Neurology Care, Diagnostics, and Fawn Leigh, MD,​a,b​ Alessandra Ferlini, PhD,c​ Doug Biggar, MD,d​ Katharine Bushby, MD,e​ EmergingRichard Finkel, MD,f​ Lauren P. Morgenroth, Therapies MS,​g Kathryn R. Wagner, of MD, thePhDh,i​ Patient With Duchenne Muscular Dystrophy abstract DMD Duchenne muscular dystrophy is the most common form of childhood muscular dystrophy.‍ A mutation in the disrupts dystrophin (protein) production, causing damage to muscle integrity, weakness, loss of ambulation, and cardiopulmonary compromise by the second decade of life.‍ Life expectancy has improved from mid-teenage years to mid- 20s with the use of glucocorticoids and beyond the third decade with ventilator support and multidisciplinary care.‍ However, Duchenne muscular dystrophy is associated with comorbidities and is a fatal disease.‍ Glucocorticoids prolong ambulation, but their side effects are significant.‍ Emerging investigational therapies have surfaced over the past decade and have rapidly been tested in clinical trials.‍ Gene-specific strategies include nonsense readthrough, , gene editing, utrophin modulation, and gene replacement.‍ Other mechanisms include muscle regeneration, antioxidants, and antifibrosis and anti-inflammatory pathways.‍ With potential therapies emerging, early diagnosis is needed to initiate treatment early enough to minimize morbidity and mortality.‍ Newborn screening can be used to significantly improve early diagnosis, especially for gene-specific therapeutics.‍

– DMD Duchenne muscular dystrophyDMD 1 (DMD),3 although rare, is the most common muscular dystrophy, with a prevalence of 1 in 5000.‍ ‍‍ It is an X-linked disorder caused by a mutation in the gene located on Xp21.‍ The gene is large (79 exons; 427 Kilodalton) and encodes a 427-Kilodalton muscle isoform protein dystrophin.‍ The gene is located in the dystrophin-associated glycoprotein complex at the sarcolemma and in the cytoskeleton.‍ The protein is located primarily in4,5​ skeletal muscle to stabilize the plasma membrane and maintain the strength of muscle fibers.‍ The absence of dystrophin causes muscle membrane damage, elevated serum creatinine kinase (CK), aMassachusetts General Hospital and Harvard Medical School, Harvard University, Cambridge, Massachusetts; cUniversity of Ferrara, Ferrara, Italy; dHolland Bloorview Kids Rehabilitation Hospital, Toronto, Ontario, Canada; eJohn Walton Centre for Muscular Dystrophy Research, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; fNemours Children’s Health System, Orlando, Florida; gChildren’s National Health System, Washington, District of Columbia; hKennedy Krieger Institute, Baltimore, Maryland; and iSchool of Medicine, Johns Hopkins University, Baltimore, Maryland bSeattle Children's Hospital, University of Washington, Seattle, Washington

The guidelines or recommendations in this article are not American Academy of Pediatrics policy, and publication that was partially supported herein does not imply endorsement.

Dr Wagner served as the chairperson of the Duchenne Muscular Dystrophy Care Considerations Neuromuscular Management Working Group as convened by the Centers for Disease Control and Prevention and reviewed and revised the manuscript; Dr Leigh served in the Duchenne Muscular Dystrophy Care Considerations Neuromuscular Management Working Group as convened by the Centers for Disease Control and Prevention, drafted the initial manuscript, and reviewed and revised the manuscript; Drs Biggar, Bushby, and Finkel served in the Duchenne Muscular Dystrophy Care Considerations Neuromuscular Management Working Group as convened by the Centers for Disease Control and Prevention, contributed to the development of corresponding recommendations, and reviewed and revised the manuscript; Drs Ferlini and Morgenroth served in the Duchenne Muscular Dystrophy Care Considerations Diagnostics Management Working Group as convened by the Centers for Disease Control and Prevention, contributed to the development of corresponding recommendations, and reviewed and revised the manuscript; and all authors approved the final manuscript as submitted and agree to be accountable for all aspects of the work. DOI: https://​doi.​org/​10.​1542/​peds.​2018-​0333C Accepted for publication Jul 26, 2018

Downloaded from www.aappublications.org/news by guest on September 25, 2021 PEDIATRICS Volume 142, number s2, October 2018:e20180333C SUPPLEMENT ARTICLE FIGURE 1 Summary of neuromuscular management depending on the clinical stage of patients with DMD. (Adapted with permission from Birnkrant DJ, Bushby K, Bann CM, et al. Diagnosis and management of Duchenne muscular dystrophy, part 1: diagnosis, and neuromuscular, rehabilitation, endocrine, and gastrointestinal and nutritional management. Lancet Neurol. 2018;17(3):252.)

