Treatment and Management of Disorders of the Neuromuscular Junction


Disorders of the neuromuscular junction include myasthenia gravis (MG), Lambert-Eaton myasthenic syndrome (LEMS), botulism, congenital myasthenic syndrome (CMS), snakebite myasthenia, organophosphate poisoning (OPP), and hypermagnesium-induced paralysis. 1

1 Tick paralysis is excluded from this chapter because it is due to conduction block at the node of Ranvier.

For this chapter, I reviewed all existing major literature on the subjects of management and treatment of these diseases and analyzed critically the data for evidence-based information. The literatures include review articles and large case series from MEDLINE and Cochrane reviews. On the basis of evidence-based data, the studies are categorized into four classes: class I, randomized controlled trials available; class II, controlled trials without randomization or randomized trial with small patient number; class III, uncontrolled trials; and class IV, case series. This classification and the recommendations are based on guidance by the European Federation of Neurological Societies (EFNS) scientific task forces, which were modified slightly from the American Academy of Neurology guidelines in 1999 ( ; ).

The therapeutic guidelines are formulated largely on the basis of best available evidence-based data in principles, and the ratings of recommendation are made depending on the evidence-based classification: Level A (established as effective, ineffective, or harmful) requires at least one convincing class I study or at least two consistent convincing class II studies; Level B (probably effective, ineffective, or harmful) requires at least one convincing class II study or overwhelming class III evidence; and Level C (possibly effective, ineffective, or harmful) requires at least two convincing class III studies. The European Task Force adopted “good practice point” recommendations when there is a uniform consensus for recommendation among the experts, although there is inadequate evidence for a formal recommendation ( ). Usually, this recommendation is based on class IV data. When there is no consensus among the experts, therapeutic guidelines and recommendations are made on the basis of my personal experience or my personal choice for the best possible regimen.

Myasthenia Gravis

Myasthenia gravis (MG) is due to an antibody-induced postsynaptic defect of the neuromuscular junction, producing exertional weakness or easy fatigability and, commonly, oculobulbar palsy . Electron microscopic studies show simplification of the postsynaptic fold at the neuromuscular junction, which is induced by the acetylcholine receptor (AChR) antibody.

MG is the most common disorder of neuromuscular junction. The prevalence of MG is approximately 5 to 10:100,000 population. The disease affects all ages. In the 1970s–1980s, two peaks were identified in MG: young females and old males. In more recent studies, an upsurge of MG was noted in older patients, especially males, whereas female predominance was maintained in young patients.

With the advent and widespread use of immunotherapy and the improvement in respiratory care and intensive care unit (ICU) management during MG crisis over the past 40 years, the prognosis of MG patients has improved vastly. In a Danish population-based study, the overall 3-, 5-, 10-, and 20-year survival was 85%, 81%, 69%, and 63% respectively ( ). Most MG patients can return to a full, productive life with adequate therapy.

There is considerable evidence indicative of an autoimmune mechanism in the pathogenesis of MG. The strongest evidence is seen in experimental autoimmune myasthenia gravis. Within 2 weeks after injection of AChR antigen purified from electric eels, animals develop weakness, which is reversed by edrophonium. Their electrophysiological and pharmacological responses are identical to those in human MG. The thymus seems to be the major organ initiating AChR antibody formation in MG, which eventually damages the postsynaptic fold.

MG is characterized by a fluctuating weakness of skeletal muscles with remission and exacerbation. Classically, weakness got worse with exertion or exercise and oculobulbar symptoms are common, seen in 90% of MG patients. Commonly, such patients complain of droopy eye lid, diplopia, swallowing or speech difficulty, and proximal muscle weakness. In 85% of the patients, MG becomes generalized, usually within 3 years. Rarely, respiratory difficulty is the initial symptom. Exertional weakness (myasthenic or fatigable weakness) can be documented on examination by the repetitive exercise.

Diagnosis and Evaluation

The diagnosis of MG is based on history and clinical findings. This is confirmed by the edrophonium test, AChR-Ab test, muscle-specific kinase (MuSK)-Ab test, lipoprotein receptor–related protein 4 (LRP4) and agrin antibody (Ab) tests, the repetitive nerve stimulation (RNS) test, and single-fiber electromyography (SFEMG).

The diagnosis of MG is easily confirmed by the intravenous (IV) edrophonium test, a short-acting acetylcholinesterase inhibitor (AChEI) ( ). Dramatic improvement is noted within 1 minute after injection. This test is positive in 95% of MG patients. The edrophonium test is still the most helpful, simple, and rapid test for the diagnosis of MG. To perform an adequate edrophonium test, placebo injection and objective measurement of two or three clinical parameters are crucial. Placebo injection has been helpful in sorting out many pseudo-MG patients in whom both the edrophonium test and placebo test were positive. A positive edrophonium test has also been reported in other diseases. Understandably, it is positive in penicillamine-induced MG and in MG unmasked or precipitated by drugs. The edrophonium test was positive in 89% of patients with MLOS (myasthenia-gravis Lambert-Eaton mysthenic syndrome overlap syndrome), in 37% of patients with LEMS, and in 27% of those with botulism. Among the CMSs, patients with endplate AChR deficiency and choline acetyltransferase (ChAT) deficiency (familial infantile myasthenia) showed a positive test. A false-negative edrophonium test may be seen in ocular MG (OMG), especially when ophthalmoplegia is severe, or when ophthalmoplegia is too minimal for objective measurement. In this case, the prostigmine test is often positive. A positive edrophonium test is less common in MuSK MG than in other forms of MG, being positive in half of the cases. Since 1979, a simple and inexpensive “ice-pack test” (application of ice over one eye for 5 minutes) has been found to be positive for OMG ( ). This test was used in patients with MG, normal individuals, and a few cases of third nerve palsy and was positive only for MG in 90% of cases.

The serum AChR antibody has become the more specific test for MG, although it is positive in 70%–95% of cases, with an average of 85% of cases ( ). False-positive responses have rarely been reported in other diseases. However, the titer is not correlated with disease severity. MuSK antibody was positive in an average of 20% to 49% of seronegative generalized MG (GMG), with an average rate of 39% ( ). MuSK-Ab is negative in OMG. MuSK-Ab should be tested in all AChR-Ab-negative cases. In recent years, two new antibodies were found in MG. LRP4 antibody was identified in 1.4% (United States)–50% (Germany) of AChR-Ab-negative MG patients. Agrin antibody was found in 7.5%–15% of MG patients regardless of AChR antibody status ( ; ).

Other objective diagnostic tests include the RNS test and the SFEMG ( ; ).

The RNS test is the time-honored test for the neuromuscular transmission (NMT) disorders and offers the advantages of relative simplicity and rapid results. Although certain patterns on the RNS test are indicative of MG, it is less specific than the AChR-Ab assay. The distinct advantages of the RNS test over the AChR-Ab assay are that this test can provide a rapid and objective diagnosis of MG and can be used serially for the evaluation of severity of disease. The RNS test is positive in 75% of cases when the distal muscles (usually hand muscles) and proximal muscles (usually facial and trapezius muscles) are tested together. The most common types of RNS abnormalities are those typical of postsynaptic NMT blocks: (1) normal compound muscle action potential (CMAP) amplitude, (2) normal or minimal postexercise facilitation (PEF), (3) decremental response at low-rate stimulation (LRS), (4) normal or decremental response at high-rate stimulation (HRS), and (5) posttetanic facilitation (PTF) followed by posttetanic exhaustion (PTE) ( Figs. 19.1 and 19.2 ). Among these, the decremental response at LRS is the most common and characteristic RNS finding in MG. In MuSK-MG, an RNS test of limb muscle had a relatively low yield (25%–57%), whereas an RNS test of facial muscles showed a decrement in 85% of cases. This is because of the predominant involvement of facio-bulbar muscle in MuSK-MG.

Fig. 19.1, Typical repetitive nerve stimulation pattern in the abductor digiti quinti muscle in mild myasthenia gravis. Normal compound muscle action potential (CMAP) amplitude at rest and after exercise (CMAP Ex). Decremental response (−19% at 3 Hz and −15% at 5 Hz at low-rate stimulation. Normal response at high rate response (50 Hz). Posttetanic facilitation (PTF, 5 Hz stimulation immediately after 50 Hz) and posttetanic exhaustion (PTE, 5 Hz stimulation) 4 minutes after 50 Hz stimulation are present.

Fig. 19.2, Typical repetitive nerve stimulation (RNS) pattern in the abductor digiti quinti and flexor carpi ulnaris (FCU) muscles in severe myasthenia gravis (MG). (A) Normal compound muscle action potential (CMAP) at rest and after exercise (CMAP Ex). (B) Decremental response at low-rate stimulation (2,3,5 Hz) and at high-rate stimulation (50 Hz). (C) Posttetanic facilitation (PTF) and posttetanic exhaustion (PTE) are also present.

The SFEMG is the single most sensitive clinical test for MG, being positive in 77%–100% of MG cases, with an average rate of 95%. The test is so sensitive that Stålberg and Trontelj concluded that the diagnosis of MG can be abandoned if abnormal jitter is not present in a weak muscle. The classic SFEMG pattern in MG is characterized by a definite increase in jitter with or without neuromuscular blocking. For obvious reasons, the SFEMG is most useful in “double or triple” seronegative MG cases in which the RNS test was negative. In mild GMG and OMG, in which the RNS test and AChR-Ab are often normal, this test can be the crucial means of confirming the diagnosis of MG. In none of our MG patients were all three tests found to be negative. Thus, MG can be confidently ruled out if all three tests are negative.

Treatment and Management

In 2016, an international consensus guidance for management of MG was published ( ). In 2006, the EFNS Task Force published consensus guidelines for the treatment of autoimmune NMT disorders ( ). In general, we will follow these consensus guidelines with some modification. There are two main modes of treatment in MG: (1) symptomatic treatment and (2) immunotherapy.

