533 PROFESSIONAL ARTICLE Duchenne muscular dystrophy – novelties in diagnostics and treatment Copyright (c) 2021 Slovenian Medical Journal. This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License. Duchenne muscular dystrophy – novelties in diagnostics and treatment Duchennova mišična distrofija – novosti pri diagnosticiranju in zdravljenju Tita Butenko,1 Tanja Loboda,1 Damjan Osredkar,1,2 Aleksandra Zver,3 Uroš Krivec,3 Primož Kotnik,2,4 Matjaž Homan,2,5 Mirko Topalović,6 Marko Pokorn,7,8,9 Katja Groleger Sršen,10,11 Petra Lešnik Musek,1 David Gosar,1 Alenka Piskar,1 Robert Košak,12 Tanja Golli1 Abstract Duchenne muscular dystrophy (DMD) is the most common and one of the most serious childhood diseases. It is an X – linked recessive neuromuscular disease, caused by mutations in the dystrophin gene, primarily resulting in skeletal and heart muscle abnormalities. In most boys, the first signs of the disease appear as progressive muscle weakness between the ages of 3 and 5 years. The muscle weakness is more pronounced in the proximal muscles, initially affecting the lower limbs to a greater extent than the upper limbs. Untreated, the disease progresses and will cause a serious decline of motor function by the age of 10 to 11 years, when the patients require wheelchairs even for travelling short distances. The pro- gressive respiratory muscle failure results in chronic respiratory insufficiency and the patients require ventilatory support. Heart muscle involvement is consistent, studies showing that cardiomyopathy is present in all patients by the age of 18. 1 Clinical Department of Child, Adolescent and Developmental Neurology, University Children’s Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia 2 Division of Pediatrics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia 3 Unit of Pulmonary Diseases, University Children’s Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia 4 Department of Pediatric Endocrinology, Diabetes and Metabolism, University Children’s Hospital, University Medical Centre Ljubljana 5 Clinical Department of Gastroenterology, Hepatology and Nutrition, University Children’s Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia 6 Department of Cardiology, University Children’s Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia 7 University Children’s Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia 8 Department of Infectious Diseases, University Medical Centre Ljubljana, Ljubljana, Slovenia 9 Division of Infectious Diseases and Epidemiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia 10 Department for Children Rehabilitation, University Rehabilitation Institute Republic of Slovenia, Ljubljana, Slovenia 11 Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia 12 Railway Community Health Center Ljubljana, Ljubljana, Slovenia Correspondence / Korespondenca: Tita Butenko, e: tita.butenko@kclj.si Key words: Duchenne muscular dystrophy; treatment; revised guidelines; outcome Ključne besede: Duchennova mišična distrofija; zdravljenje; nove smernice; izid Received / Prispelo: 4. 10. 2020 | Accepted / Sprejeto: 22. 2. 2021 Cite as / Citirajte kot: Butenko T, Loboda T, Osredkar D, Zver A, Krivec U, Kotnik P, et al. Duchenne muscular dystrophy – novelties in diagnostics and treatment. Zdrav Vestn. 2021;90(Epub ahead of print):1–17. DOI: https://doi.org/10.6016/ZdravVestn.3172 eng slo element en article-lang 10.6016/ZdravVestn.3172 doi 4.10.2020 date-received 22.2.2021 date-accepted Pediatrics Pediatrija discipline Professional article Strokovni članek article-type Duchenne muscular dystrophy – novelties in diagnostics and treatment Duchennova mišična distrofija - novosti pri diag- nosticiranju in zdravljenju article-title Duchenne muscular dystrophy Duchennova mišična distrofija alt-title Duchenne muscular dystrophy, treatment, revised guidelines, outcome Duchennova mišična distrofija, zdravljenje, nove smernice, izid kwd-group The authors declare that there are no conflicts of interest present. Avtorji so izjavili, da ne obstajajo nobeni konkurenčni interesi. conflict year volume first month last month first page last page 2021 90 9 10 533 549 name surname aff email Tita Butenko 1 tita.butenko@kclj.si name surname aff Tanja Loboda 1 Damjan Osredkar 1,2 Aleksandra Zver 3 Uroš Krivec 3 Primož Kotnik 2,4 Matjaž Homan 2,5 Mirko Topalović 6 Marko Pokorn 7,8,9 Katja Groleger Sršen 10,11 Petra Lešnik Musek 1 David Gosar 1 Alenka Piskar 1 Tanja Golli 1 eng slo aff-id Clinical Department of Child, Adolescent and Developmental Neurology, University Children's Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia Klinični oddelek za otroško, razvojno in mladostniško nevrologijo, Pediatrična klinika, Univerzitetni klinični center Ljubljana, Ljubljana, Slovenija 1 Division of Pediatrics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia Katedra za pediatrijo, Medicinska fakulteta, Ljubljana, Slovenija 2 Unit of Pulmonary Diseases, University Children's Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia Služba za pljučne bolezni, Pediatrična klinika, Univerzitetni klinični center Ljubljana, Ljubljana, Slovenija 3 Department of Pediatric Endocrinology, Diabetes and Metabolism, University Children's Hospital, University Medical Centre Ljubljana Klinični oddelek za endokrinologijo, diabetes in presnovne bolezni, Pediatrična klinika, Univerzitetni klinični center Ljubljana, Ljubljana, Slovenija 4 Clinical Department of Gastroenterology, Hepatology and Nutrition, University Children's Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia Klinični oddelek za gastroenterologijo, hepatologijo in nutricionistiko, Pediatrična klinika, Univerzitetni klinični center Ljubljana, Ljubljana, Slovenija 5 Department of Cardiology, University Children's Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia Služba za kardiologijo, Pediatrična klinika, Univerzitetni klinični center Ljubljana, Ljubljana, Slovenija 6 University Children's Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia Pediatrična klinika, Univerzitetni klinični center Ljubljana, Ljubljana, Slovenija 7 Department of Infectious Diseases, University Medical Centre Ljubljana, Ljubljana, Slovenia Klinika za infekcijske bolezni in vročinska stanja, Univerzitetni klinični center Ljubljana, Ljubljana, Slovenija 8 Division of Infectious Diseases and Epidemiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia Katedra za infekcijske bolezni in epidemiologijo, Medicinska fakulteta, Univerza v Ljubljani, Ljubljana, Slovenija 9 Department for Children Rehabilitation, University Rehabilitation Institute Republic of Slovenia, Ljubljana, Slovenia Oddelek za rehabilitacijo otrok, Univerzitetni rehabilitacijski inštitut Republike Slovenije, Ljubljana, Slovenija 10 Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia Medicinska fakulteta, Univerza v Ljubljani, Ljubljana, Slovenija 11 Railway Community Health Center Ljubljana, Ljubljana, Slovenia Železniški zdravstveni dom Ljubljana, Ljubljana, Slovenija 12 Slovenian Medical Journallovenian Medical Journal 534 PEDIATRICS Zdrav Vestn | september – october 2021 | Volume 90 | https://doi.org/10.6016/ZdravVestn.3172 1 Introduction Duchenne muscular dystrophy (DMD) is the most common muscular disease in children, with an inci- dence of 1 in 3,500 to 5,000 live-born boys (1-3). It is an inherited, X-linked recessive neuromuscular disease (NMD) caused by a mutation in the dystrophin gene that mainly affects men. The main symptom is progres- sive skeletal and heart muscle damage. However, there are also other associated health issues which jointly usually lead to loss of mobility and premature death. The diagnosis of DMD is usually made at the age of 5, when a difference in motor skills of sick children com- pared to healthy peers becomes apparent. In the early 1990s, the life expectancy of children with DMD was 20 years. With advances in treatment, however, both qual- ity of life and life expectancy have improved; the latter now reaches 30–40 years, and many patients may reach an even higher age. The article presents an overview of knowledge about DMD and novelties in the treatment and rehabilitation of children with DMD. 2 Genetics DMD is an inherited, X-linked recessive disease caused by a mutation in the DMD gene at the Xp21 locus, which encodes the skeletal muscle protein dys- trophin (2,4). Dystrophin is located on the sarcolemma The diagnosis of DMD is based on laboratory and genetic testing in patients with suspect clinical manifestations. If genetic testing is negative, muscle biopsy is necessary to confirm the diagnosis in highly suspect cases. Treatment of DMD patients requires a multidisciplinary approach, which coupled with corticosteroid treatment, physical therapy, supportive treat- ment and specific aids, have led to increased longevity and improved quality of life in these patients. Several drugs are being developed, targeting both the reduction of muscle damage, as well as the basic genetic cause of the disease. Izvleček Duchennova mišična distrofija (DMD) je najpogostejša in ena najresnejših mišičnih bolezni otroške dobe. Gre za na kromo- som X vezano recesivno živčno-mišično bolezen, ki jo povzroča mutacija v genu za distrofin. Primarno prizadene skeletne mišice in srčno mišico. Pri večini dečkov se klinični znaki bolezni izrazijo z napredujočo mišično šibkostjo med 3. in 5. letom starosti. Mišična šibkost je bolj izražena v proksimalnih kot distalnih mišicah in v začetni fazi bolj vpliva na poslabšanje funkcije spodnjih kot zgornjih udov. Bolezen postopno napreduje. Pri nezdravljenih otrocih večinoma po 11. do 12. letu starosti vodi v takó resno zmanjšanje zmožnosti gibanja, da le-ti že za premagovanje krajših razdalj potrebujejo invalidski voziček. Napredujoča šibkost dihalnih mišic vodi v kronično dihalno odpoved in potrebo po pomoči pri predihavanju. Okvara funkcije srca je splošno prisotna, kajti po študijah so klinični znaki kardiomiopatije prisotni pri vseh bolnikih po 18. letu starosti. Na podlagi klinične slike diagnozo DMD potrdimo z laboratorijskimi in genetskimi preiskavami. V primeru negativnih rezultatov genetskih preiskav, a ob močnem kliničnem sumu za DMD, pa se za potrditev diagnoze poslužuje- mo mišične biopsije. Zdravljenje bolnikov z DMD zahteva multidisciplinarno obravnavo. Z uporabo kortikosteroidov (KS), fizioterapije, podpornega zdravljenja in opreme s pripomočki sta se življenjska doba in kakovost življenja bolnikov z DMD izboljšala. V fazi razvoja je več zdravil, katerih delovanje se usmerja v zmanjšanje okvare mišic, a tudi zdravil, ki bi odpravile osnovni, tj. genetski vzrok bolezni. (plasma membrane) of skeletal muscle and is a compo- nent of a large glycoprotein complex. It acts as a me- chanical link between the cytoskeleton and the extra- cellular matrix (5). The primary consequence of this genetic mutation is skeletal muscle and heart muscle damage. Dystrophin provides stability, strength and functionality to muscle fibres, and its deficiency leads to their degeneration (6- 8). The DMD gene is the largest gene in the human ge- nome, containing 79 exons and is transcribed into a 14 kb-long mature messenger ribonucleic acid (mRNA). The size of the DMD gene contributes to a high rate of mutations in it (in a third of cases these occur de novo). They can shift the reading frame or form a premature stop codon, thus interrupting gene translation or caus- ing the formation of a shortened and unstable dystro- phin protein (1,5). The “reading frame” rule can explain most of the phenotypic differences between DMD and Becker muscular dystrophy (BMD); the latter is a mild- er form of dystrophinopathy. Mutations that shift the reading frame usually cause the formation of abnormal and shortened dystrophin and cause DMD. However, mutations that preserve the reading frame produce a shorter and partially functional dystrophin, which re- sults in the milder clinical picture of BMD (9,10). The majority of mutations in DMD are intragenic deletions, which account for 65–70% of all mutations and most 535 PROFESSIONAL ARTICLE Duchenne muscular dystrophy – novelties in diagnostics and treatment commonly occur in exons 45–53. Duplications are present in 7% of patients, and point mutations or small deletions/insertions are present in the rest (9,11,12). Thus, the clinical picture and prognosis of disease out- come can vary drastically depending on the type and location of the mutation (12). 