Вы находитесь на странице: 1из 9

Journal of Neurology (2018) 265:1717–1725

https://doi.org/10.1007/s00415-018-8856-1

NEUROLOGICAL UPDATE

Update on muscle disease


J. Witherick1 · S. Brady1

Received: 8 March 2018 / Accepted: 30 March 2018 / Published online: 18 April 2018
© Springer-Verlag GmbH Germany, part of Springer Nature 2018

Abstract
In this article, we highlight some of the most important developments from the last few years in the field of muscle diseases,
including new additions to the congenital myasthenic syndromes (CMS) and limb-girdle muscular dystrophies (LGMD),
advances in our understanding of the pathophysiology of certain muscle disorders and progress in diagnostics and thera-
peutics. Unsurprisingly, the most prominent developments have come from the field of genetics, with significant advances
in diagnosis and gene therapy giving hope to those with hitherto untreatable conditions.

Keywords  Myopathy · Inclusion body myositis · Myasthenia gravis · Limb-girdle muscular dystrophy · Duchenne muscular
dystrophy · Spinal muscular atrophy

Aetiology published the findings of a large molecular genetic study of


181 IBM patients [21]. They discovered the presence of rare
Inclusion body myositis missense variants in the autophagy-related genes SQSTM1
(p62) and VCP (which encodes an ATPase associated with
Inclusion body myositis (IBM) is the commonest acquired various cellular activities) in some patients, suggesting a
myopathy in those over 50 years of age. Previously, definite possible genetic susceptibility to IBM. Reconciling the
diagnosis was based on the presence of certain pathological inflammatory and degenerative doctrine of IBM, Benven-
changes identified on muscle biopsy, in particular an auto- iste et al. suggested that there may be an initial inflamma-
aggressive endomysial inflammatory infiltrate as evidenced tory event which overloads protein degradation systems in
by invasion of intact muscle fibres, the presence of rimmed genetically susceptible individuals, leading to protein depo-
vacuoles, and amyloid-positive inclusions or 15–18  nm sition within the muscles and initiating a cyclical myopathic
tubulofilaments identified using electron microscopy. More process [4].
recently, it was shown that mitochondrial changes and pro- Although it is now clear that IBM bears little clinical sim-
tein aggregates (particularly autophagy-associated protein— ilarity to other inflammatory myopathies, it was originally
p62) are, respectively, sensitive and specific pathological classified as an idiopathic inflammatory myopathy because
features [8]. Increasing knowledge of the pathological fea- of the pathological features observed. The inflammatory ele-
tures of IBM and its characteristic pattern of weakness at ment of IBM was further informed by molecular profiling
presentation (finger flexion and knee extension weakness) [24] and microarray analysis [23] of muscle tissue, including
led to the development of the most sensitive criteria to date, the identification of B-cell immunoglobulin transcripts in
the 2011 European Neuromuscular Centre (ENMC) diag- muscle tissue indicating antigen-mediated B-cell activity.
nostic criteria [45]. While pathological findings such as pro- In 2011, Greenberg et al. identified a 43 kDa muscle protein
tein aggregation and mitochondrial abnormalities suggest targeted by circulating antibodies in 52% of patients with
that autophagy may play a role in IBM, the aetiology and IBM [46]. The target antigen was subsequently identified as
pathogenesis of the condition remain uncertain, limiting the cytosolic 5′-nucleotidase 1A (cN1A) in contemporaneous
potential for therapeutic intervention. Gang et al. recently papers published in Annals of Neurology by Pluk et al. [42]
and Larman et al. [31].
Larman et al. utilised mass spectrometry of human mus-
* J. Witherick
Jonathan.Witherick@nbt.nhs.uk
cle lysate reactive with serum from a patient with IBM to
identify potential candidate autoantigens [31]. These were
1
Southmead Hospital, Bristol, UK

13
Vol.:(0123456789)

1718 Journal of Neurology (2018) 265:1717–1725

then cross-referenced with peptides that were enriched Using exome sequencing, Bohm et al. identified a het-
in whole exome sequencing of patients with IBM. cN1A erozygous missense mutation in the gene encoding stromal
was the only candidate protein within the human proteome interaction molecule 1 (STIM1) in four families affected by
homologous to the isolated peptide. Furthermore, cN1A was an autosomal dominant TAM [11]. STIM1 plays a key role
one of seven antigens isolated by mass spectrometry. West- in the activation of ORAI1, the pore-forming unit of C ­ a2+
2+
ern blotting confirmed reactivity of sera from patients with release-activated ­Ca (CRAC) channels. Analysis of cyto-
IBM with recombinant cN1A. plasmic ­Ca2+ in transfected myoblasts using flow cytometry
In a parallel study, Pluk et al. identified a Mr 44,000 techniques demonstrated enhanced store-operated ­Ca2+ entry
polypeptide (muscle protein 44—Mup44) by immunoblot- (SOCE), indicative of a gain of function as predicted by the
ting sera from patients with IBM [42]. Analysis using liquid missense mutation. Affected patients displayed predomi-
chromatography–mass spectrometry indicated that the pep- nantly proximal lower limb weakness, variable ophthalmo-
tide was analogous to cN1A. The findings were confirmed paresis without ptosis, and increased plasma creatine kinase
with consequent immunoprecipitation of sera from patients (CK) levels (4-27 × normal). It was later discovered that
with IBM with recombinant cN1A. gain-of-function mutations in STIM1 were also responsible
The development of a reliable serological marker for IBM for the rare Stormorken and York platelet syndromes, in
would alter the current investigative strategies employed in which there was platelet dysfunction manifesting as throm-
suspected cases of IBM. The most recent diagnostic crite- bocytopenia and bleeding diathesis, miosis, ophthalmople-
ria put forward—the 2011 European Neuromuscular Centre gia without ptosis, and TAM [33, 37, 38]. Gain-of-function
criteria [45]—are the most sensitive criteria to date, with mutations in ORAI1 have also been identified as a cause
a specificity of close to 100%. Diagnostic performance of Stormorken-like syndrome with TAM and miosis, but
of cN1A antibody detection is limited by area under the without haematological abnormalities [38]. Loss of function
curve parameters in which increasing sensitivity is com- mutations in both STIM1 and ORAI1 are associated with
promised by declining specificity. A specificity of 95% can reduced muscle strength and endurance, delayed motor mile-
be achieved, but with a sensitivity of approximately 33%. stones, and severe combined immunodeficiency syndrome
One study improved the sensitivity to 76% using multiple [18, 41].
immunoglobulin isotypes [22]. A further potentially limit- The effects of loss-of and gain-of function mutations in
ing factor is the identification of cN1A antibodies in other STIM1 and ORAI1 provide insights into the key role of cal-
conditions including Sjögren’s syndrome (36% of cases) and cium physiology and the central nature of CRAC channels
systemic lupus erythematous (20% of cases) [27]. Further in cell function and diseases of muscle and of the immune
research is required before antibody testing for IBM can be system. The identification of pupillary abnormalities in
incorporated into diagnostic criteria [28]. patients with TAM is a useful clinical indicator to guide
genetic testing.