NEUROLOGY fiber necrosis, muscle degeneration, ventilatory support.‍ The mean age 6 ∼ and regeneration.‍ Constant at which patients lose ambulation 11 DMD is a proximal myopathy muscle breakdown and necrosis is is 9 to 10 years.‍ Glucocorticoids affecting muscles, such as the followed by inflammatory processes are used to improve muscle strength 12 quadriceps and gluteals.‍ Fast- that lead to the– replacement of and function in patients with DMD 13 twitch (type IIb) myofibers are muscle with fibrotic7 9 tissue and fat and prolong ambulation by an 14 preferentially affected, leaving accumulation.‍ ‍‍ additional 2 to 3 years ; patients the slow-twitch18,19​ (type I) type to Common presenting symptoms with DMD who are treated with predominate.‍ ‍ The disease include delayed motor milestones, glucocorticoids become wheelchair is characterized by muscle inability to run or hop, toe walking, dependent by 12 to 13 years of age.‍ wasting, weakness, myofiber size difficulty climbing stairs, and Loss of ambulation is associated variation with muscle necrosis, fat hypertrophy of calf muscles.‍ with greater morbidity and mortality accumulation, connective tissue Nonmotor presentations include as the disease progresses; not replacement, and paradoxical failure to thrive, speech delay, surprisingly, the primary objective hypertrophy of calf muscles.‍ Gait fatigue, abnormal transaminases, of researchers in most clinical abnormality and difficulty getting myoglobinuria, and complications trials is prolonging ambulation.‍ up from a chair or from a supine because of anesthesia (ie, malignant Cardiomyopathy and respiratory position are common clinical hyperthermia or rhabdomyolysis).‍ insufficiency can shorten the life features.‍ However, the onset of symptoms can vary in age, severity, Global developmental delay is a span, but with ventilator support, common presentation.‍ Early signs and presentation.‍ Individuals survival in DMD has been extended and symptoms present at ages 2 to affected by DMD typically present to >30 years, and cardiac function 3 years, whereas the average age with delayed motor development is the remaining determinant of diagnosis is 3 to 5 years.‍ It has 15 or a gait abnormality, especially been reported that it takes >1 year of survival.‍ Investigational toe walking or delayed age of drugs that are used to improve to confirm DMD10 from the time of ambulation, and are often referred presentation,​ which is far too long cardiopulmonary function also to physical therapy.‍ Although the in a disease for which time means have emerged in clinical trials (for average age of presentation ranges loss of muscle function and the additional details, see the specialty from 2 to 5 years, the recognition familial risk of recurrence is up to article on cardiac management16 that of loss of milestone and the use of 50% in male siblings.‍ is part of this supplement ).‍ Our Bayley Scales of Infant Development, purpose in this article is to guide Third Edition can improve the – With recent advancements in clinicians in the diagnosis and identification of affected infants 20 22 clinical trials, there is an unmet treatment of DMD itself, expanding before motor impairment occurs.‍ ‍ need to diagnose affected infants 1 on principles outlined in the 2018 and children early.‍ Mendell et al GLUCOCORTICOIDS Duchenne Muscular Dystrophy Care presented data on newborn screening in Ohio initiated with a CK level from Considerations sponsored by the dried blood spot followed by genetic Centers for 17Disease Control and Prednisone prolongs ambulation Prevention.‍ In Fig 1, we provide an confirmation of11 those with elevated and has been shown to have long- overall guide for the neuromuscular CK.‍ Eagle et al reported that life ’ term benefits for– respiratory and management of patients with DMD expectancy in the 1960s was 14 cardiac function12,13,​ 23​ 25 in nonambulatory years; in the 1990s, life expectancy according to each patient s clinical patients.‍ ‍ ‍ ‍ Some researchers was 25 years for those receiving stage.‍ suggest there may be fewer side Downloaded from www.aappublications.org/news by guest on September 25, 2021 S6 LEIGH et al effects with 26,deflazacort27​ than prednisone.‍ ‍ However, the relative efficacy and side effects of different glucocorticoid regimens remains untested in well-designed, head-to-head trials.‍ The standard dose for prednisone is 0.‍75 mg/kg per day and for deflazacort is 0.‍9 mg/kg per day (Fig 2).‍ For a more detailed discussion on glucocorticoid management, see the Supplemental Information.‍ DIAGNOSIS

ClinicalDMD variability, genetic heterogeneity, and the large size of the gene contribute to the complexity of diagnosis (Fig 3).‍ Mutational analysis has made the confirmation of a molecular diagnosis possible in the majority of cases.‍ However, diagnosis and prognosis are also dependent on clinical presentation and genetic profile.‍ Genotype-phenotype correlation can help prognosticate, but age of presentation and family history are FIGURE 2 also important.‍ For example, most Care considerations for glucocorticoid (steroid) initiation and use for patients with DMD. ACTH, exon deletions are correlated with adrenocorticotropic hormone; CRH, corticotropin-releasing hormone; HPA, hypothalamic-pituitary- “ ” adrenal. (Reproduced with permission from Birnkrant DJ, Bushby K, Bann CM, et al. Diagnosis and disease severity on the basis of the ∼ management of Duchenne muscular dystrophy, part 1: diagnosis, and neuromuscular, rehabilitation, reading-frame28 rule and apply to endocrine, and gastrointestinal and nutritional management. Lancet Neurol. 2018;17(3):255.) 92% of cases.‍ The vast majority of whole-exon deletions or duplications are correlated with a pathogenic mutation.‍ The reading-frame stop biopsies were routinely performed to transcription testing to identify codon caused by an out-of-frame diagnose DMD.‍ With advancements intronic changes.‍ – mutation disrupts the open reading in genetic testing strategies, including In a child with a family history frame and ablates the translation multiplex ligation dependent probe DMD of DMD, a serum CK level is of dystrophin, causing a more amplification, comparative genomic recommended.‍ In a child without severe phenotype.‍ An in-frame hybridization, Sanger sequencing, a family history, the presence mutation can still translate the open and next-generation sequencing, of global development delay, reading frame to a partial-length a molecular diagnosis can be psychomotor delay, and/or dystrophin, resulting in Becker – obtained from genomicDMD DNA so that motor delay prompts a serum CK muscular dystrophy and a milder 29 muscle biopsies are rarely needed level (CK is elevated 50 100 times phenotype.‍ In some cases, in-frame to diagnose DMD.‍ If a gene normal in DMD).‍ The liver enzymes, mutations with worsening myopathy mutation cannot be identified by aspartate aminotransferase, are more clinically consistent with using genetic testing, clinicians may and alanine aminotransferase DMD.‍ choose to perform a muscle biopsy are also elevated but from the γ The methods used to diagnose DMD to determine the percentage of muscle instead of the liver.‍ include clinical history, physical dystrophin by using histopathological -glutamyl transferase is the examination, serum CK, liver staining and immunoblot analysis.‍ preferred laboratory test to enzymes, genetic testing, and perhaps If dystrophin staining is equivocal, check for liver disease in DMD.‍ muscle biopsy.‍ In the past, muscle the muscle can be sent for RNA If CK is elevated, then DNA should Downloaded from www.aappublications.org/news by guest on September 25, 2021 PEDIATRICS Volume 142, number s2, October 2018 S7 TABLE 1 Types of DMD Mutation Mutation % Deletions 65 Duplications 6–10 Point mutations 25 Complex mutations <2

evolution of next-generation sequencing has enabled the sequencing of millions of copies of

DNA fragments simultaneously32 and has reduced time and cost.‍ If the deletion and/or duplication test result is negative, then sequencing is conducted to identify smaller mutations.‍ Sequencing the entire coding region can reveal small mutations, splicing mutations, or single base changes.‍ If no mutation is identified after sequencing, a muscle biopsy can be obtained to determine the presence or absence of dystrophin by using histologic staining.‍ The absence of dystrophin confirms a diagnosis of DMD.‍ Reduced dystrophin could be associated with Becker muscular dystrophy or DMD depending on the presence and severity of muscle weakness on examination.‍ ≥ Dystrophin <3% is consistent with DMD, whereas 20% on muscle biopsy suggests that DMD is unlikely.‍ Some variation in muscle biopsy results occurs depending on the age of the child and what part of the affected muscle was biopsied.‍ Alternatively, tissue from the muscle can be tested through RNA FIGURE 3 transcription analysis to assess for Diagnosis of Duchene muscular dystrophy. Described early signs and symptoms of DMD are based intronic mutations that may have on Ciafaloni et al.10 (Reproduced with permission from Birnkrant DJ, Bushby K, Bann CM, et al. Diagnosis and management of Duchenne muscular dystrophy, part 1: diagnosis, and neuromuscular, been identified in the other genetic rehabilitation, endocrine, and gastrointestinal and nutritional management. Lancet Neurol. strategiesDMD .‍ Immunoblotting of the 2018;17(3):253.) muscle biopsy also can be used.‍ If no mutation is identified, the clinician can consider an alternative diagnosis, such as limb-girdle or be sent to test for exonic deletions used technique and can be used congenital muscular dystrophy.‍ ∼ and duplications, 30,which31​ are seen to test all 79 exons.‍ Comparative Imaging with quantitative magnetic in 65% of cases.‍ ‍ Table genomic hybridization is an resonance and magnetic resonance 1 includes the types of gene oligoclonal-based method that spectroscopy reveal an increase in fat – 33,34​ mutations in DMD.‍ Multiplex can be used to detect complex fraction with disease progression.‍ ‍ ligation dependent probe rearrangements, intronic 30alteration, Given the heterogeneity of affected amplification is the most widely or mutation break points.‍ The skeletal muscle in DMD, an Downloaded from www.aappublications.org/news by guest on September 25, 2021 S8 LEIGH et al FIGURE 4 Drug discovery chart for DMD clinical trials. Illustrated is the active pace of drug discovery in DMD.

