Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

Pediatric Myasthenia Gravis

Jason H. Peragallo, MD*,†

Myasthenia gravis is a disorder of neuromuscular transmission that leads to fatigue of skeletal


muscles and fluctuating weakness. Myasthenia that affects children can be classified into the
following 3 forms: transient neonatal myasthenia, congenital myasthenic syndromes, and
juvenile myasthenia gravis (JMG). JMG is an autoimmune disorder that has a tendency to
affect the extraocular muscles, but can also affect all skeletal muscles leading to generalized
weakness and fatigability. Respiratory muscles may be involved leading to respiratory failure
requiring ventilator support. Diagnosis should be suspected clinically, and confirmatory
diagnostic testing be performed, including serum acetylcholine receptor antibodies, repetitive
nerve stimulation, and electromyography. Treatment for JMG includes acetylcholinesterase
inhibitors, immunosuppressive medications, plasma exchange, intravenous immunoglobulins,
and thymectomy. Children with myasthenia gravis require monitoring by a pediatric
ophthalmologist for the development of amblyopia from ptosis or strabismus.
Semin Pediatr Neurol 24:116-121 C 2017 Elsevier Inc. All rights reserved.

Introduction Pathophysiology
The myasthenic syndromes are due to defects of transmission In normal synaptic transmission in the neuromuscular junc-
at the neuromuscular junction. Myasthenic syndromes in tion, the axon is depolarized and this depolarization travels to
children have 3 distinct forms with pathophysiologically the axon terminal. Voltage-gated calcium channels open,
different mechanisms. The congenital myasthenic syndromes leading to acetylcholine containing vesicles to fuse to the cell
(CMS) are a group of genetic disorders that lead to muscle membrane. Acetylcholine is then released from synaptic
weakness through structural or functional abnormalities of vesicles into the synaptic cleft from the axon terminal. The
the proteins involved in neuromuscular transmission itself. acetylcholine travels across the synaptic cleft to the AChR sites
Transplacental transfer of maternal antibodies of a myasthenic where binding causes sodium channels to open, depolarizing
mother causes transient neonatal myasthenia (TNM) in infants. the motor end plate of the muscle fiber. Acetylcholinesterase
Juvenile myasthenia gravis (JMG) is an autoimmune disorder degrades the acetylcholine, and the sodium channels close,
that leads to dysfunction of acetylcholine receptors (AChR). allowing repolarization of the muscle fiber.
All forms of myasthenia lead to muscle fatigue and weakness
of varying degrees. Fluctuations in weakness are a hallmark of
this disease. If weakness involves the musculature involved in Congenital Myasthenic Syndromes
respiration, these diseases can be life threatening.1,2 This review The CMS are not autoimmune diseases, but are a group of
will focus primarily on the JMG form. disorders caused by alterations in the structure or function of
proteins in the neuromuscular junction. These disorders must
be suspected to distinguish them from myopathies or other
neurogenic conditions.3 The disease can be the result of, for
From the *Department of Ophthalmology, Emory University School of example, inability to form normal acetylcholine, inability to
Medicine, Atlanta, GA. transport and release acetylcholine, abnormal nicotinic AChR,

Department of Pediatrics, Emory University School of Medicine, Atlanta, GA. primary AChR decifiency, end plate development and main-
Supported in part by an unrestricted departmental Grant (Department of tenance protein abnormalities, defects in protein glycosylation,
Ophthalmology) from Research to Prevent Blindness, Inc, New York, and and many others.4 The most common cause of CMS is
by NIH/NEI core Grant P30-EY06360 (Department of Ophthalmology).
Address reprint requests to Jason H. Peragallo, MD, Department of Ophthal-
alterations in the structure, presence, or kinetics of the AChR
mology, Emory University School of Medicine, 1365B Clifton Rd NE, itself.4 Performing a genetic analysis for the specific mutation
Atlanta, GA 30322. E-mail: jperaga@emory.edu causing CMS is necessary to direct treatment for that specific

