You are on page 1of 10

REVIEWS

Myasthenia gravis — autoantibody


characteristics and their implications
for therapy
Nils Erik Gilhus1,2, Geir Olve Skeie1,2, Fredrik Romi1,2, Konstantinos Lazaridis3,
Paraskevi Zisimopoulou3 and Socrates Tzartos3
Abstract | Myasthenia gravis (MG) is an autoimmune disorder caused by autoantibodies that
target the neuromuscular junction, leading to muscle weakness and fatigability. Currently
available treatments for the disease include symptomatic pharmacological treatment,
immunomodulatory drugs, plasma exchange, thymectomy and supportive therapies. Different
autoantibody patterns and clinical manifestations characterize different subgroups of the
disease: early-onset MG, late-onset MG, thymoma MG, muscle-specific kinase MG, low-density
lipoprotein receptor-related protein 4 MG, seronegative MG, and ocular MG. These subtypes
differ in terms of clinical characteristics, disease pathogenesis, prognosis and response to
therapies. Patients would, therefore, benefit from treatment that is tailored to their disease
subgroup, as well as other possible disease biomarkers, such as antibodies against cytoplasmic
muscle proteins. Here, we discuss the different MG subtypes, the sensitivity and specificity of the
various antibodies involved in MG for distinguishing between these subtypes, and the value of
antibody assays in guiding optimal therapy. An understanding of these elements should be useful
in determining how to adapt existing therapies to the requirements of each patient.

Myasthenia gravis (MG) is an autoimmune disorder caused rapid improvement even when the disease is in a chronic
by antibodies targeting the neuromuscular junction. In phase, as the synthesis of postsynaptic mol­ecules is con-
MG, these antibodies bind to the postsynaptic muscle tinuously increased in MG to counteract the destruction
end-plate and attack and destroy postsynaptic molecules. of molecules by autoantibodies. However, well-­controlled
This process leads to impaired signal transduction and, therapeutic studies in MG are rare, and have not taken
consequently, muscle weakness and fatigability — the hall- into account variability between MG subgroups.
mark symptoms of MG1–4. The weakness can be focal or Recommendations for clinical management, therefore,
generalized, and usually affects ocular, bulbar and proximal rely partly on consented guidelines and expert reviews.
extremity muscles. Respiratory muscle weakness develops Autoantibodies against proteins in the postsynaptic
only rarely, but can be life-­threatening. Weakness is typi- membrane are the most important biomarkers in guid-
1
Department of Clinical
Medicine, University of
cally symmetrical, except in affected external eye muscles, ing diagnosis of MG (BOX 1) and helping to define which
Bergen, Postboks 7804, in which the weakness is usually asymmetrical1–4. MG subtype a patient has. Moreover, an improved under-
5020 Bergen, Norway. In most populations, the prevalence of MG is 150–300 standing of the autoantibody-associated pathophysiology
2
Department of Neurology, per 1,000,000 individuals, with an annual incidence of in MG could facilitate therapeutic decision-making. In
Haukeland University
more than 10 in 1,000,000 people5,6. Both prevalence and this Review, we discuss the antibodies involved in MG
Hospital, Postbox 1400,
5021 Bergen, Norway. incidence increase with age. With modern treatment, pathogenesis, sensitivity and specificity of various anti-
3
Department of Neurobiology, MG should not increase mortality7. Therapeutic options body assays, as well as the value of these assays in guiding
Hellenic Pasteur Institute, in MG include symptomatic pharmacological treat- optimal therapy.
127 Vassilissis Sofias ment, immunomodulatory pharmacological treatment,
Avenue 115 21, Ampelokipi,
Athens, Greece.
plasma exchange, thymectomy and supportive treat- Autoantibodies in myasthenia gravis
Correspondence to N.E.G. ment4,8. The treatment should be tailored to the indivi­ Antigenic targets
nils.gilhus@uib.no dual patient depending on their MG subgroup, which MG serves as a prototypical antibody-mediated dis-
doi:10.1038/nrneurol.2016.44 can be defined on the basis of biomarkers and clinical ease because the physiological function, antigenic
Published online 22 Apr 2016 characteristics. Immunosuppressive therapy can lead to targets, and immune responses to autoantibodies at the

NATURE REVIEWS | NEUROLOGY VOLUME 12 | MAY 2016 | 259


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Key points exacerbations19. Such correlations remain to be convinc-


ingly demonstrated, but repeated testing for antibodies
• The characteristic muscle weakness in myasthenia gravis (MG) is caused by antibodies can influence therapeutic decisions: an increase in anti-
directed against the neuromuscular junction body concentration is thought to indicate exacerbation of
• MG is divided into subgroups on the basis of specific antibodies, other biomarkers, MG, whereas a stable or decreasing concentration could
and clinical characteristics, such as age of onset, presence of thymoma, and indicate stable disease. However, loss of functional AChRs,
involvement of ocular muscles not the AChR antibody concentration, is what leads to the
• The most common antibodies detected in MG are antibodies against acetylcholine symptoms of MG: reduced numbers of receptors correlate
receptors (AChRs), muscle-specific kinase (MuSK) and low-density lipoprotein with disease severity, and the loss of receptors depends not
receptor-related protein 4 (LRP4)
only on total AChR-antibody concentration, but also on
• Additional antibodies of interest in MG are directed against agrin, titin, KV1.4, autoantibody pattern and non-antibody factors1–4.
ryanodine receptors, collagen Q, and cortactin
• Therapy should be tailored to the individual patient and guided by MG subgroup, and Testing. A radioimmunoprecipitation assay based on
can include symptomatic drug therapy, immunosuppressive drug therapy,
a mixture of solubilized embryonic and adult AChRs
thymectomy and/or supportive therapy
is the most rigorously validated test for AChR antibod-
• The aim of treatment should be normal or near-normal function, which in most
ies, and is the most reliable among the validated AChR
patients requires long-term immunosuppressive treatment with a drug combination
antibody tests. These testing kits are commercially avail-
that is individualized for the patient for optimal effectiveness
able. Positive test results have a near 100% specificity for
MG in symptomatic individuals1–4. Such specificity is
neuromuscular junction are well understood (FIG. 1a). crucial for a first-line screening test that is used also in
The major antigen in MG is the acetylcholine receptor patients with vague muscular symptoms or fatigue as the
(AChR) on the muscle membrane1–4. Some patients with- primary symptom.
out AChR antibodies have autoantibodies against the Although ELISA and immunofluorescence assays
muscle specific kinase (MuSK), a protein responsible for have been developed as non-radioactive alternatives
AChR clustering and maintenance of the neuromuscular with good sensitivity and specificity, they are inferior to
junction4,9. A fraction of patients with MG who do not radioimmuno­precipitation20 and have not been widely
have either AChR or MuSK antibodies have antibodies adopted in clinical practice. Cell-based assays, in which
against low-density lipoprotein receptor-related protein 4 AChR molecules are clustered on the membranes of cul-
(LRP4), a receptor for neural agrin that relays the signal tured test cells, offer better sensitivity, and enable detec-
to MuSK to initiate AChR clustering4,10. Patients without tion of low-affinity antibodies: they can detect antibodies
detectable antibodies against any of these three antigens in 4–66% of patients with generalized MG in whom anti-
are referred to as seronegative. Some patients with MG bodies cannot be detected with radioimmunoassay15,21,22.
also have antibodies against cytoplasmic muscle proteins; Antibodies that are detected with different tests all belong
although these antibodies are not pathogenic, they can to the same IgG subclasses, indicating that the aetiology
serve as disease markers11–14. and pathogenesis is the same in patients with AChR anti-
bodies that can only be detected in cell-based assays as
AChR antibodies in those with antibodies that are detected in routine tests.
Prevalence. AChR antibodies can be detected with rou- The radioimmunoassay is recommended as the first-
tine assays in 70% of all patients with MG1–4. In another line test because it has been used successfully in clin-
5–10% of patients, AChR antibodies can only be detected ical practice; moreover, it not only detects the presence
with more-sensitive, cell-based assays15,16. of AChR antibodies but can also quantify their levels.
Supplementary cell-based testing should be performed
Pathophysiological role. In vivo studies have shown that when MG is suspected but the patient is negative for
the AChR antibodies bind to extracellular domains of the anti-AChR and anti-MuSK antibodies.
receptor17 (BOX 2), thereby impairing signal transduction.
AChR antibodies mostly belong to the IgG1 and IgG3 sub- MuSK antibodies
classes, which activate the complement cascade, leading Prevalence. Antibodies against MuSK can be detected in
to damage of the postsynaptic membrane. In addition, as 1–10% of patients with MG1–4. These antibodies are pres-
the antibodies are bivalent, they are capable of antigenic ent in patients from the Mediterranean area more often
modulation. Some AChR antibodies target the acetyl­ than in those from Northern Europe, possibly owing to a
Antigenic modulation choline binding site of the receptor, potentially blocking combination of genetic and environmental factors1–4.
Bivalent antibodies can the signalling pathway; however, such antibodies are rare
cause crosslinking of and are probably clinically relevant in few patients3. Pathophysiology. In contrast with AChR antibodies, IgG
receptors and subsequent
Antibodies against the α subunit of the AChR are more antibodies against MuSK belong mostly to the IgG4 sub-
receptor internalization.
pathogenic than those against the β subunit; therefore, class. Their mode of action also differs: AChR antibodies
Epitope pattern the AChR epitope pattern influences disease severity18. bind to complement and crosslink their antigenic target,
An epitope is a localized region Total AChR antibody concentration does not correlate whereas MuSK antibodies are directly pathogenic, even
on an antigen capable of with symptom severity when patients are compared1–4, if they do not bind to complement, and remain func-
eliciting an immune response,
and the epitope pattern refers
although fluctuations in AChR antibody concentration tionally monovalent23. Instead of modulating AChR
to all epitopes involved in an in an individual patient have been reported to corre- function, MuSK antibodies reduce the postsynaptic den-
immune response. late with the severity of muscle weakness and to predict sity of AChRs and impair their alignment between the

