Scolaris Content Display Scolaris Content Display

Agentes inmunosupresores para la miastenia grave

Contraer todo Desplegar todo

Referencias

References to studies included in this review

De Feo 2002 {published data only}

De Feo LG, Schottlender J, Martelli NA, Molfino NA. Use of intravenous pulsed cyclophosphamide in severe, generalized myasthenia gravis. Muscle and Nerve 2002;26(1):31‐6.

Gajdos 1993 {published data only}

Myasthenia Gravis Clinical Study Group. A randomised clinical trial comparing prednisolone and azathioprine in myasthenia gravis. Results of a second interim analysis. Journal of Neurology, Neurosurgery and Psychiatry 1993;56(11):1157‐63.

Meriggioli 2003b {published data only}

Meriggioli MN, Rowin J, Richman JG, Leurgans S. Mycophenolate mofetil for myasthenia gravis: a double‐blind, placebo‐controlled pilot study. Annals of the New York Academy of Sciences 2003;998:494‐9.

Nagane 2005 {published data only}

Nagane Y, Utsugisawa K, Obara D, Kondoh R, Terayama Y. Efficacy of low‐dose FK506 in the treatment of myasthenia gravis ‐ a randomized pilot study. European Neurology 2005;53(3):146‐50.

Palace 1998 {published data only}

Palace J, Newsom‐Davis J, Lecky B. A randomized double‐blind trial of prednisolone alone or with azathioprine in myasthenia gravis. Neurology 1998;50(6):1778‐83.

Tindall 1987 {published data only}

Tindall RSA, Rollins JA, Phillips JT, Greenlee RG, Wells L, Belendiuk G. Preliminary results of a double‐blind, randomized, placebo‐controlled trial of cyclosporine in myasthenia gravis. New England Journal of Medicine 1987;316(12):719‐24.

Tindall 1993 {published data only}

Tindall RS, Phillips JT, Rollins JA, Wells L, Hall K. A clinical therapeutic trial of cyclosporine in myasthenia gravis. Annals of the New York Academy of Sciences 1993;681:539‐51.

References to studies excluded from this review

Bromberg 1997 {published data only}

Bromberg MB, Wald JJ, Forshew DA, Feldman EL, Albers JW. Randomized trial of azathioprine or prednisolone for initial immunosuppressive treatment of myasthenia gravis. Journal of Neurological Sciences 1997;150(1):59‐62.

Schalke 1990 {published data only}

Schalke BG, Kappos L, Rohrbach E, Melms A, Dommasch D, Mertens HG. Ciclosporin A vs. azathioprine in the treatment of myasthenia gravis: final results of a randomised control double‐blind clinical trial. Jikeikai Medical Journal 1990;37(Suppl 1):165‐169.

References to ongoing studies

Sanders 2005 {unpublished data only}

Sanders DB, Ross M, Goldstein J, Pulley M, Meriggioli M, Barohn R, et al. Trial of mycophenolate mofetil in myasthenia gravis. www.clinicaltrials.gov.

Sanders 2006 {unpublished data only}

Sanders DB, Hart IK, Mantegazza R, Shukla SS, Siddiqi ZA. Results of a large, international, double‐blind, placebo‐controlled phase III trial of mycophenolate mofetil as a steroid‐sparing adjunct in the treatment of myasthenia gravis. Unpublished article.

Barohn 1998

Barohn RJ, McIntire D, Herbelin L, Wolfe GI, Nations S, Bryan W. Reliability testing of the quantitative myasthenia gravis score. Annals of the New York Academy of Sciences 1998;841:769‐72.

Besinger 1983

Besinger UA, Toyka KV, Hömberg M, Heininger K, Hohlfeld R, Fateh‐Moghadam A. Myasthenia gravis: Long‐term correlation of binding and bungarotoxin blocking antibodies against acetylcholine receptors with changes in disease severity. Neurology 1983;33(10):1316‐21.

Bonifati 1997

Bonifati DM, Angelini C. Long‐term cyclosporine treatment in a group of severe myasthenia gravis patients. Journal of Neurology 1997;244(9):542‐7.

Chaudhry 2001

Chaudhry V, Cornblath DR, Griffin JW, O'Brien R, Drachman DB. Mycophenolate mofetil: a safe and promising immunosuppressant in neuromuscular diseases. Neurology 2001;56(1):94‐6.

Ciafaloni 2000

Ciafaloni E, Nikhar NK, Massey JM, Sanders DB. Retrospective analysis of the use of cyclosporine in myasthenia gravis. Neurology 2000;55(3):448‐50.

