主题09 -临床试验和试验设计

IF 2.5 4区 医学 Q2 CLINICAL NEUROLOGY
S. Badri, J. Mason, S. Paganoni, J. Timmons, P. Yeramian, M. Cudkowicz, S. Appel, S. Ladha, N. Maragakis, M. Rivner, J. Katz, A. Genge, N. Olney, D. Lange, D. Heitzman, C. Bodkin, O., Jawdat, N. Goyal, J. Bornstein, C. Mak, S. Perrin
{"title":"主题09 -临床试验和试验设计","authors":"S. Badri, J. Mason, S. Paganoni, J. Timmons, P. Yeramian, M. Cudkowicz, S. Appel, S. Ladha, N. Maragakis, M. Rivner, J. Katz, A. Genge, N. Olney, D. Lange, D. Heitzman, C. Bodkin, O., Jawdat, N. Goyal, J. Bornstein, C. Mak, S. Perrin","doi":"10.1080/21678421.2022.2120685","DOIUrl":null,"url":null,"abstract":"Background: The safety and efficacy of sodium phenylbutyrate (PB) and taurursodiol (TURSO, also known as ursodoxicol- taurine) coformulation in ALS were evaluated in a phase 2 US multicenter trial (CENTAUR) encompassing both randomized (NCT03127514) and open-label extension (OLE; NCT03488524) phases. While overall treatment-emergent adverse event (TEAE) incidence was similar in the PB&TURSO versus placebo groups in the randomized phase and between those continuing versus switching to PB&TURSO in the OLE phase, treatment-emergent cardiac adverse events (TECAEs) and centrally read electrocardiographic abnormalities were reported more frequently among those receiving PB&TURSO in both phases. Objective: Describe the findings of independent expert reviews of reported TECAEs and electrocardiographic abnor- malities and of recorded electrocardiographic parameters in CENTAUR. Methods: Occurrence of TEAEs was assessed at each visit during both phases. Standard 12-lead electrocardiograms were performed at baseline in both phases and at a total of 2 visits in the randomized phase and up to 10 visits in the OLE phase, blinded and centrally read. Unblinded independent expert reviews assessed reported TECAEs and electrocar- diographic abnormalities for diagnostic accuracy and treatment emergence based on absence at baseline or lack Background: CD40L/CD40 costimulatory pathway mediates both humoral and adaptive immune responses in the pathophysiology of autoimmunity and transplant rejection. Studies have implicated CD40L (CD154) signaling and adaptive and innate immune cell activation in induction of neuroinflammation in neurodegenerative diseases. Methods: In this multicenter dose escalating phase 2 study 54 participants with a definite or probable diagnosis of ALS by El Escorial Criteria-revised received an anti CD154 antibody, tegoprubart, via intravenous infusion every two weeks for twelve weeks. Enrollment consisted of 9 participants in cohorts 1 and 2 (1 and 2mg/kg) and 18 participants in cohorts 3 and 4 (4 and 8mg/kg). The primary endpoint of the study was safety and tolerability. Secondary endpoints assessed the pharmacokinetics of tegoprubart as well as anti-drug antibody responses. Exploratory endpoints evaluated changes in disease progression utilizing the ALS Functional Rating Scale-Revised (ALSFRS-R), assessment of CD154 target engagement specifically looking at change in circulating CD40L and CXCL13, and changes in the levels of pro-inflam-matory biomarkers in circulation, by looking at change from baseline in circulating levels of a panel of inflammatory che-mokines and cytokines. Results: Seventy participants were screened, and fifty-four were enrolled in the study. Forty-nine completed the study (90.7%) receiving all six infusions of tegoprubart and com- pleting their final follow up visit. The most common TEAEs overall ( 10%) were fatigue (25.9%), falls (22.2%), headaches and muscle spasms (11.1%). Mean tegoprubart plasma concentrations increased proportionally with increasing dose with a half-life of approximately 24 days. Anti-drug antibody (ADA) titers were low and circulating levels of tego- prubart were as predicted for all cohorts. Tegopurbart demonstrated dose dependent target engagement as evidenced by a reduction in circulating levels of CD40L and CXCL13. While the study was not designed to detect clinical efficacy, target engagement was associated with a trend suggesting slowing in loss of function by change from baseline in ALSFRS-R. Tegoprubart also reduced multiple pro-inflamma- tory biomarkers in circulation in a dose dependent fashion, including TNF a , En-raged, IL-16, IL-18, CXCL9 and CXCL10 among others. The target engagement and inflammatory bio- markers showed increasing levels of reduction from the 1mg/ kg dose cohort to the 2mg/kg dose cohort and then pla- teaued at the 4mg/kg cohort. The