You are viewing a javascript disabled version of the site. Please enable Javascript for this site to function properly.
Go to headerGo to navigationGo to searchGo to contentsGo to footer
In content section. Select this link to jump to navigation

Draft Guidance for Industry Duchenne Muscular Dystrophy, Becker Muscular Dystrophy, and Related Dystrophinopathies – Developing Potential Treatments for the Entire Spectrum of Disease

Abstract

Background:

Duchenne muscular dystrophy (DMD) and related dystrophinopathies are neuromuscular conditions with great unmet medical needs that require the development of effective medical treatments.

Objective:

To aid sponsors in clinical development of drugs and therapeutic biological products for treating DMD across the disease spectrum by integrating advancements, patient registries, natural history studies, and more into a comprehensive guidance.

Methods:

This guidance emerged from collaboration between the FDA, the Duchenne community, and industry stakeholders. It entailed a structured approach, involving multiple committees and boards. From its inception in 2014, the guidance underwent revisions incorporating insights from gene therapy studies, cardiac function research, and innovative clinical trial designs.

Results:

The guidance provides a deeper understanding of DMD and its variants, focusing on patient engagement, diagnostic criteria, natural history, biomarkers, and clinical trials. It underscores patient-focused drug development, the significance of dystrophin as a biomarker, and the pivotal role of magnetic resonance imaging in assessing disease progression. Additionally, the guidance addresses cardiomyopathy’s prominence in DMD and the burgeoning field of gene therapy.

Conclusions:

The updated guidance offers a comprehensive understanding of DMD, emphasizing patient-centric approaches, innovative trial designs, and the importance of biomarkers. The focus on cardiomyopathy and gene therapy signifies the evolving realm of DMD research. It acts as a crucial roadmap for sponsors, potentially leading to improved treatments for DMD.

EXECUTIVE SUMMARY

This draft guidance represents an update of the first draft FDA guidance initially composed by a disease community, with input from industry, sponsors, academia, and the Duchenne muscular dystrophy patient community. When finalized, it should represent the Food and Drug Administration’s (FDA’s) current thinking on this topic. It does not create or confer any rights for or on any person and does not operate to bind FDA or the public. You can use an alternative approach if the approach satisfies the requirements of the applicable statutes and regulations. If you want to discuss an alternative approach, contact the FDA staff responsible for implementing this guidance. If you cannot identify the appropriate FDA staff, call the appropriate number listed on the title page of this guidance.

INTRODUCTION

The purpose of this guidance is to assist sponsors in the clinical development of medical products (i.e., human drugs and therapeutic biological products) for the treatment of individuals with Duchenne muscular dystrophy (DMD) and related dystrophinopathies over the entire spectrum of the disease.

This updated guidance is the result of the first collaboration between the FDA and a disease-specific community in their respective disease area. The FDA invited the Duchenne community (including patients, parents and caregivers, clinicians, academic experts, and industry) to develop the earlier version of the guidance as provided under FDA’s interpretation of Good Guidance Practice provisions.

The Duchenne muscular dystrophy community then embarked on an unprecedented journey to develop a draft guidance for industry to propose to the U.S. Food and Drug Administration (FDA) [1]. The push stemmed from a perceived inadequacy of existing draft guidelines. To remedy this, patient advocates mobilized over 200 community members, clinicians, academics, researchers, and other experts for the process of drafting the guidance. The strategy involved a structured Core Support Team, Steering Committee, expert working groups, and a Community Advisory Board. These entities ensured comprehensive desk reviews, transparent deliberations, and community representation. The guidance touched on pivotal issues, such as the willingness of the Duchenne community to accept potential treatment risks, diagnostic challenges, and innovative clinical trial designs. After meticulous development and revisions, the guidance was submitted to the FDA, emphasizing the community’s urgency.

Upon receipt of the first iteration of the guidance on June 25, 2014, the FDA opened a docket and held further meetings with the DMD community and other experts, leading to revisions based upon regulatory and statutory requirements and additional published data, released in June 2015 (see https://www.parentprojectmd.org/wp-content/uploads/2021/07/2014_Community_Guidance.pdf).

These activities provided the impetus and laid the groundwork for the FDA to develop its own streamlined guidance for industry on DMD and related dystrophinopathies— the first for a specific rare disease— focused specifically on the clinical trial process. This guidance was finalized in February 2018 (refer to https://www.fda.gov/media/92233/download) [2].

This process fortified relationships between the Duchenne community, FDA, and sponsors, fostering trust and enhancing credibility. The initiative not only spotlighted the patient’s perspective but set a precedent, demonstrating how rare disease communities can actively shape the direction of potential treatments and research.

A. Revision of the guidance

Since that time, there have been numerous advances in DMD, including an increase in gene therapy studies and a growing recognition of the need to focus on the deterioration of cardiac function as a separate process from loss of skeletal muscle function. There are updated care considerations guidelines that include the care of adult patients, progress in the FDA’s approach to patient engagement and preference studies, genotype / phenotype correlations and disease progression models, as well as new more sensitive outcome measures. In addition, there was also a need to expand the guidance to include considerations specific to development of treatments for Becker muscular dystrophy (BMD) and other dystrophinopathies.

After dialogues with the CDER and CBER divisions of the FDA, a consensus emerged that it was critical that all of the new knowledge be captured in an update to a patient-led guidance document focused on supporting the developing of treatments for DMD, BMD and related dystrophinopathies. In response, the community marshaled its resources, enlisting a technical management team to ensure project management, strategy formulation, editorial support, and to facilitate the steering committee’s oversight of the guidance development activities.

The revision of the guidance was a meticulous process, leveraging the expertise of leading specialists from academia, industry, government research, regulatory organizations, and the patient community. The working groups were composed of topic experts, community advocates, patients, and caregivers, engaged in regular Zoom calls to dissect and enhance the existing framework. They embarked on a thorough examination of the initial guidance, discerning elements that remained pertinent and identifying areas requiring updates.

A rigorous desk review was conducted, compiling and scrutinizing relevant epidemiological, basic research, and clinical data, with references to evidence substantiated in peer-reviewed articles and regulatory documents. This exercise was pivotal in unearthing key findings, addressing challenges faced by industry engaged in dystrophinopathy research, and forging consensus while shedding light on topics that demanded further exploration.

Central to these discussions was the integration of patient and caregiver perspectives, ensuring that the Duchenne community’s voice echoed throughout the document. The working groups were instrumental in steering the technical writer, meticulously reviewing draft iterations, and harnessing Google Docs for real-time, collaborative editing. Moreover, the process entailed a thorough consideration of feedback from both community and the pharmaceutical advisory boards, which was necessary for transparency and collective input.

Recognizing the distinct voices within the guidance reflects the multifaceted nature of the endeavor. Each working group lent its unique tone to the narrative: some sections adopted a more regulatory cadence, resonating with the precision required for policy and compliance, while others embraced an academic tone, allowing for a deeper exploration of the nuances inherent in natural history data, and the development of exploratory outcome measures and novel trial designs. This diversity in expression enriches the guidance, providing a spectrum of perspectives that mirror the complexity and depth of the field itself.

Like the previous community guidance, this updated guidance addresses and expands upon the FDA’s current thinking regarding the consideration that should be given to the patient engagement of the DMD and BMD community. It also reflects the FDA’s appreciation that recent evidence from patient registries, natural history studies, and clinical trial cohorts have updated both the understanding of DMD and BMD natural history and the causes for variability in outcomes. It addresses the selection of endpoints for clinical trials in populations with DMD as well as the manner in which disease modification might be demonstrated. Given the use of dystrophin as a surrogate endpoint marker for the approval of several drugs [3], this document provides up-to-date guidance on the state-of-the-art measurement of the biomarker. This updated guidance also provides similar guidance on the use of magnetic resonance (MR) imaging measures of both skeletal and cardiac muscle and function and encourages sponsors and regulators to consider their use as surrogate endpoints. Finally, the guidance expands beyond the development of pharmacological treatments (drugs) to consider the development of gene therapy products.

In the pursuit of advancing the development of therapies for DMD and related dystrophinopathies, this comprehensive draft guidance presents a level of detail that surpasses the practical constraints of this journal publication’s format. With the objective to disseminate key insights and foundational elements of this guidance, we have distilled its essence into an executive summary tailored for broad accessibility. This overview publication serves to elucidate the most consequential aspects of the guidance, focusing on regulatory matters, the evolving understanding of natural history, innovations in outcome measures and biomarkers, and considerations in clinical trial design. Furthermore, it highlights the community’s concerns, the evolving opportunities in gene therapy, and the need to consider cardiomyopathy in clinical trials. Crucially, it underscores the application of patient engagement in drug development as a necessary scientific and ethical component in our collective effort to meet the therapeutic needs of individuals with a dystrophinopathy. Thus, while the full guidance (provided as an online supplement), remains a comprehensive resource for pharmaceutical sponsors, this executive summary aims to share the elements that inform and drive the development of effective treatments within the wider scientific and medical communities.

GUIDANCE STRUCTURE OVERVIEW

The guidance is structured to provide comprehensive insights into the broad range of topics that sponsors should consider when developing treatments for dystrophinopathies. It is composed of an executive summary, eight detailed sections or chapters, some inclusive of figures and appendices, reflecting the collaborative efforts of various stakeholders.

Executive Summary: It serves as an introduction, detailing the impetus and methodology behind the guidance, FDA’s initial response, and the necessity for a revised update. It encapsulates a brief technical background on dystrophinopathies, summarizes the key considerations identified by the working groups for each section, and concludes with community imperatives and formal directives on the use of FDA guidance.

Main Guidance Sections:

  • A. The Science of Patient Engagement and Patient Experience Assessment: This section delves into the background of patient-focused drug development, existing FDA guidance, and how Duchenne patient experience data related to patient preferences can advance drug development programs.

  • B. Criteria for Diagnosis in the Clinical and Research Settings: Offers a detailed background on classic Duchenne Muscular Dystrophy, including clinical features, genetic confirmation, genotype-phenotype associations, and the role of muscle biopsy.

  • C. The Current Understanding of the Natural History of Duchenne Muscular Dystrophy: Discusses the stages of DMD disease progression, the heterogeneity and predictability of the disease, and natural history across the spectrum of dystrophinopathy.

  • D. Considerations for Outcome Measurement Selection: This segment addresses general comments on outcome measures, specific ones in DMD, including developmental scales, motor measures, pulmonary outcome measures, and considerations for cardiomyopathy.

  • E. Biomarkers in Duchenne Muscular Dystrophy: Provides a general commentary, delves into dystrophin quantification as biomarkers, considerations related to muscle biopsies, and a comprehensive look at non-biopsy-based biomarkers.

  • F. Specific Trial Design and Analysis Issues for Clinical Trials in DMD: Discusses the learnings from past DMD trials, key features of DMD trial design and analysis, the use of models and natural history data, innovations in trial designs, and specific trial considerations in BMD and other dystrophinopathies.

  • G. Cardiomyopathy: This crucial section introduces the topic, provides a background on the natural history of cardiomyopathy in dystrophinopathies, and discusses cardiac assessment, trial designs, potential outcome measures, and concludes with thoughts on the future direction.

  • H. Gene Therapy for DMD and Other Dystrophinopathies: Covers an introduction to gene therapy, background, considerations for chemistry, manufacturing, controls, preclinical and clinical trial considerations, patient engagement, and expedited programs, culminating in an appendix on informed consent in gene therapy trials.

Each of these sections could serve as a standalone FDA guidance in their respective areas, addressing the various facets of dystrophinopathy-related topics. The culmination of these sections presents a holistic and in-depth guide for the advancement of treatment and research in dystrophinopathies.

BACKGROUND ON DYSTROPHINOPATHIES

Dystrophinopathies result from genetic mutations in the DMD gene that decrease the amount of dystrophin protein and/or cause dysfunction of the protein [4]. In association with other proteins, dystrophin protects muscle fibers against the mechanical forces of contraction— in the absence of dystrophin, muscle is prone to damage, and progressive muscle degeneration [5]. Downstream pathologies including inflammation and fibrosis interfere with muscle regeneration and cause loss of ambulation, loss of upper limb function and other movement, orthopedic complications, and, ultimately, respiratory, and cardiac failure.

The most common and generally most severe dystrophinopathy is DMD, with a birth prevalence of about 1 in 3,500 to 6,000 males [6]. DMD causes delay and/or failure to reach developmental milestones, functional losses in the first decade of life, and a loss of independent ambulation before the age of 13 years in the absence of disease-modifying treatment. In nonambulatory boys and young men, there is gradual loss of upper limb and neck functions, so that grooming, toileting, bathing, dressing, and eating become impaired or impossible to perform by oneself— affecting the quality of life of patients, their caregivers, and families [7]. This is accompanied by weakness affecting respiratory muscles and the heart that contributes to decreased respiratory function and cardiomyopathy— and greatly decreased life expectancy. Heart disease is now the most common cause of death in boys and young men with DMD [8, 9].

BMD has later onset of symptoms and slower progression [10]. BMD is characterized by wide interpatient variability in severity, with some patients having a clinical course similar to that observed for DMD, while other patients remain nearly, or in some cases, completely asymptomatic, and cardiac dysfunction may progress more rapidly than for skeletal muscle [11]. The cumulative birth incidence of BMD was once estimated be at least 1 : 18,500 males (pre-genetic confirmation) [12], and more recently, at least 7.2 : 100,000 in a study with genetic confirmation [13] — but incidence and prevalence may vary by population. A small percentage of female carriers may also exhibit a range of muscle symptoms from the full Duchenne phenotype to milder skeletal muscle weakness (see more on related dystrophinopathies in the Diagnosis section) [14].

Over the past decade, patient organizations, academia, and industry have worked together to develop several patient registries, disseminate improved standards of care, and explore clinical outcome measures and biomarkers. This experience and data collection has resulted in a greatly improved understanding of the pathogenesis and the natural history of DMD and BMD, including factors that may lead to variability in the course of the disease.

Natural history studies as well as clinical trials have shown that the use of glucocorticoids and the management of spine deformity, and pulmonary and cardiac dysfunctions have altered the timing of some of the clinical milestones of the disease [7, 15, 16]. But with improved medical management have come new complications, and quality of life often suffers [17]. For instance, adverse events known to be associated with glucocorticoid usage include excessive weight gain, growth inhibition, bone fragility with a high risk of fractures, risk of diabetes, behavioral abnormalities, Cushingoid features, change in pubertal progression, and cataracts [18]. Of particular concern is the issue of weight gain since this can compound the physical limitations of a dystrophic myopathy.

At the time of this update, it should be acknowledged, with gratitude, that there have been some advances in treatment since the previous guidance with an FDA-approved corticosteroid drug, and also several FDA-approved DMD-specific exon-skipping drugs that provide some benefit for individuals with specific DMD mutations. These latter agents were approved based on surrogate marker-evidence [19], and there is increasing evidence of clinical benefit based on longer term observation on treatment [20]. We urge the sponsors, however, to complete the FDA-recommended post-marketing placebo-controlled trials in an expeditious manner, thus, better characterizing the extent of this clinical benefit.

However, these advances in no way reverse the underlying condition. Duchenne is characterized by a progressive, irreversible loss of one function after the other, from the loss of standing from the floor to the loss of ambulation, to the loss of the ability to self-feed, and the inability to breath without assisted ventilation. Once a functional capacity is lost in an individual with DMD, it is gone forever. Death can happen without warning, at any moment, even in younger boys [21]. Complications such as cardiomyopathy commonly cause early death in patients with BMD [22].

There is an urgent unmet need to develop new treatments, especially those that address the underlying cause of dystrophinopathy. With a number of potential therapeutic agents in or entering clinical development, sponsors need formal guidance on how best to demonstrate a treatment’s effectiveness and safety in this rare disease and what sort of effect would be clinically meaningful to patients and their caregivers.

GUIDANCE UPDATE OVERVIEW

This iteration of the draft guidance contains updates to the sections of the first draft guidance. As with the initial draft guidance, the community chose to place the topic of patient engagement at the start of the document, because it was recognized that sponsors should be guided by patient engagement and patient and caregiver preferences from the very start of a product’s clinical development. The diagnosis section follows, to help guide sponsors in the selection of patients, and to prepare for the introduction of newborn screening. This is followed with sections on natural history, outcome measure selection, and a section on biomarkers that has been moved before the updated clinical trials section as biomarkers are increasingly incorporated into these studies and often used as surrogate endpoints.

The following are key considerations in these updated sections.

A. The Science of Patient Engagement and Patient Experience Assessment

(Formerly the Benefit/Risk Assessment section)

Key considerations in this section:

  • Patient-focused drug development (PFDD) has evolved considerably since the 2014 community-led Duchenne Guidance was released, with FDA providing clearer direction via guidance documents on the collection of data related to patient experiences [23, 24].

  • Patient experience data comes in many forms [25, 26] and are intended to provide information about patientsexperiences with DMD and BMD. More data related to BMD patient experiences are needed to inform drug development [27].

  • Patient and caregiver preferences for treatments have been measured and are well documented in the Duchenne community and can inform all stages of drug development [27–35].

  • Preference data has shown that patients and caregivers have similar preferences and that they are willing to accept risk and uncertainty in exchange for therapies aimed at slowing disease progression [30, 33].

  • Sponsors should engage patient groups and FDA on the collection of new patient experience data related to their development programs.

B. Criteria for Diagnosis in the Clinical and Research Settings

This section provides sponsors with an overview on the diagnosis of dystrophinopathies and differs from the section in the earlier guidance in some key areas.

  • The updated section approaches dystrophinopathies as a spectrum of disorders rather than focusing solely on DMD, adding a list of clinical features for typical DMD and those with later-onset of clinical progression.

  • While it is emphasized that genetic testing remains the gold standard for diagnosis, it needs to be considered within the clinical context [7]. Muscle biopsy is usually not required in a clinical setting [36], but often still necessary in the research setting.

  • The discussion on the multiple testing options for genetic confirmation of a dystrophinopathy has been expanded and an algorithm that charts a diagnostic pathway has been added. It also reviews variants of uncertain significance in the DMD gene which have become more common with the advent of next generation sequencing and population screening (expanded carrier screening).

  • Forward-looking statements have been added regarding newborn screening for Duchenne. A nomination for the Recommended Uniform Screening Panel (RUSP) was submitted in 2022. The formal evidence review started in 2023, with a final vote anticipated in 2024. Implementation would enable diagnosis and management during the presymptomatic stage for infants.

Another change in the drafting of this guidance, was to combine the working groups drafting the Natural History, Outcome Measures and Clinical Trials sections into one larger group, as in the previous guidance, the work of each working group often informed the other. In addition, there has been increasing use and acceptance of DMD natural history data both in the development of disease progression models and as real-world evidence of disease progression and natural history, which, in the absence of novel therapies that can augment or potentially replace data from placebo arms by serving as an external comparator group [37, 38].

