Acquired, autoimmune myasthenia gravis (MG), the most common noniatrogenic disorder of neuromuscular transmission (DNMT), remains the favorite child of many neuromuscular clinicians. Arguably, it provides more professional satisfaction than any other neuromuscular disease. This fulfillment is derived in part from the intellectual satisfaction that comes from understanding disease pathogenesis. At the same time, satisfaction is gained by the ability to make meaningful improvements in patient function and quality of life through the application of rational and effective treatment. MG represents one of medicine’s most notable translational successes in bringing basic science to the bedside.
Although descriptions of individuals likely to have been affected by MG can be traced to antiquity, our current understanding of disease mechanism(s) originates from a series of seminal observations and discoveries. Thomas Willis, a seventeenth-century physiologist, is frequently credited for initially describing the clinical syndrome of MG. The concept of MG as a DNMT and the first therapeutic triumph are often credited to British clinician Mary Walker. She described the benefits of cholinesterase inhibitors in the 1930s; her discovery extrapolated from her observations related to the similarities between MG and curare toxicity. In 1960, Simpson first promoted the hypothesis of an autoimmune basis for MG on the basis of his observations of an increased prevalence in young women and in individuals with other autoimmune diseases.1 In that same decade, support for MG as a DNMT was provided by the in vitro electrophysiological demonstration that miniature end plate potential (EPP) amplitudes in MG were greatly reduced.2 In 1973 Daniel Drachman et al. solidified the concept of MG as a postsynaptic DNMT by demonstrating loss of acetylcholine receptors (AChR) in MG patients through α-bungarotoxin labeling techniques.3 In the same year, Patrick and Lindstrom confirmed the autoimmune nature of MG with the development of an experimental MG model in rabbits who became weak when immunized with AChR.4 In 1976, the seminal article describing the value of AChR autoantibody testing in the diagnosis of myasthenia was published.5 In 2001, Hoch et al. first reported the association between MG and autoantibodies directed against muscle-specific kinase (MuSK).6 In 2008, unnamed autoantibodies directed at clustered AChRs were found in low titer in the serum of approximately two-thirds of AChR and MuSK seronegative patients.7,8 In 2011, patient’s with autoantibodies directed at the lipoprotein receptor protein 4 (LPR4) were identified as a third MG serotype.9
Historically, seronegative MG referred to patients lacking AChR autoantibodies. With the discovery of MuSK autoantibodies, the concept of double seronegative MG patients was coined. As our knowledge of LPR4 MG is somewhat limited, and as LPR4 autoantibody tests are not commercially available at the time of writing this, seropositive will be used in this chapter to refer only to AChR and MuSK MG.
The incidence of MG and its serodistribution may vary with geography and/or ethnicity.10 MG has been estimated to occur in 2–10/106 individuals/year in the Eastern United States and the Netherlands but up to 20/106 people/year in Eastern Spain.11–13 The prevalence is estimated to be as infrequent as 2 and as frequent as 200/106 individuals.13–24 The age of onset may also be influenced by geographical and/or ethnic differences. Juvenile onset MG is uncommon in occidental populations but may represent more than half of cases in Asian populations.25 Heritable MG is rare, estimated to occur in approximately 2% of cases although the concordance rate in monozygotic twins is estimated at 40%.23,26–28 Under the age of 40, AChR MG is almost three times more common in women. Men and women in their 40s however, are affected with equal frequency whereas in older individuals, the prevalence is greater in men at a ratio of 1.5:1.29,30
Understanding normal and abnormal neuromuscular transmission (NMT) is relevant not only to MG but to other DNMT that may be either acquired or heritable, resulting from genetic, infectious/toxic, or paraneoplastic/autoimmune mechanisms. These mechanisms typically act individually, but be synergistic. For example, a family has been recently reported in which there appears to be synergy between genetic and autoimmune influences. Multiple family members with seropositive MG were found to have mutations in the ecto-NADH oxidase 1 gene (ENOX1), a gene expressed in both thymus and skeletal muscles.31 The mechanism by which this mutation, posited to predispose to autoimmunity, might cause MG remains unknown. It is thought to relate to sequence homologies between ENOX1 and the main immunogenic region (MIR) of the alpha-1 subunit of the AChR. This provides a theoretical explanation for a heritable and anatomically targeted autoimmune disorder.32
This chapter will focus on acquired, autoimmune MG, a postsynaptic disorder representing the prototype of DNMT. Chapter 26 will discuss the less frequently occurring infectious/toxic and genetic (i.e., CMS or congenital myasthenic syndromes) DNMT, both categories which can be conceptualized and categorized as adversely affecting NMT at the presynaptic, synaptic, or postsynaptic levels.
Acquired MG typically evolves subacutely over days to months and reaches its clinical nadir within 2 years.33 The majority of patients are seropositive and usually have similar phenotypes regardless of the existence or type of autoantibody. Individual cases may have differing clinical features and natural histories, that may allow prediction of an individual serotype and different responsiveness to treatment.10 Each section of this chapter will describe the general features of the disease in its classical form associated with binding autoantibodies directed toward the AChR, and will then distinguish the differing features of other serotypes when applicable.
The natural history of myasthenia is difficult to predict in an individual patient. Information is, in a large part, derived from historical data accumulated prior to the availability of current therapeutic options.13 It is recognized that approximately 15–20% of Caucasian patients with initial signs and symptoms restricted to the oculomotor system will retain this restricted phenotype.33 Ninety percent of those destined to develop generalized disease will do so within 2 years of symptom onset and a similar number will reach the nadir of their disease within a 7-year period.13,34,35 It has been suggested, but by no means universally accepted, that immunomodulating treatment may diminish the risk of evolving into generalized disease.36,37 Spontaneous, remissions unrelated to treatment are estimated to occur in between 10-22% of MG patients.13,30 These remissions may occur at any time in the course of the disease. Half of the patient’s who achieve spontaneous remission, relapse within 6 months and 90% within a year.13,30 Regarding therapeutic decisions, however, there is no apparent correlation between the existence and length of remission and maximal disease severity.13 Mortality statistics in MG has been undoubtedly altered by therapeutic intervention. At the turn of the twentieth century it was approximated at 70%, reduced to 23–30% by the 1950s, with contemporary estimates in the 1.2–2.2% range.13,23,38
The diagnosis of myasthenia is clinically established by two phenotypic features, the pattern of weakness and its tendency to fluctuate. There are theories underlying the selected vulnerability of certain muscles in MG that will be addressed in the pathophysiology section. The basis for the characteristic patterns of weakness in MG remains, however, largely speculative.39 It may be related to the distribution of different types of neuromuscular junctions (NMJs) in different muscles.
The fluctuating nature of myasthenic symptoms is related to the dynamic biology of the NMJ.40 It is a quality of the disease that may be a dominant feature of the patient’s history or may be overlooked. MG patients may recognize that their symptoms may vary on a minute-to-minute, diurnal, or week-to-week basis.41–44 For example, a patient may describe normal articulation at the onset of a telephone conversation and may have unintelligible speech 5 minutes later. Patients may observe normal eyelid position upon awakening and then develop ptosis as the day wears on. Fluctuation may not be simply diurnal and patients may have functional hardships one month that seem to improve on their own the next month without apparent explanation. Fluctuations may also occur in response to identifiable variables such as temperature, systemic infection, menses, anxiety, emotional stress, and pregnancy.18,45–50 Myasthenic visits to emergency rooms are known to increase in frequency in early morning hours in equatorial countries where electricity and therefore air conditioning may not be available at night, making this potentially the warmest period of the day. Variability may also occur not only in the timing but in the pattern of weakness. Alternating ptosis represents the most notable example of this phenomenon. It should be emphasized that diurnal worsening of strength and stamina is not pathognomonic of MG as the weakness of any neuromuscular disease may worsen as the day goes on.
MG may also be suspected by the pattern of weakness. MG should be considered in any patient with painless weakness, particularly when the weakness is multifocal or diffuse in distribution or when weakness of ocular and bulbar muscles predominates. Asymmetry is not uncommon, particularly with ptosis. Most myasthenics will experience ptosis, diplopia, dysphagia, dysphonia, difficulty chewing, or symptoms referable to facial or neck weakness, either in isolation or in combination. Identification of weakness in muscles innervated by anatomically unrelated cranial nerves, for example, concomitant weakness of eyelid opening and eyelid closure represents a common and diagnostically useful example.
Initial symptoms restricted to ptosis or diplopia will be the presenting symptoms of MG patients in 65–85% of cases and 95% will experience oculomotor involvement at some point in their illness.33,37,43,51,52 Overt diplopia may be preceded by nonspecific visual blurring when ocular malalignment is minimal and insufficient to produce two distinct images. The presence of other signs and symptoms of myasthenia or the resolution of blurring the covering of either eye aids in the identification of blurring due to ocular malalignment. Other than the levator palpebrae, the medial rectus appears to be most susceptible of the extraocular muscles. Any pattern of ophthalmoparesis may occur, however, potentially mimicking an individual cranial nerve palsy or intranuclear ophthalmoplegia, or at times even producing nystagmus.53,54,55
Bulbar symptoms typically refer to disordered speech and swallowing but will be extended here to include weakness of jaw, facial, and neck muscles. Bulbar onset of MG is quite common particularly in our experience in older men, and is estimated to occur in approximately 15% of cases.55 Weakness of bulbar muscles is estimated to occur in >40% of MG patients sometime in their illness. As bulbar weakness may be associated with considerable morbidity, we consider bulbar symptoms to be indicative of generalized MG; ocular MG being restricted to ptosis and diplopia. Speech may be adversely affected in a number of ways. The voice may be hypophonic due to vocal cord paresis or expiratory muscle weakness.56–58 It may have a nasal quality due to palatal insufficiency and nasal air leak. The patient may be dysarthric as a result of weakness of the lips, tongue, or cheeks. Nasal regurgitation of food and liquid, difficulty manipulating food due to tongue weakness, as well as ineffective sniffing, coughing, nose blowing, or throat clearing may be noted.
Facial weakness is common. Lower facial weakness may result in dysarthria or sialorrhea, or in difficulty whistling, inflating balloons, or drinking from a straw. It may also interfere with the accuracy of pulmonary function testing due to poor oral seal. Weakness of upper facial muscles is equally prevalent but less likely to be symptomatic. Occasionally, patients may complain of visual blurring due to lower lid weakness resulting in pooling of tears. Facial weakness can be easily missed if not sought for, particularly if bilateral. Patients who are affected may be unable to bury their eyelashes or maintain eye closure against resistance. They may be unable to whistle, make a kissing noise, or hold air in their distended cheeks against resistance. A “myasthenic snarl,” may occur in which the mid-portion of the upper lip elevates without elevation of the corners of the mouth during an attempted smile.
Symptoms and signs referable to jaw weakness, particularly jaw closing, occur fairly commonly in MG. Although jaw weakness may occur in other neuromuscular diseases, many of the other causes, for example, Guillain–Barré syndrome, are unlikely to have a phenotype readily confused with MG.59 Like most neuromuscular diseases, neck flexor weakness is more common and more pronounced than neck extensors weakness in the majority but not in all MG patients. Head drop is not rare however, and may be the presenting symptom. Like other causes of head drop, posterior neck pain relieved by head support may be the most prominent symptom associated with this sign, presumably related to the stretch placed on posterior cervical muscles and ligaments by the weight of the head. Ventilatory insufficiency is a rare presenting symptom of MG but may develop in a significant percentage of patients with untreated or refractory generalized disease.58,60 Along with dysphagia, it is undoubtedly the predominant basis for mortality in MG unrelated to complications of treatment or immobility.
Limb weakness occurs in 20–30% of affected individuals. Limb weakness preferentially affects proximal muscles.61,62 One potential but undoubtedly partial explanation is the warmer temperature of the proximal limbs.63 Typically, limb weakness occurs in concert with the signs and symptoms of oculobulbar disease. In approximately 10% of patients with limb involvement, MG may be initially restricted to distal limb muscles, with foot or finger drop being notable presentations that have been reported and that we have seen.64–69 Again, the pattern may be focal, multifocal, or diffuse.43
Approximately 7–15% of all MG patients or 40% of seronegative patients with generalized MG are estimated to have MuSK autoantibodies.6,10,55,70–76 On average, MuSK MG patients are younger and more severely affected than their AChR MG counterparts although MuSK MG rarely if ever develops in an individual in the first decade of life.10,77,78 Disease prevalence in MuSK MG is highest in the third and fourth decades of life in non-Asian populations. In some series, males and females are affected equally whereas in other series, females have predominated.73,77,79–81
Most MuSK MG patients will have a phenotypic pattern indistinguishable from AChR MG patients. Nonetheless, there are certain clinical features that suggest an increased or decreased probability of MuSK MG.70–88 For example, purely ocular disease is a rare MuSK MG phenotype.71–73,77,86,87 Patterns that suggest an increase probability of MuSK MG include persistent bulbar symptoms refractory to treatment, prominent neck, facial, and ventilatory muscle weakness and the presence of muscle atrophy in an otherwise typical MG patient, notably in the tongue.10,72 The latter finding may render the clinical distinction from bulbar amyotrophic lateral sclerosis (ALS) more difficult.55,77,79,88 As MuSK is known to facilitate reinnervation, it is plausible that the atrophy noted in this serotype may be related to a “denervating” effect of MuSK autoantibodies.89 MuSK MG patients in general have more severe disease both at onset and disease nadir, are less likely to achieve complete remission with treatment, and are more likely to experience myasthenic crises than either their AChR or seronegative counterparts.10,77–79
Seronegative MG may represent up to 34% of MG patients depending on the study and the ethnic background of the cohort studied.10 In 2008, a high sensitivity assay discovered IgG1 autoantibodies in two-thirds of these seronegative MG patients.7 In 2011, an epitope on the LPR4 protein was found to be the target of IgG1 autoantibodies, presumably the same autoantibodies described 3 years previously.9 These patients, historically included in the seronegative group, represent a very small percentage of MG patients.89 Studies to date have estimated that LPR4 autoantibodies are found in 3%, 9%, and 50% of seronegative MG patients.9,25,90 These significant differences may represent differences between ethnicities, differences in methodology, or accuracies relevant to small sample size; the 50% figure originating from a study of only 13 patients.90
The weight of current evidence suggests seronegative MG, with or without LPR4 autoantibodies, manifests phenotypic, natural history and response to treatment features similar, if not identical, to AChR MG.7,10,91 Otherwise seronegative patients possessing low titers of IgG1 autoantibodies directed toward clustered AChRs have been reported to exist in approximately two-thirds of all seronegative AChR/MuSK MG and half of ocular seronegative MG patients, potentially explaining at least in part the high prevalence of conventional seronegativity in this latter population.7,8,91
The examination of the suspected or established MG patient includes a number of unique or relatively unique features. As ptosis is such a common manifestation of MG, documentation of the baseline upper and lid positions in relationship to the pupil is recommended prior to provocative testing and to distinguish ptosis from squinting, both of which narrow the palpebral fissure.
In MG, pupil function is spared although physiological anisocoria is common enough to provide a potentially confounding feature. In order to unmask or exacerbate ptosis or extraocular muscle weakness, the suspected MG patient is asked to sustain either up or lateral gaze for a minute while limiting blinking as much as possible (Fig. 25-1A–C). Drifting of lid or eyeball position is watched for. Cogan’s lid twitch is another sign thought to be a relatively specific, although not necessarily sensitive, sign in MG assessment. With this maneuver, the patient is first asked to look down and then rapidly saccade to reassume the primary position. Normally the eyelid moves synchronously with the eyeball. A positive sign is defined by the eyelid overshooting the eyeball position leading to a transient scleral exposure and upper lid oscillation.
Figure 25-1. (A–C) Ocular myasthenia with fatigable L ptosis (A) immediately upon sustained upgaze, (B) 30 seconds into sustained upgaze, and (C) after completion of 1 minute of upgaze demonstrating left > right ptosis. In (C), note subtle elevation of left eyebrow as indicator of frontalis use in attempt to compensate for ptosis.
Another maneuver with potential diagnostic benefit has been referred to as enhanced ptosis.92 This maneuver has been described in patients with bilateral asymmetric ptosis but is conceptually of value in apparent unilateral ptosis as well. In this maneuver, the clinician manually elevates the most affected eyelid which may result in the revelation or exacerbation of ptosis on the opposite side in an MG patient. The proposed explanation for this phenomenon is Hering’s law of equal innervation. Theoretically, with manual lid elevation, there is less need for supranuclear stimulation of the levator subnucleus of the oculomotor nerve. As this is a single midline nucleus which innervates both levator palpebrae muscles, lifting the more severely affected lid leads to the need for less supranuclear stimulation of this subnucleus affecting the contralateral as well as ipsilateral levator palpebrae.
The last bedside maneuver relative to the evaluation of ptosis is the icepack test which relies on the recognized physiological enhancement of NMT by cooling. In this maneuver, an ice pack applied to a closed eyelid may result in notable improvement of existing ptosis. As the icepack is potentially noxious, exposure should be limited to a minute or less.
As motor neuron disease is often the major differential diagnostic consideration in patients with painless weakness who do not have ptosis or diplopia, the clinical assessment of the suspected MG patient includes careful observation of muscles for atrophy and fasciculations. Although muscle atrophy may occur in MG patients, particularly in those with MuSK autoantibodies, it is typically notable for its absence. Demonstrating weakness at the bedside that worsens with sustained or repetitive use in limb and trunk muscles is of theoretical, but in our experience, limited value in suspected MG patients. Any cause of neuromuscular weakness may produce reduced strength on repeated effort and normal patients may be reluctant to sustain effort resulting in a false perception of fatigable weakness. A careful assessment of cranial muscle strength is, however, very important in the assessment of suspected or known MG patients. We typically assess the strength of eyelid and lip closure, jaw opening and closing, tongue protrusion, and neck flexion and extension. In our experience, residual evidence of eye closure weakness in patients who otherwise seem to be in remission is a fairly common finding and should not by itself represent a justification to alter treatment. Again, demonstrating weakness in two or more muscles innervated by anatomically unrelated nerves, in the absence of atrophy and fasciculations, pain, sensory, or autonomic symptoms is likely to represent MG. As symptoms suggesting inadequate breathing warrant serious attention, we find bedside or telephone-based estimates of ventilatory muscle strength helpful. We do this by having the patients count out loud as far as one’s vital capacity will allow after full inspiration. The vital capacity in cubic centimeters can be estimated with reasonable accuracy by multiplying this number by one hundred.
There are special considerations in pregnant women with myasthenia, their newborn children, and children with myasthenia.93–97 In an extensive review of the literature involving 322 pregnancies in 225 myasthenic mothers, 31% of mothers had no change in their myasthenic symptoms, 28% improved, and 41% deteriorated during the pregnancy.98 During the postpartum period, 30% had a disease exacerbation. There is a theoretical risk of transmitting IgG AChR antibodies in breast milk, although most infants have no problem with breastfeeding.
Transient neonatal autoimmune MG develops in approximately 10% of infants born to mothers with MG.100–112 It has been reported to occur in MuSK MG.113 Maternal treatment seems to significantly lower the risk of infantile disease.114 Onset is usually within the first 3 days of life. The most notable features include a weak cry, difficulty feeding due to a poor suck, hypotonia, respiratory difficulty, ptosis, and diminished facial expression resulting from facial muscle weakness. The disorder is temporary in most cases, with a mean duration of about 18–20 days. In rare cases, in utero paralysis may lead to a child born with multiple joint contractures.114 Other than recognition and symptomatic treatment, the most important aspect of this disorder is that there appears to be no increased risk of the child developing MG in later life.
Juvenile MG represents a “subclassification” of autoimmune MG.96–98,115–118 It is estimated that approximately 10% of acquired (non-neonatal) autoimmune MG cases will occur before 18 years of age in occidental populations, the majority subsequent to puberty.114,115,117 This statistic may be inflated, as some reported juvenile, seronegative cases could easily represent congenital myasthenic syndromes (CMS). The clinical features are similar to adult-onset MG, with the majority of patients initially presenting with primarily ocular symptoms.117 Serum AChR antibodies are present in the majority of affected children. The electrophysiologic findings are also identical to the adult form of the disease.118
Effective management of MG requires knowledge of disorders that occur with an increased incidence in patients with MG, particularly thymic abnormalities and other autoimmune diseases which may occur separately or overlap. Thymus gland pathology is the most notorious disease association.23,71,119 As many as 80% of patients with seropositive MG have thymic hyperplasia while approximately 12% have thymoma.23,120–122 Hyperplasia is more common in younger patients. Thymomas occur equally in men and women and occur with the greatest frequency in middle-aged and older individuals.23 Patients with thymoma, on average, have more severe clinical presentations and higher AChR antibody titers than their nonthymomatous counterparts. Thymic abnormalities have been documented to occur in MuSK MG but are thought to occur far less frequently.10,23,71,74,78,81,123 Thymic pathology, either hyperplasia or thymoma, occurs in seronegative and presumably in LPR4 MG.23 In patients with thymoma, slightly more than half will be found to have or will develop MG.23
Thymomas are not uniquely associated with MG and may coexist with other autoimmune disorders, other autoantibodies, and a host of other neurological and neuromuscular disorders (Fig. 25-2A,B).124 Reported associations include granulomatous myositis, myocarditis, Isaacs’ syndrome, rippling muscle disease, limbic and cerebellar encephalitis, and autonomic neuropathy including the syndrome of intestinal pseudoobstruction.125–130 Eleven patients with a particularly severe phenotype characterized by bulbar involvement, myasthenic crises, thymoma, myocarditis, and prolonged QT electrocardiographic interval have been described associated with Kv1.4 voltage-gated potassium channel in addition to AChR-binding autoantibodies.129
Figure 25-2. CT scan of the chest in axial (A) and sagittal (B) orientation revealing thymoma in an MG patient.
Other autoimmune diseases, most notably Hashimoto’s disease, occur with increased frequency in MG in addition to rheumatoid arthritis, systemic lupus erythematosus, Sjögren’s syndrome, red blood cell aplasia, ulcerative colitis, sarcoidosis, Addison’s disease, and hyperparathyroidism.131–135 Predictably, these disorders may occur with or without thymic abnormalities. Other neurological or neuromuscular disorders reported to coexist with MG include acute and chronic inflammatory demyelinating polyneuropathies, autonomic neuropathy (e.g., intestinal pseudoobstruction) with or without encephalopathy, Lambert–Eaton myasthenic syndrome (LEMS), acquired neuromyotonia or Isaacs’ syndrome, acquired rippling muscle disease and stiff person syndrome125,128,136–153 In addition, approximately 5% of patients with MG also have an inflammatory myopathy.137,154–157 Most of these patients also have a thymoma with or without myocarditis. The histopathology often reveals a giant cell or granulomatous myositis. Serum CK levels are usually elevated with concomitant inflammatory myopathy, which would not be expected in MG alone. It is estimated that the coexistence of other autoimmune diseases approximates 30% in AChR or in seronegative MG as opposed to MuSK MG where the prevalence of other autoimmune disease is estimated to approximate 20%.71
The diagnosis of myasthenia is usually established clinically and supported by a positive response to one or more of the following tests:
• serological—autoantibodies against the AChR or MuSK
• pharmacological—response to edrophonium
• electrophysiological—repetitive nerve stimulation (RNS) or single-fiber electromyography (SFEMG)
As has been repeatedly emphasized, MG should be considered in any patient with painless weakness, occurring in a regional, multifocal, diffuse, or even a seemingly focal pattern. The likelihood of MG increases substantially with the objective demonstration of fatigable weakness, particularly in an oculobulbar distribution. Weakness may be asymmetric and occurs in the absence of fasciculations and in most cases, muscle atrophy.