changes were seen after the first dose and persisted throughout the treatment period. Conclusions: Tegoprubart was safe and well tolerated in adults with ALS demonstrating dose dependent functional activity to engage target and reduce pro-inflammatory che-mokines and cytokines associated with ALS. These results warrant further clinical studies with sufficient power and duration to assess clinical outcomes as a potential treatment for adults with ALS. Background: Platform trial approaches allow for simultan-eous evaluation of multiple investigational products by lever- aging a shared clinical trial infrastructure to reduce time and resources necessary to launch new investigations. Initiation of a platform trial requires tremendous clinical trial expertise, resources, and infrastructure to be successful. Objective: To share our operational experience from launch- ing the first Background: WVE-004 is an investigational stereopure antisense oligonucleotide that was designed to selectively lower transcripts derived from a hexanucleotide-repeat expansion mutation (G4C2) within the C9orf72 gene, which is the most common genetic cause of ALS and FTD. The repeat expan- sion reduces C9orf72 gene expression; transcription of the repeat leads to aberrant RNA foci and ultimately dipeptide repeat (DPR) proteins such as poly glycine-proline (poly(GP)). In preclinical mouse studies, WVE-004 produced substantial and durable reductions in repeat-containing C9orf72 tran- scripts and pathogenic poly(GP) without reducing C9orf72 protein expression (1). Results: FOCUS-C9 (NCT04931862) is a global, multicenter, placebo-controlled Phase 1b/2a trial with an adaptive design allowing for rapid assessment of the safety, tolerability, pharmacokinetics, and pharmacodynamic effects of single-and multiple-ascending intrathecal doses of WVE-004 in people with C9-ALS or C9-FTD. Initial results (up to day 85) showed significant reductions in CSF poly(GP) with single doses (10mg, n ¼ 2; 30mg, n ¼ 4; 60mg, n ¼ 3) with poly(GP) concentrations not apparently reaching steady-state maximal reduction by day 85. Adverse events (AEs) were balanced across treatment groups, including placebo ( n ¼ 3), and were mostly mild to moderate in intensity. Four patients, including one on placebo, experienced severe and/or serious AEs; one was reported to be related to study drug. These data suggest that single ascending doses of WVE-004 were generally safe, and reductions in CSF poly(GP) provide confirmation of tar- get engagement with a single dose. Based on these initial data, FOCUS-C9 was adapted to evaluate additional and expanded low, single-dose cohorts. Discussion: Treatment is ongoing, with additional data from single and multiple dose cohorts expected in 2022. New data emerging from the study will also be presented. FOCUS-C9 is the first clinical trial to address both ALS and FTD caused by the C9orf72 genetic variant in the same study. Further, FOCUS-C9 ’ s adaptive design has enabled data-driven modifi-cations to the study to support more rapid optimization of dose level and frequency for WVE-004. Introduction objective: Improved functional outcome measures in amyotrophic lateral sclerosis would aid amyo- trophic lateral sclerosis trial design and help hasten drug discovery. We evaluate the longitudinal performance of the Rasch-Built Overall Amyotrophic Lateral Sclerosis Disability Scale (ROADS) compared to the Amyotrophic Lateral Sclerosis Functional Rating Scale Revised for Self-Entry (ALSFRS-RSE) as patient reported outcomes of functional status in people with ALS. Methods: People with ALS completed the ROADS and the ALSFRS-RSE questionnaires at baseline, 3-, 6-, and 12-months using the web-based platform, Research Electronic Data Capture as a part of a prospective, longitudinal, remote, online survey study of fatigue in ALS from 9/2020 to 12/ 2021. The scales were compared cross-sectionally (at baseline) and longitudinally. Correlation coefficients, coefficient of variation, and descriptive statistics were assessed. Vital status was recorded at follow-up contact at approximately 12-months. Results: 182 adults with ALS consented to the study. This volunteer sample was comprised of predominantly White, non-Hispanic, non-smoking participants. Consented partici- pant survey completion was approximately 90% at baseline and greater than 40% at 12-months.The ALSFRS-RSE and the ROADS had high, significant agreement at 3- and 6 months by Cohen ’ s kappa > 71% ( p < 0.001); the