Spinal muscular atrophy

This article is about a genetic disorder associated with mutation in the SMN1 gene. For a list of other conditions with similar names, see Spinal muscular atrophies.
Spinal muscular atrophy
Location of neurons affected by spinal muscular atrophy in the spinal cord
Classification and external resources
Specialty Medical genetics
ICD-10 G12.0-G12.1
ICD-9-CM 335.0-335.1
OMIM 253300 253550 253400 271150
DiseasesDB 14093 32911
MedlinePlus 000996
eMedicine Spinal Muscular Atrophy
Spinal Muscle Atrophy
Kugelberg–Welander SMA
Patient UK Spinal muscular atrophy
MeSH D014897
GeneReviews

Spinal muscular atrophy (SMA), also called autosomal recessive proximal spinal muscular atrophy in order to distinguish it from other conditions with similar name, is a rare neuromuscular disorder characterised by loss of motor neurons and progressive muscle wasting, often leading to early death.

The disorder is caused by a genetic defect in the SMN1 gene, which encodes SMN, a protein widely expressed in all eukaryotic cells and necessary for survival of motor neurons. Lower levels of the protein results in loss of function of neuronal cells in the anterior horn of the spinal cord and subsequent system-wide muscle wasting (atrophy).

Spinal muscular atrophy manifests in various degrees of severity, which all have in common progressive muscle wasting and mobility impairment. Proximal muscles and lung muscles are affected first. Other body systems may be affected as well, particularly in early-onset forms of the disorder. SMA is the most common genetic cause of infant death.

Spinal muscular atrophy is an inherited disorder and is passed on in an autosomal recessive manner.

As of 2016, no drugs for SMA have been approved, with several compounds remaining in clinical trials and one, nusinersen, submitted for marketing authorisation.

Classification

SMA manifests over a wide range of severity, affecting infants through adults. The disease spectrum is variously divided into 3–5 types, in accordance either with the age of onset of symptoms or with the highest attained milestone of motor development.

The most commonly used classification is as follows:

Type Eponym Usual age of onset Characteristics OMIM
SMA1
(Infantile)
Werdnig–Hoffmann disease 0–6 months The severe form manifests in the first months of life, usually with a quick and unexpected onset ("floppy baby syndrome"). Rapid motor neuron death causes inefficiency of the major bodily organs - especially of the respiratory system - and pneumonia-induced respiratory failure is the most frequent cause of death. Babies diagnosed with SMA type 1 do not generally live past two years of age, with death occurring as early as within weeks in the most severe cases (sometimes termed SMA type 0). With proper respiratory support, those with milder SMA type I phenotypes, which account for around 10% of SMA1 cases, are known to live into adolescence and adulthood. 253300
SMA2
(Intermediate)
Dubowitz disease 6–18 months The intermediate form affects children who are never able to stand and walk but who are able to maintain a sitting position at least some time in their life. The onset of weakness is usually noticed some time between 6 and 18 months. The progress is known to vary greatly, some patients gradually grow weaker over time while others through careful maintenance avoid any progression. Scoliosis may be present in these children, and correction with a brace may help improve respiration. Body muscles are weakened, and the respiratory system is a major concern. Life expectancy is somewhat reduced but most SMA2 patients live well into adulthood. 253550
SMA3
(Juvenile)
Kugelberg–Welander disease >12 months The juvenile form usually manifests after 12 months of age and describes patients who are able to walk without support at some time, although many later lose this ability. Respiratory involvement is less noticeable, and life expectancy is normal or near normal. 253400
SMA4
(Adult-onset)
Adulthood The adult-onset form (sometimes classified as a late-onset SMA type 3) usually manifests after the third decade of life with gradual weakening of muscles – mainly affects proximal muscles of the extremities – frequently requiring the patient to use a wheelchair for mobility. Other complications are rare, and life expectancy is unaffected. 271150

The most severe form of SMA type I is sometimes termed SMA type 0 (or, severe infantile SMA) and is diagnosed in babies that are born so weak that they can survive only a few weeks even with intensive respiratory support. SMA type 0 should not be confused with SMARD1 which may have very similar symptoms and course but has a different genetic cause than SMA.

Motor development in SMA patients is usually assessed using validated functional scales – CHOP INTEND (The Children's Hospital of Philadelphia Infant Test of Neuromuscular Disorders) in SMA1; and either the Motor Function Measure scale or one of a few variants of Hammersmith Functional Motor Scale[1][2][3][4] in SMA types 2 and 3.

The eponymous label Werdnig–Hoffmann disease (sometimes misspelled with a single n) refers to the earliest clinical descriptions of childhood SMA by Johann Hoffmann and Guido Werdnig. The eponymous term Kugelberg–Welander disease is after Erik Klas Hendrik Kugelberg (1913-1983) and Lisa Welander (1909-2001), who distinguished SMA from muscular dystrophy.[5] Rarely used Dubowitz disease (not to be confused with Dubowitz syndrome) is named after Victor Dubowitz, an English neurologist who authored several studies on the intermediate SMA phenotype.

Signs and symptoms

The symptoms vary greatly depending on the SMA type involved, the stage of the disease, and individual factors; they commonly include:

  • Areflexia, particularly in extremities
  • Overall muscle weakness, poor muscle tone, limpness or a tendency to flop
  • Difficulty achieving developmental milestones, difficulty sitting/standing/walking
  • In small children: adopting of a frog-leg position when sitting (hips abducted and knees flexed)
  • Loss of strength of the respiratory muscles: weak cough, weak cry (infants), accumulation of secretions in the lungs or throat, respiratory distress
  • Bell-shaped torso (caused by using only abdominal muscles for respiration) in weaker SMA types
  • Fasciculations (twitching) of the tongue
  • Difficulty sucking or swallowing, poor feeding

Causes

Spinal muscular atrophy has an autosomal recessive pattern of inheritance.

Spinal muscular atrophy is linked to a genetic mutation in the SMN1 gene.[6]

Human chromosome 5 contains two nearly identical genes at location 5q13: a telomeric copy SMN1 and a centromeric copy SMN2. In healthy individuals, the SMN1 gene codes the survival of motor neuron protein (SMN) which, as its name says, plays a crucial role in survival of motor neurons. The SMN2 gene, on the other hand - due to a variation in a single nucleotide (840.C→T) - undergoes alternative splicing at the junction of intron 6 to exon 8, with only 10-20% of SMN2 transcripts coding a fully functional survival of motor neuron protein (SMN-fl) and 80-90% of transcripts resulting in a truncated protein compound (SMNΔ7) which is rapidly degraded in the cell.

