Skip to main content
The Cochrane Database of Systematic Reviews logoLink to The Cochrane Database of Systematic Reviews
. 2021 Nov 8;2021(11):CD013720. doi: 10.1002/14651858.CD013720.pub2

Antioxidants to prevent respiratory decline in people with Duchenne muscular dystrophy and progressive respiratory decline

Luis Garegnani 1,, Martin Hyland 2, Pablo Roson Rodriguez 3, Camila Micaela E Escobar Liquitay 4, Juan VA Franco 1
Editor: Cochrane Neuromuscular Group
PMCID: PMC8574769  PMID: 34748221

Abstract

Background

Duchenne muscular dystrophy (DMD) is an X‐linked recessive disorder characterised by progressive muscle weakness beginning in early childhood. Respiratory failure and weak cough develop in all patients as a consequence of muscle weakness leading to a risk of atelectasis, pneumonia, or the need for ventilatory support. There is no curative treatment for DMD. Corticosteroids are the only pharmacological intervention proven to delay the onset and progression of muscle weakness and thus respiratory decline in DMD. Antioxidant treatment has been proposed to try to reduce muscle weakness in general, and respiratory decline in particular. 

Objectives

To assess the effects of antioxidant agents on preventing respiratory decline in people with Duchenne muscular dystrophy during the respiratory decline phase of the condition. 

Search methods

We searched CENTRAL, MEDLINE, Embase, and two trials registers to 23 March 2021, together with reference checking, citation searching, and contact with study authors to identify additional studies.

Selection criteria

We included randomised controlled trials (RCTs) and quasi‐RCTs that met our inclusion criteria. We included male patients with a diagnosis of DMD who had respiratory decline evidenced by a forced vital capacity (FVC%) less than 80% but greater than 30% of predicted values, receiving any antioxidant agent compared with other therapies for the management of DMD or placebo. 

Data collection and analysis

Two review authors screened studies for eligibility, assessed risk of bias of studies, and extracted data. We used standard methods expected by Cochrane. We assessed the certainty of the evidence using the GRADE approach. The primary outcomes were FVC and hospitalisation due to respiratory infections. Secondary outcomes were quality of life, adverse events, change in muscle function, forced expiratory volume in the first second (FEV1), and peak expiratory flow (PEF). 

Main results

We included one study with 66 participants who were not co‐treated with corticosteroids, which was the only study to contribute data to our main analysis. We also included a study that enrolled 255 participants treated with corticosteroids, which was only available as a press release without numerical results. The studies were parallel‐group RCTs that assessed the effect of idebenone on respiratory function compared to placebo. The trial that contributed numerical data included patients with a mean (standard deviation) age of 14.3 (2.7) years at the time of inclusion, with a documented diagnosis of DMD or severe dystrophinopathy with clinical features consistent with typical DMD. The overall risk of bias across most outcomes was similar and judged as 'low'. 

Idebenone may result in a slightly less of a decline in FVC from baseline to one year compared to placebo (mean difference (MD) 3.28%, 95% confidence interval (CI) −0.41 to 6.97; 64 participants; low‐certainty evidence), and probably has little or no effect on change in quality of life (MD −3.80, 95% CI −10.09 to 2.49; 63 participants; moderate‐certainty evidence) (Pediatric Quality of Life Inventory (PedsQL), range 0 to 100, 0 = worst, 100 = best quality of life). As a related but secondary outcome, idebenone may result in less of a decline from baseline in FEV1 (MD 8.28%, 95% CI 0.89 to 15.67; 53 participants) and PEF (MD 6.27%, 95% CI 0.61 to 11.93; 1 trial, 64 participants) compared to placebo.

Idebenone was associated with fewer serious adverse events (RR 0.42, 95% CI 0.09 to 2.04; 66 participants; low‐certainty evidence) and little to no difference in non‐serious adverse events (RR 1.00, 95% CI 0.88 to 1.13; 66 participants; low‐certainty evidence) compared to placebo. Idebenone may result in little to no difference in change in arm muscle function (MD −2.45 N, 95% CI −8.60 to 3.70 for elbow flexors and MD −1.06 N, 95% CI −6.77 to 4.65 for elbow extensors; both 52 participants) compared to placebo. We found no studies evaluating the outcome hospitalisation due to respiratory infection. 

The second trial, involving 255 participants, for which data were available only as a press release without numerical data, was prematurely discontinued due to futility after an interim efficacy analysis based on FVC. There were no safety concerns.

The certainty of the evidence was low for most outcomes due to imprecision and publication bias (the lack of a full report of the larger trial, which was prematurely terminated).

Authors' conclusions

Idebenone is the only antioxidant agent tested in RCTs for preventing respiratory decline in people with DMD for which evidence was available for assessment. Idebenone may result in slightly less of a decline in FVC and less of a decline in FEV1 and PEF, but probably has little to no measurable effect on change in quality of life. Idebenone is associated with fewer serious adverse events than placebo. Idebenone may result in little to no difference in change in muscle function.

Discontinuation due to the futility of the SIDEROS trial and its expanded access programmes may indicate that idebenone research in this condition is no longer needed, but we await the trial data. Further research is needed to establish the effect of different antioxidant agents on preventing respiratory decline in people with DMD during the respiratory decline phase of the condition. 

Plain language summary

Antioxidants to prevent respiratory decline in people with Duchenne muscular dystrophy

Review question

What are the effects (benefits and harms) of antioxidants for preventing breathing problems becoming worse in people with Duchenne muscular dystrophy (DMD)?

Background

Duchenne muscular dystrophy is an inherited condition where boys show signs of muscle weakness in early childhood that become worse over time. The muscles used in breathing become involved, which leads to shortness of breath and the need for artificial ventilation (a machine to support breathing). Treatment with antioxidants has been proposed to slow down the loss of muscle strength and the decline in breathing.

Study characteristics

We searched the evidence up to 23 March 2021. We included two studies in boys with DMD whose breathing was affected. Both studies compared the antioxidant medicine idebenone with a dummy medicine. One study included 66 participants between 10 and 18 years of age. The participants in this study were not receiving corticosteroids (medicines shown to be beneficial in DMD). The other study involved 255 children with DMD who were also taking corticosteroids. This study was stopped early for lack of benefit. The full results of the study are not yet available.

Study funding sources

The studies were sponsored by Santhera Pharmaceuticals, the maker of idebenone.

Key results

Idebenone may result in slightly less of a decline in forced vital capacity (a measure of lung capacity), but probably has little or no effect on quality of life in patients with worsening breathing. Idebenone may result in less of a decline in the ability to force air out of the lungs and airways (based on tests of forced expiratory volume in the first second and peak expiratory flow). Idebenone was associated with fewer serious side effects than the dummy drug and has little or no effect on non‐serious side effects. Idebenone may have little or no effect on muscle function (arm strength). We found no studies that looked at hospitalisation due to respiratory infection.

Quality of the evidence

The overall certainty of the evidence was low. 

Summary of findings

Summary of findings 1. Idebenone compared to placebo in people with Duchenne muscular dystrophy and progressive respiratory decline.

Idebenone compared to placebo in people with Duchenne muscular dystrophy and progressive respiratory decline
Patient or population: people with Duchenne muscular dystrophy and progressive respiratory decline  
Setting: outpatients
Intervention: idebenone  
Comparison: placebo  
Outcomes Anticipated absolute effects* (95% CI) Relative effect(95% CI) No. of participants(studies) Certainty of the evidence(GRADE)
Risk with placebo Risk with idebenone
Change in respiratory function from baseline to 12 months, assessed by forced vital capacity (FVC)
Assessed by: change from baseline in per cent of predicted values. 
Follow‐up: 12 months
 
The mean change from baseline in 
FVC was −8.95%.
MD 3.28% higher
(0.41 lower to 6.97 higher)a 64
(1 RCT) ⨁⨁◯◯
LOWb,c
Hospitalisation due to respiratory infection
  No studies measured this outcome.
Quality of life
Assessed by: Pediatric Quality of Life Inventory, change from baseline
(0 to 100; 0 = worst outcome, 100 = best outcome)
Follow‐up: 12 months
The mean change in quality of life was 2.46. MD 3.8 lower
(10.09 lower to 2.49 higher) 63
(1 RCT) ⨁⨁⨁◯
MODERATEb
Serious adverse events
Follow‐up: 12 months
147 per 1000 62 per 1000
(13 to 300) RR 0.42
(0.09 to 2.04)a 66
(1 RCT) ⨁⨁◯◯
LOWc,d
Non‐serious adverse events
Follow‐up: 12 months
941 per 1000 941 per 1000
(828 to 1000) RR 1.00
(0.88 to 1.13)a 66
(1 RCT) ⨁⨁◯◯
LOWc,d
*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: confidence interval; MD: mean difference; RCT: randomised controlled trial; RR: risk ratio
GRADE Working Group grades of evidenceHigh certainty: We are very confident that the true effect lies close to that of the estimate of the effect.
Moderate certainty: We are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low certainty: Our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low certainty: We have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

aAnother study was stopped early due to lack of efficacy in the primary endpoint (FVC) with "no safety concerns". No numerical data were available; this information was reported in a press release (SIDEROS 2021).  
bDowngraded one level due to the small number of participants in the included study.
cDowngraded one level due to publication bias; there were no available data on the SIDEROS 2021 study beyond the press release from the pharmaceutical company.  
dDowngraded one level due to the small number of participants and events in the included study.

Background

Description of the condition

Duchenne muscular dystrophy (DMD) is an X‐linked recessive disorder, caused by deletions, duplications, and mutations in the dystrophin gene leading to either absent or deficient production of the dystrophin protein. It is the most common muscular dystrophy of childhood, with a reported incidence of 1:5000 in male newborns (Mendell 2013). DMD is characterised by progressive muscle weakness beginning in early childhood, which, in the absence of steroid treatment, leads to loss of independent walking between seven and 13 years of age (Birnkrant 2018aBirnkrant 2018b). Cardiac (heart) muscle is affected; progressive dilated cardiomyopathy is present in all patients by 18 years of age and accounts for up to 40% of premature death in the late teens and early 20s. Early aggressive treatment with angiotensin‐converting enzyme (ACE) inhibitors and beta‐blockers may benefit patients (Birnkrant 2018bStromberg 2012). Respiratory failure and weak cough develops in all patients as a consequence of muscle weakness in the late teens and early 20s leading to a risk of atelectasis (lung collapse), mucous plugging, and pneumonia. Proactive management using cough augmentation, vaccination against flu and pneumococcal infection, and nighttime non‐invasive ventilation (NIV) are vital components of care for individuals with DMD and can prolong life into the fourth and even fifth decades in some patients. Scoliosis occurs during the adolescent growth spurt and is common in patients not treated with corticosteroids. Most patients with scoliosis will undergo spinal fusion. Severe, untreated scoliosis may exacerbate respiratory complications (LoMauro 2018). Individuals with DMD therefore require periodic pulmonary function testing and sleep studies to monitor for nocturnal hypoventilation, as well as the use of lung volume recruitment and assisted coughing. NIV with bilevel positive airway pressure (BiPAP) at nighttime, progressing with advancing age to daytime NIV, is ultimately required for all patients (Birnkrant 2018b). 

There is at present no curative treatment for DMD. Corticosteroids are the only pharmacological intervention proven to delay the onset and progression of muscle weakness and thus respiratory decline in DMD (Matthews 2016). However, long‐term corticosteroid use leads to significant unacceptable side effects, in particular osteoporosis, growth and pubertal delay, obesity, and behavioural and mood issues, so that by 18 years of age more than 40% of patients will have stopped steroid treatment (Henricson 2013).

Description of the intervention

Dystrophin is a large cytoskeletal protein located on the cytoplasmic membrane of the sarcolemma (cell membrane of skeletal and cardiac muscle fibres). It plays a complex role in maintaining the integrity of the muscle fibre, providing structural reinforcement and stabilisation of the glycoprotein complex found within the sarcolemma, protecting it from degradation during muscle contraction. Dystrophin also plays a role in regulating neuronal nitric oxide synthase (nNOS) (Petrof 1993), which is involved in muscle blood flow to prevent early muscle fatigue during exercise (Sander 2000). Sarcolemmal fragility also leads to intracellular calcium dysregulation, a consequence of which is mitochondrial dysfunction leading to a reduction in resting adenosine triphosphate (ATP) levels, increased production of cell‐damaging reactive oxygen species (ROS), and ultimately mitochondrial damage (Brookes 2004Cardoso 1999Peng 2010). Thus, when dystrophin is deficient, muscle fibres necrose and degenerate through a complex pathway of apoptosis which ultimately causes muscle wasting and weakness (Aslesh 2018Bellinger 2009Mázala 2015Petrof 1993Turner 1988). Treatment with antioxidants has been proposed to try to reduce secondary mitochondrial damage and therefore reduce secondary apoptosis. The mdx mouse, a strain of mice arising from a spontaneous X chromosome‐linked mutation that produces lack of muscle dystrophin and possesses histological lesions similar to human muscular dystrophy, was used as an animal model for the following interventions.

  1. N‐acetylcysteine 2% solution was added to drinking water of mdx mice for six weeks and demonstrated some improvements in muscle function (Head 2017).

  2. A combination of alpha‐lipoic acid and L‐carnitine decreased plasma creatine kinase (CK) levels and decreased muscle fibre central nucleation and variations in fibre size, antioxidant activity, lipid peroxidation, and matrix metalloproteinase activity in a diaphragm preparation (Hnia 2007).

  3. Similar findings occurred with ascorbic acid (vitamin C), coenzyme Q10 (CoQ10), vitamin E, cilostazol, diacerein, and melatonin, which decreased plasma CK levels and diaphragm myonecrosis and inflammation (Hermes 2016Hibaoui 2011Mâncio 2017aMâncio 2017bTonon 2012Woodman 2016). Epigallocatechin‐3‐gallate, a green tea extract, is described as having a similar effect (Nakae 2008).

  4. Pentoxifylline and sildenafil have been found to increase diaphragm force‐generating capacity (Burdi 2009Percival 2012).

  5. Idebenone and L‐arginine reduce muscle inflammation and fibrosis (Buyse 2015Marques 2010).

However, considering the generally accepted limitations of animal models, any potential benefits need to be confirmed in people with DMD. 

How the intervention might work

Free‐radical accumulation and oxidative stress have been proposed as contributing factors to the progression of DMD. Markers of oxidative stress including by‐products of lipid peroxidation and protein oxidation have been found to be elevated in people with DMD (Haycock 1996Rodriguez 2003). Since oxidative stress results from an imbalance between the production and removal of ROS, there is a rationale to consider antioxidant therapy as a potential intervention in DMD for muscle dysfunction in general, and respiratory decline in particular. Antioxidant therapy might be capable of the inhibition of lipid peroxidation and consequent stimulation of mitochondrial electron flux and cellular energy production, thus improving respiratory muscle performance and respiratory function (Buyse 2015).

Why it is important to do this review

Given that there is no cure for DMD, and that corticosteroid use is limited by potential and real significant side effects and has a variable response, there is a need to look for other interventions that might reduce the effect of secondary muscle damage caused by dystrophin deficiency, such as antioxidants to protect mitochondrial function. Although the efficacy of antioxidant therapy has been reviewed for other neuromuscular diseases (e.g. amyotrophic lateral sclerosis) (Orrell 2007), to our knowledge there is no high‐quality systematic review evaluating the effectiveness of antioxidants to reduce respiratory decline in DMD.

Objectives

To assess the effects of antioxidant agents on preventing respiratory decline in people with Duchenne muscular dystrophy during the respiratory decline phase of the condition.

Methods

Criteria for considering studies for this review

Types of studies

We included randomised controlled trials (RCTs) and quasi‐RCTs that met our inclusion criteria. We did not include cluster‐ or cross‐over RCTs as they are not relevant to our review question. We included studies reported as full text, those published as abstract only, and unpublished data where it was possible to establish eligibility for inclusion based on limited data. There were no language restrictions. 

