128
Views
15
CrossRef citations to date
0
Altmetric
Review

Current and emerging treatments for the management of myasthenia gravis

Pages 313-323 | Published online: 22 Jul 2011

Abstract

Myasthenia gravis is an autoimmune neuromuscular disorder. There are several treatment options, including symptomatic treatment (acetylcholinesterase inhibitors), short-term immunosuppression (corticosteroids), long-term immunosuppression (azathioprine, cyclosporine, cyclophosphamide, methotrexate, mycophenolate mofetil, rituximab, tacrolimus), rapid acting short-term immunomodulation (intravenous immunoglobulin, plasma exchange), and long-term immunomodulation (thymectomy). This review explores in detail these different treatment options. Potential future treatments are also discussed.

Introduction

Myasthenia gravis (MG) is a neuromuscular disorder characterized by a clinical course of fluctuating, painless muscle weakness. Typically it begins in the extraocular muscles and remains purely ocular in 15% of patients. MG generalizes by descending to involve the bulbar muscles, and subsequently the neck, proximal limb, and sometimes respiratory muscles. Even with modern treatments, at least 20% of patients experience a myasthenic crisis which requires intubation and mechanical ventilation, usually within the first 2 years of the diagnosis.

MG is an autoimmune disorder usually caused by antibodies to postsynaptic proteins, mainly nicotinic acetylcholine receptor (AChR) and muscle-specific kinase (MuSK), but there are other as yet undiscovered antigens. These antibodies reduce the number of functional AChRs and thus impair neuromuscular transmission. The prevalence of MG has increased from around 5 per million population between 1915 and 1934Citation1 to about 200 per million population now,Citation2 in part due to improved detection of the antibodies to the postsynaptic proteins. The estimated annual incidence of MG is between 1 in 10,000 to 1 in 50,000 of the population,Citation3 but the clinical recognition of this rare disease remains difficult with many patients going undiagnosed for many months from symptom onset, and the diagnosis only correctly made after several physician consultations. In the past 70 years, treatment advances have reduced the mortality of MG from 70% between 1915 and 1934Citation1 to 5% or less now.Citation4

In this review an overview will be given of the mechanism, evidence, indication, and relevant adverse effect profile of the different treatment options in generalized MG. Several potential future therapies will also be discussed.

Symptomatic treatment

Acetylcholinesterase inhibitors

In MG, the first-line option is symptomatic treatment with acetylcholinesterase inhibitors. Pyridostigmine bromide is the most commonly used drug. Other acetylcholinesterase inhibitors such as neostigmine are rarely used because of their poorer pharmacodynamic profiles and tolerability. In an observational study of 14 MG patients comparing pyridostigmine with neostigmine, it was concluded that over 1 year, pyridostigmine was more effective with less adverse events.Citation5 Similar conclusions were reached in another observational study of 69 patients which compared the use of pyridostigmine with neostigmine.Citation6 There is no large randomized controlled trial of acetylcholinesterase inhibitors in MG, but the clear response of this drug in observational studies would make depriving patients in the placebo arm of a randomized controlled trial unethical and unjustifiable.Citation7

Pyridostigmine is most effective early in the course of MG and over time increasing tolerance to the drug develops which may necessitate dose escalation. Most MG patients do not achieve adequate response with acetylcholinesterase inhibitor treatment and will require further immunosuppression. It is also noteworthy that some MuSK antibody-positive patients may show nonresponsiveness to acetylcholinesterase inhibitors. In one study, 71% of MuSK antibody positive patients failed to respond to acetylcholinesterase inhibitors, compared to 18% respectively of AChR antibody positive and seronegative patients.Citation8 Pyridostigmine is generally well tolerated. Adverse events include muscarinic side effects such as nausea, vomiting, abdominal cramping, diarrhea, diaphoresis, increased lacrimation, excessive respiratory secretions, bradycardia, and atrioventricular block. Antimuscarinics such as propantheline bromide provide effective symptomatic relief against the abdominal adverse events induced by pyridostigmine. Pyridostigmine may also cause nicotinic adverse events such as muscle cramps and fasciculations, but these rarely require a change in the dose of the drug. High doses of pyridostigmine may desensitize AChRs and induce weakness resulting in a cholinergic crisis. If there is such a concern, cholinesterase inhibitors need to be temporarily withdrawn and the patient carefully monitored for improvement.

Short-term immunosuppression

Corticosteroids

Corticosteroids are thought to act on the immune system by inhibiting the activation of T-cells and impairing the function of cells of the monocyte/macrophage lineage. Adrenocorticotrophic hormone (ACTH) was first described to have a beneficial effect in MG in 1935.Citation9 ‘Good improvement’ was reported in a study of 100 patients with severe refractory MG given ACTH.Citation10 In four large retrospective studies of generalized MG using various doses of corticosteroids and with different follow-up durations, 74% of a total of 422 patients achieved good overall improvement of muscle strength or remission.Citation11Citation14 A prospective study of 600 MG patients (151 generalized, 449 pure ocular) treated with moderate doses of corticosteroids followed by low-dose maintenance showed an overall improvement in 95% of cases, but no clear breakdown between the generalized and ocular cases were given.Citation15

A randomized double-blind trial of prednisolone versus placebo in 13 patients with generalized MG showed no significant improvement of muscle strength at 6 months.Citation16 Another randomized double-blind trial of intravenous methylprednisolone versus placebo in 19 patients with generalized MG showed a significant short-term benefit from corticosteroids 2 weeks after treatment.Citation17 An open-label randomized trial comparing high-dose intravenous methylprednisolone and low-dose oral prednisolone in 39 patients with juvenile MG (eight generalized and 31 ocular) did not report any significant difference in improvement between the two groups, although the exact time of measurement and breakdown between the generalized and ocular cases were unclear from the paper.Citation18

Corticosteroids are useful as short-term immunosuppressants in MG. Oral prednisolone is the most commonly used first-line drug. Corticosteroids are usually used as an interim measure while titrating up the doses of other immunosuppressants and waiting for those immunosuppressants to take full effect. A temporary worsening of MG known as a ‘steroid dip’ may occur if corticosteroids are started at a high dose. This steroid dip usually occurs 4 to 10 days after starting treatment and may precipitate a MG crisis. To overcome this problem, treatment should be started at a low dose on alternate daysCitation19 and gradually titrated upwards.Citation20 In critically ill patients, high-dose daily corticosteroids can be started and additional short-term treatments such as intravenous immunoglobulin or plasma exchange can be used to overcome any temporary worsening.Citation21 The potential adverse events from prolonged corticosteroid use necessitates a slow dose reduction to the minimum effective dose given on alternate days,Citation20 but there is no clear evidence on the best time to reduce corticosteroids, how quickly or safely to do so, or how long patients should be kept on corticosteroids.Citation21

Long-term corticosteroid use is associated with many adverse events, including cushingoid features, hypertension, diabetes, osteoporosis, infections, psychiatric disorders, insomnia, and elevations in white blood cell count. Calcium, vitamin D and bisphosphonate supplement should be started concurrently with corticosteroids to protect against osteoporosis. In some patients, corticosteroid-induced myopathy needs to be differentiated from myasthenic weakness.