Symptomatic Treatment of Myasthenia Gravis

Acetylcholinesterase Inhibitors

The mainstay of symptomatic treatment is AChEI, which is effective in almost all cases. Poorer response was reported in MuSK-MG. One series showed unresponsiveness in 71% of 14 cases ( ).

AChEIs inhibit the breakdown of ACh at the neuromuscular junction and increase the availability of ACh to stimulate AChR and facilitate muscle activation and contraction. AChEIs used in MG bind reversibly to AChE as opposed to organophosphate (OP) AChEIs, which bind irreversibly ( Table 19.1 ). These drugs cross the blood-brain barrier poorly and generally remain in the extracellular space.

Table 19.1
Pharmacological Properties of Acetylcholine-Esterase Inhibitors
Drug Dosage Form Equivalent Doses Onset, Peak, and Duration of Drugs Initial Dose (Adults) Side Reactions Other Comments
Pyridostigmine bromide (Mestinon tablet) 60 mg/tablet 60 mg Onset: 30 minutes
Peak: 1–2 hours
Duration: 3–5 hours
60 mg tid in daytime
Up to 1 g a day
Most common side effect: GI symptoms
Pyridostigmine bromide TS (extended-release tablet; Mestinon TS) 180 mg/tablet 180 mg Onset: 30–60 minutes

Duration: 6–10 hours

180 mg at night
Up to 360 mg/day
Less than pyridostigmine tablet Absorption is erratic
Pyridostigmine bromide syrup (liquid; Mestinon syrup) 60 mg/5 mL 5 mL Onset: 30 minutes
Peak: 1–2 hours
Duration: 3–5 hours
60 mg tid in daytime
Up to 1 g a day
Same as pyridostigmine tablet Patient who has swallowing difficulty
Pyridostigmine bromide IV injection (Regonol) IV 5 mg/mL (2 and 5 mL ampule) 2 mg Onset: 2–5 minutes
Duration: 3–5 hours
1–2 mg q 2–3 hours
Administer slowly
Cardiac dysrhythmia During MG crisis
Neostigmine bromide (Prostigmine tablet) 15 mg/tablet 15 mg Onset: 30 minutes
Duration: 2–3 hours
15 mg qid in daytime
Up to 150 mg/day
More GI side effects than pyridostigmine Patients allergic to pyridostigmine
Neostigmine methyl sulfate (Prostigmine solution)
SC, IM
0.5 mg/mL
1 mg/mL
2.5 mg/mL
1 mg Onset: 20–30 minutes
Duration: 2–3 hours
0.5–1 mg q 3 hours
Up to 10 mg/day
Neostigmine methylsulfate (ProstgmineÆ solution) IV 0.5 mg/mL
1 mg/mL
2.5 mg/mL
0.5 mg Onset: 4–8 minutes
Duration: 2–3 hours
0.5 mg administered slowly every hour
Up to 10 mg/day
Ambenonium chloride (Mytelase) 10 mg tablet
25 mg tablet
10 mg Duration: 6–8 hours 10 mg bid
Up to 100 mg/day
Fewer GI side effects than pyridostigmine Headache Patients allergic to pyridostigmine
Edrophonium chloride (Tensilon) 10 mg/ml Onset: immediately
Duration: 2–5 minutes
2–10 mg IV/IM Bradycardia
Bronchospasm
Increased weakness
Used for diagnostic test
GI , Gastrointestinal; IM , intramuscular; IV , intravenous; MG , myasthenia gravis; qid , four times a day; SC , subcutaneous; tid , three times a day; TS , time span.

The adverse effects of AChEI are caused by the increased concentration of ACh at both nicotinic and muscarinic synapses. The common muscarinic effects are gut hypermotility (diarrhea and stomach cramps), increased sweating, salivation, and, when severe, bradycardia. The main nicotinic adverse effects are fasciculations and, sometimes, muscle cramps. Muscarinic effects are relieved by oral atropine (usual dose 0.4 mg bid or tid), glycopyrrolate (Robinul; usual dose, 1 mg or 2 mg bid or tid), or 5 mg diphenoxylate with atropine (Lomotil), which have no effects on the neuromuscular junction. Glycopyrrolate is better for this purpose because it does not enter the brain and so will not cause blurring of vision or toxic delirium expected with a high dose of atropine. However, it does not dry up secretions as much as atropine. Lomotil can be obtained without any prescription. It is important to tell patients in advance about the possible side reactions of AChEIs and relieve side reactions with glycopyrrolate or atropine so that they would not be alarmed about them. This is because this is the most common cause for MG patients not taking AChEIs.

The most serious side reaction of AChEIs is cholinergic weakness and crisis. This is due to too much AChEI medications, which lead to overaccumulation of Ach at the neuromuscular junction, producing sustained endplate depolarization without ample time for repolarization (depolarization block). This was a common phenomenon before immunotherapy in MG and was one of the well-known causes for MG crisis. However, this is extremely rare these days because we do not use a higher dose of AChEIs anymore, but it has been observed in MuSK-MG patients at the usual dose of AChEIs for MG ( ).

Although this drug has been extensively used since 1935 when Walker used Prostigmine as the first miracle drug for MG, there were no placebo-controlled randomized studies for these drugs ( ; ). However, case series demonstrate an objective and marked clinical effect (class IV evidence). Among the various AChEIs, Mestinon (pyridostigmine) is the most commonly used drug because of the 6 hours duration of the drug effect. The starting dose is 60 mg three to four times a day, depending on symptoms. This is usually given in daytime and at night; pyridostgmine time span (TS) is the preferred agent because of its slow-releasing benefit. Pyridostigmine syrup is used for children or for those who experience difficulty swallowing pills. Parenteral use of AChEIs is rarely necessary during the postoperative period or during MG crisis by some. IV pyridostigmine should be given at about 1/30 of the oral dose. In my practice, I have not used the intramuscular or IV forms of AChEIs in recent years. Neostigmine (Prostigmine) is an alternative choice of AChEIs in patients who are allergic to pyridostigmine. It has a slightly shorter duration of action and slightly greater muscarinic effects. Ambenonium (Myetelase) has been suggested as the last resort for patients who cannot take pyridostigmine or Prostigmine for whatever reason.

The optimal dose is determined by the balance between clinical improvement and adverse effects and can vary over time with concomitant treatment. The adjustment of dose should be gradual and achieved usually by the patient. The patient’s report of definite improvement following AChEI intake or the physician’s assessment of the patient’s condition at its peak of effectiveness is a reliable index of appropriate dose and effectiveness. The latter is almost impossible for nonhospitalized patients. Thus, the patients should be taught to adjust the dose. The maximal daily dose of pyridostigmine is 1 g a day. If the patients need more than 720 mg (12 tablets) of pyridostigmine for control of MG symptoms, we recommend immunotherapy. Patients with mild disease can often be managed adequately with AChEIs alone. In patients with immunotherapy, patients’ symptoms can be controlled with AChEIs.

Another symptomatic agent is ephedrine. This can be used for relief of fatigue as an adjunctive symptomatic treatment at dose of 25 mg of ephedrine sulfate. This drug is banned by the Food and Drug Administration (FDA) in the United States for safety reasons.

Potassium Channel Blocker: 3,4-Diaminopyridine

3,4-Diaminopyridine (DAP), a recently FDA-approved potassium channel blocker, was tested in MG in two studies: two MG patients by Lundh et al., with clinical and electrophysiological improvement with IV and oral 3,4-DAP, and 3,4-DAP in eight patients with MG by Sanders et al., with an improvement of >3 quantitative MG (QMG) score in two patients on a 15–45-mg daily dose ( ).

Immunotherapy

Since MG is an autoimmune disease, it is natural to treat this disorder with various immunotherapies. These include immunosuppressive therapy, immunomodulating therapy, and thymectomy.

Immunosuppressive Therapy

Steroids

In a large nonrandomized study, remission or marked improvement was reported in 63%–82% of GMG and in 66%–86% of OMG patients treated with oral corticosteroids, usually prednisone (class IV evidence) ( ) ( Table 19.2 ).

Table 19.2
Literature Review of Nonrandomized Studies of Various Immunosuppressants in Myasthenia Gravis
References: ; ; ; , ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; .
Study Number of Patients Patients with Good Overall Improvement Other Information
Improvement Remission
Steroid
9 9 Multiple short course of methylprednisone. Severe MG
15 12 IV MP followed by oral prednisone
116 61 (52.6%) marked 32 (27.6)
60 18 (30.0%) marked 25 (41.7%)
142 42 (29.6%) marked 48 (33.8%)
104 44 (42.3%) marked 41 (39.4%)
600 95% 151 generalized; 449 pure ocular
32 21 (66%) Ocular MG
45 19 (86) Ocular MG
Azathioprine (AZA)
26 (78%) Unresponsive to steroid or ACTH
78 91% Alone, in combination with steroid, thymectomy or both
99 72% (38% marked) Alone, in combination with steroid, thymectomy or both
18 15 (83%) Alone for more than 6 months
41 41 (100%) Alone or with prednisone for more than 3 years
32 75% Alone
57 70% With prednisone
50% 2-year class 2 study. Early high dose with prednisone vs. 16% in low dose control
Cyclophosphamide (CPP)
47 47 (100%) With steroid in 33; thymectomy in 5; at 3 years
Cyclosporin
10 8 (80%) marked 12-month open-label trial. Severe MG, unresponsive to AChEI or thymectomy plus steroid or AZA
9 7 (78%) marked 2-year open-label trial. Unresponsive to thymectomy, AZA, and steroid
52 44 (85%) marked 30-month open-label trial. Severe MG unresponsive to thymectomy, steroid, and AZA
Mycophenolate mofetil
22 15 (68%) 2–18-month open-label trial. 3/5 alone; 4/7 unresponsive to AZA; 7/10 with steroid
12 8 (67%) marked 6-month open-label trial. Unresponsive to AZA, steroids, CsA for <6 months
32 19 (59%) Alone, 4; 28 on other immunosuppresants
85 73% 48 thymectomy; 66 with CsA, steroid, AZA, MTX
Tacrolimus
19 7 (37%) 16-week open-label study: thymectomy and steroid
12 8 (67%) 2-year treatment
17 12 (71%) Open-label trial. Mean follow-up of 19.2 months
79 69 (87%) Pharmacologic Open-label trial. Severe MG. Thymectomy, steroid, and CSA. Mean follow-up of 2.5 years
Etanercept
11 6 (55%) Open-label trial
AChEI , Anticholinesterase inhibitor; ACTH , adrenocorticotrophic hormone; CsA , cyclosporine A; IV , intravenous; MG , myasthenia gravis; MP , methylprednisone; MTX , methotrexate.