Restore Protein (Dystrophin)

MRI-guided muscle biopsy can be uses antisense oligonucleotides.‍ ’ applied in the diagnosis and/or Two chemical constructs, Exon skipping and nonsense confirmation of DMD when genetic 2 -O-methyl phosphorothioate and readthrough can restore the testing is unrevealing.‍ phosphorodiamidate expression of the gene product oligomers, have been used in dystrophin.‍ These agents generate EMERGING THERAPIES clinical studies to bind to exon- a functional or partially functional ’ splice junctions and generate exon protein in DMD.‍ 35,36​ Exon Skipping skipping.‍ ‍ The 2 -O-methyl phosphorothioate drug, drisapersen, Numerous treatment strategies have for exon 51 skipping, which initially been investigated for DMD.‍ After The concept behind exon skipping showed improvement on the decades of searching for targets, is targeted at the messenger RNA 6-minute walk test in early clinical identifying molecular pathways, and level and is based on the reading- trials, progressed to a larger phase preclinical investigations in animal frame rule.‍ The intent is to skip (or III study in which it did37, not38​ reach models, potential therapeutic targets delete) an out-of-frame mutation its primary end point.‍ ‍ The have been translated to clinical and restore the reading frame by phosphorodiamidate morpholino trials in DMD.‍ In this section, we making an in-frame mutation that oligomers drug, , is also review some of the main strategies encodes a more functional protein designed for exon 51 skipping.‍ – and active clinical trials. ‍Figure 4 is that is consistent with the milder, Researchers in an initial study a chart of the potential drugs in the Becker muscular dystrophy like enrolled 12 patients for >3 years, and DMD drug discovery pipeline.‍ phenotype.‍ This technology these treated patients demonstrated Downloaded from www.aappublications.org/news by guest on September 25, 2021 PEDIATRICS Volume 142, number s2, October 2018 S9 TABLE 2 Exon Skipping–Amenable Mutations Exon Amenable to Skip % a slower rate of decline in ambulation an oral anti-inflammatory steroid – compared with untreated historical 51 13.0 with fewer side effects in preclinical 39,40​ 45 8.1 controls.‍ ‍ Exon skipping and phase I trials compared with ∼ ∼ 53 7.7 amenable mutations constitute 44 6.2 traditional glucocorticoids, has 80% of all DMD, of which 13% 46 4.3 advanced to phase II in an ongoing, 52 4.1 of cases are41 amenable to exon 51 larger clinical trial to assess safety 50 4.0 Antioxidantsand efficacy.‍ skipping (Table 2).‍ Drisapersen 43 3.8 is administered subcutaneously, 6 and 7 3.0 and eteplirsen is delivered by 8 2.3 intravenous infusion.‍ A drisapersen 55 2.0 Oxidative stress can damage cellular application to the US Food and function, specifically mitochondria.‍ Drug Administration (FDA) was not Replace Lost Dystrophin The overproduction and/or approved, but eteplirsen was granted accumulation of reactive oxygen accelerated approval by the FDA species can lead to mitochondrial ∼ dysfunction in neuromuscular while an ongoing42 confirmatory study Utrophin is an analog of dystrophin– is underway.‍ disease.‍ Studies have revealed that that shares 80% sequence Nonsense Suppression 50 52 increased reactive oxygen species homology with dystrophin.‍ ‍ ‍ in DMD contribute to membrane SMT C1100 is a molecule that permeability and protein degradation works as a utrophin modulator and and activates inflammatory is an investigational drug with the Nonsense mutations generate a pathways, thereby exacerbating potential to replace lost dystrophin stop codon, resulting in a truncated, 53 necrosis and fibrosis.‍ Idebenone, with utrophin.‍ Safety and nonfunctional protein.‍ Nonsense an antioxidant that inhibits lipid ∼ tolerability has been shown in (stop-codon) mutations comprise43,44​ 54 peroxidation, has shown efficacy in a study of 12 patients with DMD.‍ 10% to 15% of DMD cases.‍ ‍ The a phase III trial, with improvement A phase II study is ongoing to readthrough strategy, which involves seen in respiratory function (ie, evaluate the activity and safety 57 the suppression of the stop-codon forced vital capacity).‍ Idebenone of this orally administered drug mutation, encourages the ribosome is orally administered and currently Targetin a larger Signaling cohort Pathways of patients.‍ to read through the stop codon, undergoingAntifibrosis regulatory review.‍ which promotes45 the production of Anti-inflammatory Approaches dystrophin.‍ Proof of concept for nonsense suppression was initially In DMD, endomysial fibrosis is a established 46with an aminoglycoside: Muscle fibers in DMD undergo hallmark clinical feature associated gentamicin.‍ Translarna (), inflammatory and fibrotic changes with muscle weakness and poor β β an oral medication with a better that have been responsive to long-term outcome; transforming safety profile compared with glucocorticoids.‍ As a result, anti- growth factor (TGF- ) is a target gentamicin (formerly referred to as inflammatory approaches to in antifibrosis therapy.‍ Therapeutic β PTC124), promotes the readthrough treat DMD remain a focus in drug agents aimed at blocking cytokine of premature translation termination discovery.‍ Myofiber necrosis is signaling by inhibiting the TGF- κ codons.‍ Safety and tolerability was reported to result from chronic pathway in preclinical models α established, and a phase IIa proof- NF- B activation9,55,​ 56​ and tumor necrosis have been shown– to decrease of-concept trial revealed increased factor .‍ ‍ ‍ Prednisone and fibrosis in some58 61studies, although dystrophin47 posttreatment on muscle deflazacort, used to treat DMD, not in others.‍ ‍ ‍ Losartan, an biopsy.‍ Due to the unmet need for have anti-inflammatory effects antihypertensive drug with a known κ treatments in DMD and promising and are presumed to inhibit the safety profile, was an attractive early phase findings, this drug has NF- B pathways.‍ However, these therapeutic target, but the preclinical been approved by 48the European drugs are generally nonspecific studies that initially revealed reduced Medicines Agency.‍ A phase III anti-inflammatory agents with fibrosis later revealed minimal κ 62,63​ trial revealed enrichment of effect multisystem side effects.‍ New drugs functional benefit.‍ ‍ Another agent, α in a subgroup of patients with a aimed at specifically targeting NF- B oral halofuginone hydrobromide baseline 6-minute walk test between49 or tumor necrosis factor are being (also known as HT-100), is currently 300 and 400 m at enrollment.‍ moved forward in drug discovery and in a phase II trial to assess safety, However, the FDA voted that the are currently in the preclinical stages, tolerability, and dose selection for data used to support Translarna poised to move to clinical trials.‍ future trials.‍ Givinostat, an oral were inconclusive.‍ Vamorolone (also known as VBP15), inhibitor of histone deacetylases Downloaded from www.aappublications.org/news by guest on September 25, 2021 S10 LEIGH et al aimed at reducing fibrosis of muscle I or II trials in ambulatory DMD transfer has addressed76,77​ some of the fibers and promoting muscle patients.‍ This agent is delivered technicalDMD barriers.‍ ‍ However, AAV regeneration in DMD,64 is in a phase III Improvesubcutaneously Cardiac.‍ Function has a limited capacity to package the globalPhosphodiesterase clinical trial.‍ 5A Inhibition Geneentire Replacement gene.‍ DMD CAP-1002 is a novel therapeutic Diminished blood flow can cause65,66​ approach in which cell therapy is In DMD, the large size of the muscle damage.‍ Mendell et al ‍ used to treat cardiomyopathy in gene poses the challenge of packagingDMD showed in the mouse model that DMD.‍ This investigational treatment into an AAV.‍ Mini-dystrophins (ie, functional ischemia produces is already being investigated as miniaturized versions of the78, 79​ similar changes seen in muscle fiber a treatment of postmyocardial gene) have been developed.