3 Clinical picture In most boys with DMD, the clinical signs of the dis- ease are expressed between the ages of 3 and 5. Devel- opment is usually normal in the first few years of life, or there is only a slight delay in reaching developmental milestones (usually children start walking by the age of 18 months) (13). During infancy, the disease may man- ifest as mildly decreased muscle tone and poor head control. Muscle weakness usually begins to show be- tween the ages of 2 and 3, with gait abnormalities (e.g. walking on toes), difficulty running, difficulty walking up stairs or getting up from the floor, and frequent falls. Less commonly, the disease manifests as speech or global developmental delay. The random discovery of elevated serum creatine kinase (CK) or hepatic trans- aminases can also raise the suspicion of the disease in pre-symptomatic children. Since these findings may al- ready be elevated when the child is still asymptomatic, considering the differential diagnosis of DMD in these instances is important. It is characteristic for DMD that initially the proximal muscles of the lower limbs and the muscles of the trunk are more affected, but later al- so the muscles of the upper limbs and the distal muscles (3,6,14). Clinical symptoms and signs that gradually devel- op are: waddling gait, positive Gower’s sign (a result of weakness of the hip and thigh muscles and a character- istic manner of getting up from a lying and/or sitting position to an upright position; for a person to get up from a sitting or supine position, they must first turn around and become prone on the elbows and knees and extend them so that they can lift their torso; then, lean- ing on the ground, they gradually bring their arms and their legs closer in order to move their body’s centre of gravity over the legs; at this point, the person can release one hand and take hold of or lean on one knee, which is followed by the same movement with the other hand; then they gradually ‘climb’ up the legs to reach an upright or standing position; Gower’s sign is considered positive as soon as a person has to turn to a redirected position to lift themselves from a supine position, as ‘climbing’ up the legs in mild proximal muscle weakness might not take place; Gower’s sign is a non-specific sign of proximal muscle weakness that is not pathognomonic for DMD), (pseudo)hypertrophic and stiff calves, which are also occasionally painful, and in later stages, shortening of the Achilles and other tendons of the lower limb muscles and weakness of the neck muscles also occur. As the disease progresses, the tendon reflexes, distal following proximal, also gradu- ally dissipate (13,15,16). Most boys progress in muscle strength and motor skills (albeit to a lesser extent than their peers) by about the age of 6; after this age there is a progressive deteri- oration of muscle strength (14). The disease progresses rapidly after the age of 7 to 8, and untreated children become wheelchair dependent between the ages 11 and 12. Scoliosis and joint contractures develop due to loss of muscle function, ability to maintain posture and movement. A typical form of scoliosis is a long para- lytic C-curve involving the thoracic and lumbar spine, and in the advanced stage, pelvic tilt also occurs in most cases. Scoliosis progresses faster compared to idiopath- ic scoliosis. At the same time, the risk of vertebral and long bone fractures increases with the development of osteoporosis due to inactivity. After the loss of the abili- ty to move, problems with the heart, respiratory system and the already mentioned orthopaedic problems be- come more pronounced. It is also known that patients with DMD are more likely to develop puberty later, they are of shorter stature, and have an increased risk of malignant hyperthermia with anaesthesia and the use of depolarizing muscle relaxants (3,13,17). As already mentioned, the disease also affects the heart and lungs. Cardiac involvement is usually man- ifested by dilated cardiomyopathy and cardiac arrhyth- mias (10). The degree of cardiomyopathy in 50–80% of patients is not proportional to muscle weakness (18). Heart disease, which mostly manifests by persistent tachycardia, is present in up to 25% of children under the age of 6. Despite the high proportion of boys with heart muscle involvement, most of them are relatively asymptomatic due to physical inactivity (10). Nigro et al. (19) in a study published in 1990 found that clinical signs of cardiomyopathy are expressed after the age of 10 and are present in 30% of patients up to the age of 14, while after the age of 18 they are present in all patients. Lung damage occurs due to the progressive weakness of the respiratory muscles. The latter leads to ineffec- tive coughing, reduced nocturnal respiration (noctur- nal hypoventilation) and sleep-disordered breathing, and respiratory infections are also more common than in the general population (3,20,21). Progressive sco- liosis often impairs respiratory function even further. 536 PEDIATRICS Zdrav Vestn | september – october 2021 | Volume 90 | https://doi.org/10.6016/ZdravVestn.3172 Chronic respiratory failure as a result of restrictive lung disease is a common feature of these patients. Studies have shown that the vital capacity of this group of chil- dren increases until about 12 years of age, and then be- gins to decline by 4–8% per year (22,23). Respiratory complications are also a major cause of morbidity and mortality in patients with DMD (3,20,21). Various dystrophin isoforms have also been shown to be present in the brain, retina, and Purkinje cells. Mutations in these specific isoforms are most likely to cause extramuscular manifestations of the disease, manifested by delayed speech development, cogni- tion disorders, behavioural and learning difficulties, as well as attention and hyperactivity disorders, and autism. Various studies have shown that some degree of non-progressive cognitive impairment is present in some boys with DMD, but not all. In addition, the re- sults of studies have also shown that mutations in the distal parts of the dystrophin gene are more likely to be associated with cognitive impairment and that cu- mulative loss of dystrophin isoforms expressed in the central nervous system increases the risk of cognitive deficits (13,17). The described average IQ in boys with DMD is 85 or 1 standard deviation below the popula- tion average (3). Girls who carry mutations in the DMD gene are usually asymptomatic, but it is known that they may develop symptoms of skeletal and heart muscle dam- age; these patients are defined as ‘symptomatic carriers’ (24). The authors of some studies report that the prev- alence of skeletal muscle damage in carriers is 3–46%, the prevalence of elevated serum CK values is 53–100%, and dilated cardiomyopathy is 8–18%. Dilated cardio- myopathy, in particular, is a serious problem, as its in- cidence increases with age even in carriers with normal electrocardiogram (ECG) and without symptoms asso- ciated with skeletal muscle damage (25). Cardiac dys- trophinopathy is also a risk factor for myocarditis. Be- cause of all this, female carriers need proper treatment and regular monitoring by a cardiologist (8,21,25,26). 4 Diagnostic tests DMD should be considered by any physician who comes in contact with a boy whose parents say he often falls, that he has weak muscles, and who finds a positive Gower’s sign in the boy’s clinical status. These problems can also be accompanied by delayed speech develop- ment. The disease should also be considered in any boy with global developmental delay or any girl with unex- plained impaired heart function. Serum analysis of blood shows elevated levels of muscle enzymes which are released into the blood- stream – CK and transaminases (mainly alanine ami- notransferase – ALT and aspartate aminotransferase – AST). Based on the previouosly described clinical picture and laboratory results, the decision to perform genetic tests for DMD confirmation is made. Given that deletions and duplications in one or more exons are present in most patients (up to 70%), the multiplex li- gation-dependent probe amplification (MLPA) method is the genetic test of choice. While MLPA detects dele- tions and duplications in exons, it does not detect mu- tations in introns. For this purpose, microarray-based comparative genomic hybridization, aCGH, is used as a second test. In addition to these two, single-condition amplification/internal primer and multiplex amplifi- able probe hybridization are used to diagnose DMD. If by using one or more of these methods a mutation in dystrophin is identified, no further testing is required. However, if the deletion or duplication cannot be de- termined by these methods, it is necessary to perform sequencing of the dystrophin gene, which means carry- ing out a targeted search for point mutations or small deletions or insertions. Muscle biopsy is not required in case of a positive genetic result for DMD. However, if genetic testing is negative while CK levels are elevated and DMD-con- sistent clinical symptoms and signs are present, muscle biopsy should be performed to quantify the dystro- phin expressed in the sample. The latter should also be performed in case of suspicion of DMD and a posi- tive family history of a still unknown family mutation. Nowadays, electrophysiological examinations (electro- myography, nerve conduction studies) are rarely re- quired in the process of diagnosing DMD. They are not mandatory in diagnosing DMD (8,25). The comprehensive patient evaluation also includes genetic counselling of family members of a patient with DMD in order to identify who is a potential car- rier of the mutation in the dystrophin gene. Such test- ing is recommended to female relatives of a boy with a confirmed diagnosis of DMD. If the test confirms that the woman is a carrier, a prenatal genetic diagnosis is available to her (7,18). At the Department of Child, Adolescent and Developmental Neurology of the Uni- versity Children’s Hospital, University Medical Centre (UMCL) Ljubljana, all female relatives of an ill boy are advised to undergo genetic counselling with a clinical geneticist; further genetic diagnostics is in the domain of the clinical geneticist. 