116 http://dx.doi.org/10.1016/j.spen.2017.04.003
1071-9091/11/& 2017 Elsevier Inc. All rights reserved.
Pediatric myasthenia gravis 117

syndrome. A treatment modality for CMS caused by one B-cell–mediated disease, recent studies have proposed B-cell–
mutation may actually be harmful to the patient if a different activating factor as playing a role in its pathogenesis and as a
mutation is present, and treatment must be tailored to the marker of disease activity in JMG.15,16
specific mutation causing CMS. Children with CMS often will
have a relative who is similarly affected. They will present with
fatigable weakness, usually at a very early age, and can present Demographics
in infancy. The affected muscles may be ocular alone, which
can lead to isolated fatigable ptosis, or all skeletal muscles may There have been varying estimates of the incidence of
be affected leading to more widespread weakness. Investiga- myasthenia gravis occurring in the general population in the
tions for autoantibodies found in JMG and TNM will be literature ranging from 1.7-30.0 cases per million person years,
negative. Making the distinction between CMS and JMG may with a prevalence of 77.7 cases per million persons.17,18 Many
be difficult if a known genetic mutation causative for CMS studies estimating the incidence and prevalence of JMG have
cannot be identified and autoantibodies leading to JMG are not also been published. A meta-analysis of the literature on this
detected. subject concluded that the incidence of JMG is approximately
between 1.0 and 5.0 cases per million person years.17 Pediatric
patients were found to make up approximately 10%-15% of all
Transient Neonatal Myasthenia patients with autoimmune myasthenia gravis in a prevalence
TNM is an antibody-mediated disorder that differs from JMG study in Virginia.19 Among Asian populations, there appears to
and CMS. This disease presents shortly after birth in infants of be a higher prevalence of JMG compared to white populations,
mothers who have autoimmune myasthenia gravis. The with JMG representing up to 50% of all autoimmune
circulating maternal autoantibodies cross the placenta to the myasthenia gravis cases.20,21
fetus, leading to an affected child after birth. Children of
mothers who have myasthenia gravis will develop TNM
approximately 5%-30% of the time.5,6 Infants can present Clinical Presentation
with generalized hypotonia, feeble cry, respiratory distress, Fatigable skeletal muscle weakness that fluctuates is the
poor suck, and extraocular muscle weakness.6 Symptoms are characteristic sign of myasthenia gravis. Myasthenia gravis is
usually self-limited but respiratory support may be necessary. known to preferentially target the extraocular muscles. Cases in
Patients respond well to neostigmine treatment and plasma which only the extraocular muscles are affected are termed
exchange. TNM can be distinguished from CMS by the ocular myasthenia gravis (OMG). If any other skeletal muscles
detection of autoantibodies, and when the disease is demon- are involved, the patient is diagnosed with generalized
strated to be transient once autoantibodies are cleared from the myasthenia gravis. The vast majority of patients who have
infant and symptoms resolve. No long-term treatment is myasthenia gravis will have involvement of the extraocular
required once the autoantibodies are no longer present. muscles at some point during the course of their disease.

Juvenile Myasthenia Gravis Ocular Myasthenia Gravis


Juvenile myasthenia gravis is an autoimmune disorder pre- Approximately 10%-35% of all cases of JMG are of the OMG
senting with fatigability before the age of 19 years, which in subtype.1,22 OMG is a more common form of JMG in Asian
contrast to the CMS is not due to a structural abnormality in populations than in other populations.20,21 The muscles
the neuromuscular junction.7 JMG is also not a transient involved include the vertical and horizontal rectus muscles,
disease, such as TNM. Autoantibodies against targets such as inferior and superior oblique muscles, orbicularis oculi, and
the AChR and muscle-specific kinase (MuSK) lead to the levator palperae superioris. The initial presentation in these
decreased AChR activity though complement-mediated lysis children may be ptosis, strabismus, or diplopia. In one
of the postsynaptic membrane, increased rate of degradation of community-based case-control study, JMG was the underlying
the receptor, and direct inhibition of activity.8-10 Lipoprotein cause of ptosis in 0.81% of pediatric patients.23 JMG was
receptor–related membrane protein 4 (LRP-4) is another target present in children with ptosis at 26.7-fold the rate of children
of autoantibodies implicated in the development of myasthenia who did not have ptosis.23 Patients may actually have eyelid
gravis in patients in whom the antibodies to the AChR or retraction, most prominent upon awakening, of the eyelid
MuSK were not detected.11 LRP-4 is a protein necessary for that is found to be ptotic in the clinic. Directed questions
MuSK activation.12 Up to 18.4% of patients who do not have about ptosis should address not only if it is worse in the
antibodies to the AChR or MuSK were found to have evenings, but also if it appears resolved upon awakening in the
antibodies to LRP-4 in one analysis.12 Other autoimmune morning.
diseases, such as Hashimoto disease and autoimmune Patients with JMG with or without the purely ocular form of
polymyositis, may be present in the same individual who has disease can present with pseudo-cranial nerve palsies, and may
JMG.13 A recent study analyzed data from patients who had have variable strabismus measurements when presenting to a
both myasthenia gravis and aquaporin 4 antibody–positive pediatric ophthalmologist for evaluation for strabismus sur-
neuromyelitis optica.14 In this study, 20% of patients had gery. It is important to note that the pupils are not involved to a
JMG before developing neuromyelitis optica.14 As JMG is a clinically detectable degree in myasthenia gravis, distinguishing
118 J.H. Peragallo

