchenne Muscular Dystrophy (DMD), extracellular matrix (ECM), dystrophin-associated protein complex (DAPC), satellite cell (SC), vascular endothelial growth factor (VEGF), compound annual growth rate (CAGR), antisense oligonucleotides (AONs), adenoassociated virus serotype-9 (AAV9), European Medicines Agency (EMA), Dystrophin Expressing Chimeric (DEC), decellularized extracellular matrix (dECM), hyaluronic acid-photoinitiator (HA-PI), poly(ethylene oxide) (PEO), polycaprolactone (PCL), poly-L-lactic acid (PLLA), insulin-like growth factor 1 (IGF-1), polyethylene glycol (PEG), green fluorescent protein (GFP), muscle progenitor cells (MPCs), hepatocyte growth factor (HGF), polyethylene glycol-fibrinogen (PF), poly(lactic-co-glycolic acid) (PLGA), gelatin–poly(ethylene glycol)–tyramine (GPT), human adipose-derived stem cells (h-ADSCs), basic fibroblast growth factor (bFGF), gingival mesenchymal stem cells (GMSCs)

Introduction

Muscular dystrophy encompasses a group of genetic muscle diseases [1]. In particular, Duchenne Muscular Dystrophy (DMD) is a prevalent muscular disease, which occurs during early childhood and is one of the most severe forms of inherited muscular dystrophy [1]. DMD is caused by an X-linked recessive gene mutation, found to primarily affect males [1]. DMD has an incidence rate of approximately 1 in 3500 male infants, and is a leading genetic killer in young boys worldwide [2]. DMD is characterized by progressive striated muscle degeneration and weakness, primarily in the skeletal and cardiac muscle [3,4]. This disease is due to a mutation in the dystrophin gene, leading to inadequate and faulty dystrophin protein in the patient [3,4]. Initially, affected patients develop weakness and atrophy in the proximal trunk muscles, including upper arms and legs [5]. As the DMD symptoms progress, they spread to the lower legs, forearms, neck, and trunk [5]. Additional secondary complications commonly arise as the disease progresses, including scoliosis, abnormal joint fixations, deformity, cardiomyopathy, respiratory infections, and dysmotility in the gastrointestinal system [5].

Currently, no direct cure for DMD exists [6]. Instead, the available therapeutic options act to alleviate and control symptoms, slow disease progression, and stabilize associated conditions [6]. Therefore, the market demand for long-term treatment for young patients struggling with DMD is prevalent [7]. Of particular interest, in addition to the expanding application of treatments such as gene therapy, exon skipping, and cell transplantation, increasing preclinical research is being supported for the designing of tissue engineering products to integrate cells and bioactive molecules into the patient for therapeutic applications [7]. In this review, we will compare healthy to diseased muscle tissue, analyze the current DMD market size and trends, investigate the existing therapeutics on the market and the products within clinical trials, and look into some of the novel tissue engineering treatments in the preclinical stage.

Healthy vs diseased muscle tissue
Healthy muscle tissue

Healthy striated muscles, mainly skeletal and cardiac, consist of highly organized muscle tissue made up of fascicles [8]. Fascicles comprise bundles of myofibers (muscle cells) that enable contraction, force generation, and movement [8]. Each myofiber contains sarcomeres, the basic muscle functional units that consist of thin actin filaments and thick myosin filaments responsible for generating contractile force [8]. The bundles of myofibrils are surrounded by an extracellular matrix (ECM) and supported by a cytoskeleton, which acts as a scaffold for sarcomeres, contributing to muscle plasticity and preventing damage from repeated contraction and relaxation [8].

Dystrophin, a substantial protein, plays a pivotal role in maintaining the synchrony of muscle stretching and contraction [8]. It achieves this by anchoring the sarcomere to the ECM [8]. Dystrophin is a key component of the dystrophin-associated protein complex (DAPC), which links cytoskeletal proteins and sarcomere to the ECM, forming the structural unit of muscle tissue (Figure 1) [8,9]. In healthy muscle tissue, the DAPC stabilizes the plasma membrane of striated muscle cells during contraction, preventing contractile-induced damage [8-10].

Figure 1: Diagram of the dystrophin-associated protein complex
and the main cytoskeletal proteins of healthy skeletal muscle
[9]. <br> 
The image shows dystrophin as a key component of the
dystrophin-associated protein complex (DAPC), which links
cytoskeletal proteins and sarcomere to the ECM, forming the
structural unit of muscle tissue.

Figure 1: Diagram of the dystrophin-associated protein complex and the main cytoskeletal proteins of healthy skeletal muscle [9].
The image shows dystrophin as a key component of the dystrophin-associated protein complex (DAPC), which links cytoskeletal proteins and sarcomere to the ECM, forming the structural unit of muscle tissue.