260 | MAY 2016 | VOLUME 12 www.nature.com/nrneurol


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

motor nerve terminal and the postsynaptic membrane The fact that LRP4 antibodies are primarily reported
(FIG. 1b). Most MuSK antibodies bind to the extracellular in MG patients without AChR or MuSK antibodies sug-
N‑terminal Ig‑like domains of the AChR (BOX 3). gests that LRP4-MG is a distinct disorder26. However,
LRP4 antibodies have recently been detected in several
Testing. In patients with MuSK-MG, concentrations of patients with either AChR or MuSK antibodies, in patients
anti-MuSK antibody tend to correlate with disease sever- with other autoimmune disorders, and in patients with
ity, and changes in antibody concentration over time amyotrophic lateral sclerosis26,27,31. The presence of LRP4
can reflect disease activity24. Immunoprecipitation with antibodies is associated with milder MG symptoms, and
radioimmunoassays is the standard technique for MuSK LRP4‑MG can manifest purely as ocular MG26,27,31.
antibody detection, and ELISA tests are also available,
although neither are as sensitive as cell-based assays23,25: Testing. No commercial tests for LRP4 antibody are
a large screening study showed that a cell-based assay available. Optimal testing conditions and thresholds for
could detect MuSK antibodies in 13% of MG serum sensitivity and specificity in a clinical setting have not yet
samples that had been classified as sero­negative using been defined.
radioimmunassay or ELISA11. Some of these patients
had ocular MG. The specificity of the cell-based assay is Other antibodies
estimated to be 97–98%. Agrin antibodies. Agrin autoantibodies can be detected
MuSK‑MG and AChR‑MG are distinct disease enti- in a minority of patients with MG, either with or without
ties and rarely occur in the same patient. Nevertheless, a antibodies against AChR, MuSK or LRP4 (REFS 32,33).
few single-patient reports have demonstrated the exist- Agrin antibodies have been detected only in patients
ence of both AChR and MuSK antibodies in the same with MG, suggesting that these antibodies are specific to
patient, particularly when the most sensitive antibody the disease.
detection techniques are used16. Agrin is a heparan sulfate proteoglycan released from
the motor nerve terminal34. It regulates the formation,
LRP4 antibodies maintenance and regeneration of the neuromuscular
Prevalence. The likelihood of detecting LRP4 anti- junction34, and interference with agrin function leads to
bodies in MG depends on the assay and the examined insufficient neuromuscular transmission34. To date, no
population. LRP4 antibodies are detected in 1–5% directly pathogenic effect of agrin antibodies has been
of patients with MG of any type26,27, and in 7–33% of MG established, although such antibodies inhibit MuSK
patients without AChR and MuSK antibodies27,28. phosphorylation and AChR clustering in vitro32–34.

Pathophysiology. In LRP4‑immunized mice, LRP4 Titin antibodies. Antibodies against titin — the largest of
antibodies are directly pathogenic and induce mus- all known proteins — can be detected in 20–30% of MG
cular weakness through disruption of the inter­action patients with AChR antibodies, mostly in patients with
between LRP4 and agrin (BOX 4), and inhibition of AChR- thymoma-associated disease or late-onset MG11,35. Titin
mediated neuromuscular transmission29 (FIG 1). The dis- is located intracellularly and is essential for muscle con-
ease in the LRP4‑antibody mouse model was similar tractility36. Given its intracellular location, titin antibodies
to that seen in mice immunized with AChRs, and to should not interfere with muscle function. Nevertheless,
the muscle weakness seen in the human disease. LRP4 their presence indicates a more severe form of MG with
antibodies observed in these mice mainly belong to the mild myopathy and a need for immunosuppressive treat-
complement-binding IgG1 subclass30. ment11. Moreover, antibodies against titin are a sensitive
marker of thymoma in patients with MG whose symptom
onset occurs before the age of 50 years11,35. Most titin anti-
Box 1 | Differential diagnosis of myasthenia gravis bodies target a 30 kDa region of the protein, called myas-
thenia gravis titin‑30, that is located near the A–I junction
Myasthenia gravis (MG) needs to be distinguished from other neuromuscular disorders, in muscle, thereby potentially interfering with the role of
such as:
titin in mediating muscle elasticity37. A commercial kit is
• Genetic and toxic myasthenic syndromes available for titin antibody testing.
• Autoantibody-mediated Lambert–Eaton myasthenic syndrome
• Neuromyotonia KV1.4 antibodies. Antibodies against the α subunit of
• Miller Fisher variant of Guillain–Barré syndrome the voltage-gated K+ channel KV1.4 in skeletal muscle
In patients with muscle symptoms and a positive test for antibodies against the are detected in 10–20% of patients with MG14,38. KV1.4
acetylcholine receptor (AChR) or muscle-specific kinase (MuSK), the diagnosis of MG channels are widely expressed in the CNS, where they are
is considered established and no further tests are needed to confirm the disease. concentrated in axonal membranes or near axons, and are
In patients with no detectable antibodies, neurophysiological tests are necessary. also present in the endocardium. In MG, KV1.4 antibod-
Such tests include: ies seem to cross-react with voltage-gated K+ channels in
• Repetitive nerve stimulation67 the heart muscle14,38. In a Japanese patient cohort39, KV1.4
• Single-fibre electromyography67 antibodies were associated with severe MG and heart
The sensitivity and specificity of the diagnostic rely on the quality of the investigation. complications, but the findings could not be confirmed
The response (or lack thereof) to pharmacological treatment with anticholinesterase in European patients38. In vivo antibody binding to KV1.4
drugs also has diagnostic value. in MG has not been confirmed.