The differential diagnosis of MG includes other disorders in which signs and symptoms reside predominantly if not exclusively within the voluntary motor system (Table 25-1). Considerations include other DNMT such as the CMS, other acquired DNMT such as LEMS or botulism, motor neuron diseases such as spinal muscular atrophy (SMA), ALS, or X-linked spinal bulbar muscular atrophy (SBMA), numerous myopathies, particularly those with a predilection for cranial musculature, or motor neuropathies when the weakness is largely confined to the limbs.
TABLE 25-1. DIFFERENTIAL DIAGNOSIS OF MG
Brain
Wernicke encephalopathya
Rabiesa
Brainstem neoplasma
Anterior horn cell
ALS
Kennedy syndromea
Spinal muscular atrophya
Enterovirus infectiona
Root/nerve
Chronic meningitisa
Miller Fisher syndromea
Diphtheriaa
Immune-mediated motor neuropathiesa
NMJ
Congenital myasthenic syndromes
Lambert Eaton myasthenic syndrome
Botulism
Tick paralysis
Muscle
Oculopharyngeal MD
Myotonic MDa
Mitochondrial myopathy
Congenital myopathya
Inflammatory myopathya
Miscellaneous
Depressiona
Chronic fatiguea
Dysthyroid ophthalmopathya
Orbital pseudotumora
aUnlikely source of confusion.
Due to frequently overlapping phenotypes, and potentially similar electrophysiological features and pharmacological response, the most vexing of these considerations are the CMS. CMS should be seriously considered in any child, adolescent, or young adult with apparent seronegative MG. A history suggesting other involved relatives and/or consanguinity increases the probability of CMS. The CMS will be considered in more detail in the subsequent chapter.
In adults with bulbar weakness, considerations other than MG include but are not limited to the progressive bulbar palsy form of ALS, Kennedy disease, LEMS, botulism, and both acquired and hereditary myopathies. Congenital myasthenia cannot be entirely excluded from consideration as the DOK-7 and rapsyn mutations in particular may be associated with a late-onset phenotype and be readily misidentified as seronegative MG.158 Most causes of multiple cranial neuropathies typically produce sensory in addition to motor symptoms but occasionally these syndromes may be motor predominant. As the presence of ptosis or ophthalmoparesis essentially excludes motor neuron disease or inflammatory myopathy from consideration, careful surveillance for these abnormalities provides valuable differential diagnostic insight. LEMS is usually dominated by symptoms referable to proximal limb muscles with prominent fatigue and symptoms of cholinergic dysautonomia, but ptosis and bulbar symptoms do occur and can make its distinction from MG challenging in some cases.159,160 Botulism can affect children and adults and as a presynaptic disorder, can produce cholinergic dysautonomia with constipation and enlarged, unreactive pupils. Its acuity and the clinical context in which it occurs are helpful features to help distinguish it from MG. A number of inherited muscle diseases such as oculopharyngeal muscular dystrophy, mitochondrial myopathy, myotonic muscular dystrophy, and rare adult-onset cases of congenital myopathy such as centronuclear myopathy may produce oculobulbar syndromes.
As mentioned above, MG may rarely present with weakness restricted itself to limb or trunk muscles. The pattern of weakness may be predominantly proximal and symmetric or distal and asymmetric. Accordingly, the differential diagnostic considerations are broad and include anterior horn cell diseases, myopathies, neuropathies with motor predominance including some cases of inflammatory demyelinating polyneuropathy, multifocal motor neuropathy in more chronic and focal cases, as well as other forms of DNMT such as LEMS, botulism and acute organophosphate poisoning. Many of these diseases commonly have cranial nerve involvement as well. In children, SMA, congenital myasthenia, botulism, tick paralysis, and various myopathies would be the primary considerations. Poliomyelitis or other enteroviral infections would readily distinguish themselves in most cases but require consideration along with botulism and tick paralysis in any acute–subacute-onset case due to their pure motor characteristics.
Successful NMT is dependent on the anatomical and physiological capabilities of the NMJ to translate and amplify a peripheral nerve action potential (NAP) into a transsynaptic chemical signal mediated by the neurotransmitter acetylcholine (ACh). Subsequently, the NMJ promotes the generation of a postsynaptic EPP, which unlike an action potential, varies in amplitude. If this EPP achieves the necessary magnitude, an action potential will be generated in the corresponding muscle fiber. This single muscle fiber action potential (SMFAP) tranduces the electrical event into a chemical and eventually mechanical event. The SFMAP promotes calcium release into the sarcoplasmic reticulum which is responsible for myofiber contraction and the generation of force.161 The amount of force generated is dependent on the number of motor units activated, and ultimately, the number of muscle fibers in which this sequence of events takes place.
NMT is a process that is highly conserved between species. It has evolved into an extremely efficient system empowered with a substantial safety factor allowing for fail-safe repetitive and sustained muscle contraction in normal individuals. We have obtained considerable knowledge about the complexities of both normal and abnormal NMT. This section will address NMT and DNMT from a very superficial, clinically relevant perspective.
In view of the sophisticated evolution of NMT into a highly efficient system, external influences are more likely to compromise rather than enhance this efficiency. When NMT is compromised, it typically results in a phenotype typically dominated by fatigue and skeletal muscle weakness. As the neuromuscular junction lies beyond the protection of the blood–nerve barrier and is composed of a large number of proteins essential to its optimal function, it is vulnerable to a large number of immune-mediated, toxic, and genetic influences that can adversely affect both its function and structure. DMNT may result from anatomical and/or physiological disruption of one or more of the three components of the NMJ: (1) the presynaptic nerve terminal in which the synthesis, packaging, storage, presynaptic membrane binding, and/or release of ACh and the vesicles (quanta) that contain it take place, (2) the synaptic cleft through which ACh migrates and is eventually metabolized, and/or (3) the postsynaptic muscle membrane where specialized ACh receptors/channels are optimally positioned and organized (Figs. 23-3 and 23-4). Postsynaptic DNMT may result from multiple potential mechanisms including interference with ACh binding, AChR organization, increased AChR turnover, or overt anatomic disruption (Fig. 23-5).38,55 This section will review the anatomical, biochemical, and physiological aspects of normal and abnormal NMT.
Although DNMT are usually categorized as belonging to one of the three aforementioned anatomical domains, it would be overly simplistic to assume that all NMJs are the same, that each of the three anatomic NMJ domains develops embryologically in isolation, that any domain functions independently of the other two, or that any DNMT results solely from dysfunction of an individual domain. For example, the presynaptic configurations may differ between different NMJs in different muscles with terminal twigs having either an “en plaque” or “en grappe configuration.” The former refers to large, single contacts on each muscle fiber. It is the predominant form in most mammalian muscles. The latter array refers to multiple, smaller contact points on individual fibers. This configuration seems to correlate with the need for tonic muscle contraction and is most prevalent in nonmammalian systems, but exists in humans in extraocular muscles in particular and in the tensor tympani, stapedius, laryngeal muscles, and tongue as well.39 The greater concentration of fetal-type AChR in extraocular muscles represents one hypothesis as to why these muscles appear to be disproportionately susceptible to AChBR autoantibodies and why they are relatively spared in MuSK MG.162
In addition, AChR structure may differ between muscles as well. Muscles innervated by terminal twigs with en grappe morphologies are more likely to have fetal-type AChRs (described below) whose physiological properties may differ from their en plaque counterparts.1 NMT also differs between different fiber types. Type 2 muscle fibers (fast twitch) have greater sensitivity to ACh than their type 1 counterparts translating to a larger safety factor in NMT. This results from larger nerve terminals, a greater average quantal content, an increased number of postsynaptic folds, and a greater density of sodium channels.39,162 This is teleologically logical, as firing frequencies in fast twitch fibers are much higher than their slow twitch counterparts, translating to greater ACh depletion in the active zone of the presynaptic region, thereby requiring a greater safety factor in type 2 fibers to ensure uniformly successful NMT.162
Lastly, there is a significant interdependence on the three anatomical domains of the NMJ that is evident not only during synaptogenesis but in disease. Optimal postsynaptic architecture and function is very dependent on presynaptic influence.90 As an example, acquired postsynaptic MuSK MG and the inherited synaptic form (end plate acetylcholinesterase (AChE) deficiency of CMG both have adverse effects on more than one domain of the NMJ anatomy.90 AChE deficiency will have presynaptic effects such as reduction in quantal content and in the size of the presynaptic nerve terminal as well as a postsynaptic effect in the generation of an end plate myopathy.163
The sequence of events in normal, successful NMT can be conceptualized as beginning with the synthesis and resynthesis of ACh in the presynaptic terminal and can be schematically followed in (Figs. 25-3 and 25-4). This is accomplished primarily by the enzyme choline acetyltransferase (ChaT) that combines acetate and choline after their reuptake into the presynaptic terminal. Synthesized ACh molecules are packaged into vesicles or quanta that exist in three separate zones; a large storage pool, a mobilization pool, and in clusters close to the presynaptic membrane referred to as the active (immediate release) zone.164 Although there are non–calcium-dependent mechanisms of ACh release, the efficient function of these active zones is dependent on calcium entry into the distal motor nerve terminal. The P/Q type voltage-gated calcium channels (VGCC) integral to this process are distributed along the active zones at vesicle fusion sites on the presynaptic membrane. In response to a NAP, the presynaptic calcium concentration increases to the 100–1000 μM range.162,165 Vesicle release occurs after a delay of approximately 100 μs following the NAP with the presynaptic calcium concentration dissipating after approximately 200 μs. As calcium channels are not fully activated by a normal NAP, the capacity to increase quantal content exists by other mechanisms not involved in normal NMT.
Figure 25-3. Normal neuromuscular junction—presynapse.
Figure 25-4. Normal neuromuscular junction—postsynapse.
In mammalian systems, the contents of 50–300 vesicles are typically released in response to a single NAP, referred to in the aggregate as the quantal content.39 There are approximately 200–400 × 103 individual synaptic vesicles contained in the average nerve terminal. In mammalian NMJs, approximately 20% of these are positioned for immediate release in the active zones.166,167 Within each vesicle, there are between 5–10 × 103 molecules of ACh, the number varying somewhat between individual vesicles.168,169 In at least one form of congenital MG associated with the impaired synthesis of ACh due to a mutation of the ChaT gene, reduction of the number of ACh molecules within a single synaptic vesicle is reduced sufficiently to interfere with NMT.39
The mobilization pool is estimated to contain 300 × 103 vesicles that can be moved readily to the active zone region.168 Following vesicle fusion with the presynaptic membrane and exocytosis, ACh resynthesis and repackaging (endocytosis) take place. Experimental data suggest that the rate of resynthesis parallels the rate of ACh release under normal physiological conditions and is capable of increasing to keep pace with neuromuscular activation.170
The process of ACh synthesis and resynthesis, vesicle packaging, migration, docking, and exocytotic release into the synaptic cleft is dependent on greater than 1000 functional presynaptic proteins (Fig. 25-3). Detailed description of this obviously complex system is incompletely understood and beyond the scope of this chapter. The key proteins underlying vesicular fusion with the presynaptic membrane are referred to as the SNARE protein complex (soluble NSF attachment protein receptor). Key components of the SNARE complex are synaptotagmin and synaptobrevin (bound to the vesicular membrane), and syntaxin-1 and SNAP 25 (bound to the presynaptic plasmalemma). Interaction between synaptobrevin and syntaxin-1 and SNAP 25 prime the binding and fusion process that culminates by the subsequent binding of calcium to synaptotagmin that triggers quantal release into the synaptic space.39 Subsequent to its role in this process, calcium may freely diffuse away from the active sites, be removed from the nerve terminal by a coupled sodium/calcium exchange mechanism, or be sequestered in the smooth endoplasmic reticulum or mitochondria.171 To the best of our knowledge, there are no recognized acquired or heritable disorders related to the proteins discussed in this paragraph.
The quantal content varies in response to each NAP. Under normal circumstances, the quantal content produces an EPP that far exceeds that necessary to produce a postsynaptic muscle fiber action potential ensuring the fail-safe generation of muscle fiber action potentials with repetitive or sustained attempts at voluntary muscle activation. Although largely irrelevant to normal physiology, presynaptic release of ACh can be augmented as mentioned previously. This effect may be either pathological or therapeutic, depending on the context in which it occurs. In disorders of NMT, the EPP may be augmented by pharmacological intervention at the presynaptic terminal that either prolongs depolarization by blocking potassium channels or the duration and effect of calcium. 3,4 diaminopyridine is an example of the former and guanidine is an example of the latter.39,162,165 In addition, autoantibodies directed at components of the presynaptic potassium channels may produce a pathological condition of muscular hyperactivity (Isaacs’ syndrome) as reviewed in Chapter 10.
Another presynaptic contribution to NMJ development in particular, and to its maintenance and function as well, is the protein agrin. Agrin, synthesized in and released from the presynapse, contributes significantly to postsynaptic differentiation and to the stabilization of end plate receptors172 This process of end plate differentiation requires interaction with a number of crucial postsynaptic proteins including LPR4, MuSK, downstream of kinase-7 (Dok7), and receptoraggregating protein at the synapse (rapsyn).90 As will be described in this and the subsequent chapter, impaired NMT may result as a consequence of either heritable defects in many of these proteins (agrin, MuSK, rapsyn, Dok7) or in some cases autoantibodies directed against them (MuSK, LPR4).90
The morphology of the synaptic space may be subdivided into the major or primary gap (cleft) between the nerve terminal and muscle and multiple secondary clefts formed by the postjunctional folds extending into the postsynaptic region.39 This folding increases the surface area of the postjunctional membrane by 10-fold in comparison to the presynaptic membrane.173,174 This allows for an increase in the density of AChRs/channels as described below, thereby improving the efficiency of NMT. The synaptic cleft is narrow with an average distance of 50 nm between the presynaptic membrane and the summits of the postsynaptic folds.39 This narrow gap facilitates rapid NMT. Once released into the synapse, the quanta will briefly bind to and interact with ACh receptors or channels that control the ingress of cations into the muscle fibers on which the receptors are located.
ACh binding to the end plate is short-lived, due in part to diffusion away from the receptor and in large part due to catabolism by the enzyme AChE that is anchored to the basal lamina by its collagen tail, the outer layer of the postsynaptic muscle membrane.39 It is encoded by the triple-stranded collagen Q gene (COLQ) which is relevant to one form of CMS that will be described in Chapter 26. Drugs that reversibly inhibit AChE are used both diagnostically (edrophonium) and therapeutically (pyridostigmine) in MG whereas irreversible anticholinesterases (organophosphates) are utilized for their toxic properties as insecticides or in warfare. There are multiple isoforms of AChE.175 The primary form is AChE-S (synaptic) which is bound to the basal lamina. Other forms, AChE-E (erythropoietic) and AChE-R (read-through) do not play significant roles in ACh catalysis under normal circumstances but compensatory increases in AChR-R in response to chronic treatment with cholinesterase inhibitors may have detrimental clinical effects as described below.175
ACh catalysis by AChE is one of the fastest enzymatic processes known and occurs at a rate of five ACh molecules per millisecond.176 Despite this, ACh enjoys a competitive advantage as the density of AChR (15–30 K × m2) is 5–10 times greater than the molecular density of AChE (2–3 × 103/m2).177 This allows 50–75% of the quantal content to achieve successful interaction with AChR under normal circumstances.178
Temperature changes affect NMT in a variety of ways that may have both electrodiagnostic (EDX) and clinical significance.44,45,48,49,179,180 These effects will be addressed here as the most notable physiological effect of temperature change on NMT is on AChE. Reduced temperature slows the rate of AChE hydrolysis of ACh, prolonging the duration of EPPs by allowing ion channels to remain open for longer periods of time and enhancing end-plate responsiveness to ACh.181,182 The net effect of cooling is to enhance NMT. Electrodiagnostically, this may diminish the probability of demonstrating a decremental response to slow repetitive stimulation. Clinically, cooling may improve function, for example, patient recognition that cold liquids are easier to swallow than warm ones.
Cooling has other non-AChE effects on the physiology of NMT. Both the duration and amplitude of the NAP at the presynaptic terminal are increased by cooling.163–185 This may be a consequence of prolonged calcium channel open time and augmented quantal content.186 Reducing the affected muscle’s temperature is known to increase the AChR’s open time as well.182 Finally, a reduction in muscle temperature leads to a lowering of the resting membrane potential, bringing it closer to threshold, allowing a myofiber action potential to be triggered with a smaller EPP. These four factors, and perhaps others as well, serve to improve NMT in response to cooling.
The organization of the postsynaptic membrane provides for efficiency in NMT (Fig. 25-4). The development and maintenance of end plate complexity is related to the proximity of nerve terminals both embryologically and during adult life and to the influence of ACh and agrin produced and released by these nerve terminals. The key anatomic structure on the end plate is the AChR receptor or channel. Each AChR channel is a glycoprotein composed of five subunits that are arranged in a manner similar to barrel staves turned inside out, resulting in a transmembrane structure with a sagittal appearance similar to the cooling tower of a nuclear power plant. In an adult, the channel consists of two α subunits with singular copies of β, δ, and ε subunits.39,162,187 Embryologically, the AChR has a γ subunit instead of an ε subunit. The transition to an adult configuration occurs at least in part due to the trophic influence of the presynaptic nerve terminal during the innervation process. Fetal AChRs are typically downregulated during adult life but their presence may have two notable clinical influences. As mentioned above, they may persist to some degree in certain adult muscles, particularly those with en grappe nerve presynaptic morphology, and may contribute to the selected vulnerability of ocular and bulbar muscles in acquired MG. Their persistence in some forms of congenital myasthenia may allow for survival in what would otherwise be a lethal condition.
At the NMJ, AChRs preferentially reside in at the apices of the junctional folds of the muscle end plate and span the postsynaptic membrane. These channels are topographically clustered with their density estimated to approach 10,000 molecules per μm2.188 The density of these channels falls to approximately 10 molecules per μm2 within a few microns of the end plate.39,162 The topography of AChR channel distribution both on and within the muscle end plate is essential for optimal NMT. The placement of these channels is established embryologically and maintained during adult life through the contributions of a number of numerous proteins that are essential for the development and maintenance of channel distribution and their optimal function.114 The initiation of this process is through the effects of the presynaptically synthesized and released protein agrin and ACh, both of which are essential to this process. Agrin knockout mice have normal numbers of AChRs but no evidence of clustering. Agrin appears to bind postsynaptically to LRP4 which in turn results in MuSK activation.189 MuSK is required not only for proper synaptogenesis, but for the stabilization and maintenance of end plates in postnatal life. Knockout MuSK mice embryos fail both to develop AChR clusters or to survive.39 Deletion of MuSK in adult muscle leads to the degradation and complete loss of NMJs. MuSK reacts in turn with Dok-7 to activate certain downstream signaling pathways and with rapsyn. ACh clustering is maintained by rapsyn which directly binds with the cytoskeleton and dystrophin-glycoprotein complex, specifically through α- and β- dystroglycan.39,162,190,191 Despite the apparent importance of Dok-7 in this process, defects in this protein do not appear to adversely affect AChR clustering on junctional folds.162 An additional protein, neuregulin (NRG-1) also appears to play a role in the clustering of AChRs at the NMJ.39,162
As mentioned, the AChR is a ligand-gated channel that spans the postsynaptic muscle membrane with its long axis oriented perpendicular to this membrane. It has a hydrophilic central pore with the ACh-binding site located on the extracellular surface of each subunit.192 In the resting state, the central narrow region or waist created by the apex of the convexity of all five subunits meet in opposition, effecting channel closure. Channel opening is dependent on the simultaneous binding of two molecules of ACh with each channel, which then leads to a conformational change, allowing transient opening of the channel pore and, as a result, ion movement. The agonist binding sites for ACh straddle the α/δ or α/ε (α/γ when relevant), identical to the binding site for α bungarotoxin.114,162 They appear to be distinct from, but close to the MIR which resides on the α subunit and is the locus for AChR autoantibody binding.114,162 The α/γ binding site appears to have the highest affinity for ACh which could in turn have relevance in regard to the selective vulnerability of certain muscles which have a greater prevalence of this channel type. Although potassium, calcium, and sodium ions are all capable of traversing the channel, sodium conductance is most dynamic due to favorable size, concentration, and electrical gradient considerations.
In response to the random spontaneous release of singular quanta, unrelated to NAPs, individual channels will open and a miniature end plate potential (MEPP) will be created in that muscle fiber. This is presumed to have a trophic influence on muscle but produces no myofiber contraction. MEPPs occur at a frequency of about 0.2–0.03 times per second, resulting in the activation of 1–2 × 103 AChR channels and the generation of a nonpropagated waveform with a magnitude of 0.5–1 mV.193,194
The nonpropagating EPP generated by the normal quantal content release typically exceeds 50 mV in amplitude, and in normal individuals, produces a propagating SMFAP in each muscle fiber stimulated.195 In health there is a “safety factor”, that being an EPP whose magnitude far exceeds that which is required to depolarize the muscle fiber. A typical mammalian resting membrane potential is approximately –80 mV. The threshold for depolarization may be achieved by a change in voltage of only 10–15 mV, thus providing the three- to fourfold safety margin that normally exists in NMT transmission. The magnitude of the EPP is decreased with repetitive stimuli occurring at a frequency of 5 Hz or less, which, at least initially, deplete ACh-containing vesicles in the active zone. Because of the aforementioned safety margin however, this effect has no significance in the normal individual.
The decline in the EPP in response to “slow” repetitive stimulation will not persist indefinitely as the EPP amplitudes begin to increase after the fourth or fifth stimulus attributed to ACh arrival from the mobilization pool. Conversely, and perhaps counterintuitively, the EPP may be augmented substantially by repetitive stimuli at frequencies of 5 Hz or more. This phenomenon of post-tetanic facilitation is attributed, in large part, to enhanced quantal release related to lingering calcium effects within the presynaptic terminal. Again, this phenomenon bears no consequence in the normal individual, as MFAP occurs in each muscle fiber in response to each and every stimulus. This post-tetanic facilitation does not last indefinitely, and the EPP will begin to decline after approximately 1 minute in normal people due to declining ACh availability. This latter phenomenon is referred to as post-tetanic or postexercise exhaustion and can also be utilized as a diagnostic tool with repetitive stimulation studies in patients with suspected DNMT.196,197 Although post-tetanic exhaustion will not result in NMT failure in normal individuals, it may do so in patients with DNMT whose safety margin for the generation of muscle fiber action potentials is compromised at baseline by disease. In all DNMT, reduction and eventual loss of the EPP safety margin by whatever means is the universal mechanism by which NMT failure and weakness are created.
In addition to the AChR channel, Nav 1.4 sodium channels are also integral to the generation of the muscle fiber action potential.166 Unlike the AChR, they are clustered at the base rather than the peak of the synaptic folds.39,175 Their density is 5–10-fold higher in the end plate than in other regions of the sarcolemma and have a greater density in type 2 than in type 1 muscle fibers.162 Sodium ingress at the NMJ facilitates the EPP generated by ACh channel opening and adds to the safety margin of NMT. Mutation of Nav 1.4 channel gene is a rare form of CMS.
The half-life of an AChR in the junctional membrane is about 8–10 days.198 Under normal circumstances, there is a normal turnover of AChR which are internalized and degraded. The senescent receptors are internalized by the process of endocytosis and transported to lysosomes for degradation through an intricate network of intracellular tubules. This process is accelerated in disease as described in more detail below, being expedited by cross-linking of channels with AChR autoantibodies.162 The AChR are not recycled but are replaced by newly synthesized receptors, one reason why DNMT are more treatment responsive than other neuromuscular disorders in which damaged components are not as readily restored, even if the disease process is arrested.