‍ ‍ degeneration and regeneration in infarction.‍ The agent is an allogenic Investigators also have packaged the DMD.‍ Researchers in more studies cell therapy derived from human gene into 2 vectors in a dual-AAV have since implicated the nitric oxide heart tissue that is administered approach and showed that it restored synthase pathway in ischemia and directly to the heart through sarcolemmal neuronal nitric oxide thus inhibition of phosphodiesterase the coronary arteries by cardiac synthase expression80 in dystrophin- (PDE) 67as a potential therapeutic catheterization.‍ This drug is currently deficient mice.‍ Transgene delivery target.‍ Sildenafil, an oral in the phase I or II study stage to by an AAV through intramuscular PDE 5 inhibitor, was studied in investigate safety and tolerability.‍ injection in a phase I clinical trial adult patients with DMD and in humans revealed an immune cardiomyopathy.‍ The trial was Eplerenone, a drug that is already response to the transgene product, terminated early due to worsening used in heart failure and has a known bringing attention to the role of safety profile, has been investigated left-ventricular end-systolic68 volume T-cell immunity to self- and nonself- in a randomized, placebo-controlled in subjects on sildenafil.‍ Tadalafil, dystrophin in the study design81 of another oral PDE inhibitor, was clinical trial in patients with DMD future trials.‍ Limb and early cardiomyopathy who shown to restore blood supply to vascular delivery is another method82 muscle in the mdx mouse.‍ However, are already taking an angiotensin- to regionally deliver the gene.‍ tadalafil had no effect on the primary converting enzyme inhibitor.‍ After Systemic delivery can be achieved outcome and did not lessen the 12 months, it was reported that a intravenously, but targeting the gene statistically significant reduction in decline in ambulatory69 ability in to muscle and limiting its off-target patientsMyostatin with Inhibition DMD.‍ left-ventricular strain occurred in delivery are important aspects of the the treatment73 arm compared with gene delivery.‍ Although gene therapy a placebo.‍ Although long-term is still in early stages, the knowledge The inhibition of the myostatin benefits cannot be concluded from gained from the pioneering work pathway is a muscle-building these findings, longitudinal data of many dedicated investigators therapeutic approach for muscular from this cohort will provide greater over several decades has taken the dystrophy.‍ Myostatin regulates insight on the potential benefit of this field closer to becoming a potential muscle growth by breaking down cardiac intervention in DMD.‍ Results therapy.‍ Phase I and II studies to muscle protein, and blocking after a 2-year study revealed that deliver microdystrophin C with AAV myostatin has been shown to eplerenone can be cardioprotective74 vectors are anticipated to begin this inRestore patients Gene with Function DMD.‍ Gene Editing increase70, muscle71​ mass and72 reduce year.‍ fibrosis.‍ ‍ Wagner et al conducted an early phase clinical trial in adult patients with muscular dystrophy In a monogenetic disorder, such as A novel genetic engineering “ ” using a recombinant human antibody DMD, the idea of replacing a defective technology, clustered regularly and showed safety, although not gene with a corrected gene seems interspaced short palindromic efficacy.‍ New strategies and the simple, but many challenges have repeats (CRISPR) coupled with an formulation of an antimyostatin been encountered to date.‍ Strategies endonuclease (CRISPR-associatedDMD antibody, PF-06252616, have since on how to target the desired muscle, protein 9), has been applied to occurred, and the agent is being circumvent the immune response, edit mutations in the gene.‍ used in an ongoing phase II clinical and achieve efficient systemic Researchers in several laboratories trial in patients with DMD.‍ This delivery75 are some of the major have reported that gene editing can agent is delivered intravenously.‍ hurdles.‍ The discovery of an adeno- be used to improve muscle function Similarly, an antimyostatin adnectin, associated virus (AAV) to be a safe in the mouse model and muscle BMS-0986089, is currently in phase and effective therapeutic tool in gene stem cells.‍ Investigators have shown Downloaded from www.aappublications.org/news by guest on September 25, 2021 PEDIATRICS Volume 142, number s2, October 2018 S11 that the editing tool was used– to as possible.‍ Care centers will need intervention are necessary.‍ As the resect the faulty exons and83 87 partially to incorporate health service translational community shepherds restore protein in mice.‍ ‍ The considerations into their care plans.‍ novel therapeutics into the clinic, “ ” mice treated with CRISPR did better The molecular diagnoses stored other initiatives are equally important, on functional tests compared with in unified databases, which including newborn screening, the untreated group but not as well include molecular profiles and well- management strategies for infants and as the normal mice.‍ Although gene characterized phenotypes, can be toddlers, neurobehavioral management editing with CRISPR is closer to used to augment the identification for school-aged children, transition clinical translation now than ever and selection of more homogeneous management, and adult considerations before, it may be years before these candidates for specific therapies for affected men.‍ Newborn screening technologies achieve efficiency as gene-derived strategies are and effective treatments may towardTarget Genetic clinical Modifierstrials.‍ translated to the clinics.‍ As genetic ultimately extend survival in DMD modifiers are validated, biomarkers beyond the third decade.‍ and novel therapeutic targets can ACKNOWLEDGMENT The vast clinical variability in DMD be developed.‍ Putative disease- is well known, and environmental modifying strategies related to factors can contribute to the genetic cardiac function and neuronal We thank Joan Scott (acting modifier effect and heterogeneity involvement in DMD warrant further director, Division of Services for β of phenotypic features.‍ Latent exploration for novel targets.‍ Children with Special Health Needs, CONCLUSIONS TGF- binding proteinSPP1 4 can be Maternal and Child Health Bureau, used to predict88, the89​ age of loss DMD ∼ Health Resources and Services of ambulation.‍ ‍ , a gene Administration), who served on the encoding osteopontin, acts as a The gene was identified 3 Duchenne Muscular Dystrophy Care decades ago, and the presteroid pharmacodynamic89 biomarker of Considerations steering committee era changed after prednisone was steroidANXA6 response.‍ These 2 pathways 12 and diagnostic subcommittee.‍ β shown to prolong ambulation.‍ converge in the regulation of ABBREVIATIONS TGF- .‍ encodes annexin A6, Glucocorticoids remain the standard a calcium-binding protein, and is of care, but significant side effects correlated with sarcolemmal repair limit their therapeutic window.‍ 90,91​ AAV: adeno-associated virus in the mouse model.‍ These As survival has improved with the CK: creatinine kinase modifier are correlated with launch of specialized care centers CRISPR: clustered regularly phenotypic variation and serve as and better respiratory support, interspaced short novel therapeutic targets for future cardiac disease has become a more palindromic repeats drug discovery.‍ important cause of death.‍ The DMD: Duchenne muscular FUTURE DIRECTIONS majority of the investigational drugs dystrophy improve skeletal and pulmonary FDA: Food and Drug function, and cardiomyopathy Administration As disease-modifying treatments remains a critical area in need of β PDE: phosphodiesterase emerge, patients with DMD will effective treatment.‍ Furthermore, β TGF- : transforming growth live longer, and they will need to be addressing neurodevelopmental factor prepared to live as independently needs and providing early