NATURE REVIEWS | NEUROLOGY VOLUME 12 | MAY 2016 | 261


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

a
Pathogenic Axonal terminal Action potential
Present in MG
(unproven pathogenicity) Agrin
Ca2+ Ca2+ Choline
Acetic acid

Acetylcholine Acetylcholine
Acetylcholinesterase
ColQ
K+
AChR
KV1.4

Muscle LRP4
Na2+ AChR MuSK MuSK
clustering
Folding
RyR
Cortactin Muscle contraction
Actin
Myosin Muscle fibre Sarcoplasmic
Titin reticulum

b Thymoma Auto-reactive T cell


Action potential
B cell activation
and auto-antibody
production Ca2+ Ca2+

Complement
activation
C1 complement Antigenic
Fold modulation
1 destruction 2 3 Blocking 4
antibody

MAC ↓Clustering
AChR
internalization ↓Folding

Cell lysis ↓Muscle contraction

AChR degradation

c Thymectomy Thymoma Azathioprine Acetylcholinesterase


Mycophenolate inhibitor
mofetil Action potential Pyridostigmine
B cell activation Methotrexate
and auto-antibody Ambenonium chloride
Cyclosporine Neostigmine
production Ca2+

Rituximab ↓Complement Ca2+


activation
Plasma exchange ↓Antigenic
immunoadsorption ↓Fold modulation
destruction

↓AChR Clustering
internalization Folding

Cell lysis
Muscle contraction

↓AChR degradation

Nature Reviews | Neurology


262 | MAY 2016 | VOLUME 12 www.nature.com/nrneurol
©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Ryanodine receptor antibodies. Antibodies against with various autoimmune disorders, including myosi-
the ryanodine receptor (RyR) are present in 70% of tis11. Any relevance to MG pathogenesis, diagnosis or
AChR-MG patients with thymoma and in 14% treatment remains to be proven.
of patients with late-onset AChR-MG8,40. The RyR is
the Ca2+ channel in sarcoplasmic reticulum12. It opens Subgroups of myasthenia gravis
upon depolarization of the sarcolemma and partic- Patients with MG can be stratified into subgroups
ipates in muscle contraction through release of cal- according to clinical presentation and biomarkers3,4.
cium from the sarcolemma into the cytoplasm12. The The criteria for these subtypes include clinical symp-
presence of anti-RyR antibodies indicates severe MG, toms, age of onset, thymic pathology and — most
though the pathogenic role of these antibodies has not importantly — autoantibodies (TABLE 1). Each patient
been established12. should be assigned to only one subgroup. In the fol-
lowing discussion, we have excluded neonatal MG
Collagen Q antibodies. Collagen Q is a protein that and arthrogryposis, as these disorders are usually not
concentrates and anchors acetylcholinesterase at the regarded as autoimmune MG4.
neuromuscular junction, where it is localized in the
extracellular matrix and is, thus, accessible to anti- Autoantibody-based subgroups
bodies; collagen Q is found only at the neuromuscular AChR-MG. MG with anti-AChR antibodies is divided
junction. Antibodies against collagen Q were recently into early-onset MG (symptom onset before 50 years)
detected in the serum of 12 of 415 (3%) patients with and late-onset MG (onset after 50 years). In both sub-
MG41. In seven of these patients, no other anti­bodies groups, symptoms are generalized. In non-AChR-MG
were found. As mutations in collagen Q can lead to subtypes, the distinction between early and late onset
myasthenic syndromes41, anti-collagen Q antibodies does not have any proven value for prognostication
might be pathogenic. However, anti-­collagen Q anti- or treatment.
bodies have also been detected in healthy controls11, and In AChR-MG, the early-onset and late-onset dis-
any diagnostic or pathogenic significance of these anti- ease types differ with respect to thymic pathology, HLA
gens, or a capacity to interfere with anticholinesterase genotype and other genetic variables, autoimmune
therapies, remains to be proven. comorbidity, and response to therapy. For example,
thymectomy has clear clinical benefits in early-onset
Cortactin antibodies. Cortactin is a cytosolic actin-­ MG, but its benefits in late-onset MG are more ques-
binding protein in the skeletal muscle that promotes tionable and possibly non-existent8,43. Moreover, the
actin assembly42. Moreover, it has a role as a signalling response to immunosuppressive drugs, as well as the
protein involved in AChR clustering mediated by the risks and clinical relevance of adverse effects, can differ
agrin–MuSK complex 42. Cortactin antibodies were between early-onset and late-onset forms2,4,8 (TABLE 1).
recently detected in 20% of MG patients without AChR MG onset can occur in childhood. The clinical
or MuSK antibodies, but also in 5% of MG patients with course of childhood and juvenile MG is similar to
AChR anti­bodies, healthy controls, and 13% of patients that of early-onset MG in adults4. Juvenile MG is most
common in East Asian populations44.

