Development and IND-enabling studies of a novel Cas9 genome-edited autologous CD34+ cell therapy to induce fetal hemoglobin for sickle cell disease.

IF 12.1 1区 医学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Molecular Therapy Pub Date : 2024-07-31 DOI:10.1016/j.ymthe.2024.07.022
Varun Katta, Kiera O'Keefe, Yichao Li, Thiyagaraj Mayuranathan, Cicera R Lazzarotto, Rachael K Wood, Rachel M Levine, Alicia Powers, Kalin Mayberry, Garret Manquen, Yu Yao, Jingjing Zhang, Yoonjeong Jang, Nikitha Nimmagadda, Erin A Dempsey, GaHyun Lee, Naoya Uchida, Yong Cheng, Frank Fazio, Tim Lockey, Mike Meagher, Akshay Sharma, John F Tisdale, Sheng Zhou, Jonathan S Yen, Mitchell J Weiss, Shengdar Q Tsai
{"title":"Development and IND-enabling studies of a novel Cas9 genome-edited autologous CD34<sup>+</sup> cell therapy to induce fetal hemoglobin for sickle cell disease.","authors":"Varun Katta, Kiera O'Keefe, Yichao Li, Thiyagaraj Mayuranathan, Cicera R Lazzarotto, Rachael K Wood, Rachel M Levine, Alicia Powers, Kalin Mayberry, Garret Manquen, Yu Yao, Jingjing Zhang, Yoonjeong Jang, Nikitha Nimmagadda, Erin A Dempsey, GaHyun Lee, Naoya Uchida, Yong Cheng, Frank Fazio, Tim Lockey, Mike Meagher, Akshay Sharma, John F Tisdale, Sheng Zhou, Jonathan S Yen, Mitchell J Weiss, Shengdar Q Tsai","doi":"10.1016/j.ymthe.2024.07.022","DOIUrl":null,"url":null,"abstract":"<p><p>Sickle cell disease (SCD) is a common, severe genetic blood disorder. Current pharmacotherapies are partially effective and allogeneic hematopoietic stem cell transplantation is associated with immune toxicities. Genome editing of patient hematopoietic stem cells (HSCs) to reactivate fetal hemoglobin (HbF) in erythroid progeny offers an alternative potentially curative approach to treat SCD. Although the FDA released guidelines for evaluating genome editing risks, it remains unclear how best to approach pre-clinical assessment of genome-edited cell products. Here, we describe rigorous pre-clinical development of a therapeutic γ-globin gene promoter editing strategy that supported an investigational new drug application cleared by the FDA. We compared γ-globin promoter and BCL11A enhancer targets, identified a potent HbF-inducing lead candidate, and tested our approach in mobilized CD34<sup>+</sup> hematopoietic stem progenitor cells (HSPCs) from SCD patients. We observed efficient editing, HbF induction to predicted therapeutic levels, and reduced sickling. With single-cell analyses, we defined the heterogeneity of HbF induction and HBG1/HBG2 transcription. With CHANGE-seq for sensitive and unbiased off-target discovery followed by targeted sequencing, we did not detect off-target activity in edited HSPCs. Our study provides a blueprint for translating new ex vivo HSC genome editing strategies toward clinical trials for treating SCD and other blood disorders.</p>","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":null,"pages":null},"PeriodicalIF":12.1000,"publicationDate":"2024-07-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Molecular Therapy","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1016/j.ymthe.2024.07.022","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"BIOTECHNOLOGY & APPLIED MICROBIOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Sickle cell disease (SCD) is a common, severe genetic blood disorder. Current pharmacotherapies are partially effective and allogeneic hematopoietic stem cell transplantation is associated with immune toxicities. Genome editing of patient hematopoietic stem cells (HSCs) to reactivate fetal hemoglobin (HbF) in erythroid progeny offers an alternative potentially curative approach to treat SCD. Although the FDA released guidelines for evaluating genome editing risks, it remains unclear how best to approach pre-clinical assessment of genome-edited cell products. Here, we describe rigorous pre-clinical development of a therapeutic γ-globin gene promoter editing strategy that supported an investigational new drug application cleared by the FDA. We compared γ-globin promoter and BCL11A enhancer targets, identified a potent HbF-inducing lead candidate, and tested our approach in mobilized CD34+ hematopoietic stem progenitor cells (HSPCs) from SCD patients. We observed efficient editing, HbF induction to predicted therapeutic levels, and reduced sickling. With single-cell analyses, we defined the heterogeneity of HbF induction and HBG1/HBG2 transcription. With CHANGE-seq for sensitive and unbiased off-target discovery followed by targeted sequencing, we did not detect off-target activity in edited HSPCs. Our study provides a blueprint for translating new ex vivo HSC genome editing strategies toward clinical trials for treating SCD and other blood disorders.