Healthy muscle tissue also has the ability to regenerate due to interactions between the adult muscle stem cells, or satellite cells, and the surrounding microenvironment [8]. Satellite cells remain dormant until an injury occurs [8]. Upon muscle injury, chemoattractant growth factors, such as VEGF, fibroblast growth factors, and insulin growth factors, are released by damaged resident cells and inflammatory cells [8]. These growth factors activate the satellite cells, promoting them to migrate to the injury site, where they proliferate and differentiate into muscle fibers for muscle regeneration [8].

Diseased duchenne muscular dystrophy tissue

In DMD patients, mutations in the dystrophin gene result in a complete or near-complete lack of dystrophin protein production [11]. Without functional dystrophin protein, the DAPC is not properly formed, leading to a compromised myofiber membrane [11]. This results in a fragile and permeable membrane, disruption of calcium homeostasis, immune cell infiltration, and oxidative damage [11]. These issues increase the muscle tissue’s susceptibility to damage, leading to repeated cycles of muscle cell death and regeneration [11]. Over time, the regenerative capacity of muscle satellite cells in DMD patients becomes exhausted, and fibrous connective tissue and adipose tissue begin to replace muscle fibers as the disease progresses [11]. Figure 2 displays a side-by-side comparison of healthy and DMD muscle tissue [12].

Figure 2: Healthy muscle tissue vs Duchenne Muscle Tissue
[12]. <br>
The image shows a visual comparing healthy muscle tissue and
Duchenne Muscular Dystrophy tissue.

Figure 2: Healthy muscle tissue vs Duchenne Muscle Tissue [12].
The image shows a visual comparing healthy muscle tissue and Duchenne Muscular Dystrophy tissue.

Market size, products, and companies

DMD market size and trends

DMD is one of the most common and severe hereditary neuromuscular diseases, impacting approximately 1 in 3500 male infants worldwide [5]. Approximately 250,000 individuals are affected by muscular dystrophy in the United States [2,5]. DMD patients are primarily diagnosed between 3-5 years old and have a median expected lifespan of 22 years [13]. Figure 3 demonstrates a Kaplan-Meier survival curve for DMD patients, illustrating survival probabilities of 99.8%, 59.5%, 26.1%, and 13.3% at 10, 20, 30, and 40 years, respectively [13].

Figure 3: Kaplan-Meier Survival Curve for DMD patients from
14 pooled studies [12]. <br> 
The image shows a Kaplan-Meier survival curve for DMD
patients of age (years) vs probability of survival, illustrating
survival probabilities of 99.8%, 59.5%, 26.1%, and 13.3% at 10,
20, 30, and 40 years, respectively.

Figure 3: Kaplan-Meier Survival Curve for DMD patients from 14 pooled studies [12].
The image shows a Kaplan-Meier survival curve for DMD patients of age (years) vs probability of survival, illustrating survival probabilities of 99.8%, 59.5%, 26.1%, and 13.3% at 10, 20, 30, and 40 years, respectively.

As DMD patients age, more and worse symptoms and secondary issues present, such as cardiac and respiratory failure, thus increasing the cost of healthcare expenditures [14]. According to a 2014 study by Landfeldt., et al. the average annual direct cost of illness per patient ranged from $23,920 to $54,270 (in 2012 international dollars) [15]. The total societal burden per patient was estimated to be between $80,120 and $120,910 [15]. As seen in Figure 4a, there is an increasing trend in the per-patient annual cost as the patient’s ambulatory state progressively worsens [15]. This is similarly depicted in Figure 4b as we also see an increase in the annual cost per patient as they age, comparing 0-13 years of age to the 14-29 years of age [15,16].

Figure 4: Mean per-patient annual cost of DMD [14,15]. <br>
(a) Mean per-patient annual cost of illness (columns) and mean
proxy-assessed patient utility (connected markers), by country
and ambulatory class.14 (b) Average total healthcare costs for
DMD and matched control cohorts [15].

Figure 4: Mean per-patient annual cost of DMD [14,15].
(a) Mean per-patient annual cost of illness (columns) and mean proxy-assessed patient utility (connected markers), by country and ambulatory class.14 (b) Average total healthcare costs for DMD and matched control cohorts [15].

According to Mordor Intelligence, the DMD treatment market size was estimated at 2.56 billion USD in 2024 and is projected to reach 4 [32]. billion USD by 2029, with a compound annual growth rate (CAGR) of 11.10% (Figure 5a) [7]. Business Wire predicted a higher trending global market, projecting the market for DMD medication to grow from USD 2 billion in 2022 to USD 27.4 billion by 2030, with a predicted global CAGR of 39.1% between 20222030; particularly with China having the largest CAGR of 46%, Japan with a 32.6% CAGR, Canada with a 31.7% CAGR, and Europe and Germany with a 34.8% CAGR [17]. Figure 5b demonstrates geographically where the DMD treatment market is expected to see the greatest growth rate [7].