◀ Figure 1 | Neuromuscular junction in myasthenia gravis (MG). a | Normal function MuSK-MG. The presence of anti-MuSK antibodies
of neuromuscular junction, with major components implicated in MG shown. Action is usually associated with more-severe and general-
potential at the presynaptic nerve terminal causes opening of voltage-dependent ized muscle weakness and, sometimes, with muscular
Ca2+ channels, triggering release of acetylcholine and agrin into the synaptic cleft. atrophy45. The great majority of anti-MuSK-positive
Acetylcholine binds to acetylcholine receptors (AChRs), which promote sodium channel patients with MG are female25,45. In MuSK-MG, mus-
opening, which in turn triggers muscle contraction. Agrin binds to the complex formed cle strength fluctuates less than it does in other MG
by low-density lipoprotein receptor-related protein 4 (LRP4) and muscle-specific kinase subtypes, and bulbar and facial muscles are usually also
(MuSK), causing acetylcholine receptor (AChR) clustering, which is required for
involved early in the disease25,45. Limb weakness is less
maintenance of the postsynaptic structures of the neuromuscular junction. b | Major
pathogenic mechanisms of the AChR antibodies in MG include complement activation common than in AChR-MG, and ocular muscles can
at the neuromuscular junction, which causes formation of membrane attack complexes be spared45. Importantly, respiratory weakness is more
(MACs) on the muscle membrane and destruction of the typical folds in the sarcolemma likely in patients with MuSK-MG than in the other sub-
(1); antigenic modulation that results in internalization and degradation of surface groups45. The clinical presentation can lead to a suspi-
AChRs (2); and binding of AChR antibodies at the AChR ligand binding site (3), which cion of MuSK-MG, but there are no specific, absolute
could directly block acetylcholine binding and, consequently, channel opening. clinical criteria for this subgroup.
Anti-MuSK and anti-LRP4 antibodies have been shown to block the intermolecular
interactions of MuSK or LRP4 respectively, and could thus inhibit the normal mechanisms LRP4-MG. MG with LRP4 antibodies is less well char-
for maintenance of the organization of the neuromuscular junction (4). Antibodies with acterized than the other subgroups. Patients in this
known pathogenic involvement in MG are shown in red. c | MG treatment can restore
group tend to have milder symptoms, both at initial
function of the neuromuscular junction by increasing the levels of available acetylcholine
(acetylcholinesterase inhibitors; green), which improves signal transduction, or by presentation and over the course of the disease pro-
reducing the concentration of autoantibodies (immunosuppressive drugs, plasma gression26,27. Only very rarely do these patients expe-
exchange/immunoadsorption, B-cell-targeting therapies; red), which alleviates the rience a myasthenic crisis that necessitates respiratory
pathogenic mechanisms described in (b). KV1.4, voltage-gated potassium channel 1.4; support. Thymoma is not reported. A few patients have
RyR, ryanodine receptor. been reported to have a combination of anti-AChR,

NATURE REVIEWS | NEUROLOGY VOLUME 12 | MAY 2016 | 263


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Box 2 | Structure and function of AChR Ocular MG. Approximately 60% of patients with
MG have ptosis and/or diplopia at onset, and nearly
The acetylcholine receptor (AChR; top and side views shown) is composed of five all experience ocular symptoms during the course of
homologous subunits (pictured in different colours) that form a cation pore (middle). the disease46. In a minority of patients, the symptoms
Subunit composition changes with the age of the individual: α2βγδ is the predominant remain restricted to the extraocular muscles. Patients
subunit composition in embryonic muscle tissue, and α2βδε in adult muscle tissue17,68.
with ocular symptoms are always categorized as having
Although AChRs have several targeted epitopes, the majority of anti-AChR antibodies
are directed against the main immunogenic region (MIR [17), a group of overlapping ocular MG, irrespective of antibodies, thymic pathol-
epitopes located on the AChR α subunit; the central core of these subunits is formed by ogy (except thymoma) and disease duration. At 2 years
amino acids 67–76. after onset, 15–20% of patients with ocular MG still have
AChR MIR MIR purely ocular MG, and MG will persist as a focal weak-
ness in more than 90% of these patients46. The preva-
lence of ocular MG is similar among MG patients with
anti-AChR and anti-LRP4 anti­bodies46, whereas ocu-
lar MG with MuSK antibodies is nearly non-existent.
Patients with purely ocular weakness but MG‑specific
pathology detected with neuro­physiological testing
of non-ocular muscles are still regarded as having
ocular MG. Such patients have an increased risk of
generalization.

Therapies
Symptomatic drug therapy
The acetylcholinesterase inhibitor pyridostigmine
represents the first-choice treatment in all types of
auto­i mmune MG 4,8. Neostigmine, another acetyl­
cholinesterase inhibitor with a shorter half-life, can also
be used1–4. Ambenonium chloride is another acetyl­
anti-MuSK and anti-LRP4Nature Reviews
antibodies; | Neurology
these patients cholinesterase inhibitor, though for most patients, it is
tend to have more-severe MG and should, in our
16
less effective than pyridostigmine or neostigmine. All
opinion, be categorized according to the relevant AChR these drugs inhibit acetylcholine degradation, thereby
antibody subgroup. increasing the availability of acetylcholine in the
synapse. All MG subgroups besides MuSK-MG usually
Subgroups based on other characteristics respond well to this treatment1–4, but individual variation
Seronegative MG. Patients with MG but no positive is considerable.
test results for anti-AChR, anti-MuSK or anti-LRP4 In MuSK-MG, the response to acetylcholinesterase
anti­b odies are characterized as having seronegative inhibition is often insufficient, reflecting differences
MG, and constitute about 10% of generalized (non- between MG subgroups with respect to antibody-­
ocular) MG patients1–4, depending on the sensitivity of induced pathology in the postsynaptic muscle mem-
the antibody tests used. The seronegative MG group is brane. A recent study reported a good response in
heterogeneous, as it includes patients with antibodies only 50% of patients with MuSK‑MG, and 10% did not
that have affinities or concentrations too low to detect, respond at all16. In such patients, 3,4‑diaminopyridine,
patients with antibodies against relevant antigens that which increases presynaptic release of acetylcholine,
have not yet been defined and cannot be tested for, and should be tried45. This drug blocks neuronal efflux
patients with myasthenic symptoms that are not medi- through K+ channels, which increases the duration of
ated by anti­bodies, including late-onset genetic forms action potentials and results in prolonged Ca2+ chan-
associ­ated with mutations in rapsyn or other relevant nel opening, thereby promoting acetylcholine release
muscle proteins. Repeated testing can, in some patients, at the muscle endplate45. The beneficial effect of pyri-
lead to a diagnostic revision from the seronegative dostigmine is specific to MG. The dose should be
MG subgroup to one of the other subgroups owing to adjusted to achieve the optimum according to thera-
increased antibody concentration, epitope spreading, peutic effect and adverse effects; most patients are able
or increased test sensitivity. We recommend repeated to do this on their own. Unfortunately, 3,4‑diamino-
testing in seronegative MG 6–18 months after the initial pyridine provides only a mild positive effect for most
assessment. patients1–4.

Thymoma-associated MG. MG attributed to thymoma Immunosuppressive drug therapy


constitutes about 10% of all MG cases. Approximately Nearly all patients with late-onset MG, thymoma MG,
30% of patients with thymoma develop MG, and and MuSK-MG require immunosuppressive therapy to
another 15% have anti-AChR antibodies without MG suppress autoantibody production and autoantibody-­
symptoms2,4. Thymoma is not associated with MG that induced detrimental effects at the neuromuscular
involves antibodies other than those against AChR, and junction. Early-onset MG can sometimes be alleviated
not with autoimmune disorders in general. by symptomatic therapy alone, but the majority of

