Skip to main content
Advertisement
  • Neurology.org
  • Journals
    • Neurology
    • Clinical Practice
    • Genetics
    • Neuroimmunology & Neuroinflammation
    • Education
  • Online Sections
    • COVID-19
    • Inclusion, Diversity, Equity, Anti-racism, & Social Justice (IDEAS)
    • Innovations in Care Delivery
    • Practice Buzz
    • Practice Current
    • Residents & Fellows
    • Without Borders
  • Collections
    • Topics A-Z
    • Disputes & Debates
    • Health Disparities
    • Infographics
    • Patient Pages
    • Null Hypothesis
    • Translations
  • Podcast
  • CME
  • About
    • About the Journals
    • Contact Us
    • Editorial Board
  • Authors
    • Submit a Manuscript
    • Author Center

Advanced Search

Main menu

  • Neurology.org
  • Journals
    • Neurology
    • Clinical Practice
    • Genetics
    • Neuroimmunology & Neuroinflammation
    • Education
  • Online Sections
    • COVID-19
    • Inclusion, Diversity, Equity, Anti-racism, & Social Justice (IDEAS)
    • Innovations in Care Delivery
    • Practice Buzz
    • Practice Current
    • Residents & Fellows
    • Without Borders
  • Collections
    • Topics A-Z
    • Disputes & Debates
    • Health Disparities
    • Infographics
    • Patient Pages
    • Null Hypothesis
    • Translations
  • Podcast
  • CME
  • About
    • About the Journals
    • Contact Us
    • Editorial Board
  • Authors
    • Submit a Manuscript
    • Author Center
  • Home
  • Latest Articles
  • Current Issue
  • Past Issues
  • Residents & Fellows

User menu

  • Subscribe
  • My Alerts
  • Log in
  • Log out

Search

  • Advanced search
Neurology
Home
The most widely read and highly cited peer-reviewed neurology journal
  • Subscribe
  • My Alerts
  • Log in
  • Log out
Site Logo
  • Home
  • Latest Articles
  • Current Issue
  • Past Issues
  • Residents & Fellows

Share

August 05, 2008; 71 (6) Articles

An international, phase III, randomized trial of mycophenolate mofetil in myasthenia gravis

D. B. Sanders, I. K. Hart, R. Mantegazza, S. S. Shukla, Z. A. Siddiqi, M.H.V. De Baets, A. Melms, M. W. Nicolle, N. Solomons, D. P. Richman
First published April 23, 2008, DOI: https://doi.org/10.1212/01.wnl.0000312374.95186.cc
D. B. Sanders
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
I. K. Hart
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
R. Mantegazza
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
S. S. Shukla
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Z. A. Siddiqi
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
M.H.V. De Baets
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
A. Melms
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
M. W. Nicolle
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
N. Solomons
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
D. P. Richman
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Full PDF
Citation
An international, phase III, randomized trial of mycophenolate mofetil in myasthenia gravis
D. B. Sanders, I. K. Hart, R. Mantegazza, S. S. Shukla, Z. A. Siddiqi, M.H.V. De Baets, A. Melms, M. W. Nicolle, N. Solomons, D. P. Richman
Neurology Aug 2008, 71 (6) 400-406; DOI: 10.1212/01.wnl.0000312374.95186.cc

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Permissions

Make Comment

See Comments

Downloads
1560

Share

  • Article
  • Figures & Data
  • Info & Disclosures
Loading

Abstract

Background: This prospective, randomized, double-blind, placebo-controlled, phase III trial assessed the efficacy, safety, and tolerability of mycophenolate mofetil (MMF) as a steroid-sparing agent in patients with myasthenia gravis (MG).

Methods: Patients with acetylcholine receptor antibody-positive class II-IVa MG (MG Foundation of America [MGFA] criteria) taking corticosteroids for at least 4 weeks were randomized to MMF (2 g/day) or placebo for 36 weeks. The primary endpoint was a composite measure defined as achievement of minimal manifestations or pharmacologic remission (MGFA post-intervention status), with reduction of corticosteroid dose on a set schedule. Secondary endpoints included disease severity, quality-of-life scores, and safety.

Results: A total of 44% of MMF-treated (n = 88) and 39% of placebo-receiving (n = 88) patients achieved the primary endpoint (p = 0.541). Improvements in mean quantitative MG, MG activities of daily living, and 36-item Short-Form health survey scores were similar in both groups. Numbers of adverse events were similar in both groups. The most commonly reported adverse events in the MMF-treated group were headache (12.5%) and worsening of MG (11.4%), and in the placebo group, worsening of MG (20.5%) and diarrhea (10.2%).

Conclusions: Initiation of mycophenolate mofetil (MMF) treatment was not superior to placebo in maintaining myasthenia gravis (MG) control during a 36-week schedule of prednisone tapering. There were no significant differences in the primary or secondary endpoints between the study groups. MMF was well tolerated and adverse events were consistent with previous studies. Experience from this large, international, multicenter, phase III study employing full MG Foundation of America guidelines will aid the design of future MG studies.