Figure 5: Schematic of DMD Treatment Market [7].<br>
(a) DMD treatment market size based on 2019-2029 study
period [7]. (b) Growth rate of DMD treatment by region [7].

Figure 5: Schematic of DMD Treatment Market [7].
(a) DMD treatment market size based on 2019-2029 study period [7]. (b) Growth rate of DMD treatment by region [7].

The DMD market is expected to grow due to new product innovations, high healthcare expenditures, government awareness programs, and an increase in global clinical trials [7]. With increased R&D investment in the DMD market, patient advocacy, and regulatory incentives for drug development for rare diseases like DMD, we have seen advancements in gene therapy, extended use of steroids, and other novel treatments surface [17]. As DMD therapies advance, we expect these treatments to act as key driving factors for the DMD market [7]. In particular, the U.S. is expected to be a leader in clinical trials and remain dominant in the DMD regional market [7]. As anticipated by Market Research, with improved diagnosis methods, R&D focus and pipelines, and increased disease awareness, the market is anticipate to grow from 3.5 to 11 [7]. billion USD from 2023 to 2033 (Figure 6) [18]. Also of interest, with the recent COVID-19 pandemic, there was a rise in the DMD incidence rate, as patients with DMD were considered a high-risk population as they were more prone to experience severe COVID-19 symptoms [7]. This led to a greater DMD treatment adoption rate to combat the COVID-19 complications for DMD patients, thus increasing the expected market growth which is anticipated to maintain this upward trend through 2029 [7].

Figure 6: Duchenne Muscluar Dystrophy Market [17]. <br>
The image shows the DMD market size and trends for
molecular-based therapies, steroid therapy, and other
treatments as reported by Market Research over 2023-2033.

Figure 6: Duchenne Muscluar Dystrophy Market [17].
The image shows the DMD market size and trends for molecular-based therapies, steroid therapy, and other treatments as reported by Market Research over 2023-2033.

Existing DMD products

Currently, there is no cure for DMD on the market, but several therapeutic options are available to help slow disease progression, stabilize symptoms, and treat associated conditions [19]. These therapies primarily include corticosteroids, disease-modifying treatments like exon skipping and gene therapy, supportive care such as physical therapy, respiratory support, cardiac care, and investigational therapies undergoing clinical trials (Table 1) [3,1926]. Some leading companies in the DMD market are Sarepta Therapeutics, Pfizer Inc., PTC Therapeutics, FibroGen Inc., and F. Hoffman-La Roche AG [7]. Glucocorticoid therapy remains the gold standard for DMD patients due to its effectiveness in improving muscle strength and slowing the progression of muscle loss, inflammation, and other related symptoms [21]. The most commonly prescribed steroids for DMD are prednisone and deflazacort, primarily supplied by companies like EUROAPI, Coral Drugs Private Limited, Symbiotec Pharmalab, LGM Pharma, and Transo-Pharm USA LLC [22]. In 2023, the FDA approved vamorolone, a drug with a similar molecular structure as traditional corticosteroids created by ReveraGen BioPharma Inc [23]. Results from clinical trials showed vamorolone promoted improved muscle strength and stature without some of the side effects of the steroids such as stunted growth and weakened bones [23].

Table 1: DMD Therapeutic Approaches [3,19-26].

A more novel drug type that has more recently made its way to the market for DMD is exon-skipping agents [7]. Exon-skipping is a very effective treatment strategies for restoring the expression of a functional dystrophin protein in DMD patients [7]. In some DMD patients, a deletion nonsense mutation in the dystrophin gene leads to premature termination of the transcript and improper dystrophin production [27]. As depicted in Figure 7, exon skipping restores the dystrophin protein by antisense oligonucleotides (AONs) binding to specific exons and inducing splicing in premRNA [27]. The growing focus on exon-skipping technology has significantly propelled market growth [7]. For instance, eteplirsen (Exondys 51), produced by Sarepta Therapeutics, received fasttrack approval from the FDA in 2016 for treating DMD patients with a mutation in the dystrophin gene amenable to exon 51 skipping [28]. In 2019, the FDA approved Sarepta Therapeutics’ golodirsen (Vyondys 53) for DMD patients with a mutation amenable to exon 53 skipping [29]. In 2021, casimersen (AMONDYS 45), also manufactured by Sarepta Therapeutics, was approved by the FDA for DMD treatment targeting exon 45 skipping [30]. 

Figure 7: Illustration of the mechanism of exon skipping
therapy by skipping of exon 52 [27]. <br>
The image shows the mechanism of exon skipping therapy of
skipping exon 52. Exon skipping restores the dystrophin protein
by antisense oligonucleotides (AONs) binding to specific exons
and inducing splicing in pre-mRNA.

Figure 7: Illustration of the mechanism of exon skipping therapy by skipping of exon 52 [27].
The image shows the mechanism of exon skipping therapy of skipping exon 52. Exon skipping restores the dystrophin protein by antisense oligonucleotides (AONs) binding to specific exons and inducing splicing in pre-mRNA.

