Oral decitabine–cedazuridine versus intravenous decitabine for myelodysplastic syndromes and chronic myelomonocytic leukaemia (ASCERTAIN): a registrational, randomised, crossover, pharmacokinetics, phase 3 study

Guillermo Garcia-Manero, James McCloskey, Elizabeth A Griffiths, Karen W L Yee, Amer M Zeidan, Aref Al-Kali, H Joachim Deeg, Prapti A Patel, Mitchell Sabloff, Mary-Margaret Keating, Nancy Zhu, Nashat Y Gabrail, Salman Fazal, Joseph Maly, Olatoyosi Odenike, Hagop Kantarjian, Amy E DeZern, Casey L O’Connell, Gail J Roboz, Lambert Busque, Michael R Savona
{"title":"Oral decitabine–cedazuridine versus intravenous decitabine for myelodysplastic syndromes and chronic myelomonocytic leukaemia (ASCERTAIN): a registrational, randomised, crossover, pharmacokinetics, phase 3 study","authors":"Guillermo Garcia-Manero, James McCloskey, Elizabeth A Griffiths, Karen W L Yee, Amer M Zeidan, Aref Al-Kali, H Joachim Deeg, Prapti A Patel, Mitchell Sabloff, Mary-Margaret Keating, Nancy Zhu, Nashat Y Gabrail, Salman Fazal, Joseph Maly, Olatoyosi Odenike, Hagop Kantarjian, Amy E DeZern, Casey L O’Connell, Gail J Roboz, Lambert Busque, Michael R Savona","doi":"10.1016/s2352-3026(23)00338-1","DOIUrl":null,"url":null,"abstract":"<h3>Background</h3><p><span><span>The DNA methyltransferase inhibitors </span>azacitidine and </span>decitabine<span><span><span> for individuals with myelodysplastic syndromes or chronic myelomonocytic leukaemia are available in parenteral form. Oral therapy with similar exposure for these diseases would offer potential treatment benefits. We aimed to compare the safety and </span>pharmacokinetics of oral decitabine plus the </span>cytidine deaminase<span> inhibitor cedazuridine versus intravenous decitabine.</span></span></p><h3>Methods</h3><p><span>We did a registrational, multicentre, open-label, crossover, phase 3 trial of individuals with myelodysplastic syndromes or chronic myelomonocytic leukaemia and individuals with acute myeloid leukaemia<span>, enrolled as separate cohorts; results for only participants with myelodysplastic syndromes or chronic myelomonocytic leukaemia are reported here. In 37 academic and community-based clinics in Canada and the USA, we enrolled individuals aged 18 years or older who were candidates to receive intravenous decitabine, with Eastern Cooperative Oncology Group performance status 0 or 1 and a life expectancy of at least 3 months. Participants were randomly assigned (1:1) to receive 5 days of oral decitabine–cedazuridine (one tablet once daily containing 35 mg decitabine and 100 mg cedazuridine as a fixed-dose combination) or intravenous decitabine (20 mg/m</span></span><sup>2</sup> per day by continuous 1-h intravenous infusion) in a 28-day treatment cycle, followed by 5 days of the other formulation in the next treatment cycle. Thereafter, all participants received oral decitabine–cedazuridine from the third cycle on until treatment discontinuation. The primary endpoint was total decitabine exposure over 5 days with oral decitabine–cedazuridine versus intravenous decitabine for cycles 1 and 2, measured as area under the curve in participants who received the full treatment dose in cycles 1 and 2 and had decitabine daily AUC<sub>0–24</sub> for both oral decitabine–cedazuridine and intravenous decitabine (ie, paired cycles). On completion of the study, all patients were rolled over to a maintenance study. This study is registered with <span>ClinicalTrials.gov</span><svg aria-label=\"Opens in new window\" focusable=\"false\" height=\"8px\" viewbox=\"0 0 8 8\" width=\"8px\"><path d=\"M1.12949 2.1072V1H7V6.85795H5.89111V2.90281L0.784057 8L0 7.21635L5.11902 2.1072H1.12949Z\"></path></svg>, <span>NCT03306264</span><svg aria-label=\"Opens in new window\" focusable=\"false\" height=\"8px\" viewbox=\"0 0 8 8\" width=\"8px\"><path d=\"M1.12949 2.1072V1H7V6.85795H5.89111V2.90281L0.784057 8L0 7.21635L5.11902 2.1072H1.12949Z\"></path></svg>.</p><h3>Findings</h3><p>Between Feb 8, 2018, and June 7, 2021, 173 individuals were screened, 138 (80%) participants were randomly assigned to a treatment sequence, and 133 (96%) participants (87 [65%] men and 46 [35%] women; 121 [91%] White, four [3%] Black or African-American, three [2%] Asian, and five [4%] not reported) received treatment. Median follow-up was 966 days (IQR 917–1050). Primary endpoint of total exposure of oral decitabine–cedazuridine versus intravenous decitabine was 98·93% (90% CI 92·66–105·60), indicating equivalent pharmacokinetic exposure on the basis of area under the curve. The safety profiles of oral decitabine–cedazuridine and intravenous decitabine were similar. The most frequent adverse events of grade 3 or worse were thrombocytopenia (81 [61%] of 133 participants), neutropenia (76 [57%] participants), and anaemia (67 [50%] participants). The incidence of serious adverse events in cycles 1–2 was 31% (40 of 130 participants) with oral decitabine–cedazuridine and 18% (24 of 132 participants) with intravenous decitabine. There were five treatment-related deaths; two deemed related to oral therapy (sepsis and pneumonia) and three to intravenous treatment (septic shock [n=2] and pneumonia [n=1]).</p><h3>Interpretation</h3><p>Oral decitabine–cedazuridine was pharmacologically and pharmacodynamically equivalent to intravenous decitabine. The results support use of oral decitabine–cedazuridine as a safe and effective alternative to intravenous decitabine for treatment of individuals with myelodysplastic syndromes or chronic myelomonocytic leukaemia.</p><h3>Funding</h3><p>Astex Pharmaceuticals.</p>","PeriodicalId":501011,"journal":{"name":"The Lancet Haematology","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2023-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"The Lancet Haematology","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1016/s2352-3026(23)00338-1","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

