中国科技论文统计源期刊 中文核心期刊  
美国《化学文摘》《国际药学文摘》
《乌利希期刊指南》
WHO《西太平洋地区医学索引》来源期刊  
日本科学技术振兴机构数据库(JST)
第七届湖北十大名刊提名奖  
医药导报, 2017, 36(6): 597-600
doi: 10.3870/j.issn.1004-0781.2017.06.002
他克莫司治疗重症肌无力临床研究进展
Progress of Clinical Research on Tacrolimus in Treatment of Myasthenia Gravis
辛华雯, 李冉, 刘飞

摘要:

他克莫司是一种新型的免疫抑制药,被用于多种自身免疫性疾病的治疗。越来越多的研究证明,他克莫司对重症肌无力(MG)具有一定的治疗作用。该文综述了他克莫司治疗MG的作用机制、临床研究、不良反应、用法用量及临床评价等,供临床运用参考。

关键词: 他克莫司 ; 免疫抑制药 ; 重症肌无力

Abstract:

Tacrolimus is a novel immunosuppressant used in the treatment of a variety of autoimmune diseases.More and more studies have shown that tacrolimus has a certain therapeutic effect on myasthenia gravis (MG).This article reviews the mechanism,clinical researches,adverse reactions,dosage and clinical evaluation of tacrolimus in the treatment of MG.

Key words: Tacrolimus ; Immunosuppressant ; Myasthenia gravis

重症肌无力(myasthenia gravis,MG)是一种以神经-肌肉接头处传导障碍为特点的获得性自身免疫性疾病,主要由乙酰胆碱受体抗体(AChR-Ab)介导[1]。其临床表现为肌无力、易疲劳、眼睑下垂、咀嚼和吞咽困难等症状,常在活动后加重,休息后减轻,严重时患者丧失劳动力甚至死亡。MG可发生于任何年龄段,但以20~30岁女性及50 岁以上男性居多。

MG的发生率较低,但近50年来其发生率不断升高,而且治疗费用昂贵,患者需伴随长期免疫抑制药治疗,对MG的临床治疗研究也成了近年来的热点之一。合理的手术、药物治疗可缓解大多数MG患者的症状。通常需要根据患者的年龄、性别、疾病进程、器官功能状态等因素来制定个体化治疗方案[2]

胸腺切除术是治疗MG最有效的方法之一。大多数MG患者伴有胸腺异常,其中10%~20%患者伴有胸腺瘤,约70%患者伴有胸腺滤泡性增生[3],伴发胸腺瘤为胸腺切除术的绝对指征。其他治疗方案如静脉大剂量免疫球蛋白和血浆置换法可显著改善症状,一般用于MG危象的治疗。

药物治疗方面,胆碱酯酶抑制药的对症治疗是一线治疗方案之一,但该类药物仅能改善症状,并不能阻止MG的病理性进展,且长期使用会造成敏感性降低,一般用于轻型MG患者的治疗。免疫抑制疗法多用于胆碱酯酶抑制药无效的中重度MG的长期治疗,常用药物包括糖皮质激素,非激素类免疫抑制药,如环孢素、硫唑嘌呤、环磷酰胺,以及新型免疫抑制药麦考酚酸酯及他克莫司等[4],糖皮质激素联合环孢素或他克莫司能减少糖皮质激素用量,从而减轻长期糖皮质激素治疗带来的副作用,是临床最常用的治疗方案之一。

他克莫司是一种大环内酯类免疫抑制药,其临床应用与环孢素相似,常替代环孢素用于器官移植,也可用于多种自身免疫性疾病的治疗。他克莫司治疗MG的机制尚未完全阐明,一般认为他克莫司的免疫抑制是选择性抑制T细胞活化介导[5-6]。他克莫司结合到作为细胞内受体的结合蛋白12(FKBP12),形成FKBP12-TAC复合物,该复合物抑制钙调神经磷酸酶介导的核因子(NF-AT)的去磷酸化,从而阻止NF-AT转移到细胞核而导致白细胞介素(IL)-2的转录受阻。

另一方面,各种细胞因子在MG的发病机制中起重要作用。研究发现[7],他克莫司能降低MG患者IL-12、IL-17、 γ干扰素(IFN-γ)、粒细胞巨噬细胞刺激因子(granulocyte-macrophage stimulating factor,GM-CSF)、肿瘤坏死因子α(TNF-α)及巨噬细胞炎性蛋白-1β (MIP-1β)等细胞因子的水平,提高IL-10的水平,提示他克莫司的免疫抑制作用可能与巨噬细胞的抑制有关。另有一项国内研究表明,他克莫司能降低MG患者IFN-γ、IL-2、IL-10和IL-13等细胞因子水平,诱导耐受性浆细胞样树突状细胞的增殖,提示他克莫司的免疫抑制作用可能与IL-13及CD19阳性(C D 19 + )B细胞活化因子受体阳性(BAFF-R+)B细胞水平的降低有关。这两个研究在IL-10细胞因子变化上的差异可能由试验方法不同造成,前者检测外周血单核细胞中细胞因子含量,后者直接检测患者血清细胞因子含量。此外,他克莫司还能通过促进肌肉细胞中(肌浆网)钙离子释放,提高肌肉兴奋-收缩耦联,从而提高肌肉收缩能力[8]