Glossary

AChR=
acetylcholine receptor;
AUC=
area under the time/dose curve;
ITT=
intention-to-treat;
MG=
myasthenia gravis;
MG-ADL=
MG Activities of Daily Living;
MGFA=
MG Foundation of America;
MM=
minimal manifestations;
MMF=
mycophenolate mofetil;
PR=
pharmacologic remission;
QMGS=
quantitative MG score;
QoL=
quality of life;
SF-36=
Short-Form 36.

Myasthenia gravis (MG) is an antibody-mediated, T cell-dependent autoimmune disorder that targets the acetylcholine receptor (AChR) at the neuromuscular junction in 85–90% of patients. The use of corticosteroids and immunosuppressants such as azathioprine has transformed MG management.1 However, many patients do not tolerate or respond sufficiently to these drugs.2 In addition, corticosteroids—the main current therapy for generalized MG3—have formidable side effects. These problems have prompted researchers to look at newer immunosuppressants,4 including mycophenolate mofetil (MMF).

The first report of MMF use in MG was its rapid induction of pharmacologic remission in a patient refractory to prednisone plus azathioprine or cyclosporine.5 The efficacy and safety of MMF in patients with MG have since been evaluated in case reports,6–11 open-label prospective12,13 and retrospective cohort14–17 studies, and one double-blind randomized pilot study.18,19 However, no international, large, randomized controlled trial of MMF—or of any other immunosuppressive drug—has been performed in MG.20,21

Here we report the results of a phase III trial designed to study the efficacy and safety of MMF as a steroid-sparing adjunct in patients with mild-to-moderate, generalized, AChR antibody-positive MG. We also discuss methodologic and practical issues arising from one of the largest ever therapeutic trials in MG and implications for future studies.

METHODS

This prospective, randomized, double-blind, placebo-controlled, parallel-group, international, multicenter, 36-week trial assessed the efficacy and safety of adjunct MMF (CellCept, Roche Pharma AG, Grenzach-Wyhlen, Germany) in improving disease control while reducing corticosteroid dose in patients with MG. The study ran from August 2004 to October 2006. Initially, patients were recruited at 25 neurology centers in the United States, Canada, United Kingdom, Germany, and The Netherlands. In 2005, a further 18 centers in Ukraine, Israel, Spain, India, Italy, France, Russia, Serbia and Montenegro, and Mexico were added because of difficulties in recruiting. All patients gave written informed consent to participate in the study, which was conducted in accordance with the Declaration of Helsinki and the principles of Good Clinical Practice.

Patients.

All patients had a diagnosis of generalized MG (MGFA class II, III, or IVa22) with elevated AChR antibodies, and were treated with ≥20 mg/day oral prednisone (or equivalent alternate day dose) for at least 4 weeks before randomization, without any other immunosuppressive therapy. Full details of the inclusion and exclusion criteria are in the e-appendix on the Neurology® Web site at www.neurology.org.

Objectives and outcome measures.

The overall objectives were to assess the efficacy (good disease control with steroid sparing), safety, and tolerability of MMF compared with placebo in patients with MG receiving prednisone. A novel composite primary endpoint was developed to be meaningful to clinical practice and included outcome measures recommended by the MGFA Task Force on clinical research standards in MG.22 Patients were defined as having a treatment response if they met all three efficacy criteria: 1) MGFA post-intervention status of minimal manifestations (MM) or pharmacologic remission (PR)22 for weeks 32–36; 2) 7.5 mg/day (or alternate day equivalent) prednisone for weeks 32–36; and 3) ≤120 mg/day pyridostigmine for weeks 33–36. The last criterion ensured that higher doses of pyridostigmine could not mask continuing, marked MG weakness. It also obviated physician concerns about tapering prednisone in patients still needing high doses of pyridostigmine to control MG.

Secondary efficacy outcomes included 1) time to initial treatment response—the first visit week when a patient met the efficacy criteria and maintained this status at the next visit; 2) time to sustained treatment response—the first visit week when a patient met all efficacy criteria and maintained this status until week 36; 3) prednisone area under the time/dose curve (AUC); 4) prednisone dose at study termination; 5) cholinesterase inhibitor dose at study termination; 6) number of IV immunoglobulin and plasma exchange treatments received during the study; 7) change from baseline in quantitative MG score (QMGS)22,23; 8) change from baseline in quality of life (QoL) assessed using the Short-Form 36 (SF-36) health survey and the MG Activities of Daily Living (MG-ADL) scale24; 9) change from baseline in investigator and patient Global Assessment of disease severity; and 10) AChR antibody titers. Patients also completed a healthcare utilization questionnaire.

Safety was assessed by the incidence, severity, and relation of adverse events to treatment (probable, possible, or unknown), and the incidence of serious adverse events and adverse events leading to study withdrawal.

Treatments.

The protocol included schedules to taper prednisone and any cholinesterase inhibitor treatment to a minimum level.

Investigational drug.

Randomized patients received prednisone (or if unavailable, prednisolone) plus either oral MMF 2 g/day or matching placebo tablets for 36 weeks. Two 500-mg tablets of trial medication were taken morning and evening, on an empty stomach, with a glass of water. If a gastrointestinal disturbance occurred, the regimen could be changed to one 500-mg tablet four times daily, with or without food.