Integral to the pathogenesis of ACHR MG is the reduction of AChR at the end plate as initially demonstrated by Fambrough and Drachman in 1973 through radiolabeled α-bungarotoxin techniques.3 The pathogenic AChBR autoantibodies of MG are of the IgG1 and IgG3 types and bind predominantly to the MIRs of the AChR which exist on the two alpha subunits of the channel.32,73,114,199–201 Once bound to the AChR, these antibodies initiate a number of irreversible processes, all of which are directed at the AChR and postsynaptic membrane. The most potent of these appears to be complement-mediated, membrane-attack complex lysis of AChR.202,203 Resultant end plate changes not only reduce receptor number but have other anatomic consequence of physiological significance that include simplification of the normal corrugated structure of the end plate (Fig. 25-5). This not only reduces the cross-sectional area but widens the synaptic cleft, thus further reducing the probability of ACh/AChR interaction.173,204,205 Other potential pathophysiological mechanisms include steric hindrance, with autoantibodies physically blocking ACh binding sites on adjacent channels or preventing the conformational change resulting in opening of the ion pore.206
Figure 25-5. Neuromuscular junction in AChBR MG.
The autoantibodies not only bind to the AChR but also cross-link with other antibodies.207,208 When the AChRs are cross-linked in this manner, these are reabsorbed by the postsynaptic membrane by a process known as endocytosis. This process takes place under normal circumstances but accelerates up to three times faster in the presence of AChBR autoantibodies. As a result, the normal NMJ AChR half-life of 5–10 days is dramatically reduced and the AChR population diminished.209–211 As synthesis of new AChRs remains unchanged, there is a significant net reduction of 70–90% AChRs per NMJ.3 Although AChR MG is conceptually a disorder mediated by autoantibodies, there is a significant T-cell-mediated, CD4 lymphocyte component as well.55,212 T lymphocytes specific to AChRs are found in patients with myasthenia.213,214 T cell-targeted therapies hold therapeutic promise for MG.212,215
MG also reduces the number of sodium channels that are clustered at the depths of the end plate folds.166 This appears to increase the threshold for muscle fiber action potentials.162 In any event, reduction in the number or function of sodium channels at the muscle end plate will further erode the safety margin of NMT.
The pathogenesis of MuSK myasthenia is not as well understood as AChR MG but is clearly different. The association between MuSK and MG is based on a number of lines of evidence. There appears to be a strong association between the presence of MuSK autoantibodies and patients with seronegative myasthenic phenotypes. MuSK autoantibodies produce weakness and reduction in MEPP amplitudes in mice and a weakness associated with impaired AChR clustering in rabbits when passively transferred. Children born of MuSK MG mothers may develop transient neonatal MG and respond to plasma exchange (PLEX) treatment.74,83,216,217 Lastly, MEPP amplitude reduction has been demonstrated in vitro in an intercostal muscle biopsy specimen acquired from a MuSK MG patient.218
Current knowledge suggests that MuSK MG is a disease of disordered AChR distribution as opposed to one of AChR destruction. There appears to be a good clinical pathological correlation between those muscles where clustering is impacted the most and selectively vulnerable muscles such as sternocleidomastoid, diaphragm, and masseter.79 Histological studies of MuSK MG patients suggest that unlike AChBR MG, substantial loss of AChR content and IgG/complement does not occur.79 Unlike AChBR autoantibodies, MuSK autoantibodies are of the IgG4 subtype and do not activate complement.72,219 Their primary action in hampering NMT in MuSK MG appears to be fragmentation of the normal AChR clustering at the junctional peaks of the postsynaptic membrane. In addition, MuSK autoantibodies may have synaptic and presynaptic effects including impaired binding of the collagen tail of the AChE molecule and presynaptic reduction in quantal content instead of the usual compensatory increase that occurs in other forms of MG.178,219 In fact, recent studies suggest that the primary binding site for MuSK autoantibodies is the site of interaction between MuSK and the collagen tail of AChE (ColQ) responsible for the anchoring of AChE on the basal lamina.220 This may provide an explanation for why MuSK MG patients, as will be subsequently described, uncommonly respond favorably to cholinesterase inhibitors.79 Accordingly, MuSK MG may be eventually classified as a synaptic rather than postsynaptic disorder. Like most other postsynaptic proteins, mutation of the MuSK gene may result in rare reported cases of CMG.221
The pathogenesis of LPR4 MG is incompletely understood. In support of a pathogenetic role of LPR4 autoantibodies, serum from LPR4 MG patients reduces MEPP amplitudes in mice.8 The preponderance of evidence suggests that LPR4 MG is an IgG1-complement–mediated disease similar to AChBR MG.8,9,25,222 Accordingly, patients with LPR4 MG respond to immunomodulating agents in a manner similar to AChR MG patients although these two autoantibodies rarely, if ever, coexist.25 In contrast, as the normal function of LPR4 involves interaction with MuSK in AChR clustering during synaptogenesis, one would predict a disease mechanism similar to MuSK MG in which complement-mediated end plate destruction does not appear to have a role. Unlike AChR MG, a small percentage of LPR4 MG patients will also harbor MuSK autoantibodies.9,25
The pathophysiology of transient neonatal myasthenia results from the passive transplacental transfer of the mother’s IgG AChR antibodies which bind to the interface of alpha and gamma subunits.114 The reason why this disorder does not happen with greater frequency is unknown.
No discussion of MG pathophysiology would be complete without attempting to provide a cogent explanation for the selective vulnerability of certain muscle groups and the notable asymmetries that may occur in a disease occurring as a consequence of equitable distribution of circulating autoantibodies. Two potential explanations have already been discussed: (1) differing pathophysiological mechanisms with differing autoantibodies and (2) differences in the AChR channel types in different muscles. It has also been hypothesized that the preferential involvement of the external ocular muscles may be related to the elevated temperature of the head compared to limbs. This of course, would provide an inadequate explanation for asymmetry. In the end, adequate pathophysiological explanations for the myasthenic phenotype remain elusive.223,224
Lastly, the major gap that remains in our understanding of acquired, autoimmune MG pathogenesis is the knowledge of what initiates the disease process.32 As mentioned, Mendelian genetics appears to have a minimal causative role in acquired, autoimmune MG. We agree with those who believe that the future will identify a significant genetic role in disease susceptibility to adverse environmental influence.23,26,31,32 The role of infectious agents such as the Epstein–Barr virus as one of these potential environmental precipitants remains uncertain.225 The thymus gland continues to occupy center stage in any MG etiology discussion.23,226–228 One potential explanation involves the recognition that the thymus contains myoid cells and other types of stem cells that may serve as autoantigens by the expression of AChRs or AChR antigens on their surface.23 In this same vein, AChR-specific B lymphocytes are found within the thymus gland of MG patients that are capable of generating antibodies to AChR in culture.
The reader is referred to Chapter 2 where the testing modalities used in support of an MG diagnosis are reviewed. In this section, we will briefly review the serological, EDX, pharmacological, and imaging methods available to aid in the diagnosis and management of myasthenic patients. We will focus on their strengths and weaknesses, and the strategies that we employ in their use. The relative sensitivities of different tests used to support the clinical diagnosis of MG have been estimated and are summarized in Table 25-2.6,9,10,25,37,55,72–74,77,91,160,229–232
TABLE 25-2. DIAGNOSTIC YIELD OF TESTS USED IN THE DIAGNOSIS OF AUTOIMMUNE MG6,9,10,25,37,55,71–74,76,91,230,232,275,296,417
Diagnostic strategies in MG undoubtedly vary between clinicians and institutions due to individual bias and preference, test availability, and relevant differential diagnostic considerations in individual cases. Even in the most clinically straightforward cases, we believe that diagnostic confirmation should be obtained whenever possible, particularly if immunomodulating treatment or thymectomy is contemplated. The most sensitive test for MG is SFEMG (92–100%) followed by RNS of distal and proximal nerves (0–99%) depending on the muscle tested and whether the disease is limited or generalized in its manifestations.229 Neither modality is, however, specific for MG. AChR antibody testing is slightly less sensitive (36–94%) depending also on whether the patient has ocular or generalized disease as well as the nature of the assay utilized, but is highly specific.
Identification of AChR autoantibodies is the most expeditious means to confirm MG. Although there are different types of AChR autoantibodies as will be subsequently described, the AChR binding antibody is the principal pathogenic antibody tested for. It will be considered synonymous with AChR autoantibodies throughout this chapter unless otherwise specified. Typically, this is the only diagnostic test that we initially order unless there are phenotypic features that would suggest MuSK MG. As mentioned, the sensitivity is estimated at approximately 36–79% in ocular MG, 75–94% in generalized MG, and 66–93% in all MG patients.5,37,114,231,233 They are however, highly specific for the MG, being rarely reported as false positives in patients with other autoimmune diseases such as systemic lupus, rheumatoid arthritis, hepatitis, thymoma without MG, inflammatory neuropathy, motor neuron disease, 13% of patients with LEMS, 3% of patients with lung cancer without an apparent neurological disorder and in some asymptomatic relatives of MG patients.160 We consider the diagnosis to be confirmed if these autoantibodies are present in the appropriate clinical context.
The value of these autoantibodies is for all intents and purposes in establishing the diagnosis initially. Although titers may decline with treatment, in particular following thymectomy, it is generally held that this test cannot reliably determine disease severity, response to treatment, or to predict either remission or relapse.55,234 Unlike many other tests used in everyday practice, mild elevations in the AChR autoantibody titer are often significant. Conversely, patients without AChR autoantibodies may have a different disease, harbor a different MG autoantibody, have seronegative MG, or on occasion have a false negative result. This latter situation may arise with testing that has been done too early, or in an individual in whom autoantibody formation has been suppressed by immunomodulating treatment or thymectomy.233,234 For these reasons, testing prior to the initiation of immunomodulating treatment or thymectomy is ideal. As initially seronegative patients may develop autoantibodies over time, repeat testing in a recommended interval of 6 months in the appropriate clinical context may be considered.231
AChR-modulating and AChR-blocking autoantibodies are also commercially available but play a less significant clinical role.160,235 AChR modulating autoantibodies measure degradation of the AChR in cultured human myotubes.232 Both of these autoantibodies are most likely to coexist in individuals who are AChR binding autoantibody seropositive and are unlikely to occur in isolation.126,160,235 In one report, AChR modulating autoantibodies were found in 75% of patients with AChR binding autoantibodies but in only 5% of seronegative patients.232 It is our practice to order these autoantibodies only in this latter population. High titers of AChR modulating autoantibodies also play a potential role in the detection of thymoma. Seventy three percent of patients afflicted with both thymoma and MG harbor AChR modulating autoantibodies producing a >90% receptor loss.125,126,142 Although this potentially justifies their use as a screening tool for thymoma detection, we preferentially rely on imaging for this purpose.
AChR-blocking autoantibodies bind to the same site as ACh or α-bungarotoxin, close to but distinct from the MIR on the extracellular domain of the ACh channel.232 They are found in approximately half of patients with generalized MG, but in only 30% of patients with ocular disease.160,235 In one study, these autoantibodies were found in 30% of MG patients seropositive for AChR-binding autoantibodies but in no seronegative MG patient. As a result of this insensitivity, we find AChR-blocking autoantibody testing to have limited clinical value.
It is estimated that approximately 40–70% of AChR seronegative patients will have MuSK autoantibodies.6,73,236 Our practice is to screen for MuSK autoantibodies as a first step along with AChR-binding autoantibodies if the phenotype is suggestive of MuSK MG. We also order MuSK autoantibodies in any MG suspect who is AChR seronegative. MuSK and AChR antibodies rarely coexist.6,73,89,237 Although somewhat controversial, there appears, unlike AChR seropositive patients, to be a correlation between anti-MuSK titers, disease severity, and treatment responsiveness, thymectomy being the notable exception.73,79,89
Striated muscle antibodies refer to a class of antibodies directed against components of skeletal muscle including titin, the ryanodine receptor, myosin, and α-actinin.238 These are found in approximately 30% of adult patients with MG without thymoma, 24% of patients with thymoma without MG, and 70–80% of patients who have both.126,160,239 We have had numerous experiences where the occurrence of these autoantibodies even in high titer appears to have no detectable clinical correlation. Again, we largely rely on imaging to detect thymoma, and in view of the questionable sensitivity of the test, we find their utility limited. It is reasonable however, to measure these antibodies before and after thymectomy as a failure to reduce the titer suggests incomplete resection and as an increasing striational autoantibody titer postthymectomy has been reported to herald thymoma recurrence.
We routinely obtain a thyroid stimulating hormone level as the incidence of hypo- or hyperthyroidism is fairly high in MG patients and as a failure to recognize dysthyroidism may affect treatment efficacy. It is not uncommon for other autoantibodies to coexist with MG and/or thymoma such as ganglionic (as opposed to the nicotinic found on muscle) AChR antibodies, voltage-gated potassium channel antibodies, and CRMP-5-IgG autoantibodies.125,126,130 We do not routinely test for autoantibodies more closely related to other autoimmune diseases in MG patients unless there is clinical suspicion to do so. Antibodies directed against the Kv1.4 subunit of voltage-gated potassium channels have been described to occur in a percentage of patients with MG but not in patients with thymoma, inflammatory myopathy, or in healthy controls.129 These were found exclusively in patients who were also AChR-binding antibody positive and are not commercially available. Their role in the diagnosis of MG, if any, has yet to be defined.
The role of EDX testing in MG has undoubtedly diminished in an era of readily available, affordable, and accurate serological testing. EDX provides the greatest utility in the seronegative patient where its purpose is to identify a pattern of abnormalities that support a diagnosis of a postsynaptic DNMT, in turn consistent with MG.240 At the same time, it attempts to identify or exclude a pattern of abnormalities suggestive of an alternative cause of painless weakness. Electrodiagnosis is used by some in MG to monitor the response to treatment but we do not find this necessary in the vast majority of patients. One uncommon use of EDX in the MG patient is to identify afterdischarges on routine motor conduction studies that would support overdosing with cholinesterase inhibitors.241 The actual mechanics involved in the performance of repetitive stimulation of motor nerves and SFEMG are provided in Chapter 2.218,242 In this section, we will focus on the strategies and pitfalls involved in MG EDX and its interpretation, including a summary of characteristic pattern of abnormalities.243
The EDX evaluation of a patient with suspected MG begins with routine nerve conductions. Typically we obtain, one motor and one sensory conduction from an upper and lower limb, four conductions in total. We would expect these to be normal in a postsynaptic DNMT in the absence of an alternative or additional confounding diagnosis. If the compound muscle action potential (CMAP) amplitudes are reduced in a patient with a phenotype resembling MG, a motor neuron disease or a presynaptic DNMT such as LEMS or botulism should be considered. If this occurs, we would attempt facilitation with 10 seconds of exercise followed by a second, supramaximal stimulus immediately thereafter, attempting to identify a presynaptic DNMT. We then proceed to repetitive stimulation testing at 2–3 Hz. In general, we typically study the ulnar, accessory, and facial nerves although readily adapt this menu if warranted by clinical circumstance. If possible, we start with a weak muscle, particularly if it is in the hand where the study is technically easier to perform. If there is no weakness, or the weakness is proximal, we would go straight to the accessory nerve as the best compromise between diagnostic yield and technical ease. If a patient has only ocular or bulbar weakness, we usually start with a facial muscle such as the orbicularis oculi or the nasalis.
If repetitive stimulation reveals a decrement at baseline, we reproduce it to ensure its validity and then exercise the muscle studied. We then attempt to repair the decrement by exercising the muscle for 10 seconds and then repeating the RNS. However, if no convincing decrement is seen, we then exercise the muscle for a full minute and then repeat repetitive stimulation trains of 10 at 2–3 Hz every 30 seconds for up to 5 minutes, looking for postexercise exhaustion.
If RNS is normal, we proceed to EMG to look for motor unit instability as evidence of DNMT, or features of other possible neuromuscular disorders that may mimic MG (e.g., myopathy, motor neuron disease). If both repetitive stimulation and concentric needle EMG are nondiagnostic, we proceed to SFEMG. This testing order is based on the knowledge that SFEMG is a very sensitive but not specific test. The choice of muscle(s) for SFEMG depends on the patient phenotype. The majority of patients in whom SFEMG is performed in our laboratories have either isolated ocular complaints or on occasion generalized fatigue for which the referring physician wishes to exclude MG. In the former circumstance, we typically study the frontalis or orbicularis oculi muscle whereas in the latter, the extensor digitorum communis is our typical site. Each case should be individually assessed however, with the choice of nerves and muscles tested based on clinical evaluation. In a myasthenic, diagnostic yield will always be greatest in a clinically weak muscle. Again, failure to demonstrate a decremental response, increased jitter, and neuromuscular blockade in a clinically weak muscle implicates a diagnosis other than MG.
There are a number of important caveats to consider in the EDX testing of an MG suspect. Temperature change has a number of significant and at times conflicting physiological effects on NMT.45,244 In the aggregate, however, the maintenance of surface temperature to an optimal temperature of 35°C or above is required to minimize the risk of either a false negative repetitive stimulation or single-fiber study. A false negative study may also occur as a result of anticholinesterase medication exposure. Ideally, potential MG patients should be tested prior to exposure to any medication that might significantly affect the disease, particularly through interference with NMT (Table 25-3). In particular, pyridostigmine should be discontinued if possible at least 12–24 hours before the study.
TABLE 25-3. DRUGS THAT MAY ADVERSELY AFFECT NEUROMUSCULAR TRANSMISSION303,418
1. Drugs that may unmask or exacerbate myasthenia gravis
a. Antiarrhythmic agents—lidocaine, quinidine, quinine, procainamide, and trimethaphan camsylate
b. Antimicrobial agents including aminoglycosides, polymyxin B, colistin, clindamycin, ciprofloxacin, netilmicin, azithromycin, pefloxacin, norfloxacin, and erythromycin
c. Corticosteroids
d. Magnesium (parenteral)
e. Neuromuscular blocking agents—depolarizing and nondepolarizing including botulinum toxin, d-tubocurarine, succinylcholine, vecuronium, pancuronium, atracurium, and gallamine
2. Drugs potentially implicated in unmasking or exacerbating myasthenia gravis
a. Anesthetics—diazepam, ketamine
b. Anticonvulsants—phenytoin, mephenytoin, ethosuximide, barbiturates, carbamazepine, and gabapentin
c. Antimicrobial agents—tetracyclines and ampicillin
d. Antirheumatics—chloroquine
e. Beta blockers—propranolol, oxprenolol, timolol, practolol, and betaxolol
f. Drugs of abuse—cocaine
g. Gastrointestinal—cimetidine
h. Miscellaneous—D–L-carnitine, tropicamide, iodinated radiographic contrast, and trihexyphenidyl
i. Ophthalmics—echothiophate
j. Psychotropic drugs—phenothiazines and lithium
3. Drugs that cause myasthenia gravis or mimic myasthenia gravis
a. D-Penicillamine
b. Alpha-interferon
c. Case reports: trimethadione, riluzole, ritonavir, chloroquine, statins, and beta-interferon
d. Tandutinib
False positives are an even larger potential pitfall in the EDX testing of an MG suspect. The most common cause of a false positive examination is undoubtedly technical, particularly with repetitive stimulation testing. Unwanted movement by either the patient or technician can result in movement of the stimulator resulting in submaximal stimulation and the appearance of a “pseudodecrement.” Unwanted movement resulting in baseline deviation is a particular problem both in large proximal muscles and in the face. The latter often occurs from grimacing in response to facial nerve stimulation and the undesired coactivation of other facial innervated muscles. Any technique that will reduce unwanted muscle contraction and movement and unwanted artifact is desirable.
To minimize the risk of a false positive interpretation, we require that all characteristics of a typical postsynaptic decremental response be fulfilled. These include: (1) a normal baseline CMAP amplitude, (2) a nadir that occurs at the fourth or fifth response of a 10-stimulus train, (3) the greatest decrease in CMAP amplitude between consecutive responses occurring between the first and second responses and (4) a steady rise in the CMAP amplitude after the fifth response that allows the CMAP amplitude to approach but not achieve that of the first response, producing a “tilted saucer,” “ski jump,” or “inverted banana” configuration (Fig. 25-6).
Figure 25-6. Typical pattern of a pathological decremental response to “slow” (2–5 Hz) repetitive stimulation.
The other false positive scenario is assigning EDX abnormalities to MG, when the abnormal NMT is due to a different disease. As discussed below, there is potential overlap between the needle EMG findings in MG and other neuromuscular diseases. In addition, an abnormal repetitive stimulation or SFEMG study is not specific for MG and can be seen in any neuromuscular disease that may cause a secondary effect on NMT. Of these disorders, ALS may be the most common typically resulting in a decremental response of <10%.245–249 The EDX features characteristic of a postsynaptic DNMT that occur in other neuromuscular disease may have multiple mechanisms. In ALS and other denervating disorders, they are hypothesized to result from immature NMJ occurring as a consequence of attempted reinnervation.
Neither increased insertional activity nor abnormal spontaneous activity is typically identified in an MG patient. Fibrillation potentials and positive waves may occur however, particularly in paraspinal, bulbar, and proximal muscles.250 Presumably, they result from severe end plate destruction and effective denervation. In support of this, their prevalence appears to correlate with patients who are significantly affected by generalized disease.
Two different types of abnormal motor unit action potential (MUAP) change may occur in MG. Both may be easily overlooked. Short-duration, low-amplitude MUAPs associated with early recruitment may occur. This pattern results from neuromuscular blockade and an effective reduction in the number of functional myofibers within a given motor unit, analogous to what may occur with myopathies.251,252 Small MUAPs have been reported in MuSK MG as well, also consistent with a reduced number of normally functioning muscle fibers per motor unit resulting from either NM blockade, muscle fiber loss, or muscle fiber atrophy.
A more sensitive and valuable needle EMG tool is the demonstration of MUAP variability (instability) (Fig. 25-7). MUAP variability is the analog of clinical weakness and fatigue, a decremental response with repetitive stimulation, and blocking on SFEMG. It represents NMT failure of one or more single-fiber components to the MUAP that is typically intermittent, resulting in constant variation in the size, shape, and sound produced by an isolated MUAP. Unstable MUAPs are readily identified by the trained ear, and visualized with the use of a trigger and delay line. Their value is to provide evidence of abnormal NMT in muscles not easily studied by repetitive stimulation without necessarily undergoing the rigor of the SFEMG examination.
Figure 25-7. Motor unit instability—that is, variable MUAP size and shape with consecutive firings.
The EDX yield in MG varies depending on the serotype, severity, the EDX technique chosen, and the nerve/muscle studied. To reinforce a previous point, absent technical difficulties, the diagnostic yield in a muscle rendered weak by myasthenia should be 100%. Conversely, the value of EDX relies on its ability to demonstrate disordered NMT in muscles that do not appear to be clinically affected.
To be considered abnormal, most laboratories require demonstration of a decrement of ≥10% in two muscles. These criteria are met in response to “slow” repetitive stimulation (2–5 Hz) in 37–62% of all individuals with MG.252–256 The yield is lowest with intrinsic hand muscles which lack a decremental response in 50–70% of patients with moderately severe MG.257 Proximal muscles, for example, the biceps, deltoid, and the trapezius, have higher yields. Erb’s point stimulation with deltoid recording may be 80–90% sensitive in patients with myasthenia, but brachial plexus stimulation is both uncomfortable and technically difficult in view of the movement it creates. Accessory nerve stimulation has similar sensitivity and is usually better tolerated with an easier-to-establish stable baseline.258 Facial nerve activation with recordings from the orbicularis oculi or nasalis muscles has a higher yield than limb muscles.