笔者综述他克莫司治疗MG的临床研究、不良反应、用法用量及临床评价等,以便为他克莫司在MG中的合理应用提供参考依据。

1 临床研究

根据美国重症肌无力协会的临床分级来评价患者的疾病进程,根据临床表现严重程度一般将MG分成Ⅰ~Ⅴ级,Ⅰ级最轻微,而Ⅴ级则需要插管,其中,Ⅱ、Ⅲ和Ⅳ级又被分成了a和b两类,a类主要累及四肢,b类主要累及口咽及呼吸道[9]

在相关临床试验中,一般通过分析治疗过程中患者乙酰胆碱受体抗体水平、MG 评分、MG肌力定量 (quantitative myasthenia gravis,QMG) 评分、徒手肌力检查(manual muscle testing,MMT)评分、肌肉强度(test to evaluate muscular strength,TEMS)评分、日常活动能力(activities of daily living,ADL)评分、完全稳定缓解率(complete stable remission,CSR)、糖皮质激素剂量等临床特征来评价治疗的有效性。

1.1 单用糖皮质激素治疗效果不理想或激素依赖的MG患者

KONISHI等[10-11]对1997—2000年间19例单用糖皮质激素不能达到理想效果的全身型MG患者做了一项前瞻性研究,患者使用他克莫司联合糖皮质激素,16周后,患者乙酰胆碱受体抗体水平显著降低,其中9例患者的MG评分及ADL评分显著提升。随后,其中12例患者继续接受长达2年的小剂量他克莫司治疗,8例患者MG评分及ADL评分显著提升,7例患者糖皮质激素成功减量。

YUTAKA等[4]对1993—2004年86例全身型MG患者进行两项回顾性研究。研究者将其中29例患者纳入研究一,他克莫司组13例在6个月内开始联用他克莫司,而对照组16例仅使用糖皮质激素治疗。两组患者初始糖皮质激素稳定剂量相同,之后根据患者情况将糖皮质激素逐渐减量。在糖皮质激素治疗开始第30个月和第36个月,两组患者糖皮质激素剂量比较差异有统计学意义,第30个月他克莫司组中位剂量为1.2 mg·d-1,对照组为6.0 mg·d-1;第36个月他克莫司组为0.0 mg·d-1,对照组为5.5 mg·d-1。研究者将其中9例由于肌无力症状的恶化导致激素无法减量的糖皮质激素依赖患者纳入研究二,在联合使用他克莫司后6个月,患者糖皮质激素剂量由起始12.5 mg·d-1减至9.0 mg·d-1,并在3年内逐渐减量至8.0 mg·d-1。这两个研究证明小剂量他克莫司有助于全身型MG患者激素减量。而且,小剂量他克莫司长期应用似乎比短期应用更有效。

国内的一个研究同样证实他克莫司有助于患者的糖皮质激素减量。ZHAO等[7]对2007—2008年47例全身型MG患者进行多中心前瞻性研究。纳入研究均为单用糖皮质激素不能耐受或不能达到理想治疗效果及激素依赖的患者,其中43例完成24周的研究,有31例患者在研究期间联用糖皮质激素,初始剂量31 mg·d-1。经过24周的治疗,患者的QMG评分、MMT评分、ADL评分都有显著提升。31例使用糖皮质激素患者,有24例成功将糖皮质激素剂量减至平均19.2 mg·d-1

MINAMI等[12]对2001—2008年9例全身型MG患者进行一项回顾性研究。MG患者给予他克莫司联合糖皮质激素治疗,疗程长达5年。经过5年治疗,患者糖皮质激素用量显著下降(由24.0 mg·d-1降至10.2 mg·d-1),但其中5例患者在治疗期间经历不同程度的病情恶化和激素增量。患者ADL评分有所改善,但差异无统计学意义。研究者推测,纳入的9例患者均糖皮质激素依赖,或病情严重但不能耐受糖皮质激素,这些因素可能影响小剂量他克莫司对病情的控制。所以研究者认为他克莫司仍是MG治疗的重要选择,但也提示长期使用他克莫司可能需要管理和监测。

对于单用糖皮质激素不能达到理想治疗效果及激素依赖的患者,大部分研究均证明他克莫司治疗的有效性及他克莫司在糖皮质激素减量方面的积极影响。但唯一一项多中心、随机、对照、双盲试验中,他克莫司的糖皮质激素减量作用并未得到证实。YOSHIKAWA等[13]对2006—2008年80例分级为I~V级MG患者进行研究。所有患者都被允许使用固定剂量的胆碱酯酶抑制药,其中他克莫司组40例,初始糖皮质激素使用量为10~20 mg·d-1,联合使用他克莫司。对照组40例,仅使用糖皮质激素而不联用任何其他免疫抑制药。在试验开始4周后,根据患者病情稳定程度酌情减量糖皮质激素用药剂量。28周,他克莫司组糖皮质激素平均4.91 mg·d-1,对照组平均6.51 mg·d-1,两组差异无统计学意义。研究者推测,纳入病情轻的I级眼肌型患者,患者病程过长及研究期较短等因素可能导致研究并没有达到预期治疗终点。但针对实验的二次分析结果显示小剂量他克莫司对糖皮质激素减量有着积极作用,预示了他克莫司在MG治疗中有着潜在优势。