C. The Current Understanding of the Natural History of Duchenne Muscular Dystrophy

Key considerations in this section:

  • This section provides an updated overview of DMD natural history concepts, including a new schematic illustrating the typical progression of DMD using violin plots to depict the median and range of timing when milestones occur, (e.g., loss of ambulation, loss of standing ability from the floor, etc.), derived from extensive natural history cohorts. It also highlights key outcome measures employed to monitor disease progression across the different stages of disease.

  • Progression models such as the HERCULES Model [39] and the UC Davis Model [17] link events and outcome measures to add granularity to the characterization of disease stage and trajectory. One important feature of these models is the recognition of a brief transitional stage starting during late ambulation where individuals are able to either stand independently or with assistance and can transfer their own weight.

  • There now exists a more comprehensive body of DMD natural history data [16], allowing for a refined characterization an individuals disease course [17] and the sources of heterogeneity that sponsors can take into account when designing clinical trials [40].

D. Outcome Measurement Selection

Key considerations in this section:

  • This section describes outcome measure selection for staging disease, stratifying cohorts, and for monitoring disease progression.

  • Certain outcome measures, including the North Star Ambulatory Assessment, time function tests, 6-minute walk tests, 10-meter walk run tests, and upper limb functional measures can serve as intermediate clinical endpoints or can be used to identify populations of participants at risk of progression and functional losses during the course of a DMD trial [17, 41–45]. By using these outcome measures during participant screening and for stratification, the risk of underpowering a study and failing to reach a conclusive answer regarding a potential therapys effectiveness can be mitigated. While a study might have broad inclusion criteria, stratification can enrich a group that the studys primary prespecified analysis is based on.

  • Recent data highlight specific changes in outcome measures that are clinically meaningful to patients and families at different disease stages [44, 46–49]. The performance measured by certain tools is predictive of progression to disease milestones, potentially serving as intermediate clinical endpoints.

  • In addition to developmental and motor measures, this section reviews the use of pulmonary outcome measures [50], upper limb function measures [45], and activities monitored by digital technologies and wearable devices [51–54]. These tools can track the course of progression during the transitional phase, through loss of ambulation, and through the nonambulatory stages of DMD.

E. Biomarkers in Duchenne Muscular Dystrophy

Key considerations within this section:

  • Dystrophin quantification has been used been used as an informative biomarker to support accelerated approval of several genetic therapies in DMD [3]. A variety of quantification methods have been used for assessing dystrophin; underscoring the potential need for multiple methodologies to accurately represent the expression and biodistribution of protein [55].

  • Sponsors are advised to minimize patient trauma associated with muscle biopsies [56]. It is imperative to have well-defined protocols for the handling and preparing samples to prevent the loss of invaluable tissue [57].

  • There is a robust body of evidence that MR measures are related to patient function [58–62]. These measures are not only predictive of future functional changes but are also suitable for use in both ambulatory [59, 62, 63] and nonambulatory patients [60, 61]. To further substantiate MR measures as potential surrogate endpoints, sponsors are encouraged to incorporate them in their trials.

  • While circulating biomarkers can offer insights into disease progression and response to treatment, there remains a need for further research to connect circulating biomarkers to specific mechanisms of action [64, 65].

F. Specific Trial Design and Analysis Issues for Clinical Trials In DMD

Key considerations within this section:

  • This section contains key learnings from past trials,[66, 67] including the chief finding that baseline disease severity characteristics are better than age as criteria for enrichment of patient trajectories [46, 68, 69].

  • The section describes key considerations in DMD trial design and analysis, including recommendations on concurrent therapy, particularly corticosteroid therapy [17, 70], trial duration, and cohort selection required to measure clinical benefit at different disease stages.

  • The section considers advances in the collection and analysis of natural history data and real-world data and their use in informing the design of clinical trials [37, 40, 71]. Innovative trial designs can also include delayed placebo (or run-in trials, in which natural history data are used in the run-in to the trial) and roll-over trials in order to make trials more efficient and reduce participantsexposure to placebo [38]. For instance, the DMD community has been working on a master protocol for a platform trial that can share placebo patients and reduce the proportion of individuals randomized to placebo.

  • Finally, a brief discussion of clinical trial considerations in BMD and other dystrophinopathies is included. For instance, there is a significant body of natural history data to support the design of clinical trials in BMD and other dystrophinopathies, although decreased disease severity and slower rate of clinical progression may affect endpoint selection and trial duration [72–74].

This updated version of the guidance also contains two new sections, one on cardiomyopathy and the other on gene therapy. Cardiomyopathy is now the leading cause of death among young men with DMD [8], and some sponsors are looking specifically at heart function in patients with dystrophinopathies, and the use of imaging methodologies to monitor pathogenic changes to the heart. The guidance calls on sponsors to gather evidence linking these pathogenic changes to clinical progression to support regulatory acceptance of these imaging biomarkers as surrogate markers.

Finally, at the time of the original guidance drafting, most of gene therapy research was preclinical. Now, with a few years’ worth of data in clinic, the community saw a need to engage with the Center for Biologics Evaluation and Research to develop a section that consolidates the existing FDA guidance on gene therapy and provides specific recommendations on patient considerations unique to DMD and related dystrophinopathies.

G. Cardiomyopathy

Key considerations within this section:

  • DMD-related cardiomyopathy is characterized by fibrofatty replacement of the myocardium, with an extended timeline of cardiac disease progression culminating in full thickness fatty replacement of the myocardium [75–77]. This suggests maximum therapeutic benefit will be garnered only by developing trials focused on BOTH early and later stage disease. A singular focus on trials powered to examine late-stage disease in order to incorporate mortality outcomes may miss an important therapeutic window prior to irreversible, fatty replacement of the myocardium [78].

  • Harmonization of diagnostic evaluation and therapeutics between trial centers is integral to trial design but must be balanced with the need for inclusivity and access. Consensus recommendations regarding potential cardiac biomarkers and their consideration in trial design will not only facilitate effective trial design but would also provide a means to develop a more robust real-world data infrastructure [79]. This infrastructure is currently needed to assess ongoing clinical trials and for future trials, both cardiac and noncardiac.

  • The understanding of cardiac disease progression has evolved as longitudinal, granular cardiac data has emerged over the last decade. These data and the creation of multicenter networks have made cardiac clinical trials in DMD more feasible [80]. Creation of a roadmap to assess effectiveness of cardiac therapies in DMD will further facilitate the timely development of therapies.

H. Gene Therapy for DMD and Other Dystrophinopathies: Approaches, Patient-Centered Considerations, and Development Pathway

Key considerations within this section:

  • This section draws upon existing FDA guidance on gene therapy (GT) and considers how sponsors can apply it to the development of GT products for DMD.

  • Technical challenges for the development of GT products that are unique to dystrophinopathies include the target tissuesboth skeletal muscle and cardiac muscleas well as the size and complexity of DMD gene that a GT would be designed to restore or correct [81–85].

  • Sponsors should consider the implications of the immune responses and safety issues that currently limit the administration, and preclude re-administration, of some of the GT products furthest along in development [86].

  • Priorities for preclinical studies include dose selection so that clinical trials start with a dose expected to have a therapeutic effect, as well as early evaluation of the effects of GT on the heart [87, 88].

  • While well-controlled placebo-controlled studies are recommended for GT products that are not expected to have large, self-evident effects, sponsors are encouraged to discuss novel trial designs with FDA that limit the time or necessity that a trial participant is on placebo.

  • The section includes guidance on corticosteroid treatment prior to and during clinical trials, participant selection criteria and safety considerations including long-term monitoring of GT trial participants.

  • Efficacy endpoints considerations are the same as in trials of non-GT product for DMD. Intermediate clinical endpoints and surrogate endpoints reasonably likely to lead to or predict clinical benefit could be the basis for a GT to be granted accelerated approval. Given the inability to repeat dosing at the present time, there should be some evidence suggestive of clinical benefit, whether through demonstration of high levels of expression of a functional transgene or demonstration of restored expression of the endogenous gene after gene editing, for the proteins produced by gene therapy to be considered a surrogate endpoint meeting the “reasonably likely” standard for accelerated approval. Evidence from other candidate surrogate endpoints (such as imaging) could support an application.

IMPERATIVES AND IMPLICATIONS

Finally, the guidance document concluded with a section on the community’s imperatives for regulators and sponsors in the development of treatments across the spectrum of dystrophinopathies, based upon consultation with our community guidance board consisting of patients, caregivers, and other representatives of the DMD and BMD community (this is not included in the online supplement but can be found at https://www.parentprojectmd.org/wp-content/uploads/2022/10/Dystrophinopathy-Guidan ce-Master_Sept30Final.pdf.

It should be noted that the FDA has acknowledged the concerns expressed by the DMD and BMD community that flexibility be exercised in the review of products for these diseases— recognizing that many patients and caregivers are willing to take greater risks for a treatment that may slow clinical deterioration or delay the loss of functional milestones, each of which is clinically meaningful [2]. The FDA shares the Duchenne and Becker community’s goal to work with industry to get new therapeutic agents onto the market as rapidly and responsibly as possible. This updated community-drafted guidance for industry is but a step towards achieving that goal.

This guidance is intended to serve as a focus for continued discussions among the FDA, the medical industry, sponsors, academic community, the patient and caregiver community, and the public.

FDA’s guidance documents, including this community guidance, do not establish legally enforceable responsibilities. Instead, guidance documents describe the Agency’s current thinking on a topic and should be viewed only as recommendations, unless specific regulatory or statutory requirements are cited. The use of the word should in Agency guidance documents means that something is suggested or recommended, but not required.

ACKNOWLEDGMENTS INCLUDING SOURCES OF SUPPORT

We thank Theodore Smart, BAIS, for writing assistance and manuscript preparation, Peter Kendall, MS, and Lance Sherriff for editing assistance; each received payment for their services from Parent Project Muscular Dystrophy (PPMD), which also convened the drafting of the guidance.

We would also like to thank Voz Advisors, including Pritha Kuchaculla, Gabrielle France, and Katherine Krieger, who provided project management support.

We are grateful for the support of Tim Franson and David Zook of Faegre Drinker, who consulted with us on regulatory strategies. Likewise, our thanks to all the staff at PPMD, in particular, our chief business officer, Kaylan Moitoso, without whose support this project would not have been possible.

PROJECT FUNDING DECLARATION

This project was funded by Parent Project Muscular Dystrophy.

AUTHORS’ DISCLOSURES/CONFLICTS OF INTEREST DECLARATIONS

Eric Camino: Has no conflicts of interest to declare.

Rafael Escandon: Has consulted for Adrenas Therapeutics and BridgeBio Pharma.

Richard S. Finkel: Participated in DMD studies/activities with Capricor, Catabasis, Dyne, Eli Lilly, Italfarmaco, Pfizer, PTC Therapeutics, ReveraGen, Sarepta Therapeutics, and Summit. Dr. Finkel is an Editorial Board member of this journal, but was not involved in the peer review process nor had access to any information regarding its peer-review.

Ryan Fischer: Has no conflicts of interest to declare.

Kevin Flanigan: Has participated in DMD studies, consultancies and other activities with 4D Therapeutics, Abeona Therapeutics, Akashi, Audentes, Biomarin, Dynacure, Eli Lilly, Prosensa, PTC Therapeutics, and Sarepta Therapeutics. Dr. Flanigan is an Editorial Board member of this journal, but was not involved in the peer review process nor had access to any information regarding its peer-review.

Pat Furlong: Has no conflicts of interest to declare.

Rose Juhasz: Has no conflicts of interest to declare.

Craig McDonald: Has participated in DMD studies, consultancies and other activities with Capricor, Catabasis, Edgewise Therapeutics, Epirium Bio, Italfarmaco, Pfizer, PTC Therapeutics, Santhera Pharmaceuticals, and Sarepta Therapeutics. Dr. McDonald is an Editorial Board member of this journal, but was not involved in the peer review process nor had access to any information regarding its peer-review.

Ann S. Martin: Has participated in a DMD advisory board with Sarepta Therapeutics.

H. Lee Sweeney: Has no disclosures relevant to this project.

Chet Villa: Has consulted for PTC Therapeutics, Capricor Therapeutics, and Antisense.

SUPPLEMENTARY MATERIAL

[1] The main body (or text) of the guidance is available in the electronic version of this article: https://dx.doi.org/10.3233/JND-230219.

REFERENCES

[1] 

Furlong P , Bridges JF , Charnas L , Fallon JR , Fischer R , Flanigan KM et al. How a patient advocacy group developed the first proposed draft guidance document for industry for submission to the U.S. Food and Drug Administration. Orphanet J Rare Dis. (2015) ;10: :82.

[2] 

United States Food and Drug Administration (FDA): Center for Drug Evaluation and Research (CDER) and Center for Biologics Evaluation and Research (CBER). Duchenne Muscular Dystrophy and Related Dystrophinopathies: Developing Drugs for Treatment Guidance for Industry. Silver Spring, Maryland: FDA; 2018.

[3] 

Aartsma-Rus A , Corey DR . The 10th Oligonucleotide Therapy Approved: Golodirsen for Duchenne Muscular Dystrophy. Nucleic Acid Ther. (2020) ;30: (2):67–70.

[4] 

Kunkel LM , Hejtmancik JF , Caskey CT , Speer A , Monaco AP , Middlesworth W et al. Analysis of deletions in DNA from patients with Becker and Duchenne muscular dystrophy. NatuRep. (1986) ;322: (6074):73–7.

[5] 

Weller B , Karpati G , Carpenter S . Dystrophin-deficient mdx muscle fibers are preferentially vulnerable to necrosis induced by experimental lengthening contractions. J Neurol Sci. (1990) ;100: (1-2):9–13.

[6] 

Bushby K , Finkel R , Birnkrant DJ , Case LE , Clemens PR , Cripe L et al. Diagnosis and management of Duchenne muscular dystrophy, part diagnosis, and pharmacological and psychosocial management. Lancet Neurol. (2010) ;9: (1):77–93.

[7] 

Birnkrant DJ , Bushby K , Bann CM , Apkon SD , Blackwell A , Brumbaugh D 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):251–67.

[8] 

Cheeran D , Khan S , Khera R , Bhatt A , Garg S , Grodin JL et al. Predictors of Death in Adults With Duchenne Muscular Dystrophy-Associated Cardiomyopathy. J Am Heart Assoc. (2017) ;6: (10).

[9] 

Van Ruiten HJ , Marini Bettolo C , Cheetham T , Eagle M , Lochmuller H , Straub V et al. Why are some patients with Duchenne muscular dystrophy dying young: An analysis of causes of death in North East England. Eur J Paediatr Neurol. (2016) ;20: (6):904–9.

[10] 

Andrews JG , Lamb MM , Conway K , Street N , Westfield C , Ciafaloni E et al. Diagnostic Accuracy of Phenotype Classification in Duchenneand Becker Muscular Dystrophy Using MedicalRecord Data1. J Neuromuscul Dis. (2018) ;5: (4):481–95.

[11] 

Bushby KM , Gardner-Medwin D , Nicholson LV , Johnson MA , Haggerty ID , Cleghorn NJ et al. The clinical, genetic and dystrophin characteristics of Becker muscular dystrophy II. Correlation of phenotype with genetic and protein abnormalities. J Neurol. (1993) ;240: (2):105–12.

[12] 

Bushby KM , Thambyayah M , Gardner-Medwin D . Prevalence and incidence of Becker muscular dystrophy. Lancet. (1991) ;337: (8748):1022–4.

[13] 

Mostacciuolo ML , Miorin M , Pegoraro E , Fanin M , Schiavon F , Vitiello L et al. Reappraisal of the incidence rate of Duchenne and Becker muscular dystrophies on the basis of molecular diagnosis. Neuroepidemiology. (1993) ;12: (6):326–30.

[14] 

Ishizaki M , Kobayashi M , Adachi K , Matsumura T , Kimura E . Female dystrophinopathy: Review of current literature. Neuromuscul Disord.. (2018) ;28: (7):572–81.

[15] 

Birnkrant DJ , Bushby K , Bann CM , Alman BA , Apkon SD , Blackwell A et al. Diagnosis and management of Duchenne muscular dystrophy, part 2: respiratory, cardiac, bone health, and orthopaedic management. Lancet Neurol. (2018) ;17: (4):347–61.

[16] 

Mercuri E , Bönnemann CG , Muntoni F . Muscular dystrophies. Lancet. (2019) ;394: (10213):2025–38.

[17] 

McDonald CM , Henricson EK , Abresch RT , Duong T , Joyce NC , Hu F et al. Long-term effects of glucocorticoids on function, quality of life, and survival in patients with Duchenne muscular dystrophy: a prospective cohort study. Lancet.. (2018) ;391: (10119):451–61.

[18] 

Matthews E , Brassington R , Kuntzer T , Jichi F , Manzur AY . Corticosteroids for the treatment of Duchenne muscular dystrophy. Cochrane Database Syst Rev. (2016) ;2016: (5):Cd003725..

[19] 

United States Food and Drug Administration (FDA) Center For Drug Evaluation and Research Application Number: 206488orig1s000 Summary Review. 2016

[20] 

Clemens PR , Rao VK , Connolly AM , Harper AD , Mah JK , Smith EC et al. Safety, Tolerability, and Efficacy of Viltolarsen in Boys With Duchenne Muscular Dystrophy Amenable to Exon 53 Skipping: A Phase 2 Randomized Clinical Trial. JAMA Neurol. (2020) ;77: (8):982–91.

[21] 

Wahlgren L , Kroksmark AK , Tulinius M , Sofou K . One in five patients with Duchenne muscular dystrophy dies from other causes than cardiac or respiratory failure. Eur J Epidemiol. (2022) ;37: (2):147–56.

[22] 

Cripe LH , Tobias JD . Cardiac considerations in the operativemanagement of the patient with Duchenne or Becker musculardystrophy. Paediatr Anaesth. (2013) ;23: (9):777–84.

[23] 

United States Food and Drug Administration (FDA). Patient-Focused Drug Development: Collecting Comprehensive and Representative Input. Bethesda, Maryland: United States Food and Drug Administration; 2018.

[24] 

United States Food and Drug Administration (FDA). Patient-Focused Drug Development: Methods to Identify What Is Important to Patients. Bethesda, Maryland: FDA; 2022.

[25] 

United States Food and Drug Administration (FDA). Patient Preference Information (PPI) in Medical Device Decision-Making. Silver Spring & Rockland, Maryland, FDA; 2020.

[26] 

United States Food and Drug Administration (FDA) CfDEaRC, Center for Biologics Evaluation and Research (CBER), Center for Devices and Radiological Health (CDRH). Benefit-Risk Assessment for New Drug and Biological Products: Guidance for Industry (DRAFT GUIDANCE). Rockville, MD: FDA; 2021.

