Ez a közérdekűadat-igénylés csatolmányának HTML formátumú változata 'Semmelweis Egyetem Ritka Betegségek Intézet (MG) Myasthenia gravis diagnosztikai protokoll'.


Neurology. 2016 Jul 26; 87(4): 419–425. 
doi:  10.1212/WNL.0000000000002790 
PMCID: PMC4977114 
International consensus guidance for management of myasthenia 
gravis 

Executive summary 
Donald B. Sanders, MD, * Gil I. Wolfe, MD,* Michael Benatar, MD, PhD, Amelia Evoli, MD, Nils E. Gilhus, 
MD, Isabel Illa, MD, Nancy Kuntz, MD, Janice M. Massey, MD, Arthur Melms, MD, Hiroyuki Murai, 
MD, Michael Nicolle, MD, Jacqueline Palace, BM, DM, David P. Richman, MD, Jan Verschuuren, MD, 
and Pushpa Narayanaswami, MBBS, DM* 
Author information ► Article notes ► Copyright and License information ► 
See commentary "Treating myasthenia on consensus guide: Helpful and challenging but still unfinished business." on page 350. 
This article has been cited by other articles in PMC. 
Abstract 
Objective: 
Methods: 
Results: 
Conclusion: 
Acquired myasthenia gravis (MG) is a disorder of neuromuscular transmission, resulting from 
binding of autoantibodies to components of the neuromuscular junction, most commonly the 
acetylcholine receptor (AChR). The incidence ranges from 0.3 to 2.8 per 100,000,1 and it is 
estimated to affect more than 700,000 people worldwide. 
The increasing use of immunomodulating therapies has been a major factor in improving the 
prognosis for patients with MG in recent years.2 The various treatment options must be 
weighed in the context of individual patient factors. 
Why do we need MG guidance treatment statements? 
Although there is widespread agreement on the use of many treatments for MG, there is no 
internationally accepted standard of care. Because MG is heterogeneous, no one treatment 
approach is best for all patients. Few physicians treat enough patients with MG to be 
comfortable with all available treatments. Given its heterogeneity, the few randomized 
controlled trials (RCTs) in MG have limited generalizability, while uncontrolled trials are 
limited by potential bias. Hence, an effort to develop consensus among international experts 
was undertaken to guide clinicians worldwide on the multifaceted approach to managing MG. 
This summary condenses the extensive background information in the full guidance 
statements, available on the Neurology® Web site at Neurology.org. 
Panel constitution and method of expert consensus. 
In October 2013, a Task Force of the Myasthenia Gravis Foundation of America (MGFA) 
convened a panel of 15 international experts in MG to develop treatment guidance statements 
based on formalized consensus. The panel was chosen to represent the breadth of knowledge 
and experience and a wide variety of opinions from MG experts internationally. 

Development of preliminary definitions. 
The panel initially voted on definitions that formed the foundation for subsequent guidance 
treatment statements: goals of treatment, remission, ocular MG, impending and manifest 
myasthenic crises, and refractory MG. 
The Task Force co-chairs (D.B.S., G.I.W.) drafted initial definitions based on available 
literature.3 These were sent by e-mail to the panelists, who were asked to vote yes or no on 
each, and to provide modifications if they did not agree. Panelists were instructed not to 
discuss the definitions among themselves, and to send their votes only to the facilitator (P.N.). 
A simple consensus was used (≥80% of panelists voting yes). 
Definitions not achieving consensus were modified based on the panelists' suggestions and the 
modified definitions and discussions were shared with the panel for subsequent voting rounds. 
Development of guidance treatment statements. 
The following were agreed upon a priori: 
  1. Treatment costs and availability would not be considered, as it is not possible to 
make international consensus statements specific for all countries. 
  2. Clinical examination is assumed to have been performed by physicians skilled in 
the evaluation of neuromuscular disease. 
  3. The MGFA Clinical Classification, including remission, refers to the state of the 
patient at the time of evaluation. 
A formal systematic review of the literature was not performed. The Task Force co-chairs and 
facilitator drafted initial guidance statements based on literature cited in recent national and 
regional MG treatment guidelines,4,–supplemented by other literature. 
Guidance statements were developed for the following: 
  1. Symptomatic and immunosuppressive (IS) treatments 
  2. IV immunoglobulin (IVIg) and plasma exchange (PLEX) 
  3. Impending and manifest myasthenic crisis 
  4. Thymectomy 
  5. Juvenile MG (JMG) 
  6. MG with antibodies to muscle-specific tyrosine kinase (MuSK-MG) 
  7. MG in pregnancy 
Voting process for consensus guidance treatment statements. 
We used the RAND/UCLA Appropriateness Method (RAM) for formal consensus to quantify 
agreement.10 RAM uses a multi-round modified Delphi process to obtain a quantitative 
assessment that reflects the judgment of an expert group. Appropriateness refers to the 
relative benefit vs harm of the intervention. We obtained anonymous votes and feedback on 
each draft statement from the panelists, who rated each for appropriateness on a 9-point scale 
(1–3 inappropriate, 4–6 uncertain, and 7–9 appropriate). Panelists responded by e-mail to the 
facilitator, who tallied the votes and collated the discussions. Following each round of voting, 
statements were modified by the Task Force co-chairs and facilitator based on the panel 
feedback. Statements that did not achieve consensus within 3 rounds were excluded. 

