个人资料
正文

Tofersen治疗萎缩侧索硬化症副作用

(2025-10-10 23:26:32) 下一个

反义寡核苷酸Tofersen治疗SOD1基因突变型肌萎缩侧索硬化症(ALS)的试验

https://www.nejm.org/doi/full/10.1056/NEJMoa2204705

作者:Timothy M. Miller,医学博士,理学博士,Merit奖
发表于2022年9月21日
N Engl J Med 2022;387:1099-1110
DOI: 10.1056/NEJMoa2204705
第387卷 第12期

摘要
背景
鞘内注射反义寡核苷酸Tofersen可降低超氧化物歧化酶1 (SOD1) 蛋白的合成,目前正在SOD1基因突变相关的肌萎缩侧索硬化症 (ALS) 患者中进行研究。

方法
在这项3期试验中,我们以2:1的比例随机分配患有SOD1 ALS的成年人,在24周内分别接受8剂托弗森(100毫克)或安慰剂治疗。主要终点是预测病情进展较快的参与者,其ALS功能评定量表修订版(ALSFRS-R;范围:0至48,分数越高表示功能越好)总分从基线到第28周的变化。次要终点包括脑脊液(CSF)中SOD1蛋白总浓度的变化、血浆中神经丝轻链浓度的变化、慢肺活量的变化以及16块肌肉的手持式测力计的变化。对该试验的随机部分及其52周开放标签扩展部分的综合分析,比较了试验开始时开始使用托弗森(早期开始队列)的参与者与第28周从安慰剂转为该药物的参与者(延迟开始队列)的结果。

研究摘要
反义寡核苷酸托弗森治疗SOD1型肌萎缩侧索硬化症(ALS)的试验

结果
共72名参与者接受了托弗森治疗(其中39名预计进展更快),36名接受了安慰剂治疗(其中21名预计进展更快)。与安慰剂相比,托弗森使脑脊液中SOD1浓度和血浆中神经丝轻链浓度的降低幅度更大。在进展较快的亚组(主要分析)中,tofersen 组和安慰剂组患者至第 28 周的 ALSFRS-R 评分变化分别为 -6.98 和 -8.14(差异为 1.2 分;95% 置信区间 [CI],-3.2 至 5.5;P=0.97)。两组患者的次要临床终点结果无显著差异。共有 95 名参与者(88%)进入开放标签延伸研究。52 周时,早期开始队列的 ALSFRS-R 评分变化为 -6.0,延迟开始队列为 -9.5(差异为 3.5 分;95% CI,0.4 至 6.7);其他终点的非多重校正差异有利于早期开始 tofersen 组。腰椎穿刺相关不良事件常见。7% 的 tofersen 接受者发生神经系统严重不良事件。
结论
在SOD1型ALS患者中,托弗森(tofersen)在28周内降低了脑脊液中SOD1的浓度和血浆中神经丝轻链的浓度,但并未改善临床终点,且与不良事件相关。托弗森(tofersen)早期给药与延迟给药的潜在效果正在扩展阶段进一步评估。 (由 Biogen 资助;VALOR 和 OLE ClinicalTrials.gov 编号为 NCT02623699 和 NCT03070119;EudraCT 编号为 2015-004098-33 和 2016-003225-41。)