[27] 

Hollin IL , Peay H , Fischer R , Janssen EM , Bridges JFP . Engaging patients and caregivers in prioritizing symptomsimpacting quality of life for Duchenne and Becker muscular dystrophy. Qual Life Res.. (2018) ;27: (9):2261–73.

[28] 

Peay HL , Hollin I , Fischer R , Bridges JF . A community-engaged approach to quantifying caregiver preferences for the benefits and risks of emerging therapies for Duchenne muscular dystrophy. Clin Ther. (2014) ;36: (5):624–37.

[29] 

Peay HL , Hollin IL , Bridges JF . Prioritizing Parental Worry Associated with Duchenne Muscular Dystrophy Using Best-Worst Scaling. J Genet Couns. (2016) ;25: (2):305–13.

[30] 

Hollin IL , Peay HL , Apkon SD , Bridges JFP . Patient-centered benefit-risk assessment in duchenne muscular dystrophy. Muscle Nerve. (2017) ;55: (5):626–34.

[31] 

Landrum Peay H , Fischer R , Tzeng JP , Hesterlee SE , Morris C , Strong Martin A et al. Gene therapy as a potential therapeutic option for Duchenne muscular dystrophy: A qualitative preference study of patients and parents. PLoS One. (2019) ;14: (5):e0213649.

[32] 

Paquin RS , Fischer R , Mansfield C , Mange B , Beaverson K , Ganot A et al. Priorities when deciding on participation in early-phase gene therapy trials for Duchenne muscular dystrophy: a best-worst scaling experiment in caregivers and adult patients. Orphanet J Rare Dis. (2019) ;14: (1):102.

[33] 

Peay HL , Fischer R , Mange B , Paquin RS , Smith EC , Sadosky A et al. Patients’ and caregivers’ maximum acceptable risk of death for non-curative gene therapy to treat Duchenne muscular dystrophy. Mol Genet Genomic Med. (2021) ;9: (5):e1664.

[34] 

Parent Project Muscular Dystrophy (PPMD). Patients are Waiting: Messages from Duchenne Muscular Dystrophy Families to the FDA (White Paper) 2018.

[35] 

Parent Project Muscular Dystrophy CAB, Faegre Baker Daniels, The Kith Collective, RTI International, editor. FDA Patient Focused Drug Development (PFDD) Initiative. Compass Meeting Report. March 2018.Washington, D.C.

[36] 

Aartsma-Rus A , Ginjaar IB , Bushby K . The importance of genetic diagnosis for Duchenne muscular dystrophy. J Med Genet. (2016) ;53: (3):145–51.

[37] 

United States Food and Drug Administration (FDA): Center for Biologics Evaluation and Research (CBER) and Center for Drug Evaluation and Research (CDER). Demonstrating Substantial Evidence of Effectiveness for Human Drug and Biological Products; Guidance for Industry: Draft Guidance. Rockville, Maryland: FDA; 2019.

[38] 

United States Food and Drug Administration (FDA): Center for Drug Evaluation and Research (CDER) and Center for Biologics Evaluation and Research (CBER). Rare Diseases: Common Issues in Drug Development. Guidance for Industry: DRAFT GUIDANCE. Revision 1. Silver Spring, Maryland: FDA; 2019.

[39] 

Broomfield JHM , Crowther MJ et al. [on behalf of Project HERCULES]. Project HERCULES: The Challenges of Estimating Multi-State Model Transitions in Rare Diseases: Creating a Natural History Model For Duchenne Muscular Dystrophy (DMD). The 10th European Conference on Rare Diseases (ECRD) 2020; Online2020.

[40] 

Lingineni K , Aggarwal V , Morales JF , Conrado DJ , Corey D , Vong C et al. Development of a model-based clinical trial simulation platform to optimize the design of clinical trials for Duchenne muscular dystrophy. CPT Pharmacometrics Syst Pharmacol. (2022) ;11: (3):318–32.

[41] 

Mayhew AG , Cano SJ , Scott E , Eagle M , Bushby K , Manzur A et al. Detecting meaningful change using the North Star AmbulatoryAssessment in Duchenne muscular dystrophy. Dev Med Child Neurol. (2013) ;55: (11):1046–52.

[42] 

McDonald C SH , Luo X et al. Use of the Six-MinuteWalk Distance (6MWD) Across Duchenne Muscular Dystrophy (DMD) Studies (P3.121). Neurology. 2016;86 P3.121.

[43] 

Goemans N , Vanden Hauwe M , Signorovitch J , Swallow E , Song J . Individualized Prediction of Changes in 6-Minute Walk Distance for Patients with Duchenne Muscular Dystrophy. PLoS One. (2016) ;11: (10):e0164684.

[44] 

Goemans N , Wong B , Van den Hauwe M , Signorovitch J , Sajeev G , Cox D et al. Prognostic factors for changes in the timed 4-stair climb in patients with Duchenne muscular dystrophy, and implications for measuring drug efficacy: A multi-institutional collaboration. PLoS One. (2020) ;15: (6):e0232870.

[45] 

Pane M , Coratti G , Brogna C , Mazzone ES , Mayhew A , Fanelli L et al. Upper limb function in Duchenne muscular dystrophy: 24 month longitudinal data. PLoS One. (2018) ;13: (6):e0199223.

[46] 

Muntoni F , Domingos J , Manzur AY , Mayhew A , Guglieri M , Sajeev G et al. Categorising trajectories and individual item changes of the North Star Ambulatory Assessment in patients with Duchenne muscular dystrophy. PLoS One. (2019) ;14: (9):e0221097.

[47] 

Henricson E , Abresch R , Han JJ , Nicorici A , Goude Keller E , de Bie E et al. The 6-Minute Walk Test and Person-Reported Outcomes in Boys with Duchenne Muscular Dystrophy and Typically Developing Controls: Longitudinal Comparisons and Clinically-Meaningful Changes Over One Year. PLoS Curr. 2013;5.

[48] 

McDonald CM , Henricson EK , Abresch RT , Florence J , Eagle M , Gappmaier E et al. The 6-minute walk test and other clinical endpoints in duchenne muscular dystrophy: reliability, concurrent validity, and minimal clinically important differences from a multicenter study. Muscle Nerve. (2013) ;48: (3):357–68.

[49] 

McDonald CM , Gordish-Dressman H , Henricson EK , Duong T , Joyce NC , Jhawar S et al. Longitudinal pulmonary function testing outcome measures in Duchenne muscular dystrophy: Long-term natural history with and without glucocorticoids. Neuromuscul Disord. (2018) ;28: (11):897–909.

[50] 

Mayer OH . Clinical pulmonary function testing in Duchenne muscular dystrophy. Paediatr Respir Rev. (2019) ;30: :9–12.

[51] 

Servais L , Yen K , Guridi M , Lukawy J , Vissière D , Strijbos P . Stride Velocity 95th Centile: Insights into Gaining Regulatory Qualification of the First Wearable-Derived Digital Endpoint for use in Duchenne Muscular Dystrophy Trials. J Neuromuscul Dis. (2022) ;9: (2):335–46.

[52] 

Veerapandiyan A , Wagner KR , Apkon S , McDonald CM , Mathews KD , Parsons JA et al. The care of patients with Duchenne, Becker, and other muscular dystrophies in the COVID-19 pandemic. Muscle Nerve. (2020) ;62: (1):41–5.

[53] 

Geronimo A , Simmons Z . Evaluation of remote pulmonary function testing in motor neuron disease. Amyotroph Lateral Scler Frontotemporal Degener. (2019) ;20: (5-6):348–55.

[54] 

Contesse MG , Sapp ATL , Apkon SD , Lowes LP , Dalle Pazze L , Leffler MG . Reliability and construct validity of the Duchenne Video Assessment. Muscle Nerve. (2021) ;64: (2):180–9.

[55] 

Aartsma-Rus A , Morgan J , Lonkar P , Neubert H , Owens J , Binks M et al. Report of a TREAT-NMD/World Duchenne Organisation Meeting on Dystrophin Quantification Methodology. J Neuromuscul Dis. (2019) ;6: (1):147–59.

[56] 

Verhaart IEC , Johnson A , Thakrar S , Vroom E , De Angelis F , Muntoni F et al. Muscle biopsies in clinical trials for Duchenne musculardystrophy - Patients’ and caregivers’ perspective. NeuromusculDisord.. (2019) ;29: (8):576–84.

[57] 

Aartsma-Rus A , Muntoni F . 194th ENMC international worksho 3rd ENMC workshop on exon skipping: towards clinical application of antisense-mediated exon skipping for Duchenne muscular dystrophy 8-10 December Naarden, The Netherlands. Neuromuscul Disord. (2013) ;23: (11):934–44.

[58] 

Rooney WD , Berlow YA , Triplett WT , Forbes SC , Willcocks RJ , Wang DJ et al. Modeling disease trajectory in Duchenne muscular dystrophy. Neurology. (2020) ;94: (15):e1622–e33.

[59] 

Ropars J , Gravot F , Ben Salem D , Rousseau F , Brochard S , Pons C . Muscle MRI: A biomarker of disease severity in Duchenne muscular dystrophy? A systematic review. Neurology. (2020) ;94: (3):117–33.

[60] 

Naarding KJ , van der Holst M , van Zwet EW , van de Velde NM , de Groot IJM , Verschuuren J et al. Association of Elbow Flexor MRI Fat Fraction With Loss of Hand-to-Mouth Movement in Patients With Duchenne Muscular Dystrophy. Neurology. (2021) ;97: (17):e1737–e42.

[61] 

Wary C , Azzabou N , Giraudeau C , Le Louër J , Montus M , Voit T et al. Quantitative NMRI and NMRS identify augmented diseaseprogression after loss of ambulation in forearms of boys withDuchenne muscular dystrophy. NMR Biomed. (2015) ;28: (9):1150–62.

[62] 

Barnard AM , Willcocks RJ , Triplett WT , Forbes SC , Daniels MJ , Chakraborty S et al. MR biomarkers predict clinical function in Duchenne muscular dystrophy. Neurology. (2020) ;94: (9):e897–e909.

[63] 

Naarding KJ , Reyngoudt H , van Zwet EW , Hooijmans MT , Tian C , Rybalsky I et al. MRI vastus lateralis fat fraction predicts loss of ambulation in Duchenne muscular dystrophy. Neurology. (2020) ;94: (13):e1386–e94.

[64] 

Hathout Y , Seol H , Han MH , Zhang A , Brown KJ , Hoffman EP . Clinical utility of serum biomarkers in Duchenne muscular dystrophy. Clin Proteomics. (2016) ;13: :9.

[65] 

Hathout Y , Liang C , Ogundele M , Xu G , Tawalbeh SM , Dang UJ et al. Disease-specific and glucocorticoid-responsive serum biomarkers for Duchenne Muscular Dystrophy. Sci Rep. (2019) ;9: (1):12167.

[66] 

Markati T , De Waele L , Schara-Schmidt U , Servais L . Lessons Learned from Discontinued Clinical Developments in Duchenne Muscular Dystrophy. Front Pharmacol. (2021) ;12: :735912.

[67] 

Crow RA , Hart KA , McDermott MP , Tawil R , Martens WB , Herr BE et al. A checklist for clinical trials in rare disease: obstacles and anticipatory actions-lessons learned from the FOR-DMD trial. Trials. (2018) ;19: (1):291.

[68] 

Mercuri E , Signorovitch JE , Swallow E , Song J , Ward SJ . Categorizing natural history trajectories of ambulatory function measured by the 6-minute walk distance in patients with Duchenne muscular dystrophy. Neuromuscul Disord. (2016) ;26: (9):576–83.

[69] 

Fang Y , McDonald CM , Clemens PR , Gordish HD , Illei K , Hoffman EP et al. Modeling Early Heterogeneous Rates of Progression in Boys with Duchenne Muscular Dystrophy. J Neuromuscul Dis. (2023) ;10: (3):349–64.

[70] 

Guglieri M , Bushby K , McDermott MP , Hart KA , Tawil R , Martens WB et al. Effect of Different Corticosteroid Dosing Regimens on Clinical Outcomes in Boys With Duchenne Muscular Dystrophy: A Randomized Clinical Trial. Jama. (2022) ;327: (15):1456–68.

[71] 

United States Food and Drug Administration (FDA). Framework for FDA’s Real-World Evidence Program. FDA; 2018.

[72] 

Bello L , Campadello P , Barp A , Fanin M , Semplicini C , Sorarù G et al. Functional changes in Becker muscular dystrophy: implicationsfor clinical trials in dystrophinopathies. Sci Rep. (2016) ;6: :32439.

[73] 

Clemens PR , Niizawa G , Feng J , Florence J , D’Alessandro AS , Morgenroth LP et al. The CINRG Becker Natural History Study:Baseline characteristics. Muscle Nerve. (2020) ;62: (3):369–76.

[74] 

Barp A , Bello L , Caumo L , Campadello P , Semplicini C , Lazzarotto A et al. Muscle MRI and functional outcome measures in Becker muscular dystrophy. Sci Rep. (2017) ;7: (1):16060.

[75] 

Sanyal SK , Johnson WW , Thapar MK , Pitner SE . An ultrastructural basis for electrocardiographic alterations associated with Duchenne’s progressive muscular dystrophy. Circulation. (1978) ;57: (6):1122–9.

[76] 

Tandon A , Villa CR , Hor KN , Jefferies JL , Gao Z , Towbin JA et al. Myocardial fibrosis burden predicts left ventricular ejection fraction and is associated with age and steroid treatment duration in duchenne muscular dystrophy. J Am Heart Assoc. (2015) ;4: (4).

[77] 

Hor KN , Taylor MD , Al-Khalidi HR , Cripe LH , Raman SV , Jefferies JL et al. Prevalence and distribution of late gadolinium enhancement in a large population of patients with Duchenne muscular dystrophy: effect of age and left ventricular systolic function. J Cardiovasc Magn Reson. (2013) ;15: (1):107.

[78] 

Spurney C , Shimizu R , Morgenroth LP , Kolski H , Gordish-Dressman H , Clemens PR . Cooperative International Neuromuscular Research Group Duchenne Natural History Study demonstrates insufficient diagnosis and treatment of cardiomyopathy in Duchenne muscular dystrophy. Muscle Nerve. (2014) ;50: (2):250–6.

[79] 

Fiuzat M , Hamo CE , Butler J , Abraham WT , DeFilippis EM , Fonarow GC et al. Optimal Background Pharmacological Therapy for Heart Failure Patients in Clinical Trials: JACC Review Topic of the Week. J Am Coll Cardiol. (2022) ;79: (5):504–10.

[80] 

Villa C , Auerbach SR , Bansal N , Birnbaum BF , Conway J , Esteso P et al. Current Practices in Treating Cardiomyopathy and Heart Failure in Duchenne Muscular Dystrophy (DMD): Understanding Care Practices in Order to Optimize DMD Heart Failure Through ACTION. Pediatr Cardiol. (2022) ;43: (5):977–85.

[81] 

Mendell JR , Clark KR . Challenges for gene therapy for muscular dystrophy. Curr Neurol Neurosci Rep. (2006) ;6: (1):47–56.

[82] 

Nance ME , Duan D . Perspective on Adeno-Associated Virus Capsid Modification for Duchenne Muscular Dystrophy Gene Therapy. Hum Gene Ther. (2015) ;26: (12):786–800.

[83] 

Colella P , Ronzitti G , Mingozzi F . Emerging Issues in AAV-Mediated In Vivo Gene Therapy. Mol Ther Methods Clin Dev. (2018) ;8: :87–104.

[84] 

Blankinship MGP , Chamberlain J Muscular Dystrophy Using Adeno-Associated Viral Vectors: Promises and Limitation: In Duchenne Muscular Dystrophy: Advances in Therapeutics. 1st Edition. CRC Press; 2006.

[85] 

Seto JT , Ramos JN , Muir L , Chamberlain JS , Odom GL . Gene replacement therapies for duchenne muscular dystrophy using adeno-associated viral vectors. Curr Gene Ther. (2012) ;12: (3):139–51.

[86] 

United States Food and Drug Administration (FDA). Briefing Document. Food and Drug Administration (FDA) Cellular, Tissue, and Gene Therapies Advisory Committee (CTGTAC) Meeting #70. Toxicity Risks of Adenoassociated Virus (AAV) Vectors for Gene Therapy (GT). September 2-3, 2021.: United States Food and Drug Administration (FDA),; 2021 2021.

[87] 

United States Food and Drug Administration (FDA): Center for Biologics Evaluation and Research (CBER). Human Gene Therapy for Rare Diseases. Guidance for Industry. FDA; 2020.

[88] 

United States Food and Drug Administration (FDA): Center for Biologics Evaluation and Research (CBER). Guidance for Industry: Preclinical Assessment of Investigational Cellular and Gene Therapy Products. FDA; 2013.

[89] 

United States Food and Drug Administration (FDA). CDER Patient-Focused Drug Development: FDA; 2022.

[90] 

United States Food and Drug Administration (FDA): Center for Biologics Evaluation and Research (CBER) Office of Tissues and Advanced Therapies (OTAT). Patient Perspectives on Gene Therapy Products, November 15, 2022. Patient-Focused Drug Development Listening Meeting Summary. FDA; 2023.

[91] 

United States Food and Drug Administration (FDA). Patient-Focused Drug Development Guidance: Methods to Identify What is Important to Patients and Select, Develop or Modify Fit-for-Purpose Clinical Outcome Assessments. Silver Spring, Maryland: FDA; 2018.

[92] 

Medical Device Innovation Consortium (MDIC). Framework for Patient Input in Clinical Trial Design. Washington DC: MDIC; 2018.

[93] 

United States Food and Drug Adminstration (FDA): Center for Drug Evaluation and Research (CDER), Center for Biologics Evaluation and Research (CBER), Center for Devices and Radiological Health (CDRH). Guidance for Industry - Patient-Reported Outcome Measures: Use in Medical Product Development to Support Labeling Claims. Silver Spring & Rockville, MD: FDA; 2009.

[94] 

United States Food and Drug Administration (FDA). Benefit-Risk Assessment for New Drug and Biological Products Guidance for Industry. DRAFT GUIDANCE. Bethesda, Maryland: FDA; 2021.

[95] 

Hollin IL , Caroline Y , Hanson C , Bridges JFP , Peay H . Developing a Patient-Centered Benefit-Risk Survey: A Community-Engaged Process. Value Health. (2016) ;19: (6):751–7.

[96] 

Fischer R , Porter K , Donovan JM , Scavina MT , Armstrong N , Denger B et al. A Mixed-Method Study ExploringPatient-Experienced and Caregiver-Reported Benefits and Side Effects of Corticosteroid Use in Duchenne MuscularDystrophy. J Neuromuscul Dis.. (2023) ;10: (4):593–613.

[97] 

Griggs RC , Mendell JR , Brooke MH , Fenichel GM , Miller JP , Province M et al. Clinical investigation in Duchenne dystrophy: V. Use of creatine kinase and pyruvate kinase in carrier detection. Muscle Nerve. (1985) ;8: (1):60–7.