We included studies that included both eligible and ineligible participants (e.g. a subset of participants in the respiratory decline phase of DMD) if the study authors presented separate data for eligible participants.

Types of participants

We included male participants with a diagnosis of Duchenne muscular dystrophy (DMD) with respiratory decline evidenced by a forced vital capacity (FVC%) < 80% and > 30% of predicted values.

FVC% > 80%, regardless of age, is considered normal (Miller 2005), and FVC < 30% represents a major decline in respiratory function probably requiring ventilatory support, where antioxidants are highly unlikely to be of value (Lyager 1995).

We included participants who were being treated with corticosteroids as well as those who were corticosteroid naïve or who stopped corticosteroid treatment for any reason (Matthews 2016). 

Types of interventions

We included trials comparing antioxidant agents (given alone or in combination) with other therapies for the management of DMD (e.g. corticosteroids, physical therapy, respiratory muscle training, angiotensin‐converting enzyme (ACE) inhibitors) or with placebo. We included head‐to‐head comparisons between antioxidant agents. 

Co‐interventions (e.g. corticosteroids) were allowed provided that they were provided to each group equally.

We included any of the antioxidant agents used in animal studies as above. Our inclusion criteria included, but were not limited to, the following.

  1. Idebenone

  2. Coenzyme Q10

  3. Green tea extract

  4. N‐acetylcysteine

  5. Metformin

  6. Citrulline

We also included any other intervention considered to have action as an antioxidant identified through our search strategy.

Types of outcome measures

The outcomes listed here were not inclusion criteria for the review. A minimal clinically important difference (MCID) was not available for all of the outcomes considered. We reported an MCID when it was available.    

Primary outcomes
  • Change in respiratory function from baseline to 12 months or beyond, assessed as forced vital capacity (FVC): measured with standard spirometry at hospital visits with the aid of a qualified, trained, and certified operator and in accordance with the American Thoracic Society/European Respiratory Society guidelines (Miller 2005), converted to per cent of predicted values according to age, sex, race, and height. We considered an absolute change of 2% for improvement and 6% for worsening of the FVC predicted value as MCID (du Bois 2011).

  • Hospitalisation due to respiratory infections, measured in the number of admissions per year since the beginning of treatment.

Secondary outcomes
  • Quality of life: measured using the EuroQoL‐5D, Wille 2010, or the Pediatric Quality of Life Inventory (PedsQL), Varni 2001, or any other well‐known and validated scale. We considered a mean change of 0.074 in EuroQol‐5D (range 0 to 1; 0 = death, 1 = perfect health) (Walters 2005), and a mean change of 5 in PedsQL (range 0 to 100; 0 = worst outcome, 100 = best outcome) as MCID (Hilliard 2013).

  • Any adverse events: defined as any undesirable or harmful occurrence in a participant from the beginning of treatment. We reported non‐serious and serious adverse events (including death, disability, life‐threatening events, or those requiring hospitalisation) separately.

  • Change in muscle function: assessed by timed functional testing: walking a certain distance as fast as possible, climbing four standardised stairs or rising up from the floor to a standing position without assistance (Phillips 2008). We considered an absolute change of 2.3 seconds for the 10‐metre walk/run test, 2.1 seconds for the climbing stairs test, 3.7 seconds for the supine to stand test, and 31 m for the 6‐minute walk distance timed test as MCID (McDonald 2013). In non‐ambulant participants, we considered the change in pinch, grip and/or wrist flexion and extension strength (Servais 2013), or change in the Motor Function Measure (Bérard 2005), or similar. MCID for the given strength was identified where possible and used in the results (McDonald 2013).

  • Forced expiratory volume in the first second (FEV1): measured with standard spirometry at hospital visits with the aid of a qualified, trained, and certified operator and in accordance with the American Thoracic Society/European Respiratory Society guidelines (Graham 2019), converted to per cent of predicted values according to age, sex, race, and height or z‐scores.

  • Peak expiratory flow (PEF): measured with standard spirometry at hospital visits with the aid of a qualified, trained, and certified operator and in accordance with the American Thoracic Society/European Respiratory Society guidelines (Miller 2005), converted to per cent of predicted values according to age, sex, race, and height.

Timing of outcome measurement

We considered outcomes measured up to and including six months after randomisation as short term, from six to 12 months after randomisation as medium term, and more than 12 months as long term. When multiple results were reported for a given outcome, we included the longest follow‐up in each category.

Search methods for identification of studies

Electronic searches

The review authors prepared and ran the search strategies.  We also identified trials from the Cochrane Neuromuscular Specialised Register, which is maintained by the Information Specialist for the Group. We searched the following databases (strategies provided in the appendices):

  • Cochrane Central Register of Controlled Trials (CENTRAL) searched 23 March 2021 (Appendix 1);

  • MEDLINE (PubMed; 1945 to present) searched 23 March 2021 (Appendix 2);

  • Embase (Elsevier; 1947 to present) searched 23 March 2021 (Appendix 3).

We also conducted a search of the following clinical trials registries (Appendix 4):

  • US National Institutes of Health Ongoing Trials Register ClinicalTrials.gov (www.clinicaltrials.gov) searched 23 March 2021;

  • World Health Organization International Clinical Trials Registry Platform (apps.who.int/trialsearch/) searched 23 March 2021.

We searched all databases from their inception to the date of search, and imposed no restrictions on language of publication or publication status. We did not use a filter for RCTs in these searches as we expected a low recall.

Searching other resources

We searched the reference lists of all primary studies and reviewed articles for additional references. We searched relevant manufacturers' websites for trial information. We searched for errata or retractions from the included studies. We contacted the authors of the included studies to identify other unpublished studies.  

Data collection and analysis

Selection of studies

Two review authors (LG, PR) independently screened the titles and abstracts of all the studies identified as a result of the search, coding them as 'retrieve' (eligible or potentially eligible/unclear) or 'do not retrieve'. We retrieved the full‐text study reports or publications, and two review authors (LG, PR) independently screened these and identified studies for inclusion. We identified and recorded reasons for exclusion of the ineligible studies. Any disagreements were resolved through discussion or by consulting a third review author (JF or MH) if necessary. We identified and excluded duplicates and collated multiple reports of the same study so that each study, rather than each report, was the unit of interest in the review. We recorded the selection process in sufficient detail to complete a PRISMA flow diagram, Liberati 2009, and 'Characteristics of excluded studies' table. We used Covidence software for study selection (Covidence). 

Data extraction and management

We used a data extraction form for study characteristics and outcome data. Two review authors (LG, PR) independently extracted study characteristics from the included studies. We extracted the following study characteristics.

  1. Methods: study design, total duration of study, details of any 'run‐in' period, number of study centres and location, study setting, withdrawals, and date of study.

  2. Participants: number of participants, mean age, age range, gender, severity of condition, diagnostic criteria, baseline characteristics, inclusion criteria, and exclusion criteria.

  3. Interventions: intervention, comparison, concomitant medications, and excluded medications.

  4. Outcomes: primary and secondary outcomes specified and collected, and time points reported.

  5. Notes: funding for trial and notable conflicts of interest of trial authors.

Two review authors (LG, PR) independently extracted outcome data from the included studies. We noted in the 'Characteristics of included studies' table if outcome data were not reported in a useable way. Any disagreements were resolved by consensus or by involving a third review author (JF). One review author (LG) transferred data into Review Manager 5 (Review Manager 2020), and a second review author (PR) checked the outcome data entries. A third review author (JF) spot‐checked study characteristics for accuracy against the trial report.

Had we found reports that required translation, the translator would have extracted data directly using a data extraction form, or the review authors would have extracted data from the provided translation. Where possible, a review author would have checked numerical data in the translation against the study report. 

Assessment of risk of bias in included studies

Two review authors (LG, PR or MH) independently assessed the risk of bias for the results of the main outcomes (those included in the summary of findings table, see below) in each included study using the recently developed revision of the Cochrane risk of bias tool, RoB 2 (Higgins 2021bSterne 2019). Any disagreements were resolved by discussion or by involving another review author (JF). We assessed risk of bias according to the following domains, focusing on the effect of assignment to the intervention at baseline:

  1. the randomisation process;

  2. deviations from intended interventions;

  3. missing outcome data;

  4. measurement of the outcome;

  5. selection of the reported results.

Answers to signalling questions and supporting information collectively lead to a domain‐level judgement of 'low risk', 'some concerns', or 'high risk' of bias. These domain‐level judgements informed an overall risk of bias judgement for the outcome. We provided a quote from the study report together with a justification for our judgement in the risk of bias table. We aimed to source published protocols in order to assess selective reporting. Where information on risk of bias related to unpublished data or correspondence with a trialist, we noted this in the risk of bias table.

When considering treatment effects, we took into account the risk of bias for the studies that contributed to that outcome. We made summary assessments of the risk of bias for each important outcome (across domains), within and across studies (Higgins 2021b).

We used the 22 August 2019 RoB 2 Excel tool to manage data supporting the answers to the signalling questions and risk of bias judgements (https://www.riskofbias.info/). All of these data are publicly available as supplementary material in the Open Science Framework platform (osf.io/).

Assessment of bias in conducting the systematic review

We conducted the review according to this published protocol and reported any deviations from it in the Differences between protocol and review section of the systematic review.

Measures of treatment effect

We analysed dichotomous data (adverse events) as risk ratios (RRs) and continuous data (all other outcomes) as mean differences (MDs). We reported corresponding 95% confidence intervals (CIs). We planned to analyse standardised mean differences for pooled results across studies for outcomes that were conceptually the same but measured in different ways. We entered data presented as a scale with a consistent direction of effect.  

We planned to undertake meta‐analyses only where this was meaningful, that is if the treatments, participants, and the underlying clinical question were similar enough for pooling to make sense.

Unit of analysis issues

Where multiple trial arms were reported in a single trial, we would have included only the treatment arms relevant to the review topic. Had two comparisons from the same trial (e.g. drug A versus placebo and drug B versus placebo) been combined in the same meta‐analysis, we would have followed the guidance in Section 6.2 of the Cochrane Handbook for Systematic Reviews of Interventions to avoid double‐counting (Higgins 2021a). Our preferred approach was to combine groups to create a single pair‐wise comparison.

Dealing with missing data

We contacted investigators or study sponsors to verify key study characteristics and to obtain missing numerical outcome data where possible (e.g. when a study was available as an abstract only).

Where numerical outcome data were missing, such as standard deviations or correlation coefficients, and they could not be obtained from the authors, we calculated them from other available statistics such as 95% CI or P values, according to the methods described in the Cochrane Handbook for Systematic Reviews of Interventions (Higgins 2021a). If this was not possible, and the missing data could have introduced serious bias, we would explore the impact of including such studies in the overall assessment of results by a sensitivity analysis. 

Assessment of heterogeneity

We planned to use the I² statistic to measure heterogeneity amongst the trials in each analysis. Had we identified substantial unexplained heterogeneity, we would have reported it and explored possible causes by prespecified subgroup analysis. We would have used the rough guide to interpretation as outlined in Chapter 10 of the Cochrane Handbook for Systematic Reviews of Interventions (Higgins 2021a), as follows:

  • 0% to 40%: might not be important;

  • 30% to 60%: may represent moderate heterogeneity;

  • 50% to 90%: may represent substantial heterogeneity;

  • 75% to 100%: considerable heterogeneity.

We would have avoided the use of absolute cut‐off values, but interpreted I² in relation to the size and direction of effects and strength of evidence for heterogeneity (e.g. P value from the Chi²test, or CI for I²).

Assessment of reporting biases

Had we included more than 10 trials, we would have created and examined a funnel plot to explore possible small‐study biases. Had our searches identified trial protocols, clinical trial registrations, or abstracts indicating the existence of unpublished studies, we would have attempted to determine the status of any unpublished studies through contact with the investigators.

We considered outcome reporting bias in our risk of bias assessment.

Data synthesis

As a general rule, we planned to use a random‐effects model in Review Manager 5, as this is usually a more conservative approach. Had the analyses included both small and large studies, we would have performed a sensitivity analysis to determine whether their results were systematically different, since, in such circumstances, the use of a random‐effects meta‐analysis would exacerbate the effects of the bias. Had they been systematically different, we would have performed a sensitivity analysis in which small studies would have been excluded.

Subgroup analysis and investigation of heterogeneity

We planned to carry out the following subgroup analyses for our primary outcome.

  1. Participants with and without corticosteroids as co‐intervention.

  2. Participants with differing degrees of severity of spinal scoliosis: null or mild (Cobb angle < 20°), moderate (20º to 40º), and severe (> 40º).

We would use the formal test for subgroup interactions in Review Manager 5 (Review Manager 2020).

Sensitivity analysis

We planned the following sensitivity analyses for our primary outcome.

  1. Repeat the analysis excluding unpublished studies (if there were any).

  2. Repeat the analysis excluding studies at an overall high risk of bias for the selected outcome.

Reaching conclusions

We based our conclusions only on the findings from the quantitative or narrative synthesis of the included studies for this review. We avoided making recommendations for practice, and our implications for research suggest priorities for future research and outline the remaining uncertainties in the area.

Summary of findings and assessment of the certainty of the evidence

We created a summary of findings table using GRADEpro GDT software (GRADEpro GDT), presenting the following outcomes.

  1. Change in respiratory function from baseline to 12 months or beyond, assessed by forced vital capacity (FVC): measured with standard spirometry at hospital visits with the aid of a qualified, trained, and certified operator and in accordance with the American Thoracic Society/European Respiratory Society guidelines (Miller 2005), converted to per cent of predicted values according to age, sex, race, and height. We considered an absolute change of 2% for improvement and 6% for worsening of the FVC predicted value as MCID (du Bois 2011).

  2. Hospitalisation due to respiratory infections, measured in the number of admissions per year since the beginning of treatment.

  3. Quality of life from baseline to 12 months or beyond, measured using the EuroQoL‐5D, Wille 2010, or the Pediatric Quality of Life Inventory (PedsQL), Varni 2001, or other well‐known scales. We considered a mean change of 0.074 in EuroQol‐5D (range 0 to 1; 0 = death, 1 = perfect health) (Walters 2005), and a mean change of 5 in PedsQL (range 0 to 100, 0 = worst outcome, 100 = best outcome) as MCID (Hilliard 2013).

  4. Serious adverse events, including death, disability, life‐threatening events, or those requiring hospitalisation.

  5. Non‐serious adverse events.

Two review authors (LG, PR, MH) used the five GRADE considerations (overall risk of bias, consistency of effect, imprecision, indirectness, and publication bias) to independently assess the certainty of the body of evidence (studies that contribute data for the prespecified outcomes). We used the methods and recommendations in Chapters 14 and 15 of the Cochrane Handbook for Systematic Reviews of Interventions (Schünemann 2021aSchünemann 2021b). Any disagreements were resolved by discussion or by involving another review author (JF). We considered RCTs as high‐certainty evidence if the five factors above were not present to any serious degree, but downgraded the certainty of evidence once if a GRADE consideration was present to a serious extent and twice if it was very serious. We justified all decisions to downgrade the certainty of evidence using footnotes and made comments to aid readers' understanding of the review where necessary.

Results

Description of studies

For details of the included studies, see Characteristics of included studies.

Results of the search

We identified 1267 records through database searching and one additional record through other sources, of which 189 records were removed as duplicates. We excluded 1008 records after title and abstract screening and obtained the full texts for 71 studies. Screening the reference lists of the included publications did not reveal any additional RCTs. We excluded 25 studies with 35 records after full‐text assessment (see Characteristics of excluded studies). We identified one ongoing study with four records (see Characteristics of ongoing studies). We finally included two studies reported in 32 records. A study flow diagram illustrating the study selection process is presented in Figure 1.

1.

1

Study flow diagram.

Included studies

We included one study with 66 participants that contributed to our main analysis (DELOS 2015). We also included the results of a study with 255 participants that was only available as a press release (SIDEROS 2021). See Characteristics of included studies.

Design

The included studies were parallel RCTs. 

Sample size

One study included 66 participants (DELOS 2015). The press release of the other study did not state the number of participants; however, the clinical trial registry reported an enrolment of 255 participants (SIDEROS 2021).