快速摘要
Tofersen 治疗 SOD1 ALS
2 分 18 秒
约 2% 的肌萎缩侧索硬化症 (ALS) 病例与编码超氧化物歧化酶 1 (SOD1) 基因的突变有关。1,2 已描述了 200 多种 ALS 相关的 SOD1 突变,并且这些突变与不同的疾病进展速度相关。3-7 该疾病中的神经元变性被认为是由突变 SOD1 蛋白的毒性功能获得引起的。1,8–12 Tofersen 是一种鞘内注射的反义寡核苷酸,旨在减少通过诱导核糖核酸酶 H 介导的 SOD1 信使 RNA 降解来抑制 SOD1 蛋白。10,13–15 我们开展了一项为期 28 周的 3 期随机试验,评估托弗森 (tofersen) 对 SOD1 ALS 成人患者的疗效和安全性。本试验为三部分试验的 C 部分 (VALOR)。前两部分为剂量递增试验,旨在评估 C 部分中托弗森的剂量(参见方案,方案与本文全文可在 NEJM.org 获取)。15 已参加 A 部分和 B 部分的受试者未参加 C 部分。完成本试验后,受试者有机会参加正在进行的开放标签扩展研究。
方法
试验监督
本试验按照国际协调会 (ICHC) 的《药物临床试验质量管理规范》(GCP) 指南和《赫尔辛基宣言》中概述的伦理原则进行。VALOR 及其开放标签扩展研究的方案均已获得相关伦理委员会的批准。独立数据监测委员会审查了安全性数据。随机阶段和开放标签扩展阶段均已签署书面知情同意书。由参与者或其法定代表人提供。
申办方Biogen和作者设计了这些试验。Biogen提供了托法森和安慰剂,监督了试验,进行了统计学分析,并支付了医学写作协助费用。Biogen和作者分析了数据。论文初稿由第一作者和Biogen雇用的一位作者撰写。申办方审阅了论文稿,但不能延迟或阻止结果的发表。作者审阅并批准了论文稿的修订,并保证数据的准确性和完整性、试验对方案的忠实度以及不良事件报告的准确性。作者与Biogen之间签订了保密协议。
试验设计
本试验的3期、双盲、随机、安慰剂对照VALOR部分于2019年3月至2021年7月进行。16 参与者在10个国家的32个研究中心入组(参见补充附录S1部分,可在NEJM.org获取)。该试验包括4周的筛选期、24周的治疗期和4至8周的随访期,随后是持续的延长期。
参与者按2:1的比例随机分配,通过腰椎穿刺接受鞘内推注15毫升托弗森溶液(100毫克)或等量的安慰剂(人工脑脊液[CSF]),给药时间为24周,每2周注射3剂,之后每4周注射5剂(补充附录图S1)。随机化分层依据基线时是否使用依达拉奉、利鲁唑或两者同时使用,以及参与者是否符合疾病进展更快的预后标准,这些预后标准基于 SOD1 突变类型和肌萎缩侧索硬化症功能评定量表修订版 (ALSFRS-R) 评分的估计斜率,从症状出现到筛选时计算(“随机化前 ALSFRS-R 斜率”)。由于 ALSFRS-R 评分可能出现非线性进展,突变内变异性可能混淆这些指标的预后价值,且文献支持使用神经丝轻链作为疾病进展的预后标志物,16-24 因此在获得 VALOR 结果之前,预先设定了根据血浆中神经丝轻链基线浓度(高于或低于试验人群的中位浓度)定义的亚组分析(参见补充附录 S2 部分)。
完成 VALOR 后,参与者可以选择参加最长 236 周的开放标签扩展试验,同时不了解他们在 VALOR 中的试验组分配。VALOR 及其开放标签扩展试验第 52 周的合并分析是预先设定的,旨在比较整个意向治疗人群中提前开始和延迟开始托弗森治疗的情况。扩展阶段仍在进行中,计划在所有参与者完成至少3.5年的随访后进行分析,但目前尚未达到。
参与者
我们纳入了因ALS导致肌萎缩并确诊SOD1突变的成年人。主要分析人群是符合试验定义的快速进展疾病预后标准(参见补充附录S2部分)的参与者亚组,称为“快速进展亚组”。此外,还进行了随机分组,纳入了不符合这些扩展标准、预计疾病进展较慢的参与者亚组,即慢速进展亚组。这些参与者未被纳入主要终点分析,但有机会参加开放标签扩展研究,接受托弗森治疗。在评估 VALOR 及其开放标签扩展试验的综合数据时,无论在试验的随机部分预测其疾病进展速度是快还是慢,在 VALOR 中开始使用 tofersen 的参与者都被称为“早期启动队列”。在 VALOR 中接受安慰剂治疗,并在约 28 周后的开放标签扩展试验中有机会换用 tofersen 的参与者被称为“延迟启动队列”。
终点
VALOR 的主要疗效终点是快速进展亚组患者从基线到第 28 周的 ALSFRS-R 总分变化。ALSFRS-R 包含 12 个项目,涵盖四个功能子领域(延髓运动、精细运动、粗大运动和呼吸),总分范围为 0 至 48 分,分数越高表示功能越好。预先指定的次要终点包括脑脊液中 SOD1 蛋白总浓度与基线的变化、血浆中神经丝轻链浓度的变化、预测慢肺活量百分比(体积标准化为预测正常值的百分比)ue(根据年龄、性别和身高确定)、手持式测力计 megascore(手臂和腿部 16 个肌肉群的 z 分数平均值,值越高表示力量越大)、死亡或永久通气时间(连续 ≥21 天,每天机械通气≥22 小时)、死亡时间和安全性。预先指定的探索性终点包括参与者报告的结果测量,例如五项肌萎缩侧索硬化症评估问卷、疲劳(疲劳严重程度量表)和生活质量(EuroQol 五维问卷)。在 VALOR 和开放标签扩展的合并分析中也评估了相同的终点。
统计分析
我们计算得出,在快速进展的主要分析亚组中,样本量为60名参与者(随机化比例为2:1),根据联合秩和检验(如下所述),将提供84%的检验功效来检测组间差异。假设托弗森组ALSFRS-R评分从基线到第28周的变化为-4.8,安慰剂组为-24.7,标准差为20.39;双侧α水平为0.05,托弗森组生存率为90%,安慰剂组生存率为82%。在快速进展亚组中,对试验28周随机部分的所有主要和次要终点进行了正式检验。在慢速进展亚组中,只有脑脊液中总SOD1浓度具有统计学显著性检验功效,并且是该人群的主要终点(表S3)。在分析 ALSFRS-R 评分变化时,采用联合秩和检验进行统计推断。该检验方法同时考虑了功能衰退和生存率,并允许对治疗效果进行统计检验,同时考虑了因死亡导致的数据截断。联合秩和评分的计算方法是将每位参与者的 ALSFRS-R 评分从基线到第 28 周的变化与试验中其他每位参与者的变化进行比较,如果结果优于被比较的参与者,则得分为 1,如果结果更差,则得分为 -1,如果结果相同,则得分为 0。根据死亡时间,将死亡的参与者排名最低,首次服药后死亡时间最短的参与者排名依次降低。使用协方差分析 (ANCOVA) 评估每位参与者的个人分数总和(即排序分数)。
ALSFRS-R 排序评分的 ANCOVA 模型将试验组作为固定效应,并根据协变量(自症状出现以来的基线疾病持续时间、基线 ALSFRS-R 总分以及利鲁唑或依达拉奉的使用情况)进行了校正。估计的组间差异是通过 ALSFRS-R 评分相对于基线变化的 ANCOVA 模型获得的。VALOR 未指定对所有随机分配的参与者(无论预测进展更快或更慢)的总体人群进行正式的统计检验,但 ANCOVA 提供了相对于基线变化的估计值。联合秩分析与多重填补相结合,以解释由于未计入死亡的退出而导致的缺失数据。多重填补模型包括试验组、利鲁唑或依达拉奉的使用情况以及基线 ALSFRS-R 评分。其他亚组和探索性终点及分析在补充附录 S3 和 S4 部分中描述。
如果两个试验组的主要终点结果存在显著差异,则采用序贯封闭式检验程序按以下顺序对进展较快亚组的次要终点进行检验:脑脊液中SOD1蛋白总浓度从基线(与基线的比值)至第28周的变化、血浆中神经丝轻链浓度从基线(与基线的比值)至第28周的变化、慢肺活量占预计值百分比从基线至第28周的变化、手持式测力计Meascore从基线至第28周的变化、无呼吸机辅助生存期和总生存期。所有连续终点均采用ANCOVA分析相对于基线的变化,并结合多重填补法处理因退出而产生的缺失数据。慢肺活量的主要统计学推断采用联合秩分析和多重填补法。对于生存分析,对于不符合终点定义的参与者,在试验结束时或退出之日将数据删失。仅纳入经独立终点裁定委员会裁定的事件。治疗效果评估采用0.05的双侧显著性水平。
对VALOR及其开放标签扩展试验的数据进行合并分析,以评估托弗森早期启动与延迟启动的疗效,首次数据截止时间为2021年7月。开放标签扩展试验的第二次数据截止时间为2021年7月。

张力试验于 2022 年 1 月 16 日进行,当时在 VALOR 中接受随机分组的最后一名参与者有机会从 VALOR 开始进行至少 52 周的随访。这些数据的综合分析在此处呈现。在分析 2022 年 1 月数据截止日期的数据时,VALOR 的最终结果以及 VALOR 及其开放标签扩展的原始分析已在科学大会上发表;然而,参与者、研究人员和现场工作人员以及试验团队在扩展阶段仍然不知道 VALOR 中最初的试验组分配。
根据富集标准(快速进展和缓慢进展亚组)和基线血浆神经丝轻链中位浓度定义的分类亚组,对 VALOR 数据和开放标签扩展第一个数据截止日期的数据进行了预先指定的分析。认识到将连续变量作为协变量进行调整比将人群二分为分类亚组更能精确地控制个体异质性,我们在分析2022年1月数据截止日期之前修改了统计分析计划,将血浆中神经丝轻链的基线浓度作为分析之间的协变量(补充附录S2和S4部分)。
对2022年1月数据截止日期的数据进行合并分析是基于意向治疗原则,即所有在VALOR中接受随机分组的参与者(108名参与者)均根据其原始试验组分配纳入,无论其病情进展快慢、是否依从试验药物、是否提前终止试验或是否交叉到托弗森组。ANCOVA分析与多重填补相结合的方法与VALOR中的分析相同。 Kaplan-Meier生存分析纳入了截至2022年1月16日的所有数据,包括死亡时间或持续通气时间以及死亡时间;这些终点的组间比较基于对数秩检验,该检验根据试验组和基线血浆神经丝轻链中位浓度进行分层(补充附录S4部分和表S5)。由于合并分析中未计划调整多重比较的置信区间宽度,因此无法从这些结果中得出结论。
结果
参与者
VALOR试验共纳入108名携带42个独特SOD1突变(表S1)的参与者;72名参与者被分配接受托弗森治疗,36名参与者被分配接受安慰剂治疗。108名参与者中,共有60名构成了快速进展亚组,主要分析在该亚组中进行。共有 95 名 VALOR 参与者(88%)参与了开放标签扩展研究(图 S3)。合并分析的缺失数据量如下所示。两组参与者的基线临床特征相似,包括使用利鲁唑、依达拉奉或两者合用、疾病症状出现时间、基线 ALSFRS-R 评分以及预计慢肺活量百分比。然而,接受托弗森治疗的参与者的神经丝轻链基线浓度比接受安慰剂治疗的参与者高 15% 至 25%,并且接受托弗森治疗的参与者从筛选至第 15 天(约 42 天)的 ALSFRS-R 评分下降速度更快(表 1)。两组基线平均 ALSFRS-R 评分约为 37 分。
表 1