Address correspondence to Fawn Leigh, MD, Seattle Children's Hospital, University of Washington, 4800 Sandpoint Way NE, M/S MB.7.420, Seattle, WA 98105. Email: [email protected] PEDIATRICS (ISSN Numbers: Print, 0031-4005; Online, 1098-4275). Copyright © 2018 by the American Academy of Pediatrics FINANCIAL DISCLOSURE: Other than those listed under potential conflict of interest, the authors have indicated they have no financial relationships relevant to this article to disclose. FUNDING: Supported in part by Cooperative Agreement NU38OT000167, funded by the Centers for Disease Control and Prevention. POTENTIAL CONFLICT OF INTEREST: Dr Leigh has served as a consultant to Sarepta Therapeutics and Summit Therapeutics and as principal investigator on clinical research grants sponsored by Sarepta Therapeutics, Pfizer, Atlas 5D, and Solid Biosciences; Dr Bushby has given expert advice to Solid Ventures, Gerson Lehrman Group research, Catabasis Pharmaceuticals, Bristol-Myers Squibb, PTC Therapeutics, BioMarin Pharmaceutical, and Laboratory of the Government Chemist Ltd and has received a grant and speaker fees from PTC Therapeutics; Dr Finkel has received grant support for Duchenne muscular dystrophy–related

Downloaded from www.aappublications.org/news by guest on September 25, 2021 S12 LEIGH et al topics from Bristol-Myers-Squib, Catabasis Pharmaceuticals, the National Institutes of Health, Eli Lilly and Company, the Muscular Dystrophy Association, PTC Therapeutics, ReveraGen BioPharma, Santhera Pharmaceuticals, Sarepta Therapeutics, and Summit Therapeutics and has served as a paid consultant to Catabasis Pharmaceuticals, Eli Lilly and Company, PTC Therapeutics, Sarepta Therapeutics, and Summit Therapeutics, and none of these relationships are directly related to Dr Finkel’s participation in this activity or article; the other authors have indicated they have no potential conflicts of interest to disclose.