In patients with ocular MG, repetitive stimulation of a distal upper extremity nerve yields positive results in up to 35% of patients. Adding a proximal nerve increases the yield slightly to 45%. Patients who are MuSK antibody positive tend to have both a lesser incidence estimated at 60% and a lesser degree of decrement in limb muscles compared to other MG populations.73,78,81 A higher yield of abnormal decrement will be found in facial muscles in the anti-MuSK patient population.259 Estimates of incidence of abnormal decrement in these patients range from 25% to 50% in limb muscles as opposed to 50–85% in facial muscles.77,82
The EDX assessment of children with suspected MG is analogous to adults. In normal infants, the limited data available suggest that with stimulation rates between 1 and 2 Hz, there is no alteration in the CMAP.260–262 Repetitive stimulation at rates between 2 and 5 Hz yields variable results, with some normal infants demonstrating a decrement and others revealing no change. In summary, premature infants and some term infants will have a reduced NMJ reserve capacity, especially at the higher rates of stimulation. Repetitive stimulation can be performed and interpreted confidently in term infants as long as this caveat is kept in mind.
The electrical findings in transient neonatal MG are analogous to those found in adults.101,263 A decremental CMAP response is typically demonstrable at low rates of stimulation, which can be minimized with postactivation excitation (20–50 Hz repetitive stimulation) and augmented with postactivation exhaustion (repetitive trains of 2–5 Hz stimulation performed at 30 seconds to 1 minute intervals over a protracted period). Frequently, a decrement occurring at high rates of stimulation occurs as well, depending on the severity of the disease.
SFEMG is the most sensitive and least specific of EDX testing procedures for MG.264 It can be performed either voluntary activation of a tested muscle or by electrical stimulation of a motor nerve branch innervating that muscle. Either method has its benefits and drawbacks. One strength of stimulated SFEMG is that it is less reliant on patient cooperation than voluntary SFEMG, making it the technique of choice in infants, an elderly person who may not be able to steadily sustain a minimal level of muscle activation, or any potentially uncooperative individual.265–269 Stimulated SFEMG is typically faster to perform than voluntary SFEMG, providing an additional advantage. Its major disadvantage occurs when a muscle rather than nerve fiber is stimulated leading to a falsely reduced jitter value. Both voluntary and stimulated SFEMG are specialized techniques that many electromyographers do not receive training in. One benefit of SFEMG, stimulated or voluntary, is that it allows access to muscles frequently affected in MG not accessible to repetitive stimulation. The major benefit of SFEMG, however, and the one that translates to its high diagnostic sensitivity is its ability to detect abnormalities in NMT prior to development of overt NMT failure.
As described in Chapter 2, SFEMG assesses NMT by comparing the discharge intervals of two or more SMFAPs belonging to the same MUAP. This is technically accomplished by limiting the recording radius of the EMG needle. Historically, this was accomplished both by increasing the low frequency filter settings of the EMG machine and by using a special needle with a very limited recording radius. These needles are, however, expensive and reusable, necessitating sterilization after each use as well as periodic sharpening. For practical reasons, many institutions now use disposable, concentric needles which are adequate surrogates providing acceptable accuracy if attention is paid to detail during performance and interpretation.270–273
Two parameters are typically measured in the SFEMG assessment of a suspected MG patient. Jitter refers to the variation in the interval between the two single-muscle fiber action potentials mentioned above. Because of the normal variation in EPP amplitude, jitter is a property of normal muscle, typically lying in the 15–45 μs range depending on age and the muscle studied.274,275 It becomes abnormal only when it is either smaller or in the case of MG, larger than normal (Fig. 25-8A,B). Increased jitter reflects abnormal or delayed, not failed NMT. As a result, abnormal jitter does not translate to weakness. With an increase in jitter values to 80 μs or above, however, NMT becomes tenuous and begins to intermittently fail. As a result, blocking, the equivalent of a decremental response to repetitive stimulation, motor unit instability, and clinical weakness, becomes manifest (Fig. 25-8B).
Figure 25-8. Single-fiber electromyography demonstrating a normal recording (A) and increased jitter and blocking (B —seventh pair from top—arrow) from an MG patient.
SFEMG is abnormal in 77–100% of patients, depending on disease severity and the number and distribution of muscles tested.252,276–280 Specifically, the sensitivity of SFEMG is estimated at 97% if both a limb and a facial muscle are studied.263 SFEMG abnormalities are found in the frontalis or orbicularis oculi in 87–99% of patients with oculobulbar weakness.264 Studying the frontalis as opposed to a limb muscle will increase diagnostic yield from the 22–66% to the 54–100% range in all MG patients.276,278 SFEMG abnormalities, like every other aspect of MG, can be patchy. We have observed instances while recording three SMFAPs belonging to the same motor unit where jitter is markedly increased in one pair while normal in the other. As a quantitative reflection of this, one study of 433 potential pairs from 32 patients with MG revealed normal jitter in 9%, increased jitter in 38%, and abnormal jitter with blocking in 53% of the potentials.252,277 Although jitter measurements improve following successful treatment,276,281–283 jitter abnormalities may persist even in those in clinical remission.276,284 As in “slow” repetitive stimulation, SFEMG abnormalities in patients with MuSK MG are more likely to be found in facial or proximal muscles rather than in distal limb muscles such as the extensor digitorum communis, with a lower yield in limb muscles in general than expected in other MG populations.75,86
Historically, regional applications of both ischemia and d-tubocurarine curare were used in addition to “slow” repetitive stimulation, in an effort to increase diagnostic yield in MG. As these are cumbersome and potentially risky maneuvers, SFEMG represents the preferred EDX alternative in most EMG laboratories where “slow” repetitive stimulation is nondiagnostic.
The edrophonium (i.e., Tensilon) test is a useful diagnostic adjunct in MG.285–287 Edrophonium is a short-acting anticholinesterase, the administration of which will result in a transient increase in ACh availability at the NMJ and amplify the EPP. To perform the edrophonium test, a butterfly needle is placed in an accessible vein. A 2 mg (0.2 mL) test dose of edrophonium is administered initially. A small initial dose is necessary as some patients are extremely sensitive to the medication and may respond favorably to a small dose but not respond to a full 10 mg dose, leading to a potential false-negative conclusion in these individuals. If there is no response to the initial 2 mg after 60 seconds, the remaining 8 mg are administered in 2 mg increments every 10–15 seconds. If the patient has an objective improvement or a severe side effect, the rest of the injection is aborted.
Performance of edrophonium testing should only be done in patients in whom objective weakness is present. In this regard, evaluating improvement of ptosis, ophthalmoparesis, or dysarthria is more useful than a complaint of dysphagia or fatigue. We have attempted tensilon testing in conjunction with modified barium swallow or pulmonary function testing as a means of identifying an objective response but have not had great success.
The sensitivity of edrophonium testing in MG is reported to be 86% for ocular and 95% for generalized MG.288 Like repetitive stimulation, an abnormal test signifies abnormal NMT but does not define a singular disease. False positive responses have been reported in LEMS, ALS, CMS, botulism, Guillain–Barré syndrome, dysthyroid ophthalmopathy, and brainstem tumors.37,289–292 Edrophonium testing is reported to have less utility in MuSK MG with a reported sensitivity of 50–70%.78,82
Edrophonium testing is not without risk. Historically, these were office-based procedures, performed without incident in most cases. Some patients would experience transient, disquieting but ultimately harmless symptoms such as nausea, vomiting, increased tearing, lacrimation, fasciculations, borborygmi, and eructation. Rarely however, serious reactions including bradycardia or heart block may occur. For consideration of patient safety, these tests are now routinely performed in monitored settings with individuals who are trained in resuscitation with adequate resources including parenteral atropine available.
Edrophonium has also been used in an attempt to distinguish weakness resulting from MG as opposed to that created by cholinergic excess. The availability of other effective MG treatments has limited the need for high dose anticholinesterase treatments in recent years. In addition, myasthenic crises and cholinergic crisis can be clinically distinguished in most individuals based on both the amount of anticholinesterase received and the presence of muscarinic symptoms that frequently occur in association with cholinergic crisis. For these reasons, this application of edrophonium is largely of historical interest.
Pathological changes in the thymus are found in >80% of patients with MG, detectable by imaging in many cases. In approximately 50% of those with MG, the histology will be that of lymphoreticular hyperplasia.293 In the minority estimated at between 10–30%, either a benign low-grade thymoma of the thymic epithelium may be encountered, or on occasion an invasive thymoma.23 Although chest x-ray may detect thymic enlargement from either hyperplasia or thymoma, either computerized tomography or magnetic resonance provide better resolution and are preferable studies. Although the thymus is typically located in the anterior mediastinum, it may be found ectopically in the neck or other regions of the chest.293,294 In normal individuals, the CT appearance of normal thymus is homogeneous with attenuation characteristics similar to muscle, decreasing in size with age as fatty replacement takes place. The gland loses its appearance as a discreet structure somewhere between ages 25 and 40.293 With MR, the normal childhood thymic appearance is also homogeneous with signal characteristics somewhere between muscle and fat. In addition to these features, a normal thymus gland is also characterized by its size and shape. A focal abnormality of the contour of the gland suggests an underlying neoplasm.
With thymic hyperplasia, the CT attenuation properties are normal. In slightly more than half of cases, the gland will appear abnormal because of its enlargement or in some cases, a focal mass.293 MR characteristics are similar with gland enlargement but with normal signal characteristics. With thymoma, CT typically identifies sharply demarcated round or lobulated masses, commonly associated with low attenuation components that represent cysts, hemorrhage or necrosis (Fig. 25-2A,B). Calcifications occur and do not distinguish between invasive and noninvasive tumors.293 Thymomas may enhance with iodinated contrast but the benefit is likely outweighed by the risk of an allergic response or a myasthenic exacerbation.295
The MR appearance of thymoma is also that of a gland incorporating a round, oval, or lobulated mass.293 Signal intensity is low on T1-weighted images similar to that of muscle and relatively high signal intensity with T2-weighting. T2-weighted images may also define a lobulated structure characteristic of thymoma. One potential advantage of MR over CT in the evaluation of thymoma is the identification of characteristics suggestive of invasive thymoma such as obliteration of the adjacent fat planes.293 Thymic abnormalities in patients with anti-MuSK antibodies are uncommon and typically minimal when they occur.77 This point is emphasized in a study of 167 patients with thymoma.126 Of the 92 who had MG, only one was seropositive for MuSK antibodies.
Recently, the bright tongue sign has been reported in ALS patients, representing fatty replacement of the genioglossus muscle demonstrable with MR imaging. This finding is not unique to ALS and has been reported in MuSK MG patients, in keeping with the muscle atrophy that can be seen in some of these patients, and providing another potentially confounding feature in the distinction of MG from bulbar ALS.89
Presumably, the majority of muscle biopsies performed in MG patients is done inadvertently in seronegative individuals with phenotypes that mimic myopathy. Muscle biopsy has no role in the majority of MG patients and much of what we know about myasthenic muscle histology comes from postmortem specimens.296 These reports need to be interpreted with caution. Although MG is a systemic disease, described abnormalities often existed in limb muscles whereas the majority of these patients had oculobulbar phenotypes. Consequently, the abnormalities described are not necessarily disease-related.296 Conversely, if muscle biopsy were to be performed, consideration of a false negative result should be given in view of sampling error and the patchy nature of the disease. Interpretation of historical publications of muscle histology in MG should be made with the knowledge that many preceded the availability of contemporary histochemical analysis. Lastly, interpretation of muscle histology in MG should be made with consideration of the effects from potential coexistence of other disorders that occur with increased frequency in MG such as dysthyroidism or autoimmune muscle disease.
The findings on light microscopic analysis of MG muscle are nonspecific. The muscle may be normal in appearance.296 When abnormal, the most common findings are type 1 fiber predominance, mild fiber type grouping, or type 2 fiber atrophy.296,297 Focal interstitial inflammatory infiltrates, historically referred to as lymphorrhages, are not uncommon, and have been described predominantly within proximity of necrotic fibers, blood vessels, and muscle end plates (Fig. 25-9).296–299 Histological features of myopathy are not a characteristic feature of myasthenic muscle.296
Figure 25-9. Cross section of muscle biopsy in a myasthenic patient demonstrating a focal inflammatory response “lymphorrhage.”
Muscle biopsies in MuSK MG patients have been reported to demonstrate myofiber atrophy and minor simplification of some end plates.73 Despite the known function of MuSK in synaptogenesis, the density and distribution of AChR in patients with MuSK appear normal in the studies reported to date. Complement deposition or other markers of an immune-mediated disease have not been identified.212,300
Ultrastructural abnormalities are more evident in MG. Immunoelectron microscopy of the postsynaptic membrane region in patients with myasthenia demonstrates IgG and complement precipitation on the membrane, a widened synaptic space, reduced postsynaptic membrane complexity with fewer postjunctional folds, and decreased numbers of AChRs. Many of the remaining AChRs are bound with IgG.173,203–205,301 In contrast, the presynaptic portion of the NMJ appears completely normal.
There has been some historical concern pertaining to potential adverse structural effects on muscle in response to chronic or excessive anticholinesterase exposure.176 In animals, degeneration of postsynaptic folds has been described. SFEMG studies have suggested increased fiber density in patients treated with anticholinesterase medications, suggested of denervation and reinnervation and consistent with some of the histological findings described above. In addition, prolonged exposure to anticholinesterases leads to an overexpression of the AChE-R isoform. Overexpressed AChE-R in transgenic mice leads to myopathic muscle changes including atrophic and vacuolated muscle fibers as well as neurogenic muscle changes.302 The clinical relevance of these observations is uncertain, given the relatively minor histological findings reported in patients largely studied in an era when anticholinesterase medications were the primary therapy for MG, commonly used in large doses for prolonged periods of time.
Although relevant to myasthenia, the histological abnormalities of the thymus gland are beyond the scope of this chapter. The reader is referred to an excellent review of the thymus gland in myasthenia for more detailed information on this subject.23
Managing a myasthenic patient requires consideration of numerous variables. Patient age, gender, comorbidities, concurrent medication, patient functional expectations, serotype, clinician experience and preference, treatment availability and cost, and whenever available, an evidence-based perspective all require consideration in treatment decisions. Many excellent reviews are available that address these options and strategies.38,55,303 The ideal of a fully evidence-based therapeutic approach is hampered by the lack of adequate studies. This impediment is multifactorial. MG is a relatively uncommon disease with multiple treatment options, many of which are considered effective, if imperfectly so. In support of this perception of therapeutic efficacy is the previously mentioned mortality statistics which unequivocally demonstrate an improvement in MG care over the course of the last century.13,23,38 The strategies suggested in this chapter represent our approach, based in part on the teaching of our mentors and in part our personal experience, heavily blended with guidance from the literature. Our approach is intended to provide guidance and is not intended to be dogmatic or inflexible in its application.
One management consideration is disease serotype. AChR and seronegative MG are largely considered to have overlapping but non-identical phenotypes and treatment responsiveness. Reports suggest that seronegative MG patients respond better to most treatment modalities than either MuSK or AChR MG but less well to thymectomy than AChBr disease.71,80,304 In general, the weight of existing evidence suggests that MuSK MG is a more severe disease, less treatment responsive, and requires in part a different therapeutic strategy.10,71 Cholinesterase inhibitors are thought to be ineffective and may worsen the disease. Current thinking suggests that thymectomy has a limited role in MuSK MG unless thymoma is identified, an extremely rare occurrence.71,73,77,83,123 Steroids may or may not be effective.78 Expert opinion indicates that MuSK MG responds to other immunomodulating therapies.73,74,80,81,82,305,306–308 Intravenous immunoglobulin (IVIG) may work in some cases of MuSK MG patients refractory to other modalities.308,311 We and others have been impressed with rituximab in refractory MG, both AChR-MG and MuSK-MG.310–313 With LPR4 MG, a distinct therapeutic strategy has not been developed. Current strategy is to treat it identically to traditional AChR MG.
Therapeutic strategies in MG include attempts to treat the disease symptomatically (cholinesterase inhibitors, noninvasive positive pressure ventilation, percutaneous gastrostomy), remove a potential contributor to disease pathogenesis (thymectomy), suppress components of disease immunopathogenesis (immunomodulation through the use of drugs, IVIG, or PLEX), prevent when possible and address when necessary potential exacerbating factors (e.g., avoidance of drugs with neuromuscular blocking properties, treating intercurrent infections and comorbidities such as dysthyroidism), and potentially prevent the escalation from mild to more severe disease. In that regard, it has been suggested that corticosteroids may reduce the conversion frequency of purely ocular to generalized disease.36,37 Although we frequently treat disabling ocular symptoms refractory to pyridostigmine with immunomodulating agents, we do not do so routinely with the goal of reducing the probability of disease generalization.
In general, our philosophy is to treat the patient, not the disease. We attempt to achieve an optimal sense of patient well-being and function, while at the same time considering what an acceptable level of risk might be. This would include consideration of both probability and magnitude of potential side effects. Along those lines, we do not dogmatically adhere to historical therapeutic recommendations such as avoidance of thymectomy in patients >65 years of age or in patients with ocular myasthenia. The reluctance to thymectomize older individuals (without thymoma) is based largely on the atrophic nature of the gland in older individuals. We are unaware of any evidence that supports either decreased efficacy or increased procedural risk based on chronological age alone. Similarly we would consider thymectomy or immunosuppression in a pyridostigmine-resistant individual with limited ocular disease if their visual morbidity significantly interfered with their vocation or quality of life.314–316 Our management strategies are summarized at the end of this section.
Although most patients will not adequately respond to peripheral cholinesterase inhibitors alone, they are frequently used as the initial treatment. This practice evolves from the consideration of their cost and favorable side effect profile. In patients with minor morbidity, they may suffice as the sole treatment. Pyridostigmine is the drug of choice as it has a more favorable duration of action and side effect profile than other drugs of its class. The initial oral dose is typically 15–60 mg three times a day. In a patient with dysphagia, it is typically prescribed 30–60 minutes before meals. We rarely use more than 240 mg a day as higher doses are commonly associated with adverse rather than beneficial effect. If the patient does not achieve the desired therapeutic effect by 360 mg per day, it is our experience that they are unlikely to benefit by further dose escalation. It is also important to recognize that the beneficial effects of pyridostigmine may wane with time if autoimmunity remains unchecked, with unabated destruction of motor end plates resulting in depletion of ACh binding sites. For these reasons, we typically initiate immunomodulating therapy if 240 mg of daily pyridostigmine (60 mg every 6 hours) does not achieve or maintain the desired effect. As we tend to avoid large anticholinesterase doses, cholinergic crisis is a disorder of largely historical interest in our experience.
There are different peripheral cholinesterase inhibitors with different delivery models. Neostigmine can be given parenterally at a dose of 7.5–15 every 4–6 hours and may be the drug of choice for intramuscular delivery.303 Pyridostigmine is also available in a 180 timespan capsule, utilized primarily for treatment of nocturnal symptoms. The timespan formulation is rarely used diurnally due to its variable absorption.38 It is also available in a liquid form (12 mg/ml) and in a parenteral form that can be used in patients who cannot or should not swallow. It is important to be aware that the parenteral dose pyridostigmine is 1/30th to 1/60th of its oral counterpart. The most common side effects are related to the gastrointestinal tract such as abdominal discomfort, nausea and diarrhea. As these symptoms are related to muscarinic, not nicotinic side effects, they may be treated with anticholinergic drugs without adversely affecting the beneficial effects on nicotinic NMT.
The role of parenteral pyridostigmine is primarily in the treatment of MG patients undergoing elective surgery during the period where their oral intake is prohibited. We are less likely to use it during myasthenic crisis in an intubated patient as its potential beneficial effect may be outweighed by increased secretion production as an unwanted side effect.38 Pyridostigmine by itself will rarely, if ever, prevent myasthenic crises or rescue patients from it once it occurs.
Noninvasive positive pressure ventilation has a limited but important role in the management of MG patients. It is used to delay or avoid intubation in a patient with imminent crisis, to help wean a patient from invasive ventilation recovering from crisis, or to treat sleep-disordered breathing when present, which MG patients, like all neuromuscular patients, are susceptible to.317 Percutaneous gastrostomy is virtually never required in MG due to the typical efficacy and relative rapid onset of available MG treatments in restoring functional swallowing and minimizing aspiration risk. We have encountered rare MG patients who have received percutaneous gastrostomy as a result of diagnostic delay, for example, for an anterior cervical osteophyte misidentified as the cause of dysphagia, the gastrostomy tube subsequently removed once effective swallowing was restored.
Symptomatic treatments may be used locally as well. Lid crutches for the treatment of ptosis may be tried but is in our experience rarely tolerated. Topical naphazoline, a sympathomimetic drug with preferential α2 activity, has been reported to provide a marked response in 30% and a worthwhile benefit in an additional 40% of MG patients with ptosis.318 Presumably, the drug acts on the sympathetically innervated lid elevator, Müller’s muscle.
The association with thymic hyperplasia and an apparent therapeutic benefit of thymectomy were first described by Blalock in MG patients in the World War II era319–321 Thymectomy for nonthymomatous patients remains an accepted, but unproven therapeutic option for MG treatment.71,294,322 Estimates suggest the 50–60% of MG patients undergoing thymectomy, particularly those with thymic hyperplasia, will achieve symptomatic remission. This data, however does not account for potential contamination by the known spontaneous remission that may occur as part of the natural history of the disease.71,303 Regarding thymectomy and serotype, complete symptomatic remission is estimated to occur in 54% in AChBR MG, in 40% of patients with seronegative disease, and in 20% of MuSK MG patients with or without thymic hyperplasia.323 Current consensus recommends avoidance of thymectomy in nonthymomatous MuSK MG patients.71,73,77,83,123
The American Academy of Neurology Practice Parameter that reviewed 21 published level II studies of nonthymomatous thymectomy in MG patients found that patients undergoing thymectomy were 1.7 times as likely to improve, 1.6 times as likely to become asymptomatic, and twice as likely to attain medication-free remission.322 The apparent benefits of thymectomy are often delayed with a median time to achieve complete stable remission judged to be in the 17–20 month range.322 Current beliefs are that the benefits of thymectomy are greatest if performed within 3 years of initial symptoms.38 The neuromuscular community awaits the results of an international prospective trial intended to identify the frequency and magnitude of thymectomy benefit in MG patients.323–325
Response appears to vary by thymectomy technique and disease serotype. Historically, remission rate is thought to increase proportionately to the extent of thymic removal, and a transcervical, transsternal “complete” thymectomy is thought to provide the greatest chance of achieving this goal.294 We acknowledge however, that more recent data suggest similar efficacy with less invasive techniques such as video-assisted thoracoscopic surgery.326,327
There is a consensus that all patients with thymoma should have thymectomy to minimize the risk of the morbidity and potential mortality of local invasion, or the smaller risk of disseminated metastasis. It is generally held that MG in patients with thymoma are less responsive to thymectomy in comparison to MG patients with thymic hyperplasia.38
Details pertaining to specific immunomodulating treatments have been provided in Chapter 4. This chapter will focus on specific principles and practices related to the management of the MG patient.
Prior to the initiation of any immunomodulating agent, we attempt to exclude any indolent infectious disease such as tuberculosis or strongyloidiasis that may become symptomatic with treatment. We attempt to identify patients at increased risk because of potential prior exposure to these diseases (e.g., someone who has emigrated from a country where strongyloidiasis is endemic) and liberally obtain skin and serological testing, quantiferon gold assessments and chest x-rays. With corticosteroid use, we obtain baseline bone density determinations and vitamin D levels. All patients on corticosteroids are advised to take calcium carbonate (approximately 1000 mg/day) and vitamin D (approximately 2000 IU/day) supplements. There is an increased risk of symptomatic osteoporosis and pneumocystis in patients treated with corticosteroids or other immunomodulating drugs. Bisphosphonate prophylaxis of glucocorticoid-induced osteopenia has been shown to reduce adverse effects on bone density but has equivocal results regarding reducing the rate of bone fracture.328 Studies provide varying results depending on patient gender, prophylactic drug utilized and dose chosen. We do not routinely provide pneumocystis prophylaxis as we are unaware of any literature that provides adequate guidance regarding risk and risk reduction relevant to the number and type of immunomodulating agents that are used.