Long-term immunosuppression

Azathioprine

Azathioprine acts through its metabolite 6-mercaptopurine as a purine antagonist by inhibiting DNA synthesis and cell proliferation. There are several observational studies of azathioprine in MG. In a series of patients previously unresponsive to ACTH or glucocorticoids, 78% of 26 patients improved on azathioprine.Citation22 In another study, 91% of 78 patients improved when treated with azathioprine, given alone or in combination with corticosteroids, thymectomy or both.Citation23 Another study showed 83% of 18 patients improved with azathioprine treatment alone for more than 6 months.Citation24 In a further study, all 41 MG patients followed for more than 3 years improved when azathioprine was used either alone or in combination with prednisolone.Citation25 In another study, 75% of 32 patients treated with azathioprine monotherapy improved compared with 70% of 57 patients treated with a combination of azathioprine and corticosteroids.Citation26 The use of early ‘high-dose’ immunosuppression with azathioprine and prednisolone resulted in 50% of MG patients achieving remission after 2 years, compared with a remission rate of only 16% in those on a ‘low-dose’ regimen.Citation27

A randomized unblinded trial of azathioprine plus initial prednisolone versus prednisolone alone in 41 patients with generalized MG showed less clinical deterioration within the first 60 months (the primary endpoint) in the azathioprine group compared to the prednisolone monotherapy group, although no differences in muscle strength measurements were seen between the two groups.Citation28 A further randomized double-blind trial of azathioprine plus prednisolone versus prednisolone plus placebo in 34 patients with generalized MG reported no significant differences between the two treatment groups with regard to objective or subjective muscle strength measurements.Citation29 In the same study, the median prednisolone dose did not differ significantly between the two treatment groups at 12 months, but was significantly reduced at 36 months in the azathioprine plus prednisolone group compared with the prednisolone plus placebo group, suggesting that azathioprine had a corticosteroid-sparing effect.Citation29

In many countries, azathioprine is the first choice long-term immunosuppressant drug which is started together with corticosteroids to allow tapering of the latter to the lowest possible dose. Common adverse events of azathioprine are hepatotoxicity, nausea, vomiting, rash, cytopenia, and pancreatitis. Malignancy, primarily lymphoma, is a potential long-term complication but the absolute risk is difficult to ascertain because it is difficult to differentiate the effects of the drug from age-related increases in the background incidence of cancer. Patients with deficiency of thiopurine S-methyltransferase may develop severe bone marrow-related toxicity. Red cell thiopurine S-methyltransferase activity can be measured to identify at risk patients prior to starting treatment.

Cyclosporine

The effect of cyclosporine is mediated by calcineurin inhibition of T-cell interleukin-2 production. There are three uncontrolled trials of cyclosporine in patients with severe MG. In one trial, where patients recruited were unresponsive either to acethylcholinesterase inhibitors alone or to the combination of thymectomy plus corticosteroids or azathioprine, 80% of ten patients showed marked improvement after 12 months of treatment.Citation30 In another trial of patients who had failed to respond to thymectomy, corticosteroids, azathioprine, or all three, 78% of nine patients showed marked improvement after a mean of 2 years’ treatment.Citation31 In a third trial of patients who had not responded to thymectomy, corticosteroids and azathioprine, 85% of 52 patients showed marked improvement after an average follow-up duration of 30 months’ treatment.Citation32

A randomized double-blind trial of cyclosporine monotherapy versus placebo in 20 patients with generalized MG reported significantly greater increases in muscle strength in the cyclosporine group compared with the placebo group both at 6 and 12 months.Citation33 Another randomized double-blind trial of cyclosporine plus prednisolone versus prednisolone plus placebo in 39 patients with generalized MG demonstrated that at 6 months, the cyclosporine group had a significantly greater increase in muscle strength than the placebo group.Citation34 Rather surprisingly, there was no significant difference in the percentage change of corticosteroid dose between the two groups at the end of 6 months;Citation34 if cyclosporine is effective, it might be expected to have a corticosteroid-sparing effect.

Although cyclosporine has been shown to be clinically effective in MG, its use is limited by the potentially serious adverse events associated with this drug such as nephrotoxicity, hypertension, and malignancy. Other common adverse events include flu-like symptoms, gingival hyperplasia, hypertrichosis, myalgia, and tremor.

Cyclophosphamide

Cyclophosphamide is a DNA-alkylating drug and nonspecific cell-cycle inhibitor. One study reported stable remissions in 42 patients who were given cyclophosphamide for 2 to 37 months. Of these patients, 33 received concomitant corticosteroid treatment and five underwent thymectomy.Citation35 A randomized double-blind trial of intravenous pulsed cyclophosphamide plus prednisolone versus prednisolone plus placebo in 23 patients with severe generalized MG, demonstrated that cyclophosphamide significantly improved muscle strength at 12 months, but not at 6 months. The cyclophosphamide group also had significantly lower corticosteroid doses at 6 and 12 months than at the start of the trial.Citation36

Although there is evidence of clinical efficacy of cyclophosphamide in MG, the use of the drug is restricted by its relatively high risk of severe adverse events such as bladder toxicity, bone marrow suppression, opportunistic infections, infertility, and malignancy. Other common adverse events associated with cyclophosphamide include nausea, vomiting, alopecia, abdominal pain, and diarrhea.

Methotrexate

Methotrexate is a folate antagonist that inhibits de novo synthesis of purines and pyrimidines. There are no good quality published studies of methotrexate in MG. However, methotrexate is still often used as second-line treatment in patients who do not tolerate or are unresponsive to azathioprine for several reasons: (a) data extrapolated from other autoimmune disorders suggest that it should be effective in MG,Citation20 (b) expert panel guidelines support its use as second-line treatment in MG,Citation20 and (c) many physicians have extensive personal experience of successfully using it in MG.Citation21 A clinical trial to assess the efficacy of methotrexate in MG is underway.

Adverse events associated with methotrexate are usually mild such as alopecia, mucositis, gastrointestinal intolerance, and mild elevation of liver enzymes. Nevertheless, severe adverse events including hepatotoxicity, hematopoietic suppression, and pneumonitis may sometimes occur.