Tapering of corticosteroid.

From week 2, the prednisone dose was tapered at each visit when patients had achieved MM or PR, with the target of reaching a prednisone dose of 7.5 mg/day by week 24 (see appendix e-1). Patients were withdrawn from the study if prednisone could not be reduced below the baseline dose by week 16. These withdrawn patients were deemed nonresponders for all analyses.

Tapering of cholinesterase inhibitor.

Patients taking cholinesterase inhibitors must have been on a stable regimen for ≥2 weeks before study entry. This dose was maintained until patients achieved MM and the dose of prednisone was reduced to 7.5 mg/day. Thereafter, the cholinesterase inhibitor dose was reduced to ≤120 mg/day over 1–7 days.

Randomization and blinding.

Randomization was performed by a central, computerized interactive voice response system. Each patient was assigned a randomization number and corresponding bottles of investigational drug or placebo. All study personnel and patients were blinded to the study medication and the randomization code key set was not available at the study center, or to the study monitors, project statisticians, or study sponsor. The medical monitor had access to sealed emergency code keys, which could be broken in an emergency (e.g., an adverse event or pregnancy) upon request by the investigator and agreement by the monitor.

Assessments.

MM and PR were assessed by site personnel at weeks 2, 4, 8, 12, 16, 20, 24, 28, 32, and 36. The QMGS, MG-ADL, SF-36, and Global Assessment were performed at baseline and weeks 2, 12, 24, and 36. Serum AChR antibody titers were measured at screening, baseline, and weeks 12, 24, and 36.

Safety was evaluated by clinical assessment, electrocardiograms, hematology and blood chemistry, and vital signs at screening, baseline, and weeks 2, 4, 8, 12, 16, 20, 24, 28, 32, and 36.

Statistical analysis.

Available data2,12,14,17 and a survey of MG specialists led to the prediction that 35% of patients in the placebo group and at least 65% in the MMF group would reach treatment response. Power calculations using the two-sided Fisher exact test with a significance level of 0.05 determined that 136 patients—68 in each treatment arm—would provide 92% power to detect a 30% difference in treatment response rate between study groups. Efficacy analyses were performed using the intention-to-treat (ITT) population. The randomized population, the ITT population, and the safety population each consisted of the same 176 patients (88 in each treatment arm).

For the primary endpoint, the proportion of patients who achieved the defined treatment response in the two study groups were compared using the two-sided Fisher exact test with a 0.05 level of significance. Patients who did not continue to week 36 were considered nonresponders. Logistic regression analysis was performed to adjust for covariate factors potentially prognostic of outcome: age at MG onset (<40 years and ≥40 years), disease duration, sex, previous thymectomy, and the interactions of treatment with these factors.

For all secondary efficacy endpoints, descriptive analyses were performed by assessment at each visit and at study termination, and included calculation of 95% confidence limits for means and difference between means. If a patient was evaluated at early termination, analyses included the observed cases (no imputation); missing data were imputed using the last observation carried forward method for the week 36 visit.

Time to the defined treatment response was summarized using the Kaplan-Meier analysis. Mean prednisone and cholinesterase inhibitor doses were calculated for the total study duration (weeks 0–36) and by interval (weeks 0–2, 2–4, 4–8, 8–12, 12–16, 16–20, 20–24, 24–28, 28–32, and 32–36). To compare total cumulative prednisone dose between the two treatment groups, mean prednisone AUC was calculated over the total study duration and by interval (weeks 0–12, 12–24, and 24–36).

All patients who had at least one dose of trial medication and a post-baseline safety assessment, whether they withdrew from the study or not, were included in the safety analysis. Missing safety data were not imputed.

RESULTS

Patients.

Of the 176 patients enrolled, 88 (50%) were randomized to the MMF group and 88 (50%) to placebo. A total of 144 patients (81.8%) completed the study: 73 (83.0%) in the MMF and 71 (80.7%) in the placebo group (figure 1).

Figure
  • Download figure
  • Open in new tab
  • Download powerpoint

Figure 1 Patient flow and reasons for withdrawal throughout the study

*All patients who were randomized received the allocated intervention and were included in the intention-to-treat analysis of efficacy. IVIg = IV immunoglobulin; MMF = mycophenolate mofetil; PE = plasma exchange.

There were no substantial differences in patient demographics or disease characteristics between the two treatment groups at baseline (table 1). Exposure to study drug after randomization, analyzed to assess compliance, was similar in the MMF group (treatment duration 229.7 ± 71.9 days; mean daily dose 1.97 ± 0.1 g/day) and the placebo group (236.0 ± 48.2 days; 1.97 ± 0.2 g/day).

View this table:
  • View inline
  • View popup
  • Download powerpoint

Table 1 Baseline patient demographics and disease characteristics in the mycophenolate mofetil (MMF) and placebo treatment groups (intention-to-treat population)

Responses to treatment.