Ciafaloni 2001

Ciafaloni E, Massey JM, Tucker‐Lipscomb B, Sanders DB. Mycophenolate mofetil for myasthenia gravis: an open‐label pilot study. Neurology 2001;56(1):97‐9.

Cos 2000

Cos L, Mankodi AK, Tawil R, Thornton CA. Mycophenolate mofetil (MyM) is safe and well tolerated in myasthenia gravis (MG). Neurology 2000;54(Suppl 3):A137.

Drachman 1994

Drachman DB. Myasthenia gravis. New England Journal of Medicine 1994;330(25):1797‐810.

Dukes 2000

Dukes MNG, Aronson JK. Meyler's Side Effects of Drugs. 14. Elsevier, 2000.

Gajdos 2002

Gajdos P, Chevret S, Toyka K. Plasma exchange for myasthenia gravis. Cochrane Database of Systematic Reviews 2002, Issue 4.

Gajdos 2003

Gajdos P, Chevret S, Toyka K. Intravenous immunoglobulin for myasthenia gravis. Cochrane Database of Systematic Reviews 2003, Issue 2.

Goulon 1988

Goulon M, Elkharrat D, Lokiec F, Gajdos P. Results of a one‐year open trial of cyclosporine in ten patients with severe myasthenia gravis. Transplantation Proceedings 1988;20(3 Suppl 4):211‐7.

Grob 1981

Grob D, Brunner NG, Namba T. The natural course of myasthenia gravis and effect of therapeutic measures. Annals of the New York Academy of Sciences 1981;377:652‐69.

Haberal 2002

Haberal M, Karakayali H, Emiroglu R, Basaran O, Moray G, Bilgin N. Malignant tumors after renal transplantation. Artificial organs 2002;26(9):778‐81.

Heckmann 2001

Heckmann JM, LeePan EB, Eastman RW. High‐dose immunosuppressive therapy in generalised myasthenia gravis ‐ a 2‐year follow‐up study. South African Medical Journal 2001;91(9):765‐70.

Hohlfeld 1985

Hohlfeld R, Toyka KV, Besinger UA, Gerthold B, Heininger K. Myasthenia gravis: Reactivation of clinical disease and of autoimmune factors after discontinuation of long‐term azathioprine. Annals of Neurology 1985;17(3):238‐42.

Hohlfeld 1988

Hohlfeld R, Michels M, Heininger K, Besinger U, Toyka KV. Azathioprine toxicity during long‐term immunosuppression of generalized myasthenia gravis. Neurology 1988;38(2):258‐61.

Hohlfeld 2003

Hohlfeld R, Melms A, Schneider C, Toyka KV, Drachman DB. In Brandt T, Caplan LR, Dichgans J, Diener HC, Kennard C (eds.). Therapy of myasthenia gravis and myasthenic syndromes. Amsterdam: Elsevier, 2003.

Imao 2007

Imao T, Ichimaru N, Takahara S, Kokado Y, Okumi M, Imamura R, Namba Y, Isaka Y, Nonomura N, Okuyama A. Risk factors for malignancy in Japanese renal transplant recipients. Cancer2007:Epub ahead of print.

Jaretzki 2000

Jaretzki A, Barohn RJ, Ernstoff RM, et al. Myasthenia gravis. Recommendations for clinical research standards. Neurology 2000;55(1):16‐23.

Kawaguchi 2004

Kawaguchi N, Yoshiyama Y, Nemoto Y, Munakata S, Fukutake T, Hattori T. Low‐dose tacrolimus treatment in thymectomised and steroid‐dependent myasthenia gravis. Current Medical Research and Opinion 2004;20(8):1269‐73.

Kissel 1986

Kissel JT, Levy RJ, Mendell JR, Griggs RC. Azathioprine toxicity in neuromuscular disease. Neurology 1986;36(11):35‐9.

Knight 2004

Knight A, Askling J, Granath F, Sparen P, Ekbom A. Urinary bladder cancer in Wegener's granulomatosis: risks and relation to cyclophosphamide. Annals of the rheumatic diseases 2004;63(10):1307‐11, comment 1183‐5.

Konishi 2003

Konishi T, Yoshiyama Y, Takamori M, et al. Clinical study of FK506 in patients with myasthenia gravis. Muscle & Nerve 2003;28(5):570‐4.

Konishi 2005

Konishi T, Yoshiyama Y, Takamori M, Saida T, the Japanese FK506 MG Study Group. Long‐term treatment of generalised myasthenia gravis with FK506 (tacrolimus). Journal of Neurology, Neurosurgery and Psychiatry 2005;76(3):448‐50.