537 PROFESSIONAL ARTICLE Duchenne muscular dystrophy – novelties in diagnostics and treatment 5 Treatment In January 2018, The Lancet Neurology published new recommendations for the holistic treatment of patients with DMD (8,21,28). They emphasize the importance of early detection of the disease, earlier identification and treatment of possible and expected complications, all with the aim of prolonging the life expectancy as well as improving the quality of life of pa- tients with DMD. 5.1 Corticosteroids The exact cellular mechanism of the positive effects of corticosteroids (CS) in the treatment of DMD has not been fully elucidated. By working on mouse models with muscular dystrophy and in patient studies, researchers found that the positive effects of CS may be due to the following possibilities of their action: 1. altering the mRNA levels of structural, signalling, and immune re- sponse genes; 2. reducing the number of cytotoxic T lymphocytes; 3. lowering calcium influx and concentra- tion in the cytosol; 4. increasing laminin expression and muscle regeneration; 5. inhibiting muscle apoptosis and cellular infiltration; 6. increasing dystrophin expression; 7. affecting neuromuscular transmission; 8. protecting against mechanical damage to the fibres; 9. slowing down muscle fibre necrosis; 10. slowing down the rate of skeletal muscle breakdown; 11. increasing the level of taurine and creatine in the muscles. It was conclud- ed that additional studies are needed to identify cellular mechanisms of action that contribute to the positive ef- fects of CS in the treatment of DMD (29). One of the goals of CS treatment is to prolong the time when the patient walks independently or has the preserved ability to move. Using CS extends the ability to move independently without the use of a wheelchair for roughly three years (30). Randomized controlled studies have shown that CS significantly improve mus- cle strength and movement ability in boys with DMD (8,31), maintain respiratory and cardiac function, upper limb function, and reduce the need for scoliosis surgery, at least in the short term. Therefore, this treatment is al- so included in the latest international recommendations for the treatment of children with DMD (8,10,32,33). The authors of the current recommendations of CS treatment initiation believe that CS should be intro- duced when the child’s motor skill development stops at the latest, but prior to their loss, which is between the ages 5 and 15. Prednisone (at a final dose of 0.75 mg/ kg BW/day) or deflazacort (at a final dose of 0.9 mg/kg BW/day) are recommended for treatment. If the child does not tolerate the administered dose or more severe side effects occur, it is advisable to reduce the dose by 25–33% and re-evaluate the treatment after one month. If the functional movement ability is reduced, it is ad- visable to re-increase the dose of CS to the target dose according to the weight and based on the initial dose and to re-evaluate after 2 to 3 months. The introduc- tion of CS is also recommended for patients who are no longer able to move independently. We also follow these recommendations at the Department of Child, Adolescent and Developmental Neurology of the Uni- versity Children’s Hospital, University Medical Centre (UMCL) Ljubljana. Despite the instructions given, the preferred CS, the optimal dose and the dosing regimen are not known. In some studies, daily doses of prednisone/prednisolone or deflazacort have been shown to be more effective than intermittent dosing regimens regarding the effect on preserving the ability to move, but such doses have been associated with an increased risk of side effects (34,35). On the other hand, studies have also been published in which prednisone administered once weekly has been shown to be as effective as daily prednisone intake. Phase 3 of double-blind, randomized controlled trials compared deflazacort 0.9 mg/kg BW/day, deflazacort 1–2 mg/kg BW/day, prednisone 0.75 mg/kg BW/day, and placebo. In all groups treated, improved muscle strength was observed compared to the placebo group. They also found that deflazacort was associated with lower weight gain compared to prednisone. However, a study comparing the safety and efficacy of deflazacort with prednisone is ongoing (8,36-38). We believe that additional studies are needed to fur- ther define the effect of CS on prolonging the time when a child is still able to move independently and other positive effects on the health of children with DMD, the most appropriate time to start treatment, type of CS and dosing regimen, but also studies that, in addition to the already known side effects of CS (the most common being overweight, short stature, behavioural chang- es, Cushingoid appearance and excessive hair growth) (36,38), would exactly specify the long-term side effects of their use in patients with DMD. The desire for a drug that would be as effective or more effective than CS and at the same time have fewer side effects, has lead to the development of vamorolone. Vamorolone is a new (first in its class) anti-inflamma- tory synthetic steroid drug that appears to optimize the classic anti-inflammatory effects of CS. Studies on the efficacy and safety of vamorolone in animal models with 538 PEDIATRICS Zdrav Vestn | september – october 2021 | Volume 90 | https://doi.org/10.6016/ZdravVestn.3172 chronic inflammatory conditions, including mouse models with DMD, have shown, when comparing vam- orolone with prednisolone, that the anti-inflammatory effects of prednisolone are maintained and that there are no side effects. The maintenance of anti-inflammatory effects and loss of side effects in preclinical models is consistent with the mechanism of action of vamorolone; namely, it blocks the nuclear factor κB (NF–κB) associ- ated proinflammatory signals as a ligand/receptor in the monomeric state, instead of in the classical molecular model of ligand/receptor dimer complexes (39,40). Conklin et al. (39) conducted a multicentre, open-la- bel, two-week phase IIa study involving 48 mobile boys with DMD aged 4 to <7 years who had never received CS. The boys were divided into four groups of 12 chil- dren to study the effectiveness and safety of different doses of vamorolone; in the first group, children re- ceived vamorolone at a daily dose of 0.25 mg/kg BW/ day, in the second at a dose of 0.75 mg/kg BW/day, in the third at 2.0 mg/kg BW/day and in the last group at a dose of 6.0 mg/kg BW/day. The results showed that vamorolone was safe and well tolerated by patients even at the highest daily dose studied (6.0 mg/kg BW/day). The pharmacokinetics of vamorolone were also found to be similar to the pharmacokinetics of prednisolone. The study also showed better safety of vamorolone compared to glucocorticoids, as the results showed a decrease in insulin resistance, positive effects in bone turnover and a decrease in adrenal inhibition. In addi- tion to the above, the anti-inflammatory mechanism of action and a positive effect on the stability of the plasma membrane were also noted; all of this was demonstrated by a decrease in serum CK, dependent on the dose of vamorolone. The positive effects of vamorolone for the treatment of DMD were also found in a recently published study by Hoffman et al. (40). After 24 weeks of vamorolone treatment, improved muscle function (estimated by the time to rise) and an increase in osteocalcin (a biological marker for bone formation) were observed. In addition, lower values of adrenal inhibition and insulin resistance biomarkers were found when comparing the results with previously published studies in patients with DMD treated with glucocorticoids. To optimize the treatment of DMD, various authors have studied the safety and efficacy of many drugs of different groups (anticholinesterase drug galantam- ine; antioxidants acting on biochemical and metabol- ic pathways, such as allopurinol, creatine, glutamine, idebenone; drugs that affect growth, height and muscle function – mazindol, growth hormone; drugs that cause changes in sarcolemma and calcium accumulation – ve- rapamil; drugs that affect blood flow in the muscles, an- ti-inflammatory drugs – azathioprine, cyclosporine, CS ...). For the time being, CS (prednisone/prednisolone/ deflazacort) are still considered a credible treatment that can change the natural course of the disease. Coen- zyme Q10 and creatine had a positive effect on certain muscle functions in individual studies without side ef- fects, whereas based on the results of a 52-week study of 31 patients with DMD, idebenone (a synthetic ana- logue of coenzyme Q10) was found to supposed to have a long-term positive effect on pulmonary function (41). 5.2 Treatment of cardiovascular problems Cardiovascular complications are the leading cause of disease-related morbidity and mortality in patients with DMD. Dystrophin deficiency in the heart muscle is manifested by cardiomyopathy. As the disease progress- es, heart function deteriorates, so clinical signs of heart failure develop; besides aforementioned there is also a risk of developing life-threatening arrhythmias (21). As already mentioned, according to a study pub- lished by Nigra et al., dilated cardiomyopathy is present in all patients with DMD after the age of 18 (19). How- ever, the age at onset of dilated cardiomyopathy and its degree vary widely and do not depend on an individ- ual mutation in the dystrophin gene (42). In addition, it is important to realize that the signs and symptoms in patients who have a greatly reduced ability to move (and do not walk) are often subtle (21). Because of all of the above, boys with DMD need regular monitoring by a cardiologist for the early detection of left ventric- ular (LV) ejection fraction decline (42). Recent recom- mendations for the treatment of children with DMD, published by Birnkrant et al. (21), also state that a car- diologist should be included in the multidisciplinary treatment of these children at an early stage, if possible with additional knowledge of the treatment of cardio- myopathy and heart failure associated with NMD. When DMD is diagnosed, patients who are still able to walk and patients who have recently been able to do so are advised to have a regular, yearly examination (up to the age of 10) by a cardiologist, including an ECG and cardiac imaging. The method of choice for follow up is magnetic resonance imaging (MRI) of the heart, which, due to the need for anaesthesia, is generally performed from the age of 6 to 7 on; until this age, regular year- ly heart ultrasound examination (US) is recommend- ed. After the age of 10, however, due to the increased risk of LV dysfunction, an annual cardiac assessment 539 PROFESSIONAL ARTICLE Duchenne muscular dystrophy – novelties in diagnostics and treatment is advised. However, if symptoms and/or signs of heart failure occur during examination or upon imaging (e.g. LV enlargement, LV dysfunction), the cardiologist will assess whether more frequent monitoring is required. The biggest change in recent recommendations is that angiotensin-converting enzyme inhibitors (ACEI) may be introduced into treatment in asymptomatic boys less than 10 years of age, with no signs of abnormality in cardiac MRI or cardiac US. ACEI selection as well as dosing are in the domain of the cardiologist (21). Stud- ies conducted by Duboc et al. (2005 and 2007), which examined the effect of the ACEI perindopril on the on- set and progression of LV dysfunction in patients with DMD, have shown that early introduction of ACEI was associated with later onset of LV dysfunction and low- er mortality (43-45). However, pharmacological treat- ment, regardless of age, should be initiated in the case of abnormal US or MRI of the heart or in case of clinical symptoms of heart failure in accordance with the gener- al guidelines for the treatment of heart failure. In addi- tion to ACEI or angiotensin receptor blockers (ARBs), additional beta-blocker and mineralocorticoid receptor antagonist therapy is initiated after an observed deterio- ration of LV function. With further deterioration of LV function, anticongestive treatment is intensified further (21). In patients in the late stage of the disease, i.e. long after they have lost the ability to walk, in light of the ex- pected progression of the deterioration of cardiac func- tion or escalation of heart failure and for the purpose of early optimal treatment, more frequent treatment by a cardiologist is recommended within the time frame at their discretion. Symptomatic heart failure is diffi- cult to diagnose in patients who have lost independent movement, as the initial symptoms of heart failure are identified on the basis of physical performance (NYHA heart failure classification). In these patients, only signs of already advanced heart failure are visible, such as inability to perform daily activities, weight loss, vom- iting, abdominal pain, and sleep disturbances. It is al- so extremely important to provide optimal respiratory support, as abnormal pulmonary function affects heart function. In addition to heart failure, patients with DMD with severely impaired LV function are also at risk of throm- boembolism and cardiac arrhythmias. Most common rhythm disorders include atrial fibrillation, ventricu- lar tachycardia, and ventricular fibrillation. In patients with impaired LV function or myocardial fibrosis, a 24-hour Holter ECG is recommended once a year. Among patients with episodic, non-sustained rhythm disturbances, the method of choice are event monitors/ event recorders. The introduction of antithrombotic agents, antiarrhythmics or the insertion of specific car- dioverter defibrillators is in the domain of the cardiol- ogist (21). 