[98] 

Cyrulnik SE , Fee RJ , De Vivo DC , Goldstein E , Hinton VJ . Delayed developmental language milestones in children with Duchenne’s muscular dystrophy. J Pediatr. (2007) ;150: (5):474–8.

[99] 

Mirski KT , Crawford TO . Motor and cognitive delay in Duchenne muscular dystrophy: implication for early diagnosis. J Pediatr. (2014) ;165: (5):1008–10.

[100] 

Ciafaloni E , Fox DJ , Pandya S , Westfield CP , Puzhankara S , Romitti PA et al. Delayed diagnosis in duchenne muscular dystrophy: data from the Muscular Dystrophy Surveillance, Tracking, and Research Network (MD STARnet). J Pediatr. (2009) ;155: (3):380–5.

[101] 

Holtzer C , Meaney FJ , Andrews J , Ciafaloni E , Fox DJ , James KA et al. Disparities in the diagnostic process of Duchenne and Becker muscular dystrophy. Genet Med. (2011) ;13: (11):942–7.

[102] 

Brooke MH A Clinician’s View of Neuromuscular Diseases, 2nd ed., 2nd ed: Williams & Wilkins; 1986.

[103] 

Koeks Z , Janson AA , Beekman C , Signorelli M , van Duyvenvoorde HA , van den Bergen JC et al. Low dystrophin variability between muscles and stable expression over time in Becker muscular dystrophy using capillary Western immunoassay. Sci Rep. (2021) ;11: (1):5952.

[104] 

Farrokhi V , Walsh J , Palandra J , Brodfuehrer J , Caiazzo T , Owens J et al. Dystrophin and mini-dystrophin quantification by mass spectrometry in skeletal muscle for gene therapy development in Duchenne muscular dystrophy. Gene Ther. 2021.

[105] 

Lee T , Takeshima Y , Kusunoki N , Awano H , Yagi M , Matsuo M et al. Differences in carrier frequency between mothers of Duchenne and Becker muscular dystrophy patients. J Hum Genet. (2014) ;59: (1):46–50.

[106] 

Lin J , Li H , Liao Z , Wang L , Zhang C . Comparison of Carrier and de novo Pathogenic Variants in a Chinese DMD/BMD Cohort. Front Neurol. (2021) ;12: :714677.

[107] 

Thomas S , Conway KM , Fapo O , Street N , Mathews KD , Mann JR et al. Time to diagnosis of Duchenne muscular dystrophy remains unchanged: Findings from the Muscular Dystrophy Surveillance, Tracking, and Research Network, 2000-2015. Muscle Nerve. (2022) ;66: (2):193–7.

[108] 

Willcocks RJ , Rooney WD , Triplett WT , Forbes SC , Lott DJ , Senesac CR et al. Multicenter prospective longitudinal study of magnetic resonance biomarkers in a large duchenne muscular dystrophy cohort. Ann Neurol. (2016) ;79: (4):535–47.

[109] 

Goemans N , van den Hauwe M , Wilson R , van Impe A , Klingels K , Buyse G . Ambulatory capacity and disease progression as measured by the 6-minute-walk-distance in Duchenne muscular dystrophy subjects on daily corticosteroids. Neuromuscul Disord. (2013) ;23: (8):618–23.

[110] 

McDonald CM , Henricson EK , Abresch RT , Florence JM , Eagle M , Gappmaier E et al. The 6-minute walk test and other endpoints in Duchenne muscular dystrophy: longitudinal natural history observations over 48 weeks from a multicenter study. Muscle Nerve. (2013) ;48: (3):343–56.

[111] 

Lynn S , Aartsma-Rus A , Bushby K , Furlong P , Goemans N , De Luca A et al. Measuring clinical effectiveness of medicinal products for the treatment of Duchenne muscular dystrophy. Neuromuscul Disord. (2015) ;25: (1):96–105.

[112] 

Kim J , Jung IY , Kim SJ , Lee JY , Park SK , Shin HI et al. A New Functional Scale and Ambulatory Functional Classification of Duchenne Muscular Dystrophy: Scale Development and Preliminary Analyses of Reliability and Validity. Ann Rehabil Med. (2018) ;42: (5):690–701.

[113] 

Mazzone ES , Coratti G , Sormani MP , Messina S , Pane M , D’Amico A et al. Timed Rise from Floor as a Predictor of Disease Progression in Duchenne Muscular Dystrophy: An Observational Study. PLoS One.e. (2016) ;11: (3):0151445.

[114] 

Jin JB , Carter JC , Sheehan DW , Birnkrant DJ . Cardiopulmonary phenotypic discordance is common in Duchenne muscular dystrophy. Pediatr Pulmonol. (2019) ;54: (2):186–93.

[115] 

McDonald CM DT , Henricson E et al. CINRG Duchenne Natural HistoryStudy: relationship of longitudinal measures of ambulatory timedfunction tests and loss of clinical milestones. Neuromuscular Disorders. (2013) ;23: :752.

[116] 

Mayhew A , Mazzone ES , Eagle M , Duong T , Ash M , Decostre V et al. Development of the Performance of the Upper Limb module for Duchenne muscular dystrophy. Dev Med Child Neurol. (2013) ;55: (11):1038–45.

[117] 

Goemans N , Signorovitch J , Sajeev G , Yao Z , Gordish-Dressman H , McDonald CM et al. Suitability of external controls for drug evaluation in Duchenne muscular dystrophy. Neurology. (1381) ;95: (10):e1381–e91.

[118] 

Smith RA , Sibert JR , Wallace SJ , Harper PS . Early diagnosis and secondary prevention of Duchenne muscular dystrophy. Arch Dis Child. (1989) ;64: (6):787–90.

[119] 

Connolly AM , Florence JM , Cradock MM , Eagle M , Flanigan KM , McDonald CM et al. One year outcome of boys with Duchenne muscular dystrophy using the Bayley-III scales of infant and toddler development. Pediatr Neurol. (2014) ;50: (6):557–63.

[120] 

Connolly AM , Florence JM , Cradock MM , Malkus EC , Schierbecker JR , Siener CA et al. Motor and cognitive assessment of infants and young boys with Duchenne Muscular Dystrophy: results from the Muscular Dystrophy Association DMD Clinical Research Network. Neuromuscul Disord. (2013) ;23: (7):529–39.

[121] 

Connolly AM , Zaidman CM , Golumbek PT , Cradock MM , Flanigan KM , Kuntz NL et al. Twice-weekly glucocorticosteroidsin infants and young boys with Duchenne muscular dystrophy. Muscle Nerve.. (2019) ;59: (6):650–7.

[122] 

Pane M , Scalise R , Berardinelli A , D’Angelo G , Ricotti V , Alfieri P et al. Early neurodevelopmental assessment in Duchenne muscular dystrophy. Neuromuscul Disord. (2013) ;23: (6):451–5.

[123] 

Uttley L , Carlton J , Woods HB , Brazier J . A review of quality of life themes in Duchenne muscular dystrophy for patients and carers. Health Qual Life Outcomes. (2018) ;16: (1):237.

[124] 

Peay HKA , Fischer R , Bronson A , Furlong P Promoting meaningful clinical trial outcome measures for Duchenne muscular dystrophy. Neuromuscular Disorders. Neuromuscular Disorders. 2016;26.

[125] 

Passamano L , Taglia A , Palladino A , Viggiano E , D’Ambrosio P , Scutifero M et al. Improvement of survival in Duchenne Muscular Dystrophy: retrospective analysis of 835 patients. Acta Myol. (2012) ;31: (2):121–5.

[126] 

Humbertclaude V , Hamroun D , Bezzou K , Bérard C , Boespflug-Tanguy O , Bommelaer C et al. Motor and respiratory heterogeneity inDuchenne patients: implication for clinical trials. Eur J PaediatrNeurol. (2012) ;16: (2):149–60.

[127] 

Henricson EK , Abresch RT , Cnaan A , Hu F , Duong T , Arrieta A et al. The cooperative international neuromuscular research group Duchenne natural history study: glucocorticoid treatment preserves clinically meaningful functional milestones and reduces rate of disease progression as measured by manual muscle testing and other commonly used clinical trial outcome measures. Muscle Nerve. (2013) ;48: (1):55–67.

[128] 

Bulman DE , Murphy EG , Zubrzycka-Gaarn EE , Worton RG , Ray PN . Differentiation of Duchenne and Becker muscular dystrophy phenotypes with amino- and carboxy-terminal antisera specific for dystrophin. Am J Hum Genet. (1991) ;48: (2):295–304.

[129] 

Pane M , Mazzone ES , Sormani MP , Messina S , Vita GL , Fanelli L et al. 6 Minute walk test in Duchenne MD patients with different mutations: 12 month changes. PLoS One. (2014) ;9: (1):e83400.

[130] 

Bello L , Morgenroth LP , Gordish-Dressman H , Hoffman EP , McDonald CM , Cirak S . DMD genotypes and loss of ambulation in the CINRG Duchenne Natural History Study. Neurology. (2016) ;87: (4):401–9.

[131] 

de Feraudy Y , Ben Yaou R , Wahbi K , Stalens C , Stantzou A , Laugel V et al. Very Low Residual Dystrophin Quantity Is Associated with Milder Dystrophinopathy. Ann Neurol. (2021) ;89: (2):280–92.

[132] 

Muntoni F , Torelli S , Ferlini A . Dystrophin and mutations: one gene, several proteins, multiple phenotypes. Lancet Neurol. (2003) ;2: (12):731–40.

[133] 

D’Angelo MG , Lorusso ML , Civati F , Comi GP , Magri F , Del Bo R et al. Neurocognitive profiles in Duchenne muscular dystrophy and gene mutation site. Pediatr Neurol. (2011) ;45: (5):292–9.

[134] 

Taylor PJ , Betts GA , Maroulis S , Gilissen C , Pedersen RL , Mowat DR et al. Dystrophin gene mutation location and the risk of cognitive impairment in Duchenne muscular dystrophy. PLoS One. (2010) ;5: (1):e8803.

[135] 

Chesshyre M , Ridout D , Hashimoto Y , Ookubo Y , Torelli S , Maresh K et al. Investigating the role of dystrophin isoform deficiency in motor function in Duchenne muscular dystrophy. J Cachexia Sarcopenia Muscle. (2022) ;13: (2):1360–72.

[136] 

Zambon AA , Ayyar Gupta V , Ridout D , Manzur AY , Baranello G , Trucco F et al. Peak functional ability and age at loss of ambulation in Duchenne muscular dystrophy. Dev Med Child Neurol. (2022) ;64: (8):979–88.

[137] 

Flanigan KM , Ceco E , Lamar KM , Kaminoh Y , Dunn DM , Mendell JR et al. LTBP4 genotype predicts age of ambulatory loss in Duchenne muscular dystrophy. Ann Neurol. (2013) ;73: (4):481–8.

[138] 

Pegoraro E , Hoffman EP , Piva L , Gavassini BF , Cagnin S , Ermani M et al. SPP1 genotype is a determinant of disease severity in Duchenne muscular dystrophy. Neurology. (2011) ;76: (3):219–26.

[139] 

Bello L , Flanigan KM , Weiss RB , Spitali P , Aartsma-Rus A , Muntoni F et al. Association Study of Exon Variants in the NF-κB and TGFβ Pathways Identifies CD40 as a Modifier of Duchenne Muscular Dystrophy. Am J Hum Genet. (2016) ;99: (5):1163–71.

[140] 

Spitali P , Zaharieva I , Bohringer S , Hiller M , Chaouch A , Roos A et al. TCTEX1D1 is a genetic modifier of disease progression in Duchenne muscular dystrophy. Eur J Hum Genet. (2020) ;28: (6):815–25.

[141] 

Weiss RB , Vieland VJ , Dunn DM , Kaminoh Y , Flanigan KM . Long-range genomic regulators of THBS1 and LTBP4 modify disease severity in duchenne muscular dystrophy. Ann Neurol. (2018) ;84: (2):234–45.

[142] 

Kelley EF , Cross TJ , McDonald CM , Hoffman EP , Bello L . Influence of β(2) adrenergic receptor genotype on longitudinal measures of forced vital capacity in patients with Duchenne muscular dystrophy. Neuromuscul Disord. (2022) ;32: (2):150–8.

[143] 

Ricotti V , Ridout DA , Scott E , Quinlivan R , Robb SA , Manzur AY et al. Long-term benefits and adverse effects of intermittent versus daily glucocorticoids in boys with Duchenne muscular dystrophy. J Neurol Neurosurg Psychiatry. (2013) ;84: (6):698–705.

[144] 

Marden JR , Freimark J , Yao Z , Signorovitch J , Tian C , Wong BL . Real-world outcomes of long-term prednisone and deflazacort use in patients with Duchenne muscular dystrophy: experience at a single, large care center. J Comp Eff Res. (2020) ;9: (3):177–89.

[145] 

McDonald CM , Sajeev G , Yao Z , McDonnell E , Elfring G , Souza M et al. Deflazacort vs prednisone treatment for Duchenne muscular dystrophy: A meta-analysis of disease progression rates in recent multicenter clinical trials. Muscle Nerve. (2020) ;61: (1):26–35.

[146] 

Shieh PB , McIntosh J , Jin F , Souza M , Elfring G , Narayanan S et al. Deflazacort versus prednisone/prednisolone for maintaining motor function and delaying loss of ambulation: A post HOC analysis from the ACT DMD trial. Muscle Nerve. (2018) ;58: (5):639–45.

[147] 

Hoffman EP , Schwartz BD , Mengle-Gaw LJ , Smith EC , Castro D , Mah JK et al. Vamorolone trial in Duchenne muscular dystrophy shows dose-related improvement of muscle function. Neurology. (1312) ;93: (13):e1312–e23.

[148] 

Mah JK , Clemens PR , Guglieri M , Smith EC , Finkel RS , Tulinius M et al. Efficacy and Safety of Vamorolone in Duchenne Muscular Dystrophy: A 30-Month Nonrandomized Controlled Open-Label Extension Trial. JAMA Netw Open. (2022) ;5: (1):e2144178.

[149] 

Bushby K , Finkel R , Birnkrant DJ , Case LE , Clemens PR , Cripe L et al. Diagnosis and management of Duchenne muscular dystrophy, part 2: implementation of multidisciplinary care. Lancet Neurol. (2010) ;9: (2):177–89.

[150] 

Duong T , Canbek J , Fernandez-Fernandez A , Henricson E , Birkmeier M , Siener C et al. Knee Strength and Ankle Range of Motion Impacts on Timed Function Tests in Duchenne Muscular Dystrophy: In the Era of Glucocorticoids. J Neuromuscul Dis. (2022) ;9: (1):147–59.

[151] 

Jansen M , van Alfen N , Geurts AC , de Groot IJ . Assisted bicycletraining delays functional deterioration in boys with Duchennemuscular dystrophy: the randomized controlled trial “no use isdisuse”. Neurorehabil Neural Repair. (2013) ;27: (9):816–27.

[152] 

Eagle M , Bourke J , Bullock R , Gibson M , Mehta J , Giddings D et al. Managing Duchenne muscular dystrophy–the additive effect of spinal surgery and home nocturnal ventilation in improving survival. Neuromuscul Disord. (2007) ;17: (6):470–5.

[153] 

Finder JD , Birnkrant D , Carl J , Farber HJ , Gozal D , Iannaccone ST et al. Respiratory care of the patient with Duchenne muscular dystrophy: ATS consensus statement. Am J Respir Crit Care Med. (2004) ;170: (4):456–65.

[154] 

Rall S , Grimm T . Survival in Duchenne muscular dystrophy. Acta Myol. (2012) ;31: (2):117–20.

[155] 

Duboc D , Meune C , Pierre B , Wahbi K , Eymard B , Toutain A et al. Perindopril preventive treatment on mortality in Duchenne muscular dystrophy: 10 years’ follow-u Am Heart J. (2007) ;154: (3):596–602.

[156] 

Thayer S , Bell C , McDonald CM . The Direct Cost of Managing a Rare Disease: Assessing Medical and Pharmacy Costs Associated with Duchenne Muscular Dystrophy in the United States. J Manag Care Spec Pharm. (2017) ;23: (6):633–41.

[157] 

Kamdar F , Garry DJ . Dystrophin-Deficient Cardiomyopathy. J Am Coll Cardiol. (2016) ;67: (21):2533–46.

[158] 

Hoffman EP , Kunkel LM , Angelini C , Clarke A , Johnson M , Harris JB . Improved diagnosis of Becker muscular dystrophy by dystrophin testing. Neurology. (1989) ;39: (8):1011–7.

[159] 

Flanigan KM . Duchenne and Becker muscular dystrophies. Neurol Clin. (2014) ;32: (3):671–88, viii.

[160] 

Finsterer J , Stöllberger C . Cardiac involvement in Becker muscular dystrophy. Can J Cardiol. (2008) ;24: (10):786–92.

[161] 

Fornander F , Solheim T , Eisum AV , Poulsen NS , Andersen AG , Dahlqvist JR et al. Quantitative Muscle MRI and Clinical Findings in Women With Pathogenic Dystrophin Gene Variants. Front Neurol. (2021) ;12: :707837.

[162] 

Juan-Mateu J , Rodríguez MJ , Nascimento A , Jiménez-Mallebrera C , González-Quereda L , Rivas E et al. Prognostic value of X-chromosome inactivation in symptomatic female carriers of dystrophinopathy. Orphanet J RareDis. (2012) ;7: :82.

[163] 

Politano L , Nigro V , Nigro G , Petretta VR , Passamano L , Papparella S et al. Development of cardiomyopathy in female carriers of Duchenne and Becker muscular dystrophies. Jama. (1996) ;275: (17):1335–8.

[164] 

Florian A , Rösch S , Bietenbeck M , Engelen M , Stypmann J , Waltenberger J et al. Cardiac involvement in female Duchenne and Becker muscular dystrophy carriers in comparison to their first-degree male relatives: a comparative cardiovascular magnetic resonance study. Eur Heart J Cardiovasc Imaging. (2016) ;17: (3):326–33.

[165] 

Mah ML , Cripe L , Slawinski MK , Al-Zaidy SA , Camino E , Lehman KJ et al. Duchenne and Becker muscular dystrophycarriers: Evidence of cardiomyopathy by exercise and cardiac MRI testing. Int J Cardiol.. (2020) ;316: :257–65.

[166] 

Miyagoe-Suzuki Y , Nishiyama T , Nakamura M , Narita A , Takemura F , Masuda S et al. Induction of Pluripotent Stem Cells from a Manifesting Carrier of Duchenne Muscular Dystrophy and Characterization of Their X-Inactivation Status. Stem Cells Int. (2017) ;2017: 7906843.

[167] 

De Sanctis R , Pane M , Sivo S , Ricotti V , Baranello G , Frosini S et al. Suitability of North Star Ambulatory Assessment in young boys with Duchenne muscular dystrophy. Neuromuscul Disord. (2015) ;25: (1):14–8.