In individuals affected by SMA, the SMN1 gene is mutated in such a way that it is unable to correctly code the SMN protein - due to either a deletion occurring at exon 7 or to other point mutations (frequently resulting in the functional conversion of the SMN1 sequence into SMN2). All patients, however, retain at least one copy of the SMN2 gene (with most having 2-4 of them) which still codes small amounts of SMN protein - around 10-20% of the normal level - allowing some neurons to survive. In the long run, however, reduced availability of the SMN protein results in gradual death of motor neuron cells in the anterior horn of spinal cord and the brain. Muscles that depend on these motor neurons for neural input now have decreased innervation (also called denervation), and therefore have decreased input from the central nervous system (CNS). Denervated skeletal muscle is more difficult for the body to control. Decreased impulse transmission through the motor neurons leads to decreased contractile activity of the denervated muscle. Consequently, denervated muscles undergo progressive atrophy.

Muscles of lower extremities are usually affected first, followed by muscles of upper extremities, spine and neck and, in more severe cases, pulmonary and mastication muscles. Proximal muscles are always affected earlier and to a greater degree than distal.

The severity of SMA symptoms is broadly related to how well the remaining SMN2 genes can make up for the loss of function of SMN1. This is partly related to the number of SMN2 gene copies present on the chromosome. Whilst healthy individuals carry two SMN2 gene copies, patients with SMA can have anything between 1 and 4 (or more) of them, with the greater the number of SMN2 copies, the milder the disease severity. Thus, most SMA type I babies have one or two SMN2 copies; SMA II and III patients usually have at least three SMN2 copies; and SMA IV patients normally have at least four of them. However, the correlation between symptom severity and SMN2 copy number is not absolute, and there seem to exist other factors affecting the disease phenotype.[7]

Spinal muscular atrophy is inherited in an autosomal recessive pattern, which means that the defective gene is located on an autosome. Two copies of the defective gene - one from each parent - are required to inherit the disorder: the parents may be carriers and not personally affected. SMA seems to appear de novo (i.e., without any hereditary causes) in around 2-4% of cases.

Spinal muscular atrophy affects individuals of all ethnic groups, unlike other well known autosomal recessive disorders, such as sickle cell disease and cystic fibrosis, which have significant differences in occurrence rate among ethnic groups. The overall prevalence of SMA, of all types and across all ethnic groups, is in the range of 1 per 10,000 individuals; the gene frequency is around 1:100, therefore, approximately one in 50 persons are carriers.[8][9] There are no known health consequences of being a carrier. A person may learn carrier status only if one's child is affected by SMA or by having the SMN1 gene sequenced.

Affected siblings usually have a very similar form of SMA. However, occurrences of different SMA types among siblings do exist – while rare, these cases might be due to additional de novo deletions of the SMN gene, not involving the NAIP gene, or the differences in SMN2 copy numbers.

Diagnosis

Very severe SMA (type 0/1) can be sometimes evident before birth - reduction in foetal movement in the final months of pregnancy. Otherwise SMA1 manifests within the first few weeks or months of life when abnormally low muscle tone is observed in the infant (the "floppy baby syndrome").

For all SMA types,

While the above symptoms point towards SMA, the diagnosis can only be confirmed with absolute certainty through genetic testing for bi-allelic deletion of exon 7 of the SMN1 gene. Genetic test is usually carried out using a blood sample, and MLPA is one of more frequently used techniques as it also allows establishing the number of SMN2 gene copies.

Preimplantation testing

Preimplantation genetic diagnosis can be used to detect SMA-affected foetuses, especially when undergoing in-vitro fertilisation.

Prenatal testing

Prenatal testing towards SMA is possible through chorionic villus sampling, cell-free fetal DNA analysis and other methods.

Carrier testing

Those at risk of being carriers of SMN1 deletion, and thus at risk of having offspring affected by SMA, can undergo carrier analysis using blood or saliva sample.

Routine screening

Routine prenatal or neonatal screening for SMA is controversial, because of the cost and because of the severity of the disease as well as lack of approved treatment. Some researchers have concluded that population screening for SMA is not cost-effective, at a cost of $5 million per case averted in United States.[11] Others conclude that SMA meets the criteria for screening programs and relevant testing should be offered to all couples.[12]

Management

There is no pharmacological treatment to spinal muscular atrophy. Care is symptomatic. Main areas of concern are as follows:

Orthopaedics

Weak spine muscles may lead to development of kyphosis, scoliosis and other orthopaedic problems. Spine fusion is sometimes performed in SMA I/II patients once they reach the age of 8-10 to relieve the pressure of a deformed spine on the lungs. Patients with SMA might also benefit greatly from various forms of physiotherapy and occupational therapy.

Mobility support

Orthotic devices can be used to support the body and to aid walking. For example, orthotics such as AFO's (ankle foot orthosis) are used to stabilise the foot and to aid gait, TLSO's (thoracic lumbar sacral orthosis) are used to stabilise the torso. Assistive technologies may help in managing movement and daily activity and greatly increase the quality of life.

Respiratory care

Respiratory system requires utmost attention in SMA as once weakened it never fully recovers. Weakened pulmonary muscles in SMA type I/II patients can make breathing more difficult and pose a risk of hypoxiation, especially in sleep when muscles are more relaxed. Impaired cough reflex poses a constant risk of respiratory infection and pneumonia. Non-invasive ventilation (BiPAP) is frequently used and tracheostomy may be sometimes performed in more severe cases;[13] both methods of ventilation prolong survival in a comparable degree, although tracheostomy prevents speech development.[14]

Nutrition

Difficulties in jaw opening, chewing and swallowing food might put patients with SMA at risk of malnutrition. A feeding tube or gastrostomy can be necessary in SMA type I and more severe type II patients.[15][16][17] Additionally, metabolic abnormalities resulting from SMA impair β-oxidation of fatty acids in muscles and can lead to organic acidemia and consequent muscle damage, especially when fasting.[18][19] It is suggested that patients with SMA, especially those with more severe forms of the disease, reduce intake of fat and avoid prolonged fasting (i.e., eat more frequently than healthy people).[20]

Cardiology

Although the heart is not a matter of routine concern, a link between SMA and certain heart conditions has been suggested.[21][22][23][24]

Mental health

SMA children do not differ from the general population in their behaviour; their cognitive development can be slightly faster, and certain aspects of their intelligence are above the average.[25][26][27] Despite their disability, SMA-affected people report high degree of satisfaction from life.[28]

Palliative care in SMA has been standardised in the Consensus Statement for Standard of Care in Spinal Muscular Atrophy which has been recommended for standard adoption worldwide.