For statements on symptomatic and IS therapies and thymectomy, an initial round of e-mail 
voting was followed by a meeting in Durham, North Carolina, on March 1, 2014. During this 
meeting, statements that had undergone prior voting by e-mail were refined with panel input, 
and a second round of voting was completed. All subsequent voting was by e-mail. 
The level of appropriateness and presence of agreement were determined for each statement 
as per RAM.10 
Go to: 
RESULTS 
All definitions below obtained simple consensus and all guidance statements below were 
agreed upon as being appropriate by the panel. Literature summaries and tables for 
medication dosing guidance and medication cautions are available as supplemental data 
at Neurology.org. 
Preliminary definitions. 
1. Goals for the treatment of MG. 
MGFA Task Force Post-Intervention Status (PIS) classification Minimal Manifestation Status 
(MMS) or better,3 with no more than grade 1 Common Terminology Criteria for Adverse 
Events (CTCAE) medication side effects.11 
MMS: The patient has no symptoms or functional limitations from MG but has some 
weakness on examination of some muscles. This class recognizes that some patients who 
otherwise meet the definition of remission have mild weakness. 
CTCAE grade 1 medication side effects: asymptomatic or only mild symptoms; intervention 
not indicated. 
2. Definition of remission. 
The patient has no symptoms or signs of MG. Weakness of eyelid closure is accepted, but 
there is no weakness of any other muscle on careful examination. Patients taking 
cholinesterase inhibitors (ChEIs) every day with reasonable evidence to support symptomatic 
benefit are therefore excluded from this category. 
3. Definition of ocular MG (based on dysfunction due to MG at a specified point 
in time, and not dependent upon the duration of disease). 
MGFA Class I3: Any ocular muscle weakness. May have weakness of eye closure. Strength in 
all other facial, bulbar, and limb muscles is normal. (It is recognized that some patients report 
fatigue when strength testing is normal. The physician should use clinical judgment in 
attributing fatigue to generalized MG in the absence of objective nonocular weakness). 
4. Definition of impending myasthenic crisis. 
Rapid clinical worsening of MG that, in the opinion of the treating physician, could lead to 
crisis in the short term (days to weeks). 
5. Definition of manifest myasthenic crisis (the concept of crisis focuses on the 
clinical implications—it represents a serious, life-threatening, rapid worsening 