1.2 环孢素联合糖皮质激素治疗失败的MG患者

AZEM等[14]对15例分级为Ⅲa-Ⅳb级的全身型MG患者做一项前瞻性研究。患者使用环孢素联合糖皮质激素不能达到理想治疗效果或不能耐受,改用他克莫司联合糖皮质激素。经过12个月的治疗,所有患者都成功达到药物缓解,乙酰胆碱受体抗体水平及MG评分显著降低。同一个团队对2000—2004年79例分级为Ⅱb-Ⅴ级的全身型MG患者做了一项回顾性研究[15],患者使用环孢素联合糖皮质激素不能达到理想治疗效果或不能耐受。经过平均2.5年的他克莫司联合糖皮质激素治疗,除了2例患者,其他77例均成功减少糖皮质激素用量,使用量由平均58.9 mg·d-1显著降至2.4 mg·d-1。患者乙酰胆碱受体抗体水平和QMG评分均显著降低,TEMS评分也得到显著提升。

以上两个研究结果证明,对于环孢素联用糖皮质激素不能达到理想治疗效果或不能耐受的患者,小剂量他克莫司联合糖皮质激素治疗是有效的。而且他克莫司可能代替糖皮质激素和环孢素单一用药治疗MG。

1.3 胸腺切除的MG患者

针对胸腺切除术后的MG患者,PONSETI等[16]做了一项前瞻性研究。49例患者在术后24 h后即开始给予他克莫司联合糖皮质激素治疗。在经过平均24.4个月的治疗之后,患者乙酰胆碱受体抗体水平和QMG评分均显著降低,所有患者均成功停用糖皮质激素。

同一个团队对2000—2006年间212例MG患者做了一项回顾性研究[17],其中110例患者在胸腺切除后因环孢素或糖皮质激素依赖而改用他克莫司,另68例患者在胸腺切除术后24 h即给予他克莫司治疗。研究结果与前一项研究基本一致,在经过平均49.3个月的治疗之后,患者乙酰胆碱受体抗体水平和QMG评分均显著降低,TEMS评分显著增加,95%患者成功停用糖皮质激素。

PONSETI等[18]对1997—2002年80例分级为Ⅲa-V级接受胸腺切除的全身型MG患者做了一项回顾性研究,患者根据术后免疫抑制药使用情况被分为两组,第一组1997—1999年手术39例患者,术后仅服用糖皮质激素,另一组2000—2002年手术的41例患者,服用糖皮质激素联合他克莫司。结果证明,接受糖皮质激素联用他克莫司治疗的早期胸腺切除术后的患者CSR明显优于单用糖皮质激素治疗的患者。

值得一提的是,在第一项研究中,67%非胸腺瘤患者(占总例数71%)在治疗3年后达到完全稳定缓解,而伴胸腺瘤患者均未达到完全稳定缓解。这说明伴胸腺瘤是MG患者的不良预后指标,而后一项研究同样验证这个结论。但无论是否伴胸腺瘤,这两个研究均证明他克莫司对胸腺切除MG患者的有效性。

1.4 眼肌型MG患者

YAGI等[19]对2005—2010年23例分级为Ⅰ级的眼肌型MG患者做了一项回顾性研究。其中4例患者因年龄(平均74岁)及胃肠道出血等并发症不能使用糖皮质激素,采用小剂量他克莫司(平均2.4 mg·d-1)联合胆碱酯酶抑制剂治疗,对照组19例患者则采用激素等其他疗法。研究表明,经过24个月的治疗,他克莫司组患者的乙酰胆碱受体抗体水平显著下降,且较对照组获得更高的眼部QMG评分,这表明小剂量他克莫司作为单一免疫抑制药的长期疗法对眼肌型MG患者有效。

2 不良反应

文献报道他克莫司治疗MG引起的大部分不良反应较轻微,可自行缓解或在对症处理后缓解,包括呼吸道感染、白细胞增多、淋巴细胞减少、中性粒细胞异常、肌酐清除率降低、转氨酶异常、高血糖、高血脂、低钾血症、低镁血症、皮疹、腹泻、手颤、痉挛等。严重不良反应包括高血压、突发性失聪等[20]。ZHAO等[7]研究发现1例患者因肌无力危象在治疗9 d后去世。由于目前大部分研究均使用他克莫司联合糖皮质激素治疗MG,缺乏对照,很难确认不良反应与他克莫司之间的相关性。PONSETI等[15]一项研究证明,患者使用他克莫司替代环孢素联合糖皮质激素治疗MG,发生激素相关不良反应的概率显著降低,如多毛症发生率由50.6%降至2.3%,痤疮发生率由54.1%降至1.2%,其他相关不良反应如感觉异常、牙龈增生、脚踝水肿、手颤、头痛、抑郁、腹泻、高胆固醇血症、血尿素氮增高、血清肌酐浓度增高及高血糖症的发生率显著降低。由此可见,与环孢素联合糖皮质激素的治疗方案比较,小剂量他克莫司联合糖皮质激素治疗MG似乎更安全,更适合MG患者的长期维持治疗。