Corticosteroids are estimated to benefit 80–90% of myasthenic patients, typically within 2 weeks with maximum benefit occurring on average within 3 months and within 6 months in all patients destined to respond.38,329–332
There are two approaches commonly used in treating patients with corticosteroids. Regardless of the dosage used, the prednisone is typically prescribed as a single morning dose, attempting to mimic the normal physiological diurnal variation of endogenous glucocorticoids as much as possible. Unfortunately, again there is no literature to support any one approach being more effective than the others.
The first approach is the start low, go slow approach advocated by the Johns Hopkins group.44 We typically use this in patients with ocular myasthenia or mild generalized MG. In such cases, we usually initiate prednisone at 20 mg daily with instructions for the patient to increase the dose by 5 mg every 5–7 days as needed to control their symptoms up to a total daily dose of 1.0 mg/kg. We have patients stay on whatever the effective dosage is for one month before slowly tapering down by 5 mg a month to 20 mg daily and then by 2.5 mg every month until we find the lowest possible dose that controls their disease. The downside of this start low, go slow approach is that it takes longer from initiation of steroids to see an effect and does not guarantee avoidance of corticosteroid exacerbation.305
The second approach is to start patients on high dose daily prednisone (e.g., 0.75–1.0 mg/kg) at onset. The benefit of this approach is that patients usually improve faster. The downside is that approximately 10–15% of myasthenics who are started on high-dose prednisone may experience an initial decline in strength during the first week or so.330,332–334 For this reason, we usually reserve this approach to patients with moderate-to-severe MG who are or who will be hospitalized. The mechanism of the exacerbation is not clear. It would appear to be related to NMT as opposed to adverse nerve or muscle effect as it is associated with worsening of the decremental response to repetitive stimulation if employed.334 This weakness tends to dissipate even with continued use of high steroid dosage. It is also possible that the apparent worsening represents situational bias. That is, the patients who are started on high dose prednisone most often have newly diagnosed or progressive MG. The worsening of the MG may be because of the progression of the disease itself and not the addition of the steroids.
There are different approaches even with high dose prednisone treatment. Some authorities advocate for maintaining patients on daily steroids, while others would taper to an alternate day schedule. We typically start patients on daily prednisone for at least 2 weeks and then try the alternate day approach unless the patient has diabetes as this approach renders it more difficult to control glucose levels. As soon as the patient demonstrates clinical benefit, we begin to wean on an every other day basis. Again, there are multiple approaches. One is to double the dose on the odd day and provide no steroids on the even day. The odd day dosage is then gradually reduced. Alternatively, and perhaps preferable due to its less abrupt pattern, we will wean by maintaining the initial dose on the odd day, for example, 60 mg, and with the dose reduction occurring initially on even days, for example, 50 mg. The subsequent wean occurs by continuing the reduction on the lower dose day until it reaches zero. At that point, the wean is continued by sequential reduction of the odd day amount. The rate of weaning depends on how the patient responds. Typically, if the patient is responding, we reduce the dose by 10 mg at a time. If the patient is deemed unresponsive to steroids, or develops unacceptablel side effects, the wean occurs much more rapidly. Once the total dose reaches a semi-arbitrary dose of 20 mg every other day, we typically reduce to the dose by no more than 5 mg or less at each interval. We consider 20 mg every other day, again based on arbitrary data, as the highest acceptable steroid dose that we feel comfortable using on a chronic basis. If another immunomodulating agent is started simultaneously as is commonly the case (see below), we space the dose reduction intervals so as to ideally discontinue the steroids within a 6–12-month time frame. In our experience on average, more problems are created by weaning too fast, than weaning too slow. If the patient demonstrates no benefit within 2 months of institution of corticosteroids, they are rapidly weaned and discontinued.
If immunomodulating treatment is indicated, we commonly initiate a second-line drug along with corticosteroids. The benefit of steroids is their relatively rapid onset of action whereas many of the second- and third-line agents require at least 3 months and typically more time in order to demonstrate efficacy. As we prefer to avoid the long-term effects of corticosteroids that seem inevitable at higher doses, we attempt to provide long-term therapeutic remission with other immunomodulating drugs that are often better tolerated in the long term. Our goal is to achieve a satisfactory response with a second-line, or if necessary a third-line, agent as a monotherapy over the course of the first year. We recognize that the evidence basis by which to justify the use of or to choose a specific nonsteroidal immunomodulating drug for MG treatment is limited. We consider azathioprine333,335–339 as the second-line treatment of choice. Some prefer mycophenolate341–351 as a second-line choice though two randomized clinical trials (discussed below) failed to demonstrate efficacy. Improvement is noted in 70–90% of patients with myasthenia treated with azathioprine, including some patients who are steroid- resistant.333 Although some patients will display early intolerance to azathioprine, we have numerous patients in clinical and side-effect-free remission for over 20 years while on azathioprine monotherapy. Deficiency of enzyme thiopurine methyltransferase predisposes to bone marrow toxicity in patients exposed to azathioprine. We remain uncertain as to whether routine screening for levels of this enzyme is beneficial prior to the initiation of this drug.
Mycophenolate is attractive both for its ease of use and relatively benign side effect profile for an immunomodulating drug. Although the two randomized, blinded, clinical trials reported in 2008 failed to demonstrate a benefit, this may have been a consequence of trial design (studies too short in duration for a response to occur).340,341 A retrospective study demonstrating a therapeutic benefit after 6 months would support this contention.351 Alternatively, the concurrent small prednisone dose, 20 mg daily, that patients received in both the treatment and placebo arm in the MSG study suggests efficacy from this drug that might have masked a potential benefit from mycophenolate. We, as well as others, believe it to be an effective, well-tolerated drug in some patients.
We have had positive experiences with other immunomodulating agents, typically prescribed in those patients who have had inadequate or adverse responses to pyridostigmine, corticosteroids, azathioprine, and/or mycophenolate. In view of their side effect profiles, uncertainty of benefit, monitoring requirements or cost considerations, we tend to use cyclosporine,352–356 tacrolimus,357–367 cyclophosphamide,368–371 and rituximab305–307,310–313,372–374 as third-line treatment agents. We also use methotrexate as third-line agent while awaiting the results of a large randomized, double-blind trial. As mentioned above, rituximab, cyclophosphamide, and cyclosporine have been specifically, albeit anecdotally, reported to benefit MuSK MG patients.305,307,356,369 We have used rituxan in either two or four infusions over the course of 1 month and found it to be quite effective in refractory MuSK and AChR patients (Chapter 4). In our experience, the response to rituximab is durable and often exceeds a year of more before tretreatment is required. We have had a limited experience with sirolimus and remain uncertain regarding its efficacy. We reserve cyclophosphamide for patients’ refractory to other aforementioned treatments.
As the specter of opportunistic infection and neoplasm exists in all patients treated with long-term immunomodulating agents, we give consideration to gradual withdrawal of these agents in any patient who has been in a stable, apparent remission for 2 years or more. It is our impression that many MG patients will have mild, asymptomatic, residual weakness of eye closure despite otherwise excellent disease control. We do not consider this finding to represent a contraindication to attempted weaning from immunomodulating therapy. Given the statistical uncertainty of risk versus benefit, this decision is heavily influenced by patient perspective. We have been successful in this approach in a number of individuals, with or without thymectomy, although are uncertain whether this represents the effect of treatment or the natural history of the disease.
IVIG98,116,375–385 and PLEX381,384,386–398 are considered to be effective treatments for MG.381,384,393,399–402 Their therapeutic support however, has not been without controversy. Historically, many neurologists have considered PLEX superior to IVIG.393 IVIG, however, has been demonstrated to benefit MG patients in a prospective randomized trial whereas no analogous PLEX study has been conducted.377 In addition, IVIG has been suggested to be particularly effective in the treatment of MuSK MG.73,77,308,309,403 As a result, American Academy of Neurology guidelines have identified IVIG as “probably effective and should be considered” in contrast to PLEX where “there is insufficient evidence to support or refute the use of plasma exchange” in MG.385,390 The latter position generated a considerable editorial response from the journal’s readership including many individuals recognized for their expertise in MG management.397 These critics aptly pointed out that PLEX was penalized due to its competitive disadvantage, that is, the unlikelihood of a class 1 study of PLEX in MG ever being conducted. A subsequently published head-to-head class 1 study reinforced the opinion that PLEX performed slightly better than IVIG, although both treatments were found to be statistically equivalent relative to both the degree of efficacy and the number of MG patients that benefitted from their use.399,402
We primarily use IVIG and PLEX in two situations, to avert or treat myasthenic crisis or to “tune up” an MG patient with residual weakness prior to thymectomy.404 This is done hoping to minimize the risk of postoperative complications such as aspiration or prolonged ventilator dependency.400 Typical algorithms involving five infusions or exchanges are used for each. Due to the cost, lifestyle inconvenience and in some cases risk (e.g., need for central venous access in PLEX), we tend to avoid chronic treatment with these two modalities unless other therapeutic options are precluded.
Their greatest benefit of IVIG and PLEX is their relatively rapid onset of action, often within days.381,401 Both are costly although IVIG may enjoy a cost advantage, particularly in the hospitalized patient as it can characteristically be completed in a shorter period of time resulting in a potentially shorter length of stay. PLEX has been reported to have a 90% efficacy in one retrospective study and 65% in a prospective one.396,399 IVIG has been reported to achieve a significant benefit in 69% of patients.399 Exchange can be successfully accomplished in an outpatient setting through peripheral access in the majority of patients. Peripheral versus central venous access minimizes risk, particularly that of a serious risk.396 One additional potential drawback of PLEX is the need to coordinate its timing in relation to immunomodulatory drug administration.
Other novel treatments for MG have been suggested, all intended to address adverse effects while sparing beneficial aspects of immune surveillance.215 One such attempt that appears to have been successful in an in vitro rat model of autoimmune myasthenia is vaccination with cytoplasmic epitopes of the ACh subunits. The intended strategy is to deflect the potentially destructive aspects of autoimmunity away from the extracellular domains of these ACh subunits that the disease targets.405 A second strategy is the development of synthesized anti-sense RNA molecules that target the gene expression of the AChE-R isoform. By doing so, NMT is augmented and potential adverse effects of increased AChE-R levels on muscle that may result from chronic anticholinesterase use are avoided.215 Another apparent successful in vitro strategy is to utilize genetically engineered dendritic cells to present AChR epitopes. The intent here is to specifically kill the T cells responsible for the initiation of the autoimmune response.212 The engineering of monoclonal antibodies that target specific components of disease-specific autoimmunity represents an additional strategy. Blocking of complement activation by eculizumab that is currently being studied in clinical trial is one such strategy. The C5 complement inhibitor rEV576 has also been utilized.406 The tumor necrosis factor alpha blocker etanercept was reported in a small trial to have modest efficacy.407 Allogenic hematopoietic stem cell transplantation has been utilized successfully in one case.408
The treatment of MG in children and adolescents provides a number of additional challenges.96–98,115–117 In adolescents with MG, we attempt to avoid corticosteroids and other immunomodulating drugs if possible in view of the adverse effects on growth and the concern for the suspected increased risk of neoplasm over a patient’s lifetime. Avoidance in adolescent females is of particular concern due to the potential teratogenicity in future pregnancies. We are uncertain of the potential for teratogenicity resulting from immunomodulating drug exposure in males. We have been very supportive of thymectomy in the adolescent female population in the hope of achieving drug-free remission during the patient’s reproductive years. Removal of the thymus in children does not appear to have any deleterious effect on immune system development.333 In a large retrospective series of 149 patients with juvenile MG, 85 patients had a thymectomy while 64 patients were managed medically.97 In the thymectomy group, 82% of patients improved, while 48% went into remission compared to a 63% improvement rate and a 34% remission rate in the patients who are nonthymectomized.97 In another retrospective series of 79 patients with juvenile MG, 65 patients (82%) underwent thymectomy. Of the patients who were thymectomized, remission occurred in 60% compared to 29% in the nonthymectomized group.96 Neither of these studies controlled for baseline severity or concomitant medical treatment, thus the role of thymectomy in juvenile MG like adults remaining unproven.322
Management of MG is challenging both in anticipation of and during pregnancy. Evidence-based guidance is limited. Myasthenics should be treated as high-risk pregnancies both for the mother’s and the child’s welfare. In general, the pregnant myasthenic may be managed with a philosophy similar to the pregnant epileptic. Concerns regarding potential teratogenic effects of drugs need to be balanced with the recognition of harm that could befall both mother and child with inadequate treatment. In both diseases, the smallest effective doses of the safest potential therapies are sought.
MG is not recognized to have any adverse effects on pregnancy, for example, increasing the risk of eclampsia. Pregnancy may influence MG, however, particularly regarding management decisions. Existing information suggests that 1/3 of MG patients will improve, worsen or remain the same during pregnancy.99,409 The first trimester and the postpartum periods are the periods where exacerbation is estimated to most commonly occur.409 Myasthenic morbidity may stem not only from the disease itself but may result from the mechanical effects of the enlarging fetus on diaphragmatic movement.
Regarding management, cholinesterase inhibitors have been used anecdotally without apparent harm. There appears to be little, if any, risk of unwanted stimulation of the myometrium. Magnesium, potentially used for the treatment of pregnancy-related hypertension, is optimally avoided in a pregnant MG patient. There are theoretical concerns that PLEX may adversely affect pregnancy by unwanted effects on hormonal levels, potentially increasing the risk of premature delivery.409 The risks of IVIG during pregnancy are largely unknown. Of the immunomodulating treatment options, corticosteroids are probably the safest although they do pose a slightly increased risk of fetal cleft lip and of premature rupture of the membranes.409 As a general rule, although successful pregnancies have been accomplished under their influence, other immunomodulating drugs are avoided prior to conception and during pregnancy if at all possible.
The effects of pregnancy on MG have already been described. Intercurrent infections and other coexisting autoimmune diseases if uncontrolled are both believed to aggravate MG. Drugs remain the most noteworthy category of influences that aggravates, and in some cases even causes, MG (Table 25-3).
We monitor our patient’s treatment response primarily through clinical assessment. We feel that in most cases, it is at least as effective and undoubtedly more efficient and cost-effective than other testing modalities. As mentioned in Chapter 1, hand-held dynamometry can be a very effective tool for this purpose. As mentioned, monitoring autoantibody titers has little or no role other than potentially assessing the efficacy of thymectomy or monitoring for thymoma reoccurrence. Although electrophysiological parameters can be utilized as an indicator of treatment responsiveness, we find them to be unnecessary in the vast majority of cases. We tend to use them only in confounding situations, that is, attempting to separate myasthenic weakness from that of another potential cause. Cost, time expenditure, and patient comfort are factors that dissuade us from their routine use in patient monitoring. Again, in the spirit of treating the patient, not the disease, electrophysiological monitoring may be too sensitive and potentially lead to excessive treatment. As an example, increased jitter in an asymptomatic muscle is not an indication for treatment initiation or modification.
Assessment scales designed specifically for MG have been used both to stage disease severity, to accurately monitor response to treatment, and to assess quality of life in MG patients.410,411 They are, to the best of our knowledge, utilized more as clinical research tools rather than assessment tools routinely used in the clinic. The Osserman scale is primarily a staging tool.18 Adult MG is subdivided into Group 1 (ocular: 15–20%), Group 2A (mild generalized: 30%), Group 2B (moderately severe generalized: 20%), Group 3 (acute fulminating: 11%), and Group 4 (late severe: 9%). In Europe, the myasthenic muscle score (MMS) developed in 1983 is the preferred metric for determining efficacy in MG clinical trials.412 It is a 100-point scale that assesses trunk as well as oculobulbar, limb and muscle strength. In the United States, the quantified MG score (QMS), also initially proposed in 1983 and later modified, is the preferred instrument. It is a 39-point scale that has considerable overlap with the MMS although it incorporates ventilatory muscle strength and does not address trunk strength beyond neck flexors.412–414
In summary, our typical management strategy for MG is not dogmatic and often varies in consideration of patient and physician preference and individual patient context and comorbidities. We attempt to document a patient’s clinical deficits as quantitatively as possible at baseline in order to improve our ability to make future rational treatment decisions. We obtain baseline thyroid function studies and chest imaging (typically CT) in all patients including those with MuSK autoantibodies in whom demonstration of thymic abnormalities would be extremely unlikely. We would be vigilant for any clinical clues suggesting a synergistic cause of weakness (e.g., hypokalemia) or coexistent autoimmune disease and test accordingly. In patients with minor morbidity, we initiate pyridostigmine, typically at a dose of 60 mg tid. In patients with generalized myasthenia who do not appear to be at the imminent risk of aspiration or ventilatory failure, we typically initiate both corticosteroids at a 1 mg/kg dose (prednisone or methylprednisolone if parenteral therapy is required) along with a second immunomodulating drug, potentially as an outpatient if adequate monitoring can be assured. Hospital admission is suggested to any generalized MG patient placed on high dose corticosteroids; (1) who cannot be reliably monitored as an outpatient, (2) who is symptomatic from a breathing or swallowing perspective, or (3) who appears to be at increased risk of falling if their limb weakness transiently worsens. We have a very low threshold to admitting anyone with symptoms of swallowing or ventilatory muscle weakness to the ICU and would recommend prophylactic intubation when the forced vital capacity declines to <15 mL/kg or the negative inspiratory pressure is <30 cm H2O. Myasthenic crisis is typically treated with intravenous corticosteroids and either IVIG or PLEX with a second-line immunomodulating agent typically added prior to discharge.415,416
Thymectomy is offered to virtually every patient with thymoma, unless comorbidities negate any reasonable chance of benefit. We also offer thymectomy to the majority of adolescents and adults with generalized myasthenia without MuSK autoantibodies assuming that they are strong enough and otherwise healthy enough to safely undergo this treatment. We do so based upon the anecdotal belief that it will increase the probability of future drug-free remission. As a general rule, we do not offer this treatment initially, allowing the patient the opportunity to accept and adapt to their diagnosis. By the same token, based again on the anecdotal suggestion that the efficacy of thymectomy wanes in patients with longstanding MG, we typically offer this option within the first two years of disease if possible.
1. Simpson JA. Myasthenia gravis: A new hypothesis. Scott Med J. 1960;5:419–436.
2. Vincent A. Unravelling the pathogenesis of myasthenia gravis. Nat Rev Immunol. 2002;2(10):797–804.
3. Fambrough DM, Drachman DB, Satyamurti S. Neuromuscular junction in myasthenia gravis: Decreased acetylcholine receptors. Science. 1973; 182:293–295.
4. Patrick J, Lindstrom J. Autoimmune response to acetylcholine receptor. Science. 1973;180(4088):871–872.
5. Lindstrom JM, Seybold ME, Lennon VA, Whittingham S, Duane DD. Antibody to acetylcholine receptor in myasthenia gravis: Prevalence, clinical correlates and diagnostic value. Neurology. 1976;26(11):1054–1059.
6. Hoch W, McConville J, Hels S, Newsom-Davis J, Melms A, Vincent A. Autoantibodies to the receptor tyrosine kinase MuSK in patients with myasthenia gravis without acetylcholine receptor antibodies. Nat Med. 2001;7:365–368.
7. Leite MI, Jacob S, Viegas S, et al. IgG1 antibodies to acetylcholine receptors in seronegative myasthenia gravis. Brain. 2008;131:1940–1952.
8. Jacob S, Viegas S, Leite MI, et al. Presence and pathogenic relevance of antibodies to clustered acetylcholine receptor in ocular and generalized myasthenia gravis. Arch Neurol. 2012; 69(8):994–1001.
9. Higuchi O, Hamuro J, Motomura M, Yamanashi Y. Autoantibodies to low-density lipoprotein receptor-related protein 4 in myasthenia gravis. Ann Neurol. 2011;69:418–422.
10. Baggi F, Andreetta F, Maggi L, et al. Complete stable remission and autoantibody specificity in myasthenia gravis. Neurology. 2013;80:188–195.
11. Phillips LH 2nd, Torner JC. Epidemiologic evidence for a changing natural history of myasthenia gravis. Neurology. 1996;47(5):1233–1238.
12. Aragones JM, Bolibar I, Bonfill X, et al. Myasthenia gravis. A higher than expected incidence in the elderly. Neurology. 2003;60(6):1024–1026.
13. Oosterhuis HJ. The natural course of myasthenia gravis: A long term follow up study. J Neurol Neurosurg Psychiatry. 1989;52:1121–1127.
14. Alter M, Talbert OR, Kurland LT. Myasthenia gravis in a southern community. Arch Neurol. 1960;3:65–69.
15. Cohen MS. Epidemiology of myasthenia gravis. Mongr Allergy. 1987;21:246–251.
16. Garland H, Clark AN. Myasthenia gravis: A personal study of 60 cases. Br Med J. 1956;1:1259–1262.
17. Hokkanen E. Epidemiology of myasthenia gravis in Finland. J Neurol Sci. 1969;9:463–478.
18. Osserman KE, Genkins G. Studies in myasthenia gravis: A review of a 20-year experience in over 1200 patients. Mt Sinai J Med. 1971;38:497–537.
19. Phillips LH, Torner JC, Anderson MS, Cox GM. The epidemiology of myasthenia gravis in central and western Virginia. Neurology. 1992;42:1888–1893.
20. Somnier FE, Keidling N, Paulson OB. Epidemiology of myasthenia gravis in Denmark: A longitudinal and comprehensive population survey. Arch Neurol. 1991;48:733–739.
21. Storm-Mathisen A. Epidemiological and prognostical aspects of myasthenia gravis in Norway. Acta Neurol Scand. 1976;54:120.
22. Storm-Mathisen A. Epidemiology of myasthenia gravis in Norway. Acta Neurol Scand. 1984;70:274–284.
23. Cavalcante P, Le Panse R, Berrih-Aknin S, et al. The thymus in myasthenia gravis: Site of “innate autoimmunity”. Muscle Nerve. 2011;44:467–484.
24. Phillips LH 2nd. The epidemiology of myasthenia gravis. Ann N Y Acad Sci. 2003;998:407–412.
25. Zhang B,Tzartos JS, Belimezi M, et al. Autoantibodies to lipoprotein-related protein 4 in patients with double-seronegative myasthenia gravis. Arch Neurol. 2012;69(4):445–451.
26. Lisak RP. The clinical limits of myasthenia gravis and differential diagnosis do it. Neurology. 1997;48(Suppl 5):S36–S39.
27. Herrmann C. The familial occurrence of myasthenia gravis. Ann N Y Acad Sci. 1971;183:334–350.
28. Hokkanen E, Emeryk-Szajewska B, Rowinska-Marcinska K. Evaluation of the jitter phenomena in myasthenic patients and their relatives. J Neurol. 1978;219:73–82.
29. Grob D. Course and management of myasthenia gravis. JAMA. 1953;153:529–532.
30. Grob D, Brunner NG, Namba T. The natural course of myasthenia gravis and effects of therapeutic measures. Ann N Y Acad Sci. 1981;377:652–669.
31. Landouré G, Knight MA, Stanescu H, et al. A candidate gene for autoimmune myasthenia gravis. Neurology. 2012;79:342–347.
32. Lindstrom J. What initiates the autoimmune response to muscle AChRs in myasthenia gravis? Neurology. 2012;79:304–305.
33. Grob D, Brunner N, Namba T, Pagala M. Lifetime course of myasthenia gravis. Muscle Nerve. 2008;37:141–149.
34. Bever CT, Aquino AV, Penn AS, Lovelace RE, Rowland LP. Prognosis of ocular myasthenia. Ann Neurol. 1983;14:516–519.
35. Grob D, Arsura EL, Brunner NG, Namba T. The course of myasthenia gravis and therapies affecting outcome. Ann N Y Acad Sci. 1987;505:472–499.
36. Mee J, Paine M, Byrne E, King J, Reardon K, O’Day J. Immunotherapy of ocular myasthenia gravis reduces conversion to generalized myasthenia gravis. J Neuroophthalmol. 2003;23:251–255.