参与者基线人口统计学和临床??特征(意向治疗人群)。
终点
VALOR 的主要终点
在快速进展主要分析亚组的 60 名参与者中,从基线到第 28 周,托弗森组 ALSFRS-R 总分的变化为 -6.98 分,安慰剂组为 -8.14 分(差异为 1.2 分;95% 置信区间 [CI],-3.2 至 5.5;P=0.97)(表 2)。
表 2

快速进展亚组中 VALOR 的主要和次要终点。
VALOR 的次要终点
由于主要终点未达到统计学显著性,因此快速进展亚组中托弗森和安慰剂之间所有后续差异均视为无显著差异,因此未提供 P 值。在进展较快的亚组中,接受托弗森治疗的参与者脑脊液中SOD1蛋白的总浓度降低了29%(与基线的几何平均比值,0.71;95% CI,0.62~0.83),而接受安慰剂治疗的参与者脑脊液中SOD1蛋白的总浓度则增加了16%(与基线的几何平均比值,1.16;95% CI,0.96~1.40)(组间几何平均比值差异,0.62;95% CI,0.49~0.78)(表2)。在接受托弗森治疗的进展较慢的亚组中,脑脊液中SOD1蛋白的总浓度降低了40%,而安慰剂组的参与者脑脊液中SOD1蛋白的总浓度降低了19%。

接受安慰剂的慢进展亚组的ts降低(组间几何平均比值差异为0.74;95% CI,0.63~0.88)(表S4)。接受tofersen治疗的快进展亚组的血浆神经丝轻链平均浓度降低了60%,而接受安慰剂治疗的则增加了20%(组间几何平均比值差异为0.33;95% CI,0.25~0.45)(表2)。
在快进展亚组中,从基线到第28周,接受tofersen治疗的参与者的预测慢肺活量百分比下降了14.3个百分点,而接受安慰剂治疗的参与者的预测慢肺活量百分比下降了22.2个百分点(差异为7.9个百分点;95% CI,-3.5~19.3)(表2)。从基线到第 28 周,tofersen 组手持式测力计评分变化为 -0.34,安慰剂组为 -0.37(差异为 0.02;95% CI,-0.21 至 0.26)。由于事件数量少,无法估计死亡或永久通气的中位时间;tofersen 组(10%)和安慰剂组(10%)中死亡或需要永久通气的参与者百分比没有差异(风险比,1.39;95% CI,0.22 至 8.80)。无法估计死亡的中位时间,tofersen 组发生一例事件(3% 的参与者),安慰剂组无事件(表 2)。VALOR 期间进展较慢亚组的描述性分析见表 S4。
VALOR 联合开放标签扩展试验
VALOR 试验结束后,95 名参与者(88%)被纳入非随机开放标签扩展试验,其中 63 名(88%)最初被分配接受托弗森治疗,32 名(89%)最初被分配接受安慰剂治疗。截至最近一次数据截止时间(2022 年 1 月 16 日),早期启动队列中仍有 49 名参与者(68%),延迟启动队列中仍有 18 名参与者(50%)留在开放标签扩展试验中。所有 108 名在 VALOR 中接受随机分组的参与者均被纳入 VALOR 和开放标签扩展试验合并数据集的分析中,无论他们之前被纳入快速进展亚组还是慢速进展亚组。在早期启动的参与者中,脑脊液中总 SOD1 浓度和血浆中神经丝轻链浓度的降低在数值上随时间推移而持续;延迟启动队列参与者在开放标签扩展期也有类似的降低(图1)。52周时,早期启动队列参与者的ALSFRS-R评分相对于VALOR基线的变化为-6.0分,延迟启动队列参与者为-9.5分(差异为3.5分;95% CI,0.4-6.7)。早期启动队列中15名参与者(21%)和延迟启动队列中8名参与者(22%)的缺失数据需要填补(图2)。
图1

脑脊液(CSF)中总超氧化物歧化酶1(SOD1)浓度和血浆中神经丝轻链(NfL)浓度。
图2

临床功能和生存分析。
早期开始参与者的慢肺活量预测百分比相对于VALOR基线的变化为-9.4%,而延迟开始参与者的慢肺活量预测百分比相对于VALOR基线的变化为-18.6%(差异为9.2个百分点;95% CI,1.7-16.6)。早期开始参与者的手持式测力计评分相对于VALOR基线的变化为-0.17,而延迟开始参与者的手持式测力计评分相对于VALOR基线的变化为-0.45(差异为0.28;95% CI,0.05-0.52)。图2和表S5显示了VALOR和开放标签扩展的联合分析中ALSFRS-R评分、慢肺活量预测百分比和手持式测力计评分的结果。
由于事件数量有限,无法估计死亡或永久通气的中位时间以及死亡的中位时间。与延迟开始治疗的参与者相比,提前开始治疗的参与者死亡或持续使用呼吸机的时间风险比为0.36(95% CI,0.14 至 0.94),死亡时间风险比为0.27(95% CI,0.08 至 0.89)(表S5)。描述性分析显示,接受tofersen治疗的16例p.Ala5Val突变特别关注参与者的病程中位数为1.73年(范围,0.88 至 3.68年),其中3例参与者在数据截止时仍在试验中(3例仍在进行中参与者的范围为1.89 至 3.68年)(图S4)。
安全性和不良事件
VALOR试验和开放标签扩展试验中发生的大多数不良事件严重程度为轻度至中度,未导致停药或终止试验药物。大多数不良事件与ALS疾病进展、普通人群的疾病或腰椎穿刺的已知副作用一致(表3)。最常见的不良事件包括操作疼痛、头痛、手臂或腿部疼痛、跌倒和背痛。在VALOR研究中,接受tofersen治疗的参与者和接受安慰剂治疗的参与者的操作疼痛和头痛发生率相似,而