The efficacy has been established in double-blind, placebo-controlled trials with methylprednisolone but not with prednisone. A randomized trial compared double-blind IV methylprednisolone (10 patients) versus placebo (9 patients) for GMG ( Table 19.3 ). The steroid group showed a 7.2 times greater improvement in muscle strength than the placebo group did after 2 weeks of treatment, indicating a significant short-term benefit from steroid. A randomized, double-blind trial of prednisone (six patients) versus placebo (seven patients) for GMG showed no significant difference in improvement at 6 months. This study lacks credence because of the small number of patients for each group. In a randomized controlled trial with methylprednisolone vs. IV immunoglobulin (IVIg), no significant difference was noted at day 14, indicating that methylprednisolone is as good as IVIg. Thus, steroids are useful as short-term immunosuppressants (class II). Long-term use of steroids is associated with many adverse effects, including weight gain, fluid retention, diabetes, psychosis, glaucoma, cataracts, gastrointestinal (GI) bleeding, and osteoporosis. Steroids should be used ideally as short-term (60 mg/day for 3–4 months) immunosuppressants. However, long-term use of a steroid is not unusual. The risk of osteoporosis is reduced by giving bisphosphonates (class IV).

Table 19.3
Literature Review of Randomized Studies of Various Immunotherapies for Myasthenia Gravis
References: ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; .
Study Agent Method(s) Intervention(s) Outcome Notes
Steroids
ACTH ( n = 43) RCT, PC ACTH (580 units) 8 days vs. placebo No evidence of efficacy of ACTH Ocular MG
Prednisone ( n = 13) RCT, DB, PC Prednisone 100 mg ( n = 6) on alternate day vs. placebo ( n = 7) No significant difference
In 6 months, 3 improved on placebo and 3 on prednisone
Small number for statistical analysis
Methylprednisone (MP) RCT, DB, PC 2 g MP IV for 2 days ( n = 10) vs. placebo ( n = 10) Significant improvement in IV
MP at 2 weeks
Improved in 8 of 10 in MP and in 1 of 9 in placebo
Mean duration of improvement: 8 weeks
Prednisone ( n = 10) RCT Prednisone 60 mg daily ( n = 5) vs. AZA 2.5 mg/kg daily ( n = 5) No difference in improvement
Improvement in 4 of 7 in prednisone and in 1 of 3 in AZA
Small number for statistical analysis
Schuchardt (2004) MP ( n = 33) RCT MP 1–1.5 mg/kg for 14 days vs. 3 g IVIg daily for 5 days No difference between the two groups at day 14 MG with exacerbation
Immunosuppressants
Azathioprine (AZA) ( n = 41) RCT, unblinded AZA 3 mg/kg daily plus initial prednisone ( n = 21) vs. prednisone ( n = 20) 1 mg/kg daily No difference in remission or marked improvement between the two groups at 1, 2, and 3 years Prednisone treatment for months in AZA group
AZA ( n = 34) RCT, blind AZA 2.5 mg daily ( n = 15) vs. placebo ( n = 19)
All patients on prednisone 1.5 mg/kg or 100 mg on alternate days
No difference between the two groups at 12 months. Significant reduction of prednisone dose in AZA at 36 months (0 in AZA vs. 40 mg in placebo) Prednisone-sparing effect
Cyclophosphamide (CPP) ( n = 23) RCT, DB CPP 0.5 g/m 2 of body surface IV monthly ( n = 12) vs. placebo ( n = 11)
All patients on prednisone more than 50 mg initially
No significant difference between the two groups at 12 months IV every other month after the first 6 months
Cyclosporine A (CsA) ( n = 20) RCT, DB, PC Cyclosporine (6 mg/kg daily) ( n = 10) vs. placebo ( n = 10) Significant greater increase in QMG score in CsA at 6 and 12 months
Cyclosporine ( n = 39) RCT, DB Cyclosporine (5 mg/kg daily) plus prednisone 60–100 mg on alternate days ( n = 20) vs. prednisone plus placebo ( n = 19) Significant greater increase in QMG score in CsA plus prednisone at 6 months No steroid-sparing effect
Tacrolimus ( n = 34) RCT, unblinded Tacrolimus 3 mg/day plus steroids ( n = 18) vs. no tacrolimus plus steroid ( n = 16)
Plasmapheresis is given if necessary to either group
Significantly less number of treatment with plasmapheresis plus high-dose IV MP in tacrolimus during early and follow-up phase treatment at 1 year
The period of early-phase treatment is significantly shorter in tacrolimus
The oral prednisone dose is significantly lower in tacrolimus
Reduction in need for other immunotherapy, to include steroid-sparing effect
Mycophonolate mofetil (MM) ( n = 14) RCT, DB MM ( n = 7) vs. placebo ( n = 7). Cyclosporine, prednisone, or no immunosuppresants in either group No difference between the two groups
Improvement in 1 in each group
MM ( n = 80) RCT, DB MM (2.5 g daily) ( n = 41) vs. placebo ( n = 39)
All on prednisone 20 mg/day
No difference in QMG score change at 3 months
MM ( n = 176) RCT, DB MM (2 g/day) ( n = 88) vs. placebo ( n = 88)
All on prednisone 20 mg/day for 1 month
No difference in QMG score change at 9 months
Immunomodulating therapies
Plasma exchange (PE) ( n = 14) RCT Prednisone ( n = 7) vs. prednisone plus PE ( n = 7)
PE: Three PEs over a 10-day period with PE once a week if necessary
All on prednisone 1 mg/kg/day for 1 month
No difference in muscle strength score change at 1 month or 1 year
Significantly less relapse in prednisone: 1 in prednisone vs. 8 in PE in 1 year
4 in each group in MG crisis
IVIg ( n = 87) RCT 3 PEs vs. IVIg 2 g/kg or IVIg 1.2 g/kg No difference in MM score change at day 15 MG exacerbation
IVIg ( n = 173) RCT IVIg 1 g/kg vs. IVIg 2 g/kg No difference in MM score change at day 15 MG exacerbation
IVIg ( n = 12) RCT, crossover IVIg 0.4 g/kg for 3 days plus 5 PE vs. opposite schedule No difference in QMG change 7 days after treatment
IVIg ( n = 12) RCT IVIg 1 g/kg for 2 days vs. 5% albumin for 3 days No difference in QMG change at day 42
IVIg ( n = 51) RCT, DB, PC IVIg 1 g/kg for 2 days vs. 5% dextrose for 2 days Significance in QMG change and postintervention status in IVIg at day 14 Treatment effect maintained for 28 days after infusion
ACTH , Adrenocorticotrophic hormone; DB , double blind; IV , intravenous; IVIg , intravenous immunoglobulin; MG , myasthenia gravis; PC , placebo controlled; QMG , quantitative myasthenia gravis; RCT , randomized controlled trial.

Oral prednisone is recommended as the first drug of choice when immunosuppressive drugs are necessary in MG (class IV). This is because steroid is known to induce the earliest onset of improvement among the various immunosuppresants. Controversy exists regarding the most prudent method of initiating therapy (high-dose daily, high-dose alternate day [AD] or low-dose AD, with a gradually increasing regimen). Low doses (20–30 mg) are recommended by those who believe that some patients develop steroid worsening, including MG crisis, in 4 to 10 days on higher doses (class IV) ( Table 19.4 ). The low dose is gradually increased to a high dose over a period of a few weeks. High doses (>60 mg/day) are recommended by others, including this author, who believe that improvement is more rapid with high-dose daily regimens and steroid worsening represents natural worsening of MG because we start steroid in patients with unstable MG. I personally believe that steroid worsening is exaggerated and extremely rare these days since we use a smaller daily dose of AChEIs. Thus, I recommend an initial high-dose regimen unless there is any compelling reason not to do so. Once the maximal benefit or pharmacological remission is achieved, owing to the potential adverse effects with prolonged use of steroids, the dose should be slowly reduced to the minimum effective maintenance dose or withdrawal and the simultaneous use of a long-term immunosuppressant are initiated.