of MG and potential airway compromise from ventilatory or bulbar 
dysfunction). 
MGFA Class V3: Worsening of myasthenic weakness requiring intubation or noninvasive 
ventilation to avoid intubation, except when these measures are employed during routine 
postoperative management (the use of a feeding tube without intubation places the patient in 
MGFA Class IVB3). 
6. Definition of refractory MG. 
PIS3 is unchanged or worse after corticosteroids and at least 2 other IS agents, used in 
adequate doses for an adequate duration, with persistent symptoms or side effects that limit 
functioning, as defined by patient and physician. 
Consensus guidance treatment statements. 
Symptomatic and IS treatment of MG. 
  1. Pyridostigmine should be part of the initial treatment in most patients with MG. 
Pyridostigmine dose should be adjusted as needed based on symptoms. The ability to 
discontinue pyridostigmine can be an indicator that the patient has met treatment goals 
and may guide the tapering of other therapies. Corticosteroids or IS therapy should be 
used in all patients with MG who have not met treatment goals after an adequate trial 
of pyridostigmine. 
  2. A nonsteroidal IS agent should be used alone when corticosteroids are 
contraindicated or refused. A nonsteroidal IS agent should be used initially in 
conjunction with corticosteroids when the risk of steroid side effects is high based on 
medical comorbidities. A nonsteroidal IS agent should be added to corticosteroids 
when: 
o  a. Steroid side effects, deemed significant by the patient or the treating 
physician, develop; 
o  b. Response to an adequate trial (table e-1) of corticosteroids is inadequate; or 
o  c. The corticosteroid dose cannot be reduced due to symptom relapse. 
  3. Nonsteroidal IS agents that can be used in MG include azathioprine, cyclosporine, 
mycophenolate mofetil, methotrexate, and tacrolimus. The following factors should be 
considered in selecting among these agents: 
o  a. There is widespread variation in practice with respect to choice of IS agent 
since there is little literature comparing them. 
o  b. Expert consensus and some RCT evidence support the use of azathioprine as 
a first-line IS agent in MG. 
o  c. Evidence from RCTs supports the use of cyclosporine in MG, but potential 
serious adverse effects and drug interactions limit its use. 
o  d. Although available RCT evidence does not support the use of 
mycophenolate and tacrolimus in MG, both are widely used, and one or both 
are recommended in several national MG treatment guidelines.4,–
  4. Patients with refractory MG should be referred to a physician or a center with 
expertise in management of MG. In addition to the previously mentioned IS agents, 
the following therapies may also be used in refractory MG: 
o  a. Chronic IVIg and chronic PLEX (see IVIg and PLEX, no. 6); 
o  b. Cyclophosphamide; 

o  c. Rituximab, for which evidence of efficacy is building, but for which formal 
consensus could not be reached. 
  5. IS agent dosage and duration of treatment 
o  a. Once patients achieve treatment goals, the corticosteroid dose should be 
gradually tapered. In many patients, continuing a low dose of corticosteroids 
long-term can help to maintain the treatment goal. 
o  b. For nonsteroidal IS agents, once treatment goals have been achieved and 
maintained for 6 months to 2 years, the IS dose should be tapered slowly to the 
minimal effective amount. Dosage adjustments should be made no more 
frequently than every 3–6 months (table e-1). 
o  c. Tapering of IS drugs is associated with risk of relapse, which may 
necessitate upward adjustments in dose. The risk of relapse is higher in patients 
who are symptomatic, or after rapid taper. 
o  d. It is usually necessary to maintain some immunosuppression for many years, 
sometimes for life. 
  6. Patients must be monitored for potential adverse effects and complications from IS 
drugs. Changing to an alternative IS agent should be considered if adverse effects and 
complications are medically significant or create undue hardship for the patient. 
IVIg and PLEX. 
  1. PLEX and IVIg are appropriately used as short-term treatments in patients with MG 
with life-threatening signs such as respiratory insufficiency or dysphagia; in 
preparation for surgery in patients with significant bulbar dysfunction; when a rapid 
response to treatment is needed; when other treatments are insufficiently effective; and 
prior to beginning corticosteroids if deemed necessary to prevent or minimize 
exacerbations. 
  2. The choice between PLEX and IVIg depends on individual patient factors (e.g., 
PLEX cannot be used in patients with sepsis and IVIg cannot be used in renal failure) 
and on the availability of each. 
  3. IVIg and PLEX are probably equally effective in the treatment of severe 
generalized MG. 
  4. The efficacy of IVIg is less certain in milder MG or in ocular MG. 
  5. PLEX may be more effective than IVIg in MuSK-MG. 
  6. The use of IVIg as maintenance therapy can be considered for patients with 
refractory MG or for those in whom IS agents are relatively contraindicated. 
Impending and manifest myasthenic crisis. 
Impending and manifest myasthenic crisis are emergent situations requiring aggressive 
management and supportive care. 
Although cholinergic crises are now rare, excessive ChEI cannot be completely excluded as a 
cause of clinical worsening. Also, ChEIs increase airway secretions, which may exacerbate 
breathing difficulties. 
PLEX and IVIg are the mainstay of management in myasthenic crisis. 
  1. Impending crisis requires hospital admission and close observation of respiratory 
and bulbar function, with the ability to transfer to an intensive care unit if it progresses 