托弗森组手臂或腿部疼痛和背部疼痛更常见(发生率高出≥5个百分点),安慰剂组跌倒更常见。
表3

不良事件总结。
VALOR研究中接受托弗森治疗的4名参与者(6%)和开放标签扩展研究中的3名参与者(占所有接受托弗森治疗的参与者的7%)共发生8起神经系统严重不良事件,包括脊髓炎、化学性或无菌性脑膜炎、腰椎神经根病、颅内压升高和视乳头水肿。脊髓炎患者在第五次服用托弗森后约1周入院,接受了糖皮质激素和血浆置换治疗,未接受进一步的试验治疗。在最后一次服用托弗森后3个月内,该患者的神经系统体征、症状和影像学检查结果均得到缓解。
在VALOR试验中,托弗森组42名参与者(58%)和安慰剂组2名参与者(6%)至少有一次脑脊液白细胞计数超过10个/立方毫米,约40%的参与者基线时脑脊液蛋白浓度升高。托弗森组的中位脑脊液蛋白浓度增加了110毫克/升,而安慰剂组则降低了15毫克/升。在开放标签扩展试验中,也观察到了相似的脑脊液细胞增多和蛋白浓度升高发生率。
讨论
在试验的28周随机VALOR部分中,托弗森与脑脊液中SOD1蛋白总浓度(靶向作用的间接标志物)和血浆中神经丝轻链浓度(轴突损伤和神经退行性变的标志物)的降低相关。尽管有这些结果,在预测进展较快的亚组中,28 周时托弗森和安慰剂之间 ALSFRS-R 评分相对于基线的变化没有显著差异,并且该亚组中的其他临床终点也没有明显差异。在 52 周时对 VALOR 及其开放标签扩展进行的预设合并分析中,与 28 周后在开放标签扩展中开始使用托弗森的参与者相比,在 VALOR 开始时开始使用托弗森的参与者,无论进展快慢,ALSFRS-R 评分、预测慢肺活量百分比和手持式测力计评分的数值下降幅度较小。解释合并分析结果的局限性包括在分析早期开始和延迟开始队列之间的差异时未调整多重比较的置信区间宽度,大约 20% 的缺失终点数据需要填补,以及试验的 VALOR 部分的结果在分析时是已知的。
约7%接受tofersen治疗的受试者出现了神经系统严重不良事件,包括脊髓炎、化学性或无菌性脑膜炎、腰椎神经根病、颅内压增高和视乳头水肿。脊髓炎的潜在机制及其与脑脊液细胞增多和蛋白升高的关系尚不清楚。
在试验设计之初,SOD1突变类型和随机化前ALSFRS-R斜率被认为是解决SOD1 ALS疾病进展异质性的合适工具,但在短期试验期间,两者均不能始终如一地预测预后。尽管当时神经丝轻链的预后价值已被明确,但由于检测方法的局限性,我们无法根据个体受试者的神经丝轻链基线浓度进行随机分组,而这本可以更好地平衡各试验组。因此,我们的试验预先设定了亚组分析,并根据神经丝轻链的中位基线浓度进行定义。这种方法有助于解决基线特征的不平衡问题(从筛选到第 15 天,血浆中神经丝轻链的浓度和 ALSFRS-R 下降),但使用了任意亚组,而不是控制每个参与者的神经丝轻链基线浓度(参见补充附录 S2 部分)。为了解决个体疾病的异质性,将血浆中神经丝轻链的基线浓度作为分析的协变量。在获得 VALOR 结果以及 VALOR 和开放标签扩展的初步结果之后,但在进行最新的合并分析之前,对分析计划的这一改变是指定的。随着神经丝轻链检测变得越来越容易,在未来的 ALS 临床试验中可能会考虑将血浆中神经丝轻链浓度作为连续变量进行随机化。
VALOR 的持续时间和大小是根据 12 名 SOD1 突变携带者的现有但有限的数据确定的,这些患者患有快速进展的疾病

在 Tofersen 1-2 期多剂量递增研究 15 和阿利莫洛莫(一种促进新生蛋白质折叠的热休克蛋白辅助诱导剂)25 的 2 期试验中,接受安慰剂治疗的患者出现功能障碍。这些患者的功能在这些研究期间迅速下降。相比之下,在 VALOR 研究中,在强化快速进展亚组中接受安慰剂治疗的参与者的功能下降速度比数据预测的速度慢三倍。
在对VALOR和开放标签扩展试验的综合分析中,早期启动队列和延迟启动队列之间临床终点可能存在差异,且存在上述局限性,这表明可能需要超过28周的试验时间才能确定托弗森对此类疾病患者的疗效。26,27 正在进行的ATLAS试验(ClinicalTrials.gov注册号:NCT04856982)正在研究早期或症状前干预。28
在这项试验的28周VALOR部分中,SOD1型肌萎缩侧索硬化症(ALS)患者鞘内注射反义寡核苷酸托弗森,结果显示,与安慰剂相比,ALS进展综合指标的下降幅度没有显著差异。托弗森在少数参与者中发生了包括脊髓炎在内的不良事件。在正在进行的扩展阶段,正在进一步评估早期启动托弗森与延迟启动托弗森的潜在影响。
注释
作者提供的数据共享声明与本文全文可在 NEJM.org 上获取。
由 Biogen 赞助。
作者提供的披露表格与本文全文可在 NEJM.org 上获取。
我们感谢 VALOR 和开放标签扩展试验的参与者及其家人和护理人员,没有他们,本试验不可能完成;感??谢全球患者权益组织的成员;感谢研究中心工作人员(参见补充附录);感谢 Christine Nelson 药学博士(Biogen)和 Yien Liu 博士(Excel Scientific Solutions)对早期稿件提供的医学写作帮助;感谢 Cara Dickinson 文学士(Excel Scientific Solutions)根据期刊要求对早期稿件进行文字编辑和样式调整的帮助。
补充材料
方案 (nejmoa2204705_protocol.pdf)
下载
18.99 MB
补充附录 (nejmoa2204705_appendix.pdf)
下载
738.79 KB
披露表 (nejmoa2204705_disclosures.pdf)
下载
1.05 MB
数据共享声明 (nejmoa2204705_data-sharing.pdf)
下载
91.28 KB
参考文献
1.
Bunton-Stasyshyn RK, Saccon RA, Fratta P, Fisher EM. SOD1功能及其对肌萎缩侧索硬化症病理学的影响:新兴和复兴的主题。《神经科学家》2015;21:519-529。

Crossref
PubMed
Web of Science
Google Scholar
2.
Müller K, Brenner D, Weydt P 等。301 个德国 ALS 家系突变谱综合分析。J Neurol Neurosurg Psychiatry 2018;89:817-827。
前往引用
Crossref
PubMed
Web of Science
Google Scholar
3.
Huai J, Zhang Z. 家族性 ALS 相关 SOD1 突变体的结构特性及其相互作用伙伴。Front Neurol 2019;10:527-527。
前往引用
Crossref
PubMed
Web of Science
Google Scholar
4.
Bali T, Self W, Liu J 等。定义 SOD1 ALS 自然史以指导治疗性临床试验设计。J Neurol Neurosurg Psychiatry 2017;88:99-105。
前往引文
Crossref
PubMed
Web of Science
Google Scholar
5.
肌萎缩侧索硬化症在线数据库。SOD1 基因概要 (https://alsod.ac.uk/output/gene.php/SOD1)。
前往引文
Google Scholar
显示所有参考文献

职业中心
医生职位
2025年10月11日
马萨诸塞州
胃肠病学
高级或全科胃肠病学家
纽约州恩迪科特
家庭医学
家庭医学(纽约州恩迪科特)
加利福尼亚州伯林盖姆
内科
PAFMG - 内科医生(加利福尼亚州伯林盖姆)
田纳西州
血液学/肿瘤学
血液肿瘤学 | 田纳西州东部 |培训津贴
哥伦布,佐治亚州
肾病学
佐治亚州哥伦布市私人诊所肾病学职位!欢迎签证申请。
黎巴嫩,新罕布什尔州
血液科/肿瘤科
女性肿瘤科服务