Setting

One study was conducted in an outpatient setting (DELOS 2015), with international participating centres in Belgium, Germany, the Netherlands, Switzerland, France, Sweden, Austria, Italy, Spain, and the USA. All the study's reports were written in English. The press release of SIDEROS 2021 did not provide any information related to the setting; however, the clinical trial registry reported 63 centres across the USA, Europe, and Israel.

Participants

One study involved patients not receiving corticosteroid, who were 10 to 18 years of age at inclusion, with a mean (standard deviation (SD)) age of 14.3 (2.7) years and mean (SD) FVC of 52.85 (17.9) of predicted at baseline (DELOS 2015). Participants needed to have a documented diagnosis of DMD or severe dystrophinopathy with clinical features consistent with typical DMD at diagnosis, confirmed by mutation analysis in the dystrophin gene or by substantially reduced levels of dystrophin protein. The trial registry of the other included study stated that they included patients with respiratory decline who were receiving concomitant corticosteroid treatment and excluded those who needed daytime ventilator assistance; however, there was no information on age or respiratory status (SIDEROS 2021).

Interventions

One study delivered 900 mg/day of idebenone or placebo in two tablets of 150 mg each, taken orally three times daily with meals for 52 weeks. No corticosteroid co‐treatment was given (DELOS 2015). The press release of SIDEROS 2021 did not provide any information related to the interventions, but the clinical trial register indicated that the planned interventions were 900 mg/day of idebenone or placebo in two tablets of 150 mg each three times daily.

Outcomes

One study reported the effect of idebenone on change in respiratory function from baseline to 12 months, assessed as FVC (DELOS 2015). It also reported the effects of idebenone on quality of life, adverse events, change in muscle function, FEV1, and PEF. DELOS 2015 did not report on hospitalisation due to respiratory infections. Information on adverse events was reported in different ways across reports. The press release of the other study (without numerical data) stated that the trial was discontinued after an efficacy interim analysis based on FVC due to futility and that there were no safety concerns (SIDEROS 2021).

Funding sources

The included studies were sponsored by Santhera Pharmaceuticals.

Excluded studies

We excluded 25 studies after full‐text assessment.  

Risk of bias in included studies

The risk of bias assessments for each outcome, including all domain judgements and support for judgement, are provided in the risk of bias sections of the Characteristics of included studies tables. To access the detailed risk of bias assessment data, visit https://osf.io/w5dgu/ (DOI 10.17605/OSF.IO/W5DGU). 

The overall risk of bias across most outcomes in DELOS 2015 was 'low'. Some issues arose because allocation concealment might have been overridden in a pair of siblings who were already randomly allocated and who were assigned to the same group as their siblings to avoid mix‐up of study medication, but we did not consider this to be a serious concern regarding bias; our final judgement was therefore 'low'.  There was no bias related to deviations from intended interventions. Missing outcome data was not an issue for outcomes included in the summary of findings table, as data were available for more than 95% of participants, although for the outcomes change in muscle function and FEV1 there were missing data as high as 21.21% and 19.70% of participants, respectively. The methods for outcome measurement were appropriate and equally applied between groups, using standard spirometry for FVC and PedsQL for quality of life. There were no concerns regarding the selection of reported results.

We were unable to assess the risk of bias of the results from the SIDEROS 2021 study as data were only available as a press release.

Effects of interventions

See: Table 1

Idebenone versus placebo

We included the results of one study with 66 participants in this comparison (DELOS 2015). Where possible, we included a narrative statement extracted from the press release of the other study (SIDEROS 2021). See Table 1

Primary outcomes
Change in respiratory function from baseline to 12 months, assessed by forced vital capacity (FVC)

One study reported this outcome (DELOS 2015). Idebenone may result in slightly less of a decline from baseline in FVC compared to placebo at 12 months follow‐up (mean difference (MD) 3.28%, 95% confidence interval (CI) −0.41 to 6.97; 64 participants) (Analysis 1.1). We assessed the evidence for this outcome as of low certainty due to imprecision. The other study stated that the trial was discontinued after an efficacy interim analysis based on FVC due to futility (SIDEROS 2021).

1.1. Analysis.

1.1

Comparison 1: Idebenone versus placebo, Outcome 1: Forced vital capacity (FVC)

Hospitalisation due to respiratory infections

DELOS 2015 did not report the specific outcome ‘hospitalisation due to respiratory infections’. However, the study did state that one participant in the idebenone group and four participants in the placebo group were hospitalised as a result of "respiratory complications". 

Secondary outcomes
Quality of life 

One study reported this outcome (DELOS 2015), measuring the change from baseline in PedsQL (range 0 to 100; 0 = worst outcome, 100 = best outcome). Idebenone probably results in little to no difference in change from baseline in quality of life compared to placebo at 12 months follow‐up (MD −3.80, 95% CI −10.09 to 2.49; 63 participants) (Analysis 1.2). We assessed the evidence for this outcome as of moderate certainty due to imprecision. 

1.2. Analysis.

1.2

Comparison 1: Idebenone versus placebo, Outcome 2: Quality of life

Serious adverse events

DELOS 2015 reported this outcome. Idebenone was associated with a decrease in serious adverse events compared to placebo at 12 months follow‐up (risk ratio (RR) 0.42, 95% CI 0.09 to 2.04; 66 participants) (Analysis 1.3). We assessed the evidence for this outcome as of low certainty due to imprecision and publication bias. The other study stated that there were no safety concerns (no numerical data available) (SIDEROS 2021).

1.3. Analysis.

1.3

Comparison 1: Idebenone versus placebo, Outcome 3: Any adverse events

Non‐serious adverse events

DELOS 2015 reported this outcome. Idebenone was associated with little to no difference in non‐serious adverse events compared to placebo at 12 months follow‐up (RR 1.00, 95% CI 0.88 to 1.13; 66 participants) (Analysis 1.3). We assessed the evidence for this outcome as of low certainty due to imprecision and publication bias. The other study stated that there were no safety concerns (no numerical data available) (SIDEROS 2021)  

Change in muscle function (elbow flexors)

One study reported this outcome (DELOS 2015). Idebenone may result in little to no difference in change in muscle strength function (elbow flexors) compared to placebo at 12 months follow‐up (MD −2.45 N, 95% CI −8.60 to 3.70; 52 participants) (Analysis 1.4). 

1.4. Analysis.

1.4

Comparison 1: Idebenone versus placebo, Outcome 4: Change in muscle function (elbow flexors)

Change in muscle function (elbow extensors)

One study reported this outcome (DELOS 2015). Idebenone may result in little to no difference in change in muscle strength function (elbow extensors) compared to placebo at 12 months follow‐up (MD −1.06 N, 95% CI −6.77 to 4.65; 52 participants) (Analysis 1.5). 

1.5. Analysis.

1.5

Comparison 1: Idebenone versus placebo, Outcome 5: Change in muscle function (elbow extensors)

Forced expiratory volume in the first second (FEV1)

One study reported this outcome (DELOS 2015). Idebenone may result in less of a decline in FEV1 compared to placebo at 12 months follow‐up (MD 8.28%, 95% CI 0.89 to 15.67; 53 participants) (Analysis 1.6). 

1.6. Analysis.

1.6

Comparison 1: Idebenone versus placebo, Outcome 6: Forced expiratory volume in the first second (FEV1)

Peak expiratory flow (PEF)

One study reported this outcome (DELOS 2015). Idebenone may result in less of a decline in PEF compared to placebo at a 12 months follow‐up (MD 6.27%, 95% CI 0.61 to 11.93; 64 participants) (Analysis 1.7). 

1.7. Analysis.

1.7

Comparison 1: Idebenone versus placebo, Outcome 7: Peak expiratory flow (PEF)

Discussion

Summary of main results

We included two studies with a total of 321 participants. One unpublished study including 255 participants with DMD was terminated early for futility, and full results are not yet available. A study with 66 participants assessed the effects of idebenone compared to placebo in participants with DMD and signs of respiratory decline. The certainty of the evidence for all outcomes was low. 

Idebenone may result in slightly less of a decline from baseline in FVC at 12 months follow‐up, and probably results in little to no difference in change in quality of life at 12 months follow‐up. Idebenone was associated with a decrease in serious adverse events and little to no difference in non‐serious adverse events compared to placebo at 12 months follow‐up. Idebenone may result in little to no difference in change in muscle function at 12 months follow‐up. Idebenone may result in less of a decline in FEV1 and PEF at 12 months follow‐up. We found no studies evaluating the outcome hospitalisation due to respiratory infection.

Overall completeness and applicability of evidence

One study included participants 10 to 18 years old with a DMD diagnosis and signs of respiratory decline evidenced by an FVC of less than 60% of predicted with no concomitant corticosteroid treatment (DELOS 2015). The study did not include patients with symptomatic heart failure, symptomatic ventricular arrhythmias, or those using other therapeutic drugs (like carnitine, coenzyme Q10, vitamin E, or herbal medicines) within 90 days prior to inclusion. Participants were also not co‐treated with corticosteroids. The study delivered idebenone orally in two film‐coated tablets of 150 mg each, three times a day with meals for 52 weeks, for a total dose of idebenone of 900 mg/day, which is the same dose used for the drug's approved use in Leber’s hereditary optic neuropathy (EPAR 2015). The included study reported most outcomes of interest in the review, except for hospitalisation due to respiratory infections. We found heterogeneity in adverse events reporting across all study reports, but we were able to extract the adverse events data from the results uploaded in the clinical trial register (ClinicalTrials.gov ID NCT01027884). 

People with DMD often have comorbidities, such as asthma, heart disease, or severe scoliosis that requires spinal surgery or spinal fusion (LoMauro 2018). We did not find studies including participants with these comorbidities, resulting in uncertainty about the applicability of our findings in these populations. Furthermore, corticosteroid therapy in DMD has led to improvements in function, quality of life, health, and longevity (Matthews 2016), but DELOS 2015 did not include patients taking corticosteroids. Participants in the recently stopped trial were receiving corticosteroids (SIDEROS 2021).

There is currently no cure for DMD, making multidisciplinary approaches necessary to provide symptomatic treatment whilst the disease progresses. A 2018 clinical practice guideline proposed that co‐ordination of clinical care is a crucial component of DMD management, and recommended that it should include a wide range of healthcare professionals and interventions considering rehabilitation, orthopaedic, psychosocial, cardiac, gastrointestinal, speech, and corticosteroid and respiratory management (Birnkrant 2018aBirnkrant 2018b). With regard to respiratory management, no pharmacological intervention is recommended in the guidelines other than corticosteroid use.  Nevertheless, compliance with guidelines has been shown to be poor in Italy, the UK, the USA, and Germany, with fewer than 30% of patients meeting all absolute recommendations (Landfeldt 2015). This highlights the low number of therapeutic options available for DMD and the need to develop and evaluate new approaches and therapies with an adequate body of evidence to support their use.

Current access to newer DMD drugs is limited and generally restricted to participation in clinical trials. All the available evidence for idebenone in the treatment of decline in respiratory function in DMD comes from small trials and studies with multiple reports. When considering new therapies such as idebenone, a major barrier to uptake following regulatory approval is likely to be the associated cost. Furthermore, it is possible that some of the available medications could be used in combination, thereby increasing the cost much further (Shawi 2017). Idebenone is orally administered and requires multiple daily doses. This may present additional challenges for people with DMD who have swallowing disorders (Shawi 2017).

Quality of the evidence

The overall certainty of the evidence was low, but all the evidence comes from one relatively small study with few events, which leads to serious imprecision.  Another, larger study was discontinued due to futility with only a press release available and no numerical data. We judged all outcomes reported in the study as at low risk of bias. We also downgraded the certainty of the evidence for the outcomes of the SIDEROS 2021 trial for publication bias, as there were no results beyond statements from the pharmaceutical company in a press release.

Potential biases in the review process

We modified the outcome 'any adverse events' to maximise the use of the available data, reporting it as serious and non‐serious adverse events. We extracted and analysed data regarding 'change in muscle function' as two separate muscle groups (elbow flexors and elbow extensors). We took precautions to avoid bias in this process by documenting all changes in the Differences between protocol and review section of the review.

We encountered difficulties in determining the 'antioxidant effect' of several interventions reported in trials retrieved by our search strategy, as some interventions being studied in this population are hypothesised to have concomitant antioxidant properties. Nevertheless, when considering all of our predefined possible antioxidant agent interventions, we were able to screen studies with no major problems. 

We found studies reporting quality of life, adverse events, or muscle function in people with DMD; unfortunately, however, several of these studies did not consider or did not report respiratory function of participants at baseline (Bäckman 1988Bertorini 1985Bonati 2014Fenichel 1988Mendell 1979Nagy 2019NCT00308113Stern 1982Tamari 1982). Consequently, we were not able to identify whether the participants in these studies actually had respiratory decline as defined in our protocol, therefore we finally excluded all of them. 

Agreements and disagreements with other studies or reviews

Narrative reviews have addressed respiratory function in DMD, considering the impact of pharmacological interventions to delay its decline (Gogou 2020Werneck 2019). Although these reviews stated that every treatment strategy should prioritise the preservation of lung function, they concluded that corticosteroid therapy remains the best‐studied pharmacologic therapy for DMD and very likely delays the expected decline in lung function. A Cochrane Review of corticosteroids for DMD concluded that corticosteroid therapy in DMD improves muscle strength and function in the short term as well as up to two years, but did not consider respiratory function outcomes (Matthews 2016). 

The American Thoracic Society (ATS) Consensus Statement for respiratory care of DMD patients is based mostly on airway clearance, respiratory muscle training, or mechanical ventilation support (Finder 2004). The last update of this statement was published before the trial included in this review was published (Finder 2009). Regarding mechanical ventilation, a Cochrane Review concluded that there is a survival benefit of mechanical ventilation against no ventilation in DMD, but did not consider respiratory function outcomes in this population (Annane 2014). 

Along with respiratory decline, cardiomyopathy is a significant limiting factor for long‐term survival in DMD. As physical disability progresses, it is rarely possible to differentiate complications in the context of a chest infection with associated respiratory failure from complications of a primary cardiac cause.  Regarding the latter, a Cochrane Review concluded that early treatment with angiotensin‐converting enzyme inhibitors or angiotensin receptor blockers may be comparably beneficial for people with dystrophinopathy for the prevention and treatment of cardiac complications in people with DMD or Becker muscular dystrophy (Bourke 2018). 

Although the effects of idebenone on PEF in DMD patients with cardiac dysfunction appeared promising (DELPHI 2011), they have not been replicated in a different population of DMD participants with signs of respiratory decline, with or without concomitant corticosteroid treatment (DELOS 2015SIDEROS 2021).  Furthermore, the effect of any antioxidant in patients with spinal scoliosis, a common complication in DMD that affects respiratory function, remains unknown. Nor have the positive effects of idebenone on PEF been translated into benefit in different major outcomes like FVC, despite the fact that the effects of idebenone have been studied in former participants of the study included in this review (DELOS 2015), showing a consistent and sustained reduction in the rate of respiratory function decline for up to six years, although no procedure ensured the consistency of assessment methods across the research sites or the systematic collection of all concomitant medication use, which could also have influenced the observed effects (Buyse 2019SYROS 2019). The discontinuation of SIDEROS 2021 and its expanded access programmes, EUCTR2019‐004602‐94‐BENCT03603288, due to futility may indicate that research on this drug in DMD is no longer needed.

Antioxidant agents other than idebenone have been tested in people with DMD with regard to their effect on motor function of different muscle groups or serum levels of several biomarkers, with promising results (Bonati 2014Kawamura 2020), but respiratory function was neither considered as an inclusion criterion nor an outcome of interest in these studies.  

Authors' conclusions

Implications for practice.