to manifest crisis. Myasthenic crisis requires admission to an intensive care or step-
down unit to monitor for or manage respiratory failure and bulbar dysfunction. 
  2. PLEX and IVIg are used as short-term treatment for impending and manifest 
myasthenic crisis and in patients with significant respiratory or bulbar dysfunction. 
Corticosteroids or other IS agents are often started at the same time to achieve a 
sustained clinical response. (Because corticosteroids may cause transient worsening of 
myasthenic weakness, it may be appropriate to wait several days for PLEX or IVIg to 
have a beneficial effect before starting corticosteroids). 
  3. Although clinical trials suggest that IVIg and PLEX are equally effective in the 
treatment of impending or manifest myasthenic crisis, expert consensus suggests that 
PLEX is more effective and works more quickly. The choice between the 2 therapies 
depends on patient comorbidity (e.g., PLEX cannot be used in sepsis and IVIg is 
contraindicated in hypercoagulable states, renal failure, or hypersensitivity to 
immunoglobulin) and other factors, including availability. A greater risk of 
hemodynamic and venous access complications with PLEX should also be considered 
in the decision (many complications of PLEX are related to route of access and may 
be minimized by using peripheral rather than central venous access). 
Thymectomy in MG. 
  1. In non-thymomatous MG, thymectomy is performed as an option to potentially 
avoid or minimize the dose or duration of immunotherapy, or if patients fail to respond 
to an initial trial of immunotherapy or have intolerable side-effects from that therapy. 
Because of the long delay in onset of effect, thymectomy for MG is an elective 
procedure. It should be performed when the patient is stable and deemed safe to 
undergo a procedure where postoperative pain and mechanical factors can limit 
respiratory function. 
  2. The value of thymectomy in the treatment of prepubertal patients with MG is 
unclear, but thymectomy should be considered in children with generalized AChR 
antibody–positive MG: 
o  a. If the response to pyridostigmine and IS therapy is unsatisfactory; or 
o  b. In order to avoid potential complications of IS therapy. 
For children diagnosed with seronegative generalized MG, the possibility of a congenital 
myasthenic syndrome or other neuromuscular condition should be entertained, and evaluation 
at a center specializing in neuromuscular diseases is of value prior to thymectomy. 
  3. With rare exceptions, all patients with MG with thymoma should undergo surgery 
to remove the tumor. Removal of the thymoma is performed to rid the patient of the 
tumor and may not produce improvement in MG. All thymus tissue should be 
removed along with the tumor. Further treatment of thymoma will be dictated by 
histologic classification and degree of surgical excision. Incompletely resected 
thymomas should be managed after surgery with an interdisciplinary treatment 
approach (radiotherapy, chemotherapy). 
  4. In elderly or multimorbid patients with thymoma, palliative radiation therapy can be 
considered in the appropriate clinical setting. Small thymomas may be followed 
without treatment unless they are enlarging or become symptomatic. 
  5. Endoscopic and robotic approaches to thymectomy are increasingly performed and 
have a good track record for safety in experienced centers. Data from randomized, 
controlled comparison studies are not available. Based on comparisons across studies, 