Kuks 1991

Kuks JBM, Djojoatmodjo S, Oosterhuis HJHG. Azathioprine in myasthenia gravis: observations in 41 patients and a review of literature. Neuromuscular Disorders 1991;1(6):423‐31.

Lavrnic 2005

Lavrnic D, Vujic A, Rakocevic‐Stojanovic V, Stevic Z, Basta I, Pavlovic S, et al. Cyclosporine in the treatment of myasthenia gravis. Acta Neurologica Scandinavica 2005;111(4):247‐52.

Mantegazza 1988

Mantegazza R, Antozzi C, Peluchetti D, Sghirlanzoni A, Cornelio F. Azathioprine as a single drug or in combination with steroids in the treatment of myasthenia gravis. Journal of Neurology 1988;235(8):449‐53.

Matell 1976

Matell G, Bergstrom K, Franksson C, et al. Effects of some immunosuppressive procedures in myasthenia gravis. Annals of the New York Academy of Sciences 1976;274:659‐76.

Meriggioli 2003a

Meriggioli MN, Ciafaloni E, Al‐Hayk KA, et al. Mycophenolate mofetil for myasthenia gravis: an analysis of efficacy, safety, and tolerability. Neurology 2003;61(10):1438‐40.

Meriggioli 2003c

Meriggioli MN, Rowin J. Single fiber EMG as an outcome measure in myasthenia gravis: results from a double‐blind placebo‐controlled trial. Journal of Clinical Neurophysiology 2003;20(5):382‐5.

Mertens 1969

Mertens HG, Balzereit F, Leipert M. The treatment of severe myasthenia gravis with immunosuppressive agents. European Neurology 1969;2(6):321‐39.

Mertens 1981

Mertens HG, Hertel G, Reuther P, Ricker K. Effect of immunosuppressive drugs (azathioprine). Annals of the New York Academy of Sciences 1981;377:691‐9.

Michels 1988

Michels M, Hohlfeld R, Hartung HP, Heininger K, Besinger UA, Toyka KV. Myasthenia gravis: discontinuation of long‐term azathioprine. Annals of Neurology 1988;24(6):798.

Oosterhuis 1981

Oosterhuis HJ. Observations of the natural history of myasthenia gravis and the effect of thymectomy. Annals of the New York Academy of Sciences 1981;377:678‐90.

Oosterhuis 1988

Oosterhuis HJ. Long‐term effects of treatment in 374 patients with myasthenia gravis. Monographs in Allergy 1988;25:75‐85.

Perez 1981

Perez MC, Buot WL, Mercado‐Danguilan C, Bagabaldo ZG, Renales LD. Stable remissions in myasthenia gravis. Neurology 1981;31(1):32‐7.

Ponseti 2005

Ponseti JM, Azem J, Fort JM, Lopez‐Cano M, Vilallonga R, Buera M, et al. Long‐term results of tacrolimus in cyclosporine‐ and prednisolone‐dependant myasthenia gravis. Neurology 2005;64(9):1641‐3.

Richman 2003

Richman DP, Agius MA. Treatment of autoimmune myasthenia gravis. Neurology 2003;61(12):1652‐61.

Schneider‐Gold 2005

Schneider‐Gold 2005, Gajdos P, Toyka KV, Hohlfield RR. Corticosteroids for myasthenia gravis. Cochrane Database of Systematic Reviews 2005, Issue 2.

Skeie 2006

Skeie GO, Apostolski S, Evoli A, Gilhus NE, Hart IK, Harms L, et al. Guidelines for the treatment of autoimmune neuromuscular transmission disorders.. European Journal of Neurology 2006;13(7):691‐9.

Vincent 1985

Vincent A, Newsom‐Davis J. Acetylcholine receptor antibody as a diagnostic test for myasthenia gravis: results in 153 validated cases and 2967 diagnostic assays. Journal of Neurology, Neurosurgery and Psychiatry 1985;48(12):1246‐52.

Vincent 2001

Vincent A, Palace J, Hilton‐Jones D. Myasthenia gravis. Lancet 2001;357(9274):2122‐28.

Vincent 2003

Vincent A, Bowen J, Newsom‐Davis J, McConville J. Seronegative generalised myasthenia gravis: clinical features, antibodies, and their targets. Lancet Neurology 2003;2(2):99‐106.

Witte 1984

Witte AS, Cornblath DR, Parry GJ, Lisak RP, Schatz NJ. Azathioprine in the treatment of myasthenia gravis. Annals of Neurology 1984;15(6):602‐5.