this disease from other neurologic causes of strabismus. Having the patient look in downgaze causes relative rest of the
Pupillary abnormalities should prompt evaluation for other levator palpebrae, which is “overstimulated” upon upgaze.
neurological causes. Rates of progression from OMG to Another simple in office test that can suggest myasthenia
generalized myasthenia gravis has been reported from gravis if positive is the ice pack test. After measuring the initial
8%-49%, with rates of conversion for JMG specifically ranging palperbral fissure height, an ice-filled glove or ice pack is
from 8%-33%.2,24-29 Preadolescent patients with OMG may placed over the patient’s eye or eyes. The ice is left there for 1-2
be less likely to progress to the generalized form than minutes while the patient rests. Once the ice is removed, the
adolescents.25 palpebral fissure is measured again. An improvement of ptosis
of 2 mm of more is a positive result. This test has been found to
be approximately 80% sensitive for the detection of myasthe-
Generalized Myasthenia Gravis nia gravis.31,32 An alternative to the ice pack test is the rest test,
The generalized form of JMG can be life threatening, as any in which a patient is asked to rest for 30 minutes after being
skeletal muscle can be involved, including the muscles examined. Following a period of rest, the examination is
involved in respiration. If this occurs, patients may develop repeated, and measurements compared.33 The ice pack test
respiratory failure requiring intubation.1,8 Presenting signs can may be more sensitive than the rest test for the detection of
include the ocular motility abnormalities and ptosis described myasthenia gravis.34
earlier, but must include other limb or facial weakness,
difficulty breathing or swallowing, and chocking. Speech can
be affected leading to a change in voice characteristics. Severity Serologic Testing
of myasthenia gravis can be classified according to the Acetylcholine receptor antibody testing is readily available
Myasthenia Gravis Foundation of America’s classification commercially. The 3 main AChR antibodies tested are binding,
system, which allows for uniform criteria to be used cross- blocking, and modulating antibodies. Their presence is
institution for research purposes and for monitoring disease important as it can definitively make the diagnosis of myas-
severity.30 thenia gravis in uncertain cases.8,35 One study of patients
with JMG found an AChR antibody positivity rate of 72% of
those presenting with generalized symptoms, and 41% in
those presenting with ocular symptoms alone.36 Patients who
Diagnosis were initially AChR antibody negative became positive 41% of
Making the diagnosis of myasthenia gravis begins when a the time within 2 years.36 However, most of those patients
patient demonstrates some form of fatigable weakness. who converted to positive AChR antibody status did not
A thorough targeted clinical examination should include some have worsening of symptoms. Remission was associated
evaluation of fatigability to address suspicion of myasthenia with conversion from AChR antibody positive to AChR
gravis. Ancillary testing for its diagnosis includes serologic, antibody–negative status.36 Another more recent study found
pharmacologic, and electrophysiologic testing. an AChR antibody positivity rate of 86% in patients with
JMG.22 The finding that patients with the ocular form have
autoantibodies to the AchR less frequently than the generalized
Clinical Examination Techniques form is similar to what is found in adults.37 Patients who are
Fatigability on sustained upgaze is one of the simplest tests to negative for AChR antibodies can also be tested for anti-MuSK
perform on patients in whom JMG is suspected. First, antibodies, which have been found to be positive in up to 40%
palpebral fissure height is taken and an extraocular of all patients with autoimmune myasthenia gravis who are
motility examination is performed. The patient is then directed AChR antibody negative.38 Similar rates have been found in
to look at a target above their heads, and instructed to look at AChR antibody–negative JMG series.39 Children who have
the target without moving their head out of primary anti-MuSK antibodies tend to have more severe, acute,
position. The target is held there for a certain period while generalized symptoms, with respiratory distress, similar to
the eyelid and ocular alignment is closely observed. Patients what is found in adults with anti-MuSK antibodies.39,40
with JMG may demonstrate a change in palpebral height and Response to treatment for JMG due to anti-MuSK antibodies
ocular alignment while being observed, which suggests can be variable.39 Patients who are negative for both AChR
fatigability. The target can then be removed, and the ocular antibodies and MuSK antibodies but clinically have myasthe-
motility examination and measurement of palpebral fissure nia gravis are called “double seronegative”. Another antibody
height can be repeated. Changes in these measurements that has been found to be positive in double-seronegative
suggest the presence of myasthenia gravis. patients is the lipoprotein receptor-related membrane protein 4
The Cogan lid twitch is another feature that can be seen in (LRP-4).41 Pediatric patients may be affected in anti-LRP-
patients who have levator palpebrae superioris involvement 4–mediated myasthenia gravis as well.12 The incidence of
leading to ptosis. This sign can be elicited by having a patient anti-LRP-4 positivity varies widely from study to study, likely
look in downgaze for a certain period, followed by asking them due to different assays used by research teams.42 Anti-LRP-4–
to quickly look up. The eyelid overelevates briefly (this is the associated myasthenia gravis tends to manifest mild
“twitch”), followed by fatigue and return to a ptotic position. symptoms.12,41,42
Pediatric myasthenia gravis 119