3 剂量及给药方法

他克莫司联合糖皮质激素治疗MG的常用剂量为3 mg·d-1,每天1次,晚餐后服用,无需根据患者年龄、性别、疾病进程等个人因素及他克莫司血药浓度调节剂量[10]。ZHAO等[7]推荐3 mg·d-1,分两次服用,早晨服用1 mg,晚上服用2 mg,需在餐后2 h服药[7]。PONSETI等[15-18]推荐他克莫司的起始剂量达到0.1 mg·kg-1·d-1,分2次服用,后逐渐调节剂量至血药浓度为7~8 ng·mL-1

由于他克莫司药动学个体差异大,在用于预防器官移植术后免疫排斥时常需监测血药浓度,以保证达到良好治疗效果的前提下,尽量使用小剂量他克莫司,可减轻副作用。在用于治疗MG时,笔者尚未看到文献报道他克莫司剂量与有效性之间的关系。但根据以往使用经验,有理由认为将他克莫司血药浓度控制在有效治疗窗内对于治疗MG是有意义的。

4 结束语

笔者介绍他克莫司用于治疗MG的近期临床试验进展,仍缺乏多中心、随机、对照、双盲试验来进一步证明他克莫司联合糖皮质激素与单用糖皮质激素或环孢素联用糖皮质激素相比在有效性与安全性上的优势。有研究证明他克莫司作为单一免疫抑制药治疗眼肌型MG的疗效,仍缺乏单一用药治疗全身型MG的临床研究。就目前的临床研究结果来看,小剂量他克莫司在减少糖皮质激素依赖性MG患者糖皮质激素用量方面的作用已经得到较多临床试验的证实,他克莫司治疗MG的疗效和安全性已得到初步证实。综上所述,他克莫司在MG治疗中有良好的使用前景。

The authors have declared that no competing interests exist.