Mycophenolate mofetil

Mycophenolate mofetil, when converted to its active metabolite mycophenolic acid, inhibits T-cell proliferation by blocking purine synthesis. Several open-label trials of mycophenolate mofetil in MG have been published. In one trial, 68% of 22 patients who were either previously unresponsive to azathioprine, on combination therapy with corticosteroids or on mycophenolate mofetil monotherapy, treated for between 2 and 18 months demonstrated marked improvement.Citation37 In another trial, 67% of twelve patients who had not responded to corticosteroids, azathioprine, cyclosporine, or thymectomy showed marked improvement after 6 months of treatment.Citation38 In a further trial, 59% of 32 patients who had been unresponsive to corticosteroids, azathioprine, cyclosporine, methotrexate, or thymectomy showed marked improvement after an average of 11 months’ treatment.Citation39 A retrospective analysis of mycophenolate mofetil in 85 MG patients, 48 of whom had undergone thymectomy and 66 of whom were on various combinations of corticosteroids, azathioprine, cyclosporine, and methotrexate, showed an improvement in 73% of patients.Citation40 Another retrospective study of MG patients examined the effect of mycophenolate mofetil either as monotherapy or in combination with corticosteroids. In the monotherapy group, approximately 75% of 36 patients achieved a desirable outcome after 25 months of treatment, compared with approximately 16% when mycophenolate mofetil was initially started. In the mycophenolate mofetil plus corticosteroids group, approximately 75% of 66 patients achieved a desirable outcome after 24 months of treatment, compared with approximately 21% when mycophenolate mofetil was initially started.Citation41

A randomized, double-blind trial of mycophenolate mofetil plus corticosteroids versus corticosteroids plus placebo of 176 MG patients for 36 weeks did not show efficacy for mycophenolate mofetil.Citation42 Similarly, a second randomized double-blind trial of mycophenolate mofetil plus corticosteroids versus corticosteroids plus placebo of 80 MG patients for 12 weeks failed to demonstrate efficacy for mycophenolate mofetil.Citation43

The results of the two randomized controlled trials of mycophenolate mofetil were considered limited by their relatively short duration and greater than predicted benefit from the prednisolone doses used in the placebo arms of the studies.Citation44,Citation45 Since the drug is normally well tolerated with a relatively good adverse effect profile, it can be considered as third-line treatment in MG. The adverse events of mycophenolate mofetil are usually mild such as headache, nausea, and diarrhea, but more serious adverse events including infections, hematopoietic suppression, and hepatotoxicity can occasionally occur. Progressive multifocal leukoencephalopathy has been associated with mycophenolate mofetil treatment.

Rituximab

Rituximab is a chimeric monoclonal antibody against the B-cell surface marker CD20. In one series, six patients with refractory MG (four MuSK antibody positive, two AChR antibody positive) were treated with rituximab, resulting in a decreased need for immunosuppressant treatment, improved clinical function or both in all patients.Citation46 In another series, five patients with generalized MG resistant to conventional immunosuppression were treated with rituximab, with good clinical improvement reported in all.Citation47 In a retrospective study of ten patients with generalized MG (three MuSK antibody positive, seven AChR antibody positive) refractory to conventional immunosuppressant treatment, rituximab resulted in 60% of patients (including all the MuSK antibody positive cases) improving clinically or being able to decrease immunosuppressant treatment.Citation48 Another retrospective study of low-dose rituximab in refractory MG reported clinical improvement in 79% of the 14 patients (three MuSK antibody positive, eleven AChR antibody positive), including all MuSK antibody positive cases.Citation49

There is preliminary evidence that rituximab is beneficial in MG, but it should only be used in severe refractory cases unresponsive to other treatments. Ongoing trials are evaluating its use. The current prohibitive cost of this drug is a major disadvantage. Common infusion-related adverse events associated with rituximab are fever, chills, nausea, vomiting, flushing, and bronchospasm. Other more severe adverse events include neutropenia, infections, and the risk of progressive multifocal leukoencephalopathy.

Tacrolimus

Tacrolimus inhibits T-cell and interleukin-2 production via the calcineurin-mediated pathway. In a 16-week, open-label trial of 19 patients with generalized MG treated with low-dose tacrolimus, all of whom had previously undergone thymectomy and all but one of whom were on corticosteroid treatment, 37% showed clinical improvement at the end of the study.Citation50 Furthermore, 67% of twelve patients from the previous study that continued tacrolimus for up to 2 years showed clinical improvement.Citation51 In another open trial of low-dose tacrolimus in thymectomized and corticosteroid-dependent patients, 71% of the 17 patients treated improved clinically.Citation52 In a further open-label study of tacrolimus in severe MG, where all patients were on prednisolone and cyclosporine, and had undergone thymectomy, 87% of the 79 patients in the trial achieved pharmacological remission after a mean follow-up of 2.5 years.Citation53 Another study examined the use of low-dose tacrolimus in 212 MG patients. These patients were either thymectomized, cyclosporine- and prednisolone-dependent patients, or thymectomized patients who started tacrolimus early postoperatively, or patients over 60 years old with nonthymomatous generalized MG or in whom thymectomy was contraindicated. This study, with a mean follow-up of 49.3 months, showed that muscle strength increased 23% after 1 month of treatment and 29% at the end of the study.Citation54 A further published study of ten corticosteroid-dependent generalized MG patients treated with low-dose tacrolimus for a mean of 3.1 years reported that 50% of patients improved clinically at the end of the study.Citation55 However, another study reported no significant clinical improvement in nine corticosteroid-dependent generalized MG patients treated with low-dose tacrolimus for 5 years.Citation56 In another study, 86% of 47 generalized MG patients on low-dose tacrolimus over 24 weeks showed clinical improvement.Citation57

One randomized unblinded nonplacebo controlled trial of tacrolimus plus corticosteroids with or without plasma exchange versus no tacrolimus plus corticosteroids with or without plasma exchange in 34 generalized MG patients showed that tacrolimus reduced the need for other immunotherapy such as plasma exchange and corticosteroids.Citation58

Tacrolimus appears to be relatively safe at doses used in MG and can be considered as third-line treatment. Common adverse events include mild elevation of serum creatinine, hypertension, headache, hyperglycemia, tremor, parasthesia, decreased lymphocyte counts, and raised neutrophil counts. Tacrolimus may increase the risk of malignancy.