5.3 Treatment of respiratory problems As DMD progresses, the strength of the muscles of the chest, the diaphragm (the most important respirato- ry muscles) and the abdominal wall gradually deterio- rate, especially after the loss of independent movement. Inactivity, incorrect posture, obesity and changes in the shape of the chest further contribute to deepening respi- ratory failure and impaired coughing efficiency and lead to respiratory complications: ineffective shallow breath- ing patterns, uneven distribution of ventilation within the lungs with the formation of atelectasis, severe pneu- monia, and chronic respiratory failure. Without effective respiratory support, disease progression leads to chron- ic respiratory distress, long-term and severe respiratory infections, and exposes the patient to a high risk of poor disease outcome with respiratory failure or deepening cardiomyopathy. If left untreated, patients are at high risk of developing severe dyspnoea, the need for long- term hospitalizations due to atelectasis or pneumonia, and the risk of death as a result of respiratory failure or cardiac arrhythmias due to respiratory problems (7,21). The latest guidelines specifically emphasize the im- portance of early treatment, which aims to maintain long-term pulmonary function and to “delay” the de- cline in pulmonary function. The decline in pulmonary function initially shows no symptoms, so patients do not detect it in time. Therefore, at the stage when patients are still able to move independently, regular monitoring of pulmonary function by spirometry is recommended once a year – especially FVC. In addition, nocturnal polygraphy/polysomnogra- phy (PSG) is recommended in patients who move inde- pendently and have symptoms and/or signs of sleep-dis- ordered breathing, and in those who experience weight gain during CS therapy with the aim to objectively as- sess sleep-disordered breathing. Leading NMD centres perform nocturnal polygra- phy/PSG routinely once a year in patients who no longer walk independently (independent of FVC), as well as in all patients who still walk and have a FVC <50%. The presence of clinical signs of sleep-disordered breathing or frequent/severe respiratory infections are a warning sign of the need for additional targeted analysis of sleep breathing (46). 540 PEDIATRICS Zdrav Vestn | september – october 2021 | Volume 90 | https://doi.org/10.6016/ZdravVestn.3172 In addition to causing increased daily fatigue and headaches, obstructive sleep apnoea, which often co-oc- curs with snoring, also increases the risk of cardiovas- cular complications, namely hypertension, stroke, and heart failure (47). Apart from snoring, these are late signs of nocturnal hypoventilation, which we want to avoid with a proactive approach. Sleep studies can also be used as an alternative meth- od to monitor respiratory status in those patients who are unable to participate in routine pulmonary function tests. As already mentioned, more respiratory complica- tions are expected after the loss of the ability to move in- dependently. Therefore, monitoring of pulmonary func- tion every 6 months is advised at this stage of the disease. In addition to FVC, monitoring of maximal inspiratory and expiratory pressure, cough strength, blood oxygen saturation by pulse oximetry (SpO2) is recommended; transcutaneous measurement of the partial pressure of carbon dioxide in the blood (PtcCO2) or partial pressure of carbon dioxide in expired air (PetCO2) is also recom- mended every six months or whenever room air SpO2 is ≤ 95%. Based on the results of these examinations, the pa- tient is advised to use respiratory physiotherapy devic- es in accordance with their needs and abilities (e.g. vi- brating/oscillating device – a device that by causing the chest wall to vibrate and by using appropriate manual technique allows easier clearing of the lower respiratory tract; aspirator; expectorant…). If the criteria are met, non-invasive ventilation during sleep is introduced and, where necessary, also during the day (mouth-breathing support) and, in the case of markedly advanced disease, consideration should be given to the appropriateness of invasive respiratory support. Progressive scoliosis often worsens pulmonary func- tion. These patients should undergo surgical correction of scoliosis. A characteristic type of scoliosis in patients with DMD is a long paralytic C-curve involving the tho- racic and lumbar spine with pelvic tilt. It is usually nec- essary to correct scoliosis and fixate the spine, extending from the 4th thoracic to the 1st sacral vertebra by also fixing it to the pelvis. Regular follow-up of the patient and the provision of this respiratory support reduces the number of respirato- ry complications and improves the quality of life as well as prolonging survival. Adequate respiratory support is provided by a team of pulmonologists and respiratory physiotherapists/nurses with specialized skills who pro- vide parents with appropriate education to provide opti- mal respiratory support at home. Regular vaccination against infectious diseases is rec- ommended for all patients, regardless of the stage of the disease. However, those who receive a high dose of CS (more than 20 mg or more than 2 mg/kg body weight prednisolone per day) should not be vaccinated with live vaccines. If a child with DMD has not had chickenpox, it is advisable to vaccinate them with two doses of live, attenuated vaccine at 6-week intervals before initiating CS treatment, but if this is no longer feasible, it is recom- mended that those family members who have not had chickenpox be vaccinated. Patients with DMD should be vaccinated against influenza every autumn, and pneu- mococcal vaccination is also recommended. The best protection against pneumococcal infections is achieved by using the conjugate vaccine in the required number of doses for the age, followed by another dose of the polysaccharide vaccine after two months. Since 2015, the pneumococcal conjugate vaccine is available to all children in Slovenia, in patients with DMD aged 5 years and older who are already on a high dose of CS. Due to immune deficiency, the costs of the pneumococcal con- jugate vaccine are covered by the Health Insurance In- stitute of Slovenia, otherwise patients over 5 years of age who do not receive CS treatment have to pay the costs of the conjugate vaccine themselves, but the costs of the polysaccharide vaccine are borne by the Health Insur- ance Institute of Slovenia (21,48). 5.4 Treatment of endocrinological and metabolic co-morbidity Impaired muscle strength due to the progression of the underlying disease and treatment with CS are two major risk factors for decreased bone density and in- creased risk of bone fracture. A fracture can occur even after a minor fall. The most common are fractures of the bones of the lower limbs and vertebrae. In addition to clinically detectable fractures, asymptomatic fractures, especially of the vertebrae, occur in boys with DMD. Untreated fractures cause chronic back pain and spi- nal deformities, and fractures of limb bones can lead to premature loss of mobility. In recent guidelines for the treatment of boys with DMD, Birnkrant et al. (21) therefore emphasize the importance of early detection, identification and treatment of individuals at increased risk of bone fractures. The detection is aided by bone mineral density measurements with double X-ray ab- sorptiometry (densitometry), and even more import- ant is the routine X-ray imaging of the spine in later- al projection in individuals at increased risk due to CS treatment or physical inactivity. Thus, in addition to 541 PROFESSIONAL ARTICLE Duchenne muscular dystrophy – novelties in diagnostics and treatment periodic densitometry, periodic X-ray imaging of the thoracic-sacral part of the spine is also advised; in boys treated with CS every 1–2 years, and in others every 2–3 years. In addition to imaging tests, blood tests are also used to assess bone health: calcium, phosphate, alkaline phosphatase, parathyroid hormone, vitamin D and bone turnover parameters are determined. An endocrinolo- gist plays a major role in planning primary and second- ary prevention and treating low bone density. The basis of prevention and treatment is adequate calcium intake, maintenance of normal vitamin D levels and reduction of risk factors for reduced bone density (CS, physical in- activity). Mild asymptomatic fractures should be closely monitored. In moderate and severe asymptomatic frac- tures and in all fractures with symptoms in the thora- columbar spine, Birnkrant et al. recommend treatment with intravenous bisphosphonates (21). As part of endocrinological treatment, it is also im- portant to monitor physical growth, pubertal develop- ment and possible adrenal dysfunction. In boys with DMD treated with CS, decreased growth rate is quite common. As part of monitoring, it is therefore neces- sary to determine body height at each examination (at least every 6 months), and in the case of immobile pa- tients, other auxological measurements can be used to determine growth, such as measurements of the length of individual long bones. In case of short stature and/or slower growth, endocrinological treatment is required as in other children. Recombinant human growth hormone therapy has also been mentioned. Delayed or incomplete puberty as part of the treatment with CS or with poor nutrition is consistent with secondary-tertiary hypo- gonadism. With the exclusion of the causes of hypogo- nadism, testosterone treatment prescribed and dosed by an endocrinologist is warranted. An important goal of the testosterone treatment is to maintain bone health or treat decreased bone density as a consequence of hypo- gonadism. Iatrogenic adrenal insufficiency has also been described during treatment with CS. Patients receiving CS should be aware of the possibility of acute adrenal in- sufficiency, which is a life-threatening condition. In peo- ple with DMD, especially those receiving high doses of CS, with symptoms or clinical signs consistent with the Addisonian crisis, immediate hydrocortisone treatment is urgently needed. A holistic approach to the treatment of DMD also includes the identification of CS and reduced physical activity-related metabolic complications such as obesi- ty, so-called prediabetes and various components of the metabolic syndrome. 