Abstract

Background

The DNA methyltransferase inhibitors azacitidine and decitabine for individuals with myelodysplastic syndromes or chronic myelomonocytic leukaemia are available in parenteral form. Oral therapy with similar exposure for these diseases would offer potential treatment benefits. We aimed to compare the safety and pharmacokinetics of oral decitabine plus the cytidine deaminase inhibitor cedazuridine versus intravenous decitabine.

Methods

We did a registrational, multicentre, open-label, crossover, phase 3 trial of individuals with myelodysplastic syndromes or chronic myelomonocytic leukaemia and individuals with acute myeloid leukaemia, enrolled as separate cohorts; results for only participants with myelodysplastic syndromes or chronic myelomonocytic leukaemia are reported here. In 37 academic and community-based clinics in Canada and the USA, we enrolled individuals aged 18 years or older who were candidates to receive intravenous decitabine, with Eastern Cooperative Oncology Group performance status 0 or 1 and a life expectancy of at least 3 months. Participants were randomly assigned (1:1) to receive 5 days of oral decitabine–cedazuridine (one tablet once daily containing 35 mg decitabine and 100 mg cedazuridine as a fixed-dose combination) or intravenous decitabine (20 mg/m2 per day by continuous 1-h intravenous infusion) in a 28-day treatment cycle, followed by 5 days of the other formulation in the next treatment cycle. Thereafter, all participants received oral decitabine–cedazuridine from the third cycle on until treatment discontinuation. The primary endpoint was total decitabine exposure over 5 days with oral decitabine–cedazuridine versus intravenous decitabine for cycles 1 and 2, measured as area under the curve in participants who received the full treatment dose in cycles 1 and 2 and had decitabine daily AUC0–24 for both oral decitabine–cedazuridine and intravenous decitabine (ie, paired cycles). On completion of the study, all patients were rolled over to a maintenance study. This study is registered with ClinicalTrials.gov, NCT03306264.

Findings

Between Feb 8, 2018, and June 7, 2021, 173 individuals were screened, 138 (80%) participants were randomly assigned to a treatment sequence, and 133 (96%) participants (87 [65%] men and 46 [35%] women; 121 [91%] White, four [3%] Black or African-American, three [2%] Asian, and five [4%] not reported) received treatment. Median follow-up was 966 days (IQR 917–1050). Primary endpoint of total exposure of oral decitabine–cedazuridine versus intravenous decitabine was 98·93% (90% CI 92·66–105·60), indicating equivalent pharmacokinetic exposure on the basis of area under the curve. The safety profiles of oral decitabine–cedazuridine and intravenous decitabine were similar. The most frequent adverse events of grade 3 or worse were thrombocytopenia (81 [61%] of 133 participants), neutropenia (76 [57%] participants), and anaemia (67 [50%] participants). The incidence of serious adverse events in cycles 1–2 was 31% (40 of 130 participants) with oral decitabine–cedazuridine and 18% (24 of 132 participants) with intravenous decitabine. There were five treatment-related deaths; two deemed related to oral therapy (sepsis and pneumonia) and three to intravenous treatment (septic shock [n=2] and pneumonia [n=1]).