Rapid short-term immunomodulation

Intravenous immunoglobulin

The mode of action of intravenous immunoglobulin in MG is incompletely understood, but key mechanisms include interference of signalling via Fc receptors, neutralization of activated complement, suppression of idiotypic antibodies, and modulation of proinflammatory cytokines. Intravenous immunoglobulin was first used in the 1980s in MG.Citation59,Citation60 Two reviews which collated results from previously published uncontrolled studies showed that intravenous immunoglobulin improved MG in more than 70% of cases.Citation61,Citation62 Two open studies of a total of 21 patients with severe generalized MG showed improvement with intravenous immunoglobulin in all patients.Citation63,Citation64 Intravenous immunoglobulin has been shown to have comparable effectiveness to plasma exchange in preoperative thymectomy preparation of MG patients.Citation65,Citation66

Five randomized controlled trials comparing intravenous immunoglobulin with placebo or other treatments in MG have been carried out. Two trials compared intravenous immunoglobulin to placebo. One trial of 15 patients with mild-to-moderate generalized MG showed no significant difference between the two groups at 6 weeks. In the same study, the clinical efficacy of intravenous immunoglobulin was similar whether 1.2 g/kg or 2 g/kg of intravenous immunoglobulin was used.Citation67 In the other trial of 51 patients with acute exacerbation of generalized MG, intravenous immunoglobulin significantly improved muscle strength only in the group of patients with severe disease.Citation68 Two trials compared intravenous immunoglobulin to plasma exchange. In the first trial with 87 patients with acute exacerbation of generalized MG, no significant change in muscle strength between day 0 and day 15 was observed between the two treatment groups.Citation69 The second trial with a crossover design of 12 patients with moderate-to-evere MG in a stable phase showed no significant difference between the two groups after 1 and 4 weeks of treatment.Citation70 The Cochrane group reported that an unpublished, randomized controlled trial of intravenous immunoglobulin versus oral methylprednisolone in 33 patients with acute exacerbation of generalized MG failed to demonstrate any significant difference between the two treatment arms.Citation71 Another randomized controlled trial comparing two doses of intravenous immunoglobulin in 173 patients with acute exacerbation of generalized MG did not demonstrate any significant difference in efficacy between the doses of 1 g/kg and 2 g/kg.Citation72

There is some evidence that intravenous immunoglobulin is efficacious in acute severe exacerbation of generalized MG, but the evidence is less clear in chronic cases.Citation73 Intravenous immunoglobulin is typically used in acute exacerbation of MG or to optimize muscle strength before surgery. Common adverse events associated with intravenous immunoglobulin include headache, nausea, fever, and allergic reaction. In patients with IgA deficiency which may be present in 1 in 1000 people, a severe anaphylactic reaction might occur. Volume overload is a risk in cardiomyopathy and solute-induced renal failure may occur in patients with pre-existing renal impairment. High infusion rates may be associated with thrombotic complications such as myocardial infarction and stroke. However, the rate of adverse events from intravenous immunoglobulin appears to be less severe than those from plasma exchange.Citation69,Citation70

Plasma exchange

Plasma exchange is thought to work in MG by removing circulating antibodies, cytokines, immune complexes, and other inflammatory mediators. In MG, the concentration of AChR and MuSK antibodies has been shown to decrease with plasma exchange.Citation74,Citation75 Plasma exchange was first used in the 1970s in MG.Citation76 Several relatively large open studies of 20 or more patients, most of whom were already on other immunosuppressive or immunomodulatory treatment, have shown benefit from plasma exchange. Of 166 patients with generalized MG from five studies, 78% reported improvement with plasma exchange.Citation77Citation81 Three fairly large retrospective studies totaling 84 patients with generalized MG showed improvement in 96% of patients.Citation82Citation84 Another retrospective study of plasma exchange versus intravenous immunoglobulin in myasthenic crisis, demonstrated that the ventilatory status at 2 weeks and functional outcome after 1 month was better in the plasma exchange group.Citation85 A nonrandomized trial comparing different plasma exchange protocols in generalized MG failed to show any significant difference in efficacy between the treatments.Citation86 Prethymectomy plasmapheresis improves outcome after thymectomy in MG.Citation87,Citation88

One randomized controlled trial of plasma exchange versus prednisolone monotherapy in 14 patients with generalized MG did not show any significant difference in muscle strength between the two treatment arms after 1 month.Citation89 Two further randomized controlled trials comparing daily and alternate day plasma exchange in generalized MG did not show any significant difference in efficacy between the treatments.Citation90,Citation91

Plasma exchange is commonly used in acute severe exacerbation of MG to achieve temporary improvement or as a method of optimizing MG control before surgery. The choice between plasma exchange and intravenous immunoglobulin is often based on the physician’s opinion of the ability of a patient to tolerate each treatment. Since intravenous immunoglobulin is easier to administer, and associated with fewer adverse events than plasma exchange, and the efficacy of the two treatments is similar, the former is usually preferred to the latter.Citation73 Most adverse events associated with plasma exchange are due to problems with vascular access such as infection, thrombosis, pneumothorax, and air embolism. Excessive fluid volume shift can result in hypotension or fluid overload and congestive cardiac failure. Citrate infused for anticoagulation may cause disturbances in acid–base homeostasis and hypocalcemia.

Long-term immunomodulation

Thymectomy

The mechanism by which thymectomy potentially benefits non-thymomatous patients with generalized MG is incompletely understood, but may involve disrupting B-cells producing AChR antibodies. Although thymectomy has been the mainstay of treatment for non-thymomatous MG since the middle of the last centuary, a rigorous, evidence-based evaluation concluded that it only ‘might’ improve the chance of remission.Citation92 An ongoing study is examining whether the addition of thymectomy to prednisolone monotherapy in AChR antibody positive patients leads to an overall reduction in the dose of prednisolone over 3 years.

Most retrospective studies indicate a better response to thymectomy when it is performed early in the disease course of generalized MG; thus the procedure is usually recommended within the first 3 years of diagnosis.Citation93,Citation94 There is no consensus on the age of non-thymomatous patients who should undergo thymectomy, but most experts restrict the procedure to patients under the age of 60–65 years because older patients usually have an atrophic thymus.Citation93,Citation94 Numerous approaches to thymus removal have been advocated, but the procedure that allows the greatest removal of thymic tissue would be expected to be the preferred option. Thymectomy is not recommended in MuSK antibody positive patients because retrospective analyses indicate a lack of typical thymus pathology in these patients.Citation95 Whether a small proportion of seronegative MG patients do occasionally benefit from thymectomy remains controversial.

Thymectomy is always indicated in patients with thymoma to resect the neoplasm and treat the MG which occurs in about 10% of cases. Tumor removal in these cases does not always lead to remission and, indeed, the disease course in thymomatous patients is commonly more severe than in non-thymomatous patients. Local irradiation and chemotherapy may be needed in certain more aggressive thymoma cases. Long-term monitoring for recurrence with computed tomography or magnetic resonance imaging may be needed.