Trial of Antisense Oligonucleotide Tofersen for SOD1 ALS

https://www.nejm.org/doi/full/10.1056/NEJMoa2204705

Authors: Timothy M. Miller, M.D., Ph.D., Merit

E. Cudkowicz, M.D., Angela Genge, M.D., Pamela J. Shaw, M.B., B.S., M.D., Gen Sobue, M.D., Ph.D., Robert C. Bucelli, M.D., Ph.D., Adriano Chiò, M.D., +19 , for the VALOR and OLE Working Group*Author Info & Affiliations
Published September 21, 2022
N Engl J Med 2022;387:1099-1110
DOI: 10.1056/NEJMoa2204705
 

Abstract

BACKGROUND

The intrathecally administered antisense oligonucleotide tofersen reduces synthesis of the superoxide dismutase 1 (SOD1) protein and is being studied in patients with amyotrophic lateral sclerosis (ALS) associated with mutations in SOD1 (SOD1 ALS).

METHODS

In this phase 3 trial, we randomly assigned adults with SOD1 ALS in a 2:1 ratio to receive eight doses of tofersen (100 mg) or placebo over a period of 24 weeks. The primary end point was the change from baseline to week 28 in the total score on the ALS Functional Rating Scale–Revised (ALSFRS-R; range, 0 to 48, with higher scores indicating better function) among participants predicted to have faster-progressing disease. Secondary end points included changes in the total concentration of SOD1 protein in cerebrospinal fluid (CSF), in the concentration of neurofilament light chains in plasma, in slow vital capacity, and in handheld dynamometry in 16 muscles. A combined analysis of the randomized component of the trial and its open-label extension at 52 weeks compared the results in participants who started tofersen at trial entry (early-start cohort) with those in participants who switched from placebo to the drug at week 28 (delayed-start cohort).

RESULTS

A total of 72 participants received tofersen (39 predicted to have faster progression), and 36 received placebo (21 predicted to have faster progression). Tofersen led to greater reductions in concentrations of SOD1 in CSF and of neurofilament light chains in plasma than placebo. In the faster-progression subgroup (primary analysis), the change to week 28 in the ALSFRS-R score was −6.98 with tofersen and −8.14 with placebo (difference, 1.2 points; 95% confidence interval [CI], −3.2 to 5.5; P=0.97). Results for secondary clinical end points did not differ significantly between the two groups. A total of 95 participants (88%) entered the open-label extension. At 52 weeks, the change in the ALSFRS-R score was −6.0 in the early-start cohort and −9.5 in the delayed-start cohort (difference, 3.5 points; 95% CI, 0.4 to 6.7); non–multiplicity-adjusted differences favoring early-start tofersen were seen for other end points. Lumbar puncture–related adverse events were common. Neurologic serious adverse events occurred in 7% of tofersen recipients.

CONCLUSIONS

In persons with SOD1 ALS, tofersen reduced concentrations of SOD1 in CSF and of neurofilament light chains in plasma over 28 weeks but did not improve clinical end points and was associated with adverse events. The potential effects of earlier as compared with delayed initiation of tofersen are being further evaluated in the extension phase. (Funded by Biogen; VALOR and OLE ClinicalTrials.gov numbers, NCT02623699 and NCT03070119; EudraCT numbers, 2015-004098-33 and 2016-003225-41.)
Approximately 2% of cases of amyotrophic lateral sclerosis (ALS) are associated with mutations in the gene encoding superoxide dismutase 1 (SOD1).1,2 More than 200 ALS-associated SOD1 mutations have been described and are associated with variable rates of progression.3-7 Neuronal degeneration in this disorder is considered to be caused by toxic gain of function of the mutant SOD1 protein.1,8–12 Tofersen is an intrathecally administered antisense oligonucleotide designed to reduce the synthesis of SOD1 protein by inducing RNase H–mediated degradation of SOD1 messenger RNA.10,13–15 We conducted a 28-week, phase 3, randomized trial of the efficacy and safety of tofersen in adults with SOD1 ALS. This is part C (VALOR) of a three-part trial, the first two parts of which were dose-escalation trials conducted to assess the dose of tofersen to be used in part C (see the protocol, available with the full text of this article at NEJM.org).15 Participants who were enrolled in parts A and B were not enrolled in part C. After completion of this trial, participants had the opportunity to enroll in an ongoing open-label extension.

Methods

TRIAL OVERSIGHT

The trials were conducted in accordance with the Good Clinical Practice guidelines of the International Council for Harmonisation and the ethical principles outlined in the Declaration of Helsinki. The protocols of VALOR and its open-label extension were approved by relevant ethics committees. An independent data monitoring committee reviewed safety data. Written informed consent for both the randomized phase and open-label extension was provided by participants or their legal representatives.
The sponsor, Biogen, and the authors designed these trials. Biogen provided tofersen and placebo, oversaw the trial, performed the statistical analyses, and paid for medical writing assistance. Biogen and the authors analyzed the data. The first draft of the manuscript was written by the first author and an author employed by Biogen. The sponsor reviewed the manuscript but could not delay or prevent publication of the results. The authors reviewed and approved revisions of the manuscript and vouch for the accuracy and completeness of the data, the fidelity of the trials to the protocols, and the accuracy of the reporting of adverse events. There were confidentiality agreements between the authors and Biogen.

TRIAL DESIGN

The phase 3, double-blind, randomized, placebo-controlled VALOR component of the trial was conducted from March 2019 through July 2021.16 Participants were enrolled at 32 sites in 10 countries (see Section S1 in the Supplementary Appendix, available at NEJM.org). The trial included a 4-week screening period, a 24-week treatment period, and a follow-up period of 4 to 8 weeks followed by an ongoing extension phase.
Participants were randomly assigned in a 2:1 ratio to receive an intrathecal bolus injection through a lumbar puncture of a 15-ml solution of tofersen (100 mg) or an equivalent volume of placebo (artificial cerebrospinal fluid [CSF]) administered over a period of 24 weeks, as three doses once every 2 weeks, followed by five doses once every 4 weeks (Fig. S1 in the Supplementary Appendix). Randomization was stratified according to the use or nonuse of edaravone, riluzole, or both at baseline and according to whether participants met prognostic criteria for faster disease progression that were based on SOD1 mutation type and the estimated slope of the score on the Amyotrophic Lateral Sclerosis Functional Rating Scale–Revised (ALSFRS-R), calculated from the time of symptom onset until screening (“prerandomization ALSFRS-R slope”). Owing to the potential for nonlinear progression on the ALSFRS-R score and for intra-mutation variability confounding the prognostic value of these measures, as well as literature supporting the use of neurofilament light chains as a prognostic marker of disease progression,16-24 analyses in subgroups that were defined according to baseline concentrations of neurofilament light chains in plasma (above vs. below the median concentration for the trial population) were prespecified before VALOR results were available (see Section S2 in the Supplementary Appendix).
After completion of VALOR, participants were given the option to participate in an open-label extension for up to 236 weeks, while remaining unaware of their trial-group assignment in VALOR. The combined analysis at week 52 of VALOR and its open-label extension was prespecified and was intended to enable comparison of early-start and delayed-start tofersen in the full intention-to-treat population. The extension phase is ongoing, and analysis is planned when all participants have completed at least 3.5 years of follow-up, which has not been reached.