Characteristics of studies

Characteristics of included studies [ordered by study ID]

De Feo 2002

Methods

Randomised, double‐blind controlled trial of cyclophosphamide plus prednisolone versus prednisolone plus placebo.

Participants

23 participants, 12 in the cyclophosphamide plus corticosteroids group and 11 in the corticosteroids plus placebo group.

Interventions

Intravenous cyclophosphamide pulses were given at an initial dose of 500 mg/m2 of body surface and titrated according to response or side effects. During the 12‐month study period, treatment pulses were given monthly during the first 6 months and then every other month.

Outcomes

Primary endpoints were changes in the QMG, steroid and pyridostigmine requirements, and the number of treatment failures, the latter defined as ventilatory failure (forced vital capacity < 30% predicted) or the development of swallowing impairment requiring a feeding tube. Cyclophosphamide was reported to be not significantly better than placebo at 6 months as assessed by the QMG. Cyclophosphamide was reported to significantly improve the QMG at 12 months. At six months, cyclophosphamide treated participants were on significantly lower doses of prednisolone (mean 13.3 mg/day) than the placebo group (mean 24 mg/day) (P < 0.05). Data on anticholinesterase medication use were not comprehensively reported or evaluated statistically. There was no significant difference in treatment failures between the two groups.

Notes

The QMG was used for muscle strength assessment.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Low risk

A ‐ Adequate

Gajdos 1993

Methods

Randomised, unblinded, controlled trial of azathioprine plus initial prednisolone versus prednisolone.

Participants

41 participants, 21 in the azathioprine plus initial prednisolone group and 20 in the prednisolone group.

Interventions

Prednisolone (1 mg/kg daily for 1 month, subsequently reduced to 0.5 mg/kg daily by the fifth month, and to 0.25 mg/kg daily by the tenth month) versus azathioprine (3 mg/kg daily for one year and then 2 mg/kg daily). In the latter group, participants were also given prednisolone 1 mg/kg daily for 1 month, which was then gradually tapered and discontinued.

Outcomes

Efficacy of azathioprine plus initial prednisolone versus prednisolone at 6 months was not evaluated. No differences in the myasthenia muscle score or the modified Rankin scale were observed between the prednisolone or azathioprine plus initial prednisolone groups at 12 months. Primary endpoints of the study were time to the first episode of clinical deterioration within the first 60 months, defined by the occurrence of either impaired swallowing or respiratory insufficiency, or a drop in the myasthenia muscle score of at least 20 points. Among the 21 deterioration events, 9 were observed in the azathioprine group and 12 in the prednisolone group, giving a relative risk of 0.71 (95% CI 0.39 to 1.31).

Notes

A myasthenia muscle strength score and a five‐grade functional scale (modified Rankin scale) were used for muscle assessment.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Low risk

A ‐ Adequate

Meriggioli 2003b

Methods

Randomised double‐blind controlled pilot trial of mycophenolate mofetil plus either ciclosporin or prednisolone or no immunosuppressants versus placebo plus either ciclosporin or prednisolone or no immunosuppressants.

Participants

14 participants, 7 in the mycophenolate mofetil plus either ciclosporin or prednisolone or no immunosuppressants group, 7 in the placebo plus either ciclosporin or prednisolone or no immunosuppressants group.

Interventions

Mycophenolate mofetil (1 g twice daily) in one group versus placebo in the other group. Five participants in each group were also on prednisolone. One participant in each group was also on ciclosporin. Two participants in each group were on mycophenolate mofetil monotherapy.

Outcomes

The primary endpoint was the change in QMG score compared with baseline. The study only ran for 5 months. One of the five participants in the placebo group and one of the six participants in the mycophenolate mofetil group showed mild improvement in the QMG at the end of the study. There was no significant difference in the change of group medians of titre at the end of the study. The median change of antibody titre in the mycophenolate mofetil group was 1.1 nmol/L and in the placebo group 0.1 nmol/L (P = 0.52).

Notes

The QMG and manual muscle testing were used for muscle strength assessment.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Low risk

A ‐ Adequate

Nagane 2005

Methods

Randomized, unblinded, nonplacebo‐controlled pilot trial of tacrolimus plus corticosteroids with/without plasma exchange versus no tacrolimus plus corticosteroids with/without plasma exchange.

Participants

34 participants, 18 in the tacrolimus plus corticosteroids with/without plasma exchange group, 16 in the no tacrolimus plus corticosteroids with/without plasma exchange.