Table 19.4
Pharmacological Properties of Various Immunotherapies
Drug Mechanism of Action Typical Dose Onset and Duration of Action Contraindication
Steroid Inhibit the activation and antigen processing of T cells. Decrease of the number of circulating T cells
Increase muscle AChR synthesis
Initial: 60 mg/day
Maximum: 100 mg/day
Onset: 2–3 days
Peak: 3–6 months
Severe diabetes
Azathioprine (AZA) Purine antagonist
Inhibit T-cell and B-cell proliferation
Initial: 150 mg/day
Maximum: 400 mg/day
Onset: 4–10 months
Peak: 2–3 years
Pregnancy, TPMT
Deficiency, liver failure, cancer
Mycophenolate mofetil (MM) Purine antagonist
Inhibit T-cell and B-cell proliferation
Initial: 1.5 gm/day
Maximum: 4 gm/day
Onset: 4 weeks–10 months (mean, 11 weeks)
Peak: 8–26 months (mean, 7 months)
Pregnancy
Cyclosproine (CsA) Calcineurin inhibitors
Decease IL-2 production in T-cells
Initial: 300 mg/day Onset: 1 week–8 months Severe hypertension, renal failure
Tacrolimus Calcineurin inhibitors
Prevents IL-2 transcription in T cells
Initial: 3 mg/day Onset: 2 weeks–4 months
Peak: 4 months–2 years
Pregnancy, renal failure, severe diabetes
Cyclosphosphamide Alkylating agent
Nonspecific cell-cycle inhibitor, more effect on B cells
Initial: 150 mg/day Onset: 2 months
Peak: 3–12 months
Pregnancy, cancer,
Impaired bone marrow function
Rituximab Chimeric human-murine monoclonal CD20 antibody
Reduce B cell activation and proliferation
Initial: 375 mg/m 2 q week x 4 weeks. Onset: 1–2 months
Peak: 7 months
Cardiac disease
Plasma exchange (PE) Removal of AChR antibody 5 PE session qod
2–3 L removal each session
Onset 1 day
Peak: 2 weeks
Duration: 1–3 months
CI: coronary artery disease. Coagulopathy
Intravenous immunoglobulin (IVIg) Neutralize the blocking effects of MG IgG antibodies to AChR 2 gm/kg a day over 2 or 5 days Onset: 5 days

Duration: 1 m

Renal failure
IgA deficiency
Eculizumab Inhibits the formation of C5b-9 membrane attack complex 900 mg/week for 4 weeks
1200 mg/week every 2 weeks after that
Peak: 12 weeks Meningococcal infection: meningococcal vaccination 2 weeks before the first injection
Thymectomy Removal of thymic cells Onset: 3–4 minutes
Peak: 9–12 minutes
AChR , Acetylcholine receptor; AZA , azathioprine; CsA , cyclosporine A; IgG , immunoglobulin A; IL-2 , interleukin 2; IVIg , intravenous immunoglobulin; MG , myasthenia gravis; MM , mycophenolate mofetil; TPMT , thiopurine methyltransferase.

Azathioprine

Azathioprine (AZA) is extensively used as immunosuppressive. The onset of therapeutic response may be delayed for 4–10 months, and maximal effect is obtained in 1–2 years. AZA is usually well tolerated, but idiosyncratic flu-like symptoms or GI disturbances, including pancreatitis, occur in 10%, usually within the first few days of treatment. Side effects include bone marrow suppression and hepatotoxicity, which usually respond to drug reduction or withdrawal. When the white blood cell falls below 4000 mm 3 , the dose should be reduced; if it is below 3000 mm 3 , AZA should be discontinued. The incidence of malignancy was not increased over 22 years of AZA treatment in study.

Several observational series of AZA treatments showed 70%–100% improvement with AZA alone or in combination with other treatments, mostly with steroids (class IV) ( ). One study comparing early high-dose immunosuppressive with AZA and prednisone resulted in 50% of patients achieving remission after 2 years, compared with a remission rate of only 16% in those on a low-dose regimen.

One large double-blind randomized study ( ) has demonstrated the efficacy of AZA as a steroid-sparing agent with a better outcome in patients on a combination of AZA and steroids than in patients treated with steroids alone (class I). In a randomized, unblinded trial of AZA plus initial prednisone versus prednisone alone, no difference was found in remission or marked improvement between two groups in 1–3 years of long-term treatment.

Because AZA has been associated with relatively few treatment failures and steroid-sparing effect in two randomized controlled trials (class I), we do recommend AZA as the first-choice long-term immunosuppressant drug. Some prefer to start AZA together with steroids to allow tapering of steroids to the lowest dose possible. The usual recommended daily dose is 150 mg/day.

Cyclosphosphamide

In one observational study in 42 patients who were given cyclophosphamide (CPP) with a duration of treatment from 2 to 37 months, remission was achieved in 58% of patients who underwent 1 year of treatment, 85% of patients who underwent 2 years of treatment, and 100% of patients who underwent 32 months of treatment ( ). In a randomized, double-blind trial of CPP plus prednisone versus prednisone plus placebo in GMG, CPP significantly improved muscle strength at 12 months, but not at 6 months ( ). The CPP-treated patients were on significantly lower doses of prednisolone at 6 and 12 months. Thus, this study showed that CPP is clinically effective in GMG and that it has a steroid-sparing effect (class II). The usual oral dose of CPP has been 5 mg/kg/day (usual initial dose, 150 mg/day). CPP can be given as monthly pulse therapy at an IV dose of 1 gm/m 2 /month. High-dose CPP (50 mg/kg/day for 4 days) followed by granulocyte colony stimulating factor was also used for “rebooting” of the immune system in refractory MG. In three MG patients refractory to plasma exchange (PE), thymectomy, and conventional immunosuppressants, remission was achieved ( ). In view of the relative high risk of toxicity including bone marrow suppression, opportunistic infections, bladder toxicity, sterility, and neoplasm, this drug is recommended as the last choice of immunosuppressants to patients who are intolerant of or unresponsive to other immunosuppressants. The usual recommended oral daily dose is 150 mg/day.

Cyclosporine A

This is the first agent in MG that showed a definite benefit in a randomized controlled trial. Three observational studies on severe MG unresponsiveness to thymectomy, AZA, or steroid showed marked improvement in 75%–80% of patients in 1–3 years of treatment. One placebo-controlled, double-blind randomized study in 20 patients for 6 months with an open extension (class I) showed significantly improved strength and reduction in AChR antibody titers compared with the placebo group at 6 and 12 months. Another double-blind randomized controlled study with 39 patients showed significantly improved muscle strength at 6 months in the cyclosporine (CsA) plus prednisone group compared with the placebo plus prednisone group, but a steroid-sparing effect was not observed. Thus, two randomized controlled trials have shown that CsA is clinically effective in GMG. Initial daily dose is 4–6 mg/kg/day. However, because of significant side effects such as nephrotoxicity, hypertension, and malignancy, it should be considered only in patients intolerant of or unresponsive to AZA. Other common side effects include hypertrichosis, gingival hyperplasia, myalgia, and “flu-like” symptoms. Nonsteroidal anti-inflammatory drugs should be avoided with CsA because they increase nephrotoxicity. CsA should be considered in patients intolerant of or unresponsive to AZA (level B recommendation). The usual recommended daily dose is 200 mg/day.

Mycophenolate Mofetil

Two open-label trials and two observational studies in MG patients unresponsive to steroid, AZA, or CsA showed 59%–73% improvement over a treatment period of 2–18 months. Improvement occurred in monotherapy as well as with other immunosuppressants. Two randomized, double-blind trials of mycophenolate mofetil (MM) plus steroid versus steroid plus placebo for GMG failed to demonstrate any efficacy of MM in trials of 3 or 9 months’ duration. Another study of a small randomized, double-blind trial of MM plus either CsA or prednisolone or no immunosuppresants versus placebo plus either CsA or prednisolone or no immunosuppressants also did not show any benefit of MM. Although MM has been reported to be not efficacious in randomized controlled trials in GMG, this drug is well tolerated, with a relatively good adverse effect profile ( ),

The adverse effects of MM were usually mild (nausea, headache, or diarrhea). Serious adverse effects such as infections and hematopoietic suppression are rare. One advantage of MM, compared with other immunosuppressants, is a lesser carcinogenic tendency: a kidney transplant patient on MM had a lesser cancer incidence in the long-term follow-up. Lymphoma was reported as a rare complication. The European Task Force recommends MM as the second-line long-term therapy in MG patients intolerant of or unresponsive to AZA (level B recommendation). This view might have to be modified in view of the negative results of two randomized controlled studies. The usual recommended daily dose is 1.5 gm.

Tacrolimus

Although tacrolimus and CsA act in a similar fashion, the potency of tacrolimus appears to be 10–100 times greater than that of CsA. Tacrolimus at 3 mg/day for MG is 20% of the dose given to organ transplant patients. One 4-month open-label study showed an improvement in 37% of cases. Three observational studies with longer than 19 months’ duration showed an improvement in 67%–71% of patients and pharmacological remission in 87% of patients. A randomized, unblinded, non-placebo-controlled trial of tacrolimus plus steroids with or without PE versus no tacrolimus plus steroids with or without PE showed significantly fewer treatments with PE plus high-dose IV methyl-prednisone, a significantly shorter period of early-phase treatment, and significantly lower prednisone dose in tacrolimus-treated groups ( ). Thus, this study showed that tacrolimus reduces the need for other immunotherapy, indicating that this drug is effective in GMG. Tacrolimus seems to be relatively safe at the doses used in MG, so it should be recommended as third-line treatment for patients who are intolerant of or unresponsive to AZA. Common side effects of the drug include hypertension, renal insufficiency, hyperkalemia, headache, and hyperglycemia. If tacrolimus is used in patients with existing impaired glucose intolerance or diabetes mellitus, this impaired glucose tolerance will be almost always exacerbated ( ). The usual recommended daily dose is 3 mg.

Methotrexate

The European Task Force recommends methotrexate as second-line drug because it is well studied in other autoimmune disorders ( ). Two studies reported conflicting results ( ; ). On one hand, Heckmann et al. compared the effect of methotrexate (17.5 mg weekly) versus AZA (2.5 mg/kg daily) in a single-blind study. At 2 years, there was a substantial and comparable decrease in the average daily prednisone dose and in the QMG scores in both groups, suggesting both drugs as effective steroid-sparing agents ( ). On the other hand, in a randomized, double-blind, placebo-controlled trial of methotrexate 20 mg/week by mouth versus placebo in prednisone-dependent MG patients, showed no difference in the prednisone area under the curve between methotrexate and placebo over a 12-month period, suggesting no steroid-sparing effect of methotrexate. We recommend this drug as the last choice if all other immunosuppressants fail or the patient cannot take others. One advantage of this medication is the one-time weekly administration. Initial starting dose is 7.5 mg weekly and maximum weekly dose is 30 mg. The usual recommended weekly dose is 7.5 mg.