PARTICIPANTS

We enrolled adults with weakness attributable to ALS and a confirmed SOD1 mutation. The primary analysis population was the subgroup of participants who met the trial-defined prognostic criteria for faster-progressing disease (see Section S2 in the Supplementary Appendix) and is called the “faster-progression subgroup.” Also undergoing randomization was a subgroup of participants who did not meet these enrichment criteria and were predicted to have slower progression of disease, the slower-progression subgroup. These persons were not included in the primary end-point analysis but had the opportunity to enroll in the open-label extension to receive tofersen. In the evaluation of combined data from VALOR and its open-label extension, participants who initiated tofersen in VALOR are referred to as the “early-start cohort,” regardless of whether they were predicted to have faster-progressing or slower-progressing disease in the randomized part of the trial. Those who received placebo in VALOR and had the opportunity to cross over to tofersen in the open-label extension approximately 28 weeks later are referred to as the “delayed-start cohort.”

END POINTS

The primary efficacy end point in VALOR was the change from baseline to week 28 in the ALSFRS-R total score in the faster-progression subgroup. The ALSFRS-R consists of 12 items across four subdomains of function (bulbar, fine motor, gross motor, and breathing), with total scores ranging from 0 to 48 and higher scores indicating better function. Prespecified secondary end points included the change from baseline in the total concentration of SOD1 protein in CSF, the concentration of neurofilament light chains in plasma, the percentage of the predicted slow vital capacity (volumes were standardized to the percentage of the predicted normal value on the basis of age, sex, and height), the handheld dynamometry megascore (average of z-scores across 16 muscle groups in the arms and legs, with higher values indicating greater strength), the time to death or permanent ventilation (≥22 hours of mechanical ventilation per day for ≥21 consecutive days), the time to death, and safety. Prespecified exploratory end points included participant-reported outcome measures such as the five-item Amyotrophic Lateral Sclerosis Assessment Questionnaire, fatigue (Fatigue Severity Scale), and quality of life (EuroQol Group 5-Dimension questionnaire). The same end points were assessed as part of the combined analyses of VALOR and the open-label extension.

STATISTICAL ANALYSIS

We calculated that a sample size of 60 participants (2:1 randomization ratio) in the faster-progression primary analysis subgroup would provide 84% power to detect a between-group difference on the basis of the joint rank test (described below), assuming a change in the ALSFRS-R score from baseline to week 28 of −4.8 in the tofersen group and −24.7 in the placebo group, with a standard deviation of 20.39 and survival of 90% in the tofersen group and 82% in the placebo group, at a two-sided alpha level of 0.05. All primary and secondary end points for the 28-week randomized part of the trial were formally tested in the faster-progression subgroup. In the slower-progression subgroup, only the total SOD1 concentration in CSF was powered to test for statistical significance and was the primary end point in this population (Table S3). The joint rank test was used for statistical inference in the analysis of the change in the ALSFRS-R score. This accounts for both functional decline and survival and allows for a statistical test of the treatment effect while accounting for truncation of data owing to deaths. The joint rank score was calculated by comparing the change in each participant’s ALSFRS-R score from baseline to week 28 with that of every other participant in the trial, resulting in a score of 1 if the outcome was better than that of the participant being compared, –1 if worse, and 0 if the same. Participants who died were ranked lowest on the basis of their time to death, with progressively lower ranks given to those who died in the shortest period of time after the first dose. The sum of individual scores for each participant (i.e., ranked score) was assessed with the use of analysis of covariance (ANCOVA).
The ANCOVA model for ranked scores on the ALSFRS-R included trial group as a fixed effect and was adjusted for covariates (baseline disease duration since symptom onset, baseline ALSFRS-R total score, and use of riluzole or edaravone). The estimated between-group difference was obtained from the ANCOVA model for change from baseline in the ALSFRS-R score. Formal statistical testing for the overall population of all randomly assigned participants (irrespective of faster or slower predicted progression) was not specified for VALOR, but estimates are provided from the ANCOVA for change from baseline. Joint rank analysis was performed in conjunction with multiple imputation to account for missing data due to withdrawals not accounted for by death. The multiple-imputation model included trial group, use of riluzole or edaravone, and the baseline ALSFRS-R score. Additional subgroup and exploratory end points and analyses are described in Sections S3 and S4 in the Supplementary Appendix.
If the results for the primary end point differed significantly between the two trial groups, secondary end points for the faster-progression subgroup were tested with the use of a sequential closed testing procedure in order of ranking: the change from baseline (ratio to baseline) to week 28 in the total concentration of SOD1 protein in CSF, the change from baseline (ratio to baseline) to week 28 in the concentration of neurofilament light chains in plasma, the change from baseline to week 28 in the percentage of the predicted slow vital capacity, the change from baseline to week 28 in handheld dynamometry megascore, ventilation assistance–free survival, and overall survival. ANCOVA for change from baseline was used for all continuous end points and in conjunction with multiple imputation for handling missing data for withdrawals. Primary statistical inference for slow vital capacity was by joint rank analysis with the use of multiple imputation. For survival analyses, data for participants who did not meet the end-point definition were censored at the end of the trial or on the date of withdrawal. Only events that were adjudicated by the independent end-point adjudication committee were included. Treatment effects were assessed at a two-sided significance level of 0.05.
The first data cutoff for the combined analysis of data from VALOR and its open-label extension to evaluate the effects of early as compared with delayed initiation of tofersen was performed in July 2021. A second data cutoff of the open-label extension was performed on January 16, 2022, when the last participant who underwent randomization in VALOR had the opportunity for at least 52 weeks of follow-up from the start of VALOR. The combined analyses of these data are presented here. At the time that data from the January 2022 data cutoff were analyzed, the final results from VALOR and the original analysis of VALOR and its open-label extension had been presented at a scientific congress; however, participants, investigators and site staff, and the trial team remained unaware during the extension phase of the original trial-group assignments in VALOR.
Prespecified analyses of the data from VALOR and the data as of the first data cutoff of the open-label extension were performed on the basis of enrichment criteria (fast-progression and slow-progression subgroups) and of categorical subgroups defined by the median concentration of neurofilament light chains in plasma at baseline. Recognizing that adjusting for a continuous variable as a covariate more precisely controls for individual heterogeneity than dichotomizing the population into categorical subgroups, we amended the statistical analysis plan before analysis of the January 2022 data cutoff to incorporate the baseline concentration of neurofilament light chains in plasma as a covariate across analyses (Sections S2 and S4 in the Supplementary Appendix).
The combined analyses of the data as the January 2022 data cutoff are based on the intention-to-treat principle, whereby all participants who underwent randomization in VALOR (108 participants) are included according to their original trial-group assignment, regardless of fast or slow progression, adherence to the trial agent, early termination of the trial, or crossover to the tofersen group. The ANCOVA analyses in conjunction with multiple imputation were conducted identically to the analyses in VALOR. Kaplan–Meier survival analyses included all data up to January 16, 2022, for time to death or permanent ventilation and time to death; between-group comparisons for these end points were based on a log-rank test stratified according to trial group and the median concentration of neurofilament light chains in plasma at baseline (Section S4 and Table S5 in the Supplementary Appendix). Because there was no plan for adjustment of the widths of confidence intervals for multiple comparisons in the combined analysis, no conclusions can be drawn from these results.