Clinical and preclinical trials

Clinical trials

In addition to the clinically applied treatment methods, many new emerging products are in the preclinical phase and may drastically expand the DMD market [3]. Some main therapies include in vivo gene correction, in vivo mRNA correction, upregulation of supporting molecules, enhancing muscle metabolism, novel steroids, and cell transplants [3]. This section investigates some of the most promising therapeutic options currently in clinical trials. The main products within clinical trials are listed in Table 2 [3,20,31-45].

In vivo gene correction aims to deliver the dystrophin gene to patients so their bodies can naturally synthesize the dystrophin protein [3]. Sarepta Therapeutics completed Phase III of a clinical trial for an open-label single-dose gene transfer therapy, known as delandistrogene moxeparvovec, which involves intravenous microdystrophin administration in boys with DMD [31]. The study, completed in 2023, found the injections to be well tolerated and resulted in long-term gene expression and phenotype improvements [3]. The drug has recently received FDA approval [3]. Additionally, Pfizer has been developing fordadistrogene movaparvovec, a gene therapy drug using an adeno-associated virus serotype-9 (AAV9) gene-replacement construct to introduce a dystrophin transgene into patients and restore functional dystrophin [32,33]. While Phase I clinical trials showed an acceptable safety profile and potential benefits for DMD patients by slowing muscle function loss, Phase III trials have been halted due to patient deaths [3,32].

In vivo mRNA corrections have also shown clinical benefits [3]. They utilize modified antibiotics to cause ribosomes to ignore stop codon sequences during the elongation transcription process, thereby overcoming nonsense mutations [3]. PTC Therapeutics, in their Phase III clinical trial of Ataluren, an orally delivered readthrough in vivo mRNA correction drug, observed a reduction and delay in many DMD symptoms, including loss of ambulation and respiratory decline [3,34]. Ataluren is now approved by the European Medicine Agency (EMA) for treating male patients older than 2 years with DMD caused by a premature stop codon [35]. Exon skipping, another form of in vivo mRNA correction, involves drugs like eteplirsen, golodirsen, casimersen, and vitolarsen, all recently FDA-approved [20].

Another preclinical DMD therapeutic option being investigated is upregulating sarcolemmal supporting molecules, like utrophin, which have functions similar to dystrophin, such as forming dystrophin-glycoprotein complexes and assisting in muscle cell attachment to the basal lamina, respectively [3]. However, despite promising results in mouse models of DMD, these methods saw minimal efficacy in human trials [36]. A more novel supporting molecule, sarcospan, was found to alleviate muscular dystrophy in mouse models when upregulated, but sacrospan has not begun clinical trials [3].

Table 2: DMD Treatment Products in Clinical Trial [3,20,31-45].

Cell transplantation is another promising approach under investigation, offering the potential to regenerate muscle tissue, reduce degeneration, and enhance muscle function [3]. In a Phase I/II clinical trial conducted by CHU de Quebec-Universite Laval, researchers explored the safety and effects on muscle strength of transplanting healthy parent donor myoblasts into the extensor carpi radialis, supplemented with immunosuppressants [37]. The study observed an increase in the percentage of myofibers expressing donor dystrophin, ranging from 3.5% to 26% four weeks post-injections [38]. Another study by Capricor Inc. investigated intracoronary infusion of cardiosphere-derived cells for 13 patients with substantial cardiomyopathy secondary to DMD [43]. In Phase II of the trial, patients received no immunosuppression, and results showed a significant reduction in scar size, improvement in inferior wall systolic thickening, and enhanced upper limb function [3,39]. Additionally, a Phase I/IIa clinical study examined autologous myoblast transplantation in patients with oculopharyngeal muscular dystrophy (OPMD), a rare genetic condition characterized by muscle weakness in the pharynx and upper eyelids, via local intramuscular injections [41,42]. Promising results were reported, with local administration demonstrating enhanced muscle repair efficacy and no deterioration in swallowing function observed over two years, providing proof-of-concept for muscle cell therapy in muscular dystrophy [42]. Heydemann., et al. evaluated the safety and efficacy of Dystrophin Expressing Chimeric (DEC) cell therapy, where patient myoblasts were fused with healthy donor myoblasts for 3 DMD patients [43]. As illustrated in Figure 8, DEC cells, created through ex vivo fusion and delivered via systemic-intraosseous administration, interacted with the DMD patient’s myoblasts, leading to improved muscle strength and function through dystrophin delivery [43]. The first-in-human study demonstrated promising safety and efficacy outcomes [43].

