Interferon beta-1a

干扰素 β - 1a
  • 文章类型: Meta-Analysis
    目的:本研究提出了对原发进行性多发性硬化症(PPMS)临床试验中药物和安慰剂的疗效进行全面定量评估。
    方法:使用PubMed进行了文献检索,EMBASE,和Cochrane库数据库以及报告药物在PPMS治疗中疗效的临床研究被纳入分析.未确认残疾进展的患者的累积比例(wCDP%)用作主要疗效终点。基于模型的荟萃分析方法用于描述每种药物(以及安慰剂)的时间过程,以对治疗PPMS的药物功效进行排名。
    结果:共纳入15项研究,涉及3779名患者,其中,9项为安慰剂对照,6项为单臂试验.研究中包括12种药物。结果表明,除了生物素,干扰素β-1a,和干扰素β-1b,其疗效与安慰剂相当,其他9种药物的疗效明显优于安慰剂。其中,奥克瑞珠单抗表现突出,在96周时wCDP%为72.6,而其余药物的比例约在55-70%之间。
    结论:本研究的结果为原发进展性多发性硬化症的合理临床用药和未来临床试验提供了必要的定量信息。
    OBJECTIVE: This study proposes a comprehensive quantitative evaluation of the efficacy of drugs and placebo in clinical trials for primary progressive multiple sclerosis (PPMS).
    METHODS: A literature search was conducted using the PubMed, EMBASE, and Cochrane library databases and the clinical studies reporting drug efficacy in the treatment of PPMS were included in the analyses. The cumulative proportion of patients without confirmed disability progression (wCDP%) was used as the main efficacy endpoint. The model-based meta-analysis method was used to describe the time course of each drug (as well as placebo) in order to rank the drug efficacy for the treatment of PPMS.
    RESULTS: Fifteen studies involving 3779 patients were included, of which, nine were placebo-controlled and six were single-arm trials. Twelve drugs were included in the study. The results showed that, except for biotin, interferon β-1a, and interferon β-1b, whose efficacy was comparable to the placebo, the efficacy of the other 9 drugs were significantly better than placebo. Among these, ocrelizumab showed outstanding performance, with wCDP% of 72.6 at 96 weeks, while the proportions of rest of the drugs ranged between approximately 55-70%.
    CONCLUSIONS: The results of this study provide the necessary quantitative information for both the rational clinical use of drugs and future clinical trials in primary progressive multiple sclerosis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    Ocrelizumab是一种人源化抗CD20单克隆抗体,用于治疗多发性硬化症(MS)。它于2017年3月获得美国食品和药物管理局(FDA)的批准,用于复发缓解型多发性硬化症(RRMS)和原发性进行性多发性硬化症(PPMS)的成人。Ocrelizumab是唯一批准用于PPMS的疾病改善疗法(DMT)。2017年11月,欧洲药品管理局(EMA)也批准奥克瑞珠单抗作为早期PPMS患者的第一个药物。因此,评估收益很重要,危害,奥克瑞珠单抗在MS患者中的耐受性
    为了评估收益,危害,奥克瑞珠单抗在RRMS和PPMS患者中的耐受性。
    我们搜索了MEDLINE,Embase,中部,和2021年10月8日的两项审判登记。我们筛选了参考名单,联系专家,并联系了研究的主要作者。
    所有随机对照试验(RCT)涉及根据McDonald标准诊断为RRMS或PPMS的成人,比较奥利珠单抗单独或与其他药物相关,在任何持续时间内每24周600毫克的批准剂量,与安慰剂或任何其他活性药物治疗相比。
    我们使用了Cochrane期望的标准方法学程序。