Tubular aggregate myopathy Congenital myasthenic syndromes

Tubular aggregate myopathies (TAM) are a heterogene- Congenital myasthenic syndromes (CMS) are a clinically
ous group of disorders characterised by tubular aggregates and genetically heterogeneous group of rare inherited dis-
(TA) on muscle biopsy. TAs are densely packed areas of orders resulting from dysfunction of proteins integral to the
membranous tubules first identified in muscle biopsies from function of the neuromuscular junction (NMJ). The dis-
dyskalaemic patients [17]. Immunohistochemical and ultra- covery of new genetic causes of CMS continues to inform
structural studies have revealed that TAs are derived from our understanding of the NMJ. Determining the underlying
the sarcoplasmic reticulum [47]. TAs display a character- genotype in CMS is clinically relevant as it influences treat-
istic histochemical staining pattern, appearing bright red ment decisions.
on modified Gömöri trichrome, positive on nicotinamide Over the last few years, mutations in genes involved in
adenine dinucleotide tetrazolium reductase (NADH-TR) protein glycosylation have been identified in limb-girdle
and myoadenylate deaminase (ADD) stains, and negative pattern CMS (LG-CMS) which demonstrates a favour-
on cytochrome oxidase (COX) and succinate dehydrogenase able response to acetylcholinesterase inhibitors. The first
(SDH) stains. However, TAs are not specific for TAM and gene identified encoded glutamine-fructose-6-phosphate
can be seen in a number of conditions, for example normal transaminase 1 (GFPT1) on chromosome 2p13.3 [50].
ageing [7], alcohol- and drug-induced myopathies, and, as GFPT1 catalyses the transfer of an amino group from
described below, CMS [12]. Recently, several causes of an glutamine to fructose-6-phosphate, yielding glucosamine-
autosomal dominant TAM have been described in patients 6-phosphate and glutamate. This is the first, and rate-
with calcium channelopathies. limiting, step of the hexosamine biosynthesis pathway,

13
Journal of Neurology (2018) 265:1717–1725 1719

the major end product of which is UDP-N-acetylglucosa- Limb‑girdle muscular dystrophy


mine, a key substrate for protein glycosylation. Whilst
the precise function of GFPT1 in relation to the NMJ is The last few years have seen further additions to the growing
unknown, the resulting hypoglycosylation of key NMJ list of limb-girdle muscular dystrophies (LGMD). The typi-
proteins, including muscle-specific kinase and acetylcho- cal phenotype of LGMD is prominent wasting and weakness
line receptor (AChR) subunits, is thought to be central to of the pelvic and, less commonly, the shoulder girdle mus-
the pathogenesis of the condition. cles. Additional clinical features such as contractures, selec-
Following its initial identification, Guergueltcheva tive muscle hypertrophy and atrophy, asymmetry, pattern of
et al. published a case series describing the clinicopatho- weakness and cardiorespiratory involvement, in addition to
logical features of 24 patients with CMS due to mutations serum CK, the histopathological features on muscle biopsy,
in GFPT1 (CMS-GFPT1) [25]. Most patients developed and immunoblotting of muscle, can help direct genetic test-
symptoms in the first decade of life with fatigable limb- ing. Increasing use of genetic testing has revealed that clini-
girdle weakness and minimal muscle atrophy; however, cal phenotypes are shared by several different LGMD and,
onset was as late as the fifth decade in some patients. conversely, mutations in a single gene such as LMNA or
CMS-GFPT1 is classified as LG-CMS because of the CAV3 are associated with different phenotypes even within a
presence of prominent limb-girdle weakness with mini- single family. LGMD can be inherited in either an autosomal
mal facial and ocular involvement. CMS due to mutations dominant or recessive fashion (LGMD type 1 and LGMD
in DOK7 is also a common cause of LG-CMS. Clini- type 2, respectively); the greater prevalence of autosomal
cal features that can help differentiate these two condi- recessive LGMD was recognised not long after delineation
tions are the absence of ocular, facial, bulbar, and res- of the condition [14].
piratory muscle weakness, and a sustained benefit from LGMD1H, first described in 2010, is the most recently
acetylcholinesterase inhibition in CMS-GFPT1. Tubular discovered of the eight autosomal dominant LGMD that
aggregates (TA) were identified in 13/18 muscle biop- have so far been identified [5]. The cause of LGMD1G
sies from patients with CMS-GFPT1 and were best seen was subsequently identified as being due to a defect in the
with nicotinamide adenine dinucleotide (NADH) stain- mRNA-processing protein HNRPDL [57], the same research
ing. All patients treated with acetylcholinesterase inhibi- group having mapped the gene to 4p21 10 years previously
tors (22/24) showed a good response to treatment with a [52].
median dose of 217 mg/day, though the benefit was not Autosomal recessive (AR) LGMD are approximately nine
sustained in four patients from two pedigrees. times more commonly encountered than the dominantly
Guergueltcheva et al. found that not all patients with inherited LGMD. Seven new types of autosomal recessive
LG-CMS and TAs on muscle biopsy have mutations in LGMD (types 2R-T and 2 W-Z) have been described in the
GFPT1, suggesting additional genetic causes. Using last 5 years. Most are extremely rare and described only
whole exome and high throughput sequencing, Belaya in single families. These include LGMD2Y, caused by a
et al. identified mutations in the gene encoding dolichyl- truncating mutation in TOR1AIP1 and reported in a sister
phosphate (UDP-N-acetylglucosamine) N-acetylglucosa- and brother from a consanguineous Turkish family [17], and
minephosphotransferase 1 (DPAGT1) in CMS patients LGMD2Z, identified in four siblings in a consanguineous
with TA on muscle biopsy who were GFPT1-negative Spanish family with a homozygous missense mutation in
[3]. DPAGT1 plays an essential role in N-linked protein protein o-glucosyltransferase 1 (POGLUT1) [51].
glycosylation, including of AChR subunits, and in recep- The prevalence of LGMD varies in different populations
tor export with knockout resulting in reduced end plate worldwide. The commonest autosomal recessive LGMD
AChR expression. include types 2A, 2I, and 2L. LGMD2A (calpainopathy),
The initial report of five patients with CMS due the most common LGMD globally, is caused by mutation of
to mutations in DPAGT1 demonstrated a similar age CAPN3 which encodes calpain-3, a calcium-dependent pro-
of onset to CMS-GFPT1 with fatigable limb-girdle teolytic enzyme that binds to titin and has roles in cytoskel-
weakness and an absence of facial, ocular, and bulbar etal remodelling, membrane repair and muscle regeneration
involvement. In addition to TAs, muscle biopsy revealed [44]. Over 500 different mutations have been identified in
reduced 125I-a-bungarotoxin binding sites and a fivefold the CAPN3 gene. The disease subtype is characterised by
reduction in post-synaptic folding of the NMJ, a find- variability of clinical features and rate of progression (the
ing also seen in CMS due to AChR deficiency. In keep- most rapid progression is seen in individuals with two null
ing with these observations, all patients responded to mutations). The typical clinical presentation of LGMD2A
treatment with anticholinesterase medication and two is pelvic girdle weakness affecting the posterior thigh and
patients derived additional benefit from treatment with adductor muscle accompanied by scapular winging and
3,4-diaminopyridine. contractures, and without cardiorespiratory involvement.