Prognosis

Generally, patients tend to deteriorate over time, but prognosis varies with the SMA type and disease progress which shows a great degree of individual variability.

The majority of children diagnosed with SMA type 0 and 1 do not reach the age of 4, recurrent respiratory problems being the primary cause of death.[29] With proper care, milder SMA type 1 cases (which account for approx. 10% of all SMA1 cases) live into adulthood.[30] Long-term survival in SMA1 is not sufficiently evidenced; however, recent advances in respiratory support seem to have brought down mortality.[31]

In SMA type 2, the course of the disease is stable or slowly progressing and life expectancy is reduced compared to the healthy population. Death before the age of 20 is frequent, although many patients live to become parents and grandparents. SMA type 3 has normal or near-normal life expectancy if standards of care are followed. Adult-onset SMA usually means only mobility impairment and does not affect life expectancy.

In all SMA types, physiotherapy has been shown to delay the progress of disease.

Research directions

Since the underlying genetic mechanism of SMA was described in 1990, several therapeutic approaches have been proposed and investigated which primarily focused on increasing the availability of SMN protein to motor neurons.. The main research directions are as follows:

SMN1 gene replacement

Gene therapy in SMA aims at restoring the SMN1 gene function through inserting specially crafted nucleotide sequence (a SMN1 transgene) into the cell nucleus using a viral vector; scAAV-9 and scAAV-10 are the primary viral vectors under investigation.

Only one programme has reached the clinical stage:

Work on developing gene therapy for SMA is also conducted at the Institut de Myologie in Paris, at Genzyme Corporation and at the University of Oxford.

SMN2 alternative splicing modulation

This approach aims at modifying the alternative splicing of the SMN2 gene so that to force it to code for higher percentage of full-length SMN protein. Sometimes it is also called gene conversion, because it attempts to convert the SMN2 gene functionally into SMN1 gene.

The following splicing modulators have reached clinical stage development:

Of discontinued clinical-stage molecules, RG3039, also known as Quinazoline495, was a proprietary quinazoline derivative developed by Repligen and licensed to Pfizer in March 2014 which was discontinued shortly after, having only completed phase I trials. PTK-SMA1 was a proprietary small-molecule splicing modulator of the tetracyclines group developed by Paratek Pharmaceutical and about to enter clinical development in 2010 which however never happened.

Basic research has also identified other compounds which modified SMN2 splicing in vitro, like sodium orthovanadate[34] and aclarubicin.[35] Morpholino-type antisense oligonucleotides, similar to those used in nusinersen, remain a subject of intense research, including at the University College London[36] and at the University of Oxford.

SMN2 gene activation

This approach aims at increasing expression (activity) of the SMN2 gene, thus increasing the amount of full-length SMN protein available.

A few compounds initially showed promise but turned out ineffective upon more extensive research:

Compounds which increased SMN2 activity in vitro but did not make it to the clinical stage include growth hormone, various histone deacetylase inhibitors,[54] benzamide M344,[55] hydroxamic acids (CBHA, SBHA, entinostat, panobinostat,[56] trichostatin A,[57][58] vorinostat[59]), prolactin[60] as well as natural polyphenol compounds like resveratrol and curcumin.[61][62]

SMN stabilisation

SMN stabilisation aims at stabilising the SMNΔ7 protein, the short-lived defective protein coded by the SMN2 gene, so that it is able to sustain neuronal cells.[63]

No compounds have been taken forward to the clinical stage. Aminoglycosides showed capability to increase SMN protein availability in two studies.[64][65] Indoprofen offered some promise in vitro.[66]

Neuroprotection

Neuroprotective drugs aim at enabling the survival of motor neurons even with low levels of SMN protein.

Thyrotropin-releasing hormone (TRH) showed some promise in an open-label uncontrolled clinical trial[70][71][72] but did not prove effective in a subsequent double-blind placebo-controlled trial.[73]

Compounds that had some neuroprotective effect in in vitro research but never moved to in vivo studies include β-lactam antibiotics (e.g., ceftriaxone)[74][75] and follistatin.[76]

Muscle restoration

This approach aims to counter the effect of SMA by targeting the muscle tissue instead of neurons.

Stem cells

As of 2016, there has been no significant breakthrough in stem cell therapy in SMA. An experimental programme to develop a stem cell based therapeutic product for SMA was run, with financial support from the SMA community, by a US company California Stem Cell starting from 2005. It was discontinued in 2010, unable to enter the clinical stage, and the company ceased to exist shortly after.

In 2013–2014, a small number of SMA1 children in Italy received court-mandated stem cell injections following the Stamina scam, but the treatment was reported having no effect.[78][79]

Whilst stem cells never form a part of any recognised therapy for SMA, a number of private companies, usually located in countries with lax regulatory oversight, take advantage of media hype and market stem cell injections as a "cure" for a vast range of disorders, including SMA. The medical consensus is that such procedures offer no clinical benefit whilst carrying significant risk to the patient, therefore patients are advised against them.[80][81]

Patient registries

Patients with SMA can avail of an opportunity of participating in clinical research by entering their details into SMA patient registries managed by TREAT-NMD.[82]

Notable people

Notable people who lived or live with spinal muscular atrophy include:

See also

References

  1. Main, M.; Kairon, H.; Mercuri, E.; Muntoni, F. (2003). "The Hammersmith Functional Motor Scale for Children with Spinal Muscular Atrophy: A Scale to Test Ability and Monitor Progress in Children with Limited Ambulation". European Journal of Paediatric Neurology. 7 (4): 155–159. doi:10.1016/S1090-3798(03)00060-6. PMID 12865054.
  2. Krosschell, K. J.; Maczulski, J. A.; Crawford, T. O.; Scott, C.; Swoboda, K. J. (2006). "A modified Hammersmith functional motor scale for use in multi-center research on spinal muscular atrophy". Neuromuscular Disorders. 16 (7): 417–426. doi:10.1016/j.nmd.2006.03.015. PMC 3260054Freely accessible. PMID 16750368.
  3. O'Hagen, J. M.; Glanzman, A. M.; McDermott, M. P.; Ryan, P. A.; Flickinger, J.; Quigley, J.; Riley, S.; Sanborn, E.; Irvine, C.; Martens, W. B.; Annis, C.; Tawil, R.; Oskoui, M.; Darras, B. T.; Finkel, R. S.; De Vivo, D. C. (2007). "An expanded version of the Hammersmith Functional Motor Scale for SMA II and III patients". Neuromuscular Disorders. 17 (9–10): 693–697. doi:10.1016/j.nmd.2007.05.009. PMID 17658255.
  4. Glanzman, A. M.; O'Hagen, J. M.; McDermott, M. P.; Martens, W. B.; Flickinger, J.; Riley, S.; Quigley, J.; Montes, J.; Dunaway, S.; Deng, L.; Chung, W. K.; Tawil, R.; Darras, B. T.; De Vivo, D. C.; Kaufmann, P.; Finkel, R. S.; Pediatric Neuromuscular Clinical Research Network for Spinal Muscular Atrophy (PNCR) (2011). "Validation of the Expanded Hammersmith Functional Motor Scale in Spinal Muscular Atrophy Type II and III". Journal of Child Neurology. 26 (12): 1499–1507. doi:10.1177/0883073811420294. PMID 21940700.
  5. Dubowitz, V. (2009). "Ramblings in the history of spinal muscular atrophy". Neuromuscular Disorders. 19 (1): 69–73. doi:10.1016/j.nmd.2008.10.004. PMID 18951794.
  6. Brzustowicz, L. M.; Lehner, T.; Castilla, L. H.; Penchaszadeh, G. K.; Wilhelmsen, K. C.; Daniels, R.; Davies, K. E.; Leppert, M.; Ziter, F.; Wood, D.; Dubowitz, V.; Zerres, K.; Hausmanowa-Petrusewicz, I.; Ott, J.; Munsat, T. L.; Gilliam, T. C. (1990). "Genetic mapping of chronic childhood-onset spinal muscular atrophy to chromosome 5q11.2–13.3". Nature. 344 (6266): 540–541. Bibcode:1990Natur.344..540B. doi:10.1038/344540a0. PMID 2320125.
  7. Jędrzejowska, M.; Milewski, M.; Zimowski, J.; Borkowska, J.; Kostera-Pruszczyk, A.; Sielska, D.; Jurek, M.; Hausmanowa-Petrusewicz, I. (2009). "Phenotype modifiers of spinal muscular atrophy: The number of SMN2 gene copies, deletion in the NAIP gene and probably gender influence the course of the disease". Acta Biochimica Polonica. 56 (1): 103–108. PMID 19287802.
  8. Su, Y. N.; Hung, C. C.; Lin, S. Y.; Chen, F. Y.; Chern, J. P. S.; Tsai, C.; Chang, T. S.; Yang, C. C.; Li, H.; Ho, H. N.; Lee, C. N. (2011). Schrijver, Iris, ed. "Carrier Screening for Spinal Muscular Atrophy (SMA) in 107,611 Pregnant Women during the Period 2005–2009: A Prospective Population-Based Cohort Study". PLoS ONE. 6 (2): e17067. doi:10.1371/journal.pone.0017067. PMC 3045421Freely accessible. PMID 21364876.
  9. Sugarman, E. A.; Nagan, N.; Zhu, H.; Akmaev, V. R.; Zhou, Z.; Rohlfs, E. M.; Flynn, K.; Hendrickson, B. C.; Scholl, T.; Sirko-Osadsa, D. A.; Allitto, B. A. (2011). "Pan-ethnic carrier screening and prenatal diagnosis for spinal muscular atrophy: Clinical laboratory analysis of >72 400 specimens". European Journal of Human Genetics. 20 (1): 27–32. doi:10.1038/ejhg.2011.134. PMC 3234503Freely accessible. PMID 21811307.
  10. Rutkove, S. B.; Shefner, J. M.; Gregas, M.; Butler, H.; Caracciolo, J.; Lin, C.; Fogerson, P. M.; Mongiovi, P.; Darras, B. T. (2010). "Characterizing spinal muscular atrophy with electrical impedance myography". Muscle & Nerve. 42 (6): 915–921. doi:10.1002/mus.21784.
  11. Little, S. E.; Janakiraman, V.; Kaimal, A.; Musci, T.; Ecker, J.; Caughey, A. B. (2010). "The cost-effectiveness of prenatal screening for spinal muscular atrophy". American Journal of Obstetrics and Gynecology. 202 (3): 253.2e1. doi:10.1016/j.ajog.2010.01.032. PMID 20207244.
  12. Prior, T. W.; Professional Practice Guidelines Committee (2008). "Carrier screening for spinal muscular atrophy". Genetics in Medicine. 10 (11): 840–842. doi:10.1097/GIM.0b013e318188d069. PMC 3110347Freely accessible. PMID 18941424.
  13. Bach, J. R.; Niranjan, V.; Weaver, B. (2000). "Spinal Muscular Atrophy Type 1: A Noninvasive Respiratory Management Approach". Chest. 117 (4): 1100–1105. doi:10.1378/chest.117.4.1100. PMID 10767247.
  14. Bach, J. R.; Saltstein, K.; Sinquee, D.; Weaver, B.; Komaroff, E. (2007). "Long-Term Survival in Werdnig–Hoffmann Disease". American Journal of Physical Medicine & Rehabilitation. 86 (5): 339–45 quiz 346–8, 379. doi:10.1097/PHM.0b013e31804a8505. PMID 17449977.
  15. Messina, S.; Pane, M.; De Rose, P.; Vasta, I.; Sorleti, D.; Aloysius, A.; Sciarra, F.; Mangiola, F.; Kinali, M.; Bertini, E.; Mercuri, E. (2008). "Feeding problems and malnutrition in spinal muscular atrophy type II". Neuromuscular Disorders. 18 (5): 389–393. doi:10.1016/j.nmd.2008.02.008. PMID 18420410.