    四个RCT符合我们的选择标准。总体人群包括2551名参与者;1370名接受奥克瑞珠单抗600mg治疗,1181名对照。在控件中,298名参与者接受安慰剂,883名参与者接受干扰素β-1a。在一项研究中,治疗持续时间为24周,两项研究共96周,在一项研究中至少有120周。一项研究对参与者和人员进行分配隐藏和致盲的风险很高;所有四项研究对不完整的结果数据都有很高的偏倚风险。对于RRMS,与干扰素β-1a相比,ocrelizumab与:1.较低的复发率(风险比(RR)0.61,95%置信区间(CI)0.52至0.73;2项研究,1656名参与者;中等确定性证据);2。残疾进展的参与者人数较少(风险比(HR)0.60,95%CI0.43至0.84;2项研究,1656名参与者;低确定性证据);3。有任何不良事件的参与者人数几乎没有差异(RR1.00,95%CI0.96至1.04;2项研究,1651名参与者;中等确定性证据);4。有任何严重不良事件的参与者人数几乎没有差异(RR0.79,95%CI0.57至1.11;2项研究,1651名参与者;低确定性证据);5。因不良事件而停止治疗的参与者人数较少(RR0.58,95%CI0.37至0.91;2项研究,1651名参与者;低确定性证据);6。磁共振成像(MRI)上钆增强T1病变的参与者数量较少(RR0.27,95%CI0.22至0.35;2项研究,1656名参与者;低确定性证据);7.在MRI上出现新的或扩大的T2-高强度病变的参与者数量较少(RR0.63,95%CI0.57至0.69;2项研究,1656名参与者;低确定性证据)在96周。对于PPMS,与安慰剂相比,ocrelizumab与:1.残疾进展的参与者人数较少(HR0.75,95%CI0.58至0.98;1项研究,731名参与者;低确定性证据);2。有任何不良事件的参与者数量较多(RR1.06,95%CI1.01~1.11;1项研究,725名参与者;中等确定性证据);3。发生任何严重不良事件的参与者人数几乎没有差异(RR0.92,95%CI0.68至1.23;1项研究,725名参与者;低确定性证据);4。经历不良事件导致的治疗中断的参与者人数几乎没有差异(RR1.23,95%CI0.55至2.75;1项研究,725名参与者;低确定性证据)至少120周。没有关于MRI上有钆增强T1病变的参与者数量和MRI上有新的或扩大的T2高强度病变的参与者数量的数据。
    对于有RRMS的人,与96周时的干扰素β-1a相比,奥克瑞珠单抗可能导致复发率大幅降低,不良事件几乎没有差异(中度确定性证据).Ocrelizumab可能导致残疾进展的大幅减少,由不良事件引起的治疗中断,MRI上钆增强T1病变的参与者数量,以及MRI上出现新的或扩大的T2高强度病变的参与者数量,并且可能导致严重不良事件几乎没有差异(低确定性证据)。对于PPMS患者,与安慰剂组相比,至少120周的奥克瑞珠单抗可能导致更高的不良事件发生率(中度确定性证据).Ocrelizumab可能导致残疾进展减少,严重不良事件和不良事件引起的治疗中断几乎没有差异(低确定性证据)。Ocrelizumab在临床上耐受良好;最常见的不良事件是输注相关反应和鼻咽炎,泌尿道和上呼吸道感染。
    Ocrelizumab is a humanised anti-CD20 monoclonal antibody developed for the treatment of multiple sclerosis (MS). It was approved by the Food and Drug Administration (FDA) in March 2017 for using in adults with relapsing-remitting multiple sclerosis (RRMS) and primary progressive multiple sclerosis (PPMS). Ocrelizumab is the only disease-modifying therapy (DMT) approved for PPMS. In November 2017, the European Medicines Agency (EMA) also approved ocrelizumab as the first drug for people with early PPMS. Therefore, it is important to evaluate the benefits, harms, and tolerability of ocrelizumab in people with MS.
    To assess the benefits, harms, and tolerability of ocrelizumab in people with RRMS and PPMS.
    We searched MEDLINE, Embase, CENTRAL, and two trials registers on 8 October 2021. We screened reference lists, contacted experts, and contacted the main authors of studies.
    All randomised controlled trials (RCTs) involving adults diagnosed with RRMS or PPMS according to the McDonald criteria, comparing ocrelizumab alone or associated with other medications, at the approved dose of 600 mg every 24 weeks for any duration, versus placebo or any other active drug therapy.