13

1720 Journal of Neurology (2018) 265:1717–1725

Serum CK is moderately raised but may be normal, and mus- be statin induced, statin-naïve cases were also identified.
cle biopsy may show lobulated fibres and an eosinophilic These cases were particularly noted in younger patients
inflammatory infiltrate. [59]. Further case series have also reported clear variation
LGMD2I, common in Denmark and parts of the UK, was in the strength of association with statin exposure amongst
first described in 2001 by Brockington et al. [9]. It is caused patients demonstrating HMGCR antibodies [1, 58]. Recently
by a mutation in the fukutin-related protein (FKRP) gene it has been hypothesised that cases of IMNM with HMGCR
on chromosome 19q13.3. The mutation leads to abnormal antibodies but without exposure to statin medication may
glycosylation and consequent dysfunction of α-dystroglycan, be due to dietary sources of statins such as soy proteins,
a key component of the cellular architecture in skeletal mus- certain grains, and oyster mushrooms [2]. Unfortunately, the
cle (and other tissues). Clinical features which help in iden- rarity of the condition has precluded randomised controlled
tifying LGMD2I include exercise-induced symptoms, calf trials and treatment guidance is instead based on anecdote
hypertrophy (which is seen in up to 77% of patients), quadri- and case series. From these reports, it appears that younger,
ceps femoris muscle wasting, a very high serum CK, the statin-naïve patients may be more resistant to treatment.
presence of a concomitant cardiomyopathy, and inflamma- Therapies employed to achieve clinical remission include
tory changes on muscle biopsy with reduced α-dystroglycan steroids, with the addition of one (if not two) steroid-sparing
staining. As with many other LGMD, disease onset and pro- agent, or immunomodulatory therapies such as intravenous
gression are variable and the condition may be misdiagnosed immunoglobulin, cyclophosphamide or rituximab [43, 59].
as Duchenne or Becker muscular dystrophy [49]. Despite the results of several case reports and case series, the
LGMD2L is caused by mutations in ANO5 and is thought association between HMGCR-positive IMNM and malig-
to represent 10–20% of all undiagnosed LGMD pedigrees nancy remains uncertain.
[40]. More than 20 mutations have so far been identified in
the ANO5 gene on chromosome 11 which encodes anoc-
tamin 5, an integral intracellular transmembrane glycopro- Diagnostics
tein with a central role in myogenesis. Patients can present
with prominent wasting and weakness of the quadriceps Imaging
femoris muscles, a pseudometabolic syndrome, asympto-
matic hyperCKaemia, or distal myopathy. Serum CK can The use of muscle imaging, particularly MRI, in the diag-
be significantly elevated and muscle biopsy may reveal nosis and monitoring of both acquired and inherited muscle
deposition of amyloid. Several studies have shown a male diseases continues to increase. Imaging studies allow visu-
predominance [48, 56]. alisation of pathological changes previously only identifiable
following histological analysis, including oedema—a surro-
Immune‑mediated necrotising myopathy associated gate marker of inflammation, replacement of muscle tissue
with HMGCR antibodies with fat or connective tissue, and muscle atrophy. The use of
conventional sequences, T1 and short-tau inversion recov-
The immune-mediated necrotising myopathies (IMNM) ery (STIR), is well established. Novel sequences including
are characterised histologically by prominent muscle fibre diffusion tensor imaging (DTI), tractography, and magnetic
necrosis but minimal inflammation on muscle biopsy. resonance spectroscopy will enable more detailed analysis
IMNM is associated with malignancy, statins, and anti- of the structure and metabolism of muscle tissue, potentially
bodies to signal recognition protein (SRP) and 3-hydroxy- yielding information that may support or direct diagnosis.
3-methylglutaryl-coenzyme A reductase (HMGCR). The MRI is also being used to guide muscle biopsy and has
identification of antibodies to HMGCR, the target of statin consequently been shown to improve diagnostic accuracy
medications, in patients with IMNM has been one of most in various conditions, including inflammatory myopathies
interesting developments in the last few years. [55]. The pattern of muscles affected within a limb and the
The presence of novel antibodies recognising a 100- and pattern of involvement within certain muscles can aid diag-
200-kDa protein in patients with a necrotising myopathy was nosis particularly in distal myopathies, collagen VI disor-
first reported by Stine et al. [13]. It was noted that 63% of ders, and muscular dystrophies with spinal rigidity (in which
the patients had been exposed to statins prior to the devel- MRI was shown to have a diagnostic sensitivity of 0.9 [36]).
opment of clinical symptoms. The antibodies were subse- There are, however, limitations and situations where MRI
quently established to be directed against HMGCR [32]. may be misleading. For example, although muscle oedema
However, unlike self-limiting statin-associated muscle is suggestive of an inflammatory myopathy and muscle atro-
problems which resolve on drug withdrawal, patients with phy and fatty replacement of a muscular dystrophy, muscle
HMGCR antibody-positive INMN require immunotherapy. oedema may be seen in muscular dystrophy and muscle atro-
Although initial reports suggested that the condition may phy and fatty replacement can be observed in long-standing