Emerging treatments

A number of potentially useful treatments show promise in animal modelsCitation96 and theoretically.Citation97

Animal models, such as the experimental autoimmune MG (EAMG) model which can be induced in different species, have been used to investigate several therapeutic possibilities. The most widely used models nowadays are rodents, and active immunization is with AChR from Torpedo electric organ which is a rich source of the receptor. In rats, both the acute and chronic phases of MG are present, as in humans, although the thymus is not involved.Citation98 EAMG in mice is harder to induce, but mouse models have the advantage of the availability of mouse-specific reagents and knock-out strains which allow analyses which cannot be carried out in rats.

Different derivatives of AChR have been tested as potential therapeutic agents in EAMG. The first successful antigen-specific immunomodulation of EAMG was performed in 1978 with an intradermal injection of a chemically modified Torpedo AChR, where the denatured AChR derivative both prevented the induction of EAMG and suppressed the ongoing disease in rabbits.Citation99 Native Torpedo AChR has also been shown to modulate EAMG,Citation100Citation102 but the highly immunogenic nature of this native AChRCitation103,Citation104 has hampered its therapeutic application.

Advances in cloning and genetic engineering have led to the discovery that a certain region of the AChR molecule, termed the main immunogenic region, is the target of a large portion of the antibodies to AChR.Citation105 Recombinant allogenic or syngeneic fragments to the appropriate regions of AChR have been shown to suppress acute and chronic manifestations of rat EAMG.Citation106Citation109 It is thought that this approach suppresses autoimmune MG by inducing tolerance toward the self-autoantigen, rather than the directly neutralizing antibodies reacting with the autoantigen.

The use of synthetic peptides against immunodominant T- and B-cell epitopes within the AChR molecule is another potentially useful approach in MG. Several studies using such peptides have been associated with the amelioration of or protection from EAMG in rodent models.Citation110Citation113 The autoimmune response in MG is polyclonal, thus peptides which represent only one or two dominant epitopes are unlikely to be effective. However, the use of peptide mixtures has been shown to be ineffective in rats with EAMG, possibly due to their inability to meet conformation requirements even when they are relatively long.Citation114

Several approaches to influence key players in the immune process such as regulatory T-cells,Citation115 dendritic cells,Citation116 cytokines,Citation117 costimulatory molecules,Citation118 and complement pathwaysCitation119 have also been shown to ameliorate or prevent EAMG.

Several techniques have been devised as alternatives to plasma exchange. These include immunoadsorption of anti- AChR antibodies using gels or columns.Citation120Citation122 However, the nonspecificity of these approaches leads to the indiscriminate removal of all, or most, immunoglobulins, including potentially useful antibodies. More antigen-specific immunoadsorbent approaches have being explored,Citation123Citation125 with mixed results.

Conclusion

There remain major gaps in our knowledge of the treatments used in generalized MG. Much of the evidence that we use in determining the choice of treatment derives from clinical experience, observational studies, and expert opinion. Treatment regimens vary among different physicians, even from the same country, because many decisions are made primarily on the experience and familiarity of the treating physician to a particular regime. Furthermore, organizing randomized controlled trials in MG is fraught with difficulties, not least that the rarity of the condition makes recruitment into trials a major obstacle. In addition, the phenotypical variability between AChR antibody positive, MuSK antibody positive and seronegative patients, and the fluctuating nature of MG are confounding factors in evaluating response to treatments. There is also a need to comparatively evaluate the effectiveness of current and emerging treatments in MG.

Various studies in experimental models of MG aim to develop novel and improved treatments that would have less adverse events than currently used treatments. Many promising ways to modulate the immune system in EAMG have been examined, but not all of these would necessarily be successful in human patients. It is worth noting that a multifaceted approach to the management of MG, with both antigen-specific immunotherapy and the direct modulation of key immunological factors involved in the autoimmune process, is likely to yield better treatments to improve the management of MG.

Disclosure

The author declares no conflicts of interests in this work.