Pharmacologic Testing Immunosuppressive Agents


The classic pharmacologic test for myasthenia gravis uses a The most common immunosuppressive agent prescribed
short-acting antiacetylcholinesterase, edrophonium, in the for myasthenia gravis is prednisone. This medication is easily
edrophonium (Tensilon) test. By inhibiting acetylcholinester- administered orally to children with JMG, but long-term
ase, the amount of acetylcholine is effectively increased in the use can be harmful due to systemic side effects.1 Children
neuromuscular junction, and acetylcholine is able to bind to treated with prednisone demonstrated a good overall response
the AChR for a longer period, leading to improved neuro- in a retrospective study.47 Additionally, treatment with pre-
muscular transmission. This test is performed with cardiac dnisone has been found to have a protective effect on the
monitoring and cardiopulmonary resuscitation equipment conversion from ocular to generalized myasthenia.48,49
(including atropine) readily available owing to the risk of Treatment with prednisone appeared to provide control
bradycardia and asystole. During the test, attention should be of OMG in a randomized clinical trial compared with
directed to a sign of myasthenia the patient exhibits (muscle placebo.50
weakness, ptosis, etc) that can be readily monitored. Compar- Azathioprine can be considered for use in myasthenia gravis.
ison to photographs can be useful. A small dose is given Children who do not have an adequate response to steroids
initially to test for supersensitivity to edrophonium, followed and anticholinesterase treatment can be placed on azathio-
by escalating doses. Parasympathetic side effects including prine, and it can be used as a steroid-sparing agent.2,51-53
excessive salivation, hypotension, diarrhea, flushing, nausea, It should be kept in mind that azathioprine has a delayed
and sweating can occur. False negatives and false positives can treatment effect from time of initiation of therapy, is potentially
occur with edrophonium testing as other diseases can respond teratogenic, and is considered carcinogenic.52,54
to this test, or the patient may fail to respond to it.1 Neo- Other immunomodulatory medications can be considered
stigmine is an alternative drug to edrophonium that can be for use in myasthenia gravis. Rituximab has been found to be
used in young children. Another alternative is to simply treat effective in its treatment, improve symptoms, and reduce the
the patient with a trial of pyridostigmine and observe for amount of steroids required for treatment.55-57 Rituximab has
improvements in symptoms over time. also been found to be effective for long-term treatment of
refractory myasthenia gravis.58 Mycophenolate mofetil has also
been found to be effective in the treatment of myasthenia
Electrophysiological Testing gravis, but randomized control trials have not demonstrated
Two methods of electrophysiological testing are primarily used the ability to taper steroids while on this agent.59-61 However,
for diagnosing myasthenia gravis. In one test, repetitive nerve discontinuation of mycophenolate mofetil in patients taking
stimulation, trains of stimuli are delivered to a target muscle, this medication does lead to more frequent myasthenia gravis
and quantitative analysis of the motor responses is performed. exacerbations.62 Tacrolimus has been used for the treatment of
A decremental response of 10% from the first to the fourth myasthenia gravis as well, including JMG.63,64
response is considered a positive result.43 This test is more
sensitive in patients who have generalized JMG in comparison
to those who have ocular JMG.44 Currently the “gold-standard”
for testing for neuromuscular junction disorders is single-fiber
Other Medical Treatment Modalities
electromyography (sfEMG) performed on the orbicularis Intravenous immunoglobulin (IVIG) can be administered
oculi.43 The sfEMG is particularly useful in cases of suspected for myasthenia gravis in an effort to reduce the circulating
myasthenia gravis with negative serologies as it has very high autoantibodies by decreasing B-cell antibody production
sensitivity for MG.45 Unfortunately, traditional single-fiber and T-cell function. In a randomized controlled trial against
EMG studies are difficult to perform on children due to placebo, IVIG was found to be effective in improving
discomfort and the length of the test. As an alternative, a less myasthenia gravis symptoms.65 Therapy with IVIG has
invasive test termed stimulated single-fiber EMG has been been used for patients with JMG, but appeared to have
developed that can be used in children.43,46 better efficacy for short-term treatments and exacerbations
than for long-term treatment.66,67 Plasmapheresis can also
be used in patients with myasthenia gravis to directly filter
Treatment autoantibodies and cytokines from the affected patient’s
serum. A comparison in patients with JMG between IVIG
Acetylcholinesterase Inhibitors and plasmapheresis for maintenance therapy demonstrated
Pyridostigmine is the main pharmacologic agent used in the high response rates for both modalities, but a more
treatment of myasthenia gravis, whether in children or adults. consistent response with plasmapheresis.68 However, pre-
Edrophonium has too short a half-life to be of use in long-term vious studies performed in adults have shown no signifi-
treatment of myasthenia gravis. Even with the appropriate cant difference between the 2 treatments.69 Consensus
usage and dosing of pyridostigmine, symptoms and signs of statements state that IVIG and plasmapheresis are appro-
myasthenia gravis may continue.1 Pyridostigmine may be priate for acute exacerbations or crises only, and are not
combined with other agents such as immunosuppressives. appropriate for maintenance therapy unless patients are
Side effects of pyridostigmine are similar to those of edropho- unable to tolerate immunosuppressive agents or have
nium described earlier. refractory myasthenia gravis.70
120 J.H. Peragallo