Interventions

Tacrolimus was administered orally at a dose of 3 mg/day. For early‐phase therapy, participants received tacrolimus or prednisolone or both. Normal quality of life corresponding to the category 'Minimal Manifestations' (MM) of the MGFA Postintervention Status scale was achieved by administering, as required, early‐phase treatment which consisted of oral prednisolone (not exceeding a daily dose of 20 mg), and combined therapy with plasmapheresis and high‐dose intravenous methylprednisolone (1 mg/day for the first three days). The prednisolone dose was tapered and adjusted to the minimal dose needed to maintain MM. For the follow‐up phase, participants were treated to maintain MM for one year. In the follow‐up phase, plasma exchange plus high‐dose intravenous methylprednisolone, high‐dose intravenous methylprednisolone alone or pyridostigmine was administered as needed to maintain MM.

Outcomes

None of the primary and secondary outcome measures for review were available. In the study, endpoints of early‐phase therapy were frequency of plasma exchange plus high‐dose intravenous methylprednisolone, and the duration of early‐phase treatment. The number of treatments with plasma exchange plus high‐dose intravenous methylprednisolone was significantly lower in participants treated with tacrolimus compared with those treated without tacrolimus during early‐phase treatment, with a mean difference of ‐1.80 (95% CI ‐3.12, ‐0.48). The period of early‐phase treatment was significantly shorter in the group treated with tacrolimus, with a mean difference of ‐2.10 (95% CI ‐3.63 to ‐0.57). Endpoints of follow‐up phase therapy were frequency of plasma exchange plus high‐dose intravenous methylprednisolone or high‐dose intravenous methylprednisolone alone during this phase, and the dose of oral predisolone required at the endpoint. In this follow‐up phase, the number of treatments with plasma exchange plus high‐dose intravenous methylprednisolone was significantly lower for the participants treated with tacrolimus compared with those without tacrolimus, with a mean difference of ‐0.90 (95% CI ‐1.72 to ‐0.08). In addition, the number of treatments with high‐dose intravenous methylprednisolone alone were significantly lower for the participants treated with tacrolimus compared with those without tacrolimus, with a mean difference of ‐1.40 (95% CI ‐2.79 to ‐0.01). At one year into treatment, the oral prednisolone dose was significantly lower for participants treated with tacrolimus compared with those without tacrolimus, with a mean difference of ‐3.50 (95% CI ‐5.78 to ‐1.22).

Notes

The MGFA Postintervention Status scale and the QMG were used for muscle strength assessment.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

High risk

C ‐ Inadequate

Palace 1998

Methods

Randomised double‐blind controlled trial of azathioprine plus prednisolone versus prednisolone plus placebo.

Participants

34 participants, 15 in the azathioprine plus prednisolone group and 19 in the prednisolone plus placebo group.

Interventions

All participants received initial high‐dose prednisolone (1.5 mg/kg or 100 mg on alternate days). In addition, participants were randomised to receive either azathioprine (2.5 mg/kg daily) or the equivalent dose of a matching placebo. The initial prednisolone dose was maintained until remission and then tapered to the minimum required to maintain remission.

Outcomes

There were no significant differences between the two treatment groups in objective or subjective measurement scores at 12 months. Primary endpoints of the study were the maintenance dose of prednisolone, number of participants experiencing treatment failure (defined as either no remission achieved in the three year follow‐up period or clinical relapse after initial remission necessitating an increase in prednisolone dose), and the duration of the initial remission. The median prednisolone dose did not differ significantly between the two treatment groups at 12 months (exact figures not given), but was significantly reduced at 36 months in the azathioprine plus prednisolone group (0 mg) compared with the prednisolone plus placebo group (40mg) (P = 0.02). There was no significant difference between the two treatment groups in terms of number of participants experiencing treatment failure. Analysis of those who completed the trial showed that the duration of remission was significantly longer in the azathioprine plus prednisolone group compared with the prednisolone plus placebo group, giving a relative risk of 0.28 (95% CI 0.08 to 0.94). If those who withdrew or died were included as treatment failures, the duration of remission was not significantly different between the azathioprine plus prednisolone versus prednisolone plus placebo groups, with a relative risk of 0.67 (95% CI 0.43 to 1.04).

Notes

For muscle strength assessment, right‐sided strength of neck flexion, elbow extension, first finger abduction, shoulder abduction, hip flexion and hip abduction; walking time, swallowing time, forced vital capacity; and subjective scoring of strength, double vision and swallowing were used.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Low risk

A ‐ Adequate

Tindall 1987

Methods

Randomised double‐blind controlled trial of ciclosporin monotherapy versus placebo.

Participants

20 participants, 10 in the ciclosporin group, 10 in the placebo group.