Idebenone is the only antioxidant agent tested in randomised controlled trials for the prevention of respiratory decline in individuals with Duchenne muscular dystrophy (DMD). The limited available evidence is from a single published study. A larger study was stopped in October 2020 for futility, and at the time of writing is unpublished. Idebenone may result in slightly less of a decline from baseline in forced vital capacity (FVC) and probably results in little to no difference in change in quality of life. Idebenone may result in little to no difference in change in muscle function and less of a decline from baseline in forced expiratory volume in 1 second (FEV1) and peak expiratory flow (PEF).  

Implications for research.

The discontinuation of the SIDEROS trial and its expanded access programmes due to futility may indicate that research for this drug in this condition is no longer needed. Further research is needed to establish the effect of different antioxidant agents on preventing respiratory decline in DMD patients during the respiratory decline phase of the condition. Other antioxidant agents different from idebenone need to be considered and tested. Randomised controlled trials designed for this purpose need to include a greater number of participants and be powered to detect the effects of the intervention on relevant respiratory outcomes like FVC, total lung capacity, or the need for ventilatory support, both in the short and long term. These outcomes must be included as primary outcomes, alongside other outcomes relevant to patients, families, and relevant stakeholders, such as hospitalisations, quality of life, functional independence, or cost associated with treatment. Furthermore, comorbidities in patients with DMD, such as spinal scoliosis, amongst others, need to be considered as potential influencing factors in the treatment effects.

History

Protocol first published: Issue 9, 2020

Risk of bias

Risk of bias for analysis 1.1 Forced vital capacity (FVC).

Study Bias
Randomisation process Deviations from intended interventions Missing outcome data Measurement of the outcome Selection of the reported results Overall
Authors' judgement Support for judgement Authors' judgement Support for judgement Authors' judgement Support for judgement Authors' judgement Support for judgement Authors' judgement Support for judgement Authors' judgement Support for judgement
DELOS 2015 Low risk of bias A random component was used in the sequence generation process and a central interactive web system for allocation. No serious imbalance in baseline characteristics was found. While allocation concealment might have been overridden in a pair of siblings we don't consider it to be a serious concern regarding bias. Low risk of bias Participants and those involved in caring for participants were not aware of the assigned intervention. There are no concerns regarding the analysis used to estimate the effect of assignment to intervention.   Low risk of bias Data were available for nearly all randomized participants.  Low risk of bias Method for outcome measurement was appropriate and equally applied between groups. Outcome assessors were blinded to intervention status.  Low risk of bias Data were analysed in accordance with the pre‐specified plan and assessed based on pre‐specified domains and time‐points.  Low risk of bias The study is judged to be at low risk of bias for all domains for this result.

Risk of bias for analysis 1.2 Quality of life.

Study Bias
Randomisation process Deviations from intended interventions Missing outcome data Measurement of the outcome Selection of the reported results Overall
Authors' judgement Support for judgement Authors' judgement Support for judgement Authors' judgement Support for judgement Authors' judgement Support for judgement Authors' judgement Support for judgement Authors' judgement Support for judgement
DELOS 2015 Low risk of bias A random component was used in the sequence generation process and a central interactive web system for allocation. No serious imbalance in baseline characteristics was found. While allocation concealment might have been overridden in a pair of siblings we don't consider it to be a serious concern regarding bias. Low risk of bias Participants and those involved in caring for participants were not aware of the assigned intervention. There are no concerns regarding the analysis used to estimate the effect of assignment to intervention.   Low risk of bias Data were available for nearly all randomized participants. Low risk of bias Method for outcome measurement was appropriate and equally applied between groups. Outcome assessors were blinded to intervention status.  Low risk of bias Data were analysed in accordance with the pre‐specified plan and assessed based on pre‐specified domains and time‐points.  Low risk of bias The study is judged to be at low risk of bias for all domains for this result.

Risk of bias for analysis 1.3 Any adverse events.

Study Bias
Randomisation process Deviations from intended interventions Missing outcome data Measurement of the outcome Selection of the reported results Overall
Authors' judgement Support for judgement Authors' judgement Support for judgement Authors' judgement Support for judgement Authors' judgement Support for judgement Authors' judgement Support for judgement Authors' judgement Support for judgement
Subgroup 1.3.1 Serious adverse events
DELOS 2015 Low risk of bias A random component was used in the sequence generation process and a central interactive web system for allocation. No serious imbalance in baseline characteristics was found. While allocation concealment might have been overridden in a pair of siblings we don't consider it to be a serious concern regarding bias. Low risk of bias Participants and those involved in caring for participants were not aware of the assigned intervention. There are no concerns regarding the analysis used to estimate the effect of assignment to intervention.   Low risk of bias Data were available for nearly all randomized participants.  Low risk of bias Method for outcome measurement was appropriate and equally applied between groups. Outcome assessors were blinded to intervention status.  Low risk of bias Data were analysed in accordance with the pre‐specified plan and assessed based on pre‐specified domains and time‐points Low risk of bias The study is judged to be at low risk of bias for all domains for this result.
Subgroup 1.3.2 Nonserious adverse events
DELOS 2015 Low risk of bias A random component was used in the sequence generation process and a central interactive web system for allocation. No serious imbalance in baseline characteristics was found. While allocation concealment might have been overridden in a pair of siblings we don't consider it to be a serious concern regarding bias. Low risk of bias Participants and those involved in caring for participants were not aware of the assigned intervention. There are no concerns regarding the analysis used to estimate the effect of assignment to intervention.   Low risk of bias Data were available for nearly all randomized participants.  Low risk of bias Method for outcome measurement was appropriate and equally applied between groups. Outcome assessors were blinded to intervention status.  Low risk of bias Data were analysed in accordance with the pre‐specified plan and assessed based on pre‐specified domains and time‐points Low risk of bias The study is judged to be at low risk of bias for all domains for this result.

Acknowledgements

The Information Specialist of Cochrane Neuromuscular, Angela Gunn, reviewed the search strategy in consultation with the review authors.

The Methods section of this review is based on a template developed by Cochrane Neuromuscular from an original created by Cochrane Airways.

This project was supported by the National Institute for Health Research (NIHR) via Cochrane Infrastructure funding to Cochrane Neuromuscular. The views and opinions expressed therein are those of the authors and do not necessarily reflect those of the Systematic Reviews Programme, NIHR, NHS, or the Department of Health. Cochrane Neuromuscular is also supported by the MRC Centre for Neuromuscular Disease.

We thank Dr Rosaline Quinlivan for her contribution to the Background section of the protocol and full review. 

We thank the members of the Editorial Base (Ruth Brassington, Michael Lunn, Sarah Nevitt, and Farhad Shokraneh) and peer‐reviewer (Tracey Willis, Muscle team, Robert Jones and Agnes Hunt Hospital, Oswestry UK and Chester University Medical School) for their comments and feedback during the review process. 

Appendices

Appendix 1. Appendix 1. Cochrane Central Register of Controlled Trials (CENTRAL) search strategy

#1 MeSH descriptor: [Muscular Dystrophy, Duchenne] explode all trees

#2 (duchenne*):ti,ab,kw

#3 (DMD):ti,ab,kw

#4 #1 OR #2 OR #3

#5 MeSH descriptor: [Antioxidants] explode all trees

#6 MeSH descriptor: [Acetylcysteine] explode all trees

#7 MeSH descriptor: [Metformin] explode all trees

#8 MeSH descriptor: [Citrulline] explode all trees

#9 MeSH descriptor: [Tea] explode all trees

#10 (N‐acetylcysteine lysinate):ti,ab,kw

#11 (raxone):ti,ab,kw

#12 (Antioxidant*):ti,ab,kw OR  (Anti‐oxidant*):ti,ab,kw

#13 (Tea):ti,ab,kw

#14 ("Coenzyme Q10"):ti,ab,kw

#15 (idebenone):ti,ab,kw

#16 ("CoQ 10"):ti,ab,kw

#17 (metformin*):ti,ab,kw

#18 (citrulline):ti,ab,kw

#19 #5 OR #6 OR #7 OR #8 OR #9 OR #10 OR #11 OR #12 OR #13 OR #14 OR #15 OR #16 OR #17 OR #18

#20 #4 AND #19

Appendix 2. Appendix 2. MEDLINE (PubMed) search strategy

#1 "Muscular Dystrophy, Duchenne"[Mesh]

#2 duchenne*[Tiab]

#3 DMD[Tiab]

#4 #1 OR #2 OR #3

#5 "Antioxidants"[Mesh]

#6 "Acetylcysteine"[Mesh]

#7 "Metformin"[Mesh]

#8 "Citrulline"[Mesh]

#9 "Tea"[Mesh]

#10 "Antioxidants"[Pharmacological Action]

#11"N‐acetylcysteine lysinate"[Supplementary Concept]

#12 raxone[Tiab]

#13 Antioxidant*[Tiab] OR Anti‐oxidant*[Tiab]

#14 Tea[Tiab]

#15 "Coenzyme Q10"[Tiab]

#16 idebenone[Tiab]

#17 "CoQ 10"[Tiab]

#18 metformin*[Tiab]

#19 Citrulline[Tiab]

#20  #5 OR #6 OR #7 OR #8 OR #9 OR #10 OR #11 OR #12 OR #13 OR #14 OR #15 OR #16 OR #17 OR #18 OR #19

#21 #4 AND #20

Appendix 3. Appendix 3. Embase (Elsevier) search strategy

#1.  'duchenne muscular dystrophy'/exp       

#2.  duchenne*:ti,ab       

#3.  dmd:ti,ab   

#4.  #1 OR #2 OR #3     

#5.  'antioxidant'/exp   

#6.  'acetylcysteine'/exp 

#7.  'metformin'/exp                                         

#8.  'citrulline'/exp 

#9.  'herbal tea'/exp       

#10. 'antioxidants pharmacology'/exp                            

#11. 'lysine acetylsalicylate'/exp       

#12. 'idebenone'/exp                                         

#13. raxone:ti,ab                                             

#14. antioxidant*:ti,ab OR anti‐oxidant*:ti,ab

#15. tea:ti,ab 

#16. 'ubidecarenone'/exp 

#17. 'coenzyme q10':ti,ab 

#18. idebenone:ti,ab 

#19. 'coq 10':ti,ab                                          

#20. metformin*:ti,ab                                        

#21. citrulline:ti,ab

#22. #5 OR #6 OR #7 OR #8 OR #9 OR #10 OR #11 OR #12  OR #13 OR #14 OR #15 OR #16 OR #17 OR #18 OR #19 OR #20 OR #21                                                                                                                                              

#23. #4 AND #22

Appendix 4. Appendix 4. Clinical trials registries search strategies

US National Institutes for Health Clinical Trials Registry, ClinicalTrials.gov search strategy (Expert Search)

(Muscular Dystrophy, Duchenne OR Duchenne OR  DMD) [DISEASE] AND (Antioxidants OR Antioxidant OR Anti‐oxidant OR Anti‐oxidants OR Acetylcysteine OR Metformin OR Citrulline OR Tea OR "N‐acetylcysteine lysinate" OR raxone OR "Coenzyme Q10" OR idebenone OR "CoQ 10") [TREATMENT]

WHO International Clinical Trials Registry Portal (ICTRP) search strategy

Advanced search

Condition: Muscular Dystrophy, Duchenne OR Duchenne OR DMD

Intervention: Antioxidants OR Antioxidant OR Anti‐oxidant OR Anti‐oxidants OR Acetylcysteine OR Metformin OR Citrulline OR Tea OR "N‐acetylcysteine lysinate" OR raxone OR "Coenzyme Q10" OR idebenone OR "CoQ 10"

Recruitment status is ALL

Data and analyses

Comparison 1. Idebenone versus placebo.

Outcome or subgroup title No. of studies No. of participants Statistical method Effect size
1.1 Forced vital capacity (FVC) 1   Mean Difference (IV, Random, 95% CI) Totals not selected
1.2 Quality of life 1   Mean Difference (IV, Random, 95% CI) Totals not selected
1.3 Any adverse events 1   Risk Ratio (M‐H, Random, 95% CI) Totals not selected
1.3.1 Serious adverse events 1   Risk Ratio (M‐H, Random, 95% CI) Totals not selected
1.3.2 Nonserious adverse events 1   Risk Ratio (M‐H, Random, 95% CI) Totals not selected
1.4 Change in muscle function (elbow flexors) 1   Mean Difference (IV, Random, 95% CI) Totals not selected
1.5 Change in muscle function (elbow extensors) 1   Mean Difference (IV, Random, 95% CI) Totals not selected
1.6 Forced expiratory volume in the first second (FEV1) 1   Mean Difference (IV, Random, 95% CI) Totals not selected
1.7 Peak expiratory flow (PEF) 1   Mean Difference (IV, Random, 95% CI) Totals not selected

Characteristics of studies

Characteristics of included studies [ordered by study ID]

DELOS 2015.

Study characteristics
Methods Study design: prospective, randomised study.
Study dates: 27July 2009 to 14 January 2014.
Setting: outpatient, multicentre, international.
Country: Belgium, Germany, the Netherlands, Switzerland, France, Sweden, Austria, Italy, Spain, USA.
Participants Inclusion criteria:
  1. Patients 10 to 18 years of age at baseline.

  2. Signed and dated informed consent.

  3. Documented diagnosis of DMD or severe dystrophinopathy

  4. Clinical features consistent with typical DMD at diagnosis (i.e. documented delayed motor skills and muscle weakness by age 5 years). DMD should be confirmed by mutation analysis in the dystrophin gene or by substantially reduced levels of dystrophin protein (i.e. absent or < 5% of normal) on Western blot or immunostain.

  5. Ability to provide reliable and reproducible repeat PEF within 15% of the first assessment (i.e. baseline versus screening).

  6. Patients assessed by the investigator as willing and able to comply with the requirements of the study, possessing the required cognitive abilities, and able to swallow study medication.


Exclusion criteria:
  1. Dependence on assisted ventilation at screening or baseline (defined as NIV, daytime NIV, or continuous invasive ventilation).

  2. Documented DMD‐related hypoventilation for which assisted ventilation is needed according to current standard‐of‐care guidelines (e.g. FVC < 30%) or in the opinion of the investigator.

  3. Patients with a per cent predicted PEF > 80% at baseline.

  4. Patients unable to form a mouth seal to allow precise respiratory flow measurements and mouth pressures.

  5. Symptomatic heart failure (high probability of death within 1 year of baseline) or symptomatic ventricular arrhythmias, or both.

  6. Participation in the previous phase II or phase II extension study (SNT‐II‐001 or SNT‐II‐001‐E) for idebenone.

  7. Participation in any other therapeutic trial or intake of any investigational drug within 90 days prior to baseline.

  8. Use of carnitine, creatine, glutamine, oxatomide, or any herbal medicines within 30 days prior to baseline.

  9. Use of coenzyme Q10 or vitamin E (if taken at a dose of 5 times above the daily physiological requirement) within 30 days prior to baseline.

  10. Any previous use of idebenone.

  11. Any concomitant medication with a depressive or stimulating effect on respiration or the respiratory tract.

  12. Planned or expected spinal fixation surgery during the study period (as judged by the investigator).

  13. Asthma, bronchitis or COPD, bronchiectasis, emphysema, pneumonia, or the presence of any other non‐DMD respiratory illness that affects PEF.

  14. Chronic use of beta‐2 agonists or use of any other bronchodilating medication (e.g. inhaled steroids, sympathomimetics, anticholinergics) (chronic use is defined as daily intake for more than 14 days).

  15. Moderate or severe hepatic impairment or severe renal impairment.

  16. Prior or ongoing medical condition or laboratory abnormality that in the investigator's opinion could adversely affect the safety of the participant.

  17. Patients suffering from a severe, unstable condition including (but not limited to) cancer, autoimmune diseases, haematological diseases, metabolic disorders, or immunodeficiencies, and who are at risk of an aggravation unrelated to the study condition, could only be included in the study if accepted in writing by the sponsor's medical monitor.