参考文献

[1] DRACHMAN D B.Myasthenia gravis[J]. Semin Neurol, 2016, 36(5):419-424.
[本文引用:1]
[2] GOTTERER L, LI Y.Maintenance immunosuppression in myasthenia gravis[J]. J Neurol Sci, 2016,369:294-302.
61Therapy for MG is guided by disease severity, antibody status, and comorbidities.61Immunosuppression is needed in most MG patients.61Corticosteroids are first-line and effective in 80% or more of MG patients.61IVIG and PLEX can be used as maintenance therapy in refractory MG.61MuSK-MG is often severe at onset prompting early initiation of immunosuppression.
DOI:10.1016/j.jns.2016.08.057      PMID:27653912      URL    
[本文引用:1]
[3] RAICA M, CIMPEAN A M, RIBATTI D, et al.Myasthenia gravis and the thymus gland. A historical review[J]. Clin Exp Med, 2008, 8(2):61-64.
The first report of an association between (MG) and the thymus gland was in 1901. Although the underlying mechanisms are uncertain, thymic abnormalities are clearly associated with MG. This review article summarises, from a historical point of view, our knowledge of these associations, and the surgical and medical treatment of MG, including symptom management, immunosuppression, intravenous immunoglobulin and plasmapheresis.
DOI:10.1007/s10238-008-0158-y      PMID:18618214      URL    
[本文引用:1]
[4] YUTAKA F, HIROAKI Y, KAZUO Y, et al.Clinical efficacy and cytokine network-modulating effects of tacrolimus in myasthenia gravis[J]. J Neuroimmunol, 2008, 195(1/2):108-115.
Abstract To clarify the long-term efficacy, safety and the cytokine network-modulating effects of tacrolimus in myasthenia gravis, medical records of 86 newly diagnosed consecutive patients and nine steroid-dependent patients were retrospectively reviewed, and peripheral blood mononuclear cells (PBMC) were cultured for the cytokine profile. Steroid reduction effects were observed by using tacrolimus, and no serious adverse effects were observed. The culture study showed reduced IL-12, IL-17, IFN-gamma, GM-CSF, TNF-alpha and MIP-1beta, and elevated IL-10 in the PBMC from patients who received tacrolimus, which suggests inhibition of T cells and macrophages, and enhancement of type 1 regulatory T cells.
DOI:10.1016/j.jneuroim.2007.12.008      PMID:18262659      URL    
[本文引用:2]
[5] POUCHE L, STOJANOVA J, MARQUET P, et al.New challenges and promises in solid organ transplantation pharmacogenetics: the genetic variability of proteins involved in the pharmacodynamics of immunosuppressive drugs[J]. Pharmacogenomics,2016,17(3): 277-296.
Abstract Interindividual variability in immunosuppressive drug responses might be partly explained by genetic variants in proteins involved in the immune response or associated with IS pharmacodynamics. On a general basis, the pharmacogenetics of drug target proteins is less known and understood than that of proteins involved in drug disposition pathways. The aim of this review is to facilitate research related to the pharmacodynamics of the main immunosuppressive drugs used in solid organ transplantation. We elaborated a quality of evidence grading system based on a literature review and identified 'highly recommended', 'recommended' or 'potential' candidates for further research. It is likely that a number of additional rare variants might further explain drug response phenotypes in transplantation, and particularly the most severe ones. The advent of next-generation sequencing will help to identify those variants.
DOI:10.2217/pgs.15.169      PMID:26799749      URL    
[本文引用:1]
[6] MITSUI T, KURODA Y, UENO S, et al.FK506 attenuates thymic output in patients with myasthenia gravis[J]. Arch Med Sci, 2013, 9(6): 1090-1096.
Myasthenia gravis (MG) is an antibody-mediated, T-cell-dependent autoimmune disease. The symptoms are caused by high-affinity IgG against the muscle acetylcholine receptor (AChR) at the neuromuscular junction. The production of these antibodies in B-cells depends on AChR-specific CD4(+) T-cells and the thymus gland seems to play a significant role in the pathogenesis of MG. Altered thymic T-cell export seems to be associated with a pathological mechanism in myasthenia gravis. Tacrolimus (FK506) has recently been used to treat MG.We examined the effects of tacrolimus on thymic T-cell export in patients with MG. Sixteen patients with nonthymomatous and/or thymectomized MG were treated with oral administrations of tacrolimus. To assess the effect of tacrolimus on the thymic output, we assayed the levels of T-cell receptor excision circle (TREC), a molecular marker of thymus emigrants.T-cell receptor excision circle was not significantly different from those in age-matched controls before tacrolimus therapy, but they were partially decreased 4 months after tacrolimus therapy. T-cell receptor excision circle levels were significantly decreased in the thymomatous group (p < 0.05), but not in the nonthymomatous group. Tacrolimus treatment significantly attenuated TREC levels in cultured CD4(-)CD8(+) cells (p < 0.05), but total cell counts were not significantly changed.These results indicate that TREC levels may become a marker of the curative effect of tacrolimus therapy for thymomatous MG, and that tacrolimus suppresses not only activating T-lymphocytes, but also na茂ve T-cells.
DOI:10.5114/aoms.2013.39797      PMID:3902727      Magsci     URL    
[本文引用:1]
[7] ZHAO C B, ZHANG X, ZHANG H, et al.Clinical efficacy and immunological impact of tacrolimus in Chinese patients with generalized myasthenia gravis[J]. Int Immunopharmacol, 2011, 11(4): 519-524.