References

  • GrobDBrunnerNNambaTPagalaMLifetime course of myasthenia gravisMuscle Nerve200837214114918059039
  • PhillipsLH2ndTornerJCEpidemiologic evidence for a changing natural history of myasthenia gravisNeurology1996475123312388909435
  • VincentAPalaceJHilton-JonesDMyasthenia gravisLancet200135792742122212811445126
  • GrobDArsuraELBrunnerNGNambaTThe course of myasthenia gravis and therapies affecting outcomeAnn N Y Acad Sci19875054724993318620
  • SchwarzHMestinon (pyridostigmine bromide) in myasthenia gravisCan Med Assoc J19567529810013343058
  • SimpsonJFWesterbergMRMageeKRMyasthenia gravis. An analysis of 295 casesActa Neurol Scand.196642Suppl 231275957970
  • MehndirattaMMPandeySKuntzerTAcetylcholinesterase inhibitor treatment for myasthenia gravisCochrane Database Syst Rev20112CD00698621328290
  • HatanakaYHNonresponsiveness to anticholinesterase agents in patients with MuSK-antibody-positive MGNeurology20056591508150916275854
  • SimonHEMyasthenia gravis: effect of treatment with anterior pituitary extractJAMA19351042320652066
  • GenkinsGKornfeldPOssermanKENambaTGrobDBrunnerNGThe use of ACTH and corticosteroids in myasthenia gravisAnn N Y Acad Sci19711833693744330763
  • PascuzziRMCoslettHBJohnsTRLong-term corticosteroid treatment of myasthenia gravis: report of 116 patientsAnn Neurol19841532912986721451
  • SghirlanzoniAPeluchettiDMantegazzaRFiacchinoFCornelioFMyasthenia gravis: prolonged treatment with steroidsNeurology19843421701746538004
  • CosiVCitterioALombardiMPiccoloGRomaniAErbettaAEffectiveness of steroid treatment in myasthenia gravis: a retrospective studyActa Neurol Scand199184133391927259
  • EvoliABatocchiAPPalmisaniMTLo MonacoMTonaliPLong- term results of corticosteroid therapy in patients with myasthenia gravisEur Neurol199232137431563453
  • BuBYangMXuJGaoBTPanDJCaiYXA prospective study of effectiveness and safety of long-term prednisone therapy in patients with myasthenia gravisZhonghua Shen Jing Ge Za Zhi2000332831 Chinese
  • HowardFMJrDuaneDDLambertEHDaubeJRAlternate-day prednisone: preliminary report of a double-blind controlled studyAnn N Y Acad Sci1976274596607183591
  • LindbergCAndersenOLefvertAKTreatment of myasthenia gravis with methylprednisolone pulse: a double blind studyActa Neurol Scand19989763703739669469
  • ZhangJWuHEffectiveness of steroid treatment in juvenile myasthenia gravisChinese Journal of Pediatrics19983610612614
  • WarmoltsJREngelWKBenefit from alternate-day prednisone in myasthenia gravisN Engl J Med1972286117204331158
  • SkeieGOApostolskiSEvoliAGuidelines for treatment of autoimmune neuromuscular transmission disordersEur J Neurol201017789390220402760
  • SathasivamSSteroids and immunosuppressant drugs in myasthenia gravisNat Clin Pract Neurol20084631732718493241
  • MatellGBergstromKFrankssonCEffects of some immunosuppressive procedures in myasthenia gravisAnn N Y Acad Sci1976274659676183592
  • MertensHGHertelGReutherPRickerKEffect of immunosuppressive drugs (azathioprine)Ann N Y Acad Sci19813776916996951493
  • WitteASCornblathDRParryGJLisakRPSchatzNJAzathioprine in the treatment of myasthenia gravisAnn Neurol19841566026056742794
  • KuksJBMDjojoatmodjoSOosterhuisHJHGAzathioprine myasthenia gravis: observations in 41 patients and a review of literatureNeuromuscul Disord1991164234311822354
  • MantegazzaRAntozziCPeluchettiDSghirlanzoniACornelioFAzathioprine as a single drug or in combination with steroids in the treatment of myasthenia gravisJ Neurol198823584494533062134
  • HeckmannJMLeePanEBEastmanRWHigh-dose immunosuppressive therapy in generalized myasthenia gravis – a 2-year follow-up studyS Afr Med J200191976577011680327
  • Myasthenia Gravis Clinical Study GroupA randomised clinical trial comparing prednisolone and azathioprine in myasthenia gravis. Results of a second interim analysisJ Neurol Neurosurg Psychiatry19935611115711638229026
  • PalaceJNewsom-DavisJLeckyBA randomized double-blind trial of prednisolone alone or with azathioprine in myasthenia gravisNeurology1998506177817839633727
  • GoulonMElkharratDLokiecFGajdosPResults of a one-year open trial of cyclosporine in ten patients with severe myasthenia gravisTransplant Proc.1988203 Suppl 42112173260050
  • BonifatiDMAngeliniCLong-term cyclosporine treatment in a group of severe myasthenia gravis patientsJ Neurol199724495425479352450
  • LavrnicDVujicARakocevic-StojanovicVCyclosporine in the treatment of myasthenia gravisActa Neurol Scand2005111424725215740576
  • TindallRSARollinsJAPhillipsJTGreenleeRGWellsLBelendiukGPreliminary results of a double-blind, randomized, placebo-controlled trial of cyclosporine in myasthenia gravisN Engl J Med1987316127197243547126
  • TindallRSPhillipsJTRollinsJAWellsLHallKA clinical therapeutic trial of cyclosporine in myasthenia gravisAnn N Y Acad Sci19936815395518357194
  • PerezMCBuotWLMercado-DanguilanCBababaldoZGRenalesLDStable remissions in myasthenia gravisNeurology198131132377192824
  • De FeoLGSchottlenderJMartelliNAMolfinoNAUse of intravenous pulsed cyclophosphamide in severe, generalized myasthenia gravisMuscle Nerve2002261313612115946
  • CosLMankodiAKTawilRThorntonCAMycophenolate mofetil (MyM) is safe and well tolerated in myasthenia gravis (MG)Neurology200054Suppl 3A137
  • CiafaloniEMasseyJMTucker-LipscombBSandersDBMycophenolate mofetil for myasthenia gravis: an open-label pilot studyNeurology2001561979911148243
  • ChaudhryVCornblathDRGriffinJWO’BrienRDrachmanDBMycophenolate mofetil: a safe and promising immunosuppressant in neuromuscular diseasesNeurology2001561949611148242
  • MeriggioliMNCiafaloniEAl-HaykKAMycophenolate mofetil for myasthenia gravis: an analysis of efficacy, safety, and tolerabilityNeurology200361101438144014638974
  • HehirMKBurnsTMAlpersJConawayMRSawaMSandersDBMycophenolate mofetil in AChR-antibody-positive myasthenia gravis: outcomes in 102 patientsMuscle Nerve201041559359820405499
  • SandersDBHartIKMantegazzaRAn international, phase III, randomized trial of mycophenolate mofetil in myasthenia gravisNeurology200871640040618434638
  • Muscle Study GroupA trial of mycophenolate mofetil with prednisone as initial immunotherapy in myasthenia gravisNeurology200871639439918434639
  • SandersDBSiddiqiZALessons from two trials of mycophenolate mofetil in myasthenia gravisAnn N Y Acad Sci2008113224925318567876
  • PhanCSandersDBSiddiqiZAMycophenolate mofetil in myasthenia gravis: the unanswered questionExpert Opin Pharmacother20089142545255118778192
  • ZebardastNPatwaHSNovellaSPGoldsteinJMRituximab in the management of refractory myasthenia gravisMuscle Nerve201041337537819852027