5.5 Treatment of gastrointestinal problems and ensuring adequate nutritional status Constipation is one of the most common gastrointes- tinal (GIT) problems in patients with DMD, as they usu- ally have several risk factors present (inability to move, weakness of the abdominal muscles, prolonged passage of stool through the colon, dehydration). To prevent the development of constipation, it is necessary to take care of a balanced diet with sufficient intake of ballast substances and adequate fluid intake, and if this is not enough, regular/daily use of oral osmotic stool softeners and laxative suppositories, which accelerate intestinal peristalsis, is recommended. Gastroesophageal reflux disease (GERD) is also a common GIT problem, mainly due to impaired oesoph- ageal motility, delayed gastric emptying (gastroparesis) and the presence of scoliosis, as well as treatment with CS. GERD is primarily treated with drugs, namely pro- ton pump inhibitors. However, it should be taken into account that long-term treatment with proton pump in- hibitors is associated with an increased risk of reduced bone density, vitamin B12 deficiency, and infections such as pneumonia. It is also necessary to advise on proper nutrition, in terms of smaller and more frequent meals, and appropriate reduction of fat intake. Gastroparesis can lead to postprandial abdominal pain, nausea, vomiting, and loss of appetite. When gas- troparesis is confirmed by appropriate examinations, an attempt is made to treat it by changing the diet as in the GERD treatment, by prokinetic therapy and, in case of failure, also by inserting a gastro-jejunal feeding tube. Patients may also experience difficulty swallowing and contractures of the jaw muscles, especially in the later stages of the disease. These factors make feeding very difficult and affect the patient’s general condition (weight loss accelerates the gradual loss of respiratory muscle strength), so it is extremely important to inform patients and their parents/guardians in a timely fashion about the possibility of feeding via feeding tubes either through a nasogastric tube or especially through percu- taneous gastrostomy tube. In light of all the above, it should be emphasized that ensuring optimal body weight of patients with DMD is crucial. Namely, patients with DMD have an increased risk of being overweight in early life (mainly due to CS and loss of ability to move), but later, due to problems with feeding and swallowing and contractures of the jaw muscles, they are at risk of being underweight. Nutri- tional imbalance, as already mentioned, has a negative effect on skeletal muscle as well as on the respiratory 542 PEDIATRICS Zdrav Vestn | september – october 2021 | Volume 90 | https://doi.org/10.6016/ZdravVestn.3172 and cardiovascular systems. Therefore, it is important that the multidisciplinary team includes a dietitian who, by regularly monitoring the patient’s clinical condition, weight and height, advises the patient on a healthy, bal- anced diet, which must ensure optimal intake of calories, protein, fluids, as well as vitamins and electrolytes, espe- cially calcium and vitamin D (8,49). 5.6 Psychological aspects of treatment of children and adolescents with DMD Psychological treatment is one of the important as- pects of holistic treatment of children and adolescents with DMD (28), as living with this disease requires pa- tients and their relatives to adapt to different, disease-re- lated health and physical limitations. At the same time, the quality of life of patients is also affected by frequent associated neuro-developmental and psychopathologi- cal disorders. In children and adolescents with DMD, specific cog- nitive impairments and intellectual disabilities are more common. In tests of intellectual ability, they achieve on average about one standard deviation lower results compared to healthy peers. In a review study, Cotton et al. (50,51) found in 1,224 patients with DMD that as many as 35% of children with DMD achieved scores in the intellectual disability range (IQ ≤ 70), significantly more than in the general population of children (ap- proximately 2%). In addition, research over the last ten years has shown that such problems are more common in patients with a defect of the distal part of the DMD gene associated with the regulation of prenatal central nervous system development (52,53). In addition to differences in general intellectual abil- ities, various studies have also confirmed a higher in- cidence of deficits in specific cognitive abilities. These include problems with verbal comprehension and ex- pression, verbal memory and automation of thought processes, which increases the risk of specific learning difficulties in children with DMD, such as dyslexia, dy- scalculia and dysgraphia (54-57). Due to the described problems in the field of cog- nitive abilities and physical limitations resulting from the disease, children and adolescents with DMD often show poorer adaptive skills in the areas of communica- tion and social skills, practical everyday skills and motor skills (58). Increased symptoms of various psychopathological disorders have been reported in children and adolescents with DMD (59). In many cases, they are clinically signif- icant (56,60), most often in the form of anxiety (29.3%), depression (27.4%), obsessive compulsive disorder (4.8– 11%) and/or autism spectrum disorders (3.1–21%). In conjunction with the latter, but also possibly indepen- dent of it, boys with DMD may also have more difficul- ty processing sensory stimuli. Hyper- or hyposensitive reactions to auditory, visual, tactile, olfactory, oral, and vestibular stimuli have been reported (61). One of the most commonly associated disorders in DMD is atten- tion deficit hyperactivity disorder (ADHD) (9–45%). Hinton et al. (56) observed significant attention prob- lems in about a quarter of boys with DMD, accompa- nied by frequent behaviour regulation difficulties, low frustration tolerance, aggression, defiant behaviour, and mental rigidity. DMD, however, does not affect only patients, but as a complex and chronic progressive disease it is a great burden for the whole family. The burden that the family feels is reflected on a daily practical and mental level. At the daily practical level, the impact of the disease is re- flected in altered family relationships and the extent of social and leisure activities of family members, as well as in financial challenges. At the mental level, the effect of illness is recognizable in the emotional responses that family members experience due to illnesses such as feel- ings of loss, sadness, tension, feeling unable to cope with the situation (62). A bigger challenge for parents of chil- dren with DMD is also their upbringing (63). Compared to parents of healthy children, they report higher levels of stress (63,64) and feelings of guilt, sadness, and de- pression associated with the child’s illness (62,65). The severity of these problems and the effectiveness of par- ents in dealing with them depend on the level of pro- gression of the child’s illness, experiencing the illness as a burden, interpersonal support and the financial bur- den that the illness poses to the family. An important group that is often overlooked in the treatment of children and families with NMD are their healthy siblings. The illness of their brother or sister af- fects them as well. Research on their psychological ad- justment shows at least double the risk of them develop- ing emotional and behavioural problems than with their peers in the normative comparison group (66,67). Families of children with DMD, however, do not only report that coping with chronic NMD is difficult and burdensome, but often also emphasize the positive effects of the disease. Two thirds of parents of children with DMD and BMD report positive psychological ef- fects of their own parenting experience. They point out as particularly valuable the possibility of personal growth, the development of an enhanced sense of power in coping with difficulties, and cite the beneficial effect 543 PROFESSIONAL ARTICLE Duchenne muscular dystrophy – novelties in diagnostics and treatment of the disease on the interconnectedness of family mem- bers (62,67). Psychological assistance to patients with DMD and their families includes diagnostic procedures to identify associated neurodevelopmental and psychopathological disorders, as well as various psychotherapeutic measures to help cope with the disease. As members of the pro- fessional team, clinical psychologists use standardized psychodiagnostic tools to identify the presence of possi- ble intellectual disabilities and/or specific deficits and to assess the child’s adaptive skills. Based on the psycholog- ical assessment, they also offer support in integrating the child into the school and social environment, and advise schools and kindergartens on various adjustments, as- signing a companion and on the possibility of providing additional professional assistance if the child needs it. They provide psychological support and counselling to parents regarding family relationships, siblings and part- nerships, and help them integrate into various self-help groups. They provide psychological help and support in communicating with the child about their illness and strive to provide psychological support to healthy sib- lings. By providing appropriate psychological measures, they thus contribute to the empowerment of the fami- ly and the child with DMD and strive to strengthen the strong areas of all family members and the family as a whole. 5.7 Rehabilitation and supportive care programs The rehabilitation of children with DMD involves specialists in physical and rehabilitation medicine, physiotherapists, occupational therapists, speech thera- pists, special teachers, social workers, psychologists and specialists in orthotics and prosthetics at the Universi- ty Rehabilitation Institute of the Republic of Slovenia (URI-Soča). A key element in planning a rehabilitation programme is the assessment of functional status, espe- cially in the area of movement and daily activities. When assessing functional status, it is important that the as- sessment is performed by a therapist who has the appro- priate knowledge and experience and understands the relationship between test results and the clinical picture in a child with DMD (8). According to the recommendations of Birnkrant and his colleagues, mobility assessments should be per- formed every six months. They recommend regular as- sessments of muscle strength, the range of passive lower limb joint mobility, and walking tests (6-minute walking test, 10-metre walk, stand-up and walk test) (8). Among functional tests in Slovenia, regular clinical practice uses MFM, the motor function measure scale for neuromus- cular diseases (68). A child with DMD should regularly perform stretching exercises for the lower limb muscles 4 to 6 times a week. To maintain physical fitness, the au- thors of the guidelines recommend swimming, low-re- sistance exercise, and exercises to improve or maintain respiratory function. The physiotherapy programme is also important at a time when the child loses the abili- ty to move independently. They advise exercising to the extent that the child does not get too tired and that exer- cise does not cause pain (myoglobinuria in the 24-hour urine is a sign of excessive effort and additional muscle damage). To prevent contractures and prolong the abili- ty to move, the guidelines recommend the use of ortho- ses for the ankle and foot at night (to stretch the gastroc- nemius muscle), the use of orthoses for the knee, ankle and foot (stretching the muscles that bend the knees) and stand-up chairs (stretching all the muscles of lower limbs, when the child can no longer walk). Therapeutic bikes are available to strengthen the muscles of the lower limbs and overcome the medium distances. In the case of deformities of the lower limbs, orthopaedic surgical treatment is considered (8,21,28). An often-overlooked problem is pain. The authors of the guidelines have pointed out that little is known about this. Experts are encouraged to ask a child with DMD about possible pain. It is believed that pain is often the result of improper posture and inability to change position. In view of this, it is recommended that the child be provided with a suitable sitting chair, a seat for the wheelchair and an adaptive bed with a cushion for pressure ulcer prevention. In