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
开发新型 Cas9 基因组编辑自体 CD34+ 细胞疗法并进行 IND 授权研究,以诱导胎儿血红蛋白治疗镰状细胞病。
镰状细胞病(SCD)是一种常见的严重遗传性血液疾病。目前的药物疗法部分有效,而异体造血干细胞移植(HSCT)则与免疫毒性有关。对患者造血干细胞(HSCs)进行基因组编辑,重新激活红细胞后代中的胎儿血红蛋白(HbF),为治疗SCD提供了另一种潜在的治疗方法。尽管美国食品和药物管理局发布了基因组编辑风险评估指南,但如何对基因组编辑细胞产品进行最佳临床前评估仍不明确。在此,我们介绍了一种治疗性γ-球蛋白基因启动子编辑策略的严格临床前开发,该策略为美国食品药品管理局批准的新药研究(IND)申请提供了支持。我们比较了γ-球蛋白启动子和 BCL11A 增强子靶点,确定了一种有效的 HbF 诱导候选药物,并在 SCD 患者动员的 CD34+ HSPCs 中测试了我们的方法。我们观察到了高效的编辑、达到预测治疗水平的 HbF 诱导以及镰状细胞减少。通过单细胞分析,我们确定了 HbF 诱导和 HBG1/HBG2 转录的异质性。利用 CHANGE-seq 敏感、无偏见地发现脱靶,然后进行靶向测序,我们没有在编辑的 HSPC 中检测到脱靶活性。我们的研究为将新的体外造血干细胞基因组编辑策略转化为治疗 SCD 和其他血液疾病的临床试验提供了蓝图。
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 去求助
来源期刊
Molecular Therapy
Molecular Therapy 医学-生物工程与应用微生物
CiteScore
19.20
自引率
3.20%
发文量
357
审稿时长
3 months
期刊介绍: Molecular Therapy is the leading journal for research in gene transfer, vector development, stem cell manipulation, and therapeutic interventions. It covers a broad spectrum of topics including genetic and acquired disease correction, vaccine development, pre-clinical validation, safety/efficacy studies, and clinical trials. With a focus on advancing genetics, medicine, and biotechnology, Molecular Therapy publishes peer-reviewed research, reviews, and commentaries to showcase the latest advancements in the field. With an impressive impact factor of 12.4 in 2022, it continues to attract top-tier contributions.
期刊最新文献
Engineering a solution for allogeneic CAR-T rejection. Targeting Rap1b signaling cascades with CDNF: Modulating Platelet Activation, Regulating Plasma Oxylipins and Mitigating Reperfusion Injury in stroke. A CD25×TIGIT bispecific antibody induces anti-tumor activity through selective intratumoral Treg cell depletion. A chimeric anti-inflammatory and anti-vascularization immunomodulator prevents high-risk corneal transplantation rejection via ex vivo gene therapy. Case study of CD19-directed chimeric antigen receptor T-cell therapy in a subject with immune-mediate necrotizing myopathy treated in the RESET-Myositis™ phase I/II trial.
×
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
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 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 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1