37. Kupersmith MJ. Ocular myasthenia gravis: Long-term treatment successes and failures with long-term follow up. J Neurol. 2008;256:1314–1320.
38. Kumar V, Kaminski HJ. Treatment of myasthenia gravis. Curr Neurol Neurosci Rep. 2011;11:89–86.
39. Hughes BW, Kusner LL, Kaminski HJ. Molecular architecture of the neuromuscular junction. Muscle Nerve. 2006;33: 445–461.
40. Keesey JC. Clinical evaluation and management of myasthenia gravis. Muscle Nerve. 2004;29:484–505.
41. Grob D, Brunner NG, Namba T. The natural course of myasthenia gravis and effect of therapeutic measures. Ann New York Acad Sci. 1971;38:497.
42. Lopate G, Pestronk A. Autoimmune myasthenia gravis. Hosp Pract (Office Ed). 1993;28:109–112.
43. Beekman R, Kuks JB, Oosterhuis HJ. Myasthenia gravis: Diagnosis and follow-up of 100 consecutive patients. J Neurol. 1997;244:112–118.
44. Drachman DB. Myasthenia gravis. N Engl J Med. 1994;330: 1797–1810.
45. Borenstein S, Desmedt JE. Temperature and weather correlates of myasthenic fatigue. Lancet. 1974;2:63–66.
46. Borenstein S, Desmedt JE. Local cooling in myasthenia: Improvement on neuromuscular failure. Arch Neurol. 1975;32:152–157.
47. Fennell DF, Ringle SP. Myasthenia gravis and pregnancy. Obstet Gynecol Surv. 1987;41:414–421.
48. Gutmann L. Heat exacerbation of myasthenia gravis. Neurology. 1978;28:398.
49. Gutmann L. Heat-induced myasthenic crisis. Arch Neurol. 1980;37:671–672.
50. Mitchell P, Bebbington M. Myasthenia gravis in pregnancy. Obstet Gynecol. 1992;80:178–181.
51. Evoli A, Batocchi AP, Minisci C, Di Schino C, Tonali P. Therapeutic options in ocular myasthenia gravis. Neuromuscul Disord. 2001;11:208–216.
52. Simpson JF, Westerberg MR, Magee KR. Myasthenia gravis: An analysis of 295 cases. Acta Neurol Scand. 1966;42(Suppl 23): 1–27.
53. Acers TE. Ocular myasthenia gravis mimicking pseudonuclear ophthalmoplegia and variable esotropia. Am J Ophthalmol. 1979;88:319–321.
54. Spooner JW, Baloh RW. Eye movement fatigue in myasthenia gravis. Neurology. 1979;29:29–33.
55. Meriggioli MN, Sanders DB. Autoimmune myasthenia gravis: Emerging clinical and biological heterogeneity. Lancet Neurol. 2009;8(5):475–490.
56. Maher J, Grand’maison F, Nicolle MW, Strong MJ, Bolton CF. Diagnostic difficulties in myasthenia gravis. Muscle Nerve. 1998;21:577–583.
57. Mao VH, Abaza M, Speigel JR, et al. Laryngeal myasthenia gravis: Report of 40 cases. J Voice. 2001;15(1):122–130.
58. Mier A, Laroche C, Green M. Unsuspected myasthenia gravis presenting as respiratory failure. Thorax. 1990;45:422–423.
59. Pal S, Sanyal D. Jaw muscle weakness: A differential indicator of neuromuscular weakness-preliminary observations. Muscle Nerve. 2011;43:807–811.
60. Gracey DR, Divertie MB, Howard FM. Mechanical ventilation for respiratory failure in myasthenia gravis: 2-year experience with 22 patients. Mayo Clin Proc. 1983;58:597–602.
61. Oh SJ, Kuruoglu R. Chronic limb-girdle myasthenia gravis. Neurology. 1992;42:1153–1156.
62. Rodolico C, Toscano M, Autunno S, et al. Limb-girdle myasthenia: Clinical, electrophysiological and morphological features in familial and autoimmune cases. Neuromuscul Disord. 2002;12: 964–969.
63. Jablecki C, Benton A. The frequency of muscle involvement in myasthenia gravis correlates with mean muscle temperature. Muscle Nerve. 1982;5:491–492.
64. Gilad R, Sadeh M. Bilateral foot drop as a manifestation of myasthenia gravis. J Clin Neuromuscul Dis. 2000;2:22–23.
65. Musser WS, Barbano RL, Thornton CA, Moxley RT, Herrmann DN, Logigian EL. Distal myasthenia gravis with a decrement, and increment, and denervation. J Clin Neuromuscul Dis. 2001;3:16–19.
66. Nations SP, Wolfe GI, Amato AA, Jackson CE, Bryan WW, Barohn RJ. Distal myasthenia gravis. Neurology. 1999;52: 632–632.
67. Nicolle MW. Wrist and finger drop in myasthenia gravis. J Clin Neuromuscul Dis. 2006;8:65–69.
68. Ozturk A, Deymeer F, Serdarogly P, et al. Distribution of muscle weakness in myasthenia gravis [abstract]. Neuromuscul Disord. 1999;9:6–7.
69. Janssen JC, Larner AJ, Harris J, Sheean GL, Rossor MN. Myasthenic hand. Neurology. 1998;51:913–914.
70. Ponseti JM, Caritg M, Gamez J, López-Cano M, Vilallonga R, Armengol M. A comparison of long-term post-thymectomy outcome of anti-AChR positive, anti-AChR negative, and anti-MuSK-positive patients with non-thymomatous myasthenia gravis. Expert Opin Bio Ther. 2009;9(1):1–8.
71. McConville J, Farugia ME, Beeson D, et al. Detection and characterization of MuSK antibodies in seronegative myasthenia gravis. Ann Neurol. 2004;55:580–584.
72. Sanders DB, El-Salem K, Massey JM, McConville J, Vincent A. Clinical aspects of MuSK antibody positive seronegative MG. Neurology. 2003;60:1978–1980.
73. Zhou L, McConville J, Chaudhry V, et al. Clinical comparison of muscle-specific tyrosine kinase (MuSK) antibody-positive and -negative myasthenic patients. Muscle Nerve. 2004;30: 55–60.
74. Stickler DE, Massey JM, Sanders DB. MuSK-antibody positive myasthenia gravis: Clinical and electrodiagnostic patterns. Clin Neurophysiol. 2005;116:2065–2068.
75. Yeh J-H, Chen W-H, Chiu H-C, Vincent A. Low frequency of MuSk antibody in generalized seronegative myasthenia gravis among Chinese. Neurology. 2004;62:2131–2132.
76. Wolfe GI, Trivedi JR, Oh SJ. Clinical review of muscle-specific tyrosine kinase-antibody positive myasthenia gravis. J Clin Neuromuscul Dis. 2007;8:217–224.
77. Evoli A, Tonali PA, Padua L, et al. Clinical correlates with anti-MuSK antibodies in generalized seronegative myasthenia. Brain. 2003;126:2304–2311.
78. Bartoccioni E, Scuderi F, Minicuci GM, Marino M, Ciaraffa F, Evoli A. Anti-MuSK antibodies: Correlation with myasthenia gravis severity. Neurology. 2006;67(3):505–507.
79. Deymeer F, Gungor-Tuncer O, Yiolmaz V, et al. Clinical comparison of anti-MuSK- vs. anti-AChR-positive and seronegative myasthenia gravis. Neurology. 2007;60:609–611.
80. Guptill JG, Sanders DB, Evoli A. Anti-MuSK antibody myasthenia gravis: Clinical findings in response to treatment into large cohorts. Muscle Nerve. 2011;44:36–40.
81. Pasnoor M, Wolfe GI, Nations S, et al. Clinical findings in MuSK-antibody positive myasthenia gravis: A US experience. Muscle Nerve. 2010;41:370–374.
82. Lavrnic D, Losen M, Vujic A, et al. The features of myasthenia gravis with autoantibodies to MuSK. J Neurol Neurosurg Psychiatry. 2005;76:1099–1102.
83. Lee JY, Sung JJ, Cho JY, et al. MuSK antibody-positive, seronegative myasthenia gravis in Korea. J Clin Neurosci. 2006;13:353–255.
84. Niks EH, Kuks JB, Vershuuren JJ. Epidemiology of myasthenia gravis with anti-muscle specific kinase antibodies in the Netherlands. J Neurol Neurosurg Psychiatry. 2007;78:417–418.
85. Padua L, Tonali P, Aprile I, Caliandro P, Bartoccioni E, Evoli A. Seronegative myasthenia gravis: Comparison of neurophysiological picture in MuSK+ and MUSK– patients. Eur J Neurol. 2006;13:273–276.
86. Caress JB, Hunt CH, Batish SD. Anti-MuSK myasthenia gravis presenting with purely ocular findings. Arch Neurol. 2005;62:1002–1003.
87. Hanisch F, Eger K, Zierz S. MuSK-antibody positive pure ocular myasthenia gravis. J Neurol. 2006;253:659–660.
88. Farrugia ME, Robson MD, Clover L, et al. MRI and clinical studies of facial and bulbar muscle involvement in MuSK antibody-associated myasthenia gravis. Brain. 2006;129:1481–1492.
89. Punga AR, Ruegg MA. Signaling and aging at the neuromuscular synapse: Lessons learnt from neuromuscular diseases. Clin Opin Pharmacol. 2012;12:340–346.
90. Pevzner A, Schoser B, Peters K, et al. Anti-LRP4 autoantibodies in AchR- and MuSK-antibody-negative myasthenia gravis. J Neurol. 2012;259:427–435.
91. Vincent A, McConville J, Farrugia ME, Newsom-Davis J. Seronegative myasthenia gravis. Semin Neurol. 2004;24:125–133.
92. Gorelick PB, Rosenberg M, Pagano RJ. Enhanced ptosis in myasthenia gravis. Arch Neurol. 1981;38:531.
93. Batocchi AP, Majolini L, Evoli A, Lino MM, Minisci C, Tonali P. Course and treatment of myasthenia gravis during pregnancy. Neurology. 1999;52:447–452.
94. Hoff JM, Dalveit AK, Gilhus NE. Myasthenia gravis: Consequences for pregnancy, delivery, and the newborn. Neurology. 2003;61:1362–1366.
95. Lindner A, Schalke B, Toyka KV. Outcome in juvenile-onset myasthenia gravis: A retrospective study with long-term follow-up of 79 patients. J Neurol. 1997;244:515–520.
96. Rodriguez M, Gomez MR, Howard FM, Taylor WF. Myasthenia gravis in children: Long-term follow-up. Ann Neurol. 1983;13:504–510.
97. Selcen D, Dabrowski ER, Michon AM, Nigro MA. High-dose immunoglobulin therapy in juvenile myasthenia gravis. Pediatr Neurol. 2000;22:40–43.
98. Plauche WC. Myasthenia gravis in mothers and their newborns. Clin Obstet Gynecol. 1991;34:82–99.
99. Bartoccioni E, Evoli A, Casali C, Scoppetta C, Tonali P, Provenzano C. Neonatal myasthenia gravis: Clinical and immunological study of seven mothers and their newborn infants. J Neuroimmunol. 1986;12:155–161.
100. Branch CE, Swift TR, Dyken PR. Prolonged neonatal myasthenia gravis: Electrophysiological studies. Ann Neurol. 1978;3:416–418.
101. Eymard B, Vernet-der Garabedian B, Berrih-Aknin S, Pannier C, Bach JF, Morel E. Anti-acetylcholine receptor antibodies in neonatal myasthenia gravis: Heterogeneity and pathogenic significance. J Autoimmun. 1991;4:185–195.
102. Geddes AK, Kidd HM. Myasthenia gravis of newborn. Can Med Assoc J. 1951;64:152–156.
103. Keesey J, Lindstrom J, Cokeley H. Anti-acetylcholine receptor antibody in neonatal myasthenia gravis. N Engl J Med. 1977; 296:55.
104. Lefvert AK, Osterman PO. Newborn infants to myasthenic mothers: A clinical study and an investigation of acetylcholine receptor antibodies in 17 children. Neurology. 1983;33:133–138.
105. Morel E, Eymard B, Vernet-der Garabedian B, Pannier C, Dulac O, Bach JF. Neonatal myasthenia gravis: A new clinical and immunologic appraisal on 30 cases. Neurology. 1988;38:138–142.
106. Namba T, Brown SB, Grob D. Neonatal myasthenia gravis: Report of two cases and review of the literature. Pediatrics. 1970;45:488–504.
107. Ohta M, Matsubara F, Hayashi K, Nakao K, Nishitani H. Acetylcholine receptor antibodies in infants of mothers with myasthenia gravis. Neurology. 1981;31:1019–1022.
108. Papazian O. Transient neonatal myasthenia gravis. J Child Neurol. 1992;7:135–141.
109. Seybold ME, Lindstrom JM. Myasthenia gravis in infancy. Neurology. 1981;31:476–480.
110. Strickroot FL, Schaeffer BL, Bergo HL. Myasthenia gravis occurring in an infant born of a myasthenic mother. J Am Med Assoc. 1942;120:1207–1209.
111. Tzartos SJ, Efthimiadis A, Morel E, Eymard B, Bach JF. Neonatal myasthenia gravis: Antigenic specificities of antibodies in sera from mothers and their infants. Clin Exp Immunol. 1990;80:376–380.
112. Elias ST, Butler I, Appel SH. Neonatal myasthenia gravis in the infant of a myasthenic mother in remission. Ann Neurol. 1979;6:72–75.
113. Murray EL, Kedar S, Vedanarayanan VV. Transmission of maternal muscle-specific tyrosine kinase (MuSK) to offspring: Report of two cases. J Clin Neuromuscul Dis. 2010;12(2):76–79.
114. Vincent A. Neuromuscular Junction and Other Inherited Disorders. Amsterdam, Netherlands: Elsevier; 2009:575–583.
115. Evoli A, Batocchi AP, Bartoccioni E, Lino MM, Minisci C, Tonali P. Juvenile myasthenia gravis with prepubertal onset. Neuromuscul Disord. 1998;8:561–567.
116. Herrmann DN, Carney PR, Wald JJ. Juvenile myasthenia gravis: Treatment with immune globulin and thymectomy. Pediatr Neurol. 1998;18:63–66.
117. Snead OC, Benton JW, Dwyer D, et al. Juvenile myasthenia gravis. Neurology. 1980;30:732–739.
118. Vial C, Charles N, Chauplannaz G, Bady B. Myasthenia gravis in childhood and infancy: Usefulness of electrophysiologic studies. Arch Neurol. 1991;48:847–849.
119. Evoli A, Minisci C, Di Schino C, et al. Thymoma in patients with MG: Characteristics and long-term outcome. Neurology. 2002;59:1844–1850.
120. Hohlfeld R, Wekerle H. The thymus in myasthenia gravis. Neurol Clin. 1994;12:331–342.
121. Lovelace RE, Younger DS. Myasthenia gravis with thymoma. Neurology. 1997;48(Suppl 5):S76–S81.
122. Drachman DB. Myasthenia gravis. N Engl J Med. 1997; 330(25):1797–1809.
123. Leite MI, Ströbel P, Jones M, et al. Fewer thymic changes in MuSK antibody-negative than in MuSK positive MG. Ann Neurol. 2005;57:444–448.
124. Witt NJ, Bolton CF. Neuromuscular disorders and thymoma. Muscle Nerve. 1988;2:398–405.
125. Vernino S, Lennon VA. Ion channel and striational antibodies define a continuum of autoimmune neuromuscular excitability. Muscle Nerve. 2002;26:702–707.
126. Vernino S, Lennon VA. Muscle and neuronal autoantibody markers of thymoma: Neurological correlations. Ann N Y Acad Sci. 2003;998:359–361.
127. Khella SL, Souyah N, Dalmau J. Thymoma, myasthenia gravis, encephalitis, and a novel anticytoplasmic neuronal antibody. Neurology. 2007;69:1302–1303.
128. Martinelli P, Patuelli A, Minardi C, Cau A, Riviera AM, Dal Posso F. Neuromyotonia, peripheral neuropathy and myasthenia gravis. Muscle Nerve. 1996;19:505–510.
129. Suzuki S, Satoh T, Yasouka H, et al. Novel antibodies to a voltage-gated potassium channel K,1.4 in a severe form of myasthenia gravis. J Neuroimmunol. 2005;170:141–149.
130. Vernino S, Lennon VA. Autoantibody profiles and neurological correlations of thymoma. Clin Cancer Res. 2004;10:7270–7275.
131. Osserman KE, Tsairis P, Weiner LB. Myasthenia gravis and thyroid disease: Clinical and immunological correlation. J Mt Sinai Hosp. 1967;34:469–483.
132. Becker KL, Titus JH, McConahey WM, Woolner LB. Morphologic evidence of thyroiditis in myasthenia gravis. J Am Med Assoc. 1964;187:994–996.
133. Downes JM, Greenwood BM, Wray SH. Autoimmune aspects of myasthenia gravis. Q J Med. 1966;35:85–105.
134. Penn AS, Schotland DL, Rowland LP. Immunology of muscle disease. Res Publ Assoc Res Nerv Ment Dis. 1971;49:215–240.
135. Wolf SM, Rowland LP, Schotland DL, McKinney AS, Hoefer PF, Aranow H Jr. Myasthenia as an autoimmune disease: Clinical aspects. Ann N Y Acad Sci. 1966;135:517–535.
136. Lee EK, Maselli RA, Ellis WG, Agius MA. Morvan’s fibrillary chorea: A paraneoplastic manifestation of thymoma. J Neurol Neurosurg Psychiatry. 1998;65:857–862.
137. Mygland A, Vincent A, Newsom-Davis J, et al. Autoantibodies in thymoma-associated myasthenia gravis with myositis or neuromyotonia. Arch Neurol. 2000;57:527–531.
138. Aarli JA. Neuromyotonia and rippling muscles. Two infrequent concomitants to myasthenia gravis with thymoma. Acta Neurol Scand. 1997;96:342.
139. Heidenreich F, Vincent A. Antibodies to ion-channel proteins in thymoma with myasthenia, neuromyotonia, and peripheral neuropathy. Neurology. 1998;50:1483–1485.
140. Newsom-Davis J, Mills KR. Immunological associations of acquired neuromyotonia (Isaacs’ syndrome): Report of 5 cases and literature review. Brain. 1993;116:453–469.
141. Perini M, Ghezzi A, Basso PF, Montanini R. Association of neuromyotonia with peripheral neuropathy, myasthenia and thymoma: A case report. Ital J Neurol Sci. 1994;15:307–310.
142. Vernino S, Auger R, Emslie-Smith A, Harper CM, Lennon VA. Myasthenia, thymoma, presynaptic antibodies, and a continuum of neuromuscular hyperexcitability. Neurology. 1999;53:1233–1239.
143. Ansevin CF, Agamanolis DP. Rippling muscles and myasthenia gravis with rippling muscles. Arch Neurol. 1996;53:197–199.
144. Muller-Felber W, Ansevin CF, Ricker K, et al. Immunosuppressive treatment of rippling muscles in patients with myasthenia gravis. Neuromuscul Disord. 1999;9:604–607.
145. Nicholas AP, Chatterjee A, Arnold MM, Claussen GC, Zorn GL, Oh SJ. Stiff-persons’ syndrome associated with thymoma and subsequent myasthenia gravis. Muscle Nerve. 1997;20: 493–498.
146. Piccolo G, Martino G, Moglia A, Arrigo A, Cosi CV. Autoimmune myasthenia gravis with thymoma following spontaneous remission of stiff-man syndrome. Ital J Neurol Sci. 1990;11:177–180.
147. Anderson NE, Hutchinson DO, Nicholson GI, Aitcheson F, Nixon JM. Intestinal pseudo-obstruction, myasthenia gravis, and thymoma. Neurology. 1996;47:985–987.
148. Pande R, Leis AA. Myasthenia gravis, thymoma, intestinal pseudo-obstruction, and neuronal nicotinic acetylcholine receptor antibody. Muscle Nerve. 1999;1600–1602.
149. Katz JS, Wolfe GI, Bryan WW, Tintner R, Barohn FU. Acetylcholine receptor antibodies in the Lambert–Eaton myasthenic syndrome. Neurology. 1998;50:470–475.
150. Newsom-Davis J, Leys K, Ferguson I, Modi G, Mills K. Immunological evidence for the co-existence of the Lambert–Eaton myasthenic syndrome and myasthenia gravis in two patients. J Neurol Neurosurg Psychiatry. 1991;54:452–453.
151. Oh SJ. Overlap myasthenic syndrome. Neurology. 1987;37: 1411–1414.
152. Tabbaa MA, Leschner RT, Campbell WW. Malignant thymoma with dysautonomia and disordered neuromuscular transmission. Arch Neurol. 1986;43:955–957.
153. Taphoorn MJB, Van Duijn H, Wolters ECH. A neuromuscular transmission disorder: Combined myasthenia gravis and Lambert–Eaton syndrome in one patient. J Neurol Neurosurg Psychiatry. 1988;51:880–882.
154. Namba T, Brunner NG, Grob D. Idiopathic giant cell polymyositis. Report of a case and review of the literature. Arch Neurol. 1974;31:27–30.
155. Johns TR, Crowley WJ, Miller JQ, Campa JF. The syndrome of myasthenia and polymyositis with comments on therapy. Ann N Y Acad Sci. 1971;183:64–71.
156. Burke JS, Medline NM, Katz A. Giant cell myocarditis and myositis associated with thymoma and myasthenia gravis. Arch Pathol Lab Med. 1969;88:359–366.
157. Pascuzzi RM, Roos KL, Phillips LH. Granulomatous inflammatory myopathy associated with myasthenia gravis. A case report and review of the literature. Arch Neurol. 1986;43:621–623.
158. Alseth EH, Maniaol AH, Elsais A, et al. Investigation for RAPSN and DOK-7 mutations in a cohort of seronegative myasthenia gravis patients. Muscle Nerve. 2011;43:574–577.
159. Burns TM, Russell JA, LaChance D, Jones HR. Oculobulbar involvement is typical with Lambert–Eaton myasthenic syndrome. Ann Neurol. 2003;53:270–273.
160. Lennon VA. Serologic profile of myasthenia gravis and distinction from the Lambert-Eaton myasthenic syndrome. Neurology. 1997;48(Suppl 5):S23–S27.
161. Ruff RL, Rutecki P. Faster, slower, but never better: Mutations of the skeletal muscle acetylcholine receptor. Neurology. 2012;79:404–405.
162. Kaminski HJ, Suarez J, Ruff RL. Neuromuscular junction physiology in myasthenia gravis: Isoforms of the acetylcholine receptor in extraocular muscle and the contribution of sodium channels to the safety factor. Neurology. 1997;48:S8–S17.
163. Engel AG, Shen X-M, Selcen D. What we learned from the congenital myasthenic syndromes. J Mol Neurosci. 2010;40: 143–153.
164. Birks RI, Huxley HE, Katz B. The fine structure of the neuromuscular junction in the frog. J Physiol. 1960;150:134–144.
165. Ruff RL, Lennon V. End-plate voltage-gated sodium channels are lost in clinical and experimental myasthenia gravis. Ann Neurol. 1998;43:370–379.
166. Barrett EF, Magleby KL. Physiology of cholinergic transmission. In: Goldberg AM, Hahn P, eds. Biology of Cholinergic Function. New York, NY: Raven Press; 1976:29–100.
167. Steinbach JH, Stevens CF. Neuromuscular transmission. In: Llinas R, Precht W, eds. Progress Neurobiology. Berlin: Springer-Verlag; 1976:35–92.
168. Kuffler SW, Yoshikami D. The number of transmitter molecules in a quantum: An estimate from iontophoretic application of acetylcholine at the neuromuscular synapse. J Physiol. 1975;251:465–482.