  18. Relevant history of or current drug or alcohol abuse, use of any tobacco or marijuana products, or smoking.

  19. Known individual hypersensitivity to idebenone or to any of the ingredients or excipients.

  20. Systemic glucocorticoid therapy: chronic use of systemic glucocorticoid therapy for DMD‐related conditions within 12 months of baseline (the "12 month non‐use period"); more than 2 rounds of acute systemic glucocorticoid burst therapy (of ≤ 2 weeks' duration) for non‐DMD related conditions within the 12‐month non‐use period; use of any round of systemic glucocorticoid burst therapy of longer than 2 weeks' duration within the 12‐month non‐use period; use of systemic glucocorticoid burst therapy less than 8 weeks prior to baseline.


Total participants randomised: 66
Group 1: n = 32 idebenone group
Age, mean (SD): 13.5 (2.7)
Weight, mean (SD): 55.3 (18.3) kg
Height, mean (SD): 157.4 (11.3) cm
Body mass index, mean (SD): 22.0 (5.9) kg/m²
Ethnic origin, n (%):
  • White: 29 (94%)

  • Asian: 1 (3%)

  • Hispanic: 0

  • Other: 1 (3%)


Previous glucocorticoid use, n(%): 17 (55%)
Time since last glucocorticoid use, mean (SD): 2.9 (1.8) years
Patient in wheelchair, n(%): 28 (90%)
Baseline PEF as % predicted (PEF%p), n(%):
  • < 40%p: 5 (16%)

  • 40% to 80%p: 26 (84%)


Baseline respiratory function tests:
  • PEF%p, mean (SD): 53.5 (10.3)

  • PEF (L/min), mean (SD): 217.7 (48.6)

  • FVC%p, mean (SD): 55.3 (15.8)

  • FVC (L), mean (SD): 1.9 (0.5)

  • FEV1%p, mean (SD): 53.3 (15.1)

  • FEV1 (L), mean (SD): 1.54 (0.33)

  • MIP%p, mean (SD): 43.5 (22.2)

  • MIP (cm H2O), mean (SD): 47.3 (24.4)

  • MEP%p, mean (SD): 28.3 (12.2)

  • MEP (cm H2O), mean (SD): 40.6 (15.6)

  • PCF (L/min), mean (SD): 243.0 (70.7)


Group 2: n = 34 placebo group
Age, mean (SD): 15.0 (2.5)
Weight, mean (SD): 61.9 (18) kg
Height, mean (SD): 162.4 (12.4) cm
Body mass index, mean (SD): 23.4 (5.6) kg/m²
Ethnic origin, n (%): 
  • White: 31 (94%)

  • Asian: 0

  • Hispanic: 1 (3%)

  • Other: 1 (3%)


Previous glucocorticoid use, n(%): 19 (58%)
Time since last glucocorticoid use, mean (SD): 4.3 (2.2) years
Patient in wheelchair, n(%): 31 (94%)
Baseline peak expiratory flow as % predicted (PEF%p), n(%): 
  • < 40%p: 7 (21%)

  • 40% to 80%p: 26 (79%)


Baseline respiratory function tests:
  • PEF%p, mean (SD): 54.2 (13.2)

  • PEF (L/min), mean (SD): 233.8 (59.6)

  • FVC%p, mean (SD): 50.4 (20.0)

  • FVC (L), mean (SD): 1.9 (0.5)

  • FEV1%p, mean (SD): 49.7 (18.3)

  • FEV1 (L), mean (SD): 1.71 (0.57)

  • MIP%p, mean (SD): 38.5 (16.9)

  • MIP (cm H2O), mean (SD): 44.6 (16.9)

  • MEP%p, mean (SD): 25.1 (12.2)

  • MEP (cm H2O), mean (SD): 39.7 (16.6)

  • PCF (L/min), mean (SD): 256.4 (50.5)

Interventions Group 1: n = 32 idebenone group:
Treatment consisted of film‐coated tablets of idebenone (Raxone or Catena, Santhera Pharmaceuticals, Liestal, Switzerland) (900 mg/day): 2 tablets (150 mg each) 3 times orally with meals for 52 weeks.
Group 2: n = 34 placebo group:
Treatment consisted of placebo (900 mg/day) 2 tablets (150 mg each) 3 times orally with meals for 52 weeks.
Co‐interventions: not reported
Outcomes Forced vital capacity
How measured: measured by a qualified, trained, and certified evaluator at each centre in accordance with standardised procedures and international guidelines, using a Pneumotrac Spirometer 6800 (Vitalograph), and reported as change from baseline in FVC%p and as change from baseline in absolute values (FVC (L))
Time points measured: baseline, 13, 26, 39, and 52 weeks
Time points reported: baseline and 52 weeks
Subgroups: none
 
Quality of life
How measured: measured using the Pediatric Quality of Life Inventory (PedsQL), which contains the following paediatric health‐related quality of life measurements: Physical, Emotional, Social and School Functioning.
Item scaling in a 5‐point Likert scale from 0 (never) to 4 (almost always). 3‐point scale: 0 (not at all), 2 (sometimes), and 4 (a lot) for the young child (ages 5 to 7). Scores were transformed on a scale from 0 to 100 (0 = 100, 1 = 75, 2 = 50, 3 = 25, 4 = 0). Total score: sum of all the items over the number of items answered on all the scales. The overall scores ranged between 0 and 100, with 0 = worst outcome and 100 = best outcome. Reported as change from baseline.
Time points measured: baseline and 52 weeks
Time points reported: baseline and 52 weeks
Subgroups: none
 
Adverse events
How measured: number of participants reporting serious adverse events and non‐serious adverse events
Time points measured: not reported
Time points reported: 52 weeks
Subgroups: none
 
Change in muscle function
How measured: the change from baseline to week 52 in muscle strength as measured by hand‐held myometry (HHM) was performed following standardised procedures. As almost all participants were non‐ambulatory, only analyses of upper limb muscle strength were performed.  The highest value of 3 consecutive measurements with an interval of at least 10 s were recorded.
The HHM was measured using MicroFET2, a digital hand‐held muscle tester. The selected unit of measure was Newtons (N).
Time points measured: not reported
Time points reported: 52 weeks
Subgroups: none
 
Peak expiratory flow
How measured: measured by a qualified, trained, and certified evaluator at each centre in accordance with standardised procedures and international guidelines, using participant's portable ASMA‐1 device (usb model 4000, Vitalograph, Maids Moreton, UK) and reported as change from baseline in PEF%p and as change from baseline in absolute values (PEF L/min).
Time points measured: baseline and 52 weeks
Time points reported: baseline, 13, 26, 39, and 52 weeks
Subgroups: none
 
 
Forced expiratory volume
How measured: measured by a qualified, trained, and certified evaluator at each centre in accordance with standardised procedures and international guidelines, using participant's portable ASMA‐1 device (usb model 4000, Vitalograph, Maids Moreton, UK) and reported as change from baseline in FEV1%p and as change from baseline in absolute values (FEV1 (L)).
Time points measured: baseline, 13, 26, 39, and 52 weeks
Time points reported: baseline and 52 weeks
Subgroups: none
Notes The study was sponsored by Santhera Pharmaceuticals. 
Height was derived from ulnar length.
Change in absolute values of FVC and FEV1 available in online supplementary appendix.
Respiratory tract infection‐related adverse events available in online supplementary appendix.
Quality of life outcomes available from ClinicalTrials.gov.
Change in muscle function available from ClinicalTrials.gov. 
Declarations of interest: GMB was investigator for clinical trials in DMD sponsored by Santhera Pharmaceuticals, Prosensa Therapeutics, and GlaxoSmithKline. TV was an investigator for clinical trials of DMD sponsored by PTC Therapeutics, GlaxoSmithKline, Prosensa, and Santhera Pharmaceuticals; he serves as a scientific advisory board member to Prosensa. US was an investigator for clinical trials sponsored by PTC Therapeutics, Lilly Pharma, Santhera Pharmaceuticals, Prosensa, and GlaxoSmithKline. CSMS has participated in trials sponsored by GlaxoSmithKline, Prosensa, and Santhera Pharmaceuticals. RSF has participated in studies of DMD sponsored by PTC Therapeutics, the US National Institutes of Health (UDP R01NS043264, Wellstone 5U54AR052646‐03, Imaging DMD R01‐AR056973, and FOR‐DMD U01 NS061799‐01A2), Lilly, Muscular Dystrophy Association, Sarepta; served as a member of the data and safety monitoring board for the Sarepta 201 study; and served as adviser to Catabasis. CMM has served as a consultant for trials unrelated to this scope of work for PTC Therapeutics, Prosensa, Sarepta, Eli Lilly, Pfizer, Halo Therapeutics, Cardero, and Mitokyne, and serves on external advisory boards related to DMD for PTC Therapeutics and Eli Lilly. TM is a regular employee of Santhera Pharmaceuticals. GMB and TM are co‐inventors of relevant patent applications. The other authors declare no competing interests.

SIDEROS 2021.

Study characteristics
Methods A phase III double‐blind, randomised, placebo‐controlled study
Participants Patients with DMD receiving glucocorticoid steroids
Inclusion criteria:
  1. Male patients with an FVC ≥ 35% and ≤ 80% of predicted value at screening and at baseline who, in the opinion of the investigator, are in the respiratory function decline phase.

  2. Minimum 10 years of age at screening.

  3. Signed and dated informed consent form.

  4. Documented diagnosis of DMD (severe dystrophinopathy) and clinical features consistent with typical DMD at diagnosis (i.e. documented delayed motor skills and muscle weakness by age 5 years). DMD should be confirmed by mutation analysis in the dystrophin gene or by substantially reduced levels of dystrophin protein (i.e. absent or < 5% of normal) on Western blot or immunostaining.

  5. Chronic use of systemic glucocorticoid steroids for DMD‐related conditions continuously for at least 12 months prior to baseline without any dose adjustments on a mg/kg basis in the last 6 months (only dose adjustment determined by weight changes allowed).

  6. Ability to provide reliable FVC values at screening and baseline, and reproducible within 15% (relative change) at baseline compared to screening.

  7. Patients were assessed by the investigator as willing and able to comply with the requirements of the study, possessing the required cognitive abilities, and able to swallow study medication.

  8. Patients who prior to screening were immunised with 23‐valent pneumococcal polysaccharide vaccine or any other pneumococcal polysaccharide vaccine as per national recommendations, as well as annually immunised with inactivated influenza vaccine.


Exclusion criteria:
  1. Symptomatic heart failure (defined as structural heart disease, dyspnoea, fatigue, and impaired tolerance to exercise; stage C by the American College of Cardiology Foundation/American Heart Association guideline or New York Heart Association classes III‐IV) or symptomatic ventricular arrhythmias, or both.

  2. Ongoing participation in any other therapeutic trial or intake of any investigational drug within 90 days prior to baseline (the only exception is the use of deflazacort in the USA as part of the Expanded Access Program or any corticosteroid product in trial for regimen optimisation, for which the patient met the inclusion criterion 5).

  3. Ongoing exon‐skipping therapy or read‐through gene therapy for DMD; previous exon‐skipping or read‐through gene therapy allowed if the stop date was more than 6 months prior to screening.

  4. Planned or expected spinal fixation surgery during the study period (as judged by the investigator, i.e. due to rapidly progressing scoliosis); prior spinal fixation surgery allowed if it took place more than 6 months prior to screening.

  5. Asthma, bronchitis or COPD, bronchiectasis, emphysema, pneumonia, or the presence of any other non‐DMD respiratory illness that affects respiratory function.

  6. Chronic use of beta2‐agonists or any use of other bronchodilating or bronchoconstricting medication (inhaled steroids, sympathomimetics, anticholinergics, antihistamines); chronic use is defined as daily intake for more than 14 days.

  7. Any bronchopulmonary illness requiring treatment with antibiotics within 3 months prior to screening.

  8. Moderate or severe hepatic impairment as assessed and documented by the investigator (liver function tests, medical history, or, when the parameters of the formula are available to cite (use as guidance Child‐Pugh class B (7 to 9 points) or Child‐Pugh class C (10 to 15 points)) could be indicative of such conditions), or severe renal impairment (estimated glomerular filtration rate < 30 mL/min/1.73 m²).

  9. Prior or ongoing medical condition or laboratory abnormality, which in the investigator’s opinion may put the patient at significant risk, may confound the study results, or may interfere significantly with the patient’s participation in the study.

  10. History of or current drug or alcohol abuse or use of any tobacco or marijuana products or smoking.

  11. Known individual hypersensitivity to idebenone or to any of the ingredients or excipients of the study medication.

  12. Daytime ventilator assistance (defined as the use of any assisted ventilation whilst awake).

Interventions Idebenone 150 mg film‐coated tablet versus placebo
Outcomes Primary endpoint
  • Change from baseline to week 78 (or slope of changes over 78 weeks) in FVC%p assessed by clinic‐based spirometry measurements

  • Changes in PEF%p using clinic‐based spirometry

  • Time to loss of 10% of baseline FVC using clinic‐based spirometry


To assess the efficacy of idebenone compared to placebo in delaying the loss of inspiratory muscle function as measured by changes in inspiratory flow reserve (IFR) using clinic‐based spirometry.
To assess the time to clinically relevant events and disease milestones.
Secondary endpoints
The secondary endpoints will be evaluated in the following order in a hierarchical manner.
  1. Rate of bronchopulmonary adverse events.

  2. The time to first 10% decline in FVC%p during the 78‐week treatment period, assessed by clinic‐based spirometry measurements.

  3. Change from baseline to week 78 (or slope of changes over 78 weeks) in PEF%p assessed by clinic‐based spirometry measurements.

  4. Rate of use of antibiotics.

  5. Proportion of participants with hospitalisations due to respiratory causes.

  6. Change from baseline to week 78 (or slope of changes over 78 weeks) in IFR assessed by clinic‐based spirometry measurements.

Notes Source of monetary support: Santhera Pharmaceuticals (Switzerland) Limited.
A press release was shared by the pharmaceutical company explaining that the trial was prematurely terminated on 2 October 2020.

COPD: chronic obstructive pulmonary disease
DMD: Duchenne muscular dystrophy
FEV1: forced expiratory volume in 1 s
FEV1%p: forced expiratory volume in 1 s as percentage predicted
FVC: forced vital capacity
FVC%p: forced vital capacity as percentage predicted
MEP: maximum expiratory pressure
MEP%p: maximum expiratory pressure as percentage predicted
MIP: maximum inspiratory pressure
MIP%p: maximum inspiratory pressure as percentage predicted
NIV: non‐invasive ventilation
PCF: peak cough flow
PEF: peak expiratory flow
PEF%p: peak expiratory flow as percentage predicted
SD: standard deviation

Characteristics of excluded studies [ordered by study ID]

Study Reason for exclusion
Bäckman 1988 Wrong patient population. The study did not report respiratory function of participants at baseline, and we were unable to identify if included participants had respiratory decline.
Barcellona 2014 Wrong study design. Observational pilot study evaluating the safety and tolerability of flavocoxid in ambulant DMD patients
Bertorini 1985 Wrong patient population. The study did not report respiratory function of participants at baseline, and we were unable to identify if included participants had respiratory decline.
Bonati 2014 Wrong patient population. The study did not report respiratory function of participants at baseline, and we were unable to identify if included participants had respiratory decline.
Buyse 2008 Wrong patient population. The study did not report respiratory function of participants at baseline, and we were unable to identify if included participants had respiratory decline.
Buyse 2013 Wrong patient population. The study did not report respiratory function of participants at baseline, and we were unable to identify if included participants had respiratory decline.
Buyse 2015a Wrong study design. Combination of studies using idebenone
Buyse 2015b Wrong study design. Combination of studies using idebenone
Buyse 2019 Wrong study design. Combination of the extension programmes of the DELPHI and DELOS studies
DELPHI 2011 Wrong patient population. The majority of participants did not have respiratory decline. A meta‐analysis was presented by the study authors of the DELPHI study using raw data from the trial as a poster in a conference on the subset of participants with respiratory decline, but no numerical data were available (Leinonen 2017).
Escolar 2012 Wrong patient population. Participants did not have respiratory decline at baseline as considered for this review.
EUCTR2005‐002520‐33‐BE Wrong patient population with no outcomes reported. There is no statement related to participant respiratory decline, considered in this review as an inclusion criterion.
EUCTR2017‐002213‐60‐GB Wrong intervention. No antioxidant effect associated with the study drug
EUCTR2017‐004279‐30‐GB Wrong study design. Observational extension study of the active arm of the SIDEROS trial
EUCTR2019‐004602‐94‐BE Wrong intervention. No antioxidant effect associated with the study drug
Fenichel 1988 Wrong patient population. The study did not report respiratory function of participants at baseline, and we were unable to identify if included participants had respiratory decline.
Hunter 1983 Wrong study design. Cross‐over randomised controlled trial
Mendell 1979 Wrong patient population. The study did not report respiratory function of participants at baseline, and we were unable to identify if included participants had respiratory decline.
Nagy 2019 Wrong patient population. The study did not report respiratory function of participants at baseline, and we were unable to identify if included participants had respiratory decline.
NCT00308113 Wrong patient population with no outcomes reported. There is no statement related to participant respiratory decline, considered in this review as an inclusion criterion.
NCT03603288 Wrong study design. Observational extension study of the active arm of the SIDEROS trial
Stern 1981 Wrong patient population. The study did not report respiratory function of participants at baseline, and we were unable to identify if included participants had respiratory decline.
Stern 1982 Wrong patient population. The study did not report respiratory function of participants at baseline, and we were unable to identify if included participants had respiratory decline.
SYROS 2019 Wrong study design. Observational study based on the expanded access programme of the drug idebenone following the DELOS study
Tamari 1982 Wrong patient population. The study did not report respiratory function of participants at baseline, and we were unable to identify if included participants had respiratory decline.