Immunomodulating Therapy

Plasma Exchange

PE or plasmapheresis removes antibodies from patient sera by membrane filtration or centrifugation. The patients typically undergo five sessions of PE at 20–50 mL/kg body weight per session over a span of 2 weeks. The plasma is separated from blood cells using membrane filtration or centrifugation, and then the blood cells are reinfused into another vein with diluted albumin, colloids, or crystalloids, while plasma is removed. The onset of improvement is within the first week and the effect lasts for 1–3 months. Side effects are associated with the insertion of the IV catheter and hypotension. Since PE has been extensively used as the treatment of choice in acute MG exacerbation, there is no randomized, double-blind control study for its short-term benefit ( ). There are, however, two nonrandomized controlled studies ( ; ). One study in 1979 compared six patients who received PE over 2 to 2.5 weeks and six patients who were assigned to a control group. None of the control group improved, while all treated patients improved strikingly ( ). Another study in 1979 compared the long-term effect in severe patients treated with PE plus immunosuppressive drugs and seven patients treated with immunosuppressants alone ( ). PE was associated with short-term improvement in all seven patients. At 6 to 12 months, clinical outcome and AChR antibody titer decline were similar in both groups. Many observational studies have shown short-term clinical improvement in 65%–100% of cases. A Cochrane research revealed only one eligible controlled trial ( ; ). The aim of this trial was to compare the long-term effect of prednisone (seven patients) versus prednisone plus PE (seven patients). No difference was found in muscle strength score change at 1 month or 1 year between the two groups. Significantly, there were fewer relapses in the prednisone group. Thus, this trial failed to demonstrate any long-term benefit of prednisone and PE versus prednisone alone ( ). There are several randomized studies comparing the efficacy of PE with IVIg showing comparable therapeutic efficacy ( ; ; ; ). On the basis of these findings, the 1986 National Institutes of Health consensus ( ) is still valid: PE can be useful in strengthening patients with MG before thymectomy and during the postoperative period, and it can also be valuable in lessening symptoms during the initiation of immunosuppressants and during an acute crisis (class IV). Therefore, sham controlled trials would be unethical. A Cochrane review (2008) concluded that there are no adequate randomized controlled trials to establish whether PE improves the long- or short-term outcome from MG, but many case series report short-term benefit from PE in MG, especially in myasthenic crisis (MC) ( ). Repeated PE is thus not recommended as a treatment to obtain continuous and lasting immunosuppression in MG.

Intravenous Immunoglobulin (IVIG)

IVIg is a fractionated blood product consisting primarily of immunoglobulin G (IgG), derived from 3000 to 10,000 human plasma donors. The mechanism of action of IVIg is complex, including almost all aspects of the immune system. The most relevant mechanisms for MG were the neutralizing effect of commercial polyclonal IgG (IVIg) on the blocking effects of myasthenic IgG antibodies to nicotinic AChR and the inhibiting effect on the production of autoantibodies by inhibiting the body’s ability to produce globulin. IVIg is relatively easier to administer than PE. The usual IVIg dose is 0.4–1 gm/kg body weight given over 2–5 days. After infusion, serum IVIg levels increase fivefold, then decline by 50% in 72 hours and return to pretreatment levels after 21–28 days. Severe reactions from IVIg are rare. The most serious reactions so far reported include anaphylactic reaction, hepatitis C outbreak, aseptic meningitis, acute renal failure, hemolysis, and cerebral infarction ( ). Anaphylactic reaction following IVIg has been reported in patients with selective IgA deficiency due to IgG or IgE antibodies to IgA; IgA deficiency occurs in about 1 of every 200 to 800 persons. Thus, IgA deficiency is the only contraindication for the use of IVIg, and the IgA level should be checked when IVIg therapy is considered. If IVIg has to be given in an IgA-deficient patient, it should be infused with IVIg with low IgA levels (e.g., Gamimmune and Gammagard). An outbreak of hepatitis C was last reported with IVIg in early 1994. Death from acute renal failure in diabetic patients has been reported. Aseptic meningitis has become the most common troublesome side reaction to clinicians, seen in about 11% of patients. Most of other reactions are mild and self-limited and are associated with too rapid infusion of IVIg.

In a review of seven nonrandomized observational studies with 10 participants or more, Jongen et al. reported an overall rate of improvement of 76% (90/119), with a median of 87% (range, 48% to 92%) ( ; ). There were two randomized studies on IVIg versus placebo ( ; ). In one randomized controlled trial comparing IVIg (1 g/kg for 2 days) with placebo (5% dextrose for 2 days) in patients with MG worsening, there was a significant improvement in QMG in favor of the IVIg group on day 14 (class I) ( ). The treatment effect was maintained for 28 days after infusion in this study. Importantly, this effect was not observed in mild MG but was significant in moderate to severe MG. In another study comparing IVIg for 2 days versus 5% albumin for 3 days, there was not any significant difference in QMG change between the two groups ( ).

In a comparison of IVIg versus PE, four randomized controlled trials showed no difference in improvement between PE and IVIg ( ; ; ; ), suggesting that the efficacy of IVIg is as good as that of PE. Two randomized controlled trials compared the efficacy of two IVIg doses, 1 and 2 gm/kg, and found no difference in improvement between PE and IVIg and between the 1 and 2 gm/kg IVIg regimens.

IVIg was proposed as a maintenance therapy for MG. IVIg 2 gm/kg over 5 days was followed by 0.4 gm/kg once monthly or once every 6 weeks ( ; ; ). One study was conducted in 16 patients with severe GMG ( ), and another, in 11 patients with severe MG with bulbar palsy ( ). All patients were unresponsive to steroid and immunosuppresants. All patients improved after a mean period of 20 months. One study compared PE and IVIg as maintenance therapies for 27 juvenile MG patients and concluded that there is class III evidence that PE and IVIg both have high response rates as maintenance therapy ( ).

A Cochrane review concluded that IVIg is effective in severe MG exacerbation in one randomized controlled trial, but there is insufficient evidence from randomized trials to determine whether IVIg is efficacious in chronic MG ( ).

On the basis of these findings, we recommend IVIg to be used in lieu of PE for acutely worsening MG (class I) and preparation of weak patients for surgery including thymectomy (class IV) and to be used as an adjuvant to immunosuppressive therapy to minimize the long-term side effects of oral immunosuppressants and maintain an improved status of MG ( ). In MuSK-MG, a retrospective multicenter study on 52 patients showed a poorer outcome with IVIg compared with PE (class IV) ( ).

Eculizumab

Complement has been known to play an important role in the pathogenesis of MG, leading to the hypothesis that complement inhibition may lead to clinical improvement in MG ( ). Eculizumab, a complement inhibitor, is a C5 monoclonal antibody that inhibits the formation of C5b-9 membrane attack complex. Eculizumab is the first approved drug for MG by the FDA for treatment for adult patients with AChR-positive GMG after successful trials in 14 patients with severe refractory GMG (class I in Table 19.5 ) ( ). Six of seven (86%) patients treated with eculizumab for 16 weeks achieved the primary endpoint of a 3-point reduction in the QMG score ( P = 0.014 compared with placebo). In a subsequent phase 3 study in 125 patients, 62 receiving eculizumab and 63 receiving placebo for 26 weeks, the change in the QMG score showed a benefit with eculizumab compared with placebo ( P = 0.02) at week 26, but not in MG-ADL (activity of daily living) score. Most of the treatment effects occurred by week 12 and were sustained to week 26. The most common side reactions were headache and upper respiratory infection ( ). Thus, this drug is recommended for patients with moderate/severe GMG who are refractory to adequate trials with existing immunotherapy. The drug is given via IV infusion with a recommended dosage regimen of 900 mg/week for the first 4 weeks, 1200 mg for the fifth week, and 1200 mg every 2 weeks thereafter. Meningococcal vaccinations should be given 2 weeks before starting the first therapy to prevent any possible meningococcal infection.

Table 19.5
Evidence-Based Class and Recommendations of Various Immunotherapies
Drug Evidence Class a Recommendations
Anticholinesterase inhibitors (AChEIs) Class IV Symptomatic treatment.
Steroids Class II First-line immunotherapy. Mainly for short-term use. For long-term use if the dose can be reduced to a dose at which side reactions are negligible.
Azathioprine (AZA) Class I First-line long-term immunotherapy. Can be used to spare the steroid dose or for patients unresponsive to or intolerant of steroids.
Cyclosporin (CsA) Class I Second-line long-term immunotherapy. For patients intolerant of or unresponsive to steroids or AZA.
Cyclophosphamide (CPP) Class II Last-choice long-term immunotherapy. For patients unresponsive to or intolerant of steroids, AZA, CsA, or tacrolimus.
Mycophenolate mofetil Class IV Adjunct long-term immunotherapy. This can be used as an additional agent to any long-term immunosuppressant.
Tacrolimus Class I Third-line long-term immunotherapy. For patients unresponsive to or intolerant of steroid, AZA, or CsA.
Plasma exchange (PE) Class III Indicated for severe MG, acute exacerbation, and MG crisis. For patients unresponsive to or intolerant of IVIg.
IVIg Class I Indicated for severe MG and acute exacerbation and used as adjunct to immunotherapy to maintain an improved status. For patients unresponsive to or intolerant of PE.
Thymectomy Class I Any early-onset generalized MG patient who needs immunotherapy is candidate.
Eculizumab Class I Moderate/severe generalized MG patients who are refractory to adequate trials with existing immunotherapy.
IVIg , Intravenous immunoglobulin; MG , myasthenia gravis.

a Evidence class I, randomized controlled trials available; class II, controlled trials without randomization or randomized trial with small patient number; class III, uncontrolled trials; class IV, case series.