Thymectomy References
Approximately 10%-15% of all adults with myasthenia gravis 1. Andrews PI: Autoimmune myasthenia gravis in childhood. Semin Neurol
24:101-110, 2004
are found to have a thymoma, and 30% of patients who have a
2. Mullaney P, Vajsar J, Smith R, Buncic JR: The natural history and
thymoma develop myasthenia gravis.71,72 Children who have ophthalmic involvement in childhood myasthenia gravis at the Hospital
myasthenia gravis have rarely been found to harbor thymo- for Sick Children. Ophthalmology 107:504-510, 2000
mas.22,73 However, thymectomy for patients with seropositive 3. Kinali M, Beeson D, Pitt MC, et al: Congenital myasthenic syndromes in
JMG has been evaluated and has been found to be associated childhood: Diagnostic and management challenges. J Neuroimmunol
201-202:6-12, 2008
with improved remission rates.22,74 Therefore, thymectomy
4. Engel AG, Shen X, Selcen D, Sine SM: Congenital myasthenic syndromes:
may have a role for children who are unresponsive to medical Pathogenesis, diagnosis, and treatment. Lancet Neurol 14:420-434, 2015
therapy or who cannot undergo standard immunsuppression, 5. Téllez-Zenteno JF, Hernández-Ronquillo L, Salinas V, et al: Myasthenia
although the long-term effects of thymectomy in children are gravis and pregnancy: Clinical implications and neonatal outcome. BMC
unclear.70 Thymectomy may protect patients with the ocular Musculoskelet Disord 5:42, 2004
form of JMG from undergoing generalization, and may 6. Namba T, Brown SB, Grob D: Neonatal myasthenia gravis: Report of two
cases and review of the literature. Pediatrics 45:488-504, 1970
improve ocular symptoms as well.9,24 A randomized trial of 7. Evoli A: Acquired myasthenia gravis in childhood. Curr Opin Neurol
thymectomy for adult patients on prednisone for myasthenia 23:536-540, 2010
gravis demonstrated a reduction in prednisone dose require- 8. Liew WK, Kang PB: Update on juvenile myasthenia gravis. Curr Opin
ment and improvement in symptoms for those who under- Pediatr 25:694-700, 2013
9. Gadient P, Bolton J, Puri V: Juvenile myasthenia gravis: Three case reports
went thymectomy.75
and a literature review. J Child Neurol 24:584-590, 2009
10. Pal J, Rozsa C, Komoly S, Illes Z: Clinical and biological heterogeneity of
autoimmune myasthenia gravis. J Neuroimmunol 231:43-54, 2011
11. Tsivgoulis G, Dervenoulas G, Kokotis P, et al: Double seronegative
Ophthalmic Outcomes myasthenia gravis with low density lipoprotein-4 (LRP4) antibodies
Patients with JMG should be monitored for the development of presenting with isolated ocular symptoms. J Neurol Sci 346:328-330,
2014
amblyopia and strabismus.76 Amblyopia should be treated to
12. Zisimopoulou P, Evangelakou P, Tzartos J, et al: A comprehensive analysis
prevent permanent vision loss. Up to 96% of patients with the of the epidemiology and clinical characteristics of anti-LRP4 in myasthenia
ocular form of JMG will have ptosis, and up to 88% will gravis. J Autoimmun 52:139-145, 2014
present with strabismus.28 Amblyopia has been found in 21%- 13. Tsao C, Mendell JR, Lo WD, et al: Myasthenia gravis and associated
50% of children with ocular JMG, but following properly autoimmune diseases in childhood. J Child Neurol 15:767-769, 2000
14. Jarius S, Paul F, Franciotta D, et al: Neuromyelitis optica spectrum
diagnosis and treatment, that proportion of patients with
disorders in patients with myasthenia gravis: Ten new aquaporin-4