13
Journal of Neurology (2018) 265:1717–1725 1721

steroid-responsive inflammatory myopathy. Correct inter- provided further evidence as to the benefits of surgery over
pretation requires close correlation with clinical features and steroids alone, particularly the requirement for azathioprine
results of parallel investigations. and reduced hospital admissions for disease exacerbation.
The replacement of muscle tissue by fat is a common Whilst the study has provided further clarity, questions
feature of muscular dystrophy as well as other myopathies remain. These include the long-term benefit of thymec-
and can be readily detected using MRI [60]. Changes in tomy beyond 3 years, as prompted by the study data which
the quantitative fat fraction calculated from gradient echo showed a reduction in the benefit of thymectomy over the
sequences or MR spectroscopy has the potential to provide a duration of the study period. Whilst comparison with the
quantitative, objective, and sensitive end point in the assess- wide range of medical immunotherapies currently employed
ment of disease status or progression [10]. Given the vari- to treat myasthenia gravis would provide more meaningful
ability in disease progression in many muscular dystrophies clinical guidance, the logistics of such a study may prove
(depending on the age of the patient) and the difficulties insurmountable. The results of the study have increased the
inherent in assessing clinical metrics that require patient co- upper age limit for those in whom thymectomy can be con-
operation and motivation, such a measure has clear advan- sidered to 65 years of age. However, significant changes to
tages. Furthermore, MRI has the ability to detect subclinical clinical guidelines for the management of myasthenia gravis
changes enabling trials with smaller numbers of participants are not yet on the horizon. In the UK, the guideline’s cor-
over shorter time frames in these extremely rare diseases responding author wrote in a later editorial that ‘the advice
[20]. that thymectomy should be considered (rather than making
any stronger recommendation) remains’ and that more work
needs to be done to identify which groups would benefit
Therapeutics most from thymectomy [53]. Finally, there remains a debate
as to whether video-assisted thymectomy is as efficacious
Myasthenia gravis as open surgery. The former is associated with a lower
operative morbidity and is much more cosmetically accept-
The place of thymectomy in the management of myasthe- able meaning that many more patients may be prepared to
nia gravis (MG) has been a subject of debate ever since undergo the procedure.
the surgical removal of thymus tissue was first shown to
improve symptoms in MG by Ernst Sauerbruch in 1911. Gene therapies
Alfred Blalock was the first to demonstrate disease remission
following resection of a thymic tumour in a young woman Within the last few years, the first gene therapies for the
with myasthenic gravis [6]. Further operations on patients treatment of neuromuscular disorders have been tested and
without tumours supported Sauerbruch’s initial findings, licensed, heralding the onset of a potential paradigm shift in
but ultimately failed to establish any curative credentials. the management of previously incurable and largely untreat-
Subsequent practice has been based on clinical experience, able conditions.
anecdote and case series, incorporating increasingly sophis-
ticated surgical techniques, but with no real guidance as to Duchenne muscular dystrophy
the place surgery should take within disease management
protocols. Duchenne muscular dystrophy (DMD) is a significantly
A significant step forward in establishing the role of life-shortening, childhood-onset, X-linked condition result-
thymectomy in myasthenia gravis was the recent publication ing from mutations in the gene encoding dystrophin, a key
by Wolfe et al. of the international, multicentre, randomised protein for muscle stability and contractility. Novel thera-
control trial (the myasthenia gravis thymectomy study) peutic strategies employed in DMD have aimed to partially
comparing the effects of thymectomy and alternate-day ster- restore dystrophin protein expression by exon skipping using
oids versus alternate-day steroids alone in the treatment of antisense oligonucleotides and stop-codon suppression. An
myasthenia gravis [61]. The authors used a time-weighted alternative approach has been to upregulate the expression
approach to the primary end points—the Quantitative of utrophin, a sarcolemmal protein which mirrors many of
Myasthenia Gravis score (QMG) and the average pred- the structural characteristics of dystrophin.
nisone dose requirement—to address the inherent variabil- Eteplirsen is a phosphorodiamidate morpholino oligonu-
ity of the condition. Results over the 3-year time frame of cleotide, a DNA analogue, that restores the mRNA reading
the study demonstrated a clinically meaningful advantage frame of dystrophin, producing a functional but truncated
to thymectomy on both metrics, with patients undergoing protein in patients with exon 51-amenable deletions. Initial
thymectomy showing a meaningfully lower QMG and lower open-label studies demonstrating restoration of dystrophin
steroid requirement (32 versus 54 mg). Secondary end points expression in vivo provided proof of concept [15]. These