[168] 

Mercuri E , Coratti G , Messina S , Ricotti V , Baranello G , D’Amico A et al. Revised North Star Ambulatory Assessment for Young Boys with Duchenne Muscular Dystrophy. PLoS One. (2016) ;11: (8):e0160195.

[169] 

Scott E , Mawson SJ . Measurement in Duchenne muscular dystrophy: considerations in the development of a neuromuscular assessment tool. Dev Med Child Neurol. (2006) ;48: (6):540–4.

[170] 

Mazzone ES , Messina S , Vasco G , Main M , Eagle M , D’Amico A et al. Reliability of the North Star Ambulatory Assessment in a multicentric setting. Neuromuscul Disord. (2009) ;19: (7):458–61.

[171] 

Mazzone E , Martinelli D , Berardinelli A , Messina S , D’Amico A , Vasco G et al. North Star Ambulatory Assessment, 6-minute walk test and timed items in ambulant boys with Duchenne muscular dystrophy. Neuromuscul Disord. (2010) ;20: (11):712–6.

[172] 

Mayhew A , Cano S , Scott E , Eagle M , Bushby K , Muntoni F . Moving towards meaningful measurement: Rasch analysis of the North Star Ambulatory Assessment in Duchenne muscular dystrophy. Dev Med Child Neurol. (2011) ;53: (6):535–42.

[173] 

Scott E , Eagle M , Mayhew A , Freeman J , Main M , Sheehan J et al. Development of a functional assessment scale for ambulatory boys with Duchenne muscular dystrophy. Physiother Res Int. (2012) ;17: (2):101–9.

[174] 

Brooke MH , Fenichel GM , Griggs RC , Mendell JR , Moxley R , Miller JP et al. Clinical investigation in Duchenne dystrophy: 2. Determination of the “power” of therapeutic trials based on the natural history. Muscle Nerve. (1983) ;6: (2):91–103.

[175] 

Brooke MH , Griggs RC , Mendell JR , Fenichel GM , Shumate JB , Pellegrino RJ . Clinical trial in Duchenne dystrophy. I. The design of the protocol. Muscle Nerve. (1981) ;4: (3):186–97.

[176] 

Fowler WM Jr , Abresch RT , Aitkens S , Carter GT , Johnson ER , Kilmer DD et al. Profiles of neuromuscular diseases. Design of the protocol. Am J Phys Med Rehabil. (1995) ;74: (5 Suppl):S62–9.

[177] 

Mayhew JE , Florence JM , Mayhew TP , Henricson EK , Leshner RT , McCarter RJ et al. Reliable surrogate outcome measures in multicenter clinical trials of Duchenne muscular dystrophy. Muscle Nerve. (2007) ;35: (1):36–42.

[178] 

Mendell JR , Moxley RT , Griggs RC , Brooke MH , Fenichel GM , Miller JP et al. Randomized, double-blind six-month trial of prednisone in Duchenne’s muscular dystrophy. N Engl J Med. (1989) ;320: (24):1592–7.

[179] 

McDonald CM , Henricson EK , Han JJ , Abresch RT , Nicorici A , Atkinson L et al. The 6-minute walk test in Duchenne/Becker muscular dystrophy: longitudinal observations. Muscle Nerve. (2010) ;42: (6):966–74.

[180] 

Mazzone E , Vasco G , Sormani MP , Torrente Y , Berardinelli A , Messina S et al. Functional changes in Duchenne muscular dystrophy: a 12-month longitudinal cohort study. Neurology. (2011) ;77: (3):250–6.

[181] 

Mazzone ES , Pane M , Sormani MP , Scalise R , Berardinelli A , Messina S et al. 24 month longitudinal data in ambulant boys with Duchenne muscular dystrophy. PLoS One. (2013) ;8: (1):e52512.

[182] 

Alfano LN , Miller NF , Berry KM , Yin H , Rolf KE , Flanigan KM et al. The 100-meter timed test: Normative data in healthy males and comparative pilot outcome data for use in Duchenne muscular dystrophy clinical trials. Neuromuscul Disord. (2017) ;27: (5):452–7.

[183] 

Miller NAL , Flanigan K et al. , The 100-meter timed test: ability todetect change over time in Duchenne muscular dystrophy. Neuromuscular Disordorders.S. (2017) ;27: :235.

[184] 

United States Food and Drug Administration (FDA): Center for Drug Evaluation and Research (CDER) and Center for Biologics Evaluation and Research (CBER). Guidance for Industry: Expedited Programs for Serious Conditions–Drugs and Biologics. Silver Spring, Maryland: FDA; 2014.

[185] 

Finder J , Mayer OH , Sheehan D , Sawnani H , Abresch RT , Benditt J et al. Pulmonary Endpoints in Duchenne Muscular Dystrophy. A Workshop Summary. Am J Respir Crit Care Med. (2017) ;196: (4):512–9.

[186] 

Mayer OH . Pulmonary function and clinical correlation in DMD. Paediatr Respir Rev. (2019) ;30: :13–5.

[187] 

Connolly AM , Florence JM , Zaidman CM , Golumbek PT , Mendell JR , Flanigan KM et al. Clinical trial readiness in non-ambulatory boys and men with duchenne muscular dystrophy: MDA-DMD network follow-uMuscle Nerve (2016) ;54: (4):681–9.

[188] 

Phillips MF , Quinlivan RC , Edwards RH , Calverley PM . Changes in spirometry over time as a prognostic marker in patients with Duchenne muscular dystrophy. Am J Respir Crit Care Med. (2001) ;164: (12):2191–4.

[189] 

Bello L , D’Angelo G , Villa M , Fusto A , Vianello S , Merlo B et al. Genetic modifiers of respiratory function in Duchenne muscular dystrophy. Ann Clin Transl Neurol. (2020) ;7: (5):786–98.

[190] 

Capdevila XJ , Perrigault PF , Perey PJ , Roustan JP , d’Athis F . Occlusion pressure and its ratio to maximum inspiratory pressure are useful predictors for successful extubation following T-piece weaning trial. Chest. (1995) ;108: (2):482–9.

[191] 

Servais L , Camino E , Clement A , McDonald CM , Lukawy J , Lowes LP et al. First Regulatory Qualification of a Novel Digital Endpoint in Duchenne Muscular Dystrophy: A Multi-Stakeholder Perspective on the Impact for Patients and for Drug Development in Neuromuscular Diseases. Digit Biomark. (2021) ;5: (2):183–90.

[192] 

McDonald CM , Widman LM , Walsh DD , Walsh SA , Abresch RT . Use of step activity monitoring for continuous physical activity assessment in boys with Duchenne muscular dystrophy. Arch Phys Med Rehabil. (2005) ;86: (4):802–8.

[193] 

Fowler EG , Staudt LA , Heberer KR , Sienko SE , Buckon CE , Bagley AM et al. Longitudinal community walking activity in Duchenne muscular dystrophy. Muscle Nerve. (2018) ;57: (3):401–6.

[194] 

European Medicines Agency, Committee for Medicinal Products for Human Use (CHMP). Qualification Opinion for Stride velocity 95th centile as primary endpoint in studies in ambulatory Duchenne Muscular Dystrophy studies. Amsterdam, The Netherlands; 2023.

[195] 

Rutkove SB , Qi K , Shelton K , Liss J , Berisha V , Shefner JM . ALS longitudinal studies with frequent data collection at home: study design and baseline data. Amyotroph Lateral Scler Frontotemporal Degener. (2019) ;20: (1-2):61–7.

[196] 

Fettes E , Riley M , Brotherston S , Doughty C , Griffiths B , Laverty A et al. “You’re on mute!” Does pediatric CF home spirometry requirephysiologist supervision? Pediatr Pulmonol. (2022) ;57: (1):278–84.

[197] 

Contesse MG , Lowes LP , White MK , Dalle Pazze L , McSherry C , Alfano LN et al. Development of Duchenne Video Assessment scorecards to evaluate ease of movement among those with Duchenne muscular dystrophy. PLoS One. (2022) ;17: (4):e0266845.

[198] 

Sienko Thomas S , Buckon CE , Nicorici A , Bagley A , McDonald CM , Sussman MD . Classification of the gait patterns of boys with Duchenne muscular dystrophy and their relationship to function. J Child Neurol. (2010) ;25: (9):1103–9.

[199] 

Heberer K , Fowler E , Staudt L , Sienko S , Buckon CE , Bagley A et al. Hip kinetics during gait are clinically meaningful outcomes in young boys with Duchenne muscular dystrophy. Gait Postu Rep. (2016) ;48: :159–64.

[200] 

Han JJ , de Bie E , Nicorici A , Abresch RT , Anthonisen C , Bajcsy R et al. Reachable workspace and performance of upper limb (PUL) in duchenne muscular dystrophy. Muscle Nerve. (2016) ;53: (4):545–54.

[201] 

Han JJ , Kurillo G , Abresch RT , de Bie E , Nicorici A , Bajcsy R . Reachable workspace in facioscapulohumeral muscular dystrophy (FSHD) by Kinect. Muscle Nerve. (2015) ;51: (2):168–75.

[202] 

Lowes LP , Alfano LN , Crawfis R , Berry K , Yin H , Dvorchik I et al. Reliability and validity of active-seated: An outcome in dystrophinopathy. Muscle Nerve. (2015) ;52: (3):356–62.

[203] 

United States Food and Drug Administration (FDA): Center for Biologics Evaluation and Research, Center for Devices and Radiological Health, Center for Drug Evaluation and Research. Digital Health Technologies for Remote Data Acquisition in Clinical Investigations Guidance for Industry, Investigators, and Other Stakeholders DRAFT GUIDANCE. Silver Spring, Maryland: FDA; 2021.

[204] 

United States Food and Drug Administration (FDA): Center for Drug Evaluation and Research (CDER), Center for Biologics Evaluation and Research (CBER). Patient Preference Information—Voluntary Submission, Review in Premarket Approval Applications, Humanitarian Device Exemption Applications, De Novo Requests, and Inclusion in Decision Summaries and Device Labeling. Guidance for Industry, Food and Drug Administration Staff, and Other Stakeholders. Silver Spring, Maryland; 2016.

[205] 

United States Food and Drug Administration (FDA) Oncology Center of Excellence (OCE), Center for Drug Evaluation and Research (CDER) Center for Biologics Evaluation and Research (CBER) Center for Devices and Radiological Health (CDRH). Core Patient-Reported Outcomes in Cancer Clinical Trials Guidance for Industry DRAFT GUIDANCE. Silver Spring: FDA; 2021.

[206] 

United States Food and Drug Administration (FDA) Center for Devices and Radiological Health (CDRH), Center for Biologics Evaluation and Research (CBER). Principles for Selecting, Developing, Modifying, and Adapting Patient-Reported Outcome Instruments for Use in Medical Device Evaluation. Guidance for Industry. Silver Spring, Maryland: FDA; 2022.

[207] 

Landfeldt E , Lindgren P , Bell CF , Guglieri M , Straub V , Lochmüller H et al. Health-related quality of life in patients with Duchenne muscular dystrophy: a multinational, cross-sectional study. Dev Med Child Neurol. (2016) ;58: (5):508–15.

[208] 

Messina S , Vita GL , Sframeli M , Mondello S , Mazzone E , D’Amico A et al. Health-related quality of life and functional changes in DMD: A 12-month longitudinal cohort study. Neuromuscul Disord. (2016) ;26: (3):189–96.

[209] 

Otto C , Steffensen BF , Højberg AL , Barkmann C , Rahbek J , Ravens-Sieberer U et al. Predictors of Health-Related Quality of Life in boys with Duchenne muscular dystrophy from six European countries. J Neurol. (2017) ;264: (4):709–23.

[210] 

Campbell C , McColl E , McDermott MP , Martens WB , Guglieri M , Griggs RC . Health related quality of life in young,steroid-naïve boys with Duchenne muscular dystrophy. Neuromuscul Disord.. (2021) ;31: (11):1161–8.

[211] 

Landfeldt E , Iff J , Henricson E . Rasch Analysis of the Pediatric Quality of Life Inventory 4.0 Generic Core Scales Administered to Patients With Duchenne Muscular Dystrophy. Value Health. (2021) ;24: (10):1490–8.

[212] 

Lai JS , Nowinski C , Victorson D , Bode R , Podrabsky T , McKinney N et al. Quality-of-life measures in children with neurological conditions: pediatric Neuro-QOL. Neurorehabil Neural Repair. (2012) ;26: (1):36–47.

[213] 

Crescimanno G , Greco F , D’Alia R , Messina L , Marrone O . Quality of life in long term ventilated adult patients with Duchenne muscular dystrophy. Neuromuscul Disord. (2019) ;29: (8):569–75.

[214] 

Crossnohere NL , Fischer R , Lloyd A , Prosser LA , Bridges JFP . Assessing the Appropriateness of the EQ-5D for Duchenne Muscular Dystrophy: A Patient-Centered Study. Med Decis Making. (2021) ;41: (2):209–21.

[215] 

Cavazza M , Kodra Y , Armeni P , De Santis M , López-Bastida J , Linertová R et al. Social/economic costs and health-relatedquality of life in patients with Duchenne muscular dystrophy inEurope. Eur J Health Econ. (2016) ;1: 17 Suppl:19–29.

[216] 

Hartman AG , McKendry S , Bendixen R . Comparing contributors of parental sleep health in families with and without a child with Duchenne muscular dystrophy. Sleep Health. (2022) ;8: (1):107–13.

[217] 

Yayici Koken O , Gultutan P , Gurkas E , Degerliyurt A Sleep: How is it affected in patients with DMD and their mothers? Minerva Pediatr (Torino). 2021.

[218] 

Crescimanno G , Greco F , Abbate A , Canino M , Bertini M , Marrone O . Subjective sleep quality in adult patients affected by Duchenne muscular dystrophy. Beyond nocturnal hypoventilation. Sleep Med. (2020) ;69: :168–71.

[219] 

Nozoe KT , Polesel DN , Moreira GA , Pires GN , Akamine RT , Tufik S et al. Sleep quality of mother-caregivers of Duchenne muscular dystrophy patients. Sleep Breath. (2016) ;20: (1):129–34.

[220] 

Henricson EK MC , Joyce N and the CINRG DNHS Investigators. Technical Report: Development and Validation of the Duchenne Muscular Dystrophy Lifetime Mobility Scale (DMD-LMS). U.S. Army Medical Research and Materiel Command Grant #MD130062. U.S. Department of Defense / Defense Technical Information Center. December 2019.

[221] 

Uzark K , King E , Cripe L , Spicer R , Sage J , Kinnett K et al. Health-related quality of life in children and adolescents with Duchenne muscular dystrophy. Pediatrics. (2012) ;130: (6):e1559–66.

[222] 

Brunherotti MA , Sobreira C , Rodrigues-Júnior AL , de Assis MR , Terra Filho J , Baddini Martinez JA . Correlations of EgenKlassifikation and Barthel Index scores with pulmonary functionparameters in Duchenne muscular dystrophy. Heart Lung. (2007) ;36: (2):132–9.

[223] 

Lee HN , Sawnani H , Horn PS , Rybalsky I , Relucio L , Wong BL . The Performance of the Upper Limb scores correlate with pulmonary function test measures and Egen Klassifikation scores in Duchenne muscular dystrophy. Neuromuscul Disord. (2016) ;26: (4-5):264–71.

[224] 

Fagoaga J , Girabent-Farrés M , Bagur-Calafat C , Febrer A , Steffensen BF . [Functional assessment for people unable to walk dueto spinal muscular atrophy and Duchenne muscular dystrophy. Translation and validation of the Egen Klassifikation 2 scale forthe Spanish population]. Rev Neurol. (2015) ;60: (10):439–46.

[225] 

Klingels K , Mayhew AG , Mazzone ES , Duong T , Decostre V , Werlauff U et al. Development of a patient-reported outcome measure for upper limb function in Duchenne muscular dystrophy: DMD Upper Limb PROM. Dev Med Child Neurol. (2017) ;59: (2):224–31.

[226] 

Powell PA , Carlton J , Rowen D , Chandler F , Guglieri M , Brazier JE . Development of a New Quality of Life Measurefor Duchenne Muscular Dystrophy Using Mixed Methods: The DMD-QoL. Neurology.e-e. (2438) ;96: (19):e2438–e50.

[227] 

Rowen D , Powell P , Mukuria C , Carlton J , Norman R , Brazier J . Deriving a Preference-Based Measure for People With Duchenne Muscular Dystrophy From the DMD-QoL. Value Health. (2021) ;24: (10):1499–510.

[228] 

Mokkink LB , Terwee CB , Patrick DL , Alonso J , Stratford PW , Knol DL et al. The COSMIN study reached international consensus on taxonomy, terminology, and definitions of measurement properties for health-related patient-reported outcomes. J Clin Epidemiol. (2010) ;63: (7):737–45.

[229] 

Carlton J , Powell PA . Measuring carer quality of life in Duchenne muscular dystrophy: a systematic review of the reliability and validity of self-report instruments using COSMIN. Health Qual Life Outcomes. (2022) ;20: (1):57.

[230] 

Wagner JA , Williams SA , Webster CJ . Biomarkers and surrogate end points for fit-for-purpose development and regulatory evaluation of new drugs. Clin Pharmacol Ther. (2007) ;81: (1):104–7.

[231] 

Waldrop MA , Gumienny F , El Husayni S , Frank DE , Weiss RB , Flanigan KM . Low-level dystrophin expression attenuating the dystrophinopathy phenotype. Neuromuscul Disord. (2018) ;28: (2):116–21.

[232] 

Ledford H US government approves controversial drug for muscular dystrophy. Nature. 2016.

[233] 

Gallo A , Abraham A , Katzberg HD , Ilaalagan S , Bril V , Breiner A . Muscle biopsy technical safety and quality using a self-contained, vacuum-assisted biopsy technique. Neuromuscul Disord. (2018) ;28: (5):450–3.

[234] 

Barthelemy F , Woods JD , Nieves-Rodriguez S , Douine ED , Wang R , Wanagat J et al. A well-tolerated core needle muscle biopsy process suitable for children and adults. Muscle Nerve. (2020) ;62: (6):688–98.

[235] 

Beekman C , Janson AA , Baghat A , van Deutekom JC , Datson NA . Use of capillary Western immunoassay (Wes) for quantification of dystrophin levels in skeletal muscle of healthy controls and individuals with Becker and Duchenne muscular dystrophy. PLoS One. (2018) ;13: (4):e0195850.

[236] 

Vetter TA , Nicolau S , Bradley AJ , Frair EC , Flanigan KM . Automated immunofluorescence analysis for sensitive and precise dystrophin quantification in muscle biopsies. Neuropathol Appl Neurobiol. (2022) ;48: (3):e12785.