Figure 8: Mechanism of action of the Dystrophin Expressing
Chimeric Cell (DEC) cells created via ex vivo fusion of human
myoblast from normal and DMD-affected donors. Followed by
systemic-intraosseous administration causing DECs to engraft
and fuse with the myoblasts of DMD patients. The DEC cells then
deliver dystrophin and improve muscle strength and function
[43]. <br> 
This image shows how DEC cells, created through ex vivo fusion
and delivered via systemic-intraosseous administration, interact
with the DMD patient's myoblasts, leading to improved muscle
strength and function through dystrophin delivery.

Figure 8: Mechanism of action of the Dystrophin Expressing Chimeric Cell (DEC) cells created via ex vivo fusion of human myoblast from normal and DMD-affected donors. Followed by systemic-intraosseous administration causing DECs to engraft and fuse with the myoblasts of DMD patients. The DEC cells then deliver dystrophin and improve muscle strength and function [43].
This image shows how DEC cells, created through ex vivo fusion and delivered via systemic-intraosseous administration, interact with the DMD patient's myoblasts, leading to improved muscle strength and function through dystrophin delivery.

Preclinical Tissue Engineering Approaches for DMD Treatment

Cell therapy, while promising, has its challenges. The harsh environment and inflammation associated with DMD pose significant hurdles, leading to low cell viability and migration for transplanted cells [42]. These factors make it difficult to sustain the regeneration of the muscle tissue and achieve adequate improvement [42]. Therefore, increasing preclinical research has been investigating tissue engineering approaches for muscle injury and disease treatment [38]. Biomaterials and hydrogels offer a potential method to encapsulate and protect donor cells and growth factors during delivery for therapeutic repair [42]. They have the capability to transport cells or bioactive compounds for regenerating skeletal muscle tissue, either through transplanting muscle tissue fabricated in vitro or utilizing an injectable matrix that forms in the body, including pre-formed hydrogel particles (Figure 9) [42]. Table 3 displays the different tissue engineering designs, focusing on the two main approaches: pre-formed in vitro artificial skeletal muscle tissue and in vivo injectable delivery systems. 

Figure 9: Typical administration of hydrogel-based delivery
systems for therapeutic cells and/or bioactive molecules in
treatment of DMD [42]. <br>
(a) Implantation of a pre-formed hydrogel-based scaffold. (b)
Injectable approach hydrogel-based matrix, which is a liquid
precursor of the matrix that is injected and then formed in situ,
or pre-formed hydrogel particles.

Figure 9: Typical administration of hydrogel-based delivery systems for therapeutic cells and/or bioactive molecules in treatment of DMD [42].
(a) Implantation of a pre-formed hydrogel-based scaffold. (b) Injectable approach hydrogel-based matrix, which is a liquid precursor of the matrix that is injected and then formed in situ, or pre-formed hydrogel particles.

Table 3: Preclinical tissue engineering designs with pre-formed in vitro artificial skeletal muscle tissue and in vivo injectable delivery systems [46-59].

Much effort has been made to develop advanced scaffolds to mimic the skeletal muscle microenvironment and create artificial skeletal muscle tissue in vitro for reparative therapy [42]. Various approaches have been taken, with various scaffold biomaterial, cell types, and bioactive materials, to create a product that best assists with regenerating and improving muscle wasting diseases [42]. Nakayama., et al. synthesized a spatially aligned collagen scaffold embedded with C2C12 myoblasts and human microvascular endothelial cells [46,47]. When introduced into mouse models, researchers observed improved microvascularization, as well as the formation of endothelialized and aligned muscle tissue characterized by elongated myotubes, synchronized contractility, and increased secretion of angiogenic cytokines [46,47]. Lee., et al. also created a unique artificial skeletal muscle tissue using a poly(l-lactic acid) (PLLA) nonwoven scaffold synthesized with decellularized extracellular matrix (dECM) components and insulin growth factor-1 (IGF-1), embedded with C2C12 myoblasts (Figure 10) [46,49]. Other studies, such as Borselli., et al. and Falco., et al., have also investigated synthesizing scaffolds for delivering bioactive material, like growth factors or viral vectors, without cells [51,52].

Figure 10: Illustration of dECM hydrogel preparation and scaffold fabrication [49].
(A) decellularization process using rabbit skeletal muscle biopsies and (B) PLLA scaffold fabrication. (C, D) Gross appearance,
surface, and cross-sectional SEM images of the dECM/PLLA
scaffold.

Figure 10: Illustration of dECM hydrogel preparation and scaffold fabrication [49]. (A) decellularization process using rabbit skeletal muscle biopsies and (B) PLLA scaffold fabrication. (C, D) Gross appearance, surface, and cross-sectional SEM images of the dECM/PLLA scaffold.