number of functional increases or plateaus on each scale were not signifi- cantly different; and the coefficient of variation of functional decline was similar at the 6-month mark, though higher for the ROADS at 3-months and lower at 12-months. Background: Amyotrophic lateral sclerosis (ALS) therapeutic development has relied largely on staff-administered func- tional rating scales to determine treatment efficacy. Mobile applications (apps) and wearable devices offer multiple opportunities for biometric data collection. Hypothesis: Wearable and smartphone devices can collect patient-reported outcomes and passively collect sensor data that will provide quantitative measurement of ALS disease progression. Methods: Participants were remotely consented, enrolled and followed for 6 months. ALSFRS-R was administered at baseline, 3 and 6 months via phone call. The study app (Beiwe) was installed on the participants ’ personal smart-phones at baseline and delivered ALSFRS-R for Self-Entry (ALSFRS-RSE) and the Rasch Overall ALS Disability Scale (ROADS) surveys every 2 – 4 weeks. Participants also continu-ously wore either a wrist-worn activity monitor (ActiGraph Insight Watch) or an ankle-worn activity monitor (Modus StepWatch). Vendor-provided daily measures (VDMs) were obtained from each device. Investigator-derived daily meas- ures (IDMs) were calculated from ActiGraph ’ s minute-level activity counts data. Participants contributing at least two ALSFRS-RSE and ROADS surveys were included in this analysis. Days with at least 8hours of estimated activity monitor wear time were defined","PeriodicalId":7740,"journal":{"name":"Amyotrophic Lateral Sclerosis and Frontotemporal Degeneration","volume":"23 1","pages":"133 - 149"},"PeriodicalIF":2.5000,"publicationDate":"2022-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":"{\"title\":\"Theme 09 - Clinical Trials and Trial Design\",\"authors\":\"S. Badri, J. Mason, S. Paganoni, J. Timmons, P. Yeramian, M. Cudkowicz, S. Appel, S. Ladha, N. Maragakis, M. Rivner, J. Katz, A. Genge, N. Olney, D. Lange, D. Heitzman, C. Bodkin, O., Jawdat, N. Goyal, J. Bornstein, C. Mak, S. Perrin\",\"doi\":\"10.1080/21678421.2022.2120685\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"Background: The safety and efficacy of sodium phenylbutyrate (PB) and taurursodiol (TURSO, also known as ursodoxicol- taurine) coformulation in ALS were evaluated in a phase 2 US multicenter trial (CENTAUR) encompassing both randomized (NCT03127514) and open-label extension (OLE; NCT03488524) phases. While overall treatment-emergent adverse event (TEAE) incidence was similar in the PB&TURSO versus placebo groups in the randomized phase and between those continuing versus switching to PB&TURSO in the OLE phase, treatment-emergent cardiac adverse events (TECAEs) and centrally read electrocardiographic abnormalities were reported more frequently among those receiving PB&TURSO in both phases. Objective: Describe the findings of independent expert reviews of reported TECAEs and electrocardiographic abnor- malities and of recorded electrocardiographic parameters in CENTAUR. Methods: Occurrence of TEAEs was assessed at each visit during both phases. Standard 12-lead electrocardiograms were performed at baseline in both phases and at a total of 2 visits in the randomized phase and up to 10 visits in the OLE phase, blinded and centrally read. Unblinded independent expert reviews assessed reported TECAEs and electrocar- diographic abnormalities for diagnostic accuracy and treatment emergence based on absence at baseline or lack Background: CD40L/CD40 costimulatory pathway mediates both humoral and adaptive immune responses in the pathophysiology of autoimmunity and transplant rejection. Studies have implicated CD40L (CD154) signaling and adaptive and innate immune cell activation in induction of neuroinflammation in neurodegenerative diseases. Methods: In this multicenter dose escalating phase 2 study 54 participants with a definite or probable diagnosis of ALS by El Escorial Criteria-revised received an anti CD154 antibody, tegoprubart, via intravenous infusion every two weeks for twelve weeks. Enrollment consisted of 9 participants in cohorts 1 and 2 (1 and 2mg/kg) and 18 participants in cohorts 3 and 4 (4 and 8mg/kg). The primary endpoint of the study was safety and tolerability. Secondary endpoints assessed the pharmacokinetics of tegoprubart as well as anti-drug antibody responses. Exploratory endpoints evaluated changes in disease