Interpretation

Oral decitabine–cedazuridine was pharmacologically and pharmacodynamically equivalent to intravenous decitabine. The results support use of oral decitabine–cedazuridine as a safe and effective alternative to intravenous decitabine for treatment of individuals with myelodysplastic syndromes or chronic myelomonocytic leukaemia.

Funding

Astex Pharmaceuticals.

治疗骨髓增生异常综合征和慢性粒细胞白血病的口服地西他滨-卡达嘧啶与静脉注射地西他滨(ASCERTAIN):一项注册、随机、交叉、药代动力学 3 期研究
背景 用于治疗骨髓增生异常综合征或慢性粒细胞白血病患者的 DNA 甲基转移酶抑制剂阿扎胞苷和地西他滨均为肠外用药。对于这些疾病,具有类似暴露的口服疗法将提供潜在的治疗优势。我们的目的是比较口服地西他滨加细胞苷脱氨酶抑制剂西达嘧啶与静脉注射地西他滨的安全性和药代动力学。方法我们对骨髓增生异常综合征或慢性粒单核细胞白血病患者和急性髓性白血病患者分别进行了注册、多中心、开放标签、交叉、3 期试验;本文仅报告骨髓增生异常综合征或慢性粒单核细胞白血病患者的试验结果。在加拿大和美国的 37 家学术和社区诊所中,我们招募了年龄在 18 岁或 18 岁以上、适合静脉注射地西他滨的患者,这些患者的东部合作肿瘤学组(Eastern Cooperative Oncology Group)表现为 0 或 1,预期寿命至少为 3 个月。受试者被随机分配(1:1),在一个为期28天的治疗周期内接受5天的地西他滨-西达嘧啶口服治疗(每天一次,每次一片,含35毫克地西他滨和100毫克西达嘧啶的固定剂量组合)或地西他滨静脉注射治疗(每天20毫克/平方米,1小时连续静脉注射),然后在下一个治疗周期接受5天的另一种制剂治疗。此后,所有参与者从第三个治疗周期开始口服地西他滨-西达脲苷,直至治疗终止。主要终点是第1和第2周期口服地西他滨-西达嘧啶与静脉注射地西他滨5天的地西他滨总暴露量,以在第1和第2周期接受全部治疗剂量且口服地西他滨-西达嘧啶和静脉注射地西他滨的地西他滨日AUC0-24(即配对周期)的参与者的曲线下面积来衡量。研究结束后,所有患者均转入维持治疗研究。该研究已在ClinicalTrials.gov注册,编号为NCT03306264.研究结果2018年2月8日至2021年6月7日期间,173人接受了筛查,138人(80%)被随机分配到治疗序列,133人(96%)接受了治疗(87名[65%]男性,46名[35%]女性;121名[91%]白人,4名[3%]黑人或非裔美国人,3名[2%]亚裔,5名[4%]未报告)。随访中位数为 966 天(IQR 为 917-1050 天)。口服地西他滨-西达脲苷与静脉注射地西他滨的主要终点总暴露率为98-93%(90% CI 92-66-105-60),表明根据曲线下面积计算的药代动力学暴露相当。口服地西他滨-卡达嘧啶和静脉注射地西他滨的安全性相似。最常见的3级或更严重的不良反应是血小板减少(133名参与者中有81名[61%])、中性粒细胞减少(76名[57%]参与者)和贫血(67名[50%]参与者)。在第1-2周期中,口服地西他滨-西达脲苷的严重不良反应发生率为31%(130名参与者中的40人),静脉注射地西他滨的严重不良反应发生率为18%(132名参与者中的24人)。有5例治疗相关死亡,其中2例与口服治疗有关(败血症和肺炎),3例与静脉治疗有关(脓毒性休克[n=2]和肺炎[n=1])。研究结果支持将口服地西他滨-卡佐尿苷作为静脉注射地西他滨的安全有效替代药物,用于治疗骨髓增生异常综合征或慢性粒细胞白血病患者。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
自引率
0.00%
发文量
0
×
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