Additionally, biomaterials and hydrogels can be used as delivery vehicles for cells and bioactive molecules when treating skeletal muscle diseases [42]. As direct cell transplantation has seen challenges with cell viability and limited migration once implanted in the harsh in vivo environment, a delivery system may encapsulate and protect the cell and help promote integration processes with the host tissues [42]. For instance, Rossi., et al. developed an injectable photopolymerizable hyaluronan-based hydrogel to deliver MPCs in a controlled manner [53]. When this hydrogelMPC complex was implanted into the ablated tibialis anterior muscle of mice, it enhanced the capacity of myogenic precursor cells to regenerate muscle tissue and facilitate functional recovery [53]. Fuoco., et al. similarly utilized a PF hydrogel matrix to inject mesoangioblasts [54]. Other studies have also made injectable hydrogel beads or microspheres [55,56]. In particular, Kankala., et al. fabricated PLGA-based porous microspheres to deliver skeletal myoblasts (Figure 11) [56]. These microspheres provided a favorable microenvironment for the myoblasts to integrate with and deposit ECM, improving and facilitating cell adhesion, proliferation, and myogenic differentiation [56]. When investigated in the mouse model, improved cell retention and vascularization were shown, demonstrating cell-laden microcarriers’ potential to promote muscle tissue regeneration through a minimally invasive delivery method [56]. Hwang., et al. and Ansari., et al. expanded on the cell delivery system and incorporated bioactive molecules, such as bFGF [58,59].

Figure 11: Schematic of the PLGA HOPMs for minimally invasive
cell delivery-based tissue regeneration [56]. <br>
This image displays Kankala., et al. fabricated PLGA-based
porous microspheres to deliver skeletal myoblasts.

Figure 11: Schematic of the PLGA HOPMs for minimally invasive cell delivery-based tissue regeneration [56].
This image displays Kankala., et al. fabricated PLGA-based porous microspheres to deliver skeletal myoblasts.

While there is increasing promise in tissue engineering methods for treating muscle tissue diseases and injuries like DMD, numerous challenges persist. Key among these are the methods of targeted implantation (i.e., localized injection versus surgical implantation), integration of scaffolds with host tissue, and the response of scaffolds to mechanical stimulation [38]. Addressing and optimizing these challenges are critical to progress toward developing enduring treatments that surpass current gold standards in the field.

Conclusion and Future Considerations

This article comprehensively reviews Duchenne Muscular Dystrophy (DMD), including an overview of the disease, its market size and trends, existing therapeutic options, products currently in clinical trials, and preclinical tissue engineering treatments. While there is increasing promise in tissue engineering methods for treating muscle tissue diseases and injuries like DMD, numerous challenges persist. Key among these are the methods of targeted implantation (i.e., localized injection versus surgical implantation), integration of scaffolds with host tissue, and the response of scaffolds to mechanical stimulation [42]. Addressing and optimizing these challenges are critical to progress toward developing enduring treatments that surpass current gold standards in the field. Future studies of tissue engineering DMD therapeutics should work to address significant limitations of current therapies, such as longterm expression of a functional dystrophin gene, restoring muscle regeneration capacity due to depleted muscle satellite cells, noninvasive delivery, and targeted delivery.

Funding Sources

There is no funding to report for this study.

Conflict of Interest Sources

Authors declare that there is no conflict of interest.

Acknowledgements

Jarod Carol expresses appreciation to Professor Bill Tawil for overseeing the framework of this review, and for the insightful lectures and advice concerning biomaterials and tissue engineering.