progression utilizing the ALS Functional Rating Scale-Revised (ALSFRS-R), assessment of CD154 target engagement specifically looking at change in circulating CD40L and CXCL13, and changes in the levels of pro-inflam-matory biomarkers in circulation, by looking at change from baseline in circulating levels of a panel of inflammatory che-mokines and cytokines. Results: Seventy participants were screened, and fifty-four were enrolled in the study. Forty-nine completed the study (90.7%) receiving all six infusions of tegoprubart and com- pleting their final follow up visit. The most common TEAEs overall ( 10%) were fatigue (25.9%), falls (22.2%), headaches and muscle spasms (11.1%). Mean tegoprubart plasma concentrations increased proportionally with increasing dose with a half-life of approximately 24 days. Anti-drug antibody (ADA) titers were low and circulating levels of tego- prubart were as predicted for all cohorts. Tegopurbart demonstrated dose dependent target engagement as evidenced by a reduction in circulating levels of CD40L and CXCL13. While the study was not designed to detect clinical efficacy, target engagement was associated with a trend suggesting slowing in loss of function by change from baseline in ALSFRS-R. Tegoprubart also reduced multiple pro-inflamma- tory biomarkers in circulation in a dose dependent fashion, including TNF a , En-raged, IL-16, IL-18, CXCL9 and CXCL10 among others. The target engagement and inflammatory bio- markers showed increasing levels of reduction from the 1mg/ kg dose cohort to the 2mg/kg dose cohort and then pla- teaued at the 4mg/kg cohort. The changes were seen after the first dose and persisted throughout the treatment period. Conclusions: Tegoprubart was safe and well tolerated in adults with ALS demonstrating dose dependent functional activity to engage target and reduce pro-inflammatory che-mokines and cytokines associated with ALS. These results warrant further clinical studies with sufficient power and duration to assess clinical outcomes as a potential treatment for adults with ALS. Background: Platform trial approaches allow for simultan-eous evaluation of multiple investigational products by lever- aging a shared clinical trial infrastructure to reduce time and resources necessary to launch new investigations. Initiation of a platform trial requires tremendous clinical trial expertise, resources, and infrastructure to be successful. Objective: To share our operational experience from launch- ing the first Background: WVE-004 is an investigational stereopure antisense oligonucleotide that was designed to selectively lower transcripts derived from a hexanucleotide-repeat expansion mutation (G4C2) within the C9orf72 gene, which is the most common genetic cause of ALS and FTD. The repeat expan- sion reduces C9orf72 gene expression; transcription of the repeat leads to aberrant RNA foci and ultimately dipeptide repeat (DPR) proteins such as poly glycine-proline (poly(GP)). In preclinical mouse studies, WVE-004 produced substantial and durable reductions in repeat-containing C9orf72 tran- scripts and pathogenic poly(GP) without reducing C9orf72 protein expression (1). Results: FOCUS-C9 (NCT04931862) is a global, multicenter, placebo-controlled Phase 1b/2a trial with an adaptive design allowing for rapid assessment of the safety, tolerability, pharmacokinetics, and pharmacodynamic effects of single-and multiple-ascending intrathecal doses of WVE-004 in people with C9-ALS or C9-FTD. Initial results (up to day 85) showed significant reductions in CSF poly(GP) with single doses (10mg, n ¼ 2; 30mg, n ¼ 4; 60mg, n ¼ 3) with poly(GP) concentrations not apparently reaching steady-state maximal reduction by day 85. Adverse events (AEs) were balanced across treatment groups, including placebo ( n ¼ 3), and were mostly mild to moderate in intensity. Four patients, including one on placebo, experienced severe and/or serious AEs; one was reported to be related to study drug. These data suggest that single ascending doses of WVE-004 were generally safe, and reductions in CSF poly(GP) provide confirmation of tar- get engagement with a single dose. Based on these initial data, FOCUS-C9 was adapted to evaluate additional and expanded low, single-dose cohorts. Discussion: Treatment is ongoing, with additional data from single and multiple dose cohorts expected in 2022. New data emerging from the study will also be presented. FOCUS-C9 is the first clinical trial to address both ALS and FTD caused by the