Both MMF and placebo groups responded to treatment. There were, however, no significant differences in any of the primary or secondary measures of response between the two groups. The primary endpoint of treatment response was achieved by 39 patients (44.3%) in the MMF group compared with 34 patients (38.6%) in the placebo group (p = 0.541; ITT population). On regression analyses, this outcome was unaffected by sex, age at disease onset, duration of MG, previous thymectomy, or center effects. The reasons why patients failed to achieve treatment response are given in table 2. Results across the two treatment groups for the secondary endpoints of QMGS (figure 2), ADL (figure e-1), and investigator and patient global assessments (figure e-2) are summarized in table 3. All were similar and consistent with those obtained for the primary endpoint.

View this table:
  • View inline
  • View popup
  • Download powerpoint

Table 2 Reasons for failure to achieve a treatment response in the mycophenolate mofetil (MMF) and placebo groups (intention-to-treat population)

Figure
  • Download figure
  • Open in new tab
  • Download powerpoint

Figure 2 Mean total Quantitative Myasthenia Gravis Score (QMGS) in the mycophenolate mofetil (MMF) and placebo groups

The endpoint includes the week 36 and early termination visits. Early termination analyses included the observed cases (no imputation) and missing data imputed using the last observation carried forward method for the week 36 visit.

View this table:
  • View inline
  • View popup
  • Download powerpoint

Table 3 Responses to treatment: Efficacy results for the mycophenolate mofetil (MMF) and placebo groups on secondary outcome measures (intention-to-treat population)

Reductions in prednisone and cholinesterase inhibitor doses.

There were no significant differences in the mean prednisone and cholinesterase inhibitor doses between the two groups. The mean prednisone dose in the MMF group decreased from 30.7 ± 12.6 mg/day during weeks 0–2 to 10.1 ± 6.2 mg/day during weeks 32–36, and in the placebo group from 34.1 ± 15.0 mg/day during weeks 0–2 to 11.6 ± 9.5 mg/day during weeks 32–36 (figure e-3A). The mean cholinesterase inhibitor dose in the MMF group decreased from 214.9 ± 147.7 mg/day during weeks 0–2 to 142.1 ± 140.9 mg/day during weeks 32–36, and in the placebo group from 237.2 ± 167.2 mg/day during weeks 0–2 to 168.0 ± 143.5 mg/day during weeks 32–36 (figure e-3B). The reductions in mean prednisone dose were also seen in total cumulative dose results (as determined by the prednisone AUC) for both treatment groups: cumulative prednisone dose decreased in the MMF group from 1,974.6 ± 944.9 mg during weeks 0–12 to 856.6 ± 472.9 mg during weeks 24–36, and in the placebo group from 2,283.2 ± 1010.0 mg during weeks 0–12 to 968.1 ± 656.8 mg during weeks 24–36 (figure e-4).

AChR antibody titers.

Mean AChR antibody titers were similar at baseline in the MMF (3.5 ± 3.5 nmol/L) and placebo (3.1 ± 3.9 nmol/L) groups. These titers fell over time in both the MMF (endpoint: 2.6 ± 4.1 nmol/L) and placebo (endpoint: 2.8 ± 4.3 nmol/L) groups (figure e-5).

Safety.

Seventy-one patients (80.7%) in the MMF group and 74 patients (84.1%) in the placebo group had at least one adverse event during the study. The most frequent adverse events (table 4) in the MMF and placebo groups were headache (12.5% vs 6.8%), MG worsening reported as an adverse event (11.4% vs 20.5%), and nausea (9.1% vs 6.8%). The number of withdrawals owing to adverse events was low and similar in both treatment groups: 3 patients (3.4%) in the MMF group (1 lymphopenia; 1 diarrhea, nausea, and vomiting; 1 chronic bronchitis) and 4 (4.5%) in the placebo group (2 worsening of MG; 2 increased hepatic enzyme levels).

View this table:
  • View inline
  • View popup
  • Download powerpoint

Table 4 Most frequently occurring treatment-emergent adverse events (occurring in ≥5% of patients) and serious adverse events in the mycophenolate mofetil (MMF) and placebo groups

Serious adverse events occurred in 19 patients (21.6%) in the MMF group and 14 (15.9%) in the placebo group (table 4). Serious infections were more frequent with MMF (7 patients [8.0%]) than placebo (3 patients [3.4%]). Serious gastrointestinal disorders, however, were reported more frequently with placebo (3 patients [3.4%]) than MMF (1 patient [1.1%]), as was MG worsening, which occurred in 7 patients (8.0%) in the placebo group and 2 patients (2.3%) in the MMF group.

Three deaths occurred: two while the patients were enrolled in the study, and one 11 days after study completion (this patient was considered to be well at study completion). Two deaths were considered unrelated to study medication: hemorrhagic fever occurring after study completion (MMF) and alcohol intoxication (placebo). The other death—from pneumonia in the MMF group—was considered possibly related to treatment.

Healthcare utilization.

Healthcare utilization was similar between the two treatment groups. The 11 patients (12.5%) in the MMF group who required hospitalization had a mean stay of 9.7 ± 10.6 days, with 5.1 ± 11.5 days spent in intensive or critical care units. The 9 patients (10.2%) in the placebo group who required hospitalization had a mean stay of 19.0 ± 12.4 days, with 3.7 ± 7.0 days spent in intensive or critical care units.