[237] 

Anthony K , Arechavala-Gomeza V , Taylor LE , Vulin A , Kaminoh Y , Torelli S et al. Dystrophin quantification: Biological and translational research implications. Neurology. (2014) ;83: (22):2062–9.

[238] 

Sardone V , Ellis M , Torelli S , Feng L , Chambers D , Eastwood D et al. A novel high-throughput immunofluorescence analysis method for quantifying dystrophin intensity in entire transverse sections of Duchenne muscular dystrophy muscle biopsy samples. PLoS One. (2018) ;13: (3):e0194540.

[239] 

Torelli S , Scaglioni D , Sardone V , Ellis MJ , Domingos J , Jones A et al. High-Throughput Digital Image Analysis Reveals Distinct Patterns of Dystrophin Expression in Dystrophinopathy Patients. J Neuropathol Exp Neurol. (2021) ;80: (10):955–65.

[240] 

Taylor LE , Kaminoh YJ , Rodesch CK , Flanigan KM . Quantification of dystrophin immunofluorescence in dystrophinopathy muscle specimens. Neuropathol Appl Neurobiol. (2012) ;38: (6):591–601.

[241] 

Brown KJ , Marathi R , Fiorillo AA , Ciccimaro EF , Sharma S , Rowlands DS et al. Accurate Quantitation of Dystrophin Protein in Human Skeletal Muscle Using Mass Spectrometry. J Bioanal Biomed. 2012;Suppl 7.

[242] 

Canessa EH , Goswami MV , Alayi TD , Hoffman EP , Hathout Y . Absolute quantification of dystrophin protein in human muscle biopsies using parallel reaction monitoring (PRM). J Mass Spectrom. (2020) ;55: (2):e4437.

[243] 

Kleopa KA , Drousiotou A , Mavrikiou E , Ormiston A , Kyriakides T . Naturally occurring utrophin correlates with disease severity in Duchenne muscular dystrophy. Hum Mol Genet. (2006) ;15: (10):1623–8.

[244] 

Flanigan KM , Dunn DM , von Niederhausern A , Soltanzadeh P , Gappmaier E , Howard MT et al. Mutational spectrum of DMD mutations in dystrophinopathy patients: application of modern diagnostic techniques to a large cohort. Hum Mutat. (2009) ;30: (12):1657–66.

[245] 

Barnard AM , Hammers DW , Triplett WT , Kim S , Forbes SC , Willcocks RJ et al. Evaluating Genetic Modifiers of Duchenne Muscular Dystrophy Disease Progression Using Modeling and MRI. Neurology. (2022) ;99: (21):e2406–e16.

[246] 

Pegoraro E , Schimke RN , Garcia C , Stern H , Cadaldini M , Angelini C et al. Genetic and biochemical normalization in female carriers of Duchenne muscular dystrophy: evidence for failure of dystrophin production in dystrophin-competent myonuclei. Neurology. (1995) ;45: (4):677–90.

[247] 

Hoffman EP , Arahata K , Minetti C , Bonilla E , Rowland LP . Dystrophinopathy in isolated cases of myopathy in females. Neurology. (1992) ;42: (5):967–75.

[248] 

Hanson B , Wood MJA , Roberts TC . Molecular correction of Duchenne muscular dystrophy by splice modulation and gene editing. RNA Biol. (2021) ;18: (7):1048–62.

[249] 

Simmons TR , Vetter TA , Huang N , Vulin-Chaffiol A , Wein N , Flanigan KM . Pre-clinical dose-escalation studies establish a therapeutic range for U7snRNA-mediated DMD exon 2 skipping. Mol Ther Methods Clin Dev. (2021) ;21: :325–40.

[250] 

Aupy P , Zarrouki F , Sandro Q , Gastaldi C , Buclez PO , Mamchaoui K et al. Long-Term Efficacy of AAV9-U7snRNA-Mediated Exon 51 Skipping in mdx52 Mice. Mol Ther Methods Clin Dev. (2020) ;17: :1037–47.

[251] 

Lesman D , Rodriguez Y , Rajakumar D , Wein N . U7 snRNA, a Small RNA with a Big Impact in Gene Therapy. Hum Gene Ther. (2021) ;32: (21-22):1317–29.

[252] 

Verheul RC , van Deutekom JC , Datson NA . Digital Droplet PCR for the Absolute Quantification of Exon Skipping Induced by Antisense Oligonucleotides in (Pre-)Clinical Development for Duchenne Muscular Dystrophy. PLoS One. (2016) ;11: (9):e0162467.

[253] 

Wein N , Dunn DM , Waldrop MA , Gushchina LV , Frair EC , Weiss RB et al. Absence of Significant Off-Target Splicing Variation with a U7snRNA Vector Targeting DMD Exon 2 Duplications. Hum Gene Ther. (2021) ;32: (21-22):1346–59.

[254] 

Güttsches AK , Rehmann R , Schreiner A , Rohm M , Forsting J , Froeling M et al. Quantitative Muscle-MRI Correlates with Histopathology in Skeletal Muscle Biopsies. J Neuromuscul Dis. (2021) ;8: (4):669–78.

[255] 

Forbes SC , Walter GA , Rooney WD , Wang DJ , DeVos S , Pollaro J et al. Skeletal muscles of ambulant children with Duchenne muscular dystrophy: validation of multicenter study of evaluation with MR imaging and MR spectroscopy. Radiology. (2013) ;269: (1):198–207.

[256] 

Fischmann A , Hafner P , Gloor M , Schmid M , Klein A , Pohlman U et al. Quantitative MRI and loss of free ambulation in Duchenne muscular dystrophy. J Neurol. (2013) ;260: (4):969–74.

[257] 

Lilien C , Reyngoudt H , Seferian AM , Gidaro T , Annoussamy M , Chê V et al. Upper limb disease evolution in exon 53 skipping eligiblepatients with Duchenne muscular dystrophy. Ann Clin Transl Neurol. (2021) ;8: (10):1938–50.

[258] 

Arpan I , Willcocks RJ , Forbes SC , Finkel RS , Lott DJ , Rooney WD et al. Examination of effects of corticosteroids on skeletal muscles of boys with DMD using MRI and MRS. Neurology. (2014) ;83: (11):974–80.

[259] 

Bonati U , Hafner P , Schädelin S , Schmid M , Naduvilekoot Devasia A , Schroeder J et al. Quantitative muscle MRI: A powerful surrogateoutcome measure in Duchenne muscular dystrophy. Neuromuscul Disord.. (2015) ;25: (9):679–85.

[260] 

Ricotti V , Evans MR , Sinclair CD , Butler JW , Ridout DA , Hogrel JY et al. Upper Limb Evaluation in Duchenne Muscular Dystrophy: Fat-Water Quantification by MRI, Muscle Force and Function Define Endpoints for Clinical Trials. PLoS One. (2016) ;11: (9):e0162542.

[261] 

Wood CL , Hollingsworth KG , Hughes E , Punniyakodi S , Muni-Lofra R , Mayhew A et al. Pubertal induction in adolescents with DMD is associated with high satisfaction, gonadotropin release and increased muscle contractile surface area. Eur J Endocrinol. (2021) ;184: (1):67–79.

[262] 

Lott DJ , Taivassalo T , Cooke KD , Park H , Moslemi Z , Batra A et al. Safety, feasibility, and efficacy of strengthening exercise in Duchenne muscular dystrophy. Muscle Nerve. (2021) ;63: (3):320–6.

[263] 

van de Velde NM , Hooijmans MT , Sardjoe Mishre ASD , Keene KR , Koeks Z , Veeger TTJ et al. Selection Approach to Identify the Optimal Biomarker Using Quantitative Muscle MRI and Functional Assessments in Becker Muscular Dystrophy. Neurology.. (2021) ;97: (5):e513–e22.

[264] 

Miller RG , Sharma KR , Pavlath GK , Gussoni E , Mynhier M , Lanctot AM et al. Myoblast implantation in Duchenne muscular dystrophy: the San Francisco study. Muscle Nerve. (1997) ;20: (4):469–78.

[265] 

Bish LT , Sleeper MM , Forbes SC , Morine KJ , Reynolds C , Singletary GE et al. Long-term systemic myostatin inhibition via liver-targeted gene transfer in golden retriever muscular dystrophy. Hum Gene Ther. (2011) ;22: (12):1499–509.

[266] 

Fischmann A , Hafner P , Fasler S , Gloor M , Bieri O , Studler U et al. Quantitative MRI can detect subclinical disease progression in muscular dystrophy. J Neurol. (2012) ;259: (8):1648–54.

[267] 

Nagy S , Schädelin S , Hafner P , Bonati U , Scherrer D , Ebi S et al. Longitudinal reliability of outcome measures in patients with Duchenne muscular dystrophy. Muscle Nerve. (2020) ;61: (1):63–8.

[268] 

Glemser PA , Jaeger H , Nagel AM , Ziegler AE , Simons D , Schlemmer HP et al. (23)Na MRI and myometry to compare eplerenone vs glucocorticoid treatment in Duchenne dystrophy. Acta Myol. (2017) ;36: (1):2–13.

[269] 

Gerhalter T , Gast LV , Marty B , Martin J , Trollmann R , Schüssler S et al. (23) Na MRI depicts early changes in ion homeostasis in skeletal muscle tissue of patients with duchenne muscular dystrophy. J Magn Reson Imaging. (2019) ;50: (4):1103–13.

[270] 

Batra A , Vohra RS , Chrzanowski SM , Hammers DW , Lott DJ , Vandenborne K et al. Effects of PDE5 inhibition on dystrophic muscle following an acute bout of downhill running and endurance training. J Appl Physiol. (2019) ;126: (6):1737–45.

[271] 

Lopez C , Taivassalo T , Berru MG , Saavedra A , Rasmussen HC , Batra A et al. Postcontractile blood oxygenation level-dependent (BOLD) response in Duchenne muscular dystrophy. J Appl Physiol. (2021) ;131: (1):83–94.

[272] 

Meyerspeer M , Boesch C , Cameron D , Dezortová M , Forbes SC , Heerschap A et al. (31) P magnetic resonance spectroscopy inskeletal muscle: Experts’ consensus recommendations. NMR Biomed. (2020) ;34: (5):e4246.

[273] 

Leitner ML , Kapur K , Darras BT , Yang M , Wong B , Dalle Pazze L et al. Electrical impedance myography for reducing sample size in Duchenne muscular dystrophy trials. Ann Clin Transl Neurol. (2020) ;7: (1):4–14.

[274] 

Connolly AM , Malkus EC , Mendell JR , Flanigan KM , Miller JP , Schierbecker JR et al. Outcome reliability in non-ambulatory boys/men with Duchenne muscular dystrophy. Muscle Nerve. (2015) ;51: (4):522–32.

[275] 

Zaidman CM , Wu JS , Kapur K , Pasternak A , Madabusi L , Yim S et al. Quantitative muscle ultrasound detects disease progression in Duchenne muscular dystrophy. Ann Neurol. (2017) ;81: (5):633–40.

[276] 

Janssen M , Harlaar J , Koopman B , de Groot IJM . Dynamic arm study: quantitative description of upper extremityfunction and activity of boys and men with duchenne muscular dystrophy. J Neuroeng Rehabil.. (2017) ;14: (1):45.

[277] 

Weng WC , Tsui PH , Lin CW , Lu CH , Lin CY , Shieh JY et al. Evaluation of muscular changes by ultrasound Nakagami imaging in Duchenne muscular dystrophy. Sci Rep. (2017) ;7: (1):4429.

[278] 

Moore CJ , Caughey MC , Meyer DO , Emmett R , Jacobs C , Chopra M et al. In Vivo Viscoelastic Response (VisR) Ultrasound for Characterizing Mechanical Anisotropy in Lower-Limb Skeletal Muscles of Boys with and without Duchenne Muscular Dystrophy. Ultrasound Med Biol. (2018) ;44: (12):2519–30.

[279] 

Weng WC , Lin CW , Shen HC , Chang CC , Tsui PH . Instantaneous frequency as a new approach for evaluating the clinical severity of Duchenne muscular dystrophy through ultrasound imaging. Ultrasonics. (2019) ;94: :235–41.

[280] 

Yan D , Li Q , Lin CW , Shieh JY , Weng WC , Tsui PH . Clinical Evaluation of Duchenne Muscular Dystrophy Severity Using Ultrasound Small-Window Entropy Imaging. Entropy (Basel). (2020) ;22: (7).

[281] 

Rutkove SB , Darras BT . Electrical impedance myography for the assessment of children with muscular dystrophy: a preliminary study. J Phys Conf Ser. (2013) ;434: (1).

[282] 

Wagner KR , Guglieri M , Ramaiah SK , Charnas L , Marraffino S , Binks M et al. Safety and disease monitoringbiomarkers in Duchenne muscular dystrophy: results from a Phase II trial. Biomark Med.. (2021) ;15: (15):1389–96.

[283] 

Goossens N , Nakagawa S , Sun X , Hoshida Y . Cancer biomarker discovery and validation. Transl Cancer Res. (2015) ;4: (3):256–69.

[284] 

Kornegay JN , Bogan JR , Bogan DJ , Childers MK , Li J , Nghiem P et al. Canine models of Duchenne muscular dystrophy and their use in therapeutic strategies. Mamm Genome. (2012) ;23: (1-2):85–108.

[285] 

Kim HK , Laor T , Horn PS , Wong B . Quantitative assessment of the T2 relaxation time of the gluteus muscles in children with Duchenne muscular dystrophy: a comparative study before and after steroid treatment. Korean J Radiol. (2010) ;11: (3):304–11.

[286] 

Mavrogeni S , Papavasiliou A , Douskou M , Kolovou G , Papadopoulou E , Cokkinos DV . Effect of deflazacort on cardiac and sternocleidomastoid muscles in Duchenne muscular dystrophy: a magnetic resonance imaging study. Eur J Paediatr Neurol. (2009) ;13: (1):34–40.

[287] 

Walter G , Cordier L , Bloy D , Sweeney HL . Noninvasive monitoring of gene correction in dystrophic muscle. Magn Reson Med. (2005) ;54: (6):1369–76.

[288] 

Torrente Y , Belicchi M , Marchesi C , D’Antona G , Cogiamanian F , Pisati F et al. Autologous transplantation of muscle-derived CD133+stem cells in Duchenne muscle patients. Cell Transplant. (2007) ;16: (6):563–77.

[289] 

Karpati G , Ajdukovic D , Arnold D , Gledhill RB , Guttmann R , Holland P et al. Myoblast transfer in Duchenne muscular dystrophy. Ann Neurol. (1993) ;34: (1):8–17.

[290] 

Willcocks RJ , Arpan IA , Forbes SC , Lott DJ , Senesac CR , Senesac E et al. Longitudinal measurements of MRI-T2 in boys with Duchenne muscular dystrophy: effects of age and disease progression. Neuromuscul Disord. (2014) ;24: (5):393–401.

[291] 

Lehmann-Horn F , Weber MA , Nagel AM , Meinck HM , Breitenbach S , Scharrer J et al. Rationale for treating oedema in Duchenne muscular dystrophy with eplerenone. Acta Myol. (2012) ;31: (1):31–9.

[292] 

Hogrel JY , Wary C , Moraux A , Azzabou N , Decostre V , Ollivier G et al. Longitudinal functional and NMR assessment of upper limbs in Duchenne muscular dystrophy. Neurology. (2016) ;86: (11):1022–30.

[293] 

Willcocks RJ , Triplett WT , Forbes SC , Arora H , Senesac CR , Lott DJ et al. Magnetic resonance imaging of the proximal upper extremity musculature in boys with Duchenne muscular dystrophy. J Neurol. (2017) ;264: (1):64–71.

[294] 

Mankodi A , Kovacs W , Norato G , Hsieh N , Bandettini WP , Bishop CA et al. Respiratory magnetic resonance imaging biomarkers in Duchenne muscular dystrophy. Ann Clin Transl Neurol. (2017) ;4: (9):655–62.

[295] 

Barnard AM , Lott DJ , Batra A , Triplett WT , Forbes SC , Riehl SL et al. Imaging respiratory muscle quality and function in Duchenne muscular dystrophy. J Neurol. (2019) ;266: (11):2752–63.

[296] 

Forbes SC , Arora H , Willcocks RJ , Triplett WT , Rooney WD , Barnard AM et al. Upper and Lower Extremities in Duchenne Muscular Dystrophy Evaluated with Quantitative MRI and Proton MR Spectroscopy in a Multicenter Cohort. Radiology. (2020) ;295: (3):616–25.

[297] 

Naarding KJ , Keene KR , Sardjoe Mishre ASD , Veeger TTJ , van de Velde NM , Prins AJ et al. Preserved thenar muscles in non-ambulant Duchenne muscular dystrophy patients. J Cachexia Sarcopenia Muscle. (2021) ;12: (3):694–703.

[298] 

Barnard AM , Lott DJ , Batra A , Triplett WT , Willcocks RJ , Forbes SC et al. Characterizing Expiratory Respiratory Muscle Degeneration in Duchenne Muscular Dystrophy Using MRI. Chest. (2022) ;161: (3):753–63.

[299] 

Liu GC , Jong YJ , Chiang CH , Jaw TS . Duchenne muscular dystrophy: MR grading system with functional correlation. Radiology. (1993) ;186: (2):475–80.

[300] 

Matsumura K , Nakano I , Fukuda N , Ikehira H , Tateno Y , Aoki Y . Proton spin-lattice relaxation time of Duchenne dystrophy skeletal muscle by magnetic resonance imaging. Muscle Nerve. (1988) ;11: (2):97–102.

[301] 

Huang Y , Majumdar S , Genant HK , Chan WP , Sharma KR , Yu P et al. Quantitative MR relaxometry study of muscle composition and function in Duchenne muscular dystrophy. J Magn Reson Imaging. (1994) ;4: (1):59–64.

[302] 

Marden FA , Connolly AM , Siegel MJ , Rubin DA . Compositional analysis of muscle in boys with Duchenne muscular dystrophy using MR imaging. Skeletal Radiol. (2005) ;34: (3):140–8.

[303] 

Mavrogeni S , Tzelepis GE , Athanasopoulos G , Maounis T , Douskou M , Papavasiliou A et al. Cardiac and sternocleidomastoid muscle involvement in Duchenne muscular dystrophy: an MRI study. Chest. (2005) ;127: (1):143–8.

[304] 

Kim HK , Laor T , Horn PS , Racadio JM , Wong B , Dardzinski BJ . T2 mapping in Duchenne muscular dystrophy: distribution of disease activity and correlation with clinical assessments. Radiology. (2010) ;255: (3):899–908.

[305] 

Mercuri E , Jungbluth H , Muntoni F . Muscle imaging in clinical practice: diagnostic value of muscle magnetic resonance imaging in inherited neuromuscular disorders. Curr Opin Neurol. (2005) ;18: (5):526–37.