169. Martin AR. Quantal nature of synaptic transmission. Physiol Rev. 1986;46:51–66.
170. Potter LT. Synthesis, storage and release of 14 C acetylcholine in isolated rat diaphragm muscles. J Physiol. 1970;206:145–166.
171. Rahamimoff R, Erulkar SD, Lev-Tov A, Meiri H. Intracellular and extracellular calcium ions in transmitter release at the neuromuscular synapse. Ann N Y Acad Sci. 1978;307:583–598.
172. Gautam M, Noakes PG, Moscoso L, et al. Defective neuromuscular synaptogenesis in agrin-deficient mutant mice. Cell. 1996;85(4):525–535.
173. Engel AG, Santa T. Histometric analysis of the ultra-structure of the neuromuscular junction in myasthenia gravis and the myasthenic syndrome. Ann N Y Acad Sci. 1971;183:46–63.
174. Engel AG, Tsujihata M, Lindstrom JM, Lennon VA. The motor end-plate in myasthenia gravis and in experimental autoimmune myasthenia gravis. A quantitative ultrastructural study. Ann N Y Acad Sci. 1976;274:60–79.
175. Punga AR.Stalberg E. Acetylcholinesterase inhibitors in MG: To be or not to be? Muscle & Nerve. 2009;39(6):724–728.
176. Vigny M, Bon S, Massoulie J, Leterrier F. Active-site catalytic efficiency of acetylcholinesterase molecular forms in Electrophorus, torpedo, rat, and chicken. Eur J Biochem. 1978;85:317–323.
177. Hubbard JI. Microphysiology of vertebrate neuromuscular junction transmission. Physiol Rev. 1973;53:674–723.
178. Pennefather P, Quasterl DM. Relation between subsynaptic receptor blockade and response to quantal transmitter at the mouse neuromuscular junction. J Gen Physiol. 1981;78: 313–344.
179. Ricker K, Hertel G, Stodieck S. Influence of temperature on neuromuscular transmission in myasthenia gravis. J Neurol. 1977;216:273–282.
180. Stamboulis E, Lygidakis C. Local warming in myasthenia gravis. Electromyogr Clin Neurophysiol. 1984;24:429–435.
181. Foldes FF, Kuze S, Vizi ES, Deery A. The influence of temperature on neuromuscular performance. J Neural Transm. 1978;43:27–45.
182. Lass Y, Fischback GD. A discontinuous relationship between the acetylcholine-activated channel conductance and temperature. Nature. 1976;263:150–151.
183. Lang H, Trontelj J. Effect of temperature on NCV and NAP of human nerve. Muscle Nerve. 1986;9:573.
184. Bolton CF, Sawa GM, Carter K. The effects of temperature on human compound action potentials. J Neurol Neurosurg Psychiatry. 1981;44:407–413.
185. Louis AA, Hotson JR. Regional cooling of human nerve and slowed NA+ inactivation. Electroenceph Clin Neurophysiol. 1986;63:371–375.
186. Hubbard JI, Jones SF, Landau EM. The effect of temperature change upon transmitter release, facilitation and post-tetanic potentiation. J Physiol. 1971;216:591–609.
187. Kistler J, Stroud RM, Klymkowsky MW, Lalancette RA, Fairclough RH. Structure and function of an acetylcholine receptor. Biophys J. 1982;37:371–383.
188. Gillespie SK, Balasubramanian S, Fung ET, Huganir RL. Rapsyn clusters and activates the synapse-specific receptor tyrosine kinase MuSK. Neuron. 1996;16:953–962.
189. Evoli A, Lindstrom J. Myasthenia gravis with antibodies to MuSK. Neurology. 2011;77:1783–1784.
190. Frail DE. McLaughlin LL. Mudd J. Merlie JP. Identification of the mouse muscle 43,000-dalton acetylcholine receptor-associated protein (RAPsyn) by cDNA cloning. J Biol Chem. 1988;263(30):15602–15607.
191. Lonenzoni PJ, Scola RH, Kay CS, Werneck LC. Congenital myasthenic syndrome: A brief review. Pediatr Neurol. 2012; 46:141–148.
192. Conti-Tronconi B, Raferty M. The nicotinic cholinergic receptor: Correlation of molecular structure with functional properties. Annu Rev Biochem. 1982;51:491–530.
193. Anderson CR, Stevens CF. Voltage clamp analysis of acetylcholine produced end-plate current fluctuations at frog neuromuscular junction. J Physiol. 1973;235:655–691.
194. Katz B, Miledi R. The statistical nature of the acetylcholine potential and its molecular components. J Physiol. 1972; 224:665–699.
195. Boyd IA, Martin AR. Spontaneous subthreshold activity at mammalian neuromuscular junctions. J Physiol. 1956;132: 61–73.
196. Desmedt JE. Nature of the defect of neuromuscular transmission in myasthenic patients: Post-tetanic exhaustion. Nature. 1957;179:156–157.
197. Desmedt JE. Myasthenic-like features of neuromuscular transmission after administration of an inhibitor of acetylcholine synthesis. Nature. 1958;182:1673–1674.
198. Salpeter MM, Harris R. Distribution and turnover rate of acetylcholine receptors throughout the function folds at a vertebrate neuromuscular junction. J Cell Biol. 1983;96:1781–1785.
199. Tzartos S, Seybold M, Lindstrom J. Specificities of antibodies to acetylcholine receptors in sera from myasthenia gravis patient measured by monoclonal antibodies. Proc Natl Acad Sci USA. 1982;79:188–192.
200. Bray J, Drachman D. Binding affinities of anti-acetylcholine receptor autoantibodies in myasthenia gravis. J Immunol. 1982;128:105–110.
201. Masuda T, Motomura M, Utsugisawa K, et al. Antibodies against the main immunogenic region of the acetylcholine receptor correlate with disease severity of myasthenia gravis. J Neurol Neurosurg Psychiatry. 2012;83:935–940.
202. Engel AG, Sahashi K, Fumagalli G. The immunopathology of acquired myasthenia gravis. Ann N Y Acad Sci. 1981;377: 158–174.
203. Engel AG, Lambert EH, Howard FM. Immune complexes (IgG and C3) at the motor end-plate in myasthenia gravis. Ultrastructural and light microscopic localization and electrophysiological correlations. Mayo Clin Proc. 1977;52: 267–280.
204. Engel AG, Lindstrom JM, Lambert EH, Lennon VA. Ultrastructural localization of the acetylcholine receptor in myasthenia gravis and in its experimental autoimmune model. Neurology. 1977;27:307–315.
205. Santa T, Engel AG, Lambert EH. Histometric study of neuromuscular junction ultrastructure. Neurology. 1972;22:71–82.
206. Schonbeck S, Chrestel S, Hohlfeld R. Myasthenia gravis: Prototype of the antireceptor autoimmune diseases. Int Rev Neurobiol. 1990;32:175–200.
207. Fumagalli G, Engel AG, Linstrom J. Ultrastructural aspects of acetylcholine receptor turnover at the normal end-plate and in autoimmune myasthenia gravis. J Neuropathol Exp Neurol. 1982;41:567–579.
208. Stanley EF, Drachman DB. Effect of myasthenic immunoglobulin on acetylcholine receptors of intact mammalian neuromuscular junctions. Science. 1978;200:1285–1289.
209. Drachman DB. The biology of myasthenia gravis. Annu Rev Neurosci. 1981;4:195–225.
210. Drachman DB, Adams RN, Josifek LF, Pestronk A, Stanley EF. Antibody-mediated mechanisms of ACh receptor loss in myasthenia gravis: Clinical relevance. Ann N Y Acad Sci. 1981;377:175–188.
211. Kao I, Drachman DB. Myasthenic immunoglobulin accelerates acetylcholine receptor degradation. Science. 1977;196:527–529.
212. Sun W, Adams RN, Miagkov A, Lu Y, Juon HS, Drachman DB. Specific immunotherapy of experimental myasthenia gravis in vitro and in vivo: The guided missile strategy. J Neuroimmunol. 2012;251:25–32.
213. Newsom-Davis J, Willcox N, Scadding G, Calder L, Vincent A. Antiacetylcholine receptor antibody synthesis by cultured lymphocytes in myasthenia gravis: Thymic and peripheral blood cell interactions. Ann N Y Acad Sci. 1981;377:393–402.
214. Scadding GK, Vincent A, Newsom-Davis J, Henry K. Acetylcholine receptor antibody synthesis by thymic lymphocytes: Correlation with thymic histology. Neurology. 1981;31:935–943.
215. Kim JY, Park KD, Richman DP. Treatment of myasthenia gravis based on its immunopathogenesis. J Clin Neurol. 2011;7:173–183.
216. Shigemoto K, Kubo S, Maruyama N, et al. Induction of myasthenia by immunization against muscle-specific kinase. J Clin Invest. 2006;116(4):1016–1024.
217. Klooster R, Plomp JJ, Huijbers MG, et al. Muscle-specific kinase myasthenia gravis IgG4 autoantibodies cause severe neuromuscular junction dysfunction in mice. Brain. 2012; 135:1081–1101.
218. Shiraishi H, Motomura M, Yoshimura T, et al. Acetylcholine receptors loss and postsynaptic damage in MuSK antibody-positive myasthenia gravis. Ann Neurol. 2005;57:289–293.
219. Slater C. Diverse aspects of vulnerability at the neuromuscular junction. Brain. 2012;135:997–1001.
220. Kawakami Y, Ito M, Hirayama M, et al. Anti-MuSK autoantibodies block binding of collagen Q to MuSK. Neurology. 2011;77:1819–1826.
221. Mihaylova V, Salih MA, Mukhtar MM, et al. Refinement of the clinical phenotype in MuSK-related congenital myasthenic syndromes. Neurology. 2009;73:1926–1928.
222. Richman DP. Antibodies to low density lipoprotein receptor-related protein 4 in seronegative myasthenia gravis. Arch Neurol. 2012;69(4):434–435.
223. Verschuuren JJ, Palace J, Gilhus NE. Clinical aspects of myasthenia explained. Autoimmunity. 2010;43(5–6):344–352.
224. MacLennan C, Beeson D, Buijs A-M, Vincent A, Newsom-Davis J. Acetylcholine receptor expression in human extraocular muscles and their susceptibility to myasthenia gravis. Ann Neurol. 1997;41:423–431.
225. Serafini B, Cavalcante P, Bernasconi P, Aloisi F, Mantegazza R. Epstein-Barr virus in myasthenia gravis thymus: A matter of debate. Ann Neurol. 2011;70(3):519.
226. Genkins G, Papatestas AE, Horowitz SH, Kornfield P. Studies in myasthenia gravis: Early thymectomy. Electrophysiologic and pathologic correlations. Am J Med. 1975;58:517–524.
227. Papatestas AE, Genkins G, Horowitz SH, Kornfeld P. Thymectomy in myasthenia gravis: Pathologic, clinical, and electrophysiologic correlations. Ann N Y Acad Sci. 1976;274:555–573.
228. Rivner MH, Swift TR. Thymoma: Diagnosis and management. Semin Neurol. 1990;10:83–88.
229. Oh SJ, Kim DE, Kuruoglu R, Bradley RJ, Dwyer D. Diagnostic sensitivity of the laboratory tests in myasthenia gravis. Muscle Nerve. 1992;15:720–724.
230. Vincent A, Newsom-Davis J. Acetylcholine receptor antibody as a diagnostic test for myasthenia gravis: Results in 153 validated cases and 2967 diagnostic assays. J Neurol Neurosurg Psychiatry. 1985;48:1246–1252.
231. Vincent A. Acetylcholine receptor antibody as a diagnostic test for myasthenia gravis: Results in 153 validated cases and 2967 diagnostic assays. J Neurol Neurosurg Psychiatry. 2012;83(3): 237–238.
232. Sanders DB, Andrews I, Howard JF, Massey JM. Seronegative myasthenia gravis. Neurology. 1997;48(Suppl 5):S40–S45.
233. Vincent A, Newsom-Davis J, Newton P, Beck N. Acetylcholine receptor antibody and clinical response to thymectomy in myasthenia gravis. Neurology. 1983;33:1276–1282.
234. Chan KH, Lachance DH, Harper CM, Lennon VA. Frequency of seronegativity in adult-acquired generalized myasthenia gravis. Muscle Nerve. 2007;36:651–658.
235. Howard FM, Lennon VA, Finley J, Matsummoto J, Elvebach LR. Clinical correlations of antibodies that bind, block, or modulate human acetylcholine receptors in myasthenia gravis. Ann N Y Acad Sci. 1987;505:526–538.
236. Ohta K, Shigemoto K, Kubo S, et al. MuSK antibodies in AChR Ab-seropositive MG vs AChR Ab-seronegative MG. Neurology. 2004;62:2132–2133.
237. Zouvelou V, Kyriazi S, Rentzos M, et al. Double-seropositive myasthenia gravis. Muscle Nerve. 2013;47(3):465–466.
238. Yamamoto AM, Gajdos P, Eymard B, et al. Anti-titin antibodies in myasthenia gravis: Tight association with thymoma and heterogeneity of nonthymoma patients. Arch Neurol. 2001;58: 885–890.
239. Lanska DJ. Diagnosis of thymoma in myasthenics using antistriated muscle antibodies: Predictive value and gain in diagnostic certainty. Neurology. 1991;41:520–524.
240. Lange DJ. Electrophysiological testing of neuromuscular transmission. Neurology. 1995;48(suppl 5):S18–S22.
241. Punga AR, Sawada M, Stalberg EV. Electrophysiological signs and the prevalence of adverse effects of acetylcholinesterase inhibitors in patients with myasthenia gravis. Muscle Nerve. 2008;37(3):300–307.
242. Howard JF Jr, Sanders DB, Massey JM. The electrodiagnosis of myasthenia gravis and the Lambert-Eaton myasthenic syndrome. Neurol Clin. 1994;12(2):305–330.
243. Keesey JC. Electrodiagnostic approach to defects of neuromuscular transmission. Muscle Nerve. 1989;12:613–626.
244. Stalberg E, Sanders DB. Effect of temperature on neuromuscular transmission. Muscle Nerve. 1986;9:573.
245. Daube JR. Electrodiagnostic studies in ALS and other motor neuron disorders. Muscle Nerve. 2000;23:1488–1502.
246. Yamashita S, Sakaguchi H, Mori A, et al. Significant CMAP decrement by repetitive nerve stimulation is more frequent in median than ulnar nerves of patients with amyotrophic lateral sclerosis. Muscle Nerve. 2012;45(3):426–428.
247. Henderson RD, Daube JR. Decrement in surface-recorded motor unit potentials in amyotrophic lateral sclerosis. Neurology. 2004;63:1670–1674.
248. Maselli RA, Wollman RL, Leung C, et al. Neuromuscular transmission in amyotrophic lateral sclerosis. Muscle Nerve. 1993;16:1193–1203.
249. Wang FC, DePasqua V, Gérard P, Delwaide PJ. Prognostic value of decremental responses to repetitive nerve stimulation in ALS patients. Neurology. 2001;57:897–899.
250. Barbieri S, Weiss GM, Daube JR. Fibrillation potentials in myasthenia gravis. Muscle Nerve. 1982;5:S50.
251. Oosterhuis HJ, Hootsmans WJ, Veenhuyzen HB, van Zadelhoff I. The mean duration of motor unit action potential in patients with myasthenia gravis. Electroencephalogr Clin Neurophysiol. 1972;32:697–700.
252. Stalberg E. Clinical electrophysiology in myasthenia gravis. J Neurol Neurosurg Psychiatry. 1980;43:622–633.
253. Gilchrist JM, Sanders DB. Double-step repetitive stimulation in myasthenia gravis. Muscle Nerve. 1987;10:233–237.
254. Kelly JJ, Daube JR, Lennon VA, Howard FM Jr, Younge BR. The laboratory diagnosis of mild myasthenia gravis. Ann Neurol. 1982;12:238–342.
255. Liam Oey P, Wieneke GH, Hoogenraad TU, van Huffelen AC. Ocular myasthenia gravis: The diagnostic yield of repetitive stimulation and stimulated single fiber EMG of orbicularis oculi muscle and infrared reflection oculography. Muscle Nerve. 1993;16:142–149.
256. Oh SJ, Eslami N, Nishihira T, et al. Electrophysiological and clinical correlation in myasthenia gravis. Ann Neurol. 1982;12:348–354.
257. Ozdemir C, Young RR. The results to be expected from electrical testing in the diagnosis of myasthenia gravis. Ann N Y Acad Sci. 1976;274:203–222.
258. Schumm F, Stohr M. Accessory nerve stimulation in the assessment of myasthenia gravis. Muscle Nerve. 1984;7:147–151.
259. Oh SJ, Hatanaka Y, Hemmi S, et al. Repetitive nerve stimulation of facial muscles in MuSK antibody-positive myasthenia gravis. Muscle Nerve. 2006;33:500–504.
260. Churchill-Davidson HC, Wise RP. Neuromuscular transmission in the newborn infant. Anesthesiology. 1963;24:271–278.
261. Gatev V, Stamatova B, Angelova B, Ivanov I. Effects of repetitive stimulation on the electrical and mechanical activities of muscles in normal children. Electromyogr Clin Neurophysiol. 1975;15:339–355.
262. Koenigsberger MR, Patten B, Lovelace RE. Studies of neuromuscular function in the newborn: 1. A comparison of myoneural function in the full term and premature infant. Neuropaediatrie. 1973;4:350–361.
263. Hays RM, Michaud LJ. Neonatal myasthenia gravis: Specific advantages of repetitive stimulation over edrophonium testing. Pediatr Neurol. 1988;4:245–247.
264. Sanders DB. Clinical impact of single-fiber electromyography. Muscle Nerve. 2002;11(Suppl):15–20.
265. Jabre JF, Chirico-Post J, Weiner M. Stimulation SFEMG in myasthenia gravis. Muscle Nerve. 1989;12:38–42.
266. Trontelj JV, Mihelin M, Fernandez JM, Stalberg E. Axonal stimulation for end-plate jitter studies. J Neurol Neurosurg Psychiatry. 1986;49:677–685.
267. Trontelj JV, Khuraibet A, Mihelin M. The jitter in stimulated orbicularis oculi muscle: Technique and normal values. J Neurol Neurosurg Psychiatry. 1988;51:814–819.
268. Trontelj JV, Stalberg E, Mihelin M. Jitter in the muscle fiber. J Neurol Neurosurg Psychiatry. 1990;53:49–54.
269. Trontelj JV, Stalberg E, Mihelin M. Jitter of the stimulated motor axon. Muscle Nerve. 1992;15:449–454.
270. Stålberg EV, Sanders DB. Jitter recordings with concentric needle electrodes. Muscle Nerve. 2009;40:331–339.
271. Farrugia ME, Weir AI, Cleary M, Cooper S, Metcalfe R, Mallik A. Concentric and single fiber needle electrodes yield comparable jitter results in myasthenia gravis. Muscle Nerve. 2009;39(5):579–585.
272. Benatar M, Hammad M, Doss-Riney H. Concentric-needle single-fiber electromyography for the diagnosis of myasthenia gravis. Muscle Nerve. 2006;34(2):163–168.
273. Sanders DB. Measuring jitter with concentric needle electrodes. Muscle Nerve. 2013;47:317–318.
274. Gilchrist JM. Single fiber EMG reference values: A collaborative effort. Muscle Nerve. 1992;15:151–161.
275. Kouyoumdjian JA, Stålberg EV. Reference jitter values for concentric needle electrodes in voluntarily activated extensor digitorum communis and orbicularis oculi muscles. Muscle Nerve. 2008;37(6):694–699.
276. Cruz Martinez A, Ferrer MT, Diez Tejedor E, Perez Conde MC, Anciones B, Frank A. Diagnostic yield of single fiber electromyography and other electrophysiological technique in myasthenia gravis I. Electromyography, automatic analysis of the voluntary pattern, and repetitive nerve stimulation. Electromyogr Clin Neurophysiol. 1982;22:377–393.
277. Stalberg E, Ekstedt J, Broman A. Neuromuscular transmission in myasthenia gravis studied with single fiber electromyography. J Neurol Neurosurg Psychiatry. 1974;37:540–547.
278. Sanders DB, Howard JF, Johns TR. Single fiber electromyography in myasthenia gravis. Neurology. 1979;29:68–76.
279. Massey JM, Sanders DB. Single fiber electromyography in myasthenia gravis during pregnancy. Muscle Nerve. 1993;16: 458–460.
280. Murga L, Sanchez F, Menedez C, Castilla JM. Diagnostic yield of stimulated and voluntary single-fiber electromyography in myasthenia gravis. Muscle Nerve. 1998;21:1081–1083.
281. Sanders DB, Massey JM. Does change in neuromuscular jitter predict or correlate with clinical change in myasthenia gravis? Neurology. 2013;80:P02.204.
282. Konishi T, Nishitani H, Matsubara F, Ohta M. Myasthenia gravis: Relation between jitter in single-fiber EMG and antibody to acetylcholine receptor. Neurology. 1981;31:386–392.
283. Sitzer G, Brune GG. Effect of cholinesterase inhibitors and thymectomy on single fiber EMG in myasthenia gravis. Ann N Y Acad Sci. 1981;377:884–886.
284. Emeryk B, Rowinska K, Nowad-Michalska T. Do true remissions in myasthenia gravis really exist? An electrophysiological study. J Neurol. 1985;231:331–335.
285. Osserman KE, Kaplan LI. Rapid diagnostic test for myasthenia gravis: Increased muscle strength, without fasciculations, after intravenous administration of edrophonium (Tensilon) chloride. J Am Med Assoc. 1952;150:265–268.
286. Osserman KE, Genkins G. Clinical reappraisal of the use of edrophonium (Tensilon) chloride tests in myasthenia gravis and significance of clinical classification. Ann N Y Acad Sci. 1965;135:312–326.
287. Seybold ME, Daroff RB, Hachinski V. The office tensilon test for ocular myasthenia gravis. Arch Neurol. 1986;43:842–844.
288. Phillips LH 2nd, Melnick PA. Diagnosis of myasthenia gravis in the 1990s. Semin Neurol. 1990;10:62–69.
289. Moorthy G, Behrens MM, Drachman DB, et al. Ocular pseudomyasthenia or ocular myasthenia “plus”: A warning to clinicians. Neurology. 1989;39:1150–1154.
290. Mulder DW, Lambert EH, Eaton LM. Myasthenic syndrome in patients with ALS. Neurology. 1959;9:627–631.
291. Ragge NK, Hoyt WF. Midbrain myasthenia: Fatigable ptosis, lid twitch sign, and ophthalmoparesis from a dorsal midbrain glioma. Neurology. 1992;42:917–919.
292. Dirr LY, Donofrio PD, Patton JF, Troost BT. A false-positive edrophonium test in a patient with a brainstem glioma. Neurology. 1989;39:865–867.
293. Takahashi K, Al-Janabi NJ. Computed tomography and magnetic resonance imaging of mediastinal tumors. J Magn Reson Imaging. 2010;32(6):1325–1339.
294. Jaretzki A III. Thymectomy for myasthenia gravis: Analysis of the controversies regarding technique and results. Neurology. 1997;48:52S–63S.
295. Chagnac Y, Hadani M, Goldhammer Y. Myasthenic crisis after intravenous administration of iodinated contrast agent. Neurology. 1985;35(8):1219–1220.
296. Fenichel GM, Shy GM. Muscle biopsy experience in myasthenia gravis. Arch Neurol. 1963;9(3):237–243.
297. Engel WK, McFarlin DE. Muscle lesions in myasthenia gravis. Discussion. Ann N Y Acad Sci. 1966;135(1):68–78.
298. Maselli RA, Richman DP, Willmann RI. Inflammation at the neuromuscular junction in myasthenia gravis. Neurology. 1991;41:1497–1504.