amblyopia has been found to decrease as low as 3%.24,25,28 antibody positive cases and a review of the literature. Mult Scler
Strabismus is more difficult to treat in patients with ocular 18:1135-1143, 2012
JMG, with its presence at final follow-up ranging from 15%- 15. Motobayashi M, Inaba Y, Nishimura T, et al: An increase in circulating B
57%.24,25 Ptosis may remain in patients who have stable cell-activating factor in childhood-onset ocular myasthenia gravis. Pediatr
Neurol 52:404-409, 2015
disease in 46%-66% of patients.24,25 Upon stabilization of the
16. Motobayashi M, Nishimura T, Inaba Y: Usefulness of circulating B-cell-
underlying disease, surgical correction of the strabismus and activating factor in serial monitoring in childhood-onset ocular myasthe-
ptosis can be considered, with a 56% long-term success rate nia gravis. Pediatr Neurol 63:e1-e2, 2016
found in one retrospective study.77 Counseling should 17. McGrogan A, Sneddon S, de Vries CS: The incidence of myasthenia gravis:
be given about realistic expectations of strabismus surgery A systematic literature review. Neuroepidemiology 34:171-183, 2010
18. Carr AS, Cardwell CR, McCarron PO, McConville J: A systematic review
and about the possibility of requiring multiple surgical
of population based epidemiological studies in myasthenia gravis. BMC
interventions. Neurol 10:46, 2010
19. Phillips LH, Torner JC, Anderson MS, Cox GM: The epidemiology of
myasthenia gravis in central and western Virginia. Neurology 42:
1888-1893, 1992
Conclusions 20. Zhang X, Yang M, Xu J, et al: Clinical and serological study of myasthenia
gravis in HuBei Province, China. J Neurol Neurosurg Psychiatry 78:
Pediatric myasthenia gravis can present as neonatal myasthenia 386-390, 2007
gravis (CMS, TNM) or during adolescence (JMG). The latter is 21. Chiu H-C, Vincent A, Newsom-Davis J, et al: Myasthenia gravis: Population
an autoimmune disease that can have a variable presentation, differences in disease expression and acetylcholine receptor antibody titers
ranging from mild ophthalmic symptoms, such as isolated between Chinese and Caucasians. Neurology 37:1854-1857, 1987
fatigable ptosis, to myasthenic crises involving the respiratory 22. Castro D, Derisavifard S, Anderson M, et al: Juvenile myasthenia gravis:
A twenty-year experience. J Clin Neuromusc Dis 14:95-102, 2013
muscles, requiring ventilator support. Making a formal diag- 23. Nemet AY, Segal O, Mimouni M, Vinker S: Associated morbidity of
nosis of the disease can be difficult, but when suspicion is pediatric ptosis—a large, community based case-control study. Graefes
raised for a fatigable deficit, appropriate diagnostic testing can Arch Clin Exp Ophthalmol 252:1509-1514, 2014
be pursued. Treatments should be tailored to the subtype of 24. Pineles SL, Avery RA, Moss HE, et al: Visual and systemic outcomes in
disease and the severity of illness. Variable responses to pediatric ocular myasthenia gravis. Am J Ophthalmol 150:453-459, 2010
25. Ortiz S, Borchert M: Long-term outcomes of pediatric ocular myasthenia
medications and surgical interventions can be seen between
gravis. Ophthalmology 115:1245-1248, 2008
patients. Children with involvement of the extraocular mus- 26. Nagia L, Lemos J, Abusamra K, et al: Prognosis of ocular myasthenia
culature should be followed by a pediatric ophthalmologist to gravis: Retrospective multicenter analysis. Ophthalmology 122:1517
monitor for ophthalmic complications associated with JMG. 1521, 2015
Pediatric myasthenia gravis 121