13

1722 Journal of Neurology (2018) 265:1717–1725

studies were followed in 2013 by publication of the first kinase 3 (GSK-3) inhibitor that has been shown to reduce
randomised control trial (RCT) [35] which demonstrated a muscle weakness and myotonia in a mouse model of DM1
23% increase in dystrophin-positive muscle fibres at the end [29]. GSK-3 suppresses CUG-binding protein 1 activity, an
of the blinded treatment phase and a further increase of up to important skeletal muscle translational regulator, by inhibit-
52% at the study’s conclusion. Clinically, a 16 m benefit in ing activation by cyclin D3-dependent kinase 4. GSK-3 plays
the 6-min timed walk (6MTW) was observed in those treated an essential role in many biological processes beyond myo-
at 24 weeks, rising to 63 m at 48 weeks despite the initiation genesis and has been linked with several diseases including
of the placebo cohort on treatment at 24 weeks. Criticisms of diabetes, inflammation, cancers, neurodegeneration, and
the study include small sample size, phenotypic heterogene- psychiatric disorders. GSK-3 inhibitors are being studied as
ity, and the chosen outcome measures. a potential treatment in a number of diseases including Alz-
The results of an open-label extension trial were pub- heimer’s disease [16], progressive supranuclear palsy [54]
lished 3 years later in 2016 [34]. The study utilised age-, and in the promotion of natural tooth repair [39].
treatment-, and genetically matched historical control data
to draw comparisons with the natural history of the condi-
tion and demonstrated a statistically significant advantage Conclusion
in the treated cohort of 151 m on 6MWT at 36 months and
a reduction in loss of ambulation. The rate of decline of There have been many advances in the field of muscle dis-
minimum and maximum inspiratory pressures, and forced eases over the last few years. We have observed improve-
vital capacity were less than that seen in prior natural history ments in the investigation and diagnosis of muscle disease
studies [26, 30] though this data was not available for the using MRI, and the widespread use of modern molecular
matched historical controls. The results of this study and the genetic techniques has helped to elucidate the genetic basis
initial RCT have supported a successful, albeit controversial of many more muscle diseases. However, further work needs
in some quarters, application for the Food and Drug Admin- to be done on our understanding of the pathogenesis of many
istration (FDA) approval in the USA. of these disorders. Arguably, the greatest advances have
been the development of treatments for previously untreat-
Spinal muscular atrophy able hereditary neuromuscular diseases. Future advances in
our understanding and management of these rare and devas-
Spinal muscular atrophy (SMA) is an autosomal recessive tating diseases continues to depend on international collabo-
condition caused by deletion or mutation in the survival ration between scientists and clinicians in the neuromuscular
motor neuron 1 (SMN1) gene, resulting in insufficient pro- community.
duction of the SMN protein. Whilst the specific role of SMN
in muscle function is unknown, its deficiency results in pro- Compliance with ethical standards 
found muscle weakness, failure to thrive, and early death.
SMA is the commonest hereditary cause of childhood mor- Conflicts of interest  JW reports no disclosures. SB received a travel
bursary from Sanofi-Genzyme in 2016.
tality. Nusinersen is an intrathecally administered uniformly
modified 2′-O-methoxyethyl phosphorothioate antisense oli-
gonucleotide that enhances production of SMN protein from
the SMN2 gene, effectively a backup to SMN1. A phase III References
study was prematurely terminated following pre-specified
1. Allenbach Y, Drouot L, Rigolet A, Charuel JL, Jouen F, Romero
interim analysis [19]. The analysis demonstrated that 41% NB, Maisonobe T, Dubourg O, Behin A, Laforet P, Stojkovic T,
(21/51) of infants in the treatment group recorded a motor Eymard B, Costedoat-Chalumeau N, Campana-Salort E, Tourna-
milestone response (according to the Hammersmith Infant dre A, Musset L, Bader-Meunier B, Kone-Paut I, Sibilia J, Servais
L, Fain O, Larroche C, Diot E, Terrier B, De Paz R, Dossier A,
Neurological Examination-2 score) versus 0% (0/27) in the
Menard D, Morati C, Roux M, Ferrer X, Martinet J, Besnard S,
placebo group. On the basis of these results, nusinersen has Bellance R, Cacoub P, Arnaud L, Grosbois B, Herson S, Boyer O,
been granted both FDA and European Medicines Agency Benveniste O, Network FM (2014) Anti-HMGCR autoantibodies
approval within the last 18 months, giving hope to the many in European patients with autoimmune necrotizing myopathies:
inconstant exposure to statin. Medicine (Baltimore) 93:150–157
patients and families affected by SMA. 2. Barbacki A, Fallavollita SA, Karamchandani J, Hudson M (2018)
Immune-mediated necrotizing myopathy and dietary sources of
Myotonic dystrophy statins. Ann Intern Med. https​://doi.org/10.7326/L17-0620
3. Belaya K, Finlayson S, Slater CR, Cossins J, Liu WW, Maxwell S,
McGowan SJ, Maslau S, Twigg SR, Walls TJ, Pascual Pascual SI,
A phase 2 study of the use of Tideglusib in myotonic dystro- Palace J, Beeson D (2012) Mutations in DPAGT1 cause a limb-
phy type 1 (DM1) is approaching completion of recruitment girdle congenital myasthenic syndrome with tubular aggregates.
in the UK. Tideglusib is an irreversible glycogen synthase Am J Hum Genet 91:193–201

