Chemotherapy-free cure of hemoglobin disorders through base editing
通过碱基编辑无需化疗即可治愈血红蛋白疾病
基本信息
- 批准号:10754114
- 负责人:
- 金额:$ 80.77万
- 依托单位:
- 依托单位国家:美国
- 项目类别:
- 财政年份:2023
- 资助国家:美国
- 起止时间:2023-09-01 至 2027-05-31
- 项目状态:未结题
- 来源:
- 关键词:AddressAffectAllogenicAmino AcidsAntigensAutologousAutologous TransplantationBiological AssayBone MarrowBone Marrow PurgingBypassCell TransplantationCellsClinicClinicalClinical TrialsClustered Regularly Interspaced Short Palindromic RepeatsCouplingCytokine ReceptorsDNA DamageDataDefectDevelopmentDiseaseDrug KineticsEndowmentEngineeringEngraftmentEpitopesErythroid CellsExtracellular DomainFLT3 geneFaceFetal HemoglobinFutureGene ModifiedGene TransferGeneticGenetic EngineeringGlobinGoalsGuide RNAHairHematological DiseaseHematologyHematopoiesisHematopoieticHematopoietic Stem Cell TransplantationHematopoietic stem cellsHemoglobinHemoglobinopathiesHomologous TransplantationHumanIL3RA geneImmuneImmunotherapyImmunotoxinsInfectionInfertilityInfusion proceduresInterventionLifeLigandsMalignant NeoplasmsMeasurableMedicalModelingModificationMonoclonal AntibodiesMucositisNatureNauseaParameter EstimationPathologicPatientsPharmaceutical PreparationsPhysiologicalPre-Clinical ModelProductionRadiation therapyRegimenRegulationResistanceRiskSickle Cell AnemiaSpecificitySystemTestingTherapeuticTherapeutic antibodiesTimeToxic effectTransfusionTranslationsTransplantationTreatment EfficacyValidationbase editingbeta Thalassemiacellular engineeringchemotherapychimeric antigen receptor T cellsclinical efficacyconditioningcurative treatmentsderepressionefficacy validationgene therapygenome-widegenotoxicityhemoglobin Binnovationmanufacturing processnew technologynovel strategiesnucleasepreservationprotein functionreceptorresearch clinical testingresponsescale upsicklingstem cell engraftmenttool
项目摘要
PROJECT SUMMARY
Sickle cell disease (SCD) and transfusion-dependent b-thalassemia (TDT) are severe, prevalent blood disorders
for which fetal hemoglobin (HbF) induction can bypass the fundamental hemoglobin defects and hematopoietic
stem/progenitor cell (HSPC) transplantation (HSCT) offers curative potential. Allogeneic and autologous trans-
plant approaches can succeed, nonetheless, the short- and long-term toxicities of genotoxic alkylating chemo-
therapy-based conditioning regimens remain a substantial barrier to the widespread application of curative HSCT
for SCD and TDT. Immunotherapies targeting HSPC antigens have been proposed as a safer conditioning strat-
egy, however the pharmacokinetics of these agents currently hamper their clinical efficacy. The long-term goal
of our proposal is to address this unmet medical need by developing an effective, novel strategy for the engraft-
ment and progressive enrichment of autologous gene modified HSPCs in SCD and TDT by coupling non-geno-
toxic immunotherapy-based myeloablation with epitope-engineering. Our central hypothesis is that precise
multiplexed base editing of HbF determinants and targeted epitopes within HSPCs can endow hematopoietic
lineages with both HbF induction capacity and selective resistance to monoclonal Abs or CAR-T cells without
affecting protein function or regulation (so-called stealth status). We have identified defined minimal amino-acid
changes within the extracellular domains (ECD) of the cytokine receptors KIT, FLT3 and IL3RA, each expressed
in long-term repopulating HSCs, that abrogate recognition by therapeutic Abs while preserving physiologic
responses to stimulation with their respective ligands. Here, we will capitalize on these results and further expand
the reach of these innovative genetic engineering tools with the objectives to i) generate “stealth” g-globin
derepressed HSPCs by multiplex CRISPR-Cas base-editing; ii) validate efficacy of this approach on suitable
pre-clinical models of b-hemoglobinopathy, and iii) further optimize and scale the manufacturing process for
production of efficiently and precisely engineered cellular products suitable to progress to the clinic. We aim: 1)
to optimize multiplex base editing approaches to simultaneously derepress HbF and engineer stealth HSPC
epitopes that will generate immunotherapy resistant hematopoietic stem cells capable of ameliorating sickling
and globin chain imbalance in SCD and TDT patient erythroid cells; 2) to maximize the engraftment of edited
HSCs by optimizing immunotherapy regimens to enrich multiplex edited HSPCs through modeling parameters
for therapeutic selection of edited HSPCs, and thereby obtaining proof-of-concept chemotherapy-free engraft-
ment and selection of edited patient HSPCs; and 3) to identify conditions that produce efficient on-target base
edits without measurable off-targets at clinical scale. This project will provide fundamental advancement of a
new chemotherapy-free gene therapy approach to HSCT for hemoglobinopathies that should additionally have
broad applicability to other hematopoietic disorders.