DISCUSSION

Patients in the MMF group showed similar improvements to those in the placebo group on all efficacy measures examined. Although at first sight some data on softer endpoints, such as prednisone dose reduction, decline in AChR antibody titers, and mean length of hospitalization, seemed to suggest potential benefits of MMF over placebo, these differences were not significant. Thus, prednisone plus MMF for 36 weeks was not superior to prednisone alone in managing the selected study population of mild-to-moderate, AChR antibody-positive, generalized MG.

One possible conclusion is that MMF is ineffective as a steroid-sparing agent in the treatment of MG, contradicting evidence from smaller, less rigorous studies and the reported experience of many clinicians.6–19 Several factors could account for this incongruity.

Treatment response was achieved by only 44% of patients in the MMF group, which is strikingly lower than the 70–75% response rate reported in a review of the use of MMF in MG.2 One reason for the low response rate may be that our definition of treatment response was so rigorous that it masked lesser but clinically useful improvements.

The initiation of the steroid taper, as early as 2 weeks in patients who had achieved MM or PR by that time, may have been too early for a clinical effect of MMF to become apparent. Also, the study period of 36 weeks may have been too short, particularly as the proportion of patients who responded increased with duration of treatment and did not appear to have reached a plateau at week 36 in either group (figure e-6). An open-label pilot study of MMF in MG reported initial improvement at 2–8 weeks,12 although a longer average of 10.8 weeks (range 4–40 weeks) was reported from a retrospective analysis of 85 patients.17 Treatment response to other immunosuppressants used in MG can take even longer. For example, azathioprine took more than 18 months to maximally benefit patients with MG in a trial of prednisolone vs azathioprine plus prednisolone.25 In patients with mild disease in whom prednisone had been recently started, improvement may have been prednisone-related, making it hard to demonstrate additional benefits from MMF. Longer-term trials would be needed to demonstrate delayed initial responses and to study the long-term effects on maintenance of disease control and prevention of relapse months after prednisone discontinuation.

Our study entry criteria favored inclusion of older patients, those with milder disease, and (given the warnings against MMF use in pregnancy26) men, and thus may have selected an unrepresentative MG population less likely to respond to MMF. They also had MG for an average of 3 years (by which time MG is usually well-controlled) and had been treated with differing doses and durations of corticosteroids before entering the study. Data are unavailable to stratify our results by prednisone dose before randomization to assess potential confounding effects of different pre-randomization prednisone regimens.

Our study was designed with the expectation of detecting at least a 30% difference in treatment response rate between the MMF and placebo groups. Smaller but still clinically meaningful differences may have been missed; their detection would require an even larger sample size than the 136 patients needed to power our study.

A further reason why our study failed to confirm a steroid-sparing effect of MMF may lie in the unexpected finding that the prednisone dose could be tapered relatively rapidly to 7.5 mg/day and remain effective for the duration of the study in many placebo-group patients. This target prednisone dose was chosen as it is considered to be below the level where chronic use would severely compromise bone mineral density.27 The effect of MMF may have been masked by this corticosteroid dose and, potentially, cholinesterase inhibitors. This finding suggests that the current prednisone doses used in clinical practice may be higher than needed.

MMF was well tolerated with a safety profile consistent with that found in previous studies, except for a lower than expected incidence of diarrhea.26,28

The completion of this study confirms that a large, international, multicenter, phase III study can be successfully performed in a rare, orphan autoimmune neurologic disease. As the first such drug trial in MG to incorporate MGFA recommendations22 on clinical research standards in full, it should aid the design of future studies of MG immunotherapies.

ACKNOWLEDGMENT

The authors thank Tom Potter and Sam Coates of Caudex Medical (supported by Aspreva Pharmaceuticals) for their editorial assistance with the preparation of the article.