Bibliography

  1. Centers for Disease Control and “What is muscular dystrophy?”. Centers for Disease Control and Prevention (2022). 
  2. Venugopal V and Pavlakis “Duchenne Muscular Dystrophy”. In: StatPearls. Treasure Island (FL): StatPearls Publishing; (2024). 
  3. Heydemann A and Siemionow “A Brief Review of Duchenne Muscular Dystrophy Treatment Options, with an Emphasis on Two Novel Strategies”. Biomedicines 11.3 (2023): 830. 
  4. Duchenne muscular dystrophy (DMD) - diseases. Muscular Dystrophy Association (2021). 
  5. Duchenne muscular dystrophy - symptoms, causes, treatment: National Organization for Rare Disorders (2024). 
  6. S. National Library of Medicine. (n.d.). Duchenne muscular dystrophy: Medlineplus medical encyclopedia. MedlinePlus. 
  7. Duchenne muscular dystrophy treatment market Market Research Company - Mordor Intelligence TM. (n.d.). 
  8. Mukund K and Subramaniam S. “Skeletal muscle: A review of molecular structure and function, in health and disease”. Wiley interdisciplinary reviews. Systems biology and medicine 1 (2020): e1462. 
  9. Wilson DGS., et al. “The role of the dystrophin glycoprotein complex in muscle cell mechanotransduction”. Communication Biology 5 (2022): 1022. 
  10. Gao Q Q and McNally E “The Dystrophin Complex: Structure, Function, and Implications for Therapy”. Comprehensive Physiology 5.3 (2015): 1223-1239. 
  11. Venugopal V, Pavlakis “Duchenne Muscular Dystrophy”. [Updated 2023 Jul 10]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing (2024). 
  12. Duchenne muscular (DMD) | Sarepta Therapeutics. (n.d.-a). 
  13. Broomfield J., et al. “Life Expectancy in Duchenne Muscular Dystrophy: Reproduced Individual Patient Data Meta- analysis”. Neurology 23 (2021): e2304-e2314. 
  14. Broomfield J., et al. “Life expectancy in Duchenne muscular dystrophy: Reproduced individual patient data meta-analysis”. Neurology (2021). 
  15. Landfeldt , et al. “The burden of Duchenne muscular dystrophy: an international, cross-sectional study”. Neurology 83.6 (2014): 529-536. 
  16. Thayer S., et al. “The Direct Cost of Managing a Rare Disease: Assessing Medical and Pharmacy Costs Associated with Duchenne Muscular Dystrophy in the United States”. Journal of Managed Care and Specialty Pharmacy 6 (2017): 633-641. 
  17. “Duchenne muscular dystrophy drugs market poised for remarkable 39.1% CAGR, set to reach $27.4 billion by 2030 - com”. Business Wire (2023). 
  18. “Duchenne muscular dystrophy market share analysis report 2024”. biz. (2024). 
  19. “Therapeutic approaches for your patients with Duchenne Muscular Dystrophy”. com. (n.d.). 
  20. Patterson G., et al. “Duchenne muscular dystrophy: Current treatment and emerging exon skipping and gene therapy approach”. European Journal of Pharmacology 947 (2023): 
  21. “Deflazacort API manufacturers: Suppliers: Exporters”. PharmaCompass.com. (n.d.). 
  22. Children’s National HospitalChildren’s National HospitalChildren’s National Hospital has ranked as one of the top 10 pediatric hospitals in the nation - with the #1 ranked neonatology program - by U.S. News & World Report for the fifth year in a What. (n.d.). FDA approves steroid-like drug for Duchenne Muscular Dystrophy. clinicallab. 
  23. U.S. FDA Approves PREVNAR 20®, Pfizer’s 20-valent Pneumococcal Conjugate Vaccine for Infants and Children. Pfizer. (2023). 
  24. Center for Biologics Evaluation and (n.d.). PNEUMOVAX 23 - pneumococcal vaccine, Polyvalent. U.S. Food and Drug Administration. 
  25. “Profiling the seven biggest ventilator manufacturers across the world”. NS Medical Devices (2020). 
  26. Gotadki R. “Global ACE Inhibitors Market Overview”. Market Research Future. 
  27. Dzierlega K., et al. “Optimization of antisense-mediated exon skipping for Duchenne muscular dystrophy”. Gene Therapy 27 (2020): 407-416. 
  28. Walsh “FDA grants accelerated approval to first drug for Duchenne Muscular Dystrophy”. U.S. Food and Drug Administration (2016). 
  29. Kahn “FDA grants accelerated approval to first targeted treatment for Rare Duchenne muscular dystrophy mutation”. U.S. Food and Drug Administration (2019). 
  30. Sarepta Therapeutics Announces FDA Approval of AMONDYS 45TM (casimersen) Injection for the Treatment of Duchenne Muscular Dystrophy (DMD) in Patients Amenable to Skipping Exon Sarepta Therapeutics (2021). 
  31. Sarepta Therapeutics, Inc. “A Gene Transfer Therapy Study to Evaluate the Safety of Delandistrogene Moxeparvovec (SRP- 9001) in Participants With Duchenne Muscular Dystrophy (DMD)”. gov (2023). 
  32. Butterfield R., et al. “One year data from ambulatory boys in a phase 1B, open-label study of Fordadistrogene Movaparvovec (PF-06939926) for Duchenne muscular dystrophy (DMD)”. MDA Clinical and Scientific Conference (2023). 
  33. Pfizer. “A study to evaluate the safety and tolerability of PF06939926 gene therapy in Duchenne muscular dystrophy - full text view”. ClinicalTrials.gov (2022). 