  • LindbergCBokaweraMRituximab for severe myasthenia gravis – experience from five patientsActa Neurol Scand2010122422522820199513
  • MaddisonPMcConvilleJFarrugiaMEThe use of rituximab in myasthenia gravis and Lambert-Eaton myasthenic syndromeJ Neurol Neurosurg Psychiatry2011In press
  • BlumSGillisDBrownHUse and monitoring of low dose rituximab in myasthenia gravisJ Neurol Neurosurg Psychiatry201182665966321071753
  • KonishiTYoshiyamaYTakamoriMClinical study of FK506 in patients with myasthenia gravisMuscle Nerve200328557057414571458
  • KonishiTYoshiyamaYTakamoriMSaidaTthe Japanese FK506 MG Study GroupLong-term treatment of generalised myasthenia gravis with FK506 (tacrolimus)J Neurol Neurosurg Psychiatry200576344845015716549
  • KawaguchiNYoshiyamaYNemotoYMunakataSFukutakeTHattoriTLow-dose tacrolimus treatment in thymectomised and steroid-dependent myasthenia gravisCurr Med Res Opin20042081269127315324529
  • PonsetiJMAzemJFortJMLong-term results of tacrolimus in cyclosporine- and prednisolone-dependant myasthenia gravisNeurology20056491641164315883336
  • PonsetiJMGamezJAzemJLopez-CanoMVilallongaRArmengolMTacrolimus for myasthenia gravis. A clinical study of 212 patientsAnn N Y Acad Sci2008113225426318096852
  • NagaishiAYukitakeMKurodaYLong-term treatment of steroiddependent myasthenia gravis patients with low-dose tacrolimusIntern Med200847873173618421189
  • MinamiNFukijiNDoiSFive-year follow-up with low-dose tacrolimus in patients with myasthenia gravisJ Neurol Sci20113001–2596221035148
  • ZhaoCBZhangXZhangHClinical efficacy and immunological impact of tacrolimus in Chinese patients with generalized myasthenia gravisInt Immunopharmacol201111451952421195813
  • NaganeYUtsugisawaKObaraDKondohRTerayamaYEfficacy of low-dose FK506 in the treatment of myasthenia gravis – a randomized pilot studyEur Neurol200553314615015900097
  • Fateh-MoghadamAWickMBesingerUGeursenRGHigh dose intravenous gammaglobulin for myasthenia gravisLancet1984183818488496200741
  • GajdosPOutinHDElkharratDHigh dose intravenous gammaglobulin for myasthenia gravisLancet1984183734064076198570
  • ArsuraELExperience with intravenous immunoglobulin in myasthenia gravisClin Immunol Immunopathol.1989532 Pt 2S1701792791345
  • van der MercheFGvan DoornPAThe current place of high-dose immunoglobulins in the treatment of neuromuscular disordersMuscle Nerve19972021361479040650
  • AchironABarakYMironSSarova-PinhasIImmunoglobulin treatment in refractory myasthenia gravisMuscle Nerve200023455155510716766
  • HilkevichODroryVEChapmanJKorczynADThe use of intravenous immunoglobulin as maintenance therapy in myasthenia gravisClin Neuropharmacol200124317317611391130
  • Perez-NellarJDominquezAMLlorens-FigueroaJAA comparative study of intravenous immunoglobulin and plasmapheresis preoperatively in myastheniaRev Neurol200133541341611727205
  • JensenPBrilVA comparison of the effectiveness of intravenous immunoglobulin and plasma exchange as preoperative therapy of myasthenia gravisJ Clin Neuromuscul Dis20089335235518344718
  • WolfeGIBarohnRJFosterBMMyasthenia Gravis-IVIG Study Group. Randomized, controlled trial of intravenous immunoglobulin in myasthenia gravisMuscle Nerve200226454955212362423
  • ZinmanLNgEBrilVIV immunoglobulin in patients with myasthenia gravis. A randomized controlled trialNeurology2007681183784117353471
  • GajdosPChevretSClairBTranchantCChastangCClinical trial of plasma exchange and high dose immunoglobulin in myasthenia gravisAnn Neurol19974167897969189040
  • RonagerJRavnborgMHermansenIVolstrupSImmunoglobulin treatment versus plasma exchange in patients with chronic moderate to severe myasthenia gravisArtif Organs2001251296797311843764
  • GajdosPChevretSToykaKIntravenous immunoglobulin for myasthenia gravisCochrane Database Syst Rev.20081CD00227718254004
  • GajdosPTranchantCClairBTreatment of myasthenia gravis exacerbation with intravenous immunoglobulin 1 g/kg versus 2 g/kg: a randomized double blind clinical trialArch Neurol200562111689169316286541
  • SathasivamSEvidence for use of intravenous immunoglobulin and plasma exchange in generalised mysasthenia gravisAdv Clin Neurosci Rehabil200991810
  • ThorlaciusSLefvertAKAarliJAPlasma exchange in myasthenia gravis: effect on anti-AChR antibodies and other autoantibodiesActa Neurol Scand19867464864903825504
  • YehJHChenWHChiuHCBaiCHMuSK antibody clearance during serial sessions of plasmapheresis for myasthenia gravisJ Neurol20072631–2191193
  • PinchingASPetersDKRemission of myasthenia gravis following plasma exchangeLancet1976280001373137663848
  • BehanPOShakirRASimpsonJABurnettAKAllanTLHaaseAGPlasma exchange combined with immunosuppressive therapy in myasthenia gravisLancet19792814043844089500
  • DauCResponse to plasmapheresis and immunosuppressive drug therapy in sixty myasthenia gravis patientsAnn N Y Acad Sci19813777007086951494
  • OlarteMRShoenfeldtRSPennASLovelaceRERowlandLPEffect of plasmapheresis in myasthenia gravisAnn N Y Acad Sci19813777257286951496
  • FornasariPMRivaGPiccoloGCosiVLombardiMShort and long-term clinical effects of plasma exchange in 33 cases of myasthenia gravisInt J Artif Organs1985831591624030134
  • HuoJTLongHLLuoXXuJZClinical study of double filtration plasmapheresis for treatment of refractory myasthenia gravisNan Fang Yi Ke DA Xue Xue Bao [Journal of Southern Medical University]2007273355357 Chinese
  • Lazo-LangnerAEspinosa-PoblanoITirado-CardenasNTherapeutic plasma exchange in Mexico: experience from a single institutionAm J Hematol2002701162111994977
  • Carandina-MaffeisRNucciAMarquesJFJrPlasmapheresis in the treatment of myasthenia gravis: retrospective study of 26 patientsArq Neuropsiquiatr.2004622B39139515273832
  • YucesanCArslanOAratMTherapeutic plasma exchange in the treatment of neuroimmunologic disorders: review of 50 casesTransfus Apher Sci200736110310717224307
  • QureshiAIChoudhryMAAkbarMSPlasma exchange versus intravenous immunoglobulin treatment in myasthenic crisisNeurology199952362963210025801
  • MantegazzaRBruzzoneERegiBSingle donor plasma in therapeutic exchange for myasthenia gravisInt J Artif Organs19871053153183679573
  • NagayasuTYamayoshiTMatsumotoKBeneficial effects of plasmapheresis before thymectomy on the outcome in myasthenia gravisJpn J Thorac Cardiovasc Surg20055312715724495
  • YehJHChenWHHuangKMChiuHCPrethymectomy plasmapheresis in myasthenia gravisJ Clin Apher200520421722116035101