27. Bever CT, Aquino AV, Penn AS, et al: Prognosis of ocular myasthenia. 53. Palace J, Newsom-Davis J, Lecky B: A randomized double-blind trial of
Ann Neurol 14:516-519, 1983 prednisone alone or with azathioprine in myasthenia gravis: Myasthenia
28. Kim JH, Hwang JM, Hwang YS, et al: Childhood ocular myasthenia gravis. Gravis Study Group. Neurology 50:1778-1783, 1998
Ophthalmology 110:1458-1462, 2003 54. Haines SR, Thurtell MJ: Treatment of ocular myasthenia gravis. Curr Treat
29. McCreery KM, Hussein MA, Lee AG, et al: Major review: the clinical Options Neurol 14:103-112, 2012
spectrum of pediatric myasthenia gravis: blepharoptosis, ophthalmoplegia 55. Wylam ME, Anderson PM, Kuntz NL, Rodriguez V: Successful treatment
and strabismus. A report of 14 cases. Binocul Vis Strabismus Q 17: of refractory myasthenia gravis using rituximab: A pediatric case report.
181-186, 2002 J Pediatr 143:674-677, 2003
30. Jaretzki A 3rd, Barohn RJ, Enrstoff RM, et al: Myasthenia gravis: 56. Stieglbauer K, Topakian R, Schäffer V, Aichner FT: Rituximab for
Recommendations for clinical research standards. Task force of the myasthenia gravis: Three case reports and review of the literature.
Medical Scientific Advisory Board of the Myasthenia Gravis Foundation J Neurol Sci 280:120-122, 2009
of America. Neurology 55:16-23, 2000 57. Koul R, Al Futaisi A, Abdwani R: Rituximab in severe seronegative juvenile
31. Golnik KC, Pena R, Lee AG, Eggenberger ER: An ice test for the diagnosis myasthenia gravis: Review of the literature. Pediatr Neurol 47:209-212,
of myasthenia gravis. Ophthalmology 106:1282-1286, 1999
2012
32. Sethi KD, Rivner MH, Swift TR: Ice pack test for myasthenia gravis.
58. Robeson KR, Kumar A, Keung B, et al: Durability of the rituximab
Neurology 37:1383-1385, 1987
response in acetylcholine receptor autoantibody-positive myasthenia
33. Odel JG, Winterkorn JM, Behrens MM: The sleep test for myasthenia
gravis. JAMA Neurol 74:60-66, 2017
gravis. A safe alternative to Tensilon. J Clin Neuroophthalmol 11:
59. Hehir MK, Burns TM, Alpers J, et al: Mycophenolate mofetil in AChR-
288-292, 1991
antibody-positive myasthenia gravis: Outcomes in 102 patients. Muscle
34. Kubis KC, Danesh-Meyer HV, Savino PJ, Sergott RC: The ice test versus
the rest test in myasthenia gravis. Ophthalmology 107:1995-1998, 2000 Nerve 41:593-598, 2010
35. Della Marina A, Trippe H, Lutz S, Schara U: Juvenile myasthenia gravis: 60. Muscle Study Group. A trial of mycophenolate mofetil with prednisone as
Recommendations for diagnostic approaches and treatment. Neuropedi- initial immunotherapy in myasthenia gravis. Neurology 71:394-399, 2008
atrics 45:75-83, 2014 61. Sanders DB, Hart IK, Mantegazza R, et al: An international, phase III,
36. Anlar B, Şenbil N, Köse G, Değerliyurt A: Serological follow-up in juvenile randomized trial of mycophenolate mofetil in myasthenia gravis. Neurol-
myasthenia: Clinical and acetylcholine receptor antibody status of patients ogy 71:400-406, 2008
followed for at least 2 years. Neuromuscul Disord 15:355-357, 2005 62. Oskarsson B, Rocke DM, Dengel K, Richman DP: Myasthenia
37. VanderPluym J, Vajsar J, Jacob FD, et al: Clinical characteristics of pediatric gravis exacerbation after discontinuing mycophenolate. Neurology 86:
myasthenia: A surveillance study. Pediatrics 132:e939-e944, 2013 1159-1163, 2016
38. Zhou L, McConville J, Chaudhry V, et al: Clinical comparison of muscle- 63. Minami N, Fujiki N, Doi K, et al: Five-year follow-up with low-dose
specific tyrosine kinase (MuSK) antibody-positive and -negative myas- tacrolimus in patients with myasthenia gravis. J Neurol Sci 300:59-62, 2011
thenic patients. Muscle Nerve 30:55-60, 2004 64. Mori T, Mori K, Suzue M, et al: Effective treatment of a 13-year-old boy
39. Skjei KL, Lennon VA, Kuntz NL: Muscle specific kinase autoimmune with steroid-dependent ocular myasthenia gravis using tacrolimus. Brain
myasthenia gravis in children: A case series. Neuromuscul Disord 23: Dev 35:445-448, 2013