Zilucoplan

Zilucoplan is another complement inhibitor that binds to C5b with high affinity and specificity, thus preventing the cleavage of C5 into C5a and C5b complement component and also blocking binding of C5b to C6 complement component. Unlike eculizumab, this agent is administered subcutaneously by the patient daily. A phase 2 randomized, double-blind, placebo-controlled multicenter trial was conducted in 44 AChR-Ab-positive Myasthenia Gravis Foundation of America (MGFA) II–IV GMG patients using three treatment groups (placebo, 0.3 mg/kg SC, and 0.1 mg/kg SC) over a 12-week period ( ). All participants were vaccinated against Neisseria meningitidis . Clinically meaningful and statistically significant improvements ( P < 0.05) in primary and key secondary efficacy end points were achieved in primary (QMG and MG-ADL scores) and secondary (MG Composite and MG Quality of Life Score) end points in the 0.3-mg/kg SC treated group compared with the placebo group. Outcomes for the 0.1-mg/kg SC daily dose were also statistically significant but were slower in onset and less pronounced than with the 0.3-mg/kg dose. The onset of action was rapid, with separation of the 0.3-mg/kg arm from the placebo arm beginning after 1 week. Twenty-eight percent of participants did not experience an improvement as per the minimal clinically important difference of 3 points on the QMG score. The drug was well tolerated, with mild adverse events and a low incidence of local injection site reaction. Zilucoplan may have an advantage over ecluzumab in view of the easy subcutaneous injection route. As of March 1, 2020, this drug has not been approved for MG by the FDA.

Rituximab

Rituximab is a chimeric mouse/human anti-CD20 monoclonal immunoglobulin. According to a 2017 review, 169 MG (59% AChR-Ab positive; 34% MuSK-Ab positive) patients were treated with rituximab ( ). Rituximab treatment achieved minimal manifestation (LMm) or better status, according to the modified MGFA postintervention scale, in 44% of treated patients and combined pharmacologic and chronic stable status in 27%. MM or better status was achieved in 72% of MuSK MG and 30% of AChR MG patients ( P <0.001). Response predictors were MuSK MG, less severe disease, and younger age at treatment. A multicenter blinded review study showed that patients treated with rituximab met the end goal of an MG Status and Treatment Intensity score of 2 higher, which was statistically significant compared with the control group: 58% (14/24) of rituximab-treated patients versus 16% (5/21) of control patients (class IV) ( ). These two studies suggest that rituximab may be more effective in MuSK MG. However, this drug needs to be evaluated more extensively against the current immunosuppressants before it can be recommended. Common infusion-related side effects of rituximab are fever, chills, nausea, vomiting, flushing, and bronchospasm. Other, more serious side effects include neutropenia and increased risk of infection.

Thymectomy

In 1939, Blalock et al. reported the remission of GMG in a 21-year-old woman after removal of a cystic thymic tumor and subsequently performed thymectomy on MG patients without thymoma, found hyperplasia in the thymus glands, and reported improvement in at least half of their patients ( ). Since this report, thymectomy has been accepted widely as a standard form of treatment for MG on the basis of class II evidence ( ). In 2016, the first randomized trial comparing thymectomy with medical management in nonthymomatous patients was published ( ). A total of 126 recently diagnosed patients, ages 18 through 65 years, with AChR antibody-positive GMG with duration of 3 to 5 years were randomized to receive either extended transsternal thymectomy plus prednisone versus medical management with prednisone. Over a 3-year follow-up period, the time-weighted average QMG score was lower in the patients who underwent thymectomy (6.15 vs. 8.99; P < 0.001), and alternate-day prednisone dose requirement was lower (32 vs. 54 mg; P < 0.001). In the thymectomy group, fewer patients required immunosuppression with AZA or were hospitalized for exacerbation ( P < 0.001). The effects of thymectomy began to be observed as early as 3 to 4 months but became definite at 9–12 months and were maintained for the entire 3-year study. The results of this study provides class I evidence for thymectomy in all generalized AChR-positive patients to improve MG status and decrease the required dose of immunotherapy in GMG. A recent analysis on 50 patients (24 prednisone alone and 26 prednisone and thymectomy) showed that at 5 years, thymectomy plus prednisone continues to confer benefits in patients with generalized nonthymomatous GMG compared with prednisone alone ( ). Current evidence does not support an indication for thymectomy in patients with MuSK, LRP4, and agrin antibodies ( ; ).

At this time, the indication for thymectomy in AChR-negative GMG patients is controversial. Two retrospective cohort studies showed comparable responses among AChR-positive and AChR-negative groups ( ; ; ).

Age is another controversial subject in thymectomy ( ; ). Thymectomy is often avoided in children because of the theoretical possibility of impairing the developing immune system. However, reports of thymectomy in children as young as 2 to 3 years of age have shown favorable results, without adverse effects on the immune system ( ). Thymectomy has been largely discouraged in late-onset MG (>65 years of age) mainly because of frequent observations of an atrophic, involuted thymus. One series showed a less favorable outcome of thymectomy in late-onset MG: postthymectomy improvement was transitory and no longer detectable at 2 years after thymectomy (class IV) ( ). The age at MG onset is a more important factor than the age at thymectomy( ). Considering the benefit of thymectomy up to 65 years of age in the randomized study, it is reasonable to extend thymectomy age to 65 years ( ).

A randomized thymectomy trial mandated an extended transdermal thymectomy ( ). Less invasive procedures are now available for thymectomy: transcervical, video-assisted thoracoscopic surgery (VATET), and robotic video-assisted thoracoscopic surgery. VATET offers shorter hospital duration and stay and limited morbidity.

In comparing the various surgical approaches, employed Kaplan-Meier life table analysis at 5 years and concluded that the combined transcervical-transsternal, extended transsternal, extended cervical, and VATET procedures appear to produce more or less the same percentage (40%–55%) of remission at 5 years. It appears that the more thorough the removal of all tissue that may contain thymus, the better the longer-term results. Therefore, conceptually, the combined transcervical-transsternal thymectomy best fulfills these criteria.

Sonett and Jaretzki also stated that it is far preferable to leave behind small amounts of suspected thymus, or even likely thymus, than injuring the recurrent laryngeal, the left vagus, or the phrenic nerve. Injuries to these nerves can be devastating to a patient with MG ( ).

On the basis of this information, we believe that thymectomy is indicated in any early-onset (<65 years of age) GMG patients whose symptoms are not satisfactorily controlled with AChEIs alone and initial immunotherapy (class I recommendation for AChR-Ab-positive patients and level B recommendation for AChR-Ab-negative patients. We take this view on the basis of the findings that AChR antibodies were found in 66% of seronegative sera by a clustered AChR cell–based assay) ( ; , on the other hand, listed thymectomy as the first-line treatment of MG in their recommendation. Thymectomy induces long-term improvement and remission. About 33% of patients will achieve remission in the first 3 years after thymectomy, and 50% within 5 years ( ). Thymectomy produces a stable remission by eliminating thymic centers. The effects are delayed because of the long life-span of the existing pool of small immunocompetent lymphocytes.

Myasthenic Crisis

Myasthenic crisis (MC) is a life-threatening complication of MG. Traditionally, it is defined as respiratory failure due to worsening MG requiring intubation and mechanical ventilation (MGFA class V). Here, we define MC as respiratory failure due to worsening MG requiring mechanical ventilation to accommodate noninvasive mechanical ventilation, which was introduced as a treatment mode for serious respiratory failure in MG ( ). About 15% to 20% of patients with MG experience an MC at some point in the duration of this disease, but MC can be the first manifestation of MG in 14%–69% of MG ( ). Thymoma was observed in 32% of cases of MC and may be a risk factor ( ). Patients with MuSK-MG have a higher risk of MC because of predominant bulbar weakness ( ).

MC is caused by severe weakness of respiratory muscles, upper airway muscle (bulbar myasthenia), or both. It occurs in AChR-positive, MuSK-positive, and double seronegative MG. Infections are known to be the most common precipitating factor for MC, accounting for 38% of cases in one series. Other rare, recognized precipitating factors for crisis are surgery, pregnancy, and medications ( Box 19.1 ). Excessive dosing of AChEIs can potentially increase weakness due to depolarization blockade at neuromuscular junction. Previously, it was customary to differentiate between cholinergic crisis and MC by edrophonium test in MC patients with a known diagnosis of MG taking high doses of AChEIs. Cholinergic crisis is rare in today’s practice because high doses of AChEI are not used anymore ( ).