the event that scoliosis has already developed due to muscle weakness and asym- metrical posture, a well-adjusted and individually made chair or wheelchair is especially important. Scoliosis is measured on an X-ray with a Cobb measurement, which is still considered a useful standard clinical method. When determining the curvature, we first determine the upper and lower vertebrae that still belong to this plane. A line is then drawn along the upper border of the up- per vertebrae and the lower border of the lower vertebra and perpendiculars to this line. The sharp angle between these two lines is called the Cobb angle (69). With estab- lished scoliosis, regular monitoring, devices with a spi- nal orthosis and timely surgical treatment are important. The authors of the guidelines recommend that this be done in a child who is still growing and is not undergo- ing CS therapy, at a Cobb angle above 20°, or in a child with CS therapy at a Cobb angle above 40° (8,21,28). As the gross motor function decreases, difficulties 544 PEDIATRICS Zdrav Vestn | september – october 2021 | Volume 90 | https://doi.org/10.6016/ZdravVestn.3172 arise in performing daily activities. To make dressing and undressing easier, soft elastic clothing with simple fastening systems is recommended, as well as the use of a toilet chair and a shower chair. When contractures in the joints of the upper limbs begin to develop due to muscle weakness, it is appropriate to make an orthosis for the hands (most often for the wrist and palm with the fingers) to maintain a neutral position. In addition to testing and learning how to use these aids, occupation- al therapists at URI-Soča also monitor the child’s ability to perform fine movements and graphomotor skills. To help the latter, it is possible to use thicker pens with a softer filling, so that the child leaves a clear enough trace on the paper with less pressure. Later, the adolescent with DMD may also be offered the use of a computer to make notes of the subject matter more easily and to use handouts. In case of gross motor ability loss, we offer the child the use of a wheelchair. There are several types of wheel- chairs available (transport, manual, electric). Testing takes place at URI-Soča. It is important to decide on the model together with the child and parents, and at the same time provide the child with a good sitting posi- tion and an effective way to overcome longer distances. In the later stages of the disease, adolescents with DMD are usually equipped with a very complex electric wheel- chair, which has an adapted cushion, backrest with the possibility of tilting back, a seat with a cushion for skin protection, footrests that can change the tilt, wheelchair control lever with switches very sensitive to touch, and brackets for possible respiratory assist devices. Speech therapists usually meet a child with DMD ear- ly in case they have difficulty developing speech, but al- ways with a young person late in the course of the disease when they are no longer able to effectively produce voice and form words. The programme includes articulation learning, exercises to improve exhalation and volume, alternative strategies to improve speech, and the intro- duction of an alternative communication programme. At this time, it is important to talk to the adolescent with DMD in a calm environment, to take into account their state of fatigue, to repeat the question several times if necessary, and to give them enough time to answer the question. If a young person needs an alternative com- munication device for communication, they are referred to URI-Soča. There, the child rehabilitation team selects the appropriate device, prescribes it and then teaches the adolescent and their parents how to use it. All these devices, which significantly improve the ability to move, slow down the development of joint contractures and scoliosis, improve independence in performing daily activities, participation in activities and quality of life, are available to children and adoles- cents free of charge (up to the price standard); the costs are covered by the Health Insurance Institute of Slove- nia. The prescribing procedure is precisely determined by the Rules on Compulsory Health Insurance (70). 5.8 New drugs and drugs that change the course of the disease In recent years, several specific drugs have been de- veloped that alter the natural course of the disease and whose action is aimed at reducing muscle damage or reducing/eliminating the effect of the genetic cause of the disease. The first such drug to treat DMD is ataluren. It has been registered in Slovenia since August 2017 for pa- tients who are able to move independently, nowadays for those older than two years (initially only for those older than 5 years) who have a nonsense mutation lead- ing to the formation of a premature stop codon in the messenger RNA. Ataluren acts on the so-called princi- ple of the read-through mechanism. With its action, it strengthens the connection between the ribosome and the mRNA and enables the read-through of a prema- ture stop codon instead of termination of transcription, thus enabling the formation of functional dystrophin (71,72). It is estimated that 10–15% of patients with DMD/BMD have a nonsense mutation, meaning that they are candidates for treatment with ataluren (73). The effectiveness of ataluren has been studied in sev- eral studies. Preclinical studies have shown encourag- ing results, as more dystrophin is formed in the muscle cells of patients treated with ataluren (74). In the second phase of the study, which included 26 boys with DMD with a nonsense mutation, they found that, both in vi- tro and in vivo, dystrophin expression increased and that serum levels of muscle enzymes decreased after 28 days of treatment; however, only minimal chang- es were observed in muscle strength and timed tests during ataluren treatment (75). Bushby et al. performed a double-blind, randomized, placebo-controlled multi- centre study of the efficacy of ataluren in 173 patients with DMD aged 5 to 20. After 48 weeks of treatment, a 6-minute walk test (6MWT) showed a slower reduction in walking distance (up to 13 m) in the group of pa- tients receiving 40 mg/kg BW/day of ataluren (but not in those receiving ataluren at a dose of 80 mg/kg BW/ day) compared with placebo (up to 44m) (76). However, the results of a study conducted by McDonald et al. did not show a statistically significant difference in 6MWT 545 PROFESSIONAL ARTICLE Duchenne muscular dystrophy – novelties in diagnostics and treatment compared between the group of treated and untreated subjects (77). In addition to ataluren, eteplirsen and golodirsen, which are antisense oligonucleotides, are also used to treat DMD. They work by recognizing exon 51 or ex- on 53 on the dystrophin gene, respectively, and affect the transcription mechanism so that exon 51 or 53 is skipped. This preserves the reading frame and produces a shorter but functional dystrophin protein (78,79). Eteplirsen was approved by the US Federal Food and Drug Administration (FDA) under the fast-track procedure in September 2016, while golodirsen was ap- proved by the FDA under the fast-track procedure in December 2019. Most current data on the efficacy of in- travenous eteplirsen are the result of four clinical stud- ies (NCT00844597, NCT01396239/NCT01540409 and NCT02255552). In the NCT00844597 study, treatment with eteplirs- en improved dystrophin expression in 7 of 19 patients. NCT01396239/NCT01540409 is an extended clinical study carried out to address the preceding studies insuf- ficiency– the duration of the NCT00844597 study. The results of these studies, which showed a statistically sig- nificant increase in dystrophin-positive fibres (demon- strated by the immunohistochemical method) in pa- tients treated with eteplirsen, contributed to the FDA’s accelerated approval of eteplirsen (78). Data collection from a post-marketing efficacy study of eteplirsen for use in DMD patients with an appropriate mutation was completed on 14 June 2019, the results of which are not yet known (80). The FDA decided to accelerate the approval of golo- dirsen based on the positive results of clinical studies (phase 1/2) conducted in Europe that evaluated the safe- ty, tolerability, pharmacokinetics and efficacy (dystro- phin expression) of golodirsen in 25 boys (6–15 years) with DMD with confirmed deletions in the dystrophin gene corresponding to golodirsen treatment. The first part was a 12-week, double-blind, placebo-controlled phase one study in which patients were randomized to receive intravenous golodirsen at 4 incremental doses (4 mg/kg BW/week to 30 mg/kg BW/week for 2 weeks for each dose) and into a comparable group receiving placebo once a week. The second part was a 168-week, open-label phase 2 study evaluating the long-term effi- cacy and safety of intravenous administration of golo- dirsen 30 mg/kg BW/1x/week in 12 patients who par- ticipated in the first part and in 13 untreated patients. The results of the interim analysis at 48 weeks of the sec- ond part showed that the average dystrophin level (as- sessed in muscle biopsy using the Western blot test) was statistically significantly increased compared to baseline (p <0.001); the mean and median change from baseline was + 0.92% and + 0.88% of normal levels. Based on the underlying error in DMD (dystrophin deficiency), it can thus be predicted that this increase in dystrophin synthesis will result in a slower disease progression. A post-marketing confirmatory study (ESSENCE) is cur- rently underway; in the latter, patients are currently still being recruited; it is expected to be completed by 2024 (78,81). These medications have not yet been approved by the European Medicines Agency, so eteplirsen and golo- dirsen are not available in Slovenia. In Slovenia, we are trying to obtain eteplirsen for patients who are suitable candidates, but under the compassionate use protocol. Other drugs still in clinical trials include myosta- tin-targeting drugs, newer CS, anti-inflammatory mol- ecules and antioxidants, substances which reduce fi- brosis, drugs that improve vasodilation, mitochondrial function, and utrophin modulator drugs. Under devel- opment are the CRISPR/Cas9 technique, which alters genomic DNA, cell therapy with myoblast and stem cell transplantation, and also gene therapy that would, with the help of the so-called adeno-associated virus (AAV), transmit mini/microdystrophins to cells. Studies on the efficacy and safety of these medicines will be needed be- fore they actually enter the market (8,82-84). 6 Protocol for the treatment of children with DMD at the University Children’s Hospital Ljubljana Duchenne muscular dystrophy with its low inci- dence is considered a rare disease. Therefore, it is bene- ficial for patients to be managed in centres where a larg- er number of patients accumulate and where they are treated by specialized experts in various fields. Patients with DMD (as well as patients with other rare diseases) require a multidisciplinary approach (8). The Department of Child, Adolescent and Develop- mental Neurology of the University Children’s Hospi- tal, University Medical Centre (UMCL) Ljubljana is the only tertiary centre for the treatment of children with DMD in Slovenia. To establish an optimal team and uniform treatment of patients and to reduce the number of visits to various wards and clinics for children and the burden that this posed for the children as well as their families, we reorganized the treatment of children with DMD and other NMD in 2017, so that the DMD patient plays a central role. All activities have been adjusted so that the treatment is optimal for the patient. 