Appendix

Investigators: O. Abramsky, MD, Neurology Department, Hadassah Medical Organization, Jerusalem, Israel; S. Apostolski, MD, Institute of Neurology, University of Belgrade School of Medicine, Serbia and Montenegro; R. Barohn, MD, Department of Neurology at the Landon Center on Aging, University of Kansas Medical Center, Kansas City; A. Berthele, MD, Klinik und Poliklinik für Neurologie der Technischen Universitaet Muenchen, Klinikum Rechts der Isar, Muenchen, Germany; J.L.M. Blanco, MD, Neurology Department, Hospital General Universitario Gregorio Maranon, Madrid, Spain; E. Bogdanov, MD, Neurology and Rehabilitation Cathedra, Kazan State Medical University, Kazan, Russia; E. Ciafaloni, MD, Department of Neurology, University of Rochester Medical Center, NY; M. de Baets, MD, Department of Neurology, Academical Hospital Maastricht, the Netherlands; C. Desnuelle, MD, Federation Maladies Neuromusculaires, Hopital de l’Archet 1, Nice, France; S. Dwivedee, MD, Department of Neurology, Vidysagar Institute of Mental Health and Neurological Sciences, New Delhi, India; M. Freimer, MD, Department of Neurology, Ohio State University, Columbus; G. Garcia Ramos, MD, Instituto Nacional de Ciencias Médicas y Nutricion “Salvador Zubirán,” Mexico City, Mexico; A. Gustov, MD, Regional Hospital/State Educational Institution (postgraduate medical education) “Nizhniy Novgorod State Medical Academy,” Nizhniy Novgorod, Russia; I.K. Hart, FRCP, University Division of Neuroscience, The Walton Centre for Neurology and Neurosurgery, Liverpool, UK; D. Hilton-Jones, MD, University Department of Clinical Neurology, John Radcliffe Hospital, Oxford, UK; J. Howard Jr., MD, Department of Neurology, University of North Carolina, Chapel Hill; I. Illa, MD, Neurology Department, Hospital Ste. Creu i Saint Pau, Barcelona, Spain; M. Inghilleri, MD, Dipartimento di Scienze Neurologiche, Universita degli Studi “La Sapienza,” Rome, Italy; S. Jander, MD, Neurologische Klinik, Heinrich-Heine-Universitat, Düsseldorf, Germany; A. Kozelkin, MD, School of Neurology, Zaporozhye State Medical University, Zaporozhye, Ukraine; G. le Masson, MD, Service de Neurologie 2a2, Hopital Pellegrin/CHU Bordeaux, France; R. Malamut, MD, Neurological Associates of Delaware Valley, CCMC Ambulatory Care, Upland, PA; R. Mantegazza, MD, Department of Neurology IV, Neurological Institute “Carlo Besta” Foundation, Milan, Italy; A.K. Meena, MD, Department of Neurology, Nizam’s Institute of Medical Sciences, Hyderabad, India; M.M. Mehndiratta, MD, Neurology, GB Pant Hospital, New Delhi, India; A. Melms, MD, Neurologische Klinik, Universität Tübingen, Germany; G. Meola, MD, Dipartimento di Neurologia, Policlinico san Donato, San Donato Milanese, Milan, Italy; M. Nicolle, MD, London Health Sciences Centre, University of Western Ontario, London, Canada; S. Ravat, MD, Department of Neurology, GS Medical College and KEM Hospital, Mumbai, India; D. Richman, MD, Department of Neurology and the Center for Neuroscience, University of California, Davis; S. Rivera Nava, MD, private clinic, Guanajuato, Mexico City, Mexico; A. Sanadze, MD, Institute of General Pathology, Russian Academy of Medical Sciences, Moscow, Russia; D.S. Saperstein, MD, Department of Neurology at the Landon Center on Aging, University of Kansas Medical Center, Kansas City; B. Schalke, MD, Klinik und Poliklinik für Neurologie der Universitat Regensburg, Germany; K. Sharma, MD, Department of Neurology, University of Miami School of Medicine, FL; M. Shevnyuk, MD, 1 Bohoutivska Str., Kiev, Ukraine; Z. Siddiqi, MD, Department of Medicine, University of Alberta, Edmonton, Canada; N. Spirin, MD, Department of Neurology at the City Clinical Hospital No. 8, Yaroslavl State Medical Academy, Russia; J. Sussman, MD, Department of Neurology at the Greater Manchester Neuroscience Centre, Hope Hospital, Salford, UK; A. Syal, MD, Arizona Neurological Institute, Sun City; P. van Doorn, MD, Department of Neurology, Erasmus MC, Rotterdam, The Netherlands; J. Verschuuren, MD, Department of Neurology, Leiden University Medical Centre, the Netherlands; P.V. Voloshyn, MD, Institute of Neurology, Psychiatry and Narcology, Ams of Ukraine, Kharkiv, Ukraine; G. Wolfe, MD, Department of Neurology, The University of Texas Southwestern Medical Center at Dallas.

Footnotes

  • Embedded Image

  • Supplemental data at www.neurology.org

    Editorial, page 390

    See also page 394

    e-Pub ahead of print on April 23, 2008, at www.neurology.org.

    Sponsored by F Hoffmann-La Roche Ltd./Inc./AG as part of the Aspreva Pharmaceuticals Corp. Rare Disease Program.

    Disclosures: Dr. Sanders has received consultation fees from Sanofi-Aventis and Accordant Health Services. He is on the speaker’s program for Athena Diagnostics and has received research support through his institution from Roche Laboratories and Aspreva Pharmaceuticals. Drs. De Baets and Hart have received grants from F Hoffmann-La Roche Ltd./Inc./AG as part of the Aspreva Pharmaceuticals Corp. Rare Disease Program. Drs. Mantegazza, Melms, and Richman have received honoraria from F Hoffmann-La Roche Ltd./Inc./AG as part of the Aspreva Pharmaceuticals Corp. Rare Disease Program. Drs. Shukla and Solomons are employees of Aspreva Pharmaceuticals. Drs. Nicolle and Siddiqi have nothing to disclose.

    Received July 27, 2007. Accepted in final form January 7, 2008.