In this multicenter, open-label pilot study, the efficacy, safety, and immunological impact of tacrolimus in Chinese patients with generalized myasthenia gravis are assessed. Forty-seven generalized myasthenia gravis (MG) patients were enrolled into this study and given 3mg/day tacrolimus for 24 weeks. The primary efficacy measurements used to monitor response to tacrolimus in MG patients were the Osserman grade, the quantitative MG score (QMGS) recommended by the MGFA, the MG-specific manual muscle testing (MMT) score, and the MG-related activities of daily living (MG-ADL) scale. Also, reduction in steroid doses was used to monitor the effect of tacrolimus. Clinical evaluations were conducted at weeks 4, 8, 12, 16, 20, and 24, while immunological parameters were measured at weeks 4, 12, and 24. Measurements of the Osserman grade, QMGS, MMT, and MG-ADL all suggested improvement in patient health by the fourth week of treatment. Steroid dosage was reduced during the course of the study in 74.2% of the forty-three patients who completed the study. There were thirty-one reported adverse events in the study. Only one was considered serious. We found that tacrolimus reduced levels of the IFN-纬, IL-2, IL-10, and IL-13 cytokines and induced the proliferation of tolerogenic plasmacytoid dendritic cells after treatment. Tacrolimus did not change the population of T cell subtypes but did steadily reduce the population of BAFF-R(+) CD19(+) B cells over the course of the study. Our results show that tacrolimus improves the clinical condition of MG patients and is well tolerated. The decrease in IL-13 and reduction of BAFF-R(+) CD19(+) B cells may be related to the therapeutic effect of tacrolimus.
DOI:10.1016/j.intimp.2010.12.012      PMID:21195813      Magsci     URL    
[本文引用:5]
[8] IMAI T, TSUDA E, HOZUKI T, et al.Early effect of tacrolimus in improving excitation-contraction coupling in myasthenia gravis[J]. Clin Neurophysiol, 2012, 123(9): 1886-1890.
Tacrolimus (FK506) is a macrolide T-cell immunomodulator used to treat myasthenia gravis (MG). Besides immunosuppression, tacrolimus has been reported to have the potential to increase muscle strength by enhancing ryanodine receptor (RyR) function. However, few attempts have been made to demonstrate the early effect of tacrolimus as an RyR enhancer in clinical investigation. In 20 MG patients, masseteric compound muscle action potential (CMAP) and mandibular movement-related potentials (MRPs) were recorded simultaneously after stimulating the trigeminal motor nerve with a needle electrode. The excitation–contraction (E–C) coupling time (ECCT) was calculated by the latency difference between CMAP and MRP. Bite force was measured using a pressure-sensitive sheet. Serial assessments of % decrement in masseteric repetitive nerve stimulation (RNS), ECCT and bite force were performed before and within 4weeks of tacrolimus (3mgday611) treatment. The median (mean, range) interval of assessment was 2 (2.4, 1–4) weeks. We also measured serum antibodies against RyR, acetylcholine receptor and muscle-specific receptor tyrosine kinase. Bite force increased after tacrolimus treatment accompanying clinical improvement assessed by Myasthenia Gravis Foundation of America classification, but the bite force difference did not reach statistical significance. Wilcoxon matched-pairs signed-ranks test detected a significant ECCT shortening in 12 patients assessed after 1–2weeks of tacrolimus treatment as well as in eight patients assessed after 3–4weeks. In contrast, masseteric CMAP and % decrement showed no significant changes after short-term tacrolimus treatment. Tacrolimus induces ECCT shortening accompanying clinical improvement despite no improvement in % decrement within 2weeks. This early effect of tacrolimus may imply a pharmacological enhancement of RyR function to improve E–C coupling in MG.
DOI:10.1016/j.clinph.2012.01.017      PMID:22386321      Magsci     URL    
[本文引用:1]
[9] PENN A S, SANDERS D B, JARETZKI A, et al.Myasthenia gravis: recommendations for clinical research standards: task force of the medical scientific advisory board of the myasthenia gravis foundation of America[J]. Neurology, 2000, 55(1): 16-23.
Elsevier’s Scopus, the largest abstract and citation database of peer-reviewed literature. Search and access research from the science, technology, medicine, social sciences and arts and humanities fields.
DOI:10.1016/S0003-4975(00)01595-2      PMID:10891897      URL    
[本文引用:1]
[10] KONISHI T, YOSHIYAMA Y, TAKAMRI M, et al.Clinical study of FK506 in patients with myasthenia gravis[J]. Muscle Nerve, 2003, 28(5): 570-574.
To investigate the usefulness of low-dose FK506 for the treatment of (MG), we treated 19 patients with generalized MG in a 16-week open clinical trial of FK506 (3-5 mg/day). At the end of the trial, total MG scores (range: 0-27 points) improved by 3 points or more in 7 of 19 patients (37%), and activities of daily living () scores (range: 0-6 points) also improved by 1 point or more in 8 of 19 patients (42%). Nine of 19 patients (47%) showed improvement in either MG or scores. Significant reduction of anti-receptor titers and production were observed at the end of this study. Minor but commonly observed side effects were an increase in neutrophil count and a decrease in lymphocyte count. No serious adverse events such as renal toxicity or mellitus were observed during the 16-week treatment period. FK506 could safely serve as an adjunct to steroid therapy for MG at low dosage.
DOI:10.1002/mus.10472      PMID:14571458      URL    
[本文引用:2]
[11] KONISHI T, YOSHIYAMA Y, TAKAMRI M, et al.Long-term treatment of generalised myasthenia gravis with FK506 (tacrolimus)[J]. J Neurol Neurosurg Psychiatry, 2005, 76(3):448-450.
Efficacy and safety of long term use of FK506 (2-4.5 mg/ day)for a maximum of two years were evaluated in 12 patients with generalised myasthenia gravis (MG) . At the end of the study,eight patients (67%) showed improvement in either MG score or Activities in Daily Living score, and prednisolone dosage could be redu ced in seven patients (58%), with a mean reduction ratio of 37%. Long term use of FK506 for MG can be more effective than short term administration, with no s erious side effects.