13
Journal of Neurology (2018) 265:1717–1725 1723

4. Benveniste O, Stenzel W, Hilton-Jones D, Sandri M, Boyer O, van 19. Finkel RS, Mercuri E, Darras BT, Connolly AM, Kuntz NL,
Engelen BG (2015) Amyloid deposits and inflammatory infiltrates Kirschner J, Chiriboga CA, Saito K, Servais L, Tizzano E,
in sporadic inclusion body myositis: the inflammatory egg comes Topaloglu H, Tulinius M, Montes J, Glanzman AM, Bishop
before the degenerative chicken. Acta Neuropathol 129:611–624 K, Zhong ZJ, Gheuens S, Bennett CF, Schneider E, Farwell W,
5. Bisceglia L, Zoccolella S, Torraco A, Piemontese MR, Dell’Aglio De Vivo DC, Group ES (2017) Nusinersen versus Sham Con-
R, Amati A, De Bonis P, Artuso L, Copetti M, Santorelli FM, Ser- trol in Infantile-Onset Spinal Muscular Atrophy. N Engl J Med
lenga L, Zelante L, Bertini E, Petruzzella V (2010) A new locus 377:1723–1732
on 3p23–p25 for an autosomal-dominant limb-girdle muscular 20. Fischmann A, Hafner P, Fasler S, Gloor M, Bieri O, Studler U,
dystrophy, LGMD1H. Eur J Hum Genet 18:636–641 Fischer D (2012) Quantitative MRI can detect subclinical disease
6. Blalock A, Mason MF, Morgan HJ, Riven SS (1939) Myasthenia progression in muscular dystrophy. J Neurol 259:1648–1654
gravis and tumors of the thymic region: report of a case in which 21. Gang Q, Bettencourt C, Machado PM, Brady S, Holton JL, Pitt-
the tumor was removed. Ann Surg 110:544–561 man AM, Hughes D, Healy E, Parton M, Hilton-Jones D, Shieh
7. Boncompagni S, Protasi F, Franzini-Armstrong C (2012) Sequen- PB, Needham M, Liang C, Zanoteli E, de Camargo LV, De Paepe
tial stages in the age-dependent gradual formation and accumula- B, De Bleecker J, Shaibani A, Ripolone M, Violano R, Moggio
tion of tubular aggregates in fast twitch muscle fibers: SERCA and M, Barohn RJ, Dimachkie MM, Mora M, Mantegazza R, Zanotti
calsequestrin involvement. Age (Dordr) 34:27–41 S, Singleton AB, Hanna MG, Houlden H, Consortium MSGaTIIG
8. Brady S, Squier W, Sewry C, Hanna M, Hilton-Jones D, Holton (2016) Rare variants in SQSTM1 and VCP genes and risk of spo-
JL (2014) A retrospective cohort study identifying the principal radic inclusion body myositis. Neurobiol Aging 47:218.e211–218.
pathological features useful in the diagnosis of inclusion body e219
myositis. BMJ Open 4:e004552 22. Greenberg SA (2014) Cytoplasmic 5′-nucleotidase autoantibodies
9. Brockington M, Blake DJ, Prandini P, Brown SC, Torelli S, Ben- in inclusion body myositis: isotypes and diagnostic utility. Muscle
son MA, Ponting CP, Estournet B, Romero NB, Mercuri E, Voit Nerve 50:488–492
T, Sewry CA, Guicheney P, Muntoni F (2001) Mutations in the 23. Greenberg SA, Bradshaw EM, Pinkus JL, Pinkus GS, Burleson T,
fukutin-related protein gene (FKRP) cause a form of congenital Due B, Bregoli L, Bregoli LS, O’Connor KC, Amato AA (2005)
muscular dystrophy with secondary laminin alpha2 deficiency and Plasma cells in muscle in inclusion body myositis and polymy-
abnormal glycosylation of alpha-dystroglycan. Am J Hum Genet ositis. Neurology 65:1782–1787
69:1198–1209 24. Greenberg SA, Sanoudou D, Haslett JN, Kohane IS, Kunkel LM,
10. Burakiewicz J, Sinclair CDJ, Fischer D, Walter GA, Kan HE, Beggs AH, Amato AA (2002) Molecular profiles of inflammatory
Hollingsworth KG (2017) Quantifying fat replacement of myopathies. Neurology 59:1170–1182
muscle by quantitative MRI in muscular dystrophy. J Neurol 25. Guergueltcheva V, Müller JS, Dusl M, Senderek J, Oldfors A,
264(10):2053–2067 Lindbergh C, Maxwell S, Colomer J, Mallebrera CJ, Nascimento
11. Böhm J, Chevessier F, Maues De Paula A, Koch C, Attarian S, A, Vilchez JJ, Muelas N, Kirschner J, Nafissi S, Kariminejad A,
Feger C, Hantaï D, Laforêt P, Ghorab K, Vallat JM, Fardeau M, Nilipour Y, Bozorgmehr B, Najmabadi H, Rodolico C, Sieb JP,
Figarella-Branger D, Pouget J, Romero NB, Koch M, Ebel C, Schlotter B, Schoser B, Herrmann R, Voit T, Steinlein OK, Najafi
Levy N, Krahn M, Eymard B, Bartoli M, Laporte J (2013) Con- A, Urtizberea A, Soler DM, Muntoni F, Hanna MG, Chaouch A,
stitutive activation of the calcium sensor STIM1 causes tubular- Straub V, Bushby K, Palace J, Beeson D, Abicht A, Lochmüller H
aggregate myopathy. Am J Hum Genet 92:271–278 (2012) Congenital myasthenic syndrome with tubular aggregates
12. Chevessier F, Bauché-Godard S, Leroy JP, Koenig J, Paturneau- caused by GFPT1 mutations. J Neurol 259:838–850
Jouas M, Eymard B, Hantaï D, Verdière-Sahuqué M (2005) 26. Henricson EK, Abresch RT, Cnaan A, Hu F, Duong T, Arrieta
The origin of tubular aggregates in human myopathies. J Pathol A, Han J, Escolar DM, Florence JM, Clemens PR, Hoffman EP,
207:313–323 McDonald CM, Investigators C (2013) The cooperative interna-
13. Christopher-Stine L, Casciola-Rosen LA, Hong G, Chung T, tional neuromuscular research group Duchenne natural history
Corse AM, Mammen AL (2010) A novel autoantibody recogniz- study: glucocorticoid treatment preserves clinically meaningful
ing 200-kd and 100-kd proteins is associated with an immune- functional milestones and reduces rate of disease progression as
mediated necrotizing myopathy. Arthritis Rheum 62:2757–2766 measured by manual muscle testing and other commonly used
14. Chung CS, Morton NE (1959) Discrimination of genetic entities clinical trial outcome measures. Muscle Nerve 48:55–67
in muscular dystrophy. Am J Hum Genet 11:339–359 27. Herbert MK, Stammen-Vogelzangs J, Verbeek MM, Rietveld
15. Cirak S, Arechavala-Gomeza V, Guglieri M, Feng L, Torelli S, A, Lundberg IE, Chinoy H, Lamb JA, Cooper RG, Roberts M,
Anthony K, Abbs S, Garralda ME, Bourke J, Wells DJ, Dick- Badrising UA, De Bleecker JL, Machado PM, Hanna MG, Plesti-
son G, Wood MJ, Wilton SD, Straub V, Kole R, Shrewsbury SB, lova L, Vencovsky J, van Engelen BG, Pruijn GJ (2016) Disease
Sewry C, Morgan JE, Bushby K, Muntoni F (2011) Exon skipping specificity of autoantibodies to cytosolic 5′-nucleotidase 1A in
and dystrophin restoration in patients with Duchenne muscular sporadic inclusion body myositis versus known autoimmune dis-
dystrophy after systemic phosphorodiamidate morpholino oli- eases. Ann Rheum Dis 75:696–701
gomer treatment: an open-label, phase 2, dose-escalation study. 28. Hilton-Jones D, Brady S (2016) Diagnostic criteria for inclusion
Lancet 378:595–605 body myositis. J Intern Med 280:52–62
16. del Ser T, Steinwachs KC, Gertz HJ, Andrés MV, Gómez-Carrillo 29. Jones K, Wei C, Iakova P, Bugiardini E, Schneider-Gold C, Meola
B, Medina M, Vericat JA, Redondo P, Fleet D, León T (2013) G, Woodgett J, Killian J, Timchenko NA, Timchenko LT (2012)
Treatment of Alzheimer’s disease with the GSK-3 inhibitor tide- GSK3β mediates muscle pathology in myotonic dystrophy. J Clin
glusib: a pilot study. J Alzheimers Dis 33:205–215 Invest 122:4461–4472
17. Engel WK, Bishop DW, Cunningham GG (1970) Tubular aggre- 30. Khirani S, Ramirez A, Aubertin G, Boulé M, Chemouny C, Forin
gates in type II muscle fibers: ultrastructural and histochemical V, Fauroux B (2014) Respiratory muscle decline in Duchenne
correlation. J Ultrastruct Res 31:507–525 muscular dystrophy. Pediatr Pulmonol 49:473–481
18. Feske S, Gwack Y, Prakriya M, Srikanth S, Puppel SH, Tanasa B, 31. Larman HB, Salajegheh M, Nazareno R, Lam T, Sauld J, Steen
Hogan PG, Lewis RS, Daly M, Rao A (2006) A mutation in Orai1 H, Kong SW, Pinkus JL, Amato AA, Elledge SJ, Greenberg SA
causes immune deficiency by abrogating CRAC channel function. (2013) Cytosolic 5′-nucleotidase 1A autoimmunity in sporadic
Nature 441:179–185 inclusion body myositis. Ann Neurol 73:408–418