    We used standard methodological procedures expected by Cochrane.
    Four RCTs met our selection criteria. The overall population included 2551 participants; 1370 treated with ocrelizumab 600 mg and 1181 controls. Among the controls, 298 participants received placebo and 883 received interferon beta-1a. The treatment duration was 24 weeks in one study, 96 weeks in two studies, and at least 120 weeks in one study. One study was at high risk of allocation concealment and blinding of participants and personnel; all four studies were at high risk of bias for incomplete outcome data. For RRMS, compared with interferon beta-1a, ocrelizumab was associated with: 1. lower relapse rate (risk ratio (RR) 0.61, 95% confidence interval (CI) 0.52 to 0.73; 2 studies, 1656 participants; moderate-certainty evidence); 2. a lower number of participants with disability progression (hazard ratio (HR) 0.60, 95% CI 0.43 to 0.84; 2 studies, 1656 participants; low-certainty evidence); 3. little to no difference in the number of participants with any adverse event (RR 1.00, 95% CI 0.96 to 1.04; 2 studies, 1651 participants; moderate-certainty evidence); 4. little to no difference in the number of participants with any serious adverse event (RR 0.79, 95% CI 0.57 to 1.11; 2 studies, 1651 participants; low-certainty evidence); 5. a lower number of participants experiencing treatment discontinuation caused by adverse events (RR 0.58, 95% CI 0.37 to 0.91; 2 studies, 1651 participants; low-certainty evidence); 6. a lower number of participants with gadolinium-enhancing T1 lesions on magnetic resonance imaging (MRI) (RR 0.27, 95% CI 0.22 to 0.35; 2 studies, 1656 participants; low-certainty evidence); 7. a lower number of participants with new or enlarging T2-hyperintense lesions on MRI (RR 0.63, 95% CI 0.57 to 0.69; 2 studies, 1656 participants; low-certainty evidence) at 96 weeks. For PPMS, compared with placebo, ocrelizumab was associated with: 1. a lower number of participants with disability progression (HR 0.75, 95% CI 0.58 to 0.98; 1 study, 731 participants; low-certainty evidence); 2. a higher number of participants with any adverse events (RR 1.06, 95% CI 1.01 to 1.11; 1 study, 725 participants; moderate-certainty evidence); 3. little to no difference in the number of participants with any serious adverse event (RR 0.92, 95% CI 0.68 to 1.23; 1 study, 725 participants; low-certainty evidence); 4. little to no difference in the number of participants experiencing treatment discontinuation caused by adverse events (RR 1.23, 95% CI 0.55 to 2.75; 1 study, 725 participants; low-certainty evidence) for at least 120 weeks. There were no data for number of participants with gadolinium-enhancing T1 lesions on MRI and number of participants with new or enlarging T2-hyperintense lesions on MRI.