Results

PARTICIPANTS

A total of 108 participants with 42 unique SOD1 mutations (Table S1) were enrolled in VALOR; 72 were assigned to receive tofersen and 36 to receive placebo. A total of 60 of the 108 participants made up the faster-progression subgroup in which the primary analysis was performed. A total of 95 VALOR participants (88%) were enrolled in the open-label extension (Fig. S3). The amount of missing data for the combined analysis is given below. The clinical characteristics of the participants at baseline were similar in the two trial groups for use of riluzole, edaravone, or both, time from onset of disease symptoms, baseline ALSFRS-R score, and percentage of predicted slow vital capacity. However, baseline concentrations of neurofilament light chains were 15 to 25% higher in participants who received tofersen than in those who received placebo, and the rate of decline in the ALSFRS-R score from screening to day 15 (a period of approximately 42 days) was greater in the participants who received tofersen (Table 1). The mean ALSFRS-R score at baseline was approximately 37 in both groups.
TABLE 1
Demographic and Clinical Characteristics of the Participants at Baseline (Intention-to-Treat Population).

END POINTS

Primary End Point in VALOR

Among the 60 participants in the faster-progression primary analysis subgroup, the change in the ALSFRS-R total score from baseline to week 28 was –6.98 points in the tofersen group and –8.14 points in the placebo group (difference, 1.2 points; 95% confidence interval [CI], –3.2 to 5.5; P=0.97) (Table 2).
TABLE 2
Primary and Secondary End Points in VALOR in the Faster-Progression Subgroup.

Secondary End Points in VALOR

Because statistical significance was not achieved for the primary end point, all subsequent differences between tofersen and placebo in the faster-progression subgroup are considered to be not significantly different, and no P values are presented. In the faster-progression subgroup, the total concentration of SOD1 protein in CSF was reduced by 29% in participants who received tofersen (geometric mean ratio to baseline, 0.71; 95% CI, 0.62 to 0.83), as compared with an increase of 16% (geometric mean ratio to baseline, 1.16; 95% CI, 0.96 to 1.40) in those who received placebo (between-group difference in geometric mean ratio, 0.62; 95% CI, 0.49 to 0.78) (Table 2). The total concentration of SOD1 protein in CSF was reduced by 40% in the tofersen-treated slower-progression subgroup, as compared with a reduction by 19% in the participants in the slower-progression subgroup who received placebo (between-group difference in geometric mean ratio, 0.74; 95% CI, 0.63 to 0.88) (Table S4). The mean concentration of neurofilament light chains in plasma was reduced by 60% in the tofersen-treated faster-progression subgroup and increased 20% with placebo (between-group difference in geometric mean ratio, 0.33; 95% CI, 0.25 to 0.45) (Table 2).
In the faster-progression subgroup, the percentage of predicted slow vital capacity declined by 14.3 points from baseline to week 28 among participants who received tofersen and declined by 22.2 points among those who received placebo (difference, 7.9 percentage points; 95% CI, –3.5 to 19.3) (Table 2). The change from baseline to week 28 in handheld dynamometry megascore was −0.34 in the tofersen group and −0.37 in the placebo group (difference, 0.02; 95% CI, –0.21 to 0.26). The median time to death or permanent ventilation could not be estimated owing to the small number of events; no difference was observed in the percentage of participants who died or required permanent ventilation in the tofersen group (10%) or in the placebo group (10%) (hazard ratio, 1.39; 95% CI, 0.22 to 8.80). The median time to death could not be estimated, with one event (3% of participants) in the tofersen group and no events in the placebo group (Table 2). Descriptive analyses in the slower-progression subgroup during VALOR are provided in Table S4.

COMBINED VALOR AND OPEN-LABEL EXTENSION

After completion of VALOR, 95 participants (88%) were enrolled in the nonrandomized open-label extension, with 63 (88%) originally assigned to receive tofersen and 32 (89%) originally assigned to receive placebo. At the time of the most recent data cutoff (January 16, 2022), 49 participants (68%) in the early-start cohort and 18 (50%) in the delayed-start cohort remained in the open-label extension. All 108 participants who underwent randomization in VALOR were included in the analysis of the combined data set for VALOR and the open-label extension, whether they were previously included as part of the faster-progression or slower-progression subgroup. In early-start participants, reductions in the total SOD1 concentration in CSF and the concentration of neurofilament light chains in plasma were numerically sustained over time; delayed-start participants had similar reductions during the open-label extension (Figure 1). At 52 weeks, the change in the ALSFRS-R score from the VALOR baseline was −6.0 points for early-start participants and −9.5 points for delayed-start participants (difference, 3.5 points; 95% CI, 0.4 to 6.7). Imputation for week 52 was required for missing data in 15 participants (21%) in the early-start cohort and 8 participants (22%) in the delayed-start cohort (Figure 2).
FIGURE 1
Total Superoxidase Dismutase 1 (SOD1) Concentrations in Cerebrospinal Fluid (CSF) and Concentrations of Neurofilament Light Chains (NfL) in Plasma.
FIGURE 2
Analyses of Clinical Function and Survival.
The change in the percentage of predicted slow vital capacity from the VALOR baseline was −9.4% for early-start participants and −18.6% for delayed-start participants (difference, 9.2 percentage points; 95% CI, 1.7 to 16.6). The change in handheld dynamometry megascore from the VALOR baseline was −0.17 for early-start participants and −0.45 for delayed-start participants (difference, 0.28; 95% CI, 0.05 to 0.52). Figure 2 and Table S5 show the results of the ALSFRS-R score, the percentage of predicted slow vital capacity, and the handheld dynamometry megascore for the combined analyses of VALOR and the open-label extension.
The median time to death or permanent ventilation and the median time to death could not be estimated owing to the limited number of events. For early-start participants as compared with delayed-start participants, the hazard ratio for time to death or permanent ventilation was 0.36 (95% CI, 0.14 to 0.94), and the hazard ratio for time to death was 0.27 (95% CI, 0.08 to 0.89) (Table S5). In a descriptive analysis, the disease duration in the 16 participants of special interest with p.Ala5Val mutations who received tofersen was a median of 1.73 years (range, 0.88 to 3.68), with 3 of these participants remaining in the trial at the time of the data cutoff (range for the 3 ongoing participants, 1.89 to 3.68 years) (Fig. S4).

