Tacrolimus in the treatment of myasthenia gravis in patients with an inadequate response to glucocorticoid therapy: randomized, double-blind, placebo-controlled study conducted in China

Lei Zhou, Weibin Liu, Wei Li, Haifeng Li, Xu Zhang, Huifang Shang, Xu Zhang, Bitao Bu, Hui Deng, Qi Fang, Jimei Li, Hua Zhang, Zhi Song, Changyi Ou, Chuanzhu Yan, Tao Liu, Hongyu Zhou, Jianhong Bao, Jiahong Lu, Huawei Shi, Chongbo Zhao, Lei Zhou, Weibin Liu, Wei Li, Haifeng Li, Xu Zhang, Huifang Shang, Xu Zhang, Bitao Bu, Hui Deng, Qi Fang, Jimei Li, Hua Zhang, Zhi Song, Changyi Ou, Chuanzhu Yan, Tao Liu, Hongyu Zhou, Jianhong Bao, Jiahong Lu, Huawei Shi, Chongbo Zhao

Abstract

Background: To determine the efficacy of low-dose, immediate-release tacrolimus in patients with myasthenia gravis (MG) with inadequate response to glucocorticoid therapy in a randomized, double-blind, placebo-controlled study.

Methods: Eligible patients had inadequate response to glucocorticoids (GCs) after ⩾6 weeks of treatment with prednisone ⩾0.75 mg/kg/day or 60-100 mg/day. Patients were randomized to receive 3 mg tacrolimus or placebo daily (orally) for 24 weeks. Concomitant glucocorticoids and pyridostigmine were allowed. Patients continued GC therapy from weeks 1-4; from week 5, the dose was decreased at the discretion of the investigator. The primary efficacy outcome measure was a reduction, relative to baseline, in quantitative myasthenia gravis (QMG) score assessed using a generalized linear model; supportive analyses used alternative models.

Results: Of 138 patients screened, 83 [tacrolimus (n = 45); placebo (n = 38)] were enrolled and treated. The change in adjusted mean QMG score from baseline to week 24 was -4.9 for tacrolimus and -3.3 for placebo (least squares mean difference: -1.7, 95% confidence interval: -3.5, -0.1; p = 0.067). A post-hoc analysis demonstrated a statistically significant difference for QMG score reduction of ⩾4 points in the tacrolimus group (68.2%) versus the placebo group (44.7%; p = 0.044). Adverse event profiles were similar between treatment groups.

Conclusions: Tacrolimus 3 mg treatment for patients with MG and inadequate response to GCs did not demonstrate a statistically significant improvement in the primary endpoint versus placebo over 24 weeks; however, a post-hoc analysis demonstrated a statistically significant difference for QMG score reduction of ⩾4 points in the tacrolimus group versus the placebo group. This study was limited by the low number of patients, the absence of testing for acetylcholine receptor antibody and the absence of stratification by disease duration (which led to a disparity between the two groups). ClinicalTrials.gov identifier: NCT01325571.

Keywords: immunology; myasthenia gravis; tacrolimus.

Conflict of interest statement

Conflict of interest statement: Huawei Shi is an employee of Astellas Pharma China, Inc. All other authors declare no conflict of interest.

Figures

Figure 1.
Figure 1.
Patient disposition. *The patient was excluded from both the full analysis set and the per-protocol set due to no assessment of quantitative MG scale at any post-baseline visit.
Figure 2.
Figure 2.
Mean QMG score with tacrolimus versus placebo over 24 weeks [FAS (LOCF)]. FAS, full analysis set; LOCF, last observation carried forward; QMG, quantitative myasthenia gravis; SD, standard deviation.
Figure 3.
Figure 3.
Patients with clinically important improvement in QMG score (⩾4) between baseline and week 24 [FAS (LOCF)]. FAS, full analysis set; LOCF, last observation carried forward; QMG, quantitative myasthenia gravis.