13

1724 Journal of Neurology (2018) 265:1717–1725

32. Mammen AL, Chung T, Christopher-Stine L, Rosen P, Rosen A, 45. Rose MR, Group EIW (2013) 188th ENMC international work-
Doering KR, Casciola-Rosen LA (2011) Autoantibodies against shop: inclusion body myositis, 2–4 December 2011, Naarden, The
3-hydroxy-3-methylglutaryl-coenzyme A reductase in patients Netherlands. Neuromuscul Disord 23:1044–1055
with statin-associated autoimmune myopathy. Arthritis Rheum 46. Salajegheh M, Lam T, Greenberg SA (2011) Autoantibodies
63:713–721 against a 43 KDa muscle protein in inclusion body myositis. PLoS
33. Markello T, Chen D, Kwan JY, Horkayne-Szakaly I, Morrison One 6:e20266
A, Simakova O, Maric I, Lozier J, Cullinane AR, Kilo T, Meister 47. Salviati G, Pierobon-Bormioli S, Betto R, Damiani E, Angelini
L, Pakzad K, Bone W, Chainani S, Lee E, Links A, Boerkoel C, C, Ringel SP, Salvatori S, Margreth A (1985) Tubular aggregates:
Fischer R, Toro C, White JG, Gahl WA, Gunay-Aygun M (2015) sarcoplasmic reticulum origin, calcium storage ability, and func-
York platelet syndrome is a CRAC channelopathy due to gain-of- tional implications. Muscle Nerve 8:299–306
function mutations in STIM1. Mol Genet Metab 114:474–482 48. Sarkozy A, Hicks D, Hudson J, Laval SH, Barresi R, Hilton-Jones
34. Mendell JR, Goemans N, Lowes LP, Alfano LN, Berry K, Shao D, Deschauer M, Harris E, Rufibach L, Hwang E, Bashir R, Wal-
J, Kaye EM, Mercuri E, Network ESGaTFDI (2016) Longitudi- ter MC, Krause S, van den Bergh P, Illa I, Pénisson-Besnier I, De
nal effect of eteplirsen versus historical control on ambulation in Waele L, Turnbull D, Guglieri M, Schrank B, Schoser B, Seeger
Duchenne muscular dystrophy. Ann Neurol 79:257–271 J, Schreiber H, Gläser D, Eagle M, Bailey G, Walters R, Longman
35. Mendell JR, Rodino-Klapac LR, Sahenk Z, Roush K, Bird L, C, Norwood F, Winer J, Muntoni F, Hanna M, Roberts M, Bindoff
Lowes LP, Alfano L, Gomez AM, Lewis S, Kota J, Malik V, LA, Brierley C, Cooper RG, Cottrell DA, Davies NP, Gibson A,
Shontz K, Walker CM, Flanigan KM, Corridore M, Kean JR, Gorman GS, Hammans S, Jackson AP, Khan A, Lane R, McCo-
Allen HD, Shilling C, Melia KR, Sazani P, Saoud JB, Kaye EM, nville J, McEntagart M, Al-Memar A, Nixon J, Panicker J, Parton
Group ES (2013) Eteplirsen for the treatment of Duchenne mus- M, Petty R, Price CJ, Rakowicz W, Ray P, Schapira AH, Swingler
cular dystrophy. Ann Neurol 74:637–647 R, Turner C, Wagner KR, Maddison P, Shaw PJ, Straub V, Bushby
36. Mercuri E, Clements E, Offiah A, Pichiecchio A, Vasco G, Bianco K, Lochmüller H (2013) ANO5 gene analysis in a large cohort of
F, Berardinelli A, Manzur A, Pane M, Messina S, Gualandi F, patients with anoctaminopathy: confirmation of male prevalence
Ricci E, Rutherford M, Muntoni F (2010) Muscle magnetic reso- and high occurrence of the common exon 5 gene mutation. Hum
nance imaging involvement in muscular dystrophies with rigidity Mutat 34:1111–1118
of the spine. Ann Neurol 67:201–208 49. Schwartz M, Hertz JM, Sveen ML, Vissing J (2005) LGMD2I
37. Morin G, Bruechle NO, Singh AR, Knopp C, Jedraszak G, presenting with a characteristic Duchenne or Becker muscular
Elbracht M, Brémond-Gignac D, Hartmann K, Sevestre H, Deutz dystrophy phenotype. Neurology 64:1635–1637
P, Hérent D, Nürnberg P, Roméo B, Konrad K, Mathieu-Dramard 50. Senderek J, Müller JS, Dusl M, Strom TM, Guergueltcheva V,
M, Oldenburg J, Bourges-Petit E, Shen Y, Zerres K, Ouadid-Ahi- Diepolder I, Laval SH, Maxwell S, Cossins J, Krause S, Muelas
douch H, Rochette J (2014) Gain-of-function mutation in STIM1 N, Vilchez JJ, Colomer J, Mallebrera CJ, Nascimento A, Nafissi
(P.R304W) is associated with Stormorken syndrome. Hum Mutat S, Kariminejad A, Nilipour Y, Bozorgmehr B, Najmabadi H, Rod-
35:1221–1232 olico C, Sieb JP, Steinlein OK, Schlotter B, Schoser B, Kirschner
38. Nesin V, Wiley G, Kousi M, Ong EC, Lehmann T, Nicholl DJ, J, Herrmann R, Voit T, Oldfors A, Lindbergh C, Urtizberea A, von
Suri M, Shahrizaila N, Katsanis N, Gaffney PM, Wierenga KJ, der Hagen M, Hübner A, Palace J, Bushby K, Straub V, Beeson D,
Tsiokas L (2014) Activating mutations in STIM1 and ORAI1 Abicht A, Lochmüller H (2011) Hexosamine biosynthetic pathway
cause overlapping syndromes of tubular myopathy and congenital mutations cause neuromuscular transmission defect. Am J Hum
miosis. Proc Natl Acad Sci USA 111:4197–4202 Genet 88:162–172
39. Neves VC, Babb R, Chandrasekaran D, Sharpe PT (2017) Promo- 51. Servián-Morilla E, Takeuchi H, Lee TV, Clarimon J, Mavillard F,
tion of natural tooth repair by small molecule GSK3 antagonists. Area-Gómez E, Rivas E, Nieto-González JL, Rivero MC, Cabrera-
Sci Rep 7:39654 Serrano M, Gómez-Sánchez L, Martínez-López JA, Estrada B,
40. Penttilä S, Palmio J, Suominen T, Raheem O, Evilä A, Muelas Márquez C, Morgado Y, Suárez-Calvet X, Pita G, Bigot A, Gal-
Gomez N, Tasca G, Waddell LB, Clarke NF, Barboi A, Hack- lardo E, Fernández-Chacón R, Hirano M, Haltiwanger RS, Jafar-
man P, Udd B (2012) Eight new mutations and the expanding Nejad H, Paradas C (2016) A POGLUT1 mutation causes a mus-
phenotype variability in muscular dystrophy caused by ANO5. cular dystrophy with reduced Notch signaling and satellite cell
Neurology 78:897–903 loss. EMBO Mol Med 8:1289–1309
41. Picard C, McCarl CA, Papolos A, Khalil S, Lüthy K, Hivroz C, 52. Starling A, Kok F, Passos-Bueno MR, Vainzof M, Zatz M (2004)
LeDeist F, Rieux-Laucat F, Rechavi G, Rao A, Fischer A, Feske A new form of autosomal dominant limb-girdle muscular dystro-
S (2009) STIM1 mutation associated with a syndrome of immu- phy (LGMD1G) with progressive fingers and toes flexion limita-
nodeficiency and autoimmunity. N Engl J Med 360:1971–1980 tion maps to chromosome 4p21. Eur J Hum Genet 12:1033–1040
42. Pluk H, van Hoeve BJ, van Dooren SH, Stammen-Vogelzangs J, 53. Sussman J (2016) Thymectomy: the more you know, the more you
van der Heijden A, Schelhaas HJ, Verbeek MM, Badrising UA, know you don’t know. Pract Neurol 16:426–427
Arnardottir S, Gheorghe K, Lundberg IE, Boelens WC, van Enge- 54. Tolosa E, Litvan I, Höglinger GU, Burn D, Lees A, Andrés MV,
len BG, Pruijn GJ (2013) Autoantibodies to cytosolic 5′-nucleoti- Gómez-Carrillo B, León T, Del Ser T, Investigators T (2014)
dase 1A in inclusion body myositis. Ann Neurol 73:397–407 A phase 2 trial of the GSK-3 inhibitor tideglusib in progressive
43. Ramanathan S, Langguth D, Hardy TA, Garg N, Bundell C, supranuclear palsy. Mov Disord 29:470–478
Rojana-Udomsart A, Dale RC, Robertson T, Mammen AL, Red- 55. Van De Vlekkert J, Maas M, Hoogendijk JE, De Visser M, Van
del SW (2015) Clinical course and treatment of anti-HMGCR Schaik IN (2015) Combining MRI and muscle biopsy improves
antibody-associated necrotizing autoimmune myopathy. Neurol diagnostic accuracy in subacute-onset idiopathic inflammatory
Neuroimmunol Neuroinflamm 2:e96 myopathy. Muscle Nerve 51:253–258
44. Richard I, Roudaut C, Saenz A, Pogue R, Grimbergen JE, Ander- 56. van der Kooi AJ, Ten Dam L, Frankhuizen WS, Straathof CS, van
son LV, Beley C, Cobo AM, de Diego C, Eymard B, Gallano P, Doorn PA, de Visser M, Ginjaar IB (2013) ANO5 mutations in the
Ginjaar HB, Lasa A, Pollitt C, Topaloglu H, Urtizberea JA, de Dutch limb girdle muscular dystrophy population. Neuromuscul
Visser M, van der Kooi A, Bushby K, Bakker E, Lopez de Munain Disord 23:456–460
A, Fardeau M, Beckmann JS (1999) Calpainopathy—a survey of 57. Vieira NM, Naslavsky MS, Licinio L, Kok F, Schlesinger D,
mutations and polymorphisms. Am J Hum Genet 64:1524–1540 Vainzof M, Sanchez N, Kitajima JP, Gal L, Cavaçana N, Serafini