REFERENCES

  1. 1.↵
    Fonseca V, Havard CW. The natural course of myasthenia gravis. BMJ 1990;300:1409–1410.
    OpenUrlFREE Full Text
  2. 2.↵
    Ciafaloni E. Mycophenolate mofetil and myasthenia gravis. Lupus 2005;14(suppl 1):46–49.
    OpenUrl
  3. 3.↵
    Skeie GO, Apostolski S, Evoli A, et al. Guidelines for the treatment of autoimmune neuromuscular transmission disorders. Eur J Neurol 2006;13:691–699.
    OpenUrlCrossRefPubMed
  4. 4.↵
    Rozsa C, Lovas G, Fornadi L, Szabo G, Komoly S. Safety of long-term combined immunosuppressive treatment in myasthenia gravis: analysis of adverse effects of 163 patients. Eur J Neurol 2006;13:947–952.
    OpenUrlPubMed
  5. 5.↵
    Hauser RA, Malek AR, Rosen R. Successful treatment of a patient with severe refractory myasthenia gravis using mycophenolate mofetil. Neurology 1998;51:912–913.
    OpenUrlFREE Full Text
  6. 6.↵
    Caponnetto C, Rossi E, Primavera A. Mycophenolate mofetil: a new immunosuppressive approach. Successful treatment in a case of myasthenia gravis associated with incomplete lupus erythematosus syndrome and hepatitis C virus infection. Eur Neurol 2001;46:53–54.
    OpenUrlCrossRefPubMed
  7. 7.
    Kotani A, Takahashi A, Koga H, et al. Myasthenia gravis after allogeneic bone marrow transplantation treated with mycophenolate mofetil monitored by peripheral blood OX40+ CD4+ T cells. Eur J Haematol 2002;69:318–320.
    OpenUrlPubMed
  8. 8.
    Meriggioli MN, Rowin J. Treatment of myasthenia gravis with mycophenolate mofetil: a case report. Muscle Nerve 2000;23:1287–1289.
    OpenUrlCrossRefPubMed
  9. 9.
    Mowzoon N, Sussman A, Bradley WG. Mycophenolate (CellCept) treatment of myasthenia gravis, chronic inflammatory polyneuropathy and inclusion body myositis. J Neurol Sci 2001;185:119–122.
    OpenUrlCrossRefPubMed
  10. 10.
    Prakash KM, Ratnagopal P, Puvanendran K, Lo YL. Mycophenolate mofetil as an adjunctive immunosuppressive therapy in refractory myasthenia gravis: The Singapore experience. J Clin Neurosci 2007;14:278–281.
    OpenUrlPubMed
  11. 11.
    Schneider C, Gold R, Reiners K, Toyka KV. Mycophenolate mofetil in the therapy of severe myasthenia gravis. Eur Neurol 2001;46:79–82.
    OpenUrlCrossRefPubMed
  12. 12.↵
    Ciafaloni E, Massey JM, Tucker-Lipscomb B, Sanders DB. Mycophenolate mofetil for myasthenia gravis: an open-label pilot study. Neurology 2001;56:97–99.
    OpenUrlAbstract/FREE Full Text
  13. 13.
    Gogovska L, Ljapcev R. An open-label, 12-month trial of mycophenolate mofetil in myasthenia gravis. Eur J Neurol 2004;11(suppl 2):273.
    OpenUrl
  14. 14.↵
    Chaudhry V, Cornblath DR, Griffin JW, O’Brien R, Drachman DB. Mycophenolate mofetil: a safe and promising immunosuppressant in neuromuscular diseases. Neurology 2001;56:94–96.
    OpenUrlAbstract/FREE Full Text
  15. 15.
    Cos L, Mankodi AK, Tawil R, Thornton CA. Mycophenolate mofetil is safe and well-tolerated in myasthenia gravis. Neurology 2000;54(suppl 3):A137.
    OpenUrl
  16. 16.
    Dolezil D, Zapletalova O. Treatment of myasthenia gravis with mycophenolate mofetil: our experience. Eur J Neurol 2004;11(suppl 2):274.
    OpenUrl
  17. 17.↵
    Meriggioli MN, Ciafaloni E, Al-Hayk KA, et al. Mycophenolate mofetil for myasthenia gravis: an analysis of efficacy, safety, and tolerability. Neurology 2003;61:1438–1440.
    OpenUrlAbstract/FREE Full Text
  18. 18.↵
    Meriggioli MN, Rowin J. Single fiber EMG as an outcome measure in myasthenia gravis: results from a double-blind, placebo-controlled trial. J Clin Neurophysiol 2003;20:382–385.
  19. 19.
    Meriggioli MN, Rowin J, Richman JG, Leurgans S. Mycophenolate mofetil for myasthenia gravis: a double-blind, placebo-controlled pilot study. Ann NY Acad Sci 2003;998:494–499.
    OpenUrlCrossRefPubMed
  20. 20.↵
    Hart IK, Sathasivam S, Sharshar T. Immunosuppressive agents for myasthenia gravis. Cochrane Database Syst Rev 2007;4:CD005224.
    OpenUrlPubMed
  21. 21.
    Schneider-Gold C, Gajdos P, Toyka KV, Hohlfeld RR. Corticosteroids for myasthenia gravis. Cochrane Database Syst Rev 2005;2:CD002828.
  22. 22.↵
    Jaretzki A III, Barohn RJ, Ernstoff RM, et al. Myasthenia gravis: recommendations for clinical research standards. Task Force of the Medical Scientific Advisory Board of the Myasthenia Gravis Foundation of America. Neurology 2000;55:16–23.
    OpenUrlFREE Full Text
  23. 23.
    Barohn RJ, McIntire D, Herbelin L, Wolfe GI, Nations S, Bryan WW. Reliability testing of the quantitative myasthenia gravis score. Ann NY Acad Sci 1998;841:769–772.
    OpenUrlCrossRefPubMed
  24. 24.↵
    Wolfe GI, Herbelin L, Nations SP, Foster B, Bryan WW, Barohn RJ. Myasthenia gravis activities of daily living profile. Neurology 1999;52:1487–1489.
    OpenUrlAbstract/FREE Full Text
  25. 25.↵
    Palace J, Newsom-Davis J, Lecky B. A randomized double-blind trial of prednisolone alone or with azathioprine in myasthenia gravis. Myasthenia Gravis Study Group. Neurology 1998;50:1778–1783.
    OpenUrlAbstract/FREE Full Text
  26. 26.↵
    US Food and Drug Administration. Mycophenolate mofetil (CellCept®) labelling. Available at: http://www.fda.gov/cder/foi/label/2000/50759S5lbl.pdf.
  27. 27.↵
    van Staa TP, Abenhaim L, Cooper C, Zhang B, Leufkens HG. Public health impact of adverse bone effects of oral corticosteroids. Br J Clin Pharmacol 2001;51:601–607.
    OpenUrlPubMed
  28. 28.
    Behrend M. Adverse gastrointestinal effects of mycophenolate mofetil: aetiology, incidence and management. Drug Saf 2001;24:645–663.
    OpenUrlCrossRefPubMed