less invasive thymectomy approaches appear to yield similar results to more 
aggressive approaches. 
  6. Thymectomy may be considered in patients with generalized MG without 
detectable AChR antibodies if they fail to respond adequately to IS therapy, or to 
avoid/minimize intolerable adverse effects from IS therapy. Current evidence does not 
support an indication for thymectomy in patients with MuSK, LRP4, or agrin 
antibodies. 
Juvenile MG (see also Thymectomy in MG, no. 2). 
  1. Children with acquired autoimmune ocular MG are more likely than adults to go 
into spontaneous remission. Thus, young children with only ocular symptoms of MG 
can be treated initially with pyridostigmine. Immunotherapy can be initiated if goals of 
therapy are not met. 
  2. Children are at particular risk of steroid side effects, including growth failure, poor 
bone mineralization, and susceptibility to infection, due in part to a delay in live 
vaccinations. Long-term treatment with corticosteroids should use the lowest effective 
dose to minimize side effects. 
  3. Maintenance PLEX or IVIg are alternatives to IS drugs in JMG. 
MG with MuSK antibodies. 
  1. Many patients with MuSK-MG respond poorly to ChEIs, and conventional 
pyridostigmine doses frequently induce side effects. 
  2. Patients with MuSK-MG appear to respond well to corticosteroids and to many 
steroid-sparing IS agents. They tend to remain dependent on prednisone despite 
concomitant treatment with steroid-sparing agents. 
  3. MuSK-MG responds well to PLEX, while IVIg seems to be less effective. 
  4. Rituximab should be considered as an early therapeutic option in patients with 
MuSK-MG who have an unsatisfactory response to initial immunotherapy. 
MG in pregnancy. 
  1. Planning for pregnancy should be instituted well in advance to allow time for 
optimization of myasthenic clinical status and to minimize risks to the fetus. 
  2. Multidisciplinary communication among relevant specialists should occur 
throughout pregnancy, during delivery, and in the postpartum period. 
  3. Provided that their myasthenia is under good control before pregnancy, the majority 
of women can be reassured that they will remain stable throughout pregnancy. If 
worsening occurs, it may be more likely during the first few months after delivery. 
  4. Oral pyridostigmine is the first-line treatment during pregnancy. IV ChEIs may 
produce uterine contractions and should not be used during pregnancy. 
  5. Thymectomy should be postponed until after pregnancy as benefit is unlikely to 
occur during pregnancy. 
  6. Chest CT without contrast can be performed safely during pregnancy, although the 
risks of radiation to the fetus need to be carefully considered. Unless there is a 
compelling indication, postponement of diagnostic CT until after delivery is 
preferable. 
  7. Prednisone is the IS agent of choice during pregnancy. 

  8. Current information indicates that azathioprine and cyclosporine are relatively safe 
in expectant mothers who are not satisfactorily controlled with or cannot tolerate 
corticosteroids. Current evidence indicates that mycophenolate mofetil and 
methotrexate increase the risk of teratogenicity and are contraindicated during 
pregnancy. (These agents previously carried Food and Drug Administration [FDA] 
Category C (cyclosporine), D (azathioprine and mycophenolate mofetil), and X 
(methotrexate) ratings. The FDA has recently discontinued this rating system, and 
replaced it with a summary of the risks of using a drug during pregnancy and 
breastfeeding, along with supporting data and “relevant information to help health care 
providers make prescribing and counseling decisions”12). 
  Although this statement achieved consensus, there was a strong minority opinion 
against the use of azathioprine in pregnancy. Azathioprine is the nonsteroidal IS of 
choice for MG in pregnancy in Europe but is considered high risk in the United States. 
This difference is based on a small number of animal studies and case reports. 
  9. PLEX or IVIg are useful when a prompt, although temporary, response is required 
during pregnancy. Careful consideration of both maternal and fetal issues, weighing 
the risks of these treatments against the requirement for use during pregnancy and 
their potential benefits, is required. 
  10. Spontaneous vaginal delivery should be the objective and is actively encouraged. 
  11. Magnesium sulfate is not recommended for management of eclampsia in MG 
because of its neuromuscular blocking effects; barbiturates or phenytoin usually 
provide adequate treatment. 
  12. All babies born to myasthenic mothers should be examined for evidence of 
transient myasthenic weakness, even if the mother's myasthenia is well-controlled, and 
should have rapid access to neonatal critical care support. 
Go to: 
DISCUSSION 
We have developed international guidance statements for the management of JMG and adult 
MG. We utilized recent national guidelines and a regional European guideline to assemble a 
foundation of literature, supplementing their comprehensive literature reviews with additional 
articles identified by panelists. After reaching agreement on the treatment goals, a 3-round 
anonymous modified Delphi voting process was used to obtain consensus on guidance 
statements. 
A limitation of consensus-based processes is that subconscious or conscious selection of like-
minded panel members may result in opinions that are not representative of MG experts. This 
issue was addressed by selecting an international panel with variations in practice and by 
using a formal consensus process. 
Recognizing the variability of practice patterns and availability of treatment modalities, these 
statements are not absolute recommendations for management, but are intended as a guide for 
the clinician. They are also not intended for establishing payment policies or drug tiering by 
payers. 
This is a living document that will require updates as the MG treatment theater continues to 
evolve. Despite the limitations of consensus-based methods, these guidance statements reflect 
an up-to-date expert consensus to guide clinicians worldwide who strive to optimize function 
and quality of life for their patients with MG, especially for those who practice in parts of the 

world that do not have the resources to develop local treatment guidelines. Any future trial of 
treatment that provides relevant information will merit review of these guidance statements. 
Go to: 
Supplementary Material 
Data Supplement:
 