C9orf72 genetic variant in the same study. Further, FOCUS-C9 ’ s adaptive design has enabled data-driven modifi-cations to the study to support more rapid optimization of dose level and frequency for WVE-004. Introduction objective: Improved functional outcome measures in amyotrophic lateral sclerosis would aid amyo- trophic lateral sclerosis trial design and help hasten drug discovery. We evaluate the longitudinal performance of the Rasch-Built Overall Amyotrophic Lateral Sclerosis Disability Scale (ROADS) compared to the Amyotrophic Lateral Sclerosis Functional Rating Scale Revised for Self-Entry (ALSFRS-RSE) as patient reported outcomes of functional status in people with ALS. Methods: People with ALS completed the ROADS and the ALSFRS-RSE questionnaires at baseline, 3-, 6-, and 12-months using the web-based platform, Research Electronic Data Capture as a part of a prospective, longitudinal, remote, online survey study of fatigue in ALS from 9/2020 to 12/ 2021. The scales were compared cross-sectionally (at baseline) and longitudinally. Correlation coefficients, coefficient of variation, and descriptive statistics were assessed. Vital status was recorded at follow-up contact at approximately 12-months. Results: 182 adults with ALS consented to the study. This volunteer sample was comprised of predominantly White, non-Hispanic, non-smoking participants. Consented partici- pant survey completion was approximately 90% at baseline and greater than 40% at 12-months.The ALSFRS-RSE and the ROADS had high, significant agreement at 3- and 6 months by Cohen ’ s kappa > 71% ( p < 0.001); the number of functional increases or plateaus on each scale were not signifi- cantly different; and the coefficient of variation of functional decline was similar at the 6-month mark, though higher for the ROADS at 3-months and lower at 12-months. Background: Amyotrophic lateral sclerosis (ALS) therapeutic development has relied largely on staff-administered func- tional rating scales to determine treatment efficacy. Mobile applications (apps) and wearable devices offer multiple opportunities for biometric data collection. Hypothesis: Wearable and smartphone devices can collect patient-reported outcomes and passively collect sensor data that will provide quantitative measurement of ALS disease progression. Methods: Participants were remotely consented, enrolled and followed for 6 months. ALSFRS-R was administered at baseline, 3 and 6 months via phone call. The study app (Beiwe) was installed on the participants ’ personal smart-phones at baseline and delivered ALSFRS-R for Self-Entry (ALSFRS-RSE) and the Rasch Overall ALS Disability Scale (ROADS) surveys every 2 – 4 weeks. Participants also continu-ously wore either a wrist-worn activity monitor (ActiGraph Insight Watch) or an ankle-worn activity monitor (Modus StepWatch). Vendor-provided daily measures (VDMs) were obtained from each device. Investigator-derived daily meas- ures (IDMs) were calculated from ActiGraph ’ s minute-level activity counts data. Participants contributing at least two ALSFRS-RSE and ROADS surveys were included in this analysis. Days with at least 8hours of estimated activity monitor wear time were defined\",\"PeriodicalId\":7740,\"journal\":{\"name\":\"Amyotrophic Lateral Sclerosis and Frontotemporal Degeneration\",\"volume\":\"23 1\",\"pages\":\"133 - 149\"},\"PeriodicalIF\":2.5000,\"publicationDate\":\"2022-11-01\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Amyotrophic Lateral Sclerosis and Frontotemporal Degeneration\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.1080/21678421.2022.2120685\",\"RegionNum\":4,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q2\",\"JCRName\":\"CLINICAL NEUROLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Amyotrophic Lateral Sclerosis and Frontotemporal Degeneration","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1080/21678421.2022.2120685","RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q2","JCRName":"CLINICAL NEUROLOGY","Score":null,"Total":0}
引用次数: 0

摘要