874-882, 2013 65. Zinman L, Ng E, Bril V: IV immunoglobulin in patients with myasthenia
40. Evoli A, Tonali PA, Padua L, et al: Clinical correlates with anti-MuSK gravis. Neurology 68:837-841, 2007
antibodies in generalized seronegative myasthenia gravis. Brain 126: 66. Selcen D, Dabrowski ER, Michon AM, Nigro MA: High-dose intravenous
2304-2311, 2003 immunoglobulin therapy in juvenile myasthenia gravis. Pediatr Neurol
41. Pevzner A, Schoser B, Peters K, et al: Anti-LRP4 autoantibodies in AChR- 22:40-43, 2000
and MuSK-antibody-negative myasthenia gravis. J Neurol 259:427-435, 67. Feasby T, Banwell B, Benstead T, et al: Guidelines on the use of
2012 intravenous immune globulin for neurologic conditions. Transfus Med
42. Binks S, Vincent A, Palace J: Myasthenia gravis: A clinical-immunological Rev 21:S57-S107, 2007
update. J Neurol 263:826-834, 2016 68. Liew WK, Powell CA, Sloan SR, et al: Comparison of plasmapheresis and
43. Pitt M: Neurophysiological strategies for the diagnosis of disorders of the intravenous immunoglobulin as maintenance therapies for juvenile
neuromuscular junction in children. Dev Med Child Neurol 50:328-333, myasthenia gravis. JAMA Neurol 71:575-580, 2014
2008 69. Gajdos P, Chevret S, Toyka KV: Intravenous immunoglobulin for
44. Afifi AK, Bell WE: Tests for juvenile myasthenia gravis: Comparative myasthenia gravis. Cochrane Database Syst Rev 12:CD002277, 2012
diagnostic yield and prediction of outcome. J Child Neurol 8:403-411, 70. Sanders DB, Wolfe GI, Evoli A, et al: International consensus guidance
1993
for management of myasthenia gravis. Neurology 87:419-425, 2016
45. Padua L, Stalberg E, LoMonaco M, et al: SFEMG in ocular myasthenia
71. Marx A, Pfister F, Schalke B, et al: The different roles of the thymus in the
gravis diagnosis. Clin Neurophysiol 111:1203-1207, 2000
pathogenesis of the various myasthenia gravis subtypes. Autoimmun Rev
46. Verma S, Lin J: Stimulated jitter analysis for the evaluation of neuro-
12:875-884, 2013
muscular junction disorders in children. Muscle Nerve 53:471-472, 2016
72. Gilhus NE, Verschuuren JJ: Myasthenia gravis: Subgroup classification
47. Batocchi AP, Evoli A, Palmisani MT, et al: Early-onset myasthenia gravis:
and therapeutic strategies. Lancet Neurol 14:1023-1036, 2015
Clinical characteristics and response to therapy. Eur J Pediatr 150:66-68,
73. Nikolic DM, Nikolic AV, Lavrnic DV, et al: Childhood-onset myasthenia
1990
48. Kupersmith MJ, Latkany R: Development of generalized disease at two gravis with thymoma. Pediatr Neurol 46:329-331, 2012
years in patients with ocular myasthenia gravis. Arch Neurol 60:243-248, 74. Heng HS, Lim M, Absoud M, et al: Outcome of children with
2003 acetylcholine receptor (AChR) antibody positive juvenile myasthenia
49. Kupersmith MJ: Ocular myasthenia gravis: Treatment successes and gravis following thymectomy. Neuromuscul Disord 24:25-30, 2014
failures in patients with long-term follow-up. J Neurol 256:1314-1320, 75. Wolfe GI, Kaminski HJ, Aban IB, et al: Randomized trial of thymectomy in
2009 myasthenia gravis. N Engl J Med 375:511-522, 2016
50. Benatar M, McDermott MP, Sanders DB, et al: Efficacy of prednisone for 76. Emoto Y, Emoto H, Fujie W, Wakakura M: Prolonged constant eye
the treatment of ocular myasthenia (EPITOME): A randomized, con- misalignment leads to failure to develop binocular vision in childhood
trolled trial. Muscle Nerve 53:363-369, 2016 ocular myasthenia gravis. J Pediatr Ophthalmol Strabismus 46:358-361,
51. Ashraf VV, Taly AB, Veerendrakumar M, Rao S: Myasthenia gravis in 2009
children: A longitudinal study. Acta Neurol Scand 114:119-123, 2006 77. Peragallo JH, Velez FG, Demer JL, Pineles SL: Long-term follow-up of
52. Ionita CM, Acsadi G: Management of juvenile myasthenia gravis. Pediatr strabismus surgery for patients with ocular myasthenia gravis. J Neuro-
Neurol 48:95-104, 2013 ophthalmol 33:40-44, 2013

You might also like