Interventions

Ciclosporin was started at a dose of 6 mg/kg daily and then adjusted on the basis of assessments of blood drug levels, renal function, clinical response and adverse events.

Outcomes

7 of the 10 participants in the ciclosporin group and four of the 10 participants in the placebo group showed mild improvement at 6 months. Therefore, the relative rate of improvement was 1.75 (95% CI 0.74 to 4.14). The ciclosporin group demonstrated significantly greater increase in muscle strength (measured by change in the QMG score) than the placebo group. The mean difference in QMG score between the ciclosporin group and placebo group was ‐0.46 (95% CI ‐0.83 to ‐0.09). 6 of the 10 participants in the ciclosporin group improved at 12 months: 5 had mild improvement and 1 had marked improvement. 1 of the 10 participants in the placebo group had mild improvement. Therefore, the overall relative rate of improvement was 6.00 (95% CI 0.87 to 41.21). The ciclosporin group demonstrated significantly greater increase in muscle strength (measured by change in the QMG score) than the placebo group. The mean difference in QMG score between the ciclosporin group and placebo group was ‐0.72 (95% CI ‐1.10 to ‐0.34). There was no significant mean difference of AChR antibody titre between the two groups at 6 months. Six participants in the ciclosporin group and 5 participants in the placebo group experienced treatment failure, giving a relative risk of 1.20 (95% CI 0.54 to 2.67).

Notes

The QMG was used for muscle strength assessment.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Low risk

A ‐ Adequate

Tindall 1993

Methods

Randomised double‐blind controlled trial of ciclosporin plus prednisolone versus prednisolone plus placebo

Participants

39 participants, 20 in the ciclosporin plus prednisolone group and 19 in the prednisolone plus placebo group.

Interventions

Ciclosporin was started at a dose of 5 mg/kg daily. The dose was adjusted on the basis of blood drug levels, renal function and adverse reactions. Corticosteroid withdrawal was begun at 2 months with a reduction of 10 mg if the dose was 60 mg every other day or lower, and 20 mg if the dose was 80 to 100 mg. From the third to the sixth months, the dose of corticosteroids was reduced by 10 mg/month. If weakness increased following reduction in corticosteroids, the dose was increased. Participants were withdrawn from the study if treatment failed.

Outcomes

Primary endpoints, measured when participants reached 6 months of treatment or an earlier endpoint, were entry to exit change in QMG, AChR antibody titre and dose of steroids. 8 of the 20 participants in the ciclosporin plus prednisolone group improved. 2 of the 19 participants in the prednisolone plus placebo group improved. Therefore, the overall relative rate of improvement was 5.70 (95% CI 1.46 to 22.18). The ciclosporin group demonstrated significantly greater increase in muscle strength than the placebo group. The mean difference in QMG score between the ciclosporin group and placebo group was ‐0.31 (95% CI ‐0.51 to ‐0.11). The ciclosporin group had a statistically significant percentage reduction of antibody titre compared with placebo at six months, with a mean difference of 62.90 (95% CI 1.22 to 124.58). There was no significant difference in corticosteroid dose between the two groups. The mean difference was 13.60 (95% CI ‐5.64 to 32.84).

Notes

The QMG was used for muscle strength assessment.

Risk of bias

Bias

Authors' judgement

Support for judgement

Allocation concealment?

Low risk

A ‐ Adequate

QMG ‐ Quantitative MG Score for Disease Severity

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Bromberg 1997

There were major violations of the trial protocol. Patients were initially randomised to either azathioprine or prednisolone with the intention to treat for one year before they crossed over to the other drug. Only four out of the five patients initially randomised to azathioprine were crossed over to prednisolone, two within four weeks of starting the trial ‐ because they did not tolerate azathioprine ‐ and two at the end of one year. The other patient on azathioprine was kept on the drug beyond one year. None of the patients on prednisolone at initial randomisation were crossed over to azathioprine. Therefore, the intended crossover design of the trial was not achieved.

Schalke 1990

The trial set out to compare ciclosporin with azathioprine. However, it lacked key data. In particular, it is unclear which participants had prior immunosuppression (azathioprine alone, ciclosporin alone or azathioprine plus ciclosporin), and if a washout period was instituted before randomisation. These problems make it difficult to know which drug was actually being evaluated.

Characteristics of ongoing studies [ordered by study ID]

Sanders 2005

Trial name or title

Trial of mycophenolate mofetil in myasthenia gravis.

Methods

Participants

Randomised double‐blind controlled trial of mycophenolate mofetil plus prednisolone versus prednisolone versus placebo. 80 participants with seropositive MG were randomized.