[306] 

Mathur S , Lott DJ , Senesac C , Germain SA , Vohra RS , Sweeney HL et al. Age-related differences in lower-limb muscle cross-sectional area and torque production in boys with Duchenne muscular dystrophy. Arch Phys Med Rehabil. (2010) ;91: (7):1051–8.

[307] 

Kinali M , Arechavala-Gomeza V , Cirak S , Glover A , Guglieri M , Feng L et al. Muscle histology vs MRI in Duchenne muscular dystrophy. Neurology. (2011) ;76: (4):346–53.

[308] 

Torriani M , Townsend E , Thomas BJ , Bredella MA , Ghomi RH , Tseng BS . Lower leg muscle involvement in Duchenne muscular dystrophy: an MRimaging and spectroscopy study. Skeletal Radiol.. (2012) ;41: (4):437–45.

[309] 

Triplett WT , Baligand C , Forbes SC , Willcocks RJ , Lott DJ , DeVos S et al. Chemical shift-based MRI to measure fat fractions in dystrophic skeletal muscle. Magn Reson Med. (2014) ;72: (1):8–19.

[310] 

Kan HE , Scheenen TW , Wohlgemuth M , Klomp DW , van Loosbroek-Wagenmans I , Padberg GW et al. Quantitative MR imaging of individual muscle involvement in facioscapulohumeral muscular dystrophy. Neuromuscul Disord. (2009) ;19: (5):357–62.

[311] 

Brogna C , Cristiano L , Tartaglione T , Verdolotti T , Fanelli L , Ficociello L et al. Functional levels and MRIpatterns of muscle involvement in upper limbs in Duchenne muscular dystrophy. PLoS One. (2018) ;13: (6):e0199222.

[312] 

Misra LK , Luthra MG , Amtey SR , Elizondo-Riojas G , Swezey SH , Todd LE . Enhanced T1 differentiation between normal and dystrophic muscles. Magn Reson Imaging. (1984) ;2: (1):33–5.

[313] 

Schlaeger S , Sollmann N , Zoffl A , Becherucci EA , Weidlich D , Kottmaier E et al. Quantitative Muscle MRI in Patients with Neuromuscular Diseases-Association of Muscle Proton Density Fat Fraction with Semi-Quantitative Grading of Fatty Infiltration and Muscle Strength at the Thigh Region. Diagnostics (Basel). (2021) ;11: (6).

[314] 

Vohra RS , Lott D , Mathur S , Senesac C , Deol J , Germain S et al. Magnetic Resonance Assessment of Hypertrophic and Pseudo-Hypertrophic Changes in Lower Leg Muscles of Boys with Duchenne Muscular Dystrophy and Their Relationship to Functional Measurements. PLoS One.. (2015) ;10: (6):e0128915.

[315] 

Løkken N , Hedermann G , Thomsen C , Vissing J . Contractile properties are disrupted in Becker muscular dystrophy, but not in limb girdle type 2I. Ann Neurol. (2016) ;80: (3):466–71.

[316] 

Batra A , Lott DJ , Willcocks R , Forbes SC , Triplett W , Dastgir J et al. Lower Extremity Muscle Involvement in the Intermediate and Bethlem Myopathy Forms of COL6-Related Dystrophy and Duchenne Muscular Dystrophy: A Cross-Sectional Study. J Neuromuscul Dis. (2020) ;7: (4):407–17.

[317] 

Sherlock SP , Zhang Y , Binks M , Marraffino S . Quantitative muscle MRI biomarkers in Duchenne muscular dystrophy: cross-sectional correlations with age and functional tests. Biomark Med. (2021) ;15: (10):761–73.

[318] 

Fan Z , Wang J , Ahn M , Shiloh-Malawsky Y , Chahin N , Elmore S et al. Characteristics of magnetic resonance imaging biomarkers in a natural history study of golden retriever muscular dystrophy. Neuromuscul Disord. (2014) ;24: (2):178–91.

[319] 

Wang J , Fan Z , Vandenborne K , Walter G , Shiloh-Malawsky Y , An H et al. A computerized MRI biomarker quantification scheme for a canine model of Duchenne muscular dystrophy. Int J Comput Assist Radiol Surg. (2013) ;8: (5):763–74.

[320] 

Thibaud JL , Monnet A , Bertoldi D , Barthélémy I , Blot S , Carlier PG . Characterization of dystrophic muscle in goldenretriever muscular dystrophy dogs by nuclear magnetic resonanceimaging. Neuromuscul Disord. (2007) ;17: (7):575–84.

[321] 

Lott DJ , Forbes SC , Mathur S , Germain SA , Senesac CR , Lee Sweeney H et al. Assessment of intramuscular lipid and metabolites of the lower leg using magnetic resonance spectroscopy in boys with Duchenne muscular dystrophy. Neuromuscul Disord. (2014) ;24: (7):574–82.

[322] 

Wren TA , Bluml S , Tseng-Ong L , Gilsanz V . Three-point technique of fat quantification of muscle tissue as a marker of disease progression in Duchenne muscular dystrophy: preliminary study. AJR Am J Roentgenol. (2008) ;190: (1):W8–12.

[323] 

Janssen BH , Voet NB , Nabuurs CI , Kan HE , de Rooy JW , Geurts AC et al. Distinct disease phases in muscles of facioscapulohumeral dystrophy patients identified by MR detected fat infiltration. PLoS One. (2014) ;9: (1):e85416.

[324] 

Hiba B , Richard N , Hébert LJ , Coté C , Nejjari M , Vial C et al. Quantitative assessment of skeletal muscle degeneration inpatients with myotonic dystrophy type 1 using MRI. J Magn ResonImaging. (2012) ;35: (3):678–85.

[325] 

Renard D , Labauge P . Thenar and hypothenar muscle hypertrophy in Becker muscular dystrophy. Neuromuscul Disord. (2010) ;20: (4):281.

[326] 

Fischer D , Hafner P , Rubino D , Schmid M , Neuhaus C , Jung H et al. The 6-minute walk test, motor function measure and quantitative thigh muscle MRI in Becker muscular dystrophy: A cross-sectional study. Neuromuscul Disord. (2016) ;26: (7):414–22.

[327] 

Godi C , Ambrosi A , Nicastro F , Previtali SC , Santarosa C , Napolitano S et al. Longitudinal MRI quantification of muscle degeneration in Duchenne muscular dystrophy. Ann Clin Transl Neurol. (2016) ;3: (8):607–22.

[328] 

Schmidt S , Hafner P , Klein A , Rubino-Nacht D , Gocheva V , Schroeder J et al. Timed function tests, motor function measure, and quantitative thigh muscle MRI in ambulant children with Duchenne muscular dystrophy: A cross-sectional analysis. Neuromuscul Disord. (2018) ;28: (1):16–23.

[329] 

Yin L , Xie ZY , Xu HY , Zheng SS , Wang ZX , Xiao JX et al. T2 Mapping and Fat Quantification of Thigh Muscles in Children with Duchenne Muscular Dystrophy. Curr Med Sci. (2019) ;39: (1):138–45.

[330] 

Finkel RS , Finanger E , Vandenborne K , Sweeney HL , Tennekoon G , Shieh PB et al. Disease-modifying effects of edasalonexent, an NF-κB inhibitor, in young boys with Duchenne muscular dystrophy: Results of the MoveDMD phase 2 and open label extension trial. Neuromuscul Disord. (2021) ;31: (5):385–96.

[331] 

Pichiecchio A , Alessandrino F , Bortolotto C , Cerica A , Rosti C , Raciti MV et al. Muscle ultrasound elastography and MRI in preschool children with Duchenne muscular dystrophy. Neuromuscul Disord. (2018) ;28: (6):476–83.

[332] 

Frisullo G , Frusciante R , Nociti V , Tasca G , Renna R , Iorio R et al. CD8(+) T cells in facioscapulohumeral muscular dystrophy patients with inflammatory features at muscle MRI. J Clin Immunol. (2011) ;8: (2):155–66.

[333] 

Weber MA , Nagel AM , Jurkat-Rott K , Lehmann-Horn F . Sodium (23Na) MRI detects elevated muscular sodium concentration in Duchenne muscular dystrophy. Neurology. (2011) ;77: (23):2017–24.

[334] 

Mankodi A , Bishop CA , Auh S , Newbould RD , Fischbeck KH , Janiczek RL . Quantifying disease activity in fatty-infiltrated skeletal muscle by IDEAL-CPMG in Duchenne muscular dystrophy. Neuromuscul Disord. (2016) ;26: (10):650–8.

[335] 

Hooijmans MT , Niks EH , Burakiewicz J , Verschuuren JJ , Webb AG , Kan HE . Elevated phosphodiester and T(2) levels can be measured in the absence of fat infiltration in Duchenne muscular dystrophy patients. NMR Biomed. (2017) ;30: (1).

[336] 

Mankodi A , Azzabou N , Bulea T , Reyngoudt H , Shimellis H , Ren Y et al. Skeletal muscle water T(2) as a biomarker of disease status and exercise effects in patients with Duchenne muscular dystrophy. Neuromuscul Disord. (2017) ;27: (8):705–14.

[337] 

Keene KR , Beenakker JM , Hooijmans MT , Naarding KJ , Niks EH , Otto LAM et al. T(2) relaxation-time mapping in healthy and diseased skeletal muscle using extended phase graph algorithms. Magn Reson Med. (2020) ;84: (5):2656–70.

[338] 

Hooijmans MT , Froeling M , Koeks Z , Verschuuren J , Webb A , Niks EH et al. Multi-parametric MR in Becker muscular dystrophy patients. NMR Biomed. (2020) ;33: (11):e4385.

[339] 

Araujo ECA , Marty B , Carlier PG , Baudin PY , Reyngoudt H . Multiexponential Analysis of the Water T2-Relaxation in the Skeletal Muscle Provides Distinct Markers of Disease Activity Between Inflammatory and Dystrophic Myopathies. J Magn Reson Imaging. (2021) ;53: (1):181–9.

[340] 

Forbes SC , Willcocks RJ , Triplett WT , Rooney WD , Lott DJ , Wang DJ et al. Magnetic resonance imaging and spectroscopy assessment of lower extremity skeletal muscles in boys with Duchenne muscular dystrophy: a multicenter cross sectional study. PLoS One. (2014) ;9: (9):e106435.

[341] 

Wansapura JP , Hor KN , Mazur W , Fleck R , Hagenbuch S , Benson DW et al. Left ventricular T2 distribution in Duchenne muscular dystrophy. J Cardiovasc Magn Reson. (2010) ;12: (1):14.

[342] 

Bun SS , Kober F , Jacquier A , Espinosa L , Kalifa J , Bonzi MF et al. Value of in vivo T2 measurement for myocardial fibrosis assessment in diabetic mice at 11. 75 T. Invest Radiol. (2012) ;47: (5):319–23.

[343] 

Loganathan R , Bilgen M , Al-Hafez B , Smirnova IV . Characterization of alterations in diabetic myocardial tissue using high resolution MRI. Int J Cardiovasc Imaging. (2006) ;22: (1):81–90.

[344] 

Martins-Bach AB , Bachasson D , Araujo ECA , Soustelle L , Loureiro de Sousa P , Fromes Y et al. Non-invasive assessment of skeletal muscle fibrosis in mice using nuclear magnetic resonance imaging and ultrasound shear wave elastography. Sci Rep. (2021) ;11: (1):284.

[345] 

Hughes MS , Marsh JN , Wallace KD , Donahue TA , Connolly AM , Lanza GM et al. Sensitive ultrasonic detection of dystrophic skeletal muscle in patients with duchenne muscular dystrophy using an entropy-based signal receiver. Ultrasound Med Biol. (2007) ;33: (8):1236–43.

[346] 

Pillen S , Arts IM , Zwarts MJ . Muscle ultrasound in neuromuscular disorders. Muscle Nerve. (2008) ;37: (6):679–93.

[347] 

Forst R , Casser HR . [7MHz real-time sonography of the skeletal musculature in Duchenne muscular dystrophy]. Ultraschall Med. (1985) ;6: (6):336–40.

[348] 

Jansen M , van Alfen N , Nijhuis van der Sanden MW , van Dijk JP , Pillen S , de Groot IJ . Quantitative muscle ultrasound is a promising longitudinal follow-up tool in Duchenne muscular dystrophy. Neuromuscul Disord. (2012) ;22: (4):306–17.

[349] 

Koppaka S , Shklyar I , Rutkove SB , Darras BT , Anthony BW , Zaidman CM et al. Quantitative Ultrasound Assessment of Duchenne Muscular Dystrophy Using Edge Detection Analysis. J Ultrasound Med. (2016) ;35: (9):1889–97.

[350] 

Rutkove SB , Geisbush TR , Mijailovic A , Shklyar I , Pasternak A , Visyak N et al. Cross-sectional evaluation of electrical impedancemyography and quantitative ultrasound for the assessment of Duchennemuscular dystrophy in a clinical trial setting. Pediatr Neurol.. (2014) ;51: (1):88–92.

[351] 

Geisbush TR , Visyak N , Madabusi L , Rutkove SB , Darras BT . Inter-session reliability of electrical impedance myography in children in a clinical trial setting. Clin Neurophysiol. (2015) ;126: (9):1790–6.

[352] 

Rutkove SB , Kapur K , Zaidman CM , Wu JS , Pasternak A , Madabusi L et al. Electrical impedance myography for assessment of Duchenne muscular dystrophy. Ann Neurol. (2017) ;81: (5):622–32.

[353] 

Skalsky AJ , Han JJ , Abresch RT , Shin CS , McDonald CM . Assessment of regional body composition with dual-energy X-ray absorptiometry in Duchenne muscular dystrophy: correlation of regional lean mass and quantitative strength. Muscle Nerve. (2009) ;39: (5):647–51.

[354] 

Ponrartana S , Ramos-Platt L , Wren TA , Hu HH , Perkins TG , Chia JM et al. Effectiveness of diffusion tensor imaging in assessing disease severity in Duchenne muscular dystrophy: preliminary study. Pediatr Radiol. (2015) ;45: (4):582–9.

[355] 

Hooijmans MT , Damon BM , Froeling M , Versluis MJ , Burakiewicz J , Verschuuren JJ et al. Evaluation of skeletal muscle DTI in patients with duchenne muscular dystrophy. NMR Biomed. (2015) ;28: (11):1589–97.

[356] 

Li GD , Liang YY , Xu P , Ling J , Chen YM . Diffusion-Tensor Imaging of Thigh Muscles in Duchenne Muscular Dystrophy: Correlation of Apparent Diffusion Coefficient and Fractional Anisotropy Values With Fatty Infiltration. AJR Am J Roentgenol. (2016) ;206: (4):867–70.

[357] 

McDowell AR , Feiweier T , Muntoni F , Hall MG , Clark CA . Clinically feasible diffusion MRI in muscle: Time dependence and initial findings in Duchenne muscular dystrophy. Magn Reson Med. (2021) ;86: (6):3192–200.

[358] 

Roy B , Darras BT , Zaidman CM , Wu JS , Kapur K , Rutkove SB . Exploring the relationship between electrical impedance myography and quantitative ultrasound parameters in Duchenne muscular dystrophy. Clin Neurophysiol. (2019) ;130: (4):515–20.

[359] 

Arechavala-Gomeza V , Kinali M , Feng L , Brown SC , Sewry C , Morgan JE et al. Immunohistological intensity measurements as a tool to assess sarcolemma-associated protein expression. Neuropathol Appl Neurobiol. (2010) ;36: (4):265–74.

[360] 

Boca SM , Nishida M , Harris M , Rao S , Cheema AK , Gill K et al. Discovery of Metabolic Biomarkers for Duchenne Muscular Dystrophy within a Natural History Study. PLoS One. (2016) ;11: (4):e0153461.

[361] 

Srivastava NK , Annarao S , Sinha N . Metabolic status of patients with muscular dystrophy in early phase of the disease: In vitro, high resolution NMR spectroscopy based metabolomics analysis of serum. Life Sci. (2016) ;151: :122–9.

[362] 

Spitali P , Hettne K , Tsonaka R , Sabir E , Seyer A , Hemerik JBA et al. Cross-sectional serum metabolomic study of multiple forms of muscular dystrophy. J Cell Mol Med. (2018) ;22: (4):2442–8.

[363] 

Boca SM , Nishida M , Harris M , Rao S , Cheema AK , Gill K et al. Correction: Discovery of Metabolic Biomarkers for Duchenne Muscular Dystrophy within a Natural History Study. PLoS One. (2016) ;11: (7):e0159895.

[364] 

Signorelli M , Ebrahimpoor M , Veth O , Hettne K , Verwey N , García-Rodríguez R et al. Peripheral bloodtranscriptome profiling enables monitoring disease progression in dystrophic mice and patients. EMBO Mol Med. (2021) ;13: (4):e13328.

[365] 

Trifunov S , Natera-de Benito D , Exposito Escudero JM , Ortez C , Medina J , Cuadras D et al. Longitudinal Study of Three microRNAs in Duchenne Muscular Dystrophy and Becker Muscular Dystrophy. Front Neurol. (2020) ;11: :304.

[366] 

Amor F , Vu Hong A , Corre G , Sanson M , Suel L , Blaie S et al. Cholesterol metabolism is a potential therapeutic target in Duchenne muscular dystrophy. J Cachexia Sarcopenia Muscle. (2021) ;12: (3):677–93.

[367] 

Cacchiarelli D , Legnini I , Martone J , Cazzella V , D’Amico A , Bertini E et al. miRNAs as serum biomarkers for Duchenne muscular dystrophy. EMBO Mol Med. (2011) ;3: (5):258–65.

[368] 

Dang UJ , Ziemba M , Clemens PR , Hathout Y , Conklin LS , Hoffman EP . Serum biomarkers associated with baseline clinical severity in youngsteroid-naïve Duchenne muscular dystrophy boys. Hum Mol Genet. (2020) ;29: (15):2481–95.

[369] 

Signorelli M , Ayoglu B , Johansson C , Lochmüller H , Straub V , Muntoni F et al. Longitudinal serum biomarkerscreening identifies malate dehydrogenase 2 as candidate prognostic biomarker for Duchenne muscular dystrophy. JCachexia Sarcopenia Muscle.. (2020) ;11: (2):505–17.

[370] 

Ayoglu B , Chaouch A , Lochmüller H , Politano L , Bertini E , Spitali P et al. Affinity proteomics within rare diseases: a BIO-NMD study for blood biomarkers of muscular dystrophies. EMBO Mol Med. (2014) ;6: (7):918–36.

[371] 

Hathout Y , Brody E , Clemens PR , Cripe L , DeLisle RK , Furlong P et al. Large-scale serum protein biomarker discovery in Duchenne muscular dystrophy. Proc Natl Acad Sci U S A. (2015) ;112: (23):7153–8.