    For people with RRMS, ocrelizumab probably results in a large reduction in relapse rate and little to no difference in adverse events when compared with interferon beta-1a at 96 weeks (moderate-certainty evidence). Ocrelizumab may result in a large reduction in disability progression, treatment discontinuation caused by adverse events, number of participants with gadolinium-enhancing T1 lesions on MRI, and number of participants with new or enlarging T2-hyperintense lesions on MRI, and may result in little to no difference in serious adverse events (low-certainty evidence). For people with PPMS, ocrelizumab probably results in a higher rate of adverse events when compared with placebo for at least 120 weeks (moderate-certainty evidence). Ocrelizumab may result in a reduction in disability progression and little to no difference in serious adverse events and treatment discontinuation caused by adverse events (low-certainty evidence). Ocrelizumab was well tolerated clinically; the most common adverse events were infusion-related reactions and nasopharyngitis, and urinary tract and upper respiratory tract infections.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:作为一种疾病修饰疗法,鞘氨醇-1-磷酸受体(S1PR)调节剂,如芬戈莫德,奥扎曼德,和西波莫德已被批准或正在开发用于治疗多发性硬化症(MS)。已经实施了几项随机对照试验(RCT),以比较S1PR调节剂与干扰素β在治疗复发缓解型多发性硬化症(RRMS)患者中的疗效和安全性。
    方法:我们通过搜索PubMed搜索了2010年1月至2020年6月实施的RCT,Embase,Cochrane图书馆数据库,和中央受控试验登记册。最后,经过仔细选择,我们将5项随机对照试验纳入了我们的研究。
    结果:我们汇集了来自5个随机对照试验的4304例患者。主要结果是年复发率。我们发现,S1PR调节剂组的年复发率比干扰素β组低20%(95CI,-0.32至-0.07,P=0.002)。与干扰素β相比,S1PR调节剂导致每次扫描新的或扩大的T2病变数量以及钆增强病变数量显着减少。此外,S1PR调节剂还可以改善54项多发性硬化症的生活质量(MSQOL-54)身体健康综合评分(P=0.0005)。
    结论:与干扰素β相比,S1PR调节剂治疗RRMS具有良好的疗效和安全性。根据后续试验,S1PR调节剂可以改善MSQOL-54身体健康综合评分,从而可能对神经系统恢复有益,这需要更多的研究来证实。
    BACKGROUND: As one kind of disease-modifying therapies, sphingosine-1-phosphate receptor (S1PR) modulators such as fingolimod, ozanimod, and siponimod have been approved or are being developed to treat multiple sclerosis (MS). Several randomized controlled trials (RCT) have been implemented to compare the efficacy and safety of S1PR modulators versus interferon beta in the treatment of people with relapsing-remitting multiple sclerosis (RRMS).
    METHODS: We searched RCTs which were implemented from January 2010 to June 2020 by searching PubMed, Embase, Cochrane Library databases, and the Central Register of Controlled Trials. Finally, five RCTs were included in our study after carefully choosing.
    RESULTS: We pooled 4304 patients from 5 RCTs. The primary outcome was the annualized relapse rate. We found that the annualized relapse rate in the S1PR modulator group is 20% less than that in the interferon beta group (95%CI, - 0.32 to - 0.07, P = 0.002). S1PR modulators led to a significant reduction in number of new or enlarging T2 lesions per scan and number of gadolinium-enhancing lesions compared with interferon beta. Moreover, S1PR modulators can also improve 54-item multiple sclerosis quality of life (MSQOL-54) physical health composite score (P = 0.0005).
    CONCLUSIONS: S1PR modulators exhibited good efficacy and safety for the treatment of RRMS compared with interferon beta. According to follow-up trials, S1PR modulators can improve MSQOL-54 physical health composite score so that it may be beneficial to neurological recovery which need more research to confirm.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    2019年冠状病毒病(COVID-19)正在全球迅速蔓延。药物治疗是主要的治疗方法之一,但是不同个体之间的临床试验结果相互矛盾。此外,目前尚缺乏对个性化药物治疗的综合分析。在这项研究中,对用于COVID-19个性化治疗的47种特征明确的COVID-19药物进行了分析。