Box 19.1
Drugs/Factors Associated with Unmasking or Aggravating Myasthenia Gravis (MG) a

a For further information, consult www.myasthenia.org/docs/MGFA_Medications

Definite Association

  • Drugs/factors that are known to induce autoimmune MG and thus are contraindicated in patients with MG

    • Alpha interferon

    • Bone marrow transplantation

    • D-Penicillamine

  • Drugs/factors that are known definitely to unmask or aggravate MG and thus should be avoided in patients with MG

    • Botulinum toxin, type A (Botox)

    • Antibiotics—ketolide (telithromycin [Ketek])

    • Depolarizing and nondepolarizing neuromuscular blocking agents

    • Iatrogenic hypermagnesemia including that due to magnesium sulfate

Probable Association

  • Drugs/factors that are known to unmask or aggravate MG in more than two cases and thus are not recommended for use in patients with MG; if used, then close monitoring is required

    • Antibiotics

      • Aminoglycosides: gentamicin, kanamycin, netilmicin, neomycin, streptomycin

      • Fluoroquinolones: ciprofloxacin, ofloxacin

      • Other antibiotics: bacitracin, colistin, polymyxin

    • Cardiovascular drugs

      • Beta blockers: propranolol

      • Verapamil

      • Procainamide

      • Statins

    • Quinolones: chloroquine, fluoroquinolone antibiotics, quinidine, quinine

    • Iodinated contrast media

    • Ophthalmologic medication: timolol maleate eye drops

    • Psychotropic medication: lithium carbonate

Possible Association

  • Drugs/factors that are known to unmask or exacerbate MG in one or two cases and thus can be used safely in most patients with MG, with close monitoring recommended

    • Antibiotics: amino acid antibiotics, ampicillin, azithromycin, clarithromycin, clindamycin, erythromycin, imipenem and cilastatin sodium, macrolides, nitrofurantoin, pyrantel pamoate, ritonavir, sulfonamides, tetracyclines, vancomycin

    • Beta blockers: accubutol, oxprenolol, practolol

    • Other cardiac drugs: bretylium, propafenone HCl, trimethaphan

    • Anticonvulsant medications: carbamazepine, ethosuximide, gabapentin, trimethadione

    • Ophthalmologic medications: betaxolol hydrochloride, echothiophate, proparacaine, tropicamide

    • Psychotropic medication: phenothiazide

    • Other medicines neurologists often prescribe: riluzole, glatiramer acetate, methocarbamol, trihexyphenidyl (Artane)

    • Miscellaneous drugs/agents: fludarabine, cisplatin, interleukin, carnitine, nicotine patch, diatrizoate meglumine, chlorine gas

Diagnosis of MC in patients with an established diagnosis of MG is relatively easy. In those MG patients with MC as the first manifestation, the diagnosis of MG is based on the clinical findings and should be confirmed by edrophonium test, RNS test, or, rarely, SFEMG. The RNS test can provide a rapid objective diagnostic confirmation of MG in 92% of cases and identify cholinergic crisis pattern ( ).

Clinical Signs of Myasthenic Crisis

Prompt recognition of impending respiratory paralysis is the key to successful management of MC. Features of impending MC include severe bulbar weakness, marginal vital capacity (20 to 25 mL/kg), weak cough with difficulty clearing secretions from the airway, or paradoxical breathing, while supine. These patients should be admitted to the ICU and monitored closely. These patients should also receive no food or liquid by mouth to prevent aspiration. Careful observation and bedside measurements (pulse rate, blood pressure, oxymetry, vital capacity, tidal volume, and peak inspiratory and expiratory pressure) are more important than repeated monitoring of blood gases. The 20/30/40 rule (vital capacity <20 mL/kg [or 1 L]; peak inspiratory pressure >-30 cm H 2 O; and peak expiratory pressure <40 cm H 2 O) is probably the most helpful guide to decide when intubation is necessary. In MC, the clinician should anticipate the need for respiratory assistance rather than deal with emergency intubation (“when in doubt, intubate”).

Noninvasive mechanical ventilation with bilevel positive pressure ventilation (BIPAP) has been used as an alternative to intubation and mechanical ventilation in MG, but experience is still relatively limited. One retrospective study showed that in 14 (58%) of 24 cases initially treated with BIPAP, endotracheal intubation was avoided and patients treated with BIPAP had shorter stays in the ICU and the hospital than those treated initially with intubation. The only predictor of BIPAP failure was a pCO 2 level exceeding 45 mm Hg on BIPAP initiation. This study suggests the possibility that some patients with MC or impending crisis might be managed with BIPAP ( ).

Treatment of Myasthenia Gravis During Myasthenic Crisis ( Box 19.2 )

To date, treatment of MG during MC is based on clinical consensus, not on evidence-based data. There is a general agreement among experts that high-dose steroids in combination with PE or immunoglobulin are the cornerstone of MG treatment during MC.

Box 19.2
Principles of Management of Myasthenic Crisis
AChEI , Acetylcholinesterase inhibitor; IVIg , intravenous immunoglobulin; MG , myasthenia gravis.

  • 1.

    Provide airway assistance and ventilation: This step is most critical; always intubate when in doubt.

  • 2.

    Discontinue AChEIs initially.

  • 3.

    Remove and treat precipitating factors.

  • 4.

    Routine monitoring

    • Cardiac monitoring

    • Oximetry

    • Pulmonary function monitoring: forced vital capacity, peak inspiratory pressure, and expiratory pressure, every 2–4 hours

  • 5.

    General medical care

    • Institute prophylaxis for deep vein thrombosis with low-molecular-weight heparin.

    • Identify and treat infection.

    • Identify and treat cardiac arrhythmia and congestive heart failure.

  • 6.

    Initiate specific treatment for MG.

    • Plasma exchange (removal of 2 to 3 L of plasma volume on each session × 5)

    • Alternatively, IVIg (0.4 mg/kg/day × 5)

    • High-dose steroid (prednisone 1 mg/kg/day)

    • Start a low-dose AChEI when the patient gets stronger and benefit is found with trial

There have been few treatment trials in MC, and both PE and IVIg are comparable in terms of efficacy on the basis of clinical evidence. Since response to PE is more predictable and a good clinical response to PE in some myasthenic patients refractory to IVIg was reported ( ), many experts take the view that PE is probably more effective than IVIg in MC, and thus PE has become the most commonly used immunotherapy for MC ( ; ). In one retrospective multicenter review of 54 episodes of MC, PE had a superior outcome for ventilatory status at 2 weeks and functional outcome at 1 month, despite higher complication rate with PE ( ).

IVIg represents an alternative short-term immunomodulating therapy for MC in patients who are poor candidates for PE due to various medical reasons, including vascular access problems, septicemia, and cardiac failure. The wider availability and ease of administration of IVIg make it an automatic choice as the first-line therapy for MC in patients who have difficulty accessing a medical center that can perform PE.

Steroids are the drug of choice as the immunotherapeutic agent for maintaining an improved status by PE or IVIg. This is because an initial stable improvement is achieved within weeks (average, 13–39 days) of steroid treatment in contrast to within months with other immunosuppressants ( ). Most experts recommend high-dose (1 gm/kg) prednisone daily as the initial regimen.

IV pyridostigmine infusions at 1 to 2 mg/hour have been advocated by in patients during crisis. In 63 episodes with MC treatment in an acute care setting with pyridostigmine ( n = 24), pyridostigmine and prednisolone ( n = 18), and PE alone ( n = 23), they did not find any signficant differences in short-term efficacy; there were comparable long-term outcomes and side effect profiles among the different regimens. On the basis of these results, they advocated an approach tailored to the individual patient considering the precrisis condition of the patient and the precipitating factors of MC. However, because of possible cardiac complications, sometimes fatal, cholinergic crisis, and excessive secretions, most experts recommend stopping pyridostigmine during crisis (drug holiday) and reinstituting it orally when patients are getting stronger and the benefit is found with a trial ( ). This is usually around the extubation time. Some believed that a drug holiday heightens patient sensitivity to AChEIs ( ). The usual recommendation is to start at a low dose (60 mg every 8 hours enterally) and gradually increase to a dose that produces a clear benefit.

Immunosuppressants are usually not recommended because the improvement from treatment with these agents is generally not seen for several months. They may be used in place of prednisone, however, when steroid therapy is contraindicated, but not for MC. There is no indication of thymectomy during MC.

Respiratory Management and Treatment During Myasthenic Crisis

This will be discussed in detail in another chapter. When intubation is performed, nasotracheal intubation is preferred as nasotracheal tubes are more comfortable than orotracheal tubes. Tracheostomy should be considered for patients requiring more than 2 weeks of mechanical ventilation.

Weaning trials should begin after patients demonstrate a clear trend of improved respiratory muscle strength. The general guideline is the 10/20/40 rule: vital capacity >10 mL/kg, maximum negative inspiratory force <-20 cm H 2 O, and peak positive expiratory force >40 cm H 2 O.

The three most common complications are fever (69%), pneumonia (51%), and atelectasis. Aggressive treatment for pneumonia and atelectasis is mandatory.

Mortality in MC has declined from over 40% in the early 1960s to approximately 5% in the 1970s ( ), due in large part to the improvement in respiratory care and ICU management. Current statistics still report a 4% mortality rate despite newer treatments and better intensive medical care, indicating no documented improvement in fatality rates over the last several decades ( ). All deaths were ascribed to severe medical comorbidity ( ). The most common causes of death in MC are cardiac arrhythmia or infections.

Treatment in the Specific Subtypes of Myasthenia Gravis

Ocular Myasthenia Gravis

Pyridostigmine is usually effective for ptosis, but not for double vision, a more troublesome symptom. Steroids are effective in OMG as well as in GMG. AZA was also used as the second line immunosuppressive medication. Oral steroid produces improvement in 66%–85% of OMG patients ( ). However, permanent remission is associated with steroids in only 10% of patients.

A major question in the treatment of OMG is whether steroids or immunosuppressant drugs may prevent the development of GMG ( ; ). In 49%–69% of patients, OMG progressed to GMG. This occurs in 2 years in 50%–88% of cases. In AChR-Ab-positive OMG, this conversion occurs in 45%–71% of cases.

Retrospective studies have suggested reduced rates of generalization with steroids in four studies and AZA in one study ( ). Only 7%–17% of OMG patients treated with prednisone developed GMG compared with 36%–83% of those not treated with immunosuppressants. In OMG patients who received AZA treatment, GMG developed in 16% of patients in 2 years and 23% in 4 years. There are no randomized controlled trials to support this finding ( ).

Thymectomy is not generally indicated in OMG because of its benign course ( ). Two case series claimed its benefits (class IV): no generalization in 18 patients in 26 months and “cure” in 51% in 61 patients with a mean follow-up of 9 years ( ).