DOI:10.1136/jnnp.2004.042176      PMID:15716549      URL    
[本文引用:1]
[12] MINAMI N, FUJIKI N, DOI S, et al.Five-year follow-up with low-dose tacrolimus in patients with myasthenia gravis[J]. J Neurol Sci, 2011, 300(1/2):59-62.
Myasthenia gravis (MG) is an antibody-mediated autoimmune disease of the neuromuscular junction, and prednisolone (PSL) and immunosuppressive drugs are available for treatment. Tacrolimus, a macrolide that suppresses the immune system, is used as a second-line treatment for MG. There have been several reports of the effects of tacrolimus over a few years of follow-up. Here, we report data from 9 patients with steroid-dependent generalized MG treated with low-dose tacrolimus (2–302mg/day) for 502years. Following treatment with tacrolimus, mean MG-activities of daily living score improved from 4.6 at baseline to 3.3 at 502years after initiation of treatment. Mean dose of PSL could also be reduced, from 24.002mg/day at baseline to 10.202mg/day at 502years, although there were no cases of total withdrawal of PSL. By contrast, 5 of the 9 patients experienced exacerbation of symptoms and transient increases in PSL dose during the 5-year period. Tacrolimus is an important option for treatment of MG; however, careful management is needed for long-term treatment with this drug.
DOI:10.1016/j.jns.2010.09.033      PMID:21035148      URL    
[本文引用:1]
[13] YOSHIKAWA H, KIUCHI T, SAIDA Y, et al.Randomised, double-blind, placebo-controlled study of tacrolimus in myasthenia gravis[J]. J Neurol Neurosurg Psychiatry, 2011, 82(9):970-977.
Abstract OBJECTIVES: To evaluate the ability of tacrolimus to reduce the corticosteroid dose in patients with myasthenia gravis (MG) and the drug's safety in a double-blind, placebo-controlled, parallel group study. METHODS: Patients being treated with oral prednisolone at doses equivalent to 10-20 mg/day, and with stable symptoms, were randomised to tacrolimus or placebo in a 28-week double-blind study. The dose of corticosteroid was tapered with the procedures specified in the protocol. The primary efficacy endpoint was the mean daily prednisolone dose given in the last 12&emsp14;weeks of the study. RESULTS: Eighty patients received the study drug (40 patients in each group) and were included in the full analysis set. In the full analysis set, there was no significant difference in the primary efficacy endpoint between the two groups (p = 0.078). However, some secondary analyses suggested the steroid-sparing effect of tacrolimus. Tacrolimus was well tolerated, and no safety concerns were noted. CONCLUSIONS: This study suggests that tacrolimus has a potential advantage as a steroid-sparing agent in the treatment of MG patients. CLINICAL TRIAL REGISTRATION NUMBER: NCT00309088 . Name of the trial registry: FK506 Phase 3 STUDY: A STUDY for Steroid Non-Resistant MG Patients.
DOI:10.1136/jnnp-2011-300148      PMID:21784757      URL    
[本文引用:1]
[14] AZEM J, FORT J M, CODINA A, et al.Benefits of FK506 (tacrolimus) for residual, cyclosporine- and prednisone-resistant myasthenia gravis: one-year follow-up of an open-label study[J]. Clin Neurol Neurosurg, 2005, 107(3):187-190.
Thirteen patients with , unresponsive to and cyclosporin after thymectomy, received KF506 () for 12 months, at starting doses of 0.1 mg/kg per day b.i.d. and then adjusted to achieve plasma concentrations between 7 and 8 ng/mL. The doses of were progressively reduced and finally discontinued. Anti-and score for disease severity decreased significantly and muscular strength increased by 37%. All patients achieved pharmacological remission, 11 were asymptomatic and two had minimal weakness of eyelid closure. was well tolerated and appears a suitable approach after unsuccessful treatment with conventional immunosuppressants in patients with disabling .
DOI:10.1016/j.clineuro.2004.07.013      PMID:15823673      URL    
[本文引用:1]
[15] PONSETI J M, AZEM J, FORT J M, et al.Long-term results of tacrolimus in cyclosporine- and prednisone-dependent myasthenia gravis[J]. Neurology, 2005, 64(9):1641-1643.
Seventy-nine patients with cyclosporine-and prednisone-dependent myasthenia gravis (MG) after thymectomy received tacrolimus for a mean of 2.5 卤0.8 years. Prednisone was withdrawn in all but two patients. Anti-acetylcholine antibodies and MG score for disease severity decreased significantly and muscular strength increased by 39%. Complete stable remission was achieved in 5%of patients and pharmacologic remission in 87.3%. All patients resumed fu ll activities of daily living.
DOI:10.1212/01.wnl.0000218665.94920.5e      PMID:16567729      URL    
[本文引用:3]
[16] PONSETI J M, AZEM J, FORT J M, et al.Experience with starting tacrolimus postoperatively after transsternal extended thymectomy in patients with myasthenia gravis[J]. Curr Med Res Opin, 2006, 22(5): 885-895.
Thymectomy is a standard treatment of myasthenia gravis (MG). Immunomodulating agents are frequently given during the post-thymectomy latency period until complete remission is fully consolidated, but serious side effects is a relevant clinical problem for patients on long-term immunomodulating treatment.To assess the effectiveness of starting tacrolimus in the immediate postoperative period in MG patients undergoing transsternal extended thymectomy, with complete stable remission (CSR) as the primary outcome of the study.Forty-eight MG patients received tacrolimus, 0.1 mg/kg per day b.i.d. (started 24 h after thymectomy) and prednisone 1.5 mg/kg/day. Histologically, 34 patients had hyperplasia, 20 thymic involution, and 14 thymoma. Of the 48 patients, 40 completed 1 year of tacrolimus therapy, 38 completed 2 years, 27 completed 3 years, 21 completed 4 years, and 9 more than 5 years. Mean dose of tacrolimus was 4.9 mg/day (range 