    收集了已发表的COVID-19治疗药物使用结果的临床试验,以评估药物疗效。对药物相互作用(DDI)进行了总结和分类。收集并系统分析了可操作药物基因的功能变化。定义并计算“基因评分”和“药物评分”,以系统地分析基于种族的遗传差异,这对更安全地使用COVID-19药物很重要。
    我们的结果表明,四种抗病毒药物(利托那韦,darunavir,daclatasvir和sofosbuvir)和三种免疫调节剂(布地奈德,秋水仙碱和泼尼松)以及肝素和依那普利可能会产生最多的DDI,而常见的伴随使用药物。八种药物(利托那韦,daclatasvir,sofosbuvir,利巴韦林,干扰素α-2b,氯喹,羟氯喹(HCQ)和头孢曲松在所有种族中都具有可操作的药物基因组学(PGx)生物标志物。14种药物(利托那韦,daclatasvir,泼尼松,地塞米松,利巴韦林,HCQ,头孢曲松,锌,干扰素β-1a,remdesivir,左氧氟沙星,洛匹那韦,人免疫球蛋白G和氯沙坦)与患者的种族血统有关,显示出明显不同的药物基因组特征。
    我们建议特别是对于有合并症的患者,避免严重的DDI,预测,预防性,和个性化医疗(PPPM,下午3点)必须对COVID-19治疗应用策略,并且应该对具有可操作药物基因的药物进行基因测试,特别是在一些功能变异频率较高的种族群体中,正如我们的分析显示。我们还建议,与较高种族遗传差异相关的药物应在未来COVID-19管理的药物遗传学研究中优先考虑。为了促进我们的结果转化为临床实践,建议采用符合PPPM/3PM原则的方法。总之,建议的PPPM/3PM态度应该是COVID-19整体管理的强制性考虑。
    在线版本包含补充材料,可在10.1007/s13167-021-00247-0获得。
    UNASSIGNED: Coronavirus disease 2019 (COVID-19) is rapidly spreading worldwide. Drug therapy is one of the major treatments, but contradictory results of clinical trials have been reported among different individuals. Furthermore, comprehensive analysis of personalized pharmacotherapy is still lacking. In this study, analyses were performed on 47 well-characterized COVID-19 drugs used in the personalized treatment of COVID-19.
    UNASSIGNED: Clinical trials with published results of drugs use for COVID-19 treatment were collected to evaluate drug efficacy. Drug-to-Drug Interactions (DDIs) were summarized and classified. Functional variations in actionable pharmacogenes were collected and systematically analysed. \"Gene Score\" and \"Drug Score\" were defined and calculated to systematically analyse ethnicity-based genetic differences, which are important for the safer use of COVID-19 drugs.
    UNASSIGNED: Our results indicated that four antiviral agents (ritonavir, darunavir, daclatasvir and sofosbuvir) and three immune regulators (budesonide, colchicine and prednisone) as well as heparin and enalapril could generate the highest number of DDIs with common concomitantly utilized drugs. Eight drugs (ritonavir, daclatasvir, sofosbuvir, ribavirin, interferon alpha-2b, chloroquine, hydroxychloroquine (HCQ) and ceftriaxone had actionable pharmacogenomics (PGx) biomarkers among all ethnic groups. Fourteen drugs (ritonavir, daclatasvir, prednisone, dexamethasone, ribavirin, HCQ, ceftriaxone, zinc, interferon beta-1a, remdesivir, levofloxacin, lopinavir, human immunoglobulin G and losartan) showed significantly different pharmacogenomic characteristics in relation to the ethnic origin of the patient.
    UNASSIGNED: We recommend that particularly for patients with comorbidities to avoid serious DDIs, the predictive, preventive, and personalized medicine (PPPM, 3 PM) strategies have to be applied for COVID-19 treatment, and genetic tests should be performed for drugs with actionable pharmacogenes, especially in some ethnic groups with a higher frequency of functional variations, as our analysis showed. We also suggest that drugs associated with higher ethnic genetic differences should be given priority in future pharmacogenetic studies for COVID-19 management. To facilitate translation of our results into clinical practice, an approach conform with PPPM/3 PM principles was suggested. In summary, the proposed PPPM/3 PM attitude should be obligatory considered for the overall COVID-19 management.