  16. Chen, Y. S.; Shih, H. H.; Chen, T. H.; Kuo, C. H.; Jong, Y. J. (2011). "Prevalence and Risk Factors for Feeding and Swallowing Difficulties in Spinal Muscular Atrophy Types II and III". The Journal of Pediatrics. 160 (3): 447–451.e1. doi:10.1016/j.jpeds.2011.08.016. PMID 21924737.
  17. Tilton, A.; Miller, M.; Khoshoo, V. (1998). "Nutrition and swallowing in pediatric neuromuscular patients". Seminars in Pediatric Neurology. 5 (2): 106–115. doi:10.1016/S1071-9091(98)80026-0. PMID 9661244.
  18. Tein, I.; Sloane, A. E.; Donner, E. J.; Lehotay, D. C.; Millington, D. S.; Kelley, R. I. (1995). "Fatty acid oxidation abnormalities in childhood-onset spinal muscular atrophy: Primary or secondary defect(s)?". Pediatric neurology. 12 (1): 21–30. doi:10.1016/0887-8994(94)00100-G. PMID 7748356.
  19. Crawford, T. O.; Sladky, J. T.; Hurko, O.; Besner-Johnston, A.; Kelley, R. I. (1999). "Abnormal fatty acid metabolism in childhood spinal muscular atrophy". Annals of Neurology. 45 (3): 337–343. doi:10.1002/1531-8249(199903)45:3<337::AID-ANA9>3.0.CO;2-U. PMID 10072048.
  20. Leighton, S. (2003). "Nutrition issues associated with spinal muscular atrophy". Nutrition & Dietetics. 60 (2): 92–96.
  21. Rudnik-Schoneborn, S.; Heller, R.; Berg, C.; Betzler, C.; Grimm, T.; Eggermann, T.; Eggermann, K.; Wirth, R.; Wirth, B.; Zerres, K. (2008). "Congenital heart disease is a feature of severe infantile spinal muscular atrophy". Journal of Medical Genetics. 45 (10): 635–638. doi:10.1136/jmg.2008.057950. PMID 18662980.
  22. Heier, C. R.; Satta, R.; Lutz, C.; Didonato, C. J. (2010). "Arrhythmia and cardiac defects are a feature of spinal muscular atrophy model mice". Human Molecular Genetics. 19 (20): 3906–3918. doi:10.1093/hmg/ddq330. PMC 2947406Freely accessible. PMID 20693262.
  23. Shababi, M.; Habibi, J.; Yang, H. T.; Vale, S. M.; Sewell, W. A.; Lorson, C. L. (2010). "Cardiac defects contribute to the pathology of spinal muscular atrophy models". Human Molecular Genetics. 19 (20): 4059–4071. doi:10.1093/hmg/ddq329. PMID 20696672.
  24. Bevan, A. K.; Hutchinson, K. R.; Foust, K. D.; Braun, L.; McGovern, V. L.; Schmelzer, L.; Ward, J. G.; Petruska, J. C.; Lucchesi, P. A.; Burghes, A. H. M.; Kaspar, B. K. (2010). "Early heart failure in the SMNΔ7 model of spinal muscular atrophy and correction by postnatal scAAV9-SMN delivery". Human Molecular Genetics. 19 (20): 3895–3905. doi:10.1093/hmg/ddq300. PMC 2947399Freely accessible. PMID 20639395.
  25. Von Gontard, A.; Zerres, K.; Backes, M.; Laufersweiler-Plass, C.; Wendland, C.; Melchers, P.; Lehmkuhl, G.; Rudnik-Schöneborn, S. (2002). "Intelligence and cognitive function in children and adolescents with spinal muscular atrophy". Neuromuscular Disorders. 12 (2): 130–136. doi:10.1016/S0960-8966(01)00274-7. PMID 11738354.
  26. Billard, C.; Gillet, P.; Signoret, J. L.; Uicaut, E.; Bertrand, P.; Fardeau, M.; Barthez-Carpentier, M. A.; Santini, J. J. (1992). "Cognitive functions in duchenne muscular dystrophy: A reappraisal and comparison with spinal muscular atrophy". Neuromuscular Disorders. 2 (5–6): 371–378. doi:10.1016/S0960-8966(06)80008-8. PMID 1300185.
  27. Laufersweiler-Plass, C.; Rudnik-Schöneborn, S.; Zerres, K.; Backes, M.; Lehmkuhl, G.; Von Gontard, A. (2002). "Behavioural problems in children and adolescents with spinal muscular atrophy and their siblings". Developmental Medicine & Child Neurology. 45. doi:10.1017/S0012162203000082.
  28. De Oliveira, C. M.; Araújo, A. P. D. Q. C. (2011). "Self-reported quality of life has no correlation with functional status in children and adolescents with spinal muscular atrophy". European Journal of Paediatric Neurology. 15 (1): 36–39. doi:10.1016/j.ejpn.2010.07.003. PMID 20800519.
  29. Yuan, N.; Wang, C. H.; Trela, A.; Albanese, C. T. (2007). "Laparoscopic Nissen Fundoplication During Gastrostomy Tube Placement and Noninvasive Ventilation May Improve Survival in Type I and Severe Type II Spinal Muscular Atrophy". Journal of Child Neurology. 22 (6): 727–731. doi:10.1177/0883073807304009. PMID 17641258.
  30. Bach, J. R. (2007). "Medical Considerations of Long-Term Survival of Werdnig–Hoffmann Disease". American Journal of Physical Medicine & Rehabilitation. 86 (5): 349–55. doi:10.1097/PHM.0b013e31804b1d66. PMID 17449979.
  31. Oskoui, M; Levy, G; Garland, C. J.; Gray, J. M.; O'Hagen, J; De Vivo, D. C.; Kaufmann, P (2007). "The changing natural history of spinal muscular atrophy type 1". Neurology. 69 (20): 1931–6. doi:10.1212/01.wnl.0000290830.40544.b9. PMID 17998484.
  32. "AveXis Reports Data from Ongoing Phase 1 Trial of AVXS-101 in Spinal Muscular Atrophy Type 1".
  33. "Biogen and Ionis Pharmaceuticals Provide Important Update on First Ever SMA Regulatory Filings". CureSMA. 1 August 2016.