264 | MAY 2016 | VOLUME 12 www.nature.com/nrneurol


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

patients with early-onset MG require pharmacological to insufficient benefit or problematic adverse effects51.
immuno­suppression; however, in many of them, this Evidence that supports the use of this drug is strongest
need is temporary. in AChR-MG51. Even though open studies and clinical
LRP4-MG is usually relatively mild, and immuno- experience point to an effect, two well-controlled stud-
suppression is often not needed. For ocular MG, there ies failed to show an additional benefit of mycophe-
are two treatment aims: to improve symptoms (ptosis nolate mofetil as an add‑on to prednisone52,53. It should
and diplopia) and prevent generalized weakness. also be noted that mycophenolate mofetil carries a
Immunosuppression can do both46. teratogenic risk50 and should be avoided in females of
childbearing age.
First-line treatments. First-line immunosuppressive Rituximab has emerged as a potent drug in MG1–4,8.
drug therapy for MG comprises either prednisone or This monoclonal antibody binds specifically to the
prednisolone, or the combination of prednisone or pred- B-lymphocyte surface antigen CD20. For severe MG,
nisolone with azathioprine47. These drugs have a broad and for MuSK-MG in particular, rituximab can be given
action on the immune system, and have been shown to if the first-line immunosuppressive therapy fails54,55.
be beneficial in all MG subgroups, though the benefit A recent meta-analysis confirmed that rituximab was
varies depending on the subgroup. more effective in MuSK-MG than in AChR-MG56, pos-
According to current recommendations, predniso- sibly via effective depletion of cells that produce IgG4
lone alone should be given only as short-term treatment antibodies. Overall, more than 80% of patients with
(<1 year)8,46. Long-term prednisolone monotherapy severe or refractory MG responded to rituximab56.
could be considered for the treatment of ocular MG, but Optimal treatment regimens with rituximab for MG
for the majority of other patients with MG, combina- as a whole or for specific MG subtypes are not known.
tion immunosuppressive treatment is recommended to Most studies to date have used the same induction reg-
obtain maximum effect with minimal adverse effects8,46. imen as for rheumatoid arthritis and then repeated the
Controlled studies of immunosuppression in MG treatment only if symptoms recurred after many months.
are scarce, and virtually none have described subgroup-­ The major limiting factor in rituximab treatment of MG
specific effects. However, uncontrolled studies and is the risk of precipitating other autoimmune disorders
clinical experience have proven the effect of this treat-
ment for MuSK-MG, late-onset MG and thymoma MG,
as well as for early-onset MG and LRP4-MG, which Box 3 | Structure and function of MuSK
usually have milder symptoms. Whereas azathioprine
(2–3 mg/kg daily) takes 6–15 months to yield an opti- The muscle-specific kinase (MuSK; schematic drawing on
the left, domains with known structure on the right69–71)
mal effect, prednisolone exerts its full effect during the
exerts its role at the neuromuscular junction by interacting
first few weeks and months of treatment. Alternate-day via its extracellular domain with a number of proteins.
dosing and gradual dose increases are widely used in Anti-MuSK antibodies have been demonstrated to inhibit
an attempt to avoid adverse effects. Once the optimal MuSK–collagen Q and MuSK–LRP4 complexes; this
improvement has been reached, the dose of prednisolone inhibition interferes with MuSK functions, such as AChR
should be slowly reduced to the lowest effective dose. clustering24,72.
Two studies have indicated that prednisolone treatment
of ocular MG reduces the risk of MG generalization, MuSK
in addition to the beneficial effect on the ocular symp-
toms48,49. Any added value of azathioprine for ocular MG Ig1
has not been shown.
If immunosuppressive therapy induces pharmaco- Ig2 Ig1+Ig2
logical remission or a marked improvement, it should
Ig3
be maintained in the long-term, but the dose should be
reduced to avoid adverse effects. Full drug withdrawal
will often lead to new exacerbations, particularly in
MuSK‑MG, thymoma MG and late-onset MG. The Frizzled Frizzled
presence of additional antibodies, particularly against
RyR and titin, is an indication for long-term treat-
ment, as these antibodies are more common in severe
MG8,11. Prednisolone and azathioprine can be safely Transmembrane
segment
administered during pregnancy and lactation50.
Juxtamembrane
segment
Second-line treatments. Priorities for second-line
immunosuppressive therapy are debated. No controlled Kinase Kinase
studies have compared different drugs in MG. Formal
studies examining specific therapies in well-defined MG
cohorts, including different MG subgroups, are sparse.
For moderate and mild MG, mycophenolate mofetil is Ig, immunoglobulin-like domain; Nature
LRP4, low-density
Reviews |lipoprotein
Neurology
an option after the failure of first-line therapy owing receptor-related protein 4.

NATURE REVIEWS | NEUROLOGY VOLUME 12 | MAY 2016 | 265


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

MG crisis or JC‑virus-related progressive multifocal leukoenceph- 2–3 months57–60. A series of IvIg or plasma exchange is
Severe worsening of alopathy. Treatments specifically targeting B cells rep- usually combined with intensified immunosuppressive
myasthenic weakness that resent an attractive strategy for all antibody-mediated treatment. MG crisis is a reversible condition and should
requires intubation or disorders, including all MG subgroups, and emerg- be treated vigorously, even in the cases where treatment
noninvasive ventilation
to avoid intubation.
ing monoclonal antibodies are promising. However, response is delayed. All MG subgroups — even sero­
concerns about the consequences of highly effective negative MG — respond in a similar way to IvIg and
immunosuppression persist. plasma exchange, implying that seronegative patients also
have circulating antibodies that are pathogenic. Antigen-
Alternative second-line treatments and third-line treat- specific immunoadsorption with selective antibody
ments. Alternative second-line and third-line treatment removal is a promising approach61, but further studies
options for MG include methotrexate, cyclosporine, tac- are needed before its clinical application. Antigen-specific
rolimus and cyclophosphamide1–4,8. Cyclosporine has a immunosuppression or immunotolerance for AChR,
proven effect in well-controlled studies, but its use has MuSK and LRP4 should be the aim for future treatment62.
been limited by a high risk of adverse effects. Response
rates for the different MG subgroups have not been Thymectomy
defined. The presence of antibodies that have been linked Thymic abnormalities in MG, such as hyperplasia and
to more-severe disease (that is, antibodies against RyR, thymoma, strongly suggest a role for a thymus-mediated
titin or KV1.4) increases the need for active treatment. The immune response in MG43,63. Active germinal centres and
effects of these treatments in the presence of antibodies an ongoing export of disease-inducing T lymphocytes
against agrin, collagen Q and cortactin are not known. will be effectively stopped by thymectomy43,63.
Intravenous immunoglobulin (IvIg) is predominantly
used as short-term immunoactive therapy in acute situ- Early-onset MG. Many controlled studies have shown
ations. IvIg and plasma exchange have similar effects on that patients with early-onset MG who are thymecto-
MG exacerbations57–60. Such treatment should always be mized have a more favourable outcome than those who
given for an ongoing or imminent MG crisis, and is also are not63,64. However, none of the studies have been pro-
recommended shortly before situations in which muscle spective or included matched control groups. Present evi-
weakness is expected to deteriorate or lead to compli- dence clearly favours early and complete thymectomy in
cations, such as surgery. The effect of IvIg and plasma patients with early-onset MG who are not symptom-free
exchange occurs 2–5 days after treatment and lasts for on symptomatic drugs alone. The fact that thymic hyper-
plasia is common in this subgroup supports a therapeutic
effect of thymectomy performed early after MG onset.
Box 4 | Structure and function of LRP4–agrin complex Early thymectomy will prevent export of AChR-specific
Low-density lipoprotein receptor-related protein 4 (LRP4; magenta) is a single T cells from the thymus to lymph nodes and peripheral
transmembrane protein with a large extracellular domain that contains multiple lymphoid tissue43,63. For a positive outcome, removal of
low-density lipoprotein repeats as well as epidermal-growth-factor-like and β‑propeller all thymus tissue is essential; the results of various video-­
repeats; it also has a transmembrane domain and a short C‑terminal region without an assisted and robotic surgical procedures and traditional
identifiable catalytic motif29. LRP4 is expressed in various tissues, including muscle and methods are similar65.
nerve tissues, and has important functions during development and morphogenesis of
limbs, ectodermal organs, the lung and the kidney73. In adult skeletal muscle, LRP4 is
Late-onset MG. Late-onset MG is traditionally regarded
expressed by subsynaptic myonuclei, and the protein is concentrated at the
neuromuscular junction, where it binds to the extracellular matrix proteoglycan agrin74
as less responsive or nonresponsive to thymectomy63,64;
(turquoise). Formation of the LRP4–agrin complex triggers activation of muscle-specific however, the evidence regarding thymectomy in patients
kinase (MuSK) and the signalling cascade required for acetylcholine receptor (AChR) with late-onset MG is sparse. The fact that the thymus of
clustering and postsynaptic differentiation75. patients in this subgroup is usually atrophic (an age-nor-
mal finding) does not provide any support to the use of
LRP4–Agrin complex thymectomy. However, the onset age of 50 years implies
that thymus function has no strong influence on pathogen-
esis. In patients with MG onset at 50–65 years who have
thymic hyperplasia, the response to thymectomy might
be expected to be similar to that in early-­onset MG, and
thymectomy should be considered in selected patients.
Antibodies against titin and RyR are most common in
late-onset MG. Although not tested in controlled studies,
such antibodies probably indicate that no response will be
seen to thymectomy. Biomarkers are needed to establish
indications for thymectomy in late-onset MG.