    UNASSIGNED: The online version contains supplementary material available at 10.1007/s13167-021-00247-0.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Letter
    暂无摘要。
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    The virus-induced signaling adaptor (VISA) complex plays a critical role in the innate immune response to RNA viruses. However, the mechanism of VISA complex formation remains unclear. Here, we demonstrate that thioredoxin 2 (TRX2) interacts with VISA at mitochondria both in vivo and in vitro Knockdown and knockout of TRX2 enhanced the formation of the VISA-associated complex, as well as virus-triggered activation of interferon regulatory factor 3 (IRF3) and transcription of the interferon beta 1 (IFNB1) gene. TRX2 inhibits the formation of VISA aggregates by repressing reactive oxygen species (ROS) production, thereby disrupting the assembly of the VISA complex. Furthermore, our data suggest that the C93 residue of TRX2 is essential for inhibition of VISA aggregation, whereas the C283 residue of VISA is required for VISA aggregation. Collectively, these findings uncover a novel mechanism of TRX2 that negatively regulates VISA complex formation.IMPORTANCE The VISA-associated complex plays pivotal roles in inducing type I interferons (IFNs) and eliciting the innate antiviral response. Many host proteins are identified as VISA-associated-complex proteins, but how VISA complex formation is regulated by host proteins remains enigmatic. We identified the TRX2 protein as an important regulator of VISA complex formation. Knockout of TRX2 increases virus- or poly(I·C)-triggered induction of type I IFNs at the VISA level. Mechanistically, TRX2 inhibits the production of ROS at its C93 site, which impairs VISA aggregates at its C283 site, and subsequently impedes the assembly of the VISA complex. Our findings suggest that TRX2 plays an important role in the regulation of VISA complex assembly.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    暂无摘要。
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    背景:多发性硬化症(MS)是一种自身免疫性疾病,中枢神经系统脱髓鞘疾病。MS的医治一向是神经学研讨的重点。迄今为止,美国食品和药物管理局已经批准了15种改善多发性硬化症病程的药物.在这项研究中,我们研究了疾病改善疗法(DMT)对临床结局的影响.
    方法:我们根据比较DMT在复发缓解型多发性硬化症(RRMS)患者中的随机对照试验(RCT)进行了系统评价和网络荟萃分析。我们搜索了Cochrane中央对照试验登记簿,MEDLINE,Embase,ClinicalTrials.gov和世界卫生组织国际临床试验注册平台RCTs发布至2018年10月31日。主要结果是疗效(24个月以上的复发率)和可接受性(24个月以上因不良事件而停止治疗)。
    结果:我们确定了23项合适的试验,包括14,096名参与者。在2年的随访中,所有药物均显著优于安慰剂.95%可信区间的风险比如下:阿仑单抗,0.49(0.40,0.59);奥克瑞珠单抗,0.49(0.40,0.61);米托蒽醌,0.47(0.27,0.80);那他珠单抗,0.51(0.43,0.61);芬戈莫德,0.57(0.50,0.65);聚乙二醇干扰素β-1a,0.63(0.52,0.77);富马酸二甲酯,0.65(0.56,0.74);特立氟胺14毫克,0.78(0.66,0.92);醋酸格拉替雷,0.80(0.72,0.89);IFNβ-1a(Rebif),0.81(0.72,0.90);IFNβ-1b(Betaseron),0.81(0.72,0.91);特立氟胺7毫克,0.83(0.71,0.98);和IFNβ-1a(Avonex)。0.87(0.77,0.99)。与安慰剂相比,因不良事件而停药的风险比从最佳药物(芬戈莫德)的1.12到最差药物(米托蒽醌)的0.10;持续(3个月)残疾进展的0.24(阿仑单抗)到0.89(IFNβ-1b[Betaseron]);严重不良事件的参与者人数从0.85(那他珠单抗)到1.25(特立氟胺14mg)。
    结论:在2年的随访中,所有DMT在降低复发率方面均优于安慰剂。至于药物之间的比较,阿仑单抗,奥克瑞珠单抗,与其他DMT相比,那他珠单抗和芬戈莫德的缓解率相对较高,退出率相对较低.
    BACKGROUND: Multiple sclerosis (MS) is an autoimmune, demyelinating disease of the central nervous system. The treatment of MS has always been a focus of neurological research. To date, the US Food and Drug Administration has approved 15 medications for modifying the course of multiple sclerosis. In this study, we examined the effects of disease-modifying therapies (DMTs) on clinical outcomes.