  34. Zhang, M. L.; Lorson, C. L.; Androphy, E. J.; Zhou, J. (2001). "An in vivo reporter system for measuring increased inclusion of exon 7 in SMN2 mRNA: Potential therapy of SMA". Gene Therapy. 8 (20): 1532–1538. doi:10.1038/sj.gt.3301550. PMID 11704813.
  35. Andreassi, C.; Jarecki, J.; Zhou, J.; Coovert, D. D.; Monani, U. R.; Chen, X.; Whitney, M.; Pollok, B.; Zhang, M.; Androphy, E.; Burghes, A. H. (2001). "Aclarubicin treatment restores SMN levels to cells derived from type I spinal muscular atrophy patients". Human Molecular Genetics. 10 (24): 2841–2849. doi:10.1093/hmg/10.24.2841. PMID 11734549.
  36. Zhou, Haiyan; Meng, Jinhong; Marrosu, Elena; Janghra, Narinder; Morgan, Jennifer; Muntoni, Francesco (2015). "Repeated low doses of morpholino antisense oligomer: An intermediate mouse model of spinal muscular atrophy to explore the window of therapeutic response". Human Molecular Genetics. 24 (22): 6265–77; 6265. doi:10.1093/hmg/ddv329. PMC 4614699Freely accessible. PMID 26264577.
  37. Angelozzi, C.; Borgo, F.; Tiziano, F. D.; Martella, A.; Neri, G.; Brahe, C. (2007). "Salbutamol increases SMN mRNA and protein levels in spinal muscular atrophy cells". Journal of Medical Genetics. 45 (1): 29–31. doi:10.1136/jmg.2007.051177. PMID 17932121.
  38. Pane, M.; Staccioli, S.; Messina, S.; d'Amico, A.; Pelliccioni, M.; Mazzone, E. S.; Cuttini, M.; Alfieri, P.; Battini, R.; Main, M.; Muntoni, F.; Bertini, E.; Villanova, M.; Mercuri, E. (2008). "Daily salbutamol in young patients with SMA type II". Neuromuscular Disorders. 18 (7): 536–540. doi:10.1016/j.nmd.2008.05.004. PMID 18579379.
  39. Tiziano, F. D.; Lomastro, R.; Pinto, A. M.; Messina, S.; d'Amico, A.; Fiori, S.; Angelozzi, C.; Pane, M.; Mercuri, E.; Bertini, E.; Neri, G.; Brahe, C. (2010). "Salbutamol increases survival motor neuron (SMN) transcript levels in leucocytes of spinal muscular atrophy (SMA) patients: Relevance for clinical trial design". Journal of Medical Genetics. 47 (12): 856–858. doi:10.1136/jmg.2010.080366. PMID 20837492.
  40. Morandi, L. (2013). "P.6.4 Salbutamol tolerability and efficacy in adult type III SMA patients: Results of a multicentric, molecular and clinical, double-blind, placebo-controlled study". Neuromuscular Disorders. 23 (9–10): 771. doi:10.1016/j.nmd.2013.06.475.
  41. Chang, J. -G.; Hsieh-Li, H. -M.; Jong, Y. -J.; Wang, N. M.; Tsai, C. -H.; Li, H. (2001). "Treatment of spinal muscular atrophy by sodium butyrate". Proceedings of the National Academy of Sciences. 98 (17): 9808–9813. Bibcode:2001PNAS...98.9808C. doi:10.1073/pnas.171105098.
  42. Andreassi, C.; Angelozzi, C.; Tiziano, F. D.; Vitali, T.; De Vincenzi, E.; Boninsegna, A.; Villanova, M.; Bertini, E.; Pini, A.; Neri, G.; Brahe, C. (2003). "Phenylbutyrate increases SMN expression in vitro: Relevance for treatment of spinal muscular atrophy". European Journal of Human Genetics. 12 (1): 59–65. doi:10.1038/sj.ejhg.5201102. PMID 14560316.
  43. Brahe, C.; Vitali, T.; Tiziano, F. D.; Angelozzi, C.; Pinto, A. M.; Borgo, F.; Moscato, U.; Bertini, E.; Mercuri, E.; Neri, G. (2004). "Phenylbutyrate increases SMN gene expression in spinal muscular atrophy patients". European Journal of Human Genetics. 13 (2): 256–259. doi:10.1038/sj.ejhg.5201320. PMID 15523494.
  44. Mercuri, E.; Bertini, E.; Messina, S.; Solari, A.; d'Amico, A.; Angelozzi, C.; Battini, R.; Berardinelli, A.; Boffi, P.; Bruno, C.; Cini, C.; Colitto, F.; Kinali, M.; Minetti, C.; Mongini, T.; Morandi, L.; Neri, G.; Orcesi, S.; Pane, M.; Pelliccioni, M.; Pini, A.; Tiziano, F. D.; Villanova, M.; Vita, G.; Brahe, C. (2007). "Randomized, double-blind, placebo-controlled trial of phenylbutyrate in spinal muscular atrophy". Neurology. 68 (1): 51–55. doi:10.1212/01.wnl.0000249142.82285.d6. PMID 17082463.
  45. "Study to Evaluate Sodium Phenylbutyrate in Pre-symptomatic Infants With Spinal Muscular Atrophy (STOPSMA)". Retrieved 28 December 2011.
  46. Brichta, L.; Hofmann, Y.; Hahnen, E.; Siebzehnrubl, F. A.; Raschke, H.; Blumcke, I.; Eyupoglu, I. Y.; Wirth, B. (2003). "Valproic acid increases the SMN2 protein level: A well-known drug as a potential therapy for spinal muscular atrophy". Human Molecular Genetics. 12 (19): 2481–2489. doi:10.1093/hmg/ddg256. PMID 12915451.
  47. Tsai, L. K.; Tsai, M. S.; Ting, C. H.; Li, H. (2008). "Multiple therapeutic effects of valproic acid in spinal muscular atrophy model mice". Journal of Molecular Medicine. 86 (11): 1243–1254. doi:10.1007/s00109-008-0388-1. PMID 18649067.
  48. Swoboda, K. J.; Scott, C. B.; Crawford, T. O.; Simard, L. R.; Reyna, S. P.; Krosschell, K. J.; Acsadi, G.; Elsheik, B.; Schroth, M. K.; d'Anjou, G.; Lasalle, B.; Prior, T. W.; Sorenson, S. L.; MacZulski, J. A.; Bromberg, M. B.; Chan, G. M.; Kissel, J. T.; Project Cure Spinal Muscular Atrophy Investigators Network (2010). Boutron, Isabelle, ed. "SMA CARNI-VAL Trial Part I: Double-Blind, Randomized, Placebo-Controlled Trial of L-Carnitine and Valproic Acid in Spinal Muscular Atrophy". PLoS ONE. 5 (8): e12140. Bibcode:2010PLoSO...512140S. doi:10.1371/journal.pone.0012140. PMC 2924376Freely accessible. PMID 20808854.