Interventions

Thirty six weeks of treatment with 2.5 g mycophenolate mofetil/day plus at least 20 mg prednisone/day versus at least 20 mg/day prednisone plus placebo.

Outcomes

The primary outcome measure was the change from baseline in QMG score at the end of 36 weeks. Secondary outcome measures included survival analysis for treatment failure, MG‐related impairment of daily activities, functional assessment, manual muscle testing, SF‐36 Health Status, and serum concentration of anti‐AChR antibody. The trial failed to meet both primary and secondary endpoints. The trial is awaiting publication.

Starting date

January 2003

Contact information

Donald B Sanders, M.D., Principal Investigator, Duke University.

Notes

The QMG was used for muscle strength assessment.

Sanders 2006

Trial name or title

International, double‐blind, placebo‐controlled phase III trial of mycophenolate mofetil as a steroid‐sparing adjunct in the treatment of myasthenia gravis.

Methods

Participants

Randomised double‐blind controlled trial of mycophenolate mofetil plus prednisolone versus prednisolone versus placebo. 176 participants with seropositive MG were randomized.

Interventions

Thirty six weeks of treatment with 2 g mycophenolate mofetil/day plus at least 20 mg prednisone/day versus at least 20 mg/day prednisone plus placebo.

Outcomes

The primary outcome measures were: (1) minimal manifestations (MM) or pharmacologic remission (PR) on the MGFA Postintervention Status scale of disease symptoms for weeks 32‐36, and (2) =7.5 mg/day prednisolone for weeks 32‐36, and (3) =120 mg/day pyridostigmine for weeks 33‐36. Secondary efficacy outcomes included: time to initial 'treatment response' ‐ the first visit week when a participant met all three primary efficacy criteria and maintained this status at the next visit; time to sustained 'treatment response' ‐ the first visit week when a participant met all three primary efficacy criteria and maintained this status until week 36; prednisone area under the time/dose curve; prednisone dose at study termination; cholinesterase inhibitor dose at study termination; number of intravenous immunoglobulin and plasma exchange treatments received during the study; change from baseline in QMG score; change from baseline in Quality of Life (QoL) assessed using the 36‐item Short Form (SF‐36) health survey and the MG Activities of Daily Living (MG ADL) scale; change from baseline in investigator and patient Global Assessment of disease severity; and AChR antibody titres. The study is awaiting publication.

Starting date

August 2004

Contact information

Donald B Sanders, M.D., Principal Investigator, Duke University.

Notes

The QMG was used for muscle strength assessment.

Data and analyses

Open in table viewer
Comparison 1. Ciclosporin versus placebo at six months

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Six months Show forest plot

2

59

Risk Ratio (M‐H, Fixed, 95% CI)

2.44 [1.13, 5.27]

Analysis 1.1

Comparison 1 Ciclosporin versus placebo at six months, Outcome 1 Six months.

Comparison 1 Ciclosporin versus placebo at six months, Outcome 1 Six months.

Open in table viewer
Comparison 2. Ciclosporin versus placebo ‐ change in QMG at six months

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Six months Show forest plot

2

59

Mean Difference (IV, Fixed, 95% CI)

‐0.34 [‐0.52, ‐0.17]

Analysis 2.1

Comparison 2 Ciclosporin versus placebo ‐ change in QMG at six months, Outcome 1 Six months.

Comparison 2 Ciclosporin versus placebo ‐ change in QMG at six months, Outcome 1 Six months.

Comparison 1 Ciclosporin versus placebo at six months, Outcome 1 Six months.
Figuras y tablas -
Analysis 1.1

Comparison 1 Ciclosporin versus placebo at six months, Outcome 1 Six months.

Comparison 2 Ciclosporin versus placebo ‐ change in QMG at six months, Outcome 1 Six months.
Figuras y tablas -
Analysis 2.1

Comparison 2 Ciclosporin versus placebo ‐ change in QMG at six months, Outcome 1 Six months.

Table 1. Summary of Quantitative MG Score for Disease Severity (QMG)

Items tested

Time to develop diplopia with prolonged lateral gaze

Time to develop ptosis with prolonged upgaze

Facial strength

Ability to swallow 4 oz (1/2 cup) of water

Speech after counting aloud from 1 to 50

Time patient can outstretch either arm

Vital capacity

Right and left grip strength

Time patient can lift head 45° in a supine position

Time patient can outstretch either leg

Figuras y tablas -
Table 1. Summary of Quantitative MG Score for Disease Severity (QMG)
Table 2. Summary of MGFA Postintervention Status Scale

Category

Clinical state

Complete Stable Remission (CSR)

No symptoms or signs of MG. Not received therapy for MG for at least one year.