Disputes & Debates: Rapid online correspondence

No comments have been published for this article.
Comment

REQUIREMENTS

If you are uploading a letter concerning an article:
You must have updated your disclosures within six months: http://submit.neurology.org

Your co-authors must send a completed Publishing Agreement Form to Neurology Staff (not necessary for the lead/corresponding author as the form below will suffice) before you upload your comment.

If you are responding to a comment that was written about an article you originally authored:
You (and co-authors) do not need to fill out forms or check disclosures as author forms are still valid
and apply to letter.

Submission specifications:

  • Submissions must be < 200 words with < 5 references. Reference 1 must be the article on which you are commenting.
  • Submissions should not have more than 5 authors. (Exception: original author replies can include all original authors of the article)
  • Submit only on articles published within 6 months of issue date.
  • Do not be redundant. Read any comments already posted on the article prior to submission.
  • Submitted comments are subject to editing and editor review prior to posting.

More guidelines and information on Disputes & Debates

Compose Comment

More information about text formats

Plain text

  • No HTML tags allowed.
  • Web page addresses and e-mail addresses turn into links automatically.
  • Lines and paragraphs break automatically.
Author Information
NOTE: The first author must also be the corresponding author of the comment.
First or given name, e.g. 'Peter'.
Your last, or family, name, e.g. 'MacMoody'.
Your email address, e.g. higgs-boson@gmail.com
Your role and/or occupation, e.g. 'Orthopedic Surgeon'.
Your organization or institution (if applicable), e.g. 'Royal Free Hospital'.
Publishing Agreement
NOTE: All authors, besides the first/corresponding author, must complete a separate Publishing Agreement Form and provide via email to the editorial office before comments can be posted.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.

Vertical Tabs

You May Also be Interested in

Back to top
  • Article
    • Abstract
    • Glossary
    • METHODS
    • RESULTS
    • DISCUSSION
    • ACKNOWLEDGMENT
    • Appendix
    • Footnotes
    • REFERENCES
  • Figures & Data
  • Info & Disclosures
Advertisement

Related Articles

  • A trial of mycophenolate mofetil with prednisone as initial immunotherapy in myasthenia gravis

Topics Discussed

  • All Clinical Neurology
  • Clinical trials Randomized controlled (CONSORT agreement)
  • Autoimmune diseases
  • Myasthenia
  • Quality of life

Alert Me

  • Alert me when eletters are published
Neurology: 98 (24)

Articles

  • Ahead of Print
  • Current Issue
  • Past Issues
  • Popular Articles
  • Translations

About

  • About the Journals
  • Ethics Policies
  • Editors & Editorial Board
  • Contact Us
  • Advertise

Submit

  • Author Center
  • Submit a Manuscript
  • Information for Reviewers
  • AAN Guidelines
  • Permissions

Subscribers

  • Subscribe
  • Activate a Subscription
  • Sign up for eAlerts
  • RSS Feed
Site Logo
  • Visit neurology Template on Facebook
  • Follow neurology Template on Twitter
  • Visit Neurology on YouTube
  • Neurology
  • Neurology: Clinical Practice
  • Neurology: Genetics
  • Neurology: Neuroimmunology & Neuroinflammation
  • Neurology: Education
  • AAN.com
  • AANnews
  • Continuum
  • Brain & Life
  • Neurology Today

Wolters Kluwer Logo

Neurology | Print ISSN:0028-3878
Online ISSN:1526-632X

© 2022 American Academy of Neurology

  • Privacy Policy
  • Feedback
  • Advertise