  49. Kissel, J. T.; Scott, C. B.; Reyna, S. P.; Crawford, T. O.; Simard, L. R.; Krosschell, K. J.; Acsadi, G.; Elsheik, B.; Schroth, M. K.; d'Anjou, G.; Lasalle, B.; Prior, T. W.; Sorenson, S.; MacZulski, J. A.; Bromberg, M. B.; Chan, G. M.; Swoboda, K. J.; Project Cure Spinal Muscular Atrophy Investigators' Network (2011). Feany, Mel B., ed. "SMA CARNI-VAL TRIAL PART II: A Prospective, Single-Armed Trial of L-Carnitine and Valproic Acid in Ambulatory Children with Spinal Muscular Atrophy". PLoS ONE. 6 (7): e21296. Bibcode:2011PLoSO...621296K. doi:10.1371/journal.pone.0021296. PMC 3130730Freely accessible. PMID 21754985.
  50. Darbar, I. A.; Plaggert, P. G.; Resende, M. B. D.; Zanoteli, E.; Reed, U. C. (2011). "Evaluation of muscle strength and motor abilities in children with type II and III spinal muscle atrophy treated with valproic acid". BMC Neurology. 11: 36. doi:10.1186/1471-2377-11-36. PMC 3078847Freely accessible. PMID 21435220.
  51. Rak, K.; Lechner, B. D.; Schneider, C.; Drexl, H.; Sendtner, M.; Jablonka, S. (2009). "Valproic acid blocks excitability in SMA type I mouse motor neurons". Neurobiology of Disease. 36 (3): 477–487. doi:10.1016/j.nbd.2009.08.014. PMID 19733665.
  52. Grzeschik, S. M.; Ganta, M.; Prior, T. W.; Heavlin, W. D.; Wang, C. H. (2010). "Hydroxyurea enhances SMN2 gene expression in spinal muscular atrophy cells". Annals of Neurology. 58 (2): 194–202. doi:10.1002/ana.20548. PMID 16049920.
  53. Chen, T. - H.; Chang, J. - G.; Yang, Y. - H.; Mai, H. - H.; Liang, W. - C.; Wu, Y. - C.; Wang, H. - Y.; Huang, Y. - B.; Wu, S. - M.; Chen, Y. - C.; Yang, S. - N.; Jong, Y. - J. (2010). "Randomized, double-blind, placebo-controlled trial of hydroxyurea in spinal muscular atrophy". Neurology. 75 (24): 2190–2197. doi:10.1212/WNL.0b013e3182020332. PMID 21172842.
  54. Evans, M. C.; Cherry, J. J.; Androphy, E. J. (2011). "Differential regulation of the SMN2 gene by individual HDAC proteins". Biochemical and Biophysical Research Communications. 414 (1): 25–30. doi:10.1016/j.bbrc.2011.09.011. PMID 21925145.
  55. Riessland, M.; Brichta, L.; Hahnen, E.; Wirth, B. (2006). "The benzamide M344, a novel histone deacetylase inhibitor, significantly increases SMN2 RNA/protein levels in spinal muscular atrophy cells". Human Genetics. 120 (1): 101–110. doi:10.1007/s00439-006-0186-1. PMID 16724231.
  56. Garbes, L.; Riessland, M.; Hölker, I.; Heller, R.; Hauke, J.; Tränkle, C.; Coras, R.; Blümcke, I.; Hahnen, E.; Wirth, B. (2009). "LBH589 induces up to 10-fold SMN protein levels by several independent mechanisms and is effective even in cells from SMA patients non-responsive to valproate". Human Molecular Genetics. 18 (19): 3645–3658. doi:10.1093/hmg/ddp313. PMID 19584083.
  57. Narver, H. L.; Kong, L.; Burnett, B. G.; Choe, D. W.; Bosch-Marcé, M.; Taye, A. A.; Eckhaus, M. A.; Sumner, C. J. (2008). "Sustained improvement of spinal muscular atrophy mice treated with trichostatin a plus nutrition". Annals of Neurology. 64 (4): 465–470. doi:10.1002/ana.21449. PMID 18661558.
  58. Avila, A. M.; Burnett, B. G.; Taye, A. A.; Gabanella, F.; Knight, M. A.; Hartenstein, P.; Cizman, Z.; Di Prospero, N. A.; Pellizzoni, L.; Fischbeck, K. H.; Sumner, C. J. (2007). "Trichostatin a increases SMN expression and survival in a mouse model of spinal muscular atrophy". Journal of Clinical Investigation. 117 (3): 659–671. doi:10.1172/JCI29562. PMC 1797603Freely accessible. PMID 17318264.
  59. Riessland, M.; Ackermann, B.; Förster, A.; Jakubik, M.; Hauke, J.; Garbes, L.; Fritzsche, I.; Mende, Y.; Blumcke, I.; Hahnen, E.; Wirth, B. (2010). "SAHA ameliorates the SMA phenotype in two mouse models for spinal muscular atrophy". Human Molecular Genetics. 19 (8): 1492–1506. doi:10.1093/hmg/ddq023. PMID 20097677.
  60. Farooq, F.; Molina, F. A. A.; Hadwen, J.; MacKenzie, D.; Witherspoon, L.; Osmond, M.; Holcik, M.; MacKenzie, A. (2011). "Prolactin increases SMN expression and survival in a mouse model of severe spinal muscular atrophy via the STAT5 pathway". Journal of Clinical Investigation. 121 (8): 3042–3050. doi:10.1172/JCI46276. PMC 3148738Freely accessible. PMID 21785216.
  61. Sakla, M. S.; Lorson, C. L. (2007). "Induction of full-length survival motor neuron by polyphenol botanical compounds". Human Genetics. 122 (6): 635–643. doi:10.1007/s00439-007-0441-0. PMID 17962980.
  62. Dayangaç-Erden, D.; Bora, G.; Ayhan, P.; Kocaefe, Ç.; Dalkara, S.; Yelekçi, K.; Demir, A. S.; Erdem-Yurter, H. (2009). "Histone Deacetylase Inhibition Activity and Molecular Docking of (E)-Resveratrol: Its Therapeutic Potential in Spinal Muscular Atrophy". Chemical Biology & Drug Design. 73 (3): 355–364. doi:10.1111/j.1747-0285.2009.00781.x.
  63. Burnett, B. G.; Munoz, E.; Tandon, A.; Kwon, D. Y.; Sumner, C. J.; Fischbeck, K. H. (2008). "Regulation of SMN Protein Stability". Molecular and Cellular Biology. 29 (5): 1107–1115. doi:10.1128/MCB.01262-08. PMC 2643817Freely accessible. PMID 19103745.