2-8 mg/day) with a mean plasma drug concentration of 7.6 ng/mL (range 7-9 ng/mL). Prednisone could be withdrawn after the first year in 93.7% of patients and at 2 years in 100%.The mean follow-up was 24.4 months, SD 17.3 (range 6-60 months). Improvement of muscular strength and decrease of anti-AChR antibodies were statistically significant (p < 0.001) shortly after operation. CSR was obtained in 33.4% of patients, pharmacological remission in 62.6%; 4% of patients had minimal symptoms. None of the patients with thymoma achieved CSR. The estimated median follow-up to obtain a CSR was 37.9 months (95% confidence interval [CI] 26.4-49.5 months). The overall crude CSR rate was 33.4%, with 47% for non-thymoma patients. The probability to achieve CSR at 3 years was 67% for the non-thymomatous group.Long-term immune-directed treatment with tacrolimus to improve the effectiveness of thymectomy in MG is feasible and was associated with a high rate of CSR in patients without thymoma.
DOI:10.1185/030079906X104650      PMID:16709310      URL    
[本文引用:1]
[17] PONSETI J M, GAMEZ J, AZEM J, et al.Tacrolimus for myasthenia gravis: a clinical study of 212 patients[J]. Ann N Y Acad Sci, 2008, 1132:254-263.
Tacrolimus is a macrolide T cell immunomodulator that is used in myasthenia gravis (MG) patients to affect muscle contraction (ryanodine receptor by modulating intracellular calcium-release channels and increasing muscular strength), glucocorticoid receptors (increasing intracellular concentration of steroids and blocking the steroid export mechanism), and an increase in T cell apoptosis. In this study, we report the results of low-dose tacrolimus (0.165mg/kg/day) treatment in 21265MG patients. There were 110 thymectomized, cyclosporine- and prednisone-dependent patients; 68 thymectomized patients who started tacrolimus early postoperatively (2465h after operation); and 3465patients over 60 years old with nonthymomatous generalized MG or in whom thymectomy was contraindicated. The mean follow-up time was 49.365±6518.165months. Muscular strength showed an increase of 23% after 165month of treatment and 29% at the end of the study. The acetylcholine receptor antibodies decreased significantly from a mean of 33.565nmol/L at base line to 7.865nmol/L at the final visit. In the thymectomy group with combined prednisone and tacrolimus stratified by histology of the thymus, the mean probability to attain complete stable remission at 565years was 80.8% in patients with hyperplasia, 48.1% in thymic involution, and 9.3% in patients with thymoma. In 4.9% of patients, tacrolimus was withdrawn because of major adverse effects. Our results suggest that a low dose of tacrolimus is effective for MG and could be included to the armamentarium for this autoimmune disease. The present results should be interpreted considering the limitations of a retrospective clinical study. Confirmation of these results in randomized studies is desirable.
DOI:10.1196/annals.1405.000      PMID:18096852      URL    
[本文引用:1]
[18] PONSETI J M, GAMEZ J, AZEM J, et al.Post-thymectomy combined treatment of prednisone and tacrolimus versus prednisone alone for consolidation of complete stable remission in patients with myasthenia gravis: a non-randomized, non-controlled study[J]. Curr Med Res Opin, 2007, 23(6): 1269-1278.
[本文引用:2]
[19] YAGI Y, SANJO N, YOKOTA T, et al.Tacrolimus monotherapy:a promising option for ocular myasthenia gravis[J]. Eur Neurol, 2013, 69(6):344-345.
Yagi Y, Sanjo N, Yokota T, Mizusawa H.
DOI:10.1159/000347068      PMID:23549260      Magsci     URL    
[本文引用:1]
[20] CRUZ J L, WOLFF M L, VANDERMAN A J, et al.The emerging role of tacrolimus in myasthenia gravis[J]. Ther Adv Neurol Disord, 2015, 8(2): 92-103.
Abstract OBJECTIVE: To describe and evaluate the available evidence assessing the role of tacrolimus in the management of patients with myasthenia gravis (MG). DATA SOURCES: A literature search of MEDLINE (1946 to September 2014) and EMBASE (1947 to September 2014) was performed using the terms 'tacrolimus' and 'myasthenia gravis'. Citations of retrieved articles were examined for relevance. STUDY SELECTION AND DATA EXTRACTION: The search was limited to prospective clinical trials focused on clinical outcomes in patients with generalized MG. Case reports, retrospective evaluations and non-English articles were excluded. DATA SYNTHESIS: A total of 12 studies met inclusion criteria, of which seven articles evaluated tacrolimus in steroid-dependent patients and two examined the utility of tacrolimus in patients failing corticosteroids and cyclosporine. Other studies evaluated early initiation of tacrolimus after thymectomy, effectiveness of tacrolimus in de novo MG and the effectiveness of tacrolimus post-thymectomy in thymoma patients versus nonthymoma. A total of eight trials showed statistically significant improvements in quantitative MG score (QMGS) and postintervention status criteria - Myasthenia Gravis Foundation of America (PSC-MGFA). Of the trials examining steroid reduction with tacrolimus, two reported high rates of complete withdrawal; however, the most robust trial was unable to detect a difference in mean steroid dose. Long-term effects of tacrolimus (up to 5 years) were assessed in eight trials, which consistently showed positive effects on QMGS or reduction in adjunct therapies. CONCLUSIONS: There is limited yet promising information to suggest a beneficial role for tacrolimus in reducing QMGS and corticosteroid burden in patients with refractory symptoms or new-onset MG. Long-term use appears to be safe in this population.
DOI:10.1177/1756285615571873      PMID:25922621      URL    
[本文引用:1]
资源
PDF下载数    
RichHTML 浏览数    
摘要点击数    

分享
导出

相关文章:
关键词(key words)
他克莫司
免疫抑制药
重症肌无力

Tacrolimus
Immunosuppressant
Myasthenia gravis

作者
辛华雯
李冉
刘飞

XIN Huawen
LI Ran
LIU Fei