背景:美国一项2期多中心试验(CENTAUR)评估了苯丁酸钠(PB)和牛磺酸二醇(TURSO,也称为牛磺酸多醇-牛磺酸)联合配方治疗ALS的安全性和有效性,该试验包括随机(NCT03127514)和开放标签扩展(OLE;NCT03488524)阶段。虽然随机期PB&TURSO组与安慰剂组的总体治疗不良事件(TEAE)发生率相似,OLE期继续使用PB&TURSO组与改用PB&TURSO组之间的总体治疗不良事件(TEAE)发生率相似,但在两个阶段接受PB&TURSO的患者中,治疗出现的心脏不良事件(tecae)和中心阅读心电图异常的报告频率更高。目的:描述独立专家对CENTAUR报告的tecae和心电图异常以及记录的心电图参数的审查结果。方法:在两个阶段的每次访问中评估teae的发生情况。标准12导联心电图在两个阶段的基线上进行,随机阶段共2次访问,OLE阶段最多10次访问,盲法和中央读取。背景:CD40L/CD40共刺激通路在自身免疫和移植排斥的病理生理过程中介导体液和适应性免疫反应。研究表明,CD40L (CD154)信号和适应性和先天免疫细胞激活在神经退行性疾病中诱导神经炎症。方法:在这项多中心剂量递增的2期研究中,54名根据El Escorial标准修订的明确或可能诊断为ALS的参与者每两周静脉输注一次抗CD154抗体tegopruart,持续12周。入组1和2组9名受试者(1和2mg/kg), 3和4组18名受试者(4和8mg/kg)。研究的主要终点是安全性和耐受性。次要终点评估了地戈鲁巴的药代动力学以及抗药物抗体反应。探索性终点评估疾病进展的变化,使用ALS功能评定量表-修订版(ALSFRS-R),评估CD154靶点参与,特别是观察循环CD40L和CXCL13的变化,以及循环中促炎生物标志物水平的变化,通过观察一组炎症因子和细胞因子的循环水平从基线的变化。结果:70名参与者被筛选,54人被纳入研究。49例(90.7%)完成了研究,接受了全部六次输注地甲肽并完成了最后的随访。最常见的teae(10%)是疲劳(25.9%)、跌倒(22.2%)、头痛和肌肉痉挛(11.1%)。平均地鲁巴血药浓度随剂量增加成比例增加,半衰期约为24天。抗药物抗体(ADA)滴度较低,所有队列的tego- prubart循环水平与预测一致。Tegopurbart通过降低循环CD40L和CXCL13水平证明了剂量依赖性靶标作用。虽然该研究的目的不是检测临床疗效,但靶标接触与ALSFRS-R基线变化导致功能丧失减慢的趋势有关。tegopruart还以剂量依赖性的方式降低循环中的多种促炎生物标志物,包括TNF a、en -rage、IL-16、IL-18、CXCL9和CXCL10等。靶接触和炎症生物标志物显示,从1mg/ kg剂量组到2mg/kg剂量组,降低水平不断增加,然后在4mg/kg剂量组达到峰值。这种变化在第一次给药后出现,并在整个治疗期间持续存在。结论:替戈鲁巴在成人ALS患者中是安全且耐受性良好的,显示出剂量依赖性的功能活性,可以参与靶标并减少与ALS相关的促炎切因子和细胞因子。这些结果为进一步的临床研究提供了足够的力量和持续时间来评估作为成人ALS潜在治疗方法的临床结果。背景:平台试验方法允许通过杠杆化共享临床试验基础设施来同时评估多种研究产品,以减少启动新研究所需的时间和资源。平台试验的启动需要大量的临床试验专业知识、资源和基础设施才能成功。 背景:WVE-004是一种正在研究的体纯反义寡核苷酸,旨在选择性地降低来自C9orf72基因中六核苷酸重复扩增突变(G4C2)的转录本,这是ALS和FTD最常见的遗传原因。重复扩增降低了C9orf72基因的表达;重复序列的转录导致异常的RNA聚焦,最终产生二肽重复序列(DPR)蛋白,如聚甘氨酸-脯氨酸(聚(GP))。在临床前小鼠研究中,WVE-004在不降低C9orf72蛋白表达的情况下,显著且持久地降低了含有重复序列的C9orf72转录因子和致病性多聚(GP)(1)。FOCUS-C9 (NCT04931862)是一项全球性、多中心、安慰剂对照的1b/2a期临床试验,采用自适应设计,可快速评估C9-ALS或C9-FTD患者鞘内单次和多次递增剂量WVE-004的安全性、耐受性、药代动力学和药效学效应。初步结果(直至第85天)显示单剂量(10mg, n / 2;30mg, n¼4;60mg, n¼3),聚(GP)浓度在第85天没有明显达到稳态最大还原。不良事件(ae)在治疗组之间是平衡的,包括安慰剂组(n¼3),并且大多数是轻度到中度的强度。4名患者(包括1名安慰剂组患者)经历了严重和/或严重不良反应;据报道,其中一个与研究药物有关。这些数据表明,单次上升剂量的WVE-004通常是安全的,脑脊液聚(GP)的减少证实了单次剂量的目标接触。基于这些初始数据,FOCUS-C9被用于评估额外的和扩大的低单剂量队列。讨论:治疗正在进行中,预计将于2022年获得单剂量和多剂量队列的额外数据。研究中出现的新数据也将被介绍。FOCUS-C9是首个在同一项研究中解决由C9orf72基因变异引起的ALS和FTD的临床试验。此外,FOCUS-C9的自适应设计使数据驱动的研究修改成为可能,以支持WVE-004的剂量水平和频率的更快优化。前言目的:改善肌萎缩性侧索硬化症的功能结果测量将有助于肌萎缩性侧索硬化症的试验设计,并有助于加速药物的发现。我们评估了rasch构建的肌萎缩侧索硬化症总体残疾量表(ROADS)与肌萎缩侧索硬化症自我进入功能评定量表(ALSFRS-RSE)作为ALS患者报告的功能状态结果的纵向表现。方法:ALS患者在基线、3个月、6个月和12个月时使用基于网络的平台完成ROADS和ALSFRS-RSE问卷,研究电子数据采集作为2020年9月至2021年12月ALS患者疲劳的前瞻性、纵向、远程、在线调查研究的一部分。对量表进行横断面(基线)和纵向比较。评估相关系数、变异系数和描述性统计。随访约12个月时记录生命状况。结果:182名成年ALS患者同意这项研究。这个志愿者样本主要由白人、非西班牙裔、不吸烟的参与者组成。基线时同意参与者的调查完成率约为90%,12个月时超过40%。ALSFRS-RSE与ROADS在第3个月和第6个月具有高度显著的一致性,Cohen ' s kappa bb0.71 (p < 0.001);各尺度上功能增加或停滞的次数无显著性差异;功能衰退的变异系数在6个月时相似,但在3个月时更高,在12个月时更低。背景:肌萎缩性侧索硬化症(ALS)的治疗发展在很大程度上依赖于工作人员管理的功能评定量表来确定治疗效果。移动应用程序和可穿戴设备为生物识别数据收集提供了多种机会。假设:可穿戴和智能手机设备可以收集患者报告的结果,并被动收集传感器数据,为ALS疾病进展提供定量测量。方法:参与者远程同意,登记并随访6个月。ALSFRS-R在基线、3和6个月时通过电话给药。研究应用程序(Beiwe)在基线时安装在参与者的个人智能手机上,并每2 - 4周提供ALSFRS-R自我输入(ALSFRS-RSE)和Rasch整体ALS残疾量表(ROADS)调查。参与者还连续佩戴手腕上的活动监测器(ActiGraph Insight Watch)或脚踝上的活动监测器(Modus StepWatch)。 从每个设备获得供应商提供的每日测量值(vdm)。研究者衍生的每日测量(IDMs)是根据ActiGraph的分钟级活动计数数据计算的。参与至少两次ALSFRS-RSE和ROADS调查的参与者被纳入本分析。定义了至少有8小时估计活动监测器磨损时间的天数
本文章由计算机程序翻译,如有差异,请以英文原文为准。
Theme 09 - Clinical Trials and Trial Design
Background: The safety and efficacy of sodium phenylbutyrate (PB) and taurursodiol (TURSO, also known as ursodoxicol- taurine) coformulation in ALS were evaluated in a phase 2 US multicenter trial (CENTAUR) encompassing both randomized (NCT03127514) and open-label extension (OLE; NCT03488524) phases. While overall treatment-emergent adverse event (TEAE) incidence was similar in the PB&TURSO versus placebo groups in the randomized phase and between those continuing versus switching to PB&TURSO in the OLE phase, treatment-emergent cardiac adverse events (TECAEs) and centrally read electrocardiographic abnormalities were reported more frequently among those receiving PB&TURSO in both phases. Objective: Describe the findings of independent expert reviews of reported TECAEs and electrocardiographic abnor- malities and of recorded electrocardiographic parameters in CENTAUR. Methods: Occurrence of TEAEs was assessed at each visit during both phases. Standard 12-lead electrocardiograms were performed at baseline in both phases and at a total of 2 visits in the randomized phase and up to 10 visits in the OLE phase, blinded and centrally read. Unblinded independent expert reviews assessed reported TECAEs and electrocar- diographic abnormalities for diagnostic accuracy and treatment emergence based on absence at baseline or lack Background: CD40L/CD40 costimulatory pathway mediates both humoral and adaptive immune responses in the pathophysiology of autoimmunity and transplant rejection. Studies have implicated CD40L (CD154) signaling and adaptive and innate immune cell activation in induction of neuroinflammation in neurodegenerative diseases. Methods: In this multicenter dose escalating phase 2 study 54 