Thymoma-associated MG. In thymoma MG, the thy-


moma of the thymus gland should always be removed to
treat the cancer. The response of MG to thymectomy is
variable, and improvement of MG symptoms is usually
more limited than in early-onset MG.
Nature Reviews | Neurology
266 | MAY 2016 | VOLUME 12 www.nature.com/nrneurol
©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Table 1 | Characteristics of myasthenia gravis subgroups


Subgroup Antibody Additional Age at Proportion of Thymus Clinical benefit
antibodies onset patients (%) of thymectomy
Early-onset Anti-AChR Rare <50 years 15–25 Hyperplasia Proven
common
Late-onset Anti-AChR Common >50 years 35–45 Atrophy common Not proven
(but possible)
Thymoma Anti-AChR Very Any 10 Lymphoepithelioma Proven
common
MuSK Anti-MuSK Rare Any 1–10 Normal None
LRP4 Anti-LRP4 Rare Any 1–5 Normal None
Seronegative None of the Variable Any 10–15 Variable None
above detected
Ocular Variable Rare Any 15 Variable None
AChR, acetyline choline receptor; LRP4, low-density lipoprotein receptor-related protein 4; MuSK, muscle-specific kinase.

MuSK and LRP4-MG. MuSK-MG and LRP4-MG have Conclusions and perspectives
no proven connection to the thymus or thymus pathol- MG is mediated by several different autoantibod-
ogy. Case reports of single patients improving after ies, all directed against proteins in the postsynaptic
thymectomy have been published, but owing to very membrane of the neuromuscular junction, thereby
scarce evidence, thymectomy is not recommended1–4,8. inducing muscle weakness. The exact clinical pictures,
responses to symptomatic and immunoactive therapy,
Ocular MG. Thymectomy has not been shown to pre- and disease pathogenesis vary between MG subgroups
vent generalization in ocular MG, or to induce remis- defined on the basis of autoantibody pattern. AChR,
sion. Thymectomy, is therefore, not recommended for MuSK and LRP4 are the three well-defined antigen
ocular MG46. targets for symptom-inducing antibodies identified to
date. Epitope specificity and antibody characteristics
Seronegative MG. Thymectomy is not indicated in are more important for disease severity than is total
seronegative patients, except in cases where the sub- antibody concentration
group diagnosis is revised after the detection of low-­ As discussed above, patients with MG can also have
affinity anti-AChR antibodies. New and more sensitive muscle antibodies that react with non-junctional anti-
biomarkers are needed for this heterogeneous group. gens, such as titin, KV1.4 and RyR. Such antibodies
can be used as markers of disease severity and patho­
Monitoring, management, and supportive therapy genesis, as well as of response to therapy. In addition,
In MG crisis, respiratory support and treatment of any the number of junctional and non-junctional antibodies
precipitating or complicating infections are crucial. reported in patients with MG —particularly in those
Careful monitoring during pregnancy and at birth is with late-onset MG, thymoma MG and those previously
important, particularly given that 15% of babies born thought to be seronegative — is expanding, increasing
to mothers with MG have transient neonatal myasthe- diagnostic specificity and providing potential markers
nia caused by transplacental transfer of IgG antibod- for predicting response to therapies.
ies (against AChR, MuSK or LRP4)50. Moreover, some MG treatment should be personalized for each
women experience worsening of MG during pregnancy. patient. Correct MG subgrouping is a first step when
More than 30 drugs that are currently in use (includ- selecting therapy. Autoantibody pattern, age of onset,
ing muscle relaxants, penicillamine and some antibiotics) thymus pathology and the extent of muscle weakness
can theoretically interfere with neuromuscular transmis- are fundamental for such subgrouping. Therapy
sion. The clinician should always be aware of the possi- includes symptomatic and immuno­s uppressive
bility that these drugs induce increased neuro­muscular drugs, supportive measures and sometimes thymec-
impairment and worsening of MG symptoms1–4,8 and tomy. Specialized follow‑up is necessary to adapt
avoid prescribing them to patients with MG. therapy to the actual symptoms and to prevent new
Particular attention should be directed towards the exacerbations.
optimal treatment of comorbid conditions66: it can be Most MG patients have a well-controlled disease
difficult to distinguish between MG‑related reduction with minimal or moderate symptoms. The aim should
in physical capacity and heart and lung diseases and be to develop more-specific therapy that suppresses or
systemic disorders. All comorbid disorders should have induces tolerance to the well-characterized and spe-
the attention of the treating physician. Maintaining a cific autoimmune reactions that lead to autoantibody
healthy weight is beneficial, as being overweight can production and muscle weakness. Until such an anti-
further interfere with physical function. Physical activ- gen-specific therapy is available, it remains a challenge
ity should be encouraged, and adapted training should to optimize the use of available treatments for each
be instituted. individual MG patient.