DMD: Duchenne muscular dystrophy

Characteristics of ongoing studies [ordered by study ID]

SUNIMUD.

Study name Sunphenon EGCg (epigallocatechin‐gallate) in Duchenne muscular dystrophy
Methods Randomised controlled parallel trial 
Participants Inclusion criteria:
  • DMD

  • Male patients > 4 years

  • Ability to walk without help (a minimum of 75 m)

  • Maximum 2 cups of black tea, no consumption of green tea, no consumption of large amounts of grapefruit juice

  • Informed consent of parents


Exclusion criteria:
  • Serious other organic diseases

  • Known intolerance of Sunphenon

  • Massive and protracted exposure to the sun

  • Participation in other interventional clinical trials on pharmaceuticals or medical devices during the study or 3 months prior to study

  • Further primary psychiatric or neurologic disorders

  • Known allergy to Sunphenon EGCg or additives of the study medication or placebo capsules

  • Long‐term intake of liver toxic medication

Interventions Sunphenon EGCg (epigallocatechin‐gallate) 50 mg hard capsule or placebo
Outcomes Primary endpoint(s): 
  • Adverse events and high‐density lipoprotein values (EGCg versus placebo, months 0 to 12 and months 12 to 36)

  • 6‐minute walk test (difference between month 0 and 36)


Secondary objective: to compare secondary clinical endpoints between the EGCg and the placebo group
Time point(s) of evaluation of this endpoint: after 12 and 36 months
Secondary endpoint(s): 
  • Progression (Medical Research Council Score, Hammersmith Scale for Assessment of Motor Ability Score, timed function tests)

  • Contractures (neutral‐zero‐method, a standardised orthopaedic evaluation and documentation index for the mobility of joints)


Time point(s) of evaluation of this endpoint: after 36 months
Starting date 25 June 2010
Contact information Name: Paul Friedmann  
Address: Chariteplatz 1 10117 Berlin, Germany
Telephone: 4930450539755
Email: friedemann.paul@charite.de
Notes Source of monetary support: aktion benni & co. Deutsche Gesellschaft für Muskelkranke e.V. 
It is unclear whether patients with respiratory decline will be included in this study.

DMD: Duchenne muscular dystrophy

Differences between protocol and review

Change in the outcomes

We divided the outcome 'any adverse events' into 'serious adverse events' and 'non‐serious adverse events' in order to maximise the use of the available data.

We extracted and analysed data for the outcome 'change in muscle function' in two separate groups of muscles.

We planned to pilot our data extraction form on at least one study, but only one eligible study provided data.  

Methods not implemented

We were not able to assess possible small‐study effects; however, we performed judgements on publication bias (see Quality of the evidence).

We were not able to carry out the subgroup analyses planned in the protocol as we found no studies with these characteristics. 

We were not able to carry out sensitivity analyses because we did not find unpublished studies or studies with an overall high risk of bias. 

Clarification of selected studies

We noted that we included studies that involved both eligible and ineligible participants (e.g. a subset of participants in the respiratory decline phase of DMD) if data for eligible participants were presented separately.

Contributions of authors

Roles and responsibilities (protocol and review stage)
Guarantor of the review LIG
Draft the protocol LIG, PRR, CMEL, JVAF, MRH    
Develop and run the search strategy LIG, CMEL, MRH, JVAF    
Obtain copies of studies CMEL, JVAF
Select which studies to include (2 people) LIG, PRR, MRH, JVAF    
Extract data from studies (2 people) LIG, PRR, JVAF    
Enter data into Review Manager 5 LIG, PRR, JVAF    
Carry out the analysis LIG, PRR, JVAF    
Interpret the analysis LIG, PRR, MRH, JVAF
Draft the final review LIG, PRR, CMEL, MRH, JVAF 
Update the review LIG, PRR, CMEL, MRH, JVAF   

Sources of support

Internal sources

  • Instituto Universitario Hospital Italiano, Argentina

    Provided support for LIG and PRR as Research Fellows

    Provided the salary for JVAF and CMEL as staff for the Cochrane Centre

  • Servicio de Neurología ‐ Hospital Italiano de Buenos Aires, Argentina

    Provided support for the participation of MRH

External sources

  • None, Other

    None

Declarations of interest

LIG: none known.

MRH: is a paediatric neurologist in an academic hospital and provides care to children with Duchenne muscular dystrophy.

PRR: none known.

CMEL: none known.

JVAF: none known.

New

References

References to studies included in this review

DELOS 2015 {published data only}

  1. Buyse G, Meier T. Idebenone reduces loss of respiratory function in Duchenne muscular dystrophy: outcome of a phase III double blind, randomized, placebo-controlled trial. Neuropediatrics 2015;46(S1):FV03-08. [Google Scholar]
  2. Buyse G, Voit T, Schara U, Straathof CS, D'Angelo MG, Bernert G, et al. Treatment effect of idebenone on inspiratory function in patients with Duchenne muscular dystrophy. Journal of Neuromuscular Diseases 2016;3:S107-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Buyse G, Voit T, Schara U, Verschuuren J, D'Angelo M, Bernert G, et al. Idebenone reduces loss of respiratory function in Duchenne muscular dystrophy - outcome of a phase III double blind, randomised, placebo-controlled trial (DELOS). American Academy of Neurology 2015;84:Abstract: 15-1-442-AAN.
  4. Buyse GM, Rummey C, Meier T, Leinonen M, Voit T, McDonald CM, et al. Home-based monitoring of pulmonary function in patients with Duchenne muscular dystrophy. Journal of Neuromuscular Diseases 2018;5(4):419-30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Buyse GM, Voit T, Schara U, Straathof CS, D'Angelo MG, Bernert G, et al. Efficacy of idebenone on respiratory function in patients with Duchenne muscular dystrophy not using glucocorticoids (DELOS): a double-blind randomised placebo-controlled phase 3 trial. Lancet 2015;385:1748-57. [DOI] [PubMed] [Google Scholar]
  6. Buyse GM, Voit T, Schara U, Straathof CS, D'Angelo MG, Bernert G, et al. Treatment effect of idebenone on inspiratory function in patients with Duchenne muscular dystrophy. Pediatric Pulmonology 2017;52(4):508-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Cirrincione A, Leinonen M, Hasham S,  Buyse G, DELOS study group. Idebenone preserves respiratory function above clinically relevant thresholds of FVC in Duchenne muscular dystrophy (DMD). European Journal of Paediatric Neurology 2017;21(Suppl 1):e92. [Google Scholar]
  8. EUCTR2009-012037-30-FR. A phase III double-blind, randomised, placebo-controlled study of the efficacy, safety and tolerability of idebenone in 10 – 18 year old patients with Duchenne muscular dystrophy - DELOS. www.clinicaltrialsregister.eu/ctr-search/trial/2009-012037-30/FR (first received 17 December 2009).
  9. Hasham S, Meier T, Leinonen M, Voit T, Mayer OH, Buyse G for the DELOS Study Group. Evaluating the effects of baseline variables on the respiratory function benefit of idebenone in Duchenne muscular dystrophy. Journal of Neuromuscular Diseases 2018;5(Suppl 1):S185-6. [Google Scholar]
  10. Karafilidis J, Leinonen M, Buyse G. Impact of idebenone on rate of respiratory function decline in Duchenne muscular dystrophy (DMD). Neurology 2018;90(15 Suppl):P5.436. [Google Scholar]
  11. Karafilidis J, Mayer H, Rummey C, Buyse G. Effect of idebenone on respiratory function across various subgroups in patients with Duchenne muscular dystrophy (DMD). American Journal of Respiratory and Critical Care Medicine 2018;197:A5865. [Google Scholar]
  12. Karafilidis J, Mayer H, Rummey C, Buyse G. Effect of idebenone on respiratory function across various subgroups in patients with Duchenne muscular dystrophy (DMD). American Thoracic Society International Conference Abstracts 2018;197:A5865. [Google Scholar]
  13. Lawrence C, Warnock A, McDonald C, Mayer O, Meier T, Leinonen M, et al on behalf of the DELOS Study Group. Effect of idebenone on bronchopulmonary adverse events and hospitalizations in patients with Duchene muscular dystrophy (DMD). Neuromuscular Disorders 2018;28(Suppl 1):S16-7. [Google Scholar]
  14. Leinonen M,  Mayer O. Efficacy of idebenone in respiratory decline in Duchenne muscular dystrophy (DMD): comparison of analysis methods. European Respiratory Journal 2017;50(Suppl 61):OA2928. [Google Scholar]
  15. Leinonen M, Warnock A, Buyse G. Efficacy of idebenone in respiratory decline in Duchenne muscular dystrophy (DMD): comparison of analysis methods. Developmental Medicine and Child Neurology 2017;59(S4):15. [Google Scholar]
  16. Lutz S, Buyse G, Meier T, Schara U. DELOS phase III study with idebenone (Catena®) in Duchenne muscular dystrophy. Neuropediatrics 2012;43(2):PS18_07. [Google Scholar]
  17. Mayer O,  Leinonen M,  Rummey C, Meier T,  Buyse G. Efficacy of idebenone to preserve respiratory function above clinically meaningful thresholds for forced vital capacity (FVC) in patients with Duchenne muscular dystrophy. Journal of Neuromuscular Diseases 2017;4(3):189-98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Mayer O, McDonald C, Meier T,  Leinonen M,  Buyse G. Effect of idebenone on bronchopulmonary adverse events and hospitalizations in patients with Duchene muscular dystrophy (DMD). American Journal of Respiratory and Critical Care Medicine 2017;195:A6876. [Google Scholar]
  19. Mayer O, McDonald C, Meier T, Leinonen M, Buyse G. Effect of idebenone on bronchopulmonary adverse events and hospitalizations in patients with Duchene muscular dystrophy (DMD). American Journal of Respiratory and Critical Care Medicine 2017;195:A6876.
  20. Mayer OH, Leinonen M, Hasham S, Meier T, Voit T, Buyse G for the DELOS study group. Impact of idebenone on respiratory burden, including risk of bronchopulmonary complications, in Duchenne muscular dystrophy. Journal of Neuromuscular Diseases 2018;5(Suppl 1):48-9. [Google Scholar]
  21. Mayer OH, Leinonen M, Rummey C, Meier T, Hasham S, Voit T, et al. Alternative analyses of respiratory function in Duchenne muscular dystrophy: consistent treatment benefit of idebenone. Journal of Neuromuscular Diseases 2018;5(Suppl 1):S321-2. [Google Scholar]
  22. McDonald CM, Meier T, Voit T, Schara U, Straathof CS, D'Angelo MG, et al. Idebenone reduces respiratory complications in patients with Duchenne muscular dystrophy. Journal of Neuromuscular Diseases 2016;3(Suppl 1):S107. [DOI] [PubMed] [Google Scholar]
  23. McDonald CM, Meier T, Voit T, Schara U, Straathof CS, D'Angelo MG, et al. Idebenone reduces respiratory complications in patients with Duchenne muscular dystrophy. Neuromuscular Disorders 2016;26(8):473-80. [DOI] [PubMed] [Google Scholar]
  24. NCT01027884. Phase III study of idebenone in Duchenne muscular dystrophy (DMD). clinicaltrials.gov/ct2/show/NCT01027884 (first received 19 October 2015).
  25. Rummey C,  Hasham S,  Mayer O,  Buyse G. Effects of idebenone on pulmonary morbidity in Duchenne muscular dystrophy (DMD). European Respiratory Journal 2017;50(Suppl 61):OA2927. [Google Scholar]
  26. Warncok A, McDonald C, Mayer O, Meier T, Leinonen M, Buyse G. Effect of idebenone on bronchopulmonary adverse events and hospitalizations in patients with Duchene muscular dystrophy (DMD). Developmental Medicine and Child Neurology 2017;59(S4):29. [Google Scholar]
  27. Warnock A, Lawrence C, Leinonen M, Buyse G. Efficacy of idebenone in respiratory decline in Duchenne muscular dystrophy (DMD): comparison of analysis methods. Neuromuscular Disorders 2018;28(Suppl 1):S17. [Google Scholar]
  28. Warnock A, Lawrence C, Leinonen M, Buyse G. Efficacy of idebenone in respiratory decline in Duchenne muscular dystrophy (DMD): comparison of analysis methods. Neuromuscular Disorders 2018;28(Suppl 1):S17.

SIDEROS 2021 {published data only}

  1. Buyse G, Mayer OH, Donisa-Dreghici R, Couttet F, Wolff J, Coppard N. A phase III double-blind, randomized, placebo controlled study (SIDEROS) assessing the efficacy of idebenone in slowing the rate of respiratory function loss in patients with Duchenne muscular dystrophy receiving glucocorticoid steroids. Journal of Neuromuscular Diseases 2016;3(S1):S103-4. [Google Scholar]
  2. EUCTR2016-000602-10-AT. A clinical study to assess how effective and safe is idebenone treatment in patients with Duchenne muscular dystrophy (DMD) who are currently receiving glucocorticoid steroids. www.clinicaltrialsregister.eu/ctr-search/trial/2016-000602-10/AT (first received 1 December 2016).
  3. NCT02814019. A Phase III double-blind study with idebenone in patients with Duchenne muscular dystrophy (DMD) taking glucocorticoid steroids. clinicaltrials.gov/ct2/show/NCT02814019 (first received 27 June 2016).
  4. Santhera Pharmaceuticals Holding AG. Santhera to discontinue phase 3 SIDEROS study and development of Puldysa® in Duchenne muscular dystrophy (DMD) and focus on vamorolone. www.santhera.com/assets/files/press-releases/2020-10-06_SiderosPuldysa_e_final.pdf (accessed 2 August 2021).