  64. Mattis, V. B.; Rai, R.; Wang, J.; Chang, C. W. T.; Coady, T.; Lorson, C. L. (2006). "Novel aminoglycosides increase SMN levels in spinal muscular atrophy fibroblasts". Human Genetics. 120 (4): 589–601. doi:10.1007/s00439-006-0245-7. PMID 16951947.
  65. Mattis, V. B.; Fosso, M. Y.; Chang, C. W.; Lorson, C. L. (2009). "Subcutaneous administration of TC007 reduces disease severity in an animal model of SMA". BMC Neuroscience. 10: 142. doi:10.1186/1471-2202-10-142. PMC 2789732Freely accessible. PMID 19948047.
  66. Lunn, M. R.; Root, D. E.; Martino, A. M.; Flaherty, S. P.; Kelley, B. P.; Coovert, D. D.; Burghes, A. H.; Thi Man, N.; Morris, G. E.; Zhou, J.; Androphy, E. J.; Sumner, C. J.; Stockwell, B. R. (2004). "Indoprofen Upregulates the Survival Motor Neuron Protein through a Cyclooxygenase-Independent Mechanism". Chemistry & Biology. 11 (11): 1489–1493. doi:10.1016/j.chembiol.2004.08.024. PMC 3160629Freely accessible. PMID 15555999.
  67. Haddad, Hafedh; Cifuentes-Diaz, Carmen; Miroglio, Audrey; Roblot, Natacha; Joshi, Vandana; Melki, Judith (2003). "Riluzole attenuates spinal muscular atrophy disease progression in a mouse model". Muscle & Nerve. 28 (4): 432. doi:10.1002/mus.10455.
  68. Dimitriadi, M.; Kye, M. J.; Kalloo, G.; Yersak, J. M.; Sahin, M.; Hart, A. C. (2013). "The Neuroprotective Drug Riluzole Acts via Small Conductance Ca2+-Activated K+ Channels to Ameliorate Defects in Spinal Muscular Atrophy Models". Journal of Neuroscience. 33 (15): 6557–62, p. 6557. doi:10.1523/JNEUROSCI.1536-12.2013. PMC 3652322Freely accessible. PMID 23575853.
  69. "Study to Evaluate the Efficacy of Riluzole in Children and Young Adults With Spinal Muscular Atrophy (SMA)". ClinicalTrials.gov. Retrieved 2016-05-20.
  70. Takeuchi, Y.; Miyanomae, Y.; Komatsu, H.; Oomizono, Y.; Nishimura, A.; Okano, S.; Nishiki, T.; Sawada, T. (1994). "Efficacy of Thyrotropin-Releasing Hormone in the Treatment of Spinal Muscular Atrophy". Journal of Child Neurology. 9 (3): 287–289. doi:10.1177/088307389400900313. PMID 7930408.
  71. Tzeng, A. C.; Cheng, J.; Fryczynski, H.; Niranjan, V.; Stitik, T.; Sial, A.; Takeuchi, Y.; Foye, P.; Deprince, M.; Bach, J. R. (2000). "A study of thyrotropin-releasing hormone for the treatment of spinal muscular atrophy: A preliminary report". American Journal of Physical Medicine & Rehabilitation. 79 (5): 435–440. doi:10.1097/00002060-200009000-00005. PMID 10994885.
  72. Kato, Z.; Okuda, M.; Okumura, Y.; Arai, T.; Teramoto, T.; Nishimura, M.; Kaneko, H.; Kondo, N. (2009). "Oral Administration of the Thyrotropin-Releasing Hormone (TRH) Analogue, Taltireline Hydrate, in Spinal Muscular Atrophy". Journal of Child Neurology. 24 (8): 1010–1012. doi:10.1177/0883073809333535. PMID 19666885.
  73. Bosboom, W. M.; Vrancken, A. F. E.; Van Den Berg, L. H.; Wokke, J. H.; Iannaccone, S. T. (2009). Bosboom, Wendy MJ, ed. "Drug treatment for spinal muscular atrophy type I". The Cochrane Library. doi:10.1002/14651858.CD006281.pub2.
  74. Nizzardo, M.; Nardini, M.; Ronchi, D.; Salani, S.; Donadoni, C.; Fortunato, F.; Colciago, G.; Falcone, M.; Simone, C.; Riboldi, G.; Govoni, A.; Bresolin, N.; Comi, G. P.; Corti, S. (2011). "Beta-lactam antibiotic offers neuroprotection in a spinal muscular atrophy model by multiple mechanisms". Experimental Neurology. 229 (2): 214–225. doi:10.1016/j.expneurol.2011.01.017. PMID 21295027.
  75. Hedlund, E. (2011). "The protective effects of beta-lactam antibiotics in motor neuron disorders". Experimental Neurology. 231 (1): 14–18. doi:10.1016/j.expneurol.2011.06.002. PMID 21693120.
  76. Rose, F. F.; Mattis, V. B.; Rindt, H.; Lorson, C. L. (2009). "Delivery of recombinant follistatin lessens disease severity in a mouse model of spinal muscular atrophy". Human Molecular Genetics. 18 (6): 997–1005. doi:10.1093/hmg/ddn426. PMC 2649020Freely accessible. PMID 19074460.
  77. "CK-2127107".
  78. Carrozzi, Marco; Amaddeo, Alessandro; Biondi, Andrea; Zanus, Caterina; Monti, Fabrizio; Alessandro, Ventura (2012). "Stem cells in severe infantile spinal muscular atrophy (SMA1)". Neuromuscular Disorders. 22 (11): 1032. doi:10.1016/j.nmd.2012.09.005.
  79. Mercuri, Eugenio; Bertini, Enrico (2012). "Stem cells in severe infantile spinal muscular atrophy". Neuromuscular Disorders. 22 (12): 1105. doi:10.1016/j.nmd.2012.11.001.
  80. "Use of unregulated stem-cell based medicinal products". The Lancet. 376 (9740): 514. 2010. doi:10.1016/S0140-6736(10)61249-4.
  81. European Medicines Agency (16 April 2010). "Concerns over unregulated medicinal products containing stem cells" (PDF). European Medicines Agency.
  82. "National registries for DMD, SMA and DM". Archived from the original on 22 January 2011.

Further reading

This article is issued from Wikipedia - version of the 11/29/2016. The text is available under the Creative Commons Attribution/Share Alike but additional terms may apply for the media files.