  • GajdosPSimonNde Rohan-ChabotPRaphaelJCGoulonMLong-term effects of plasma exchange in myasthenia. Results of a randomized studyPresse Med198312159399426221247
  • YehJHChiuHCPlasmapheresis in myasthenia gravis. A comparative study of daily versus alternately daily scheduleActa Neurol Scand199999314715110100957
  • TrikhaISinghSGoyalVShuklaGBhasinRBehariMComparative efficacy of low dose daily versus alternate day plasma exchange in severe myasthenia gravis: a randomised trialJ Neurol2007254898999517694386
  • GronsethGSBarohnRJPractice parameter: thymectomy for autoimmune myasthenia gravis (an evidence-based review): report of the Quality Standards Subcommittee of the American Academy of NeurologyNeurology200055171510891896
  • GoldRSchneider-GoldRCurrent and future standards in treatment of myasthenia gravisNeurotherapeutics20085453554119019304
  • KumarVKaminskiHJTreatment of myasthenia gravisCurr Neurol Neurosci Rep2011111899620927659
  • LeiteMIStrobelPJonesMFewer thymic changes in MuSK antibody positive than in MuSK antibody-negative MGAnn Neurol200557344444815732104
  • SouroujonMCBrennerTFuchsSDevelopment of novel therapies for MG: studies in animal modelsAutoimmunity2010435–644646020298126
  • LagoumintzisGZisimopoulouPKordasGLazaridisKPoulasKTzartosSJRecent approaches to the development of antigen-specific immunotherapies for myasthenia gravisAutoimmunity2010435–643644520187712
  • MeinlEKlinkertWEWekerleHThe thymus in myasthenia gravis Changes typical for the human disease are absent in experimental autoimmune myasthenia gravis of the Lewis ratAm J Pathol1991139599510081951638
  • BartfeldDFuchsSSpecific immunosuppression of experimental autoimmune myasthenia gravis by denatured acetylcholine receptorProc Natl Acad Sci U S A197875840064010279016
  • WangZYQiaoJLinkHSuppression of experimental autoimmune myasthenia gravis by oral administration of acetylcholine receptorJ Neuroimmunol19934422092148505410
  • OkumuraSMcIntoshKDrachmanDBOral administration of acetylcholine receptor: effects on experimental myasthenia gravisAnn Neurol19943657047137979216
  • MaCGZhangGXXiaoBGLinkJOlssonTLinkHSuppression of experimental autoimmune myasthenia gravis by nasal administration of acetylcholine receptorJ Neuroimmunol199558151607537280
  • DrachmanDBOkumuraSAdamsRNMcIntoshKROral tolerance in myasthenia gravisAnn NY Acad Sci19967782582728610979
  • ShiFDBaiXFLiHLHuangYMVan der MeidePHLinkHNasal tolerance in experimental autoimmune myasthenia gravis (EAMG): induction of protective tolerance in primed animalsClin Exp Immunol199811135065129528890
  • TzartosSJLindstromJMMonoclonal antibodies used to probe acetylcholine receptor structure: localization of the main immunogenic region and detection of similarities between subunitsProc Natl Acad Sci U S A19807727557596153804
  • ImSHBarchanDFuchsSSouroujanMCSuppression of ongoing experimental myasthenia by oral treatment with an acetylcholine receptor recombinant fragmentJ Clin Invest1999104121723173010606626
  • BarchanDSouroujonMCImSHAntozziCFuchsSAntigen-specific modulation of experimental myasthenia gravis: nasal tolerization with recombinant fragments of the human acetylcholine receptor alpha- subunitProc Natl Acad Sci U S A199996148086809110393952
  • ImSHBarchanDFuchsSSouroujonMCMechanism of nasal tolerance induced by a recombinant fragment of acetylcholine receptor for treatment of experimental myasthenia gravisJ Neuroimmunol20001111–216116811063834
  • MaitiPKFefermanTImSHSouroujonMCFuchsSImmunosuppression of rat myasthenia gravis by oral administration of a syngeneic acetylcholine receptor fragmentJ Neuroimmunol20041521–211212015223243
  • WuBDengCGoluszkoEChristadossPTolerance to a dominant T cell epitope in the acetylcholine receptor molecule induces epitope spread and suppresses murine myasthenia gravisJ Immunol19971596301630239300727
  • KarachunskiPIOstlieNSOkitaDKGarmanRConti-FineBMSubcutaneous administration of T-epitope sequences of the acetylcholine receptor prevents experimental myasthenia gravisJ Neuroimmunol1999931–210812110378874
  • ShenoyMOshimaMAtassiMZChristadossPSuppression of experimental autoimmune myasthenia gravis by epitope-specific neonatal tolerance to synthetic region alpha 146–162 of acetylcholine receptorClin Immunol Immunopathol19936632302387679342
  • BaggiFAndreettaFCaspaniEOral administration of an immunodominant T-cell epitope downregulates Th1/Th2 cytokines and prevents experimental myasthenia gravisJ Clin Invest199910491287129510545527
  • SouroujonMCMaitiPKFefermanTImSHRavehLFuchsSSuppression of myasthenia gravis by antigen-specific mucosal tolerance and modulation of cytokines and costimulatory factorsAnn NY Acad Sci200399853353614592924
  • ShengJRLiLCGaneshBBPrabhakarBSMeriggioliMNRegulatory T cells induced by GM-CSF suppress ongoing experimental myasthenia gravisClin Immunol2008128217218018502693
  • YarilinDDuanRHuangYMXiaoBGDendritic cells exposed in vitro to TGF-beta1 ameliorate experimental autoimmune myasthenia gravisClin Exp Immunol2002127221421911876742
  • ImSHBarchanDMaitiPKRavehLSouroujonMCFuchsSSuppression of experimental myasthenia gravis, a B cell-mediated autoimmune disease, by blockade of IL-18FASEB J200115122140214811641240
  • ImSHBarchanDMaitiPKFuchsSSouroujonMCBlockade of CD40 ligand suppresses chronic experimental myasthenia gravis by down-regulation of Th1 differentiation and up-regulation of CTLA-4J Immunol2001166116893689811359850
  • SoltysJKusnerLLYoungANovel complement inhibitor limits severity of experimentally myasthenia gravisAnn Neurol2009651677519194881
  • YamazakiZFujimoriYTakahamaTEfficiency and biocompatibility of a new immunosorbentTrans Am Soc Artif Intern Organs1982283183237164255
  • MaticGWinklerRETiessMRamlowWSelective apheresis-time for a changeInt J Artif Organs20012414711266041
  • PtakJChanges of plasma proteins after immunoadsorption using Ig-Adsopak columns in patients with myasthenia gravisTransfus Apher Sci200430212512915062750
  • TakamoriMMarutaTImmunoadsorption in myasthenia gravis based on specific ligands mimicking the immunogenic sites of the acetylcholine receptorTher Apher20015534035011778918
  • GuoCYLiZYXuMQYuanJMPreparation of an immunoadsorbent coupled with a recombinant antigen to remove anti-acetylcholine receptor antibodies in abnormal serumJ Immunol Methods20053031–214214716040047
  • ZisimopoulouPLagoumintzisGPoulasKTzartosSAntigen-specific apheresis of human anti-acetylcholine receptor autoantibodies from myasthenia gravis patients’ sera using Escherichia coli-expressed receptor domainsJ Neuroimmunol20082001–213314118603305