References to studies excluded from this review

Bäckman 1988 {published data only}

  1. Bäckman E, Nylander E, Johansson I, Henriksson K, Tagesson C. Selenium and vitamin E treatment of Duchenne muscular dystrophy: no effect on muscle function. Acta Neurologica Scandinavica 1988;78(5):429-35. [DOI] [PubMed] [Google Scholar]

Barcellona 2014 {published data only}

  1. Barcellona C, Vita G,  Licata N, Sframeli M, Bitto A, Distefano M, et al. Pilot study of flavocoxid in ambulant DMD patients. Acta Myologica 2014;33(1):49. [Google Scholar]

Bertorini 1985 {published data only}

  1. Bertorini T, Palmieri G, Griffin J, Chesney C, Pifer D, Verling L, et al. Chronic allopurinol and adenine therapy in Duchenne muscular dystrophy: effects on muscle function, nucleotide degradation, and muscle ATP and ADP content. Neurology 1985;35(1):61-5. [DOI] [PubMed] [Google Scholar]

Bonati 2014 {published data only}

  1. Bonati U, Hafner P, Erne B, Neuhaus C, Gloor M, Bieri O, et al. Successful pilot trial of L-arginine and metformin in Duchenne's muscular dystrophy. Journal of Neuromuscular Diseases 2014;1:S138-9. [Google Scholar]
  2. Bonati U, Hafner P, Erne B, Thomas E, Rutz E, Frank S, et al. Improved muscle function in Duchenne muscular dystrophy using a combination of l-arginine and metformin. Neuropediatrics 2015;46:PS02-34. [Google Scholar]
  3. Hafner P, Bonati U, Klein A, Rubino D, Gocheva V, Schmidt S, et al. Effect of combination l-citrulline and metformin treatment on motor function in patients with Duchenne muscular dystrophy: a randomized clinical trial. JAMA Network Open 2019;2(10):e1914171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Hafner P, Bonati U, Klein A, Rubino D, Gocheva V, Schmidt S, et al. L-citrulline and metformin delay muscle degeneration in Duchenne muscular dystrophy: results from a randomised clinical trial. Journal of Neuromuscular Diseases 2018;5:S116. [Google Scholar]
  5. Hafner P, Bonati U, Neuhaus C, Rutz E, Frank S, Erne B, et al. Succesful pilot trial of L-arginine and metformin in Duchenne's muscular dystrophy. Clinical Neurophysiology 2014;125(S1):S30. [Google Scholar]
  6. Hafner P, Bonati U, Rubino D, Gocheva V, Zumbrunn T, Gueven N, et al. Treatment with L-citrulline and metformin in Duchenne muscular dystrophy: study protocol for a single-centre, randomised, placebo-controlled trial. Trials 2016;17(1):389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. NCT01995032. L-citrulline and metformin in Duchenne's muscular dystrophy. clinicaltrials.gov/ct2/show/NCT01995032 (first received 26 November 2013).

Buyse 2008 {published data only}

  1. Buyse G, Mertens L, Van den Hauwe M, Thijs D, De Groot I, Schara U. Double-blind randomized controlled trial of SNT-MC17/idebenone in Duchenne muscular dystrophy. Neurology 2008;70(11 Suppl 1):A79. [DOI] [PubMed] [Google Scholar]

Buyse 2013 {published data only}

  1. Buyse G, Goemans N, Van den Hauwe M, Meier T. Effects of glucocorticoids and idebenone on respiratory function in patients with Duchenne muscular dystrophy. Pediatrics Pulmonology 2013;48(9):912-20. [DOI] [PubMed] [Google Scholar]

Buyse 2015a {published data only}

  1. Buyse G. Idebenone improves respiratory function in pediatric and adolescent patients with Duchenne muscular dystrophy not using glucocorticoid steroids. Neuromuscular Disorders 2015;25:S262. [Google Scholar]

Buyse 2015b {published data only}

  1. Buyse GM, Meier T. Idebenone as a new therapy to treat respiratory function loss in Duchenne muscular dystrophy: from preclinical studies to phase III trial outcome. European Journal of Paediatric Neurology 2015;19:S13. [Google Scholar]

Buyse 2019 {published data only}

  1. Buyse G, Servais L,  Mayer O, McDonald C, Voit T, Mercuri E. Consistent long-term effect of idebenone in reducing respiratory function decline in advanced patients with Duchenne muscular dystrophy. Neuromuscular Disorders 2019;29:S143. [Google Scholar]

DELPHI 2011 {published data only}

  1. Buyse G, Goemans N, Van den Hauwe M, Thijs D, De Groot I, Schara U, et al. Idebenone as a novel, therapeutic approach for Duchenne muscular dystrophy: results from a 12 month, double-blind, randomized placebo-controlled trial. Neuromuscular Disorders 2011;21(6):396-405. [DOI] [PubMed] [Google Scholar]

Escolar 2012 {published data only}

  1. Escolar D, Zimmerman A,  Bertorini T, Clemens P, Connolly A, Mesa L, et al. Pentoxifylline as a rescue treatment for DMD: a randomized double-blind clinical trial. Neurology 2012;78(12):904-13. [DOI] [PMC free article] [PubMed] [Google Scholar]

EUCTR2005‐002520‐33‐BE {published data only}

  1. EUCTR2005-002520-33-BE. A Phase IIa double blind, randomised, placebo controlled, single centre study at the University of Leuven to assess the efficacy and tolerability of idebenone in 10 - 16 year old males with cardiac dysfunction associated with Duchenne muscular dystrophy. www.clinicaltrialsregister.eu/ctr-search/trial/2005-002520-33/BE (first received 4 October 2005).

EUCTR2017‐002213‐60‐GB {published data only}

  1. EUCTR2017-002213-60-GB. A study to investigate the safety, tolerability, and efficacy of SGT-001 in male adolescents and children with Duchenne muscular dystrophy. www.clinicaltrialsregister.eu/ctr-search/trial/2017-002213-60/GB (first received 11 December 2018).

EUCTR2017‐004279‐30‐GB {published data only}

  1. EUCTR2017-004279-30-GB. A phase III open-label extension study to assess the long-term safety and efficacy of idebenone in patients with Duchenne muscular dystrophy (DMD) who completed the SIDEROS study. www.clinicaltrialsregister.eu/ctr-search/trial/2017-004279-30/GB (first received 9 May 2018).

EUCTR2019‐004602‐94‐BE {published data only}

  1. EUCTR2019-004602-94-BE. A 3-part, randomized, double blind, adaptive seamless phase 1-3 study to evaluate the safety, efficacy, pharmacokinetics, and pharmacodynamics of BIO101 in non-ambulatory patients with a genetically confirmed diagnosis of Duchenne muscular dystrophy and evidence of respiratory deterioration. clinicaltrialsregister.eu/ctr-search/trial/2019-004602-94/BE (first received 3 February 2020).

Fenichel 1988 {published data only}

  1. Fenichel G, Brooke M, Griggs R, Mendell J, Miller J, Moxley R, et al. Clinical investigation in Duchenne muscular dystrophy: penicillamine and vitamin E. Muscle & Nerve 1988;11(11):1164-8. [DOI] [PubMed] [Google Scholar]

Hunter 1983 {published data only}

  1. Hunter J, Galloway J, Brooke M, Kutner M, Rudman D, Vogel R, et al. Effects of allopurinol in Duchenne's muscular dystrophy. Archives of Neurology 1983;40(5):294-9. [DOI] [PubMed] [Google Scholar]

Mendell 1979 {published data only}

  1. Mendell J, Wiechers D. Lack of benefit of allopurinol in Duchenne dystrophy. Muscle & Nerve 1979;2(1):53-6. [DOI] [PubMed] [Google Scholar]

Nagy 2019 {published data only}

  1. Nagy S, Hafner P, Schmidt S, Rubino-Nacht D, Schadelin S, Bieri O, et al. Tamoxifen in Duchenne muscular dystrophy (TAMDMD): study protocol for a multicenter, randomized, placebo-controlled, double-blind phase 3 trial. Trials 2019;20:637. [DOI] [PMC free article] [PubMed] [Google Scholar]

NCT00308113 {published data only}

  1. NCT00308113. CoQ10 and prednisone in non-ambulatory DMD. clinicaltrials.gov/ct2/show/NCT00308113 (first received 29 March 2006).

NCT03603288 {published data only}

  1. NCT03603288. A phase III open-label extension study to assess the long-term safety and efficacy of idebenone in patients with Duchenne muscular dystrophy (DMD) who completed the SIDEROS study. clinicaltrials.gov/ct2/show/NCT03603288 (first received 27 July 2018).

Stern 1981 {published data only}

  1. Stern L,  Fewings J, Bretag A, Ballard F, Tomas F, Cooper D, et al. The progression of Duchenne muscular dystrophy: clinical trial of allopurinol therapy. Neurology 1981;31(4):422-6. [DOI] [PubMed] [Google Scholar]

Stern 1982 {published data only}

  1. Stern L,  Ringel S, Ziter F, Menander-Huber K, Lonasescu V, Pellegrino R, et al. Drug trial of superoxide dismutase in Duchenne's muscular dystrophy. Archives of Neurology 1982;39(6):342-6. [DOI] [PubMed] [Google Scholar]

SYROS 2019 {published data only}

  1. Mayer O,  Leinonen M, Servais L, Straathof C, Schara U,  Voit T, et al. Evaluating the effect of long-term idebenone treatment on respiratory morbidity in patients with Duchenne muscular dystrophy (DMD). European Respiratory Journal 2019;54:OA5330. [Google Scholar]
  2. Mayer O, Leinonen M, Hasham S, Buyse G. Long term efficacy: idebenone reduces the rate of both inspiratory and expiratory functional loss in Duchenne muscular dystrophy (DMD). European Respiratory Journal 2019;54:OA5331. [Google Scholar]
  3. Mayer O, Leinonen M, Hasham S, Buyse G on behalf of the SYROS investigators. Treatment effect of idebenone on inspiratory flow reserve (IFR) in patients with Duchenne muscular dystrophy (DMD): long term analysis from the SYROS study. American Journal of Respiratory and Critical Care Medicine 2019;199:A7449. [Google Scholar]
  4. Servais L, Mayer O, McDonald C, Straathof C, Schara U, Voit T, et al. P.264SYROS study – long-term reduction in rate of respiratory function decline in patients with Duchenne muscular dystrophy treated with idebenone. Neuromuscular Disorders 2019;29:S144. [Google Scholar]
  5. Servais L, Straathof CSM, Schara U, Klein A, Leinonen M, Hasham S, et al. Long-term data with idebenone on respiratory function outcomes in patients with Duchenne muscular dystrophy. Neuromuscular Disorders 2020;30(1):5-16. [DOI: 10.1016/j.nmd.2019.10.008.] [DOI] [PubMed] [Google Scholar]

Tamari 1982 {published data only}

  1. Tamari H, Ohtani Y, Higashi A, Miyoshino S, Matsuda I. Xanthine oxidase inhibitor in Duchenne muscular dystrophy. Brain & Development 1982;4(2):137-43. [DOI] [PubMed] [Google Scholar]

References to ongoing studies

SUNIMUD {published data only}

  1. EUCTR2009-016482-28-DE. SUNIMUD - sunphenon EGCg (epigallocatechin-gallate) in Duchenne muscular dystrophy. www.clinicaltrialsregister.eu/ctr-search/trial/2009-016482-28/DE (first received 15 March 2010).
  2. NCT01183767. Sunphenon epigallocatechin-gallate (EGCg) in Duchenne muscular dystrophy. clinicaltrials.gov/ct2/show/NCT01183767 (first received 18 August 2010).
  3. Ruegg UT, Moers A, Paul F, Nakae Y, Dorchies O. Focused conference group: P10 - drugs for half the world: paediatric clinical pharmacology clinical trial of epigallocatechin-3-gallate in Duchenne muscular dystrophy. Basic & Clinical Pharmacology & Toxicology 2010;107 (Suppl 1):550. [Google Scholar]
  4. Von Moers A, Paul F, Schülke M, Ohlraum S, Nakae Y, Dorchies O, et al. T.P.3.08 Clinical trial of epigallocatechin-3-gallate in Duchenne muscular dystrophy. Neuromuscular Disorders 2009;19(8-9):P612. [Google Scholar]

Additional references

Annane 2014

  1. Annane D, Orlikowski D, Chevret S. Nocturnal mechanical ventilation for chronic hypoventilation in patients with neuromuscular and chest wall disorders. Cochrane Database of Systematic Reviews 2014, Issue 12. Art. No: CD001941. [DOI: 10.1002/14651858.CD001941.pub3] [DOI] [PMC free article] [PubMed] [Google Scholar]

Aslesh 2018

  1. Tejal A, Rika M, Toshifumi Y. Skipping multiple exons to treat DMD-promises and challenges. Biomedicines 2018;6(1):E1. [DOI] [PMC free article] [PubMed] [Google Scholar]

Bellinger 2009

  1. Bellinger AM, Reiken S, Carlson C, Mongillo M, Liu X, Rothman L, et al. Hypernitrosylated ryanodine receptor calcium release channels are leaky in dystrophic muscle. Nature Medicine 2009;15(3):325-30. [DOI] [PMC free article] [PubMed] [Google Scholar]

Bérard 2005

  1. Bérard C, Payan C, Hodgkinson I, Fermanian J. A motor function measure for neuromuscular diseases. Construction and validation study. Neuromuscular Disorders: NMD 2005;15(7):463-70. [PMID: ] [DOI] [PubMed] [Google Scholar]

Birnkrant 2018a

  1. Birnkrant DJ, Bushby K, Bann CM, Apkon SD, Blackwell A, Brumbaugh D, et al, DMD Care Considerations Working Group. Diagnosis and management of Duchenne muscular dystrophy, part 1: diagnosis, and neuromuscular, rehabilitation, endocrine, and gastrointestinal and nutritional management. Lancet Neurology 2018;17(3):251-67. [DOI] [PMC free article] [PubMed] [Google Scholar]

Birnkrant 2018b

  1. Birnkrant DJ, Bushby K, Bann CM, Alman BA, Apkon SD, Blackwell A, et al,  DMD Care Considerations Working Group. Diagnosis and management of Duchenne muscular dystrophy, part 2: respiratory, cardiac, bone health, and orthopaedic management. Lancet Neurology 2018;17(4):347-61. [DOI] [PMC free article] [PubMed] [Google Scholar]

Bourke 2018

  1. Bourke J, Bueser T, Quinlivan R. Interventions for preventing and treating cardiac complications in Duchenne and Becker muscular dystrophy and X-linked dilated cardiomyopathy. Cochrane Database of Systematic Reviews 2018, Issue 10. Art. No: CD009068. [DOI: 10.1002/14651858.CD009068.pub3] [DOI] [PMC free article] [PubMed] [Google Scholar]

Brookes 2004

  1. Brookes PS, Yoon Y, Robotham JL, Anders MW, Sheu S-S. Calcium, ATP, and ROS: a mitochondrial love-hate triangle. American Journal of Physiology. Cell Physiology 2004;287(4):C817-33. [DOI] [PubMed] [Google Scholar]

Burdi 2009

  1. Burdi R, Rolland J-F, Fraysse B, Litvinova K, Cozzoli A, Giannuzzi V, et al. Multiple pathological events in exercised dystrophic mdx mice are targeted by pentoxifylline: outcome of a large array of in vivo and ex vivo tests. Journal of Applied Physiology 2009;106(4):1311-24. [DOI] [PubMed] [Google Scholar]

Buyse 2015

  1. Buyse GM, Voit T, Schara U, Straathof CS, D'Angelo MG, Bernert G, et al, DELOS Study Group. Efficacy of idebenone on respiratory function in patients with Duchenne muscular dystrophy not using glucocorticoids (DELOS): a double-blind randomised placebo-controlled phase 3 trial. Lancet 2015;385(9979):1748-57. [DOI] [PubMed] [Google Scholar]

Cardoso 1999

  1. Cardoso SM, Pereira C, Oliveira R. Mitochondrial function is differentially affected upon oxidative stress. Free Radical Biology & Medicine 1999;26(1-2):3-13. [DOI] [PubMed] [Google Scholar]

Covidence [Computer program]

  1. Veritas Health Innovation Covidence. Version accessed 23 March 2021. Melbourne, Australia: Veritas Health Innovation. Available at covidence.org.

du Bois 2011

  1. Du Bois RM, Weycker D, Albera C, Bradford WZ, Costabel U, Kartashov A, et al. Forced vital capacity in patients with idiopathic pulmonary fibrosis. American Journal of Respiratory and Critical Care Medicine 2011;184(12):1382-9. [DOI] [PubMed] [Google Scholar]

EPAR 2015

  1. Raxone. www.ema.europa.eu/en/documents/overview/raxone-epar-summary-public_en.pdf (accessed prior to 27 October 2021).