REFERENCES 1. Mendell JR, Shilling C, Leslie ND, (MD STARnet). J Pediatr. 2009;155(3): dystrophy, express connexin et al. Evidence-based path to 380–385 hemichannels that lead to apoptosis. newborn screening for Duchenne 11. Eagle M, Baudouin SV, Chandler Cell Mol Life Sci. 2016;73(13):2583–2599 muscular dystrophy. Ann Neurol. C, Giddings DR, Bullock R, Bushby 20. Bayley N. Bayley Scales of Infant and 2012;71(3):304–313 K. Survival in Duchenne muscular Toddler Development: Administration 2. Cowan J, Macdessi J, Stark A, dystrophy: improvements in Manual. San Antonio, TX: Harcourt Morgan G. Incidence of Duchenne life expectancy since 1967 and Assessment; 2006 muscular dystrophy in New South the impact of home nocturnal 21. Connolly AM, Florence JM, Cradock Wales and Australian Capital ventilation. Neuromuscul Disord. MM, et al; MDA DMD Clinical Research Territory. J Med Genet. 1980;17(4): 2002;12(10):926 929 – Network. One year outcome of boys 245 249 – 12. Mendell JR, Moxley RT, Griggs RC, et al. with Duchenne muscular dystrophy 3. Romitti PA, Zhu Y, Puzhankara S, et al; Randomized, double-blind six-month using the Bayley-III scales of infant and MD STARnet. Prevalence of Duchenne trial of prednisone in Duchenne’s toddler development. Pediatr Neurol. and Becker muscular dystrophies muscular dystrophy. N Engl J Med. 2014;50(6):557–563 in the United States. Pediatrics. 1989;320(24):1592 1597 – 22. Albers CA, Grieve AJ. Test review: 2015;135(3):513 521 – 13. Matthews E, Brassington R, Kuntzer T, Bayley, N. (2006). Bayley scales of 4. Davies KE, Nowak KJ. Molecular Jichi F, Manzur AY. Corticosteroids for infant and toddler development–third mechanisms of muscular dystrophies: the treatment of Duchenne muscular edition. San Antonio, TX: Harcourt old and new players. Nat Rev Mol Cell dystrophy. Cochrane Database Syst Assessment. J Psychoeduc Assess. Biol. 2006;7(10):762–773 Rev. 2016;(5):CD003725 2007;25(2):180–190 5. Nowak K, McCullagh K, Poon E, Davies 14. Griggs RC, Herr BE, Reha A, et al. 23. Fenichel GM, Florence JM, Pestronk KE. Muscular dystrophies related to Corticosteroids in Duchenne A, et al. Long-term benefit from the cytoskeleton/nuclear envelope. muscular dystrophy: major prednisone therapy in Duchenne Novartis Found Symp. 2005;264:98–111; variations in practice. Muscle Nerve. muscular dystrophy. Neurology. discussion 112–117, 227–230 2013;48(1):27–31 1991;41(12):1874–1877 6. Kiessling WR, Beckmann R. Serum 15. Birnkrant DJ, Ararat E, Mhanna MJ. 24. Griggs RC, Moxley RT III, Mendell JR, levels of myoglobin and creatine Cardiac phenotype determines survival et al; Clinical Investigation of Duchenne kinase in Duchenne muscular in Duchenne muscular dystrophy. Dystrophy Group. Prednisone in dystrophy. Klin Wochenschr. Pediatr Pulmonol. 2016;51(1):70–76 Duchenne dystrophy. A randomized, 1981;59(7):347–348 16. Buddhe S, Cripe L, Friedland-Little controlled trial defining the time 7. Desguerre I, Mayer M, Leturcq F, J, et al. Cardiac management of the course and dose response. Arch Barbet JP, Gherardi RK, Christov C. patient with Duchenne muscular Neurol. 1991;48(4):383–388 Endomysial fibrosis in Duchenne dystrophy. Pediatrics. 2018;142(suppl 25. Manzur AY, Kuntzer T, Pike M, Swan muscular dystrophy: a marker of poor 2):e20180333I​ A. Glucocorticoid corticosteroids outcome associated with macrophage 17. Birnkrant DJ, Bushby K, Bann CM, et al; for Duchenne muscular dystrophy. alternative activation. J Neuropathol DMD Care Considerations Working Cochrane Database Syst Rev. Exp Neurol. 2009;68(7):762–773 Group. Diagnosis and management of 2008;(1):CD003725 8. Klingler W, Jurkat-Rott K, Lehmann- Duchenne muscular dystrophy, part 26. Biggar WD, Harris VA, Eliasoph L, Horn F, Schleip R. The role of fibrosis 2: respiratory, cardiac, bone health, Alman B. Long-term benefits of in Duchenne muscular dystrophy. and orthopaedic management. Lancet deflazacort treatment for boys with Acta Myol. 2012;31(3):184–195 Neurol. 2018;17(4):347–361 Duchenne muscular dystrophy in their 9. Wynn TA. Cellular and molecular 18. Webster C, Silberstein L, Hays second decade. Neuromuscul Disord. mechanisms of fibrosis.J Pathol. AP, Blau HM. Fast muscle fibers 2006;16(4):249–255 2008;214(2):199–210 are preferentially affected in 27. Mavrogeni S, Papavasiliou A, Douskou 10. Ciafaloni E, Fox DJ, Pandya S, et al. Duchenne muscular dystrophy. Cell. M, Kolovou G, Papadopoulou E, Delayed diagnosis in Duchenne 1988;52(4):503–513 Cokkinos DV. Effect of deflazacort on muscular dystrophy: data from the 19. Cea LA, Puebla C, Cisterna BA, et al. cardiac and sternocleidomastoid Muscular Dystrophy Surveillance, Fast skeletal myofibers of mdx muscles in Duchenne muscular Tracking, and Research Network mouse, model of Duchenne muscular dystrophy: a magnetic resonance

Downloaded from www.aappublications.org/news by guest on September 25, 2021 PEDIATRICS Volume 142, number s2, October 2018 S13 imaging study. Eur J Paediatr Neurol. phase 2 study. Lancet Neurol. dystrophy. Ther Adv Neurol Disorder. 2009;13(1):34–40 2014;13(10):987–996 2010;3(6):379–389 28. Koenig M, Beggs AH, Moyer M, et al. 38. Flanigan KM, Voit T, Rosales XQ, et al. 47. Hirawat S, Welch EM, Elfring GL, The molecular basis for Duchenne Pharmacokinetics and safety of single et al. Safety, tolerability, and versus Becker muscular dystrophy: doses of drisapersen in non-ambulant pharmacokinetics of PTC124, a correlation of severity with type subjects with Duchenne muscular nonaminoglycoside nonsense mutation of deletion. Am J Hum Genet. dystrophy: results of a double-blind suppressor, following single- and 1989;45(4):498–506 randomized clinical trial. Neuromuscul multiple-dose administration to 29. Monaco AP, Bertelson CJ, Liechti-Gallati Disord. 2014;24(1):16–24 healthy male and female adult volunteers. J Clin Pharmacol. S, Moser H, Kunkel LM. An explanation 39. Mendell JR, Goemans N, Lowes LP, et al; 2007;47(4):430 444 for the phenotypic differences Eteplirsen Study Group and Telethon – between patients bearing partial Foundation DMD Italian Network. 48. Mullard A. EMA reconsiders ‘read- deletions of the DMD locus. Genomics. Longitudinal effect of eteplirsen versus through’ drug against Duchenne 1988;2(1):90–95 historical control on ambulation in muscular dystrophy following appeal. 30. Abbs S, Tuffery-Giraud S, Bakker E, Duchenne muscular dystrophy. Ann Nat Biotechnol. 2014;32(8):706 Ferlini A, Sejersen T, Mueller CR. Best Neurol. 2016;79(2):257–271 49. McDonald CM, Campbell C, Torricelli practice guidelines on molecular 40. Mendell JR, Rodino-Klapac LR, Sahenk RE, et al; Clinical Evaluator Training diagnostics in Duchenne/Becker Z, et al; Eteplirsen Study Group. Group; ACT DMD Study Group. muscular dystrophies. Neuromuscul Eteplirsen for the treatment of Ataluren in patients with nonsense Disord. 2010;20(6):422–427 Duchenne muscular dystrophy. mutation Duchenne muscular 31. Cunniff C, Andrews J, Meaney FJ, Ann Neurol. 2013;74(5):637–647 dystrophy (ACT DMD): a multicentre, randomised, double-blind, placebo- et al. Mutation analysis in a 41. Aar tsma-Rus A, Fokkema I, controlled, phase 3 trial. Lancet. population-based cohort of Verschuuren J, et al. Theoretic 2017;390(10101):1489 1498 boys with Duchenne or Becker applicability of antisense-mediated – muscular dystrophy. J Child Neurol. exon skipping for Duchenne muscular 50. Love DR, Byth BC, Tinsley JM, Blake DJ, 2009;24(4):425–430 dystrophy mutations. Hum Mutat. Davies KE. Dystrophin and dystrophin- 32. Ashton EJ, Yau SC, Deans ZC, Abbs SJ. 2009;30(3):293–299 related proteins: a review of protein and RNA studies. Neuromuscul Disord. Simultaneous mutation scanning for 42. Food and Drug Administration. FDA 1993;3(1):5–21 gross deletions, duplications and point news release: FDA grants accelerated mutations in the DMD gene. Eur J Hum approval to first drug for Duchenne 51. Tinsley JM, Blake DJ, Roche Genet. 2008;16(1):53–61 muscular dystrophy. 2016. Available A, et al. Primary structure of 33. Willcocks RJ, Arpan IA, Forbes SC, et al. at: https://​www.​fda.​gov/​NewsEvents/​ dystrophin-related protein. Nature. Longitudinal measurements of MRI- Newsroom/​PressAnnouncement​s/​ 1992;360(6404):591–593 T2 in boys with Duchenne muscular ucm521263.htm.​ Accessed November 8, 52. Tinsley JM, Blake DJ, Pearce M, Knight dystrophy: effects of age and disease 2017 AE, Kendrick-Jones J, Davies KE. progression. Neuromuscul Disord. 43. Bladen CL, Salgado D, Monges S, et al. Dystrophin and related proteins. Curr 2014;24(5):393–401 The TREAT-NMD DMD global database: Opin Genet Dev. 1993;3(3):484–490 34. Willcocks RJ, Rooney WD, Triplett WT, et al. analysis of more than 7,000​ Duchenne 53. Tinsley JM, Davies KE. Utrophin: Multicenter prospective longitudinal muscular dystrophy mutations. Hum a potential replacement for study of magnetic resonance Mutat. 2015;36(4):395–402 dystrophin? Neuromuscul Disord. biomarkers in a large Duchenne 44. Mendell JR, Buzin CH, Feng J, 1993;3(5–6):537–539 muscular dystrophy cohort. Ann et al. Diagnosis of Duchenne 54. De Moor O, Dorgan CR, Johnson Neurol. 2016;79(4):535–547 dystrophy by enhanced detection PD, et al. Discovery and SAR 35. Aar tsma-Rus A, van Ommen GJ. of small mutations. Neurology. of 2-arylbenzotriazoles and Antisense-mediated exon skipping: 2001;57(4):645–650 2-arylindazoles as potential a versatile tool with therapeutic 45. Finkel RS. Read-through strategies for treatments for Duchenne muscular and research applications. RNA. suppression of nonsense mutations dystrophy. Bioorg Med Chem Lett. 2007;13(10):1609–1624 in Duchenne/ Becker muscular 2011;21(16):4828–4831 36. Aar tsma-Rus A. Overview on dystrophy: aminoglycosides and 55. Acharyya S, Villalta SA, Bakkar N, et al. AON design. Methods Mol Biol. ataluren (PTC124). J Child Neurol. Interplay of IKK/NF-kappaB signaling in 2012;867:117–129 2010;25(9):1158–1164 macrophages and myofibers promotes 37. Voit T, Topaloglu H, Straub V, et al. 46. Malik V, Rodino-Klapac LR, Viollet L, muscle degeneration in Duchenne Safety and efficacy of drisapersen for Mendell JR. Aminoglycoside-induced muscular dystrophy. J Clin Invest. the treatment of Duchenne muscular mutation suppression (stop codon 2007;117(4):889–901 dystrophy (DEMAND II): an exploratory, readthrough) as a therapeutic 56. Wynn TA, Barron L, Thompson RW, randomised, placebo-controlled strategy for Duchenne muscular et al. Quantitative assessment of