SAFETY AND ADVERSE EVENTS

Most adverse events across VALOR and the open-label extension were mild to moderate in severity and did not cause withdrawal or discontinuation of the trial agent. Most adverse events were consistent with ALS disease progression, conditions in the general population, or known side effects of lumbar puncture (Table 3). The most common adverse events included procedural pain, headache, pain in the arms or legs, falls, and back pain. In VALOR, the incidence of procedural pain and headache were similar among participants who received tofersen and among those who received placebo, whereas pain in the arms or legs and back pain were more common in the tofersen group (incidence higher by ≥5 percentage points) and falls were more common in the placebo group.
TABLE 3
Summary of Adverse Events.
Four participants who received tofersen in VALOR (6%) and three participants in the open-label extension (constituting 7% of all participants who received tofersen) had a total of eight neurologic serious adverse events, including myelitis, chemical or aseptic meningitis, lumbar radiculopathy, increased intracranial pressure, and papilledema. The participant with myelitis was hospitalized approximately 1 week after the fifth dose of tofersen, received glucocorticoids and plasma exchange, and received no further trial treatment. Within 3 months after the last dose of tofersen, this participant had resolution of neurologic signs, symptoms, and findings on imaging.
In VALOR, 42 participants (58%) in the tofersen group and 2 participants (6%) in the placebo group had at least one CSF white-cell count of more than 10 cells per cubic millimeter, and approximately 40% of the participants had elevated CSF protein concentrations at baseline. The median CSF protein concentration increased by 110 mg per liter in the tofersen group and decreased by 15 mg per liter in the placebo group. Similar incidences of CSF pleocytosis and elevated protein concentrations were observed during the open-label extension.

Discussion

In the 28-week randomized VALOR component of the trial, tofersen was associated with reductions in the total concentration of SOD1 protein in CSF, an indirect marker of target engagement, and the concentration of neurofilament light chains in plasma, a marker of axonal injury and neurodegeneration. Despite these results, no significant difference was seen at 28 weeks in the change from baseline in the ALSFRS-R score between tofersen and placebo in a subgroup predicted to have faster progression, and no definitive differences were seen in other clinical end points in this subgroup. At 52 weeks in a prespecified combined analysis of VALOR and its open-label extension, participants who started tofersen at the beginning of VALOR, irrespective of fast or slow progression, had a smaller numeric decline in the ALSFRS-R score, the percentage of predicted slow vital capacity, and handheld dynamometry megascore than those who started tofersen in the open-label extension 28 weeks later. Limitations in interpreting the results of the combined analysis include the absence of adjustment of the widths of confidence intervals for multiple comparisons in the analysis of differences between the early-start and delayed-start cohorts, approximately 20% of missing end-point data that required imputation, and the results of the VALOR component of the trial being known at the time of analysis.
Neurologic serious adverse events, including myelitis, chemical or aseptic meningitis, lumbar radiculopathy, increased intracranial pressure, and papilledema, occurred in approximately 7% of the participants receiving tofersen. The underlying mechanism of myelitis and the relationship to CSF pleocytosis and protein elevations could not be established.
At the time that the trial was designed, SOD1 mutation type and prerandomization ALSFRS-R slope were considered to be appropriate tools for addressing the heterogeneity of disease progression in SOD1 ALS, but neither is consistently prognostic over a short trial period. Although the prognostic usefulness of neurofilament light chains had been characterized at that time, assay limitations precluded randomization according to an individual participant’s baseline concentration of neurofilament light chains, which would have enabled better balance across trial groups. Instead, subgroup analyses were prespecified in our trial and defined according to the median baseline concentration of neurofilament light chains. This approach helped to address imbalances in baseline characteristics (concentration of neurofilament light chains in plasma and ALSFRS-R decline from screening to day 15) but made use of arbitrary subgrouping rather than controlling for each participant’s baseline concentration of neurofilament light chains (see Section S2 in the Supplementary Appendix). To address individual disease heterogeneity, the baseline concentration of neurofilament light chains in plasma was incorporated as a covariate across analyses. This alteration to the analysis plan was specified after the VALOR results and initial results from the combined VALOR and open-label extension were available but before the latest combined analysis was conducted. As testing of neurofilament light chains becomes more readily available, randomization based on the concentration of neurofilament light chains in plasma as a continuous variable may be considered in future ALS clinical trials.
The duration and size of VALOR were determined on the basis of available but limited data from 12 SOD1 mutation carriers with rapidly progressing disease who received placebo in the tofersen phase 1–2 multiple-ascending-dose study15 and the phase 2 trial of arimoclomol, a heat-shock protein coinducer that promotes nascent protein folding.25 These persons had a rapid decline in function over the period of these studies. In contrast, the participants who received placebo in the enriched faster-progression subgroup in VALOR had declines that were three times as slow as those projected by the data.
The possible signal of differences in clinical end points between the early-start and delayed-start cohorts in the combined analysis of VALOR and the open-label extension, with the limitations mentioned, suggests that a trial duration of more than 28 weeks may be required to determine the effect of tofersen in patients with this disorder.26,27 Earlier or presymptomatic intervention is being investigated in the ongoing ATLAS trial (ClinicalTrials.gov number, NCT04856982).28
In the 28-week VALOR component of this trial of intrathecal administration of the antisense oligonucleotide tofersen in patients with SOD1 ALS, there was not a significant difference in the decline on a composite measure of ALS progression as compared with placebo. Tofersen was associated in a limited number of participants with adverse events, including myelitis. The potential effects of earlier as compared with delayed initiation of tofersen are being further evaluated in the ongoing extension phase.

NOTES

data sharing statement provided by the authors is available with the full text of this article at NEJM.org.
Supported by Biogen.
Disclosure forms provided by the authors are available with the full text of this article at NEJM.org.
We thank the participants in VALOR and the open-label extension and their families and caregivers, without whom this trial would not have been possible; members of global patient advocacy organizations; site staff (see the Supplementary Appendix); Christine Nelson, Pharm.D. (Biogen), and Yien Liu, Ph.D. (Excel Scientific Solutions), for medical writing assistance with earlier versions of the manuscript; and Cara Dickinson, B.A. (Excel Scientific Solutions), for assistance with copyediting and styling of an earlier version of the manuscript in accordance with Journal requirements.

SUPPLEMENTARY MATERIAL

Protocol (nejmoa2204705_protocol.pdf)
Supplementary Appendix (nejmoa2204705_appendix.pdf)
Disclosure Forms (nejmoa2204705_disclosures.pdf)
Data Sharing Statement (nejmoa2204705_data-sharing.pdf)

REFERENCES

1.
Bunton-Stasyshyn RK, Saccon RA, Fratta P, Fisher EM. SOD1 function and its implications for amyotrophic lateral sclerosis pathology: new and renascent themes. Neuroscientist 2015;21:519-529.
2.
Müller K, Brenner D, Weydt P, et al. Comprehensive analysis of the mutation spectrum in 301 German ALS families. J Neurol Neurosurg Psychiatry 2018;89:817-827.
3.
Huai J, Zhang Z. Structural properties and interaction partners of familial ALS-associated SOD1 mutants. Front Neurol 2019;10:527-527.
4.
Bali T, Self W, Liu J, et al. Defining SOD1 ALS natural history to guide therapeutic clinical trial design. J Neurol Neurosurg Psychiatry 2017;88:99-105.
5.
Amyotrophic Lateral Sclerosis online Database. SOD1 gene summary (https://alsod.ac.uk/output/gene.php/SOD1).
 
[ 打印 ]
评论
目前还没有任何评论
登录后才可评论.