299. Pascuzzi RM, Campa JF. Lymphorrhage localized to the muscle end-plate on myasthenia gravis. Arch Pathol Lab Med. 1988;112:934–937.
300. Selcen D, Fukuda T, Shen X-M, Engel AG. Are MuSK antibodies the primary cause of myasthenic symptoms? Neurology. 2004;62:1945–1950.
301. Lindstrom J, Lambert EH. Content of acetylcholine receptor and antibodies bound to receptor in myasthenia gravis, experimental autoimmune myasthenia gravis, and Eaton–Lambert syndrome. Neurology. 1978;28:130–138.
302. Lev-Lehmann E, Evron T, Broide RS, et al. Synaptogeneis and myopathy under acetylcholinesterase overexpression. J Mol Neurosci. 2000;14:93–105.
303. Massey JM. Treatment of acquired myasthenia gravis. Neurology. 1997;48(Suppl 5):S46–S51.
304. Soliven BC, Lange DJ, Penn AS, et al. Seronegative myasthenia gravis. Neurology. 1988;38:514–517.
305. Baek WS, Bashey A, Sheean GL. Complete remission induced by rituximab in refractory, seronegative, muscle-specific kinase positive myasthenia gravis. J Neurol Neurosurg Psychiatry. 2007;78:771.
306. Díaz-Manera J, Martínez-Hernandez E, Querol L, et al. Long-lasting treatment effect of rituximab in MuSK myasthenia. Neurology. 2012;78:189–193.
307. Hain B, Jordan K, Deschauer M, Zierz S. Successful treatment of MuSK antibody-positive myasthenia gravis with rituximab. Muscle Nerve. 2006;33(4):575–580.
308. Shibata-Hamaguchi, A, Samuraki M, Furui E, et al. Long-term effect of intravenous immunoglobulin on anti-MuSK antibody-positive myasthenia gravis. Acta Neurol Scand. 2007;116:406–408.
309. Takahashi H, Kawaguchi N, Nemoto Y, Hattori T. High-dose intravenous immunoglobulin for the treatment of MuSK antibody-positive seronegative myasthenia gravis. J Neurol Sci. 2006;247:239–241.
310. Nowak RJ, Dicapua DB, Zebardast N, Goldstein JM. Response of patients with refractory myasthenia gravis to rituximab: A retrospective study. Ther Adv Neurol Disord. 2011;4:259–266.
311. Collongues N, Casez O, Lacour A, et al. Rituximab in refractory and non-refractory myasthenia: A retrospective multicenter study. Muscle Nerve. 2012;46:687–691.
312. Steiglbauer K, Topakian R, Schäffer G, Aichner FT. Rituximab for myasthenia gravis: Three case reports and review of the literature. J Neurol Sci. 2009;280:120–122.
313. Blum S, Gillis D, Brown H, et al. Use and monitoring of low-dose rituximab in myasthenia gravis. J Neurol Neurosurg Psychiatry. 2011;82:659–663.
314. Agius MA. Treatment of ocular myasthenia gravis with corticosteroids: Yes. Arch Neurol. 2000;57:750–751.
315. Kaminski HJ, Daroff RB. Treatment of ocular myasthenia. Steroids only when compelled. Arch Neurol. 2000;57:752–753.
316. Benatar M, Kaminski HJ. Evidence report: The medical treatment of ocular myasthenia (an evidence-based review). Neurology. 2007;68:2144–2149.
317. Nicolle MW, Rask S, Koopman WJ, George CF, Adams J, Wiebe S. Sleep apnea in patients with myasthenia gravis. Neurology. 2006;67(1):140–142.
318. Nagane Y, Utsugisawa K, Suzuki S, et al. Topical naphazoline in the treatment of myasthenic blepharoptosis. Muscle Nerve. 2011;44:41–44.
319. Blalock A, Mason MF, Morgan HJ, Riven SS. Myasthenia gravis and tumors of the thymic region. Report of a case in which the tumor was removed. Ann Surg. 1939;110:544–561
320. Blalock A. Thymectomy in the treatment of myasthenia gravis. Report of twenty cases. J Thorac Surg. 1944;13:316–339.
321. Blalock A, Harvey AM, Ford FR, Lilientha JL Jr. The treatment of myasthenia gravis by removal of the thymus gland. Preliminary report. JAMA. 1945;127:1089–1096.
322. Gronseth GA, Barohn RB. Practice parameter: Thymectomy for autoimmune myasthenia gravis (an evidence-based review): Report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology. 2000;55: 1–7.
323. Wolfe GI, Kaminski HJ, Jaretzki A III, Swan A, Newsom-Davis J. Development of a thymectomy trial in nonthymomatous myasthenia gravis patients receiving immunosuppressive therapy. Ann NY Acad Sci. 2003;998:473–480.
324. Aban IB, Wolfe GI, Cutter GR, et al. The MGTX experience: Challenges in planning and executing an international, multicenter clinical trial. J Neuroimmunol. 2008;201–202:80–84.
325. Newsome-Davis J, Cutter G, Wolfe GI, et al. Status of thymectomy trial for nonthymomatous myasthenia gravis patients receiving prednisone. Ann NY Acad Sci. 2008;1132:344–347.
326. Matee MJ, Mack MJ. Surgical approaches to the thymus in patients with myasthenia gravis. Thorac Surg Clin. 2009;19: 83–89.
327. Manlulu A, Lee TW, Wan I, et al. Video-assisted thoracic surgery thymectomy for nonthymomatous myasthenia gravis. Chest. 2005;128:3454–3460.
328. Pereira RM, Carvalho JF, Paula AP, et al. Guidelines for the prevention and treatment of glucocorticoid-induced osteoporosis. Rev Bras Rheumatol. 2012;52(4):580–593.
329. Pascuzzi RM, Coslett HB, Johns TR. Long-term corticosteroid treatment of myasthenia gravis: Report of 116 patients. Ann Neurol. 1984;15:291–298.
330. Johns TR. Long-term corticosteroid treatment of myasthenia gravis. Ann NY Acad Sci. 1987;505:568–583.
331. Schneider-Gold C, Gajdos P, Toyka KV, Hohlfeld RR. Corticosteroids for myasthenia gravis. Cochrane Database Syst Rev. 2005;2:CD002828.
332. Bae JS, Go SM, Kin BJ. Clinical predictors of steroid-induced exacerbation in myasthenia gravis. J Clin Neurosci. 2006;13:1006–2010.
333. Sanders DB, Scoppetta C. The treatment of patients with myasthenia gravis. Neurol Clin. 1994;12:343–368.
334. Miller RG, Milner-Brown HS, Mirka A. Prednisone-induced worsening of neuromuscular function in myasthenia gravis. Neurology. 1986;36:729–732.
335. Palace J, Newsom-Davis J, Lecky B. A randomized double-blind trial of prednisolone alone or with azathioprine in myasthenia gravis. Myasthenia Gravis Study Group. Neurology. 1998;50:1778–1783.
336. Herrllinger U, Weller M, Dichgans J, Melms A. Association of primary central nervous system lymphoma with long-term azathioprine therapy for myasthenia gravis. Ann Neurol. 2000;47:682–683.
337. Hohlfeld R, Michels M, Heininger K, Besinger U, Toyka KV. Azathioprine toxicity during long-term immunosuppression of generalized myasthenia gravis. Neurology. 1988;38: 258–261.
338. Kissel JT, Levy RJ, Mendell JR, Griggs RC. Azathioprine toxicity in neuromuscular disease. Neurology. 1986;36:35–39.
339. Gajdos P, Elkharrat D, Chevret S, Chastang C. A randomized clinical trial comparing prednisone and azathioprine in myasthenia gravis. Results of the second interim analysis. J Neurol Neurosurg Psychiatry. 1993;56:1157–1163.
340. The Muscle Study Group. A trial of mycophenolate mofetil with prednisone as initial immunotherapy in myasthenia gravis. Neurology. 2008;71:394–399.
341. Sanders DB, Hart IK, Mantegazza R, et al. An international, phase III, randomized trial of mycophenolate mofetil in myasthenia gravis. Neurology. 2008;71:400–406.
342. Bromberg MB, Wald JJ, Forshew DA, Feldman EL, Albers JW. Randomized trial of azathioprine or prednisone for initial immunosuppressive treatment of myasthenia. J Neurol Sci. 1997;150:59–62.
343. Meriggioli MN, Rowin J, Richman JG, Leurgans S. Mycophenolate mofetil for myasthenia gravis: A double-blind, placebo-controlled pilot study. Ann N Y Acad Sci. 2003;998:494–499.
344. Meriggioli MN, Ciafaloni E, Al-Hayk KA, et al. Mycophenolate mofetil for myasthenia gravis: An analysis of efficacy, safety, and tolerability. Neurology. 2003;61:1438–1440.
345. Sanders D, McDermott M, Thornton C, Tawil A, Barohn R; the Muscle Study Group. A trial of mycophenolate mofetil (MMF) with prednisone as initial immunotherapy in myasthenia gravis (MG) [abstract]. Neurology. 2007;68: A107.
346. Ciafaloni E, Massey JM, Tucker-Lipscomb B, Sanders DB. Mycophenolate mofetil for myasthenia gravis: An open-label pilot study. Neurology. 2001;56:97–99.
347. Vernino S, Salomao DR, Habermann TM, O’Neill BP. Primary CNS lymphoma complicating treatment of myasthenia gravis with mycophenolate mofetil. Neurology. 2005;65:639–641.
348. Hauser RA, Malek AR, Rosen R. Successful treatment of a patient with severe refractory myasthenia gravis using mycophenolate mofetil. Neurology. 1998;51:912–913.
349. Sollinger HW; Renal Transplant Mycophenolate Mofetil Study Group. Mycophenolate mofetil for the prevention of acute rejection in primary cadaveric renal allograft recipients. U.S. Renal Transplant Mycophenolate Mofetil Study Group. Transplantation. 1995;60:225–232.
350. Chaudhry V, Cornblath DR, Griffin JW, O’Brien R, Drachman DB. Mycophenolate mofetil: A safe and promising immunosuppressant in neuromuscular diseases. Neurology. 2001;56:94–96.
351. Hehir MK, Burns TM, Alpers J, Conaway MR, Sawa M, Sanders DB. Mycophenolate mofetil in AChR-antibody-positive myasthenia gravis: Outcomes in 102 patients. Muscle Nerve. 2010;41(5):593–598.
352. Ciafaloni E, Nikhar NK, Massey JM, Sanders DB. Retrospective analysis of the use of cyclosporine in myasthenia gravis. Neurology. 2000;55:448–450.
353. Sanders DB, Ciafaloni E, Nikhar NK, Massey JM. Retrospective analysis of the use of cyclosporine in myasthenia [abstract]. Neurology. 2000;54(Suppl 3):A394.
354. Tindall RS, Rollins JA, Phillips JT, Greenlee RG, Wells L, Belendiuk G. Preliminary results of a double-blind, randomized, placebo-controlled trial of cyclosporine in myasthenia gravis. N Engl J Med. 1987;316:719–724.
355. Tindall RS, Phillips JT, Rollins JA, Wells L, Hall K. A clinical therapeutic trial of cyclosporine in myasthenia gravis. Ann N Y Acad Sci. 1993;681:539–551.
356. Kurokawa T, Nishiyama T, Yamamoto R, Kishida H, Hakii Y, Kuroiwa Y. Anti-MuSK antibody positive myasthenia gravis with HIV infection successfully treated with cyclosporin: A case report. Rinsho Shinkeigaku. 2008;48(9):666–669.
357. Yoshikawa H, Kiuchi T, Saida T, Takamori M. Randomised double blind, placebo controlled study of tacrolimus in myasthenia gravis. J Neurol Neurosurg Psychiatry. 2011;82:970–977.
358. Ponseti JM, Gamez J, Azem J, et al. Post-thymectomy combined treatment of prednisone and tacrolimus versus prednisone alone for the consolidation of complete stable remission in patients with myasthenia gravis: A non-randomized, non-controlled study. Curr Med Res Opin. 2007;23:1269–1278.
359. Ponseti JM, Azem J, Fort JM, et al. Long-term results of tacrolimus in cyclosporine- and prednisone-dependent myasthenia gravis. Neurology. 2005;64:1641–1643.
360. Nagane Y, Utsugisawa K, Obara D, Kondoh R, Terayama Y. Efficacy of low-dose FK506 in the treatment of myasthenia gravis-a randomized pilot study. Eur Neurol. 2005;53: 146–150.
361. Benatar M, Sanders D. the importance of studying history: Lessons learnt from a trial of tacrolimus in myasthenia gravis. J Neurol Neurosurg Psychiatry. 2011;82:945.
362. Evoli A, Di Schino C, Marsili F, Punzi C. Successful treatment of myasthenia gravis with tacrolimus. Muscle Nerve. 2002;25(1):111–114.
363. Konishi T, Yoshiyama Y, Takamori M, Yagi K, Mukai E, Saida T; Japanese FK506 MG Study Group. Clinical study of FK506 in patients with myasthenia gravis. Muscle Nerve. 2003;28(5):570–574.
364. Sanders DB, Aarli JA, Cutter GR, Jaretzki A III, Kaminski HJ, Phillips LH II. Long-term results of tacrolimus in cyclosporine- and prednisone-dependent myasthenia gravis [comment]. Neurology. 2006;66(6):954–955.
365. Schneider-Gold C, Hartung HP, Gold R. Mycophenolate mofetil and tacrolimus: New therapeutic options in neuroimmunological diseases. Muscle Nerve. 2006;34(3):284–291.
366. Tada M, Shimohata T, Tada M, et al. Long-term therapeutic efficacy and safety of low-dose tacrolimus (FK506) for myasthenia gravis. J Neurol Sci. 2006;247(1):17–20.
367. Wakata N, Saito T, Tanaka S, Hirano T, Oka K. Tacrolimus hydrate (FK506): Therapeutic effects and selection of responders in the treatment of myasthenia gravis. Clin Neurol Neurosurg. 2003;106(1):5–8.
368. Lewis RA, Lisak RP. “Rebooting” the immune system with cyclophosphamide: Taking risks for a “cure”? Ann Neurol. 2003;53:7–9
369. Lin PT, Martin BA, Winacker AB, So YT. High-dose cyclophosphamide in refractory myasthenia with MuSK antibodies. Muscle Nerve. 2006;33:433–435.
370. DeFeo LG, Schottlender J, Martelli NA, Molfino NA. Use of intravenous pulsed cyclophosphamide in severe, generalized myasthenia gravis. Muscle Nerve. 2002;26:32–36.
371. Drachman DB, Jones RJ, Brodsky RA. Treatment of refractory myasthenia: “Rebooting” with high-dose cyclophosphamide. Ann Neurol. 2003;53:29–34.
372. Kuntzer T, Carota A, Novy J, Cavassini M, Du Pasquier RA. Rituximab is successful in an HIV positive patient with MuSK myasthenia. Neurology. 2011;76:757–758.
373. Maddison P, McConville J, Farrugia ME, et al. The use of rituximab in myasthenia gravis and Lambert-Eaton myasthenic syndrome. J Neurol Neurosurg Psychiatry. 2011;82:671–673.
374. Gajra A, Vajpayee N, Grethlein SJ. Response of myasthenia gravis to rituximab in a patient with non-Hodgkin lymphoma. Am J Hematol. 2004;77(2):196–197.
375. Gajdos P, Chevret S, Toyka K. Intravenous immunoglobulin for myasthenia gravis. Cochrane Database Syst Rev. 2006;19: CD002277.
376. Gajdos P, Tranchant C, Clair B, et al. Myasthenia Gravis Clinical Study Group Treatment of myasthenia gravis exacerbation with intravenous immunoglobulin 1 g/kg versus 2 g/kg: A randomized double blind clinical trial. Ann Neurol. 2005;62: 1689–1693.
377. Zinman L, Ng E, Bril V. IV immunoglobulin in patients with myasthenia gravis: A randomized controlled trial. Neurology. 2007;68:837–841.
378. Achiron A, Barak Y, Miron S, Sarova-Pinhas I. Immunoglobulin treatment in refractory myasthenia gravis. Muscle Nerve. 2000;23:551–555.
379. Cosi V, Lombardi M, Piccolo G, Erbetta A. Treatment of myasthenia gravis with high-dose intravenous immunoglobulin. Acta Neurol Scand. 1991;84:81–84.
380. Dwyer JM. Manipulating the immune system with immune globulin. N Engl J Med. 1992;326:107–116.
381. Gajdos P, Chevret S, Clair B, Tranchant C, Chastang C. Clinical trial of plasma exchange and high-dose intravenous immunoglobulin in myasthenia gravis. Ann Neurol. 1997;41:789–796.
382. Howard JF Jr. Intravenous immunoglobulin for the treatment of acquired myasthenia gravis. Neurology. 1998;51(Suppl 5): S30–S36.
383. Jongen JL, van Doorn PA, van der Meche FG. High-dose intravenous immunoglobulin therapy for myasthenia gravis. J Neurol. 1998;245:26–31.
384. Qureshi AI, Choudhry MA, Akbar MS, et al. Plasma exchange versus intravenous immunoglobulin treatment in myasthenic crisis. Neurology. 1999;52:629–632.
385. Li HF, Gao X, Hong Y, et al. Evidence-based guideline: Intravenous immunoglobulin in the treatment of neuromuscular disorders: Report of the Therapeutics and Technology Assessment Subcommittee of the American Academy of Neurology. Neurology. 2012;78:1009–1015.
386. NIH Consensus Conference. The utility of therapeutic plasmapheresis for neurological disorders. JAMA. 1986;256:1333–1337.
387. Pinching AF, Peters DK, Newsom-Davis J. Remission of myasthenia gravis following plasma exchange. Lancet. 1976;2:1373–1376.
388. Antozzi C, Gemma M, Regi B, et al. A short plasma exchange protocol is effective in severe myasthenia gravis. J Neurol. 1991;238:103–107.
389. Mandawat A, Mandawat A, Kaminski H, Shaker Z, Alawi AA, Alshekhlee A. Outcome of plasmapheresis in myasthenia gravis: Delayed therapy is not favorable. Muscle Nerve. 2011;43:578–584.
390. Cortese I, Chaudhry V, So YT, Cantor F, Cornblath DR, Rei-Grant A. Evidence-based guideline update: Plasmapheresis in neurologic disorders: Report of the Therapeutics and Technology Assessment Subcommittee of the American Academy of Neurology. Neurology. 2011;76:294–300.
391. Gajdos P, Chevret S, Toyka K. Plasma exchange for myasthenia gravis. Cochrane Database of Syst Rev. 2002;(4):CD002275.
392. Yeh JH, Chiu HC. Plasmapheresis in myasthenia gravis. A comparative study of daily versus alternately daily schedule. Acta Neurol Scand. 1999;99:147–151.
393. Stricker RB, Kwiatkowski BJ, Habis JA, Kiprov DD. Myasthenic crisis: Response to plasmapheresis following failure of intravenous gamma-globulin. Arch Neurol. 1993;50:837–840.
394. Howard JF. The treatment of myasthenia gravis with plasma exchange. Semin Neurol. 1982;2:273–279.
395. Mahalati K, Dawson RB, Collins JO, Mayer RF. Predictable recovery for myasthenia gravis crisis with plasma exchange: 36 cases and review of current management. J Clin Apher. 1999;14:1–8.
396. Guptill JT, Oakley D, Kuchibhatla M, et al. A retrospective study of complications of therapeutic plasma exchange in myasthenia. Muscle Nerve. 2013;47:170–176.
397. Kaminski H, Cutter G, Ruff RL, Wolfe G. Evidence-based guideline update: Plasmapheresis in neurologic disorders. Neurology. 2011;77:e101–e102.
398. Dau PC, Lindstrom JM, Cassel CK, Denys EH, Shev EE, Spitler LE. Plasmapheresis and immunosuppressive drug therapy in myasthenia gravis. N Engl J Med. 1977;297:1134–1140.
399. Barth D, Nabavi Noure M, Ng E, Nwe P, Bril V. Comparison of IVIg and PLEX in patients with myasthenia gravis. Neurology. 2011;76:2017–2023.
400. Mandawat A, Kaminski HJ, Carter G, Katirji B, Alshekhlee A. Comparative analysis of therapeutic options used for myasthenia gravis. Ann Neurol. 2010;68(6):797–805.
401. Ronager J, Ravnborg M, Hermansen I, Vorstrup S. Immunoglobulin treatment versus plasma exchange in patients with chronic moderate to severe myasthenia gravis. Artif Organs. 2001;25:967–973.
402. Bril V, Barnett-Tapia C, Barth D, Katzberg HD. IVIG and PLEX in the treatment of myasthenia gravis. Ann N Y Acad Sci. 2012;1275(1):1–6.
403. Sanders DB, Massey J, Juel V. MuSK antibody positive myasthenia gravis: Response to treatment in 31 patients [abstract]. Neurology. 2007;68:A299.
404. Huang C-S, Hsu H-S, Kao K-P, Huang M-H, Huang B-S. Intravenous immunoglobulin in the preparation of thymectomy for myasthenia gravis. Acta Neurol Scand. 2003;108: 136–138.
405. Luo J, Kuryatov A, Lindstrom JM. Specific immunotherapy of experimental myasthenia gravis by a novel mechanism. Ann Neurol. 2010;67:441–451.
406. Soltys J, Kusner LL, Young A, et al. A novel complement inhibitor limits severity of experimental myasthenia gravis. Ann Neurol. 2009;65:67–75.
407. Rowin J, Meriggioli MN, Tüzün E, Leurgans S, Christadoss P. Etanercept treatment in in corticosteroid-dependent myasthenia gravis. Neurology. 2004;63:2390–2392.
408. Strober J, Cowan MJ, Horn BN. Allogeneic hematopoietic cell transplantation for refractory myasthenia gravis. Arch Neurol. 2009;66:659–661.
409. Berlit S, Tuschy B, Spaich S, Sütterlin M, Schaffelder R. Myasthenia gravis in pregnancy: A case report. Case Rep Obstet Gynecol. 2012;2012:736024.
410. Burns TM. More than meets the eye: The benefits of listening closely to what our patients with myasthenia gravis are telling us. Muscle Nerve. 2012;46:153–154.
411. Farrugia ME, Vincent A. Autoimmune mediated neuromuscular junction defects. Curr Opin Neurol. 2010;23:489–495.
412. Gajdos P, Sharshar T, Chevret S. Standards of measurements in myasthenia gravis. Ann N Y Acad Sci. 2003;998:445–452.
413. Barohn RJ, McIntire D, Herbelin L, Wolfe GI, Nations S, Bryan WW. Reliability testing of the quantitative myasthenia gravis score. Ann N Y Acad Sci. 1998;841:769–772.
414. Bedlack RS, Simel DL, Bosworth H, Samsa G, Tucker-Lipscomb B, Sanders DB. Quantitative myasthenia gravis score: Assessment of responsiveness and longitudinal validity. Neurology. 2005;64(11):1968–1970.
415. Berrouschot J, Baumann I, Kalischewski P, Sterker M, Schneider D. Therapy of myasthenic crisis. Crit Care Med. 1997;25:1228–1235.
416. Thomas CE, Mayer SA, Gunger Y, et al. Myasthenic crisis: Clinical features, mortality, complications, and risk factors for prolonged intubation. Neurology. 1997:48–1253–1260.
417. Vincent A, Leite MI. Neuromuscular junction autoimmune disease: Muscle specific kinase antibodies and treatments for myasthenia gravis.. Curr Opin Neurol. 2005;18(5):519–525.
418. Lehky TJ, Iwamoto FM, Kreisel TN, Floeter MK, Fine HA. Neuromuscular junction toxicity with tandutanib induces and myasthenic-like syndrome. Neurology. 2011;76:236–241.