[372] 

Spitali P , Hettne K , Tsonaka R , Charrout M , van den Bergen J , Koeks Z et al. Tracking disease progression non-invasively in Duchenne and Becker muscular dystrophies. J Cachexia Sarcopenia Muscle. (2018) ;9: (4):715–26.

[373] 

Alayi TD , Tawalbeh SM , Ogundele M , Smith HR , Samsel AM , Barbieri ML et al. Tandem Mass Tag-Based Serum Proteome Profiling for Biomarker Discovery in Young Duchenne Muscular Dystrophy Boys. ACS Omega. (2020) ;5: (41):26504–17.

[374] 

Veidal SS , Vassiliadis E , Bay-Jensen AC , Tougas G , Vainer B , Karsdal MA . Procollagen type I N-terminal propeptide (PINP) is a marker for fibrogenesis in bile duct ligation-induced fibrosis in rats. Fibrogenesis Tissue Repair. (2010) ;3: (1):5.

[375] 

Leeming DJ , Byrjalsen I , Jiménez W , Christiansen C , Karsdal MA . Protein fingerprinting of the extracellular matrix remodelling in arat model of liver fibrosis–a serological evaluation. Liver Int. (2013) ;33: (3):439–47.

[376] 

Nielsen MJ , Nedergaard AF , Sun S , Veidal SS , Larsen L , Zheng Q et al. The neo-epitope specific PRO-C3 ELISA measures true formation of type III collagen associated with liver and muscle parameters. Am J Transl Res. (2013) ;5: (3):303–15.

[377] 

Hu J , Kong M , Ye Y , Hong S , Cheng L , Jiang L . Serum miR-206 and other muscle-specific microRNAs as non-invasive biomarkers for Duchenne muscular dystrophy. J Neurochem. (2014) ;129: (5):877–83.

[378] 

Zaharieva IT , Calissano M , Scoto M , Preston M , Cirak S , Feng L et al. Dystromirs as serum biomarkers for monitoring the disease severity in Duchenne muscular Dystrophy. PLoS One. (2013) ;8: (11):e80263.

[379] 

Nadarajah VD , van Putten M , Chaouch A , Garrood P , Straub V , Lochmüller H et al. Serum matrix metalloproteinase-9 (MMP-9) as a biomarker for monitoring disease progression in Duchenne muscular dystrophy (DMD). Neuromuscul Disord. (2011) ;21: (8):569–78.

[380] 

Cynthia Martin F , Hiller M , Spitali P , Oonk S , Dalebout H , Palmblad M et al. Fibronectin is a serum biomarker for Duchenne muscular dystrophy. Proteomics Clin Appl. (2014) ;8: (3-4):269–78.

[381] 

Nakagawa T , Takeuchi A , Kakiuchi R , Lee T , Yagi M , Awano H et al. A prostaglandin D2 metabolite is elevated in the urine of Duchenne muscular dystrophy patients and increases further from 8 years old. Clin Chim Acta. (2013) ;423: :10–4.

[382] 

Ellero-Simatos S , Szymańska E , Rullmann T , Dokter WH , Ramaker R , Berger R et al. Assessing the metabolic effects of prednisolone inhealthy volunteers using urine metabolic profiling. Genome Med.. (2012) ;4: (11):94.

[383] 

Wang L , Xu M , Liu D , Liang Y , Feng P , Li H et al. Serum creatinine as a biomarker for dystrophinopathy: a cross-sectional and longitudinal study. BMC Neurol. (2021) ;21: (1):372.

[384] 

Strandberg K , Ayoglu B , Roos A , Reza M , Niks E , Signorelli M et al. Blood-derived biomarkers correlate with clinical progression in Duchenne muscular dystrophy. J Neuromuscul Dis. (2020) ;7: (3):231–46.

[385] 

Tawalbeh S , Samsel A , Gordish-Dressman H , Hathout Y , Investigators CD , Dang UJ . Comparison of Serum Pharmacodynamic Biomarkers in Prednisone-Versus Deflazacort-Treated Duchenne Muscular Dystrophy Boys. J Pers Med. (2020) ;10: (4).

[386] 

Goldstein RA . Skeletal Muscle Injury Biomarkers: Assay Qualification Efforts and Translation to the Clinic. Toxicol Pathol. (2017) ;45: (7):943–51.

[387] 

United States Food and Drug Administration (FDA): Center for Drug Evaluation and Research (CDER) and Center for Biologics Evaluation and Research (CBER). Duchenne Muscular Dystrophy and Related Dystrophinopathies: Developing Drugs for Treatment Guidance for Industry. FDA; 2018.

[388] 

United States Food and Drug Administration (FDA): Center for Drug Evaluation and Research (CDER) and Center for Biologics Evaluation and Research (CBER). Guidance for Clinical Trial Sponsors Establishment and Operation of Clinical Trial Data Monitoring Committees. FDA; 2006.

[389] 

Wallach JD , Zhang AD , Skydel JJ , Bartlett VL , Dhruva SS , Shah ND et al. Feasibility of Using Real-world Data to Emulate Postapproval Confirmatory Clinical Trials of Therapeutic Agents Granted US Food and Drug Administration Accelerated Approval. JAMA Network Open. (2021) ;4: (11):e2133667.

[390] 

Mathews KD , Conway KM , Gedlinske AM , Johnson N , Street N , Butterfield RJ et al. Characteristics of Clinical Trial Participants with Duchenne Muscular Dystrophy: Data from the Muscular Dystrophy Surveillance, Tracking, and Research Network (MD STARnet). Children (Basel). (2021) ;8: (10).

[391] 

McDonald CM , Abresch RT , Carter GT , Fowler WM Jr , Johnson ER , Kilmer DD . Profiles of neuromuscular diseases. Becker’s muscular dystrophy. Am J Phys Med Rehabil. (1995) ;74: (5 Suppl):S93–103.

[392] 

Eagle M , Baudouin SV , Chandler C , Giddings DR , Bullock R , Bushby K . Survival in Duchenne muscular dystrophy: improvements in life expectancy since 1967 and the impact of home nocturnal ventilation. Neuromuscul Disord. (2002) ;12: (10):926–9.

[393] 

Kieny P , Chollet S , Delalande P , Le Fort M , Magot A , Pereon Y et al. Evolution of life expectancy of patients with Duchenne muscular dystrophy at AFM Yolaine de Kepper centre between 1981 and 2011. Ann Phys Rehabil Med. (2013) ;56: (6):443–54.

[394] 

Landfeldt E , Thompson R , Sejersen T , McMillan HJ , Kirschner J , Lochmüller H . Life expectancy at birth in Duchenne muscular dystrophy: a systematic review and meta-analysis. Eur J Epidemiol. (2020) ;35: (7):643–53.

[395] 

Schram G , Fournier A , Leduc H , Dahdah N , Therien J , Vanasse M et al. All-cause mortality and cardiovascular outcomes with prophylactic steroid therapy in Duchenne muscular dystrophy. J Am Coll Cardiol. (2013) ;61: (9):948–54.

[396] 

Birnkrant DJ , Ararat E , Mhanna MJ . Cardiac phenotype determines survival in Duchenne muscular dystrophy. Pediatr Pulmonol. (2016) ;51: (1):70–6.

[397] 

Wang M , Birnkrant DJ , Super DM , Jacobs IB , Bahler RC . Progressive left ventricular dysfunction and long-term outcomes in patients with Duchenne muscular dystrophy receiving cardiopulmonary therapies. Open Heart. (2018) ;5: (1):e000783.

[398] 

Voleti S , Olivieri L , Hamann K , Gordish-Dressman H , Spurney C . Troponin I Levels Correlate with Cardiac MR LGE and Native T1 Values in Duchenne Muscular Dystrophy Cardiomyopathy and Identify Early Disease Progression. Pediatr Cardiol. (2020) ;41: (6):1173–9.

[399] 

Hor KN , Wansapura J , Markham LW , Mazur W , Cripe LH , Fleck R et al. Circumferential strain analysis identifies strata of cardiomyopathy in Duchenne muscular dystrophy: a cardiac magnetic resonance tagging study. J Am Coll Cardiol. (2009) ;53: (14):1204–10.

[400] 

Silva MC , Meira ZM , Gurgel Giannetti J , da Silva MM , Campos AF , Barbosa Mde M et al. Myocardial delayed enhancement by magnetic resonance imaging in patients with muscular dystrophy. J Am Coll Cardiol. (2007) ;49: (18):1874–9.

[401] 

Siddiqui S , Alsaied T , Henson SE , Gandhi J , Patel P , Khoury P et al. Left Ventricular Magnetic Resonance Imaging Strain Predicts the Onset of Duchenne Muscular Dystrophy-Associated Cardiomyopathy. Circ Cardiovasc Imaging. (2020) ;13: (11):e011526.

[402] 

Sheybani A , Crum K , Raucci FJ , Burnette WB , Markham LW , Soslow JH . Duchenne muscular dystrophy patients: troponin leak in asymptomaticand implications for drug toxicity studies. Pediatr Res. (2021) ;92: (6):1613–1620.

[403] 

Trucco F , Domingos JP , Tay CG , Ridout D , Maresh K , Munot P et al. Cardiorespiratory Progression Over 5 Years and Role of Corticosteroids in Duchenne Muscular Dystrophy: A Single-Site Retrospective Longitudinal Study. Chest. (2020) ;158: (4):1606–16.

[404] 

Fayssoil A , Yaou RB , Ogna A , Leturcq F , Nardi O , Clair B et al. Clinical profiles and prognosis of acute heart failure in adult patients with dystrophinopathies on home mechanical ventilation. ESC Heart Fail. (2017) ;4: (4):527–34.

[405] 

Matsumura T , Saito T , Fujimura H , Sakoda S . [Renal dysfunction is a frequent complication in patients with advanced stage of Duchenne muscular dystrophy]. Rinsho Shinkeigaku. (2012) ;52: (4):211–7.

[406] 

Porcher R , Desguerre I , Amthor H , Chabrol B , Audic F , Rivier F et al. Association between prophylactic angiotensin-converting enzyme inhibitors and overall survival in Duchenne muscular dystrophy-analysis of registry data. Eur Heart J. (2021) ;42: (20):1976–84.

[407] 

Raman SV , Hor KN , Mazur W , Cardona A , He X , Halnon N et al. Stabilization of Early Duchenne Cardiomyopathy WithAldosterone Inhibition: Results of the Multicenter AIDMD Trial. J Am Heart Assoc. (2019) ;8: (19):e013501.

[408] 

Florian A , Ludwig A , Rösch S , Yildiz H , Sechtem U , Yilmaz A . Myocardial fibrosis imaging based on T1-mapping and extracellular volume fraction (ECV) measurement in muscular dystrophy patients: diagnostic value compared with conventional late gadolinium enhancement (LGE) imaging. Eur Heart J Cardiovasc Imaging. (2014) ;15: (9):1004–12.

[409] 

Giglio V , Puddu PE , Camastra G , Sbarbati S , Della Sala SW , Ferlini A et al. Patterns of late gadolinium enhancement in Duchenne muscular dystrophy carriers. J Cardiovasc Magn Reson. (2014) ;16: (1):45.

[410] 

Aikawa T , Takeda A , Oyama-Manabe N , Naya M , Yamazawa H , Koyanagawa K et al. Progressive left ventricular dysfunction and myocardial fibrosis in Duchenne and Becker muscular dystrophy: a longitudinal cardiovascular magnetic resonance study. Pediatr Cardiol. (2019) ;40: (2):384–92.

[411] 

Silva MC , Magalhães TA , Meira ZM , Rassi CH , Andrade AC , Gutierrez PS et al. Myocardial Fibrosis Progression in Duchenne andBecker Muscular Dystrophy: A Randomized Clinical Trial. JAMACardiol. (2017) ;2: (2):190–9.

[412] 

Villa CR , Czosek RJ , Ahmed H , Khoury PR , Anderson JB , Knilans TK et al. Ambulatory Monitoring and Arrhythmic Outcomes in Pediatric andAdolescent Patients With Duchenne Muscular Dystrophy. J Am Heart Assoc. (2015) ;5: (1):e002620.

[413] 

Chiang DY , Allen HD , Kim JJ , Valdes SO , Wang Y , Pignatelli RH et al. Relation of Cardiac Dysfunction to Rhythm Abnormalities in Patients With Duchenne or Becker Muscular Dystrophies. Am J Cardiol. (2016) ;117: (8):1349–54.

[414] 

Menon SC , Etheridge SP , Liesemer KN , Williams RV , Bardsley T , Heywood MC et al. Predictive value of myocardial delayed enhancement in Duchenne muscular dystrophy. Pediatr Cardiol. (2014) ;35: (7):1279–85.

[415] 

Sanyal SK , Johnson WW , Dische MR , Pitner SE , Beard C . Dystrophic degeneration of papillary muscle and ventricular myocardium. A basis for mitral valve prolapse in Duchenne’s muscular dystrophy. Circulation. (1980) ;62: (2):430–8.

[416] 

Frankel KA , Rosser RJ . The pathology of the heart in progressive muscular dystrophy: epimyocardial fibrosis. Hum Pathol. (1976) ;7: (4):375–86.

[417] 

Moriuchi T , Kagawa N , Mukoyama M , Hizawa K . Autopsy analyses of the muscular dystrophies. Tokushima J Exp Med. (1993) ;40: (1-2):83–93.

[418] 

Adorisio R , Calvieri C , Cantarutti N , D’Amico A , Catteruccia M , Bertini E et al. Heart rate reduction strategy using ivabradine in end-stage Duchenne cardiomyopathy. Int J Cardiol. (2019) ;280: :99–103.

[419] 

Raman SV , Hor KN , Mazur W , Halnon NJ , Kissel JT , He X et al. Eplerenone for early cardiomyopathy in Duchenne muscular dystrophy: a randomised, double-blind, placebo-controlled trial. Lancet Neurol. (2015) ;14: (2):153–61.

[420] 

Taylor M , Jefferies J , Byrne B , Lima J , Ambale-Venkatesh B , Ostovaneh MR et al. Cardiac and skeletal muscle effects in the randomized HOPE-Duchenne trial. Neurology. (2019) ;92: (8):e866–e78.

[421] 

Allen HD , Flanigan KM , Thrush PT , Dvorchik I , Yin H , Canter C et al. A randomized, double-blind trial of lisinopril and losartan for the treatment of cardiomyopathy in duchenne muscular dystrophy. PLoS Curr. 2013;5.

[422] 

Soslow JH , Xu M , Slaughter JC , Stanley M , Crum K , Markham LW et al. Evaluation of Echocardiographic Measures of Left Ventricular Function in Patients with Duchenne Muscular Dystrophy: Assessment of Reproducibility and Comparison to Cardiac Magnetic Resonance Imaging. J Am Soc Echocardiogr. (2016) ;29: (10):983–91.

[423] 

Wittlieb-Weber CA , Knecht KR , Villa CR , Cunningham C , Conway J , Bock MJ et al. Risk Factors for Cardiac and Non-cardiac Causes of Death in Males with Duchenne Muscular Dystrophy. Pediatr Cardiol. (2020) ;41: (4):764–71.

[424] 

Chenard AA , Becane HM , Tertrain F , de Kermadec JM , Weiss YA . Ventricular arrhythmia in Duchenne muscular dystrophy: prevalence, significance and prognosis. Neuromuscul Disord. (1993) ;3: (3):201–6.

[425] 

Melacini P , Vianello A , Villanova C , Fanin M , Miorin M , Angelini C et al. Cardiac and respiratory involvement in advanced stage Duchenne muscular dystrophy. Neuromuscul Disord. (1996) ;6: (5):367–76.

[426] 

Spurney CF , Ascheim D , Charnas L , Cripe L , Hor K , King N et al. Current state of cardiac troponin testing in Duchenne muscular dystrophy cardiomyopathy: review and recommendations from the Parent Project Muscular Dystrophy expert panel. Open Heart. (2021) ;8: (1).

[427] 

Feingold B , Mahle WT , Auerbach S , Clemens P , Domenighetti AA , Jefferies JL et al. Management of Cardiac Involvement Associated With Neuromuscular Diseases: A Scientific Statement From the American Heart Association. Circulation.. (2017) ;136: (13):e200–e31.

[428] 

Viollet L , Gailey S , Thornton DJ , Friedman NR , Flanigan KM , Mahan JD et al. Utility of cystatin C to monitor renal function in Duchenne muscular dystrophy. Muscle Nerve. (2009) ;40: (3):438–42.

[429] 

Villa CR , Kaddourah A , Mathew J , Ryan TD , Wong BL , Goldstein SL et al. Identifying evidence of cardio-renal syndrome in patients with Duchenne muscular dystrophy using cystatin C. Neuromuscul Disord. (2016) ;26: (10):637–42.

[430] 

Duboc D , Meune C , Lerebours G , Devaux JY , Vaksmann G , Bécane HM . Effect of perindopril on the onset andprogression of left ventricular dysfunction in Duchenne muscular dystrophy. J Am Coll Cardiol.. (2005) ;45: (6):855–7.

[431] 

Nance ME , Shi R , Hakim CH , Wasala NB , Yue Y , Pan X et al. AAV9 Edits Muscle Stem Cells in Normal and Dystrophic Adult Mice. Mol Ther. (2019) ;27: (9):1568–85.

[432] 

Verdera HC , Kuranda K , Mingozzi F . AAV Vector Immunogenicity in Humans: A Long Journey to Successful Gene Transfer. Mol Ther. (2020) ;28: (3):723–46.

[433] 

United States Food and Drug Administration (FDA): Center for Biologics Evaluation and Research (CBER). Human Gene Therapy for Neurodegenerative Diseases. Draft Guidance for Industry. Silver Spring, MD: FDA; 2021.

[434] 

United States Food and Drug Administration (FDA): Center for Biologics Evaluation and Research (CBER). Considerations for the Design of Early-Phase Clinical Trials of Cellular and Gene Therapy Products: Guidance for Industry. FDA; 2015.

[435] 

United States Food and Drug Administration (FDA): Center for Biologics Evaluation and Research (CBER). BRIEFING DOCUMENT. Cellular, Tissue, and Gene Therapies Advisory Committee (CTGTAC) Meeting #70. Toxicity Risks of Adeno-associated Virus (AAV) Vectors for Gene Therapy. FDA; 2021.

[436] 

United States Food and Drug Administration (FDA). In Vitro Companion Diagnostic Devices: Guidance for Industry and Food and Drug Administration Staff. FDA; 2014.

[437] 

United States Food and Drug Administration (FDA): Center for Biologics Evaluation and Research (CBER). Long Term Follow-Up After Administration of Human Gene Therapy Products: Guidance for Industry. FDA; 2020.

[438] 

United States Food and Drug Administration (FDA): Center for Drug Evaluation and Research (CDER) and Center for Biologics Evaluation and Research (CBER). Guidance for industry: Expedited Programs for Serious Conditions—Drugs and Biologics. Silver Spring, MD: FDA: CDER, CBER; 2014.