项目摘要
镰状细胞疾病(SCD)和依赖于输血的B-心中贫血(TDT)严重,流行的血液疾病
胎儿血红蛋白(HBF)诱导可以绕过基本血红蛋白缺陷和造血
茎/祖细胞(HSPC)移植(HSCT)具有治疗潜力。同种异体和自体反式
尽管如此,植物的方法仍可以成功,尽管如此
基于治疗的调节方案仍然是治疗性HSCT宽度应用的实质性障碍
对于SCD和TDT。已经提出针对HSPC抗原的免疫疗法作为更安全的条件层
但是,这些药物的药代动力学目前妨碍了其临床效率。长期目标
我们的建议是通过制定有效的新型策略来满足这种未满足的医疗需求。
通过耦合非生物 -
基于毒性免疫疗法的骨髓疗法和表位工程。我们的核心假设是如此精确
HBF确定剂和HSPC中的靶向表位的多路复用基础编辑可以赋予造血
具有HBF诱导能力和对单克隆ABS或CAR-T细胞的选择性抗性的谱系
影响蛋白质功能或调节(所谓的隐形状态)。我们已经确定了定义的最小氨基酸
细胞因子受体试剂盒(FLT3和IL3RA)的细胞外域(ECD)内的变化,每个都表达
在长期重新流传的HSC中,可以通过治疗ABS消除识别性的识别
对刺激的反应与各自的配体。在这里,我们将利用这些结果并进一步扩展
这些创新的基因工程工具的影响
通过多重CRISPR-CAS基础编辑的hspcs抑制了HSPC; ii)验证这种方法的效率
B-杂型细胞病的临床前模型和iii)进一步优化和扩展制造过程
有效,精确地设计的细胞产品适合于诊所进行。我们的目标:1)
优化多重基础编辑方法以简单地解开HBF和工程师隐形HSPC
将产生免疫疗法的抗造血干细胞的表位,能够改善病态
SCD和TDT患者红细胞细胞中的球蛋白链失衡; 2)最大化编辑的植入
通过优化免疫疗法方案,通过建模参数来丰富多重HSPC来丰富HSC
用于对编辑的HSPC进行热选择,从而获得概念验证的无化疗植入
编辑的患者HSPC的精神和选择; 3)确定产生有效靶向基础的条件
在临床规模上没有可测量的脱离目标的编辑。该项目将提供一个基本进步
新的无化学疗法基因治疗方法用于血红蛋白病的HSCT
对其他造血疾病的广泛适用性。
项目成果
期刊论文数量(0)
专著数量(0)
科研奖励数量(0)
会议论文数量(0)
专利数量(0)
数据更新时间:{{ journalArticles.updateTime }}
{{
item.title }}
{{ item.translation_title }}
- DOI:
{{ item.doi }} - 发表时间:
{{ item.publish_year }} - 期刊:
- 影响因子:{{ item.factor }}
- 作者:
{{ item.authors }} - 通讯作者:
{{ item.author }}
数据更新时间:{{ journalArticles.updateTime }}
{{ item.title }}
- 作者:
{{ item.author }}
数据更新时间:{{ monograph.updateTime }}
{{ item.title }}
- 作者:
{{ item.author }}
数据更新时间:{{ sciAawards.updateTime }}
{{ item.title }}
- 作者:
{{ item.author }}
数据更新时间:{{ conferencePapers.updateTime }}
{{ item.title }}
- 作者:
{{ item.author }}
数据更新时间:{{ patent.updateTime }}
Daniel Evan Bauer其他文献
Daniel Evan Bauer的其他文献
{{
item.title }}
{{ item.translation_title }}
- DOI:
{{ item.doi }} - 发表时间:
{{ item.publish_year }} - 期刊:
- 影响因子:{{ item.factor }}
- 作者:
{{ item.authors }} - 通讯作者:
{{ item.author }}
{{ truncateString('Daniel Evan Bauer', 18)}}的其他基金
Comprehensive characterization of variants underlying heart and blood diseases with CRISPR base editing
通过 CRISPR 碱基编辑全面表征心脏和血液疾病的变异
- 批准号:
10296877 - 财政年份:2021
- 资助金额:
$ 80.77万 - 项目类别:
Comprehensive characterization of variants underlying heart and blood diseases with CRISPR base editing
通过 CRISPR 碱基编辑全面表征心脏和血液疾病的变异
- 批准号:
10473734 - 财政年份:2021
- 资助金额:
$ 80.77万 - 项目类别:
Comprehensive characterization of variants underlying heart and blood diseases with CRISPR base editing
通过 CRISPR 碱基编辑全面表征心脏和血液疾病的变异
- 批准号:
10627940 - 财政年份:2021
- 资助金额:
$ 80.77万 - 项目类别:
Gene editing ELANE to understand and treat severe congenital neutropenia
基因编辑 ELANE 了解和治疗严重先天性中性粒细胞减少症
- 批准号:
10580862 - 财政年份:2020
- 资助金额:
$ 80.77万 - 项目类别:
Therapeutic BCL11A enhancer gene editing to induce fetal hemoglobin in β-hemoglobinopathy patients
治疗性 BCL11A 增强子基因编辑诱导 β 血红蛋白病患者胎儿血红蛋白
- 批准号:
10317505 - 财政年份:2020
- 资助金额:
$ 80.77万 - 项目类别:
Therapeutic BCL11A enhancer gene editing to induce fetal hemoglobin in β-hemoglobinopathy patients
治疗性 BCL11A 增强子基因编辑诱导 β 血红蛋白病患者胎儿血红蛋白
- 批准号:
10090251 - 财政年份:2020
- 资助金额:
$ 80.77万 - 项目类别:
Gene editing ELANE to understand and treat severe congenital neutropenia
基