546 PEDIATRICS Zdrav Vestn | september – october 2021 | Volume 90 | https://doi.org/10.6016/ZdravVestn.3172 References 1. Moat SJ, Bradley DM, Salmon R, Clarke A, Hartley L. Newborn bloodspot screening for Duchenne muscular dystrophy: 21 years experience in Wales (UK). Eur J Hum Genet. 2013;21(10):1049-53. DOI: 10.1038/ ejhg.2012.301 PMID: 23340516 2. Goemans N, Kirschner J, Mercuri E. New Perspectives in the Management of Duchenne Muscular Dystrophy. Eur Neurol Rev. 2014;9(1):78. DOI: 10.17925/ENR.2014.09.01.78 3. Yiu EM, Kornberg AJ. Duchenne muscular dystrophy. J Paediatr Child Health. 2015;51(8):759-64. DOI: 10.1111/jpc.12868 PMID: 25752877 4. Crispi V, Matsakas A. Duchenne muscular dystrophy: genome editing gives new hope for treatment. Postgrad Med J. 2018;94(1111):296-304. DOI: 10.1136/postgradmedj-2017-135377 PMID: 29386334 5. Rando TA. The dystrophin-glycoprotein complex, cellular signaling, and the regulation of cell survival in the muscular dystrophies. Muscle Nerve. 2001;24(12):1575-94. DOI: 10.1002/mus.1192 PMID: 11745966 6. Crispi V, Matsakas A. Duchenne muscular dystrophy: genome editing gives new hope for treatment. Postgrad Med J. 2018;94(1111):296-304. DOI: 10.1136/postgradmedj-2017-135377 PMID: 29386334 7. Annexstad EJ, Lund-Petersen I, Rasmussen M. Duchenne muscular dystrophy. Tidsskr Nor Laegeforen. 2014;134(14):1361-4. DOI: 10.4045/ tidsskr.13.0836 PMID: 25096430 A team for NMD was formed, linking experts of various specialties. The core team includes paediatric neurologists, paediatric endocrinologists, paediatric pulmonologists, paediatric gastroenterologists, clinical psychologists, physiotherapists and occupational thera- pists, dieticians and other staff with specific knowledge in the field of NMD; and last but not least, the nurses and technicians who make the first contact with the patient on the ward and are the link between the other team members. Depending on the individual needs of patients, specialists in physical and rehabilitation med- icine, ophthalmologists, orthopaedists, surgeons, oto- rhinolaryngologists, geneticists, speech therapists, social workers and other professionals are also included. With the help of a coordinator, all patients with DMD (as well as those with other forms of NMD) are invited anually for a two day inpatient stay, during which pa- tients undergo assessment by all sub-specialists that they require. Children are admitted to the Department of Child, Adolescent and Developmental Neurology or to the Unit of Pulmonary Diseases (children with non-in- vasive or invasive respiratory support), and specialists from various subspecialties perform examinations/tests in the department and plan further follow ups. If more frequent assessments are required, the coordinator tries to combine these examinations. As part of the treat- ment, attention is also paid to psychological support for patients and their families. A social worker also offers assistance in managing the rights and adjustments re- lated to schooling and the social status of children and families. In April 2020, we manage 29 boys with DMD/BMD and one girl, who is a symptomatic carrier. In accor- dance with the recommendations, in the case of the appropriate clinical picture or at the first signs of mo- bility decline, patients and their parents are advised to introduce CS therapy and all other forms of treatment. Treatment with ataluren is also available in Slovenia and fully covered by the Health Insurance Institute of Slove- nia. Thus, this therapy has already been introduced to three patients who have the appropriate mutation and meet other necessary criteria. We believe that such multidisciplinary treatment con- tributes to reducing the burden on patients, contributes to the establishment of good cooperation and patient trust, reduces the financial burden for patients, provides the best long-term outcome and, last but not least, en- sures uniform treatment in accordance with state-of- the-art guidelines. Therefore, we advise that all children whose doctor suspects that they may have DMD or any other NMD be referred for treatment to the University Children’s Hospital, which can be arranged by doctors by telephone, No. 01 522 5189. 7 Conclusion DMD is a serious muscle disease that affects boys and their families and for which we still do not have an effec- tive treatment to eliminate the basic, i.e. genetic cause of the disease. In recent years, there has been significant progress in understanding the natural course of the dis- ease. With this knowledge, with the earlier diagnosis of the disease and with multidisciplinary treatment, with the use of CS, and in cases of appropriate mutations with the use of newer drugs, physiotherapy and supportive care, the duration and quality of life of patients with DMD have improved. There are several drugs in devel- opment, whose aim is reducing muscle damage, as well as drugs that would eliminate the basic, i.e. genetic cause of the disease. They raise hopes that in the future the dis- ease will only be a thing of the past. Conflict of interest None declared. 547 PROFESSIONAL ARTICLE Duchenne muscular dystrophy – novelties in diagnostics and treatment 8. 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 Neurol. 2018;17(3):251-67. DOI: 10.1016/S1474-4422(18)30024-3 PMID: 29395989 9. Aartsma-Rus A, Van Deutekom JC, Fokkema IF, Van Ommen GJ, Den Dunnen JT. Entries in the Leiden Duchenne muscular dystrophy mutation database: an overview of mutation types and paradoxical cases that confirm the reading-frame rule. Muscle Nerve. 2006;34(2):135-44. DOI: 10.1002/mus.20586 PMID: 16770791 10. Feingold B, Mahle WT, Auerbach S, Clemens P, Domenighetti AA, Jefferies JL, et al.; American Heart Association Pediatric Heart Failure Committee of the Council on Cardiovascular Disease in the Young; Council on Clinical Cardiology; Council on Cardiovascular Radiology and Intervention; Council on Functional Genomics and Translational Biology; and Stroke Council. Management of Cardiac Involvement Associated With Neuromuscular Diseases: A Scientific Statement From the American Heart Association. Circulation. 2017;136(13):e200-31. DOI: 10.1161/CIR.0000000000000526 PMID: 28838934 11. Nallamilli BR, Ankala A, Hegde M. Molecular diagnosis of Duchenne muscular dystrophy. Curr Protoc Hum Genet. 2014;83(1):25.1-29. DOI: 10.1002/0471142905.hg0925s83 PMID: 25271841 12. Robinson-Hamm JN, Gersbach CA. Gene therapies that restore dystrophin expression for the treatment of Duchenne muscular dystrophy. Hum Genet. 2016;135(9):1029-40. DOI: 10.1007/s00439-016- 1725-z PMID: 27542949 13. Brandsema JF, Darras BT. Dystrophinopathies. Semin Neurol. 2015;35(4):369-84. DOI: 10.1055/s-0035-1558982 PMID: 26502761 14. Darras BT, Menache-Starobinski CC, Hinton V, Kunkel LM. Dystrophinopathies. In: Darras BT, Jones HR, Ryan MM, De Vivo DC. Neuromuscular Disorders of Infancy, Childhood and Adolescence: A Clinician’s Approach. 2nd ed. San Diego (CA): Academic Press; 2015. pp. 551-92. 15. Forsyth R, Newton R. Pediatric neurology. 3rd ed. Oxford, UK: Oxford University Press; 2018. pp. 36-7. 16. Do Twee T. What is the Gower sign in patients with muscular dystrophy? Medscape, Aug 17, 2020. [cited 2020 Nov 22]. Available from: https:// www.medscape.com/answers/1259041-93872/what-is-the-gower-sign- in-patients-with-muscular-dystrophy. 17. Liang WC, Wang CH, Chou PC, Chen WZ, Jong YJ. The natural history of the patients with Duchenne muscular dystrophy in Taiwan: A medical center experience. Pediatr Neonatol. 2018;59(2):176-83. DOI: 10.1016/j. pedneo.2017.02.004 PMID: 28903883 18. Sarnat HB. Duchenne and Becker muscular dystrophies. In: Kliegman RM, Stanton BF. Nelson textbook of pediatrics. 20th ed. Philadelphia (PA): Elsevier; 2016. pp. 2976-9. 19. Nigro G, Comi LI, Politano L, Bain RJ. The incidence and evolution of cardiomyopathy in Duchenne muscular dystrophy. Int J Cardiol. 1990;26(3):271-7. DOI: 10.1016/0167-5273(90)90082-G PMID: 2312196 20. McDonald CM, Meier T, Voit T, Schara U, Straathof CS, D’Angelo MG, et al.; DELOS Study Group. Idebenone reduces respiratory complications in patients with Duchenne muscular dystrophy. Neuromuscul Disord. 2016;26(8):473-80. DOI: 10.1016/j.nmd.2016.05.008 PMID: 27238057 21. 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 Neurol. 2018;17(4):347-61. DOI: 10.1016/S1474-4422(18)30025-5 PMID: 29395990 22. Khirani S, Ramirez A, Aubertin G, Boulé M, Chemouny C, Forin V, et al. Respiratory muscle decline in Duchenne muscular dystrophy. Pediatr Pulmonol. 2014;49(5):473-81. DOI: 10.1002/ppul.22847 PMID: 23836708 23. Phillips MF, Quinlivan RC, Edwards RH, Calverley PM. Changes in spirometry over time as a prognostic marker in patients with Duchenne muscular dystrophy. Am J Respir Crit Care Med. 2001;164(12):2191-4. DOI: 10.1164/ajrccm.164.12.2103052 PMID: 11751186 24. Ishizaki M, Kobayashi M, Adachi K, Matsumura T, Kimura E. Female dystrophinopathy: review of current literature. Neuromuscul Disord. 2018;28(7):572-81. DOI: 10.1016/j.nmd.2018.04.005 PMID: 29801751 25. Aartsma-Rus A, Ginjaar IB, Bushby K. The importance of genetic diagnosis for Duchenne muscular dystrophy. J Med Genet. 2016;53(3):145-51. DOI: 10.1136/jmedgenet-2015-103387 PMID: 26754139 26. Adachi K, Hashiguchi S, Saito M, Kashiwagi S, Miyazaki T, Kawai H, et al. Detection and management of cardiomyopathy in female dystrophinopathy carriers. J Neurol Sci. 2018;386:74-80. DOI: 10.1016/j. jns.2017.12.024 PMID: 29358000 27. Mavrogeni S, Papavasiliou A, Spargias K, Constandoulakis P, Papadopoulos G, Karanasios E, et al. Myocardial inflammation in Duchenne Muscular Dystrophy as a precipitating factor for heart failure: a prospective study. BMC Neurol. 2010;10:33. DOI: 10.1186/1471-2377-10- 33 PMID: 20492678 28. Birnkrant DJ, Bushby K, Bann CM, Apkon SD, Blackwell A, Colvin MK, et al.; DMD Care Considerations Working Group. Diagnosis and management of Duchenne muscular dystrophy, part 3: primary care, emergency management, psychosocial care, and transitions of care across the lifespan. Lancet Neurol. 2018;17(5):445-55. DOI: 10.1016/ S1474-4422(18)30026-7 PMID: 29398641 29. Moxley RT, Ashwal S, Pandya S, Connolly A, Florence J, Mathews K, et al.; Quality Standards Subcommittee of the American Academy of Neurology; Practice Committee of the Child Neurology Society. Practice parameter: corticosteroid treatment of Duchenne dystrophy: report of the Quality Standards Subcommittee of the American Academy of Neurology and the Practice Committee of the Child Neurology Society. Neurology. 2005;64(1):13-20. DOI: 10.1212/01.WNL.0000148485.00049.B7 PMID: 15642897 30. Wang RT, Silverstein Fadlon CA, Ulm JW, Jankovic I, Eskin A, Lu A, et al. Online self-report data for duchenne muscular dystrophy confirms natural history and can be used to assess for therapeutic benefits. PLoS Curr. 2014;6:ecurrents.md.e1e8f2be7c949f9ffe81ec6fca1cce6a. DOI: 10.1371/currents.md.e1e8f2be7c949f9ffe81ec6fca1cce6a PMID: 25635234 31. Matthews E, Brassington R, Kuntzer T, Jichi F, Manzur AY. Corticosteroids for the treatment of Duchenne muscular dystrophy. Cochrane Library. 2016(5):CD003725. DOI: 10.1002/14651858.CD003725.pub4 PMID: 27149418 32. Koeks Z, Bladen CL, Salgado D, van Zwet E, Pogoryelova O, McMacken G, et al. Clinical Outcomes in Duchenne Muscular Dystrophy: A Study of 5345 Patients from the TREAT-NMD DMD Global Database. J Neuromuscul Dis. 2017;4(4):293-306. DOI: 10.3233/JND-170280 PMID: 29125504 33. Balaban B, Matthews DJ, Clayton GH, Carry T. Corticosteroid treatment and functional improvement in Duchenne muscular dystrophy: long- term effect. Am J Phys Med Rehabil. 2005;84(11):843-50. DOI: 10.1097/01. phm.0000184156.98671.d0 PMID: 16244521 34. Ricotti V, Ridout DA, Scott E, Quinlivan R, Robb SA, Manzur AY, et al.; NorthStar Clinical Network. Long-term benefits and adverse effects of intermittent versus daily glucocorticoids in boys with Duchenne muscular dystrophy. J Neurol Neurosurg Psychiatry. 