NATURE REVIEWS | NEUROLOGY VOLUME 12 | MAY 2016 | 267


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

1. Gilhus, N. E. Myasthenia and neuromuscular junction. myasthenia gravis. J. Autoimmun. 52, 139–145 55. Keung, B. et al. Long-term benefit of rituximab in MuSK
Curr. Opin. Neurol. 25, 523–529 (2012). (2014). autoantibody myasthenia gravis patients. J. Neurol.
2. Querol, L. & Illa, I. Myasthenia and the neuromuscular 28. Pevzner, A. et al. Anti‑LRP4 autoantibodies in AChR- Neurosurg. Psychiatry 84, 1407–1409 (2013).
junction. Curr. Opin. Neurol. 26, 459–465 (2013). and MuSK-antibodynegative myasthenia gravis. 56. Iorio, R. et al. Efficacy and safety of rituximab for
3. Verschuuren, J. J.G. M. et al. Pathophysiology of J. Neurol. 259, 427–435 (2012). myasthenia gravis: a systematic review and meta-
myasthenia gravis with antibodies to the acetylcholine 29. Lu, Y. et al. A role for LRP4 in neuronal cell viability is analysis. J. Neurol. 262, 1115–1119 (2015).
receptor, muscle-specific kinase, and low-density related to apoE-binding. Brain Res. 1177, 19–28 57. Mandawat, A. et al. Comparative analysis of
lipoprotein receptor-related protein 4. Autoimmune (2007). therapeutic options used for myasthenia gravis.
Rev. 12, 918–923 (2013). 30. Shen, C. et al. Antibodies against low-density Ann. Neurol. 68, 797–805 (2010).
4. Gilhus, N. E. & Verschuuren, J. J. Myasthenia gravis: lipoprotein receptor-related protein 4 induce 58. Barth, D. et al. Comparison of IVIg and PLEX in patients
subgroup classification and therapeutic strategies. myasthenia gravis. J. Clin. Invest. 123, 5190–5202 with myasthenia gravis. Neurology 76, 2017–2023
Lancet Neurol. 14, 1023–1036 (2015). (2013). (2011).
5. Heldal, A. T., Owe, J. F., Gilhus, N. E. & Romi, F. 31. Tzartos, J. S. et al. LRP4 antibodies in serum and CSF 59. Gilhus, N. E. Acute treatment for myasthenia gravis.
Seropositive myasthenia gravis; a nationwide from amyotrophic lateral sclerosis patients. Ann. Clin. Nat. Rev. Neurol. 7, 132–134 (2011).
epidemiologic study. Neurology 73, 150–151 (2009). Trans. Neurol. 2, 80–87 (2014). 60. Gajdos, P., Chevret, S. & Toyka, K. V. Intravenous
6. Carr, A. S. et al. A systematic review of population 32. Gasperi, C. et al. Anti-agrin autoantibodies in immunoglobulin for myasthenia gravis (review).
based epidemiological studies in myasthenia gravis. myasthenia gravis. Neurology 82, 1976–1983 Cochrane Database Syst. Rev. 12, CD002277 (2012).
BMC Neurol. 10, 46 (2010). (2014). 61. Lazaridis, K. et al. Specific adsorbents for myasthenia
7. Owe, J. F., Daltveit, A. K. & Gilhus, N. E. Causes of 33. Zhang, B. et al. Autoantibodies to agrin in myasthenia gravis autoantibodies using mutants of the muscle
death among patients with myasthenia gravis in gravis. PloS ONE 9, e91816 (2014). nicotinic acetylcholine receptor extracellular domains.
Norway between 1951 and 2001. J. Neurol. 34. Witzemann, V., Chevessier, F., Pacifici, P. G. J. Neuroimmunol. 278, 19–25 (2015).
Neurosurg. Psychiatry 77, 203–207 (2006). & Yampolsky, P. The neuromuscular junction: selective 62. Steinman, L. The road not taken: antigen-specific
8. Skeie, G. O. et al. Guidelines for treatment of remodeling of synaptic regulators at the nerve/muscle therapy and neuroinflammatory disease. JAMA Neurol.
autoimmune neuromuscular transmission disorders. interface. Mech. Dev. 130, 402–411 (2013). 70, 1100–1101 (2013).
Eur. J. Neurol. 17, 893–902 (2010). 35. Szczudlik, P. et al. Anti-titin antibody in early and late 63. Marx, A. et al. The different roles of the thymus in the
9. Otsuka, K. et al. Collagen Q and anti-MuSK onset myasthenia gravis. Acta Neurol. Scand. 130, pathogenesis of the various myasthenia gravis
autoantibody competitively suppress agrin/LRP4/MuSK 229–233 (2014). subtypes. Autoimmun. Rev. 12, 875–884 (2013).
signalling. Sci. Rep. 5, 13928 (2015). 36. Powers, K. et al. Titin force is enhanced in actively 64. Cea, G., Benatar, M., Verdugo, R. J. & Salinas, R. A.
10. Messeant, J. et al. MuSK frizzled-like domain is critical stretched skeletal muscle. J. Exp. Biol. 217, Thymectomy for non-thymomatous myasthenia gravis
for mammalian neuromuscular junction formation and 3629–3636 (2014). (review). Cochrane Database Syst. Rev. 10, CD008111
maintenance. J. Neurosci. 35, 4926–4941 (2015). 37. Gautel, M. et al. Titin antibodies in myasthenia gravis: (2013).
11. Zisimopoulou, P., Brenner, T., Trakas, N. & Tzartos, S. J. identification of a major antigenic region of titin. 65. Ye, B. et al. Video-assisted thoracoscopic surgery versus
Serological diagnostics in myasthenia gravis based on Neurol. 43, 1381–1385 (1993). robotic-assisted thoracoscopic surgery in the surgical
novel assays and recently identified antigens. 38. Romi, F. et al. Anti-voltage-gated potassium channel treatment of Masaoka stage I thymoma. World J. Surg.
Autoimmune Rev. 12, 924–930 (2013). Kv1.4 antibodies in myasthenia gravis. J. Neurol. 259, Oncol. 11, 157–162 (2013).
12. Romi, F., Skeie, G. O., Gilhus, N. E. & Aarli, J. A. 1312–1316 (2012). 66. Gilhus, N. E., Nacu, A., Andersen, J. B. & Owe, J. F.
Striational antibodies in myasthenia gravis; reactivity 39. Suzuki, S. et al. Cardiac involvements in Myasthenia gravis and risks for comorbidity.
and possible clinical significance. Arch. Neurol. 62, myasthenia gravis associated with anti‑Kv1.4 Eur. J. Neurol. 22, 17–23 (2015).
442–446 (2005). antibodies. Eur. J. Neurol. 21, 223–230 (2014). 67. Chiou-Tan, F. Y. & Gilchrist, J. M. Repetitive nerve
13. Romi, F., Aarli, J. A. & Gilhus, N. E. Myasthenia gravis 40. Skeie, G. O. et al. Ryanodine receptor antibodies in stimulation and single-fiber electromyography in the
patients with ryanodine receptor antibodies have myasthenia gravis: epitope mapping and effect on evaluation of patients with suspected myasthenia gravis
distinctive clinical features. Eur. J. Neurol. 14, calcium release in vitro. Muscle Nerve 27, 81–89 or Lambert–Eaton myasthenic syndrome: review of
617–620 (2007). (2003). recent literature. Muscle Nerve 52, 455–462 (2015).
14. Suzuki, S. et al. Autoimmune targets of heart and 41. Zoltowska, K. M. et al. Collagen Q: a potential target for 68. Huijbers, M. G. et al. Pathogenic immune mechanisms
skeletal muscles in myasthenia gravis. Arch. Neurol. 66, autoantibodies in myasthenia gravis. J. Neurol. Sci. at the neuromuscular synapse: the role of specific