    METHODS: We did a systematic review and network meta-analysis based on randomized controlled trials (RCTs) comparing DMTs in patients with relapsing-remitting multiple sclerosis (RRMS). We searched the Cochrane Central Register of Controlled Trials, MEDLINE, Embase, ClinicalTrials.gov and the World Health Organization International Clinical Trials Registry Platform for RCTs published up to Oct 31, 2018. The primary outcome was efficacy (relapse rate over 24 months) and acceptability (treatment discontinuation due to adverse events over 24 months).
    RESULTS: We identified 23 suitable trials encompassing 14,096 participants. During the 2 years of follow-up, all drugs were significantly more effective than were placebos. The risk ratios with 95% credible intervals were as follows: alemtuzumab, 0.49 (0.40, 0.59); ocrelizumab, 0.49 (0.40, 0.61); mitoxantrone, 0.47 (0.27, 0.80); natalizumab, 0.51 (0.43, 0.61); fingolimod, 0.57 (0.50, 0.65); peginterferon beta-1a, 0.63 (0.52, 0.77); dimethyl fumarate, 0.65 (0.56, 0.74); teriflunomide 14 mg, 0.78 (0.66, 0.92); glatiramer acetate, 0.80 (0.72, 0.89); IFN β-1a (Rebif), 0.81 (0.72, 0.90); IFN β-1b (Betaseron), 0.81 (0.72, 0.91); teriflunomide 7 mg, 0.83 (0.71, 0.98); and IFN β-1a (Avonex). 0.87 (0.77, 0.99). Risk ratios compared with placebo for discontinuation due to adverse events ranged from 1.12 for the best drug (fingolimod) to 0.10 for the worst drug (mitoxantrone); from 0.24 (alemtuzumab) to 0.89 (IFNβ-1b [Betaseron]) for sustained (3-month) disability progression; and from 0.85 (natalizumab) to 1.25 (teriflunomide 14 mg) for the number of participants with serious adverse events.
    CONCLUSIONS: All DMTs were superior to placebo in reducing the relapse rate during the 2 years of follow-up. As to the comparison between drugs, alemtuzumab, ocrelizumab, natalizumab and fingolimod had a relatively higher response and lower dropout rates than did the other DMTs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Recombinant human interferon beta (rhIFN-β) is a glycoprotein produced by genetically engineered cells and has anti-virus, anti-tumor and immunoregulation functions. Although studies have shown that other subtypes of IFN such as IFN-γ affects cell proliferation and differentiation to some extent, the effect of rhIFN-β on chondrogenic differentiation of human bone marrow mesenchymal stem cells (hMSCs) is less known. In this study we studied the effect of rhIFN-β on the chondrogenic differentiation of hMSCs by inducing hMSCs into cartilage pellet via adding IFN-β1a into regular TGF-β3 chondrogenic differentiation medium. We collected the induced pellets and then detected GAG content, assessed pellets size, observed agreecan using alcian blue staining, and analyzed the expression of Sox and CollangenⅡusing real-time PCR and Western blotting. Addition of 100 ng/mL IFN-β1a to regular TGF-β3 chondrogenic differentiation medium could improve the concentration of GAG, increase the size of pellets, promote the formation of aggrecan and up-regulate the expression of CollangenII and Sox9. IFN-β1a combined with TGF-β3 could promote chondrogenic differentiation of hMSCs.
    重组人干扰素β (rhIFN-β) 是通过基因表达的一种糖蛋白,具有抗病毒、抗肿瘤和免疫调节作用,虽有研究表明其他亚型干扰素如干扰素γ 对细胞增殖和分化有一定的作用,但rhIFN-β 对人骨髓间充质干细胞(hMSCs) 诱导分化的影响尤其是成软骨细胞定向分化的研究尚少。文中采用成球法研究在常规TGF-β3 诱导分化培养基中添加重组IFN-β1a 后对诱导hMSCs 分化成软骨球的影响。hMSCs 经诱导分化后,收集软骨球,通过定量检测糖胺多糖 (GAG) 含量、软骨球形态测定、Alcian Blue 组织染色、Real-time PCR 和Western blotting 检测Sox9 和Collangen Ⅱ的表达。结果显示,在常规TGF-β3 诱导分化培养基中添加100 ng/mL IFN-β1a 能显著提高GAG 含量,增大软骨球尺寸,促进聚集蛋白聚糖形成,上调Sox9 和Collangen Ⅱ的表达。研究结果表明,重组人IFN-β1a 能够与TGF-β3 联合作用促进hMSCs 成软骨细胞定向分化。.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    Alemtuzumab is a humanised monoclonal antibody that alters the circulating lymphocyte pool, causing prolonged lymphopenia, thus remoulding the immune repertoire that accompanies homeostatic lymphocyte reconstitution. It has been proved more effective than interferon (IFN) 1a for the treatment of relapsing-remitting multiple sclerosis (RRMS).