Pharmacologic Remission (PR)

Same as above, but continues to take some form of therapy for MG. Patients on cholinesterase inhibitors are excluded as their use suggests the presence of weakness.

Minimal Manifestations (MM)

No symptoms of functional limitations, but has some weakness on examination.

Minimal Manifestations MM‐0

Not received MG treatment for at least one year.

Minimal Manifestations MM‐1

Continues to receive some form of immunosuppression, but no cholinesterase inhibitors or other symptomatic therapy.

Minimal Manifestations MM‐2

Received only low‐dose cholinesterase inhibitors (<120 mg pyridostigmine/day) for at least one year.

Minimal Manifestations MM‐3

Received cholinesterase inhibitors or other symptomatic therapy and some form of immunosuppression during the past year.

Change in status

Improved (I)

A substantial decrease in pretreatment clinical manifestations, or a sustained substantial reduction in medications, or a specific decrease in QMG score, as defined in the protocol.

Unchanged (U)

No substantial change in pretreatment clinical manifestations or reduction in medications, or a change in QMG score, as defined in the protocol.

Worse (W)

A substantial increase in pretreatment clinical manifestations, or a substantial increase in medications, or a specific increase in QMG score, as defined in the protocol.

Exacerbation (E)

Fulfilled criteria of CSR, PR or MM, but subsequently developed clinical findings greater than permitted by these criteria.

Died of MG (D of MG)

Died of MG, of complications of therapy, or within
30 days of a thymectomy.

Figuras y tablas -
Table 2. Summary of MGFA Postintervention Status Scale
Table 3. MGFA Clinical Classification

Class

Definition

Class I

Any ocular muscle weakness
May have weakness of eye closure
All other muscle strength is normal

Class II

Mild weakness affecting other than ocular muscles
May also have ocular muscle weakness of any severity

Class IIa

Predominantly affecting limb, axial muscles, or both
May also have lesser involvement of oropharyngeal muscles

Class IIb

Predominantly affecting oropharyngeal, respiratory muscles, or both
May also have lesser or equal involvement of limb, axial muscles, or both

Class III

Moderate weakness affecting other than ocular muscles
May also have ocular muscle weakness of any severity

Class IIIa

Predominantly affecting limb, axial muscles, or both
May also have lesser involvement of oropharyngeal muscles

Class IIIb

Predominantly affecting oropharyngeal, respiratory muscles, or both
May also have lesser or equal involvement of limb, axial muscles, or both

Class IV

Severe weakness affecting other than ocular muscles
May also have ocular muscle weakness of any severity

Class IVa

Predominantly affecting limb and/or axial muscles
May also have lesser involvement of oropharyngeal muscles

Class IVb

Predominantly affecting oropharyngeal, respiratory muscles, or both
May also have lesser or equal involvement of limb, axial muscles, or both

Class V

Defined by intubation, with or without mechanical ventilation, except when employed during routine postoperative management. The use of a feeding tube without intubation places the patient in class IVb

Figuras y tablas -
Table 3. MGFA Clinical Classification
Table 4. Methodological Quality Scores

Study ID

Alloc concealment

Patient blinding

Observer blinding

Desc of clin status

Explicit outcomes

Baseline difference

Follow‐up

Statistical power

Bromberg 1997

B

C

C

B

A

C

B

C

De Feo 2002

A

A

A

B

A

A

A

B

Gajdos 1993

A

C

C

A

A

B

A

B

Meriggioli 2003b

A

A

A

A

A

C

B

C

Nagane 2005

C

C

C

A

A

A

A

B

Palace 1998

A

A

A

A

A

A

A

B

Schalke 1990

C

A

A

C

A

A

A

B

Tindall 1987

A

A

A

A

A

A

A

B

Tindall 1993

A

A

A

A

A

A

A

B

Figuras y tablas -
Table 4. Methodological Quality Scores
Comparison 1. Ciclosporin versus placebo at six months

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Six months Show forest plot

2

59

Risk Ratio (M‐H, Fixed, 95% CI)

2.44 [1.13, 5.27]

Figuras y tablas -
Comparison 1. Ciclosporin versus placebo at six months
Comparison 2. Ciclosporin versus placebo ‐ change in QMG at six months

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Six months Show forest plot

2

59

Mean Difference (IV, Fixed, 95% CI)

‐0.34 [‐0.52, ‐0.17]

Figuras y tablas -
Comparison 2. Ciclosporin versus placebo ‐ change in QMG at six months