Downloaded from www.aappublications.org/news by guest on September 25, 2021 S14 LEIGH et al macrophage functions in repair 65. Mendell JR, Engel WK, Derrer EC. 75. Mendell JR, Rodino-Klapac L, and fibrosis. Curr Protoc Immunol. Duchenne muscular dystrophy: Sahenk Z, et al. Gene therapy for 2011;Chapter 14:Unit14.22 functional ischemia reproduces muscular dystrophy: lessons learned 57. Buyse GM, Voit T, Schara U, et al; its characteristic lesions. Science. and path forward. Neurosci Lett. DELOS Study Group. Efficacy of 1971;172(3988):1143–1145 2012;527(2):90–99 idebenone on respiratory function 66. Mendell JR, Engel WK, Derrer 76. Asokan A, Schaffer DV, Samulski in patients with Duchenne muscular EC. Increased plasma enzyme RJ. The AAV vector toolkit: poised at dystrophy not using glucocorticoids concentrations in rats with functional the clinical crossroads. Mol Ther. (DELOS): a double-blind randomised ischaemia of muscle provide 2012;20(4):699–708 placebo-controlled phase 3 trial. a possible model of Duchenne 77. Mingozzi F, High KA. Therapeutic in Lancet. 2015;385(9979):1748–1757 muscular dystrophy. Nature. vivo gene transfer for genetic disease 58. Welch RD, Billon C, Valfort AC, Burris TP, 1972;239(5374):522–524 using AAV: progress and challenges. Flaveny CA. Pharmacological inhibition 67. Asai A, Sahani N, Kaneki M, Ouchi Y, Nat Rev Genet. 2011;12(5):341–355 of REV-ERB stimulates differentiation, Martyn JA, Yasuhara SE. Primary role 78. Decrouy A, Renaud JM, Davis HL, inhibits turnover and reduces fibrosis of functional ischemia, quantitative Lunde JA, Dickson G, Jasmin BJ. Mini- in dystrophic muscle. Sci Rep. evidence for the two-hit mechanism, dystrophin gene transfer in mdx4cv 2017;7(1):17142 and phosphodiesterase-5 inhibitor diaphragm muscle fibers increases 59. Acuña MJ, Salas D, Córdova-Casanova therapy in mouse muscular dystrophy. sarcolemmal stability. Gene Ther. PLoS One. 2007;2(8):e806 A, et al. Blockade of Bradykinin 1997;4(5):401–408 receptors worsens the dystrophic 68. Leung DG, Herzka DA, Thompson WR, 79. Li S, Kimura E, Ng R, et al. A highly phenotype of mdx mice: differential et al. Sildenafil does not improve functional mini-dystrophin/GFP fusion effects for B1 and B2 receptors. cardiomyopathy in Duchenne/Becker gene for cell and gene therapy studies J Cell Commun Signal. 2018;12(3): muscular dystrophy. Ann Neurol. of Duchenne muscular dystrophy. Hum 589–601 2014;76(4):541–549 Mol Genet. 2006;15(10):1610–1622 60. Andreetta F, Bernasconi P, Baggi F, 69. Victor RG, Sweeney HL, Finkel R, et al; 80. Zhang Y, Yue Y, Li L, et al. Dual AAV et al. Immunomodulation of TGF-beta Tadalafil DMD Study Group. A phase therapy ameliorates exercise-induced 1 in mdx mouse inhibits connective 3 randomized placebo-controlled muscle injury and functional ischemia tissue proliferation in diaphragm but trial of tadalafil for Duchenne in murine models of Duchenne increases inflammatory response: muscular dystrophy. Neurology. muscular dystrophy. Hum Mol Genet. implications for antifibrotic therapy. 2017;89(17):1811–1820 2013;22(18):3720–3729 J Neuroimmunol. 2006;175(1–2):77–86 70. McPherron AC, Lawler AM, Lee SJ. 81. Mendell JR, Campbell K, Rodino-Klapac 61. Morales MG, Cabrera D, C spedes C, é Regulation of skeletal muscle L, et al. Dystrophin immunity in et al. Inhibition of the angiotensin- mass in mice by a new TGF-beta Duchenne s muscular dystrophy. converting enzyme decreases ’ superfamily member. Nature. N Engl J Med. 2010;363(15):1429 1437 skeletal muscle fibrosis in – 1997;387(6628):83–90 dystrophic mice by a diminution 82. Rodino-Klapac LR, Janssen PM, in the expression and activity of 71. Wagner KR, McPherron AC, Winik N, Montgomery CL, et al. A translational connective tissue growth factor Lee SJ. Loss of myostatin attenuates approach for limb vascular delivery (CTGF/CCN-2). Cell Tissue Res. severity of muscular dystrophy in mdx of the micro-dystrophin gene without 2013;353(1):173–187 mice. Ann Neurol. 2002;52(6):832–836 high volume or high pressure for treatment of Duchenne muscular 62. Bish LT, Yarchoan M, Sleeper MM, 72. Wagner KR, Fleckenstein JL, dystrophy. J Transl Med. 2007;5:45 et al. Chronic losartan administration Amato AA, et al. A phase I/II trial reduces mortality and preserves of MYO-029 in adult subjects with 83. Bengtsson NE, Hall JK, Odom GL, cardiac but not skeletal muscle muscular dystrophy. Ann Neurol. et al. Muscle-specific CRISPR/Cas9 function in dystrophic mice. 2008;63(5):561–571 dystrophin gene editing ameliorates PLoS One. 2011;6(6):e20856 73. Raman SV, Hor KN, Mazur W, et al. pathophysiology in a mouse model for Duchenne muscular dystrophy. Nat 63. Spurney CF, Sali A, Guerron AD, et al. Eplerenone for early cardiomyopathy Commun. 2017;8:14454 Losartan decreases cardiac muscle in Duchenne muscular dystrophy: a fibrosis and improves cardiac randomised, double-blind, placebo- 84. Nelson CE, Hakim CH, Ousterout DG, function in dystrophin-deficient mdx controlled trial. Lancet Neurol. et al. In vivo genome editing improves mice. J Cardiovasc Pharmacol Ther. 2015;14(2):153–161 muscle function in a mouse model 2011;16(1):87–95 74. Raman SV, Hor KN, Mazur W, et al. of Duchenne muscular dystrophy. Science. 2016;351(6271):403–407 64. Bettica P, Petrini S, D’Oria V, et al. Eplerenone for early cardiomyopathy Histological effects of givinostat in Duchenne muscular dystrophy: 85. Tabebordbar M, Zhu K, Cheng JKW, et al. in boys with Duchenne muscular results of a two-year open-label In vivo gene editing in dystrophic dystrophy. Neuromuscul Disord. extension trial. Orphanet J Rare Dis. mouse muscle and muscle stem cells. 2016;26(10):643–649 2017;12(1):39 Science. 2016;351(6271):407–411