Finder 2004

  1. 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. American Journal of Respiratory and Critical Care Medicine 2004;170:456-65. [DOI] [PubMed] [Google Scholar]

Finder 2009

  1. Finder JD. A 2009 perspective on the 2004 American Thoracic Society statement, "Respiratory Care of the Patient With Duchenne Muscular Dystrophy". Pediatrics 2009;123:S239–41. [DOI] [PubMed] [Google Scholar]

Gogou 2020

  1. Gogou M, Pavlou E, Haidopoulou K. Therapies that are available and under development for Duchenne muscular dystrophy: what about lung function? Pediatric Pulmonology 2020;55(2):300-15. [DOI] [PubMed] [Google Scholar]

GRADEpro GDT [Computer program]

  1. McMaster University (developed by Evidence Prime) GRADEpro GDT. Version accessed 20 August 2020. Hamilton (ON): McMaster University (developed by Evidence Prime). Available at gradepro.org.

Graham 2019

  1. Graham BL, Steenbruggen I, Miller MR, Barjaktarevic IZ, Cooper BG, Hall GL,   et al. Standardization of Spirometry 2019 Update. An Official American Thoracic Society and European Respiratory Society Technical Statement. American Journal of Respiratory and Critical Care Medicine 2019;200(8):e70-88. [DOI] [PMC free article] [PubMed] [Google Scholar]

Haycock 1996

  1. Haycock JW, MacNeil S, Jones P, Harris JB, Mantle D. Oxidative damage to muscle protein in Duchenne muscular dystrophy. Neuroreport 1996;8(1):357-61. [DOI] [PubMed] [Google Scholar]

Head 2017

  1. Head SI. Antioxidant therapy in a mouse model of Duchenne muscular dystrophy: some promising results but with a weighty caveat. Journal of Physiology 2017;595(23):7015. [DOI] [PMC free article] [PubMed] [Google Scholar]

Henricson 2013

  1. Henricson EK, Abresch RT, Cnaan A, Hu F, Duong T, Arrieta A, et al, CINRG Investigators. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]

Hermes 2016

  1. Hermes Tde A, Macedo AB, Fogaça AR, Moraes LH, Faria FM, Kido LA, et al. Beneficial cilostazol therapeutic effects in mdx dystrophic skeletal muscle. Clinical and Experimental Pharmacology & Physiology 2016;43(2):259-67. [DOI] [PubMed] [Google Scholar]

Hibaoui 2011

  1. Hibaoui Y, Reutenauer-Patte J, Patthey-Vuadens O, Ruegg UT, Dorchies OM. Melatonin improves muscle function of the dystrophic mdx5Cv mouse, a model for Duchenne muscular dystrophy. Journal of Pineal Research 2011;51(2):163-71. [DOI] [PubMed] [Google Scholar]

Higgins 2021a

  1. Higgins JP, Thomas J, Chandler J, Cumpston M, Li T, Page MJ, Welch VA, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 6.2 (updated February 2021). Cochrane, 2021. Available from training.cochrane.org/handbook. [DOI] [PMC free article] [PubMed]

Higgins 2021b

  1. Higgins JPT,  Savović J, Page MJ, Elbers RG, Sterne JAC. Chapter 8: Assessing risk of bias in a randomized trial. In: Higgins JPT, Thomas J, Chandler J, Cumpston M, Li T, Page MJ, Welch VA, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 6.2 (updated February 2021). Cochrane, 2021. Available from training.cochrane.org/handbook.

Hilliard 2013

  1. Hilliard ME, Lawrence JM, Modi AC, Anderson A, Crume T, Dolan LM, et al. Identification of minimal clinically important difference scores of the PedsQL in children, adolescents, and young adults with type 1 and type 2 diabetes. Diabetes Care 2013;36(7):1891-7. [DOI] [PMC free article] [PubMed] [Google Scholar]

Hnia 2007

  1. Hnia K, Hugon G, Rivier F, Masmoudi A, Mercier J, Mornet D. Modulation of p38 mitogen-activated protein kinase cascade and metalloproteinase activity in diaphragm muscle in response to free radical scavenger administration in dystrophin-deficient Mdx mice. American Journal of Pathology 2007;170(2):633-43. [DOI] [PMC free article] [PubMed] [Google Scholar]

Kawamura 2020

  1. Kawamura K, Fukumura S, Nikaido K, Tachi N, Kozuka N, Seino T,  et al. Resveratrol improves motor function in patients with muscular dystrophies: an open-label, single-arm, phase IIa study. Scientific Reports 2020;10(1):20585. [DOI] [PMC free article] [PubMed] [Google Scholar]

Landfeldt 2015

  1. Landfeldt E, Lindgren P, Bell CF, Schmitt C, Guglieri M, Straub V, et al. Compliance to care guidelines for Duchenne muscular dystrophy. Journal of Neuromuscular Diseases 2015;2(1):63-72. [DOI] [PMC free article] [PubMed] [Google Scholar]

Leinonen 2017

  1. Leinonen M. Meta-analysis of two clinical trials: slowing respiratory decline in Duchenne muscular dystrophy (DMD). European Respiratory Journal 2017;50(Suppl 61):1043. [Google Scholar]

Liberati 2009

  1. Liberati A, Altman DG, Tetzlaff J, Mulrow C, Gotzsche PC, Ioannidis JP, et al. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate health care interventions: explanation and elaboration. PLoS Medicine 2009;6(7):e1000100. [DOI] [PMC free article] [PubMed] [Google Scholar]

LoMauro 2018

  1. LoMauro A, Romei M, Gandossini S, Pascuzzo R, Vantini S, D'Angelo MG, et al. Evolution of respiratory function in Duchenne muscular dystrophy from childhood to adulthood. European Respiratory Journal 2018;51(2):1701418. [DOI] [PubMed] [Google Scholar]

Lyager 1995

  1. Lyager S, Steffensen B, Juhl B. Indicators of need for mechanical ventilation in Duchenne muscular dystrophy and spinal muscular atrophy. Chest 1995;108(3):779-85. [DOI] [PubMed] [Google Scholar]

Mâncio 2017a

  1. Mâncio RD, Hermes T de A, Macedo AB, Mizobuti DS, Rupcic IF, Minatel E. Dystrophic phenotype improvement in the diaphragm muscle of mdx mice by diacerhein. PLOS ONE 2017;12(8):e0182449. [DOI] [PMC free article] [PubMed] [Google Scholar]

Mâncio 2017b

  1. Mâncio RD, Hermes T de A, Macedo AB, Mizobuti DS, Valduga AH, Rupcic IF, et al. Vitamin E treatment decreases muscle injury in mdx mice. Nutrition (Burbank, Los Angeles County, Calif.) 2017;43-4:39-46. [DOI] [PubMed] [Google Scholar]

Marques 2010

  1. Marques MJ, Barbin IC, Taniguti AP, Oggian DS, Ferretti R, Santo Neto H. Myocardial fibrosis is unaltered by long-term administration of L-arginine in dystrophin deficient mdx mice: a histomorphometric analysis. Acta Biologica Hungarica 2010;61(2):168-74. [DOI] [PubMed] [Google Scholar]

Matthews 2016

  1. Matthews E, Brassington R, Kuntzer T, Jichi F, Manzur AY. Corticosteroids for the treatment of Duchenne muscular dystrophy. Cochrane Database of Systematic Reviews 2016, Issue 5. Art. No: CD003725. [DOI: 10.1002/14651858.CD003725.pub4] [DOI] [PMC free article] [PubMed] [Google Scholar]

Mázala 2015

  1. Mázala DA, Pratt SJ, Chen D, Molkentin JD, Lovering RM, Chin ER. SERCA1 overexpression minimizes skeletal muscle damage in dystrophic mouse models. American Journal of Physiology. Cell Physiology 2015;308(9):C699-709. [DOI] [PMC free article] [PubMed] [Google Scholar]

McDonald 2013

  1. McDonald CM, Henricson EK, Abresch RT, Florence J, Eagle M, Gappmaier E, et al for the DMD Study Group. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]

Mendell 2013

  1. Mendell JR, Lloyd-Puryear M. Report of MDA muscle disease symposium on newborn screening for Duchenne muscular dystrophy. Muscle & Nerve 2013;48(1):21-6. [DOI] [PubMed] [Google Scholar]

Miller 2005

  1. Miller MR, Hankinson J, Brusasco V, Burgos F, Casaburi R, Coates A, et al, ATS/ERS Task Force. Standardisation of spirometry. European Respiratory Journal 2005;26(2):319-38. [DOI] [PubMed] [Google Scholar]

Nakae 2008

  1. Nakae Y, Hirasaka K, Goto J, Nikawa T, Shono M, Yoshida M, et al. Subcutaneous injection, from birth, of epigallocatechin-3-gallate, a component of green tea, limits the onset of muscular dystrophy in mdx mice: a quantitative histological, immunohistochemical and electrophysiological study. Histochemistry and Cell Biology 2008;129(4):489-501. [DOI] [PubMed] [Google Scholar]

Orrell 2007

  1. Orrell RW, Lane RJ, Ross M. Antioxidant treatment for amyotrophic lateral sclerosis or motor neuron disease. Cochrane Database of Systematic Reviews 2007, Issue 1. Art. No: CD002829. [DOI: 10.1002/14651858.CD002829.pub2] [CD002829] [DOI] [PMC free article] [PubMed] [Google Scholar]

Peng 2010

  1. Peng TI, Jou MJ. Oxidative stress caused by mitochondrial calcium overload. Annals of the New York Academy of Sciences 2010;1201(1):183-8. [DOI] [PubMed] [Google Scholar]

Percival 2012

  1. Percival JM, Whitehead NP, Adams ME, Adamo CM, Beavo JA, Froehner SC. Sildenafil reduces respiratory muscle weakness and fibrosis in the mdx mouse model of Duchenne muscular dystrophy. Journal of Pathology 2012;228(1):77-87. [DOI] [PMC free article] [PubMed] [Google Scholar]

Petrof 1993

  1. Petrof BJ, Shrager JB, Stedman HH, Kelly AM, Sweeney HL. Dystrophin protects the sarcolemma from stresses developed during muscle contraction. Proceedings of the National Academy of Sciences 1993;90(8):3710-4. [DOI] [PMC free article] [PubMed] [Google Scholar]

Phillips 2008

  1. Phillips MF, Quinlivan R. Calcium antagonists for Duchenne muscular dystrophy. Cochrane Database of Systematic Reviews 2008, Issue 4. Art. No: CD004571. [DOI: 10.1002/14651858.CD004571.pub2] [DOI] [PMC free article] [PubMed] [Google Scholar]

Review Manager 2020 [Computer program]

  1. Nordic Cochrane Centre, The Cochrane Collaboration Review Manager 5 (RevMan 5). Version 5.4. Copenhagen: Nordic Cochrane Centre, The Cochrane Collaboration, 2020.

Rodriguez 2003

  1. Rodriguez MC, Tarnopolsky MA. Patients with dystrophinopathy show evidence of increased oxidative stress. Free Radical Biology & Medicine 2003;34(9):1217-20. [DOI] [PubMed] [Google Scholar]

Sander 2000

  1. Sander M, Chavoshan B, Harris SA, Iannaccone ST, Stull JT, Thomas GD, et al. Functional muscle ischemia in neuronal nitric oxide synthase-deficient skeletal muscle of children with Duchenne muscular dystrophy. Proceedings of the National Academy of Sciences of the United States of America 2000;97(25):13818-23. [DOI] [PMC free article] [PubMed] [Google Scholar]

Schünemann 2021a

  1. Schünemann HJ, Higgins JP, Vist GE, Glasziou P, Akl EA, Skoetz N, Guyatt GH. Chapter 14: Completing ‘Summary of findings’ tables and grading the certainty of the evidence. In: Higgins JP, Thomas J, Chandler J, Cumpston M, Li T, Page MJ, Welch VA, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 6.2 (updated February 2021). Cochrane, 2021. Available from training.cochrane.org/handbook.

Schünemann 2021b

  1. Schünemann HJ, Vist GE, Higgins JPT, Santesso N, Deeks JJ, Glasziou P, et al. Chapter 15: Interpreting results and drawing conclusions. In: Higgins JP, Thomas J, Chandler J, Cumpston M, Li T, Page MJ, Welch VA, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 6.2 (updated February 2021). Cochrane, 2021. Available from training.cochrane.org/handbook.

Servais 2013

  1. Servais L, Deconinck N, Moraux A, Benali M, Canal A, Van Parys F, et al. Innovative methods to assess upper limb strength and function in non-ambulant Duchenne patients. Neuromuscular Disorders: NMD 2013;23(2):139-48. [PMID: ] [DOI] [PubMed] [Google Scholar]

Shawi 2017

  1. Shawi F, Perras C, Severn M. Emerging Drugs for Duchenne Muscular Dystrophy. CADTH Issues in Emerging Health Technologies 2017;(161). [PubMed]

Sterne 2019

  1. Sterne JA, Savović J, Page MJ, Elbers RG, Blencowe NS, Boutron I, et al. RoB 2: a revised tool for assessing risk of bias in randomised trials. BMJ 2019;366:l4898. [DOI] [PubMed] [Google Scholar]

Stromberg 2012

  1. Stromberg A, Darin N, Kroksmark AK, Tulinius M. S.P.31 What was the age and cause of death in patients with Duchenne muscular dystrophy in Sweden during 2000–2010? Neuromuscular Disorders 2012;22(9-10):880-1. [Google Scholar]

Tonon 2012

  1. Tonon E, Ferretti R, Shiratori JH, Santo Neto H, Marques MJ, Minatel E. Ascorbic acid protects the diaphragm muscle against myonecrosis in mdx mice. Nutrition (Burbank, Los Angeles County, Calif.) 2012;28(6):686-90. [DOI] [PubMed] [Google Scholar]

Turner 1988

  1. Turner PR, Westwood T, Regen CM, Steinhardt RA. Increased protein degradation results from elevated free calcium levels found in muscle from mdx mice. Nature 1988;335(6192):735-8. [DOI] [PubMed] [Google Scholar]

Varni 2001

  1. Varni JW, Seid M, Kurtin PS. PedsQL™ 4.0: Reliability and Validity of the Pediatric Quality of Life Inventory™ Version 4.0 Generic Core Scales in Healthy and Patient Populations. Medical Care 2001;39(8):800-12. [DOI] [PubMed] [Google Scholar]

Walters 2005

  1. Walters SJ, Brazier JE. Comparison of the minimally important difference for two health state utility measures: EQ-5D and SF-6D. Quality of Life Research 2005;14(6):1523-32. [PMID: ] [DOI] [PubMed] [Google Scholar]

Werneck 2019

  1. Werneck L, Lorenzoni P, Ducci R, Fustes O, Kay C, Scola R. Duchenne muscular dystrophy: an historical treatment review. Arquivos de Neuro-Psiquiatria 2019;77(8):579-89. [DOI] [PubMed] [Google Scholar]

Wille 2010

  1. Wille N, Badia X, Bonsel G, Burström K, Cavrini G, Devlin N, et al. Development of the EQ-5D-Y: a child-friendly version of the EQ-5D. Quality of Life Research 2010;19(6):875-86. [DOI] [PMC free article] [PubMed] [Google Scholar]

Woodman 2016

  1. Woodman KG, Coles CA, Lamandé SR, White JD. Nutraceuticals and their potential to treat Duchenne muscular dystrophy: separating the credible from the conjecture. Nutrients 2016;8(11):E713. [DOI] [PMC free article] [PubMed] [Google Scholar]

References to other published versions of this review

Garegnani 2020 

  1. Garegnani L, Hyland M, Roson Rodriguez P, Escobar Liquitay CM, Quinlivan R, Franco JV. Antioxidants to prevent respiratory decline in people with Duchenne muscular dystrophy and progressive respiratory decline. Cochrane Database of Systematic Reviews 2020, Issue 9. Art. No: CD013720. [DOI: 10.1002/14651858.CD013720] [DOI] [Google Scholar]

Articles from The Cochrane Database of Systematic Reviews are provided here courtesy of Wiley

RESOURCES