  34. PTC Therapeutics. “Study of Ataluren for Previously Treated Participants With Nonsense Mutation Duchenne/Becker Muscular Dystrophy (nmDBMD) in Europe, Israel, Australia, and Canada”. gov (2020). 
  35. Michael , et al. “Long term treatment with ataluren-the Swedish experience”. BMC Musculoskeletal Disorders 22.1 (2021): 837. 
  36. Summit “Proof of Concept Study to Assess Activity and Safety of SMT C1100 (Ezutromid) in Boys With Duchenne Muscular Dystrophy (DMD)”. Clinicaltrials.gov. (2020b). 
  37. CHU de Quebec-Universite “Transplantation of Myoblasts to Duchenne Muscular Dystrophy (DMD) Patients”. Clinicaltrials.gov. (2021). 
  38. Skuk D., et al. “Dystrophin expression in muscles of duchenne muscular dystrophy patients after high-density injections of normal myogenic cells’. Journal of Neuropathology and Experimental Neurology 4 (2006): 371-386. 
  39. Capricor “HOPE-Duchenne (Halt cardiomyOPathy progrEssion in Duchenne) (HOPE)”. Clinicaltrials.gov. (2018). 
  40. Allergy and Asthma Consultants, Wichita, Kansas. “Allogeneic Human Umbilical Cord Mesenchymal Stem Cells for a Single Male Patient With Duchenne Muscular Dystrophy (DMD)”. gov. (2019). 
  41. Assistance  Publique   -   Hôpitaux    de        “Treatment of   Dysphagia   in   Oculopharyngeal   Muscular    Dystrophy by Autologous Transplantation of Myoblasts (OPMD)”. Clinicaltrials.gov. (2015). 
  42. Lev R and Seliktar “Hydrogel biomaterials and their therapeutic potential for muscle injuries and muscular dystrophies”. Journal of the Royal Society, Interface 15.138 (2018): 20170380. 
  43. Heydemann A., et al. “Dystrophin Expressing Chimeric (DEC) Cell Therapy for Duchenne Muscular Dystrophy: A First-in- Human Study with Minimum 6 Months Follow-up”. Stem Cell Reviews and Reports 19 (2023): 1340-1359. 
  44. Cossu G., et al. “Intra-arterial transplantation of HLA-matched donor mesoangioblasts in Duchenne muscular dystrophy”. EMBO Molecular Medicine 12 (2015): 1513-1528. 
  45. Klimczak , et al. “Co-Transplantation of Bone Marrow- MSCs and Myogenic Stem/Progenitor Cells from Adult Donors Improves Muscle Function of Patients with Duchenne Muscular Dystrophy”. Cells 9.5 (2020): 1119. 
  46. Fernández-Costa JM., et al. “Bioengineered in vitro skeletal muscles as new tools for muscular dystrophies preclinical studies”. Journal of Tissue Engineering. 12 (2021). 
  47. Nakayama K , et al. “Treatment of volumetric muscle loss in mice using nanofibrillar scaffolds enhances vascular organization and integration”. Communications Biology 2 (2019): 170. 
  48. Yeo M and Kim “Micro/nano-hierarchical scaffold fabricated using a cell electrospinning/3D printing process for co-culturing myoblasts and HUVECs to induce myoblast alignment and differentiation”. Acta Biomaterialia 107 (2020): 102-114. 
  49. Lee H., et al. “A novel decellularized skeletal muscle-derived ECM scaffolding system for in situ muscle regeneration”. Methods (San Diego, Calif.), 171 (2020): 77-85. 
  50. Denes L T., et al. “Culturing C2C12 myotubes on micromolded gelatin hydrogels accelerates myotube maturation”. Skeletal Muscle 1 (2019): 17. 
  51. Borselli , et al. “Functional muscle regeneration with combined delivery of angiogenesis and myogenesis factors”. Proceedings of the National Academy of Sciences of the United States of America, 107.8 (2010): 3287-3292. 
  52. Falco EE., et al. “Porous EH and EH-PEG scaffolds as gene delivery vehicles to skeletal muscle”. Pharmaceutical Research 6 (2011): 1306-1316. 
  53. Rossi C , et al. “In vivo tissue engineering of functional skeletal muscle by freshly isolated satellite cells embedded in a photopolymerizable hydrogel”. FASEB journal: official publication of the Federation of American Societies for Experimental Biology 25.7 (2011): 2296-2304. 
  54. Fuoco , et al. “Injectable polyethylene glycol-fibrinogen hydrogel adjuvant improves survival and differentiation of transplanted mesoangioblasts in acute and chronic skeletal- muscle degeneration”. Skeletal Muscle 2 (2012): 24. 
  55. Ruvinov E., et al. “The effects of controlled HGF delivery from an affinity-binding alginate biomaterial on angiogenesis and blood perfusion in a hindlimb ischemia model”. Biomaterials 16 (2010): 4573-4582. 
  56. Kankala R , et al. “Highly Porous Microcarriers for Minimally Invasive In Situ Skeletal Muscle Cell Delivery”. Small (Weinheim an der Bergstrasse, Germany) 15.25 (2019): e1901397. 
  57. Beier , et al. “Tissue engineering of injectable muscle: three-dimensional myoblast-fibrin injection in the syngeneic rat animal model”. Plastic and Reconstructive Surgery 118.5 (2006): 1113-1121. 
  58. Hwang JH., et al. “Combination therapy of human adipose- derived stem cells and basic fibroblast growth factor hydrogel in muscle regeneration”. Biomaterials 25 (2013): 6037- 6045. 
  59. Ansari S., et al. “Muscle Tissue Engineering Using Gingival Mesenchymal Stem Cells Encapsulated in Alginate Hydrogels Containing Multiple Growth Factors”. Annals of Biomedical Engineering 44 (2016): 1908-1920.

Copyright: © 2024 Carol Jarod and Tawil Bill. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Contact US