Click here to view. 
Accompanying Editorial: 
Click here to view. 
Go to: 
ACKNOWLEDGMENT 
The authors thank MGFA Board member Jurgen Venitz, MD, PhD, for providing insight from 
the patients' perspective. 
Go to: 
GLOSSARY 
AChR 
acetylcholine receptor 
ChEI 
cholinesterase inhibitor 
CTCAE  Common Terminology Criteria for Adverse Events 
FDA 
Food and Drug Administration 
IS 
immunosuppressive 
IVIg 
IV immunoglobulin 
JMG 
juvenile myasthenia gravis 

MG 
myasthenia gravis 
MGFA  Myasthenia Gravis Foundation of America 
MMS 
minimal manifestation status 
MuSK  muscle-specific tyrosine kinase 
PIS 
Post-Intervention Status 
PLEX 
plasma exchange 
RAM 
RAND/UCLA Appropriateness Method 
RCT 
randomized controlled trial 
Go to: 
Footnotes 
Supplemental data at Neurology.org 
Editorial, page 350 
Go to: 
AUTHOR CONTRIBUTIONS 
D.B. Sanders: study concept and design, grant application and management, acquisition of 
data, analysis or interpretation of data, drafting/revising the manuscript, critical revision of the 
manuscript for important intellectual content, study supervision. G.I. Wolfe: study concept 
and design, acquisition of data, analysis or interpretation of data, drafting/revising the 
manuscript, critical revision of the manuscript for important intellectual content, study 
supervision. M. Benatar: acquisition of data, drafting/revising the manuscript, critical revision 
of the manuscript for important intellectual content. A. Evoli: acquisition of data, 
drafting/revising the manuscript, critical revision of the manuscript for important intellectual 
content. N.E. Gilhus: acquisition of data, critical revision of the manuscript for important 
intellectual content. I. Illa: acquisition of data, critical revision of the manuscript for important 

intellectual content. N. Kuntz: acquisition of data, drafting/revising the manuscript, critical 
revision of the manuscript for important intellectual content. J.M. Massey: acquisition of data, 
drafting/revising the manuscript, critical revision of the manuscript for important intellectual 
content. A. Melms: acquisition of data, critical revision of the manuscript for important 
intellectual content. H. Murai: acquisition of data, critical revision of the manuscript for 
important intellectual content. J. Palace: acquisition of data, drafting/revising the manuscript, 
critical revision of the manuscript for important intellectual content. M. Nicolle: acquisition of 
data, drafting/revising the manuscript, critical revision of the manuscript for important 
intellectual content. D.P. Richman: acquisition of data, critical revision of the manuscript for 
important intellectual content. J. Verschuuren: acquisition of data, critical revision of the 
manuscript for important intellectual content. P. Narayanaswami: study concept and design, 
acquisition of data, analysis or interpretation of data, drafting/revising the manuscript, critical 
revision of the manuscript for important intellectual content, study supervision. 
Go to: 
STUDY FUNDING 
Supported by a grant from the Myasthenia Gravis Foundation of America (MGFA). 
Go to: 
DISCLOSURE 
D. Sanders: consultant for Accordant Health Services, Cytokinetics, GlaxoSmithKline, and 
Jacobus Pharmaceutical Co. G. Wolfe: advisory board for Grifols, Baxter, CSL Behring, and 
Syntimmune; and research support from Alexion. M. Benatar: research support from the FDA 
(R01FD003710), NIH (U01NS084495), and Alexion. A. Evoli: scientific award jury member 
for Grifols. N. Gilhus: speaker's honoraria from Octopharma, Baxter, and Merck Serono. I. 
Illa: research funding from Grifols and speaking fees and travel grants from Genzyme and 
Pfizer; and consultant for Alexion, UCB, and Grifols. N. Kuntz and J. Massey report no 
disclosures relevant to the manuscript. A. Melms: advisory board, UCB. H. Murai: research 
funding from Biogen, Novartis, Bayer, Tanabe Mitsubishi, and the Japan Blood Products 
Organization. J. Palace: consultant for Merck Serono, Biogen Idec, Novartis, Teva, Chugai 
Pharma, Alexion, and Bayer Schering; research funding from Merck Serono, Novartis, 
Biogen Idec, and Bayer Schering. M. Nicolle reports no disclosures relevant to the 
manuscript. D. Richman: research funding from the Muscular Dystrophy Association, 
Myasthenia Gravis Foundation of California, and the Myasthenia Gravis Foundation of 
Illinois. J. Verschuuren: research support by NIH, FP7 European grant (#602420), consultant 
for Tyr pharma, and arGEN-X. The LUMC receives royalties from IBL for antibody tests. P. 
Narayanaswami: serves on the Medical and Scientific Advisory Board, Myasthenia Gravis 
Foundation of America (MGFA), and Medical Advisory Board, MGFA, New England 
Chapter. Go to Neurology.org for full disclosures. 
Go to: 
REFERENCES 
1. Deenen JCW, Horlings CGC, Verschuuren JJGM, Verbeek ALM. The epidemiology of 
neuromuscular disorders: a comprehensive overview of the literature. J Neuromuscul 
Dis 2015;2:73–85. [PubMed] 