1334–1338 (2009). 348, 241–244 (2015). antibody-binding epitopes in myasthenia gravis.
15. Leite, M. I. et al. IgG1 antibodies to acetylcholine 42. Gallardo, E. et al. Cortactin autoantibodies in J. Intern. Med. 275, 12–26 (2013).
receptors in ‘seronegative’ myasthenia gravis. Brain myasthenia gravis. Autoimmun. Rev. 13, 1003–1007 69. Stiegler, A. L., Burden, S. J. & Hubbard, S. R. Crystal
131, 1940–1952 (2008). (2014). structure of the agrin-responsive immunoglobulin-like
16. Tsonis, A. I. et al. MuSK autoantibodies in myasthenia 43. Gronseth, G. H. & Barohn, R. J. Thymectomy for domains 1 and 2 of the receptor tyrosine kinase MuSK.
gravis detected by cell-based assay: a multinational autoimmune myasthenia gravis (an evidence-based J. Mol. Biol. 364, 424–433 (2006).
study. J. Neuroimmunol. 284, 10–17 (2015). review). Neurology 55, 7–15 (2000). 70. Till, J. H. et al. Crystal structure of the MuSK tyrosine
17. Unwin, N. Refined structure of the nicotinic 44. VanderPluym, J. et al. Clinical characteristics of kinase: insights into receptor autoregulation. Structure
acetylcholine receptor at 4 Å resolution. J. Mol. Biol. pediatric myasthenia: a surveillance study. Pediatrics 10, 1187–1196 (2002).
346, 967–989 (2005). 132, e939–944 (2013). 71. Koneczny, I., Cossins, J., Waters, P., Beeson, D.
18. Kordas, G. et al. Direct proof of the in vivo pathogenic 45. Guptill, J. T., Sanders, D. B. & Evoli, A. Anti-MuSK & Vincent, A. MuSK myasthenia gravis IgG4 disrupts
role of the AChR autoantibodies from myasthenia gravis antibody myasthenia gravis; clinical findings and the interaction of LRP4 with MuSK but both IgG4 and
patients. PLoS ONE 9, e108327 (2014). response to treatment in two large cohorts. IgG1‑3 can disperse preformed agrin-independent
19. Heldal, A. T., Eide, G. E., Romi, F., Owe, J. F. Muscle Nerve 44, 36–40 (2011). AChR clusters. PLoS ONE 8, e80695 (2013).
& Gilhus, N. E. Repeated acetylcholine receptor 46. Kerty, E., Elsais, A., Argov, Z., Evoli, A. & Gilhus, N. E. 72. Stiegler, A. L., Burden, S. J. & Hubbard, S. R. Crystal
antibody-concentrations and association to clinical EFNS/ENS guidelines for the treatment of ocular structure of the frizzled-like cysteine-rich domain of the
myasthenia gravis development. PLoS ONE 9, myasthenia gravis. Eur. J. Neurol. 21, 687–693 receptor tyrosine kinase MuSK. J. Mol. Biol. 393, 1–9
e114060 (2014). (2014). (2009).
20. Yang, L. et al. Non-radioactive serological diagnosis of 47. Palace, J., Newsom-Davis, J. & Lecky, B. A randomized 73. Choi, H. Y. et al. Lrp4, a novel receptor for dickkopf 1
myasthenia gravis and clinical features of patients from double-blind trial of prednisolone alone or with and sclerostin, is expressed by osteoblasts and
Tianjin, China. J. Neurol. Sci. 301, 71–76 (2011). azathioprine in myasthenia gravis. Neurology 50, regulates bone growth and turnover in vivo. PLoS ONE
21. Jacob, S., Viega, S. & Leite, M. I. Presence and 1778–1783 (1998). 4, e7930 (2009).
pathogenic relevance of antibodies to clustered 48. Benatar, M., Sanders, D. B., Wolfe, G. I., 74. Zong, Y. et al. Structural basis of agrin–LRP4–MuSK
acetylcholine receptor in ocular and generalized McDermott, M. P. & Tawil, R. Design of the efficacy of signaling. Genes Dev. 26, 247–258 (2012).
myasthenia gravis. Arch. Neurol. 69, 994–1001 prednisone in the treatment of ocular myasthenia 75. Zhang, B. et al. Autoantibodies to lipoprotein-related
(2012). (EPITOME) trial. Ann. NY Acad. Sci. 1275, 17–22 protein 4 in patients with double-seronegative
22. Cruz, P. M. R. et al. Clinical features and diagnostic (2012). myasthenia gravis. Arch. Neurol. 69, 445–451
usefulness of antibodies to clustered acetylcholine 49. Benatar, M. & Kaminski, H. Medical and (2012).
receptors in the diagnosis of seronegative myasthenia surgical treatment for ocular myasthenia (review).
gravis. JAMA Neurol. 72, 642–649 (2015). Cochrane Database Syst. Rev. 12, CD005081 (2012). Acknowledgements
23. Plomp, J. J., Huijbers, M. G., van der Maarel, S. M. 50. Norwood, F. et al. Myasthenia in pregnancy; best N.E.G., G.O.S. and F.R. have received funding from Torbjørg
& Verschuuren, J. J. Pathogenic IgG4 subclass practice guidelines from a UK multispeciality working Hauge’s Legacy for Neurological Research and from the
autoantibodies in MuSK myasthenia gravis. group. J. Neurol. Neurosurg. Psychiatry 85, 538–543 Norwegian Muscle Disease Association. S.J.T., K.L. and P.Z.
Ann. NY Acad. Sci. 1275, 114–122 (2012). (2014). have received grants from the Muscular Dystrophy
24. Kawakami, Y. et al. Anti-MuSK autoantibodies block 51. Hehir, M. K. et al. Mycophenolate mofetil in AChR- Association of the USA and from the Greek National Strategic
binding of collagen Q to MuSK. Neurology 77, antibody-positive myasthenia gravis; outcomes in 102 Reference Framework (NeuroID ISR‑3257).
1819–1828 (2011). patients. Muscle Nerve 41, 593–598 (2010).
25. Chang, T. et al. Clinical and serological study of 52. The Muscle Study Group. A trial of mycophenolate Author contributions
myasthenia gravis using both radioimmunoprecipitation mofetil with prednisone as initial immunotherapy in All authors researched data for the Review, made substantial
and cell-based assays in a South Asian population. myasthenia gravis. Neurology 71, 394–399 (2008). contributions to the discussion of the content of the article
J. Neurol. Sci. 343, 82–87 (2014). 53. Sanders, D. B. et al. An international, phase III, and reviewed and edited the manuscript before submission.
26. Higuchi, O. et al. Autoantibodies to low-density randomized trial of mycophenolate mofetil in N.E.G and S.T. wrote the article.
lipoprotein receptor-related protein 4 in myasthenia myasthenia gravis. Neurology 71, 400–406 (2008).
gravis. Ann. Neurol. 69, 418–422 (2011). 54. Diaz-Manera, J. et al. Long-lasting treatment of Competing interests statement
27. Zisimopoulou, P. et al. A comprehensive analysis of the rituximab in MuSK myasthenia. Neurology 78, N.E.G. has received speaker’s honoraria from Octapharma,
epidemiology and clinical characteristics of anti‑LRP4 in 189–193 (2012). Baxter and Merck Serono.

268 | MAY 2016 | VOLUME 12 www.nature.com/nrneurol


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

You might also like