Muscle-Specific Kinase–Myasthenia Gravis

MuSK is a surface receptor that plays an essential role in the clustering of AChR during development. MuSK-Ab-positive MG (MuSK-MG) was first reported in 2001. MuSK-Ab is negative in OMG. MuSK-Ab is reported to be positive in 0% in Norway to 49% in Turkey, with a mean frequency of approximately 35% of generalized seronegative MG ( ). MuSK-MG is predominantly observed in females, and the onset is younger. Patients tending to have a more severe form of MG and crisis are more common in MuSK-MG. Prominent cranial and bulbar weakness is the most consistent pattern. Profound facial and tongue atrophy has been observed in some of these cases. Predominant neck, shoulder, and respiratory involvement, but without ocular weakness, is another rare manifestation. MuSK-MG patients tend to have a more severe form of MG and crisis is common.

A positive edrophonium test is less common in MuSK-MG than other forms of MG, being positive in half of the cases. The RNS test of limb muscle had a relatively low yield (25%–57%) in MuSK–MG, but the RNS test of facial muscles showed a decrement in 85% of cases ( ). Percentage decrements in the RNS test in the facial muscle are of greater magnitude than in the other two forms. Thus, including facial muscles in the RNS protocol is important when evaluating patients with suspected MuSK-MG. In SFEMG, many series reported a relatively low yield of abnormality in the limb muscles, but when the proximal and cranial muscles are added for the study, it is abnormal in the majority of MuSK-MG patients.

The clinical response to anticholesterase agents in MuSK-MG has generally been disappointing ( ). With standard pyridostigmine doses, unresponsiveness, actual worsening, and intolerance manifested by severe muscarinic and nicotinic side effects are common. Most patients need one or a combination of immunotherapies. Among the various immunotherapies, steroid and PE seem to result in the best response ( ). However, a general impression is that MuSK-MG is refractory to the standard treatment because finding the right combination of immunotherapy is time-consuming and difficult.

3,4-DAP was used in a few MuSK-MG patients, with some success. One adult patient showed an improvement of 3 in the QMG score with 3,4-DAP 40 mg/day ( ). In one randomized study in seven patients, the end-points were met but the other randomized study did not meet the end-points, but a definite study is needed to prove this ( ).

Myasthenia Gravis with Lipoprotein Receptor–Related Protein 4 And Agrin Antibody

LRP4 and agrin antibodies were two recent additions to AChR and MuSK antibodies for MG. LRP4 and agrin antibody–positive MG is rarer than MuSK-MG. Almost all patients with LRP4 or agrin antibodies had GMG, but few had thymoma ( ; ). In terms of treatment, no specific difference is advised from AChR-negative GMG. Overall, the response of LRP4-MG patients to treatment was similar to published responses of AChR-negative MG rather than to MuSK-MG patients.

Pregnancy

The course of MG during pregnancy is unpredictable; approximately one third of patients remain the same, one third improves, and the remaining one third worsens ( ). Worsening of MG symptoms is more likely in the first trimester and the puerperal periods and is possibly related with level of alfa-fetoprotein, which is known to block the binding of AChR antibody to AChR. The clinical state of MG at the beginning of pregnancy or in one pregnancy does not predict the occurrence of exacerbations or remission during gestation. MG does not provide any adverse effect on the course of pregnancy. The coexistence of MG and preeclampsia, although rare, may produce a high degree of morbidity and mortality in both the mother and the fetus. Magnesium sulfate, the drug of choice in the treatment of preeclampsia, is contraindicated in MG because hypermagnesemia inhibits the release of ACh at the neuromusuclar junction and worsens MG muscle weakness, causing a severe MC.

Management of Myasthenia Gravis During Pregnancy

Patients should be evaluated more frequently for possible worsening of MG throughout pregnancy. Screening for asymptomatic bacteriuria should be regularly performed and appropriate treatment of urinary tract infections is required because infection is a known cause of MG worsening and crisis. Pregnant women with MG should carefully monitor fetal movements and seek medical advice as soon as they find these reduced. If indicated, fetal ultrasound evaluations should be done to detect any sign of the fetal akinesia sequence.

Oral AChEIs are the drug of choice for the symptomatic treatment of MG and can be used alone in mild cases of MG. Pyridostigmine is considered to be safe during pregnancy when used at the recommended dosage of less than 1000 mg (14 tablets) a day.

Steroids are the drug of choice for immunotherapy because they present little, if any, teratogenic risk to the fetus, and only a slight increase in the incidence of cleft palate has been reported ( ). Another advantage of steroids is that they work much faster than many immunosuppresants in achieving the stable course of MG. All other immunosuppressants should be avoided before and during pregnancy whenever possible because of potential teratogenic effects.

PE or IVIg can be and has been used as an effective treatment for severe worsening of MG or crisis during pregnancy without any major complication. Thymectomy during pregnancy has no role because of its delayed effect and possible surgical risk.

Management of Myasthenia Gravis During Labor and Delivery

Most labors can safely be completed vaginally ( ). Epidural anesthesia is preferred for vaginal and surgical delivery because many myasthenic patients are sensitive to many anesthetic agents ( ). Local anesthetics can be used, but high doses should be avoided because they may interfere with NMT. Lidocaine is the recommended local anesthetic because it is an aminoacyl amide and is not affected by AChEIs. Women taking steroids during pregnancy should continue the treatment during labor and stress doses should be given (an extra dose of 50–100 mg of prednisone IM at the first stage of labor or Solu-medrol 200 mg IV drip during labor).

During the second stage of labor, which involves the voluntary expulsive effort, the patient may become exhausted and may require immediate AChEIs. Neostigmine (1.5–2.0 mg IM or 0.5 mg IV every 3–4 hours) is preferred to pyridostigmine. Contrary to expert recommendation, many MG women prefer C-section for delivery of the baby because of an overall increased rate of delivery complications and a higher rate of intervention during delivery ( ),

Neonatal Myasthenia Gravis

Neonatal MG (NMG) is a syndrome that occurs in 10%–20% of newborns of myasthenic mothers shortly after birth. NMG is reported in all types of MG. Symptoms develop most commonly 12 to 48 hours after birth and include generalized weakness, hypotonia, difficulty feeding, feeble cry, ptosis, facial paresis, and respiratory distress. NMG usually resolves within 3 weeks but occasionally persists for as long as 4 months. This spontaneous improvement is most likely due to the gradual degradation of the maternally derived IgG in a newborn baby. NMG symptoms usually respond to anticholinesterase medications, and ventilatory support should be used as necessary until the weakness resolves.

Arthrogryposis Multiplex Congenita

Maternal MG is a rare cause of arthrogryposis multiplex congenita, which consists of nonprogressive multiple congenital joint contractures developing in utero from the lack of fetal movements caused by maternal AChR antibodies to fetal AChR.

Breastfeeding

The American Academy of Pediatrics classifies pyridostigmine and steroids as compatible with breastfeeding and methotrexate as contraindicated during breastfeeding ( ). Although there are no firm data for the safety of other immunosuppresants, in general, breastfeeding is not recommended for other immunosuppresants.

Thymoma

Thymoma is an epithelial neoplasm of the thymus and represents the most common primary neoplasm of the anterior superior mediastinum. The majority of thymomas are completely encapsulated, but in one third of cases there may be invasion of the tumor capsule and/or the surrounding structure (invasive thymoma or malignant thymoma).

MG is common in thymoma (T-MG), occurring in approximately 44% of thymoma patients. Conversely, 10%–15% of MG patients have thymoma. Thymoma occurs in MG at any age. However, it is much more common among middle-aged and older patients. MG tends to be more severe in patients with thymoma than those with early-onset MG. The MG subgroup has a poorer prognosis than the group with MG without thymoma, requiring long-term immunotherapy with prednisone, AZA, or other immunosuppressive drugs.

When the diagnosis of T-MG is established, thymoma should be removed surgically, ensuring radical excision. The removal of thymoma does cure the thymic neoplasia in most cases, but MG symptoms usually continue after thymectomy.

The pharmacological treatment of T-MG is not different from that for nonparaneoplastic MG, except for tacrolimus that should be considered in difficult cases. Tacrolimus, an immunosuppressant and enhancer of ryanodine receptor (RyR)–related sarcoplasmic calcium release, is beneficial in MG patients with RyR antibodies that in theory might block the RyR. It seems to have a purely symptomatic effect in addition to its immunosuppressive effects. Since most patients with T-MG have RyR antibodies, tacrolimus may be of more value specifically in T-MG patients.

The first antibodies described in MG in 1960 were the striational muscle (SM) antibodies, and the main antigens were later identified as titin and RyR. SM antibodies were present in 24% of patients with thymoma alone, in 38% of patients with MG without tumor, and in 80%–90% of T-MG patients ( ). There is a correlation between the titer of SM antibody and the likelihood of a thymoma. The appearance of thymic enlargement on computed tomography (CT) scan and a high titer of antistriational antibody make the diagnosis of thymoma virtually certain. More specific SM antibody tests against titin and RyR have become available in recent years. Titin and RyR antibodies have been found in 95% and 70% of thymoma MG patients, respectively ( ), and 58% and 6% of late-onset MG patients have titin and RyR antibodies, respectively. Titin and RyR antibody tests and CT scan of the mediastinum share similar sensitivities for thymoma in MG. claimed that the presence of titin and RyR antibodies in an MG patient younger than 60 years strongly suggests a thymoma, while the absence of such antibodies at any age strongly excludes thymoma.

The treatment of thymomas is determined after the lesion has been staged (class III). In general, complete surgical resection of tumor is the preferred treatment. This is the treatment of choice in stage I tumor (noninvasive thymoma), given the very low recurrence rate. In invasive thymoma (stages II, III, and IV), adjuvant radiation therapy is recommended because of the recurrence of tumor.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here