    To compare the efficacy, tolerability and safety of alemtuzumab versus interferon beta 1a in the treatment of people with RRMS to prevent disease activity.
    We searched the Cochrane Multiple Sclerosis and Rare Diseases of the CNS Group Trials Register (1 February 2017) which, among other sources, contains records from CENTRAL, MEDLINE, Embase, CINAHL, LILACS, PEDRO and the trial registry databases Clinical Trials.gov and WHO International Clinical Trials Registry Platform for all prospectively registered and ongoing trials.
    All double-blind, randomised, controlled trials comparing intravenous alemtuzumab (12 mg per day or 24 mg per day on five consecutive days during the first month and on three consecutive days at months 12 and 24) versus subcutaneous IFN beta 1a (22 μg or 44 μg three times per week (Rebif) or intramuscular injection 30 μg once a week (Avonex)) in people of any gender and age with RRMS.
    We used standard methodological procedures expected by Cochrane.
    We included three trials involving 1694 participants. All trials compared alemtuzumab 12 mg per day or 24 mg per day versus IFN beta 1a for treating RRMS. In CAMMS223, participants received either subcutaneous IFN beta 1a 44 μg three times per week or annual intravenous cycles of alemtuzumab (at a dose of 12 mg per day or 24 mg per day) for 36 months. In CARE-MS I and CARE-MS II, participants received subcutaneous IFN beta 1a 44 μg three times per week or annual intravenous cycles of alemtuzumab 12 mg per day for 24 months. The methodological quality was good for all three studies.In the alemtuzumab 12 mg per day group, the results showed statistically significant difference in reducing relapses (risk ratio (RR) 0.60, 95% confidence interval (CI) 0.52 to 0.70), preventing disease progression (RR 0.60, 95% CI 0.45 to 0.79) and developing new T2 lesions on magnetic resonance imaging (RR 0.75, 95% CI 0.61 to 0.93) after 24 and 36 months\' follow-up, but found no statistically significant difference in the changes of Expanded Disability Status Scale (EDSS) score (mean difference (MD) -0.35, 95% CI -0.73 to 0.03). In the alemtuzumab 24 mg per day group, the results showed statistically significant differences in reducing relapses (RR 0.38, 95% CI 0.23 to 0.62), preventing disease progression (RR 0.42, 95% CI 0.21 to 0.84) and the changes of EDSS score (MD -0.83, 95% CI -1.17 to -0.49) after 36 months\' follow-up.All three trials reported adverse events and serious adverse events. There was no statistically significant difference in the number of participants with at least one adverse event (RR 1.03, 95% CI 0.97 to 1.08) and the number of participants who experienced serious adverse events (RR 1.03, 95% CI 0.83 to 4.54).
    There is low- to moderate-quality evidence that annual intravenous cycles of alemtuzumab at a dose of 12 mg per day or 24 mg per day reduces the proportion of participants with relapses, disease progression, change of EDSS score and developing new T2 lesions on MRI over 24 to 36 months in comparison with subcutaneous IFN beta-1a 44 μg three times per week.Alemtuzumab appeared to be relatively well tolerated. The most frequently reported adverse events were infusion-associated reactions, infections and autoimmune events. The use of alemtuzumab requires careful monitoring so that potentially serious adverse effects can be treated early and effectively.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号