participants with a definite or probable diagnosis of ALS by El Escorial Criteria-revised received an anti CD154 antibody, tegoprubart, via intravenous infusion every two weeks for twelve weeks. Enrollment consisted of 9 participants in cohorts 1 and 2 (1 and 2mg/kg) and 18 participants in cohorts 3 and 4 (4 and 8mg/kg). The primary endpoint of the study was safety and tolerability. Secondary endpoints assessed the pharmacokinetics of tegoprubart as well as anti-drug antibody responses. Exploratory endpoints evaluated changes in disease progression utilizing the ALS Functional Rating Scale-Revised (ALSFRS-R), assessment of CD154 target engagement specifically looking at change in circulating CD40L and CXCL13, and changes in the levels of pro-inflam-matory biomarkers in circulation, by looking at change from baseline in circulating levels of a panel of inflammatory che-mokines and cytokines. Results: Seventy participants were screened, and fifty-four were enrolled in the study. Forty-nine completed the study (90.7%) receiving all six infusions of tegoprubart and com- pleting their final follow up visit. The most common TEAEs overall ( 10%) were fatigue (25.9%), falls (22.2%), headaches and muscle spasms (11.1%). Mean tegoprubart plasma concentrations increased proportionally with increasing dose with a half-life of approximately 24 days. Anti-drug antibody (ADA) titers were low and circulating levels of tego- prubart were as predicted for all cohorts. Tegopurbart demonstrated dose dependent target engagement as evidenced by a reduction in circulating levels of CD40L and CXCL13. While the study was not designed to detect clinical efficacy, target engagement was associated with a trend suggesting slowing in loss of function by change from baseline in ALSFRS-R. Tegoprubart also reduced multiple pro-inflamma- tory biomarkers in circulation in a dose dependent fashion, including TNF a , En-raged, IL-16, IL-18, CXCL9 and CXCL10 among others. The target engagement and inflammatory bio- markers showed increasing levels of reduction from the 1mg/ kg dose cohort to the 2mg/kg dose cohort and then pla- teaued at the 4mg/kg cohort. The changes were seen after the first dose and persisted throughout the treatment period. Conclusions: Tegoprubart was safe and well tolerated in adults with ALS demonstrating dose dependent functional activity to engage target and reduce pro-inflammatory che-mokines and cytokines associated with ALS. These results warrant further clinical studies with sufficient power and duration to assess clinical outcomes as a potential treatment for adults with ALS. Background: Platform trial approaches allow for simultan-eous evaluation of multiple investigational products by lever- aging a shared clinical trial infrastructure to reduce time and resources necessary to launch new investigations. Initiation of a platform trial requires tremendous clinical trial expertise, resources, and infrastructure to be successful. Objective: To share our operational experience from launch- ing the first Background: WVE-004 is an investigational stereopure antisense oligonucleotide that was designed to selectively lower transcripts derived from a hexanucleotide-repeat expansion mutation (G4C2) within the C9orf72 gene, which is the most common genetic cause of ALS and FTD. The repeat expan- sion reduces C9orf72 gene expression; transcription of the repeat leads to aberrant RNA foci and ultimately dipeptide repeat (DPR) proteins such as poly glycine-proline (poly(GP)). In preclinical mouse studies, WVE-004 produced substantial and durable reductions in repeat-containing C9orf72 tran- scripts and pathogenic poly(GP) without reducing C9orf72 protein expression (1). Results: FOCUS-C9 (NCT04931862) is a global, multicenter, placebo-controlled Phase 1b/2a trial with an adaptive design allowing for rapid assessment of the safety, tolerability, pharmacokinetics, and pharmacodynamic effects of single-and multiple-ascending intrathecal doses of WVE-004 in people with C9-ALS or C9-FTD. Initial results (up to day 85) showed significant reductions in CSF poly(GP) with single doses (10mg, n ¼ 2; 30mg, n ¼ 4; 60mg, n ¼ 3) with poly(GP) concentrations not apparently reaching steady-state maximal reduction by day 85. Adverse events (AEs) were balanced across treatment groups, including placebo ( n ¼ 3), and were mostly mild to moderate in intensity. Four patients, including one on placebo, experienced severe and/or serious AEs; one was reported to be related to