2013;84(6):698-705. DOI: 10.1136/jnnp-2012-303902 PMID: 23250964 35. Griggs RC, Herr BE, Reha A, Elfring G, Atkinson L, Cwik V, et al. Corticosteroids in Duchenne muscular dystrophy: major variations in practice. Muscle Nerve. 2013;48(1):27-31. DOI: 10.1002/mus.23831 PMID: 23483575 36. Griggs RC, Miller JP, Greenberg CR, Fehlings DL, Pestronk A, Mendell JR, et al. Efficacy and safety of deflazacort vs prednisone and placebo for Duchenne muscular dystrophy. Neurology. 2016;87(20):2123-31. DOI: 10.1212/WNL.0000000000003217 PMID: 27566742 37. Guglieri M, Bushby K, McDermott MP, Hart KA, Tawil R, Martens WB, et al. Developing standardized corticosteroid treatment for Duchenne muscular dystrophy. Contemp Clin Trials. 2017;58:34-9. DOI: 10.1016/j. cct.2017.04.008 PMID: 28450193 548 PEDIATRICS Zdrav Vestn | september – october 2021 | Volume 90 | https://doi.org/10.6016/ZdravVestn.3172 38. Gloss D, Moxley RT, Ashwal S, Oskoui M. Practice guideline update summary: Corticosteroid treatment of Duchenne muscular dystrophy: Report of the Guideline Development Subcommittee of the American Academy of Neurology. Neurology. 2016;86(5):465-72. DOI: 10.1212/ WNL.0000000000002337 PMID: 26833937 39. Conklin LS, Damsker JM, Hoffman EP, Jusko WJ, Mavroudis PD, Schwartz BD, et al. Phase IIa trial in Duchenne muscular dystrophy shows vamorolone is a first-in-class dissociative steroidal anti-inflammatory drug. Pharmacol Res. 2018;136:140-50. DOI: 10.1016/j.phrs.2018.09.007 PMID: 30219580 40. Hoffman EP, Schwartz BD, Mengle-Gaw LJ, Smith EC, Castro D, Mah JK, et al.; Cooperative International Neuromuscular Research Group. Vamorolone trial in Duchenne muscular dystrophy shows dose-related improvement of muscle function. Neurology. 2019;93(13):e1312-23. DOI: 10.1212/WNL.0000000000008168 PMID: 31451516 41. Werneck LC, Lorenzoni PJ, Ducci RD, Fustes OH, Kay CS, Scola RH. Duchenne muscular dystrophy: an historical treatment review. Arq Neuropsiquiatr. 2019;77(8):579-89. DOI: 10.1590/0004-282x20190088 PMID: 31508685 42. Ashwath ML, Jacobs IB, Crowe CA, Ashwath RC, Super DM, Bahler RC. Left ventricular dysfunction in duchenne muscular dystrophy and genotype. Am J Cardiol. 2014;114(2):284-9. DOI: 10.1016/j.amjcard.2014.04.038 PMID: 24878125 43. Duboc D, Meune C, Pierre B, Wahbi K, Eymard B, Toutain A, et al. Perindopril preventive treatment on mortality in Duchenne muscular dystrophy: 10 years’ follow-up. Am Heart J. 2007;154(3):596-602. DOI: 10.1016/j.ahj.2007.05.014 PMID: 17719312 44. Duboc D, Meune C, Lerebours G, Devaux JY, Vaksmann G, Bécane HM. Effect of perindopril on the onset and progression of left ventricular dysfunction in Duchenne muscular dystrophy. J Am Coll Cardiol. 2005;45(6):855-7. DOI: 10.1016/j.jacc.2004.09.078 PMID: 15766818 45. Allen HD, Flanigan KM, Thrush PT, Dvorchik I, Yin H, Canter C, et al. A Randomized, Double-Blind Trial of Lisinopril and Losartan for the Treatment of Cardiomyopathy in Duchenne Muscular Dystrophy. PLoS Curr. 2013;5:ecurrents.md.2cc69a1dae4be7dfe2bcb420024ea865. DOI: 10.1371/currents.md.2cc69a1dae4be7dfe2bcb420024ea865 PMID: 24459612 46. Sawnani H. Sleep disordered breathing in Duchenne muscular dystrophy. Paediatr Respir Rev. 2019;30:2-8. PMID: 30153979 47. Tsuda T. Clinical Manifestations and Overall Management Strategies for Duchenne Muscular Dystrophy. In: Bernardini C. Duchenne Muscular Dystrophy. New York: Humana Press; 2018. (Methods in Molecular Biology; vol 1687). 48. Nacionalni inštitut za javno zdravje. Priporočila za cepljenje odraslih in otrok, starih 5 let in več, proti pnevmokoknim okužbam. Ljubljana: NIJZ; 2019 [cited 2020 Sept 09]. Available from: https://www.nijz.si/sites/ www.nijz.si/files/uploaded/cepljenje_pnevmo_za_odrasle_in_otroke_ nad_5_let_posodobljen_2019.pdf. 49. Nascimento Osorio A, Medina Cantillo J, Camacho Salas A, Madruga Garrido M, Vilchez Padilla JJ. Consensus on the diagnosis, treatment and follow-up of patients with Duchenne muscular dystrophy. Neurologia (Engl Ed). 2019;34(7):469-81. PMID: 29526319 DOI: 10.1016/j. nrl.2018.01.001 50. Cotton S, Voudouris NJ, Greenwood KM. Intelligence and Duchenne muscular dystrophy: full-scale, verbal, and performance intelligence quotients. Dev Med Child Neurol. 2001;43(7):497-501. DOI: 10.1017/ S0012162201000913 PMID: 11463183 51. Cotton SM, Voudouris NJ, Greenwood KM. Association between intellectual functioning and age in children and young adults with Duchenne muscular dystrophy: further results from a meta-analysis. Dev Med Child Neurol. 2005;47(4):257-65. DOI: 10.1017/S0012162205000496 PMID: 15832549 52. Chamova T, Guergueltcheva V, Raycheva M, Todorov T, Genova J, Bichev S, et al. Association between loss of dp140 and cognitive impairment in duchenne and becker dystrophies. Balkan J Med Genet. 2013;16(1):21- 30. DOI: 10.2478/bjmg-2013-0014 PMID: 24265581 53. Doorenweerd N, Mahfouz A, van Putten M, Kaliyaperumal R. Timing and localization of human dystrophin isoform expression provide insights into the cognitive phenotype of Duchenne muscular dystrophy. Sci Rep. 2017;7(1):12575. DOI: 10.1038/s41598-017-12981-5 PMID: 28974727 54. Billard C, Gillet P, Barthez M, Hommet C, Bertrand P. Reading ability and processing in Duchenne muscular dystrophy and spinal muscular atrophy. Dev Med Child Neurol. 1998;40(1):12-20. DOI: 10.1111/j.1469- 8749.1998.tb15351.x PMID: 9459212 55. Hendriksen JG, Vles JS. Are males with Duchenne muscular dystrophy at risk for reading disabilities? Pediatr Neurol. 2006;34(4):296-300. DOI: 10.1016/j.pediatrneurol.2005.08.029 PMID: 16638505 56. Hinton VJ, Nereo NE, Fee RJ, Cyrulnik SE. Social behavior problems in boys with Duchenne muscular dystrophy. J Dev Behav Pediatr. 2006;27(6):470- 6. DOI: 10.1097/00004703-200612000-00003 PMID: 17164619 57. Hinton VJ, De Vivo DC, Fee R, Goldstein E, Stern Y. Investigation of Poor Academic Achievement in Children with Duchenne Muscular Dystrophy. Learn Disabil Res Pract. 2004;19(3):146-54. DOI: 10.1111/j.1540- 5826.2004.00098.x PMID: 17710256 58. Cyrulnik SE, Fee RJ, Batchelder A, Kiefel J, Goldstein E, Hinton VJ. Cognitive and adaptive deficits in young children with Duchenne muscular dystrophy (DMD). J Int Neuropsychol Soc. 2008;14(5):853-61. DOI: 10.1017/S135561770808106X PMID: 18764980 59. Snow WM, Anderson JE, Jakobson LS. Neuropsychological and neurobehavioral functioning in Duchenne muscular dystrophy: a review. Neurosci Biobehav Rev. 2013;37(5):743-52. DOI: 10.1016/j. neubiorev.2013.03.016 PMID: 23545331 60. Ricotti V, Mandy WP, Scoto M, Pane M, Deconinck N, Messina S, et al. Neurodevelopmental, emotional, and behavioural problems in Duchenne muscular dystrophy in relation to underlying dystrophin gene mutations. Dev Med Child Neurol. 2016;58(1):77-84. DOI: 10.1111/ dmcn.12922 PMID: 26365034 61. Ayres AJ. Treatment of sensory integrative dysfunction. Aust Occup Ther J. 2010;19(2):88. DOI: 10.1111/j.1440-1630.1972.tb00547.x PMID: 20854573 62. Magliano L, D’Angelo MG, Vita G, Pane M, D’Amico A, Balottin U, et al. Psychological and practical difficulties among parents and healthy siblings of children with Duchenne vs. Becker muscular dystrophy: an Italian comparative study. Acta Myol. 2014;33(3):136-43. PMID: 25873782 63. Boyer F, Drame M, Morrone I, Novella JL. Factors relating to carer burden for families of persons with muscular dystrophy. J Rehabil Med. 2006;38(5):309-15. DOI: 10.1080/16501970600731578 PMID: 16931461 64. Reid DT, Renwick RM. Relating familial stress to the psychosocial adjustment of adolescents with Duchenne muscular dystrophy. Int J Rehabil Res. 2001;24(2):83-93. DOI: 10.1097/00004356-200106000-00001 PMID: 11421396 65. Kenneson A, Bobo JK. The effect of caregiving on women in families with Duchenne/Becker muscular dystrophy. Health Soc Care Community. 2010;18(5):520-8. DOI: 10.1111/j.1365-2524.2010.00930.x PMID: 20561071 66. Read J, Kinali M, Muntoni F, Garralda ME. Psychosocial adjustment in siblings of young people with Duchenne muscular dystrophy. Eur J Paediatr Neurol. 2010;14(4):340-8. DOI: 10.1016/j.ejpn.2009.09.011 PMID: 19850502 67. Read J, Kinali M, Muntoni F, Weaver T, Garralda ME. Siblings of young people with Duchenne muscular dystrophy—a qualitative study of impact and coping. Eur J Paediatr Neurol. 2011;15(1):21-8. DOI: 10.1016/j. ejpn.2010.07.006 PMID: 20728383 68. Bérard C, Payan C, Hodgkinson I, Fermanian J; MFM Collaborative Study Group. A motor function measure for neuromuscular diseases. Construction and validation study. Neuromuscul Disord. 2005;15(7):463- 70. DOI: 10.1016/j.nmd.2005.03.004 PMID: 16106528 69. Skolioza. Ljubljana: Univerzitetni klinični center, Ortopedska klinika; 2020 [cited 2020 Nov 29]. Available from: http://www.ortopedska-klinika. si/upload/files/skolioza.pdf. 70. Pravilnik o medicinskih pripomočkih. Ljubljana: Služba Vlade RS za zakonodajo; 2012 [cited 2020 Aug 15]. Available from: http://www.pisrs. si/Pis.web/pregledPredpisa?id=PRAV9508. 549 PROFESSIONAL ARTICLE Duchenne muscular dystrophy – novelties in diagnostics and treatment 71. Strehle EM, Straub V. Recent advances in the management of Duchenne muscular dystrophy. Arch Dis Child. 2015;100(12):1173-7. DOI: 10.1136/ archdischild-2014-307962 PMID: 26153505 72. Siddiqui N, Sonenberg N. Proposing a mechanism of action for ataluren. Proc Natl Acad Sci USA. 2016;113(44):12353-5. DOI: 10.1073/ pnas.1615548113 PMID: 27791186 73. Finkel RS. Read-through strategies for suppression of nonsense mutations in Duchenne/ Becker muscular dystrophy: aminoglycosides and ataluren (PTC124). J Child Neurol. 2010;25(9):1158-64. DOI: 10.1177/0883073810371129 PMID: 20519671 74. Welch EM, Barton ER, Zhuo J, Tomizawa Y, Friesen WJ, Trifillis P, et al. PTC124 targets genetic disorders caused by nonsense mutations. Nature. 2007;447(7140):87-91. DOI: 10.1038/nature05756 PMID: 17450125 75. Bönnemann C, Finkel R, Wong B, Flanigan K, Sampson J, Sweeney L, et al. G.P.3.05 Phase 2 study of PTC124 for nonsense mutation suppression therapy of Duchenne muscular dystrophy (DMD). Neuromuscul Disord. 2007;17(9–10):783. DOI: 10.1016/j.nmd.2007.06.080 76. Bushby K, Finkel R, Wong B, Barohn R, Campbell C, Comi GP, et al.; PTC124-GD-007-DMD STUDY GROUP. Ataluren treatment of patients with nonsense mutation dystrophinopathy. Muscle Nerve. 2014;50(4):477-87. DOI: 10.1002/mus.24332 PMID: 25042182 77. McDonald CM, Campbell C, Torricelli RE, Finkel RS, Flanigan KM, Goemans N, et al.; Clinical Evaluator Training Group; ACT DMD Study Group. Ataluren in patients with nonsense mutation Duchenne muscular dystrophy (ACT DMD): a multicentre, randomised, double-blind, placebo- controlled, phase 3 trial. Lancet. 2017;390(10101):1489-98. DOI: 10.1016/ S0140-6736(17)31611-2 PMID: 28728956 78. Lim KR, Maruyama R, Yokota T. Eteplirsen in the treatment of Duchenne muscular dystrophy. Drug Des Devel Ther. 2017;11:533-45. DOI: 10.2147/ DDDT.S97635 PMID: 28280301 79. Shieh PB. Emerging Strategies in the Treatment of Duchenne Muscular Dystrophy. Neurotherapeutics. 2018;15(4):840-8. DOI: 10.1007/s13311- 018-00687-z PMID: 30414046 80. U.S. National Library of Medicine. ClinicalTrials.gov. Study of Eteplirsen in DMD Patients (PROMOVI). Bethesda: NLM; 2020 [cited 2020 Aug 05]. Available from: https://clinicaltrials.gov/ct2/show/study/NCT02255552. 81. Heo YA. Golodirsen: first Approval. Drugs. 2020;80(3):329-33. DOI: 10.1007/s40265-020-01267-2 PMID: 32026421 82. Duan D. Systemic AAV Micro-dystrophin Gene Therapy for Duchenne Muscular Dystrophy. Mol Ther. 2018;26(10):2337-56. DOI: 10.1016/j. ymthe.2018.07.011 PMID: 30093306 83. Galli F, Bragg L, Meggiolaro L, Rossi M, Caffarini M, Naz N, et al. Gene and Cell Therapy for Muscular Dystrophies: Are We Getting There? Hum Gene Ther. 2018;29(10):1098-105. DOI: 10.1089/hum.2018.151 PMID: 30132372 84. Shimizu-Motohashi Y, Miyatake S, Komaki H, Takeda S, Aoki Y. Recent advances in innovative therapeutic approaches for Duchenne muscular dystrophy: from discovery to clinical trials. Am J Transl Res. 2016;8(6):2471-89. PMID: 27398133