2. Grob D, Brunner NG, Namba T, Pagala M. Lifetime course of myasthenia gravis. Muscle 
Nerve2008;37:141–149. [PubMed] 
3. Jaretzki A III, Barohn RB, Ernstoff RM, et al. Myasthenia gravis: recommendations for 
clinical research standards: Task Force of the Medical Scientific Advisory Board of the 
Myasthenia Gravis Foundation of America. Neurology 2000;55:16–23. [PubMed] 
4. Wiendl H. Diagnostik und Therapie der Myasthenia gravis und des Lambert-Eaton-
Syndroms [online]. Available at: http://www.dgn.org/leitlinien/11-leitlinien-der-dgn/3005-ll-
68-ll-diagnostik-und-therapie-der-myasthenia-gravis-und-des-lambert-eaton-syndroms. 
Accessed August 14, 2015. 
5. Fuhr P, Gold R, Hohlfeld R, et al. Diagnostik und therapie der myasthenia gravis und des 
Lambert-Eaton Syndroms. In: Diener HC, Weimar C, Deuschl G, et al. eds. Leitlinien für 
Diagnostik und Therapie in der Neurologie, 5th ed Stuttgart: Thieme; 2012:830–856. 
6. Murai H. Japanese clinical guidelines for myasthenia gravis: putting into practice. Clin Exp 
Neuroimmunol 2015;6:21–31. 
7. Sussman J, Farrugia ME, Maddison P, Hill M, Leite MI, Hilton-Jones D. Myasthenia 
gravis: association of British Neurologists' management guidelines. Pract 
Neurol 2015;15:199–206. [PubMed] 
8. Norwood F, Dhanjal M, Hill M, et al. Myasthenia in pregnancy: best practice guidelines 
from a UK multispecialty working group. J Neurol Neurosurg Psychiatry 2014;85:538–
543. [PubMed] 
9. Skeie GO, Apostolski S, Evoli A, et al. Guidelines for treatment of autoimmune 
neuromuscular transmission disorders. Eur J Neurol 2010;17:893–902. [PubMed] 
10. Fitch K, Bernstein SJ, Aguilar MD, et al. The RAND/UCLA Appropriateness Method 
User's Manual. Santa Monica, CA: RAND Corporation; 2001. 
11. US Department of Health and Human Services. Common Terminology Criteria for 
Adverse Events (CTCAE) v4.03 [online]. Available 
at: http://evs.nci.nih.gov/ftp1/CTCAE/CTCAE_4.03_2010-06-14_QuickReference_5x7.pdf. 
Accessed May 25, 2016. 
12. US Food and Drug Administration. Pregnancy and Lactation Final Rule [online]. 
Available 
at: http://www.fda.gov/Drugs/DevelopmentApprovalProcess/DevelopmentResources/Labelin
g/ucm093307.htm. Ac
cessed November 11, 2015.