study drug. These data suggest that single ascending doses of WVE-004 were generally safe, and reductions in CSF poly(GP) provide confirmation of tar- get engagement with a single dose. Based on these initial data, FOCUS-C9 was adapted to evaluate additional and expanded low, single-dose cohorts. Discussion: Treatment is ongoing, with additional data from single and multiple dose cohorts expected in 2022. New data emerging from the study will also be presented. FOCUS-C9 is the first clinical trial to address both ALS and FTD caused by the C9orf72 genetic variant in the same study. Further, FOCUS-C9 ’ s adaptive design has enabled data-driven modifi-cations to the study to support more rapid optimization of dose level and frequency for WVE-004. Introduction objective: Improved functional outcome measures in amyotrophic lateral sclerosis would aid amyo- trophic lateral sclerosis trial design and help hasten drug discovery. We evaluate the longitudinal performance of the Rasch-Built Overall Amyotrophic Lateral Sclerosis Disability Scale (ROADS) compared to the Amyotrophic Lateral Sclerosis Functional Rating Scale Revised for Self-Entry (ALSFRS-RSE) as patient reported outcomes of functional status in people with ALS. Methods: People with ALS completed the ROADS and the ALSFRS-RSE questionnaires at baseline, 3-, 6-, and 12-months using the web-based platform, Research Electronic Data Capture as a part of a prospective, longitudinal, remote, online survey study of fatigue in ALS from 9/2020 to 12/ 2021. The scales were compared cross-sectionally (at baseline) and longitudinally. Correlation coefficients, coefficient of variation, and descriptive statistics were assessed. Vital status was recorded at follow-up contact at approximately 12-months. Results: 182 adults with ALS consented to the study. This volunteer sample was comprised of predominantly White, non-Hispanic, non-smoking participants. Consented partici- pant survey completion was approximately 90% at baseline and greater than 40% at 12-months.The ALSFRS-RSE and the ROADS had high, significant agreement at 3- and 6 months by Cohen ’ s kappa > 71% ( p < 0.001); the number of functional increases or plateaus on each scale were not signifi- cantly different; and the coefficient of variation of functional decline was similar at the 6-month mark, though higher for the ROADS at 3-months and lower at 12-months. Background: Amyotrophic lateral sclerosis (ALS) therapeutic development has relied largely on staff-administered func- tional rating scales to determine treatment efficacy. Mobile applications (apps) and wearable devices offer multiple opportunities for biometric data collection. Hypothesis: Wearable and smartphone devices can collect patient-reported outcomes and passively collect sensor data that will provide quantitative measurement of ALS disease progression. Methods: Participants were remotely consented, enrolled and followed for 6 months. ALSFRS-R was administered at baseline, 3 and 6 months via phone call. The study app (Beiwe) was installed on the participants ’ personal smart-phones at baseline and delivered ALSFRS-R for Self-Entry (ALSFRS-RSE) and the Rasch Overall ALS Disability Scale (ROADS) surveys every 2 – 4 weeks. Participants also continu-ously wore either a wrist-worn activity monitor (ActiGraph Insight Watch) or an ankle-worn activity monitor (Modus StepWatch). Vendor-provided daily measures (VDMs) were obtained from each device. Investigator-derived daily meas- ures (IDMs) were calculated from ActiGraph ’ s minute-level activity counts data. Participants contributing at least two ALSFRS-RSE and ROADS surveys were included in this analysis. Days with at least 8hours of estimated activity monitor wear time were defined
求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
CiteScore
5.40
自引率
10.70%
发文量
64
期刊介绍: Amyotrophic Lateral Sclerosis and Frontotemporal Degeneration is an exciting new initiative. It represents a timely expansion of the journal Amyotrophic Lateral Sclerosis in response to the clinical, imaging pathological and genetic overlap between ALS and frontotemporal dementia. The expanded journal provides outstanding coverage of research in a wide range of issues related to motor neuron diseases, especially ALS (Lou Gehrig’s disease) and cognitive decline associated with frontotemporal degeneration. The journal also covers related disorders of the neuroaxis when relevant to these core conditions.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信