References

    1. Gold R, Schneider-Gold C. Current and future standards in treatment of myasthenia gravis. Neurotherapeutics 2008; 5: 535–541.
    1. Gilhus NE. Myasthenia gravis. New Engl J Med 2016; 375: 2570–2581.
    1. Gilhus NE, Verschuuren JJ. Myasthenia gravis: subgroup classification and therapeutic strategies. Lancet Neurol 2015; 14: 1023–1036.
    1. Jaretzki A, 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.
    1. Guo J, Dang D, Li H-Z, et al. Current overview of myasthenia gravis and experience in China. Neuroimmunol Neuroinflammation 2014; 1: 127–130.
    1. Lai CH, Tseng HF. Nationwide population-based epidemiological study of myasthenia gravis in Taiwan. Neuroepidemiology 2010; 35: 66–71.
    1. Skeie GO, Apostolski S, Evoli A, et al. Guidelines for treatment of autoimmune neuromuscular transmission disorders. Eur J Neurol 2010; 17: 893–902.
    1. Drachman DB. Myasthenia gravis. N Engl J Med 1994; 330: 1797–1810.
    1. Pascuzzi RM, Coslett HB, Johns TR. Long-term corticosteroid treatment of myasthenia gravis: report of 116 patients. Ann Neurol 1984; 15: 291–298.
    1. Astellas. PROGRAF prescribing information, (accessed 11 July 2017).
    1. Cruz JL, Wolff ML, Vanderman AJ, et al. The emerging role of tacrolimus in myasthenia gravis. Ther Adv Neurol Disord 2015; 8: 92–103.
    1. Konishi T, Yoshiyama Y, Takamori M, et al. Clinical study of FK506 in patients with myasthenia gravis. Muscle Nerve 2003; 28: 570–574.
    1. Tada M, Shimohata T, Tada M, et al. Long-term therapeutic efficacy and safety of low-dose tacrolimus (FK506) for myasthenia gravis. J Neurol Sci 2006; 247: 17–20.
    1. Zhao CB, Zhang X, Zhang H, et al. Clinical efficacy and immunological impact of tacrolimus in Chinese patients with generalized myasthenia gravis. Int Immunopharmacol 2011; 11: 519–524.
    1. Nagaishi A, Yukitake M, Kuroda Y. Long-term treatment of steroid-dependent myasthenia gravis patients with low-dose tacrolimus. Intern Med 2008; 47: 731–736.
    1. Yoshikawa H, Kiuchi T, Saida T, et al. Randomised, double-blind, placebo-controlled study of tacrolimus in myasthenia gravis. J Neurol Neurosurg Psychiatry 2011; 82: 970–977.
    1. Soliven B, Rezania K, Gundogdu B, et al. Terbutaline in myasthenia gravis: a pilot study. J Neurol Sci 2009; 277: 150–154.
    1. Osserman K. Myasthenia gravis. New York: Grune and Stratton, 1958.
    1. Selcen D, Dabrowski ER, Michon AM, et al. High-dose intravenous immunoglobulin therapy in juvenile myasthenia gravis. Pediatric Neurol 2000; 22: 40–43.
    1. Tindall RS, Phillips JT, Rollins JA, et al. A clinical therapeutic trial of cyclosporine in myasthenia gravis. Ann N Y Acad Sci 1993; 681: 539–551.
    1. Shimojima Y, Matsuda M, Gono T, et al. Tacrolimus in refractory patients with myasthenia gravis: coadministration and tapering of oral prednisolone. J Clin Neurosci 2006; 13: 39–44.
    1. Ponseti JM, Azem J, Fort JM, et al. Benefits of FK506 (tacrolimus) for residual, cyclosporin- and prednisone-resistant myasthenia gravis: one-year follow-up of an open-label study. Clin Neurol Neurosurg 2005; 107: 187–190.
    1. Ponseti JM, Gamez J, Azem J, et al. Tacrolimus for myasthenia gravis: a clinical study of 212 patients. Ann N Y Acad Sci 2008; 1132: 254–263.
    1. Benatar M, Sanders DB, Burns TM, et al. Recommendations for myasthenia gravis clinical trials. Muscle Nerve 2012; 45: 909–917.

Source: PubMed

3
Iratkozz fel