13
Journal of Neurology (2018) 265:1717–1725 1725

PR, Chuartzman S, Vasquez C, Mimbacas A, Nigro V, Pavanello correlate with creatine kinase levels and strength in anti-3-hy-
RC, Schuldiner M, Kunkel LM, Zatz M (2014) A defect in the droxy-3-methylglutaryl-coenzyme A reductase-associated autoim-
RNA-processing protein HNRPDL causes limb-girdle muscular mune myopathy. Arthritis Rheum 64:4087–4093
dystrophy 1G (LGMD1G). Hum Mol Genet 23:4103–4110 60. Willcocks RJ, Rooney WD, Triplett WT, Forbes SC, Lott DJ,
58. Watanabe Y, Suzuki S, Nishimura H, Murata KY, Kurashige T, Senesac CR, Daniels MJ, Wang DJ, Harrington AT, Tennekoon
Ikawa M, Asahi M, Konishi H, Mitsuma S, Kawabata S, Suzuki GI, Russman BS, Finanger EL, Byrne BJ, Finkel RS, Walter GA,
N, Nishino I (2015) Statins and myotoxic effects associated with Sweeney HL, Vandenborne K (2016) Multicenter prospective
anti-3-hydroxy-3-methylglutaryl-coenzyme A reductase autoan- longitudinal study of magnetic resonance biomarkers in a large
tibodies: an observational study in Japan. Medicine (Baltimore) duchenne muscular dystrophy cohort. Ann Neurol 79:535–547
94:e416 6 1. Wolfe GI, Kaminski HJ, Cutter GR (2016) Randomized trial of
5 9. Werner JL, Christopher-Stine L, Ghazarian SR, Pak KS, Kus thymectomy in myasthenia gravis. N Engl J Med 375:2006–2007
JE, Daya NR, Lloyd TE, Mammen AL (2012) Antibody levels

13

Вам также может понравиться