Downloaded from www.aappublications.org/news by guest on September 25, 2021 PEDIATRICS Volume 142, number s2, October 2018 S15 86. Long C, McAnally JR, Shelton JM, 88. Flanigan KM, Ceco E, Lamar KM, et al; Group Duchenne Natural History Study. Mireault AA, Bassel-Duby R, Olson EN. United Dystrophinopathy Project. LTBP4 Ann Neurol. 2015;77(4):684–696 Prevention of muscular dystrophy genotype predicts age of ambulatory 90. Swaggar t KA, Demonbreun AR, Vo AH, in mice by CRISPR/Cas9-mediated loss in Duchenne muscular dystrophy. et al. Annexin A6 modifies muscular editing of germline DNA. Science. Ann Neurol. 2013;73(4):481–488 dystrophy by mediating sarcolemmal 2014;345(6201):1184–1188 89. Bello L, Kesari A, Gordish-Dressman repair. Proc Natl Acad Sci USA. 87. Long C, Amoasii L, Mireault AA, et al. H, et al; Cooperative International 2014;111(16):6004–6009 Postnatal genome editing partially Neuromuscular Research Group 91. Swaggar t KA, McNally EM. Modifiers restores dystrophin expression in a Investigators. Genetic modifiers of heart and muscle function: where mouse model of muscular dystrophy. of ambulation in the Cooperative genetics meets physiology. Exp Physiol. Science. 2016;351(6271):400–403 International Neuromuscular Research 2014;99(4):621–626

Downloaded from www.aappublications.org/news by guest on September 25, 2021 S16 LEIGH et al Neurology Care, Diagnostics, and Emerging Therapies of the Patient With Duchenne Muscular Dystrophy Fawn Leigh, Alessandra Ferlini, Doug Biggar, Katharine Bushby, Richard Finkel, Lauren P. Morgenroth and Kathryn R. Wagner Pediatrics 2018;142;S5 DOI: 10.1542/peds.2018-0333C

Updated Information & including high resolution figures, can be found at: Services http://pediatrics.aappublications.org/content/142/Supplement_2/S5 References This article cites 89 articles, 11 of which you can access for free at: http://pediatrics.aappublications.org/content/142/Supplement_2/S5# BIBL Permissions & Licensing Information about reproducing this article in parts (figures, tables) or in its entirety can be found online at: http://www.aappublications.org/site/misc/Permissions.xhtml Reprints Information about ordering reprints can be found online: http://www.aappublications.org/site/misc/reprints.xhtml

Downloaded from www.aappublications.org/news by guest on September 25, 2021 Neurology Care, Diagnostics, and Emerging Therapies of the Patient With Duchenne Muscular Dystrophy Fawn Leigh, Alessandra Ferlini, Doug Biggar, Katharine Bushby, Richard Finkel, Lauren P. Morgenroth and Kathryn R. Wagner Pediatrics 2018;142;S5 DOI: 10.1542/peds.2018-0333C

The online version of this article, along with updated information and services, is located on the World Wide Web at: http://pediatrics.aappublications.org/content/142/Supplement_2/S5

Data Supplement at: http://pediatrics.aappublications.org/content/suppl/2018/09/23/peds.2018-0333C.DCSupplemental

Pediatrics is the official journal of the American Academy of Pediatrics. A monthly publication, it has been published continuously since 1948. Pediatrics is owned, published, and trademarked by the American Academy of Pediatrics, 345 Park Avenue, Itasca, Illinois, 60143. Copyright © 2018 by the American Academy of Pediatrics. All rights reserved. Print ISSN: 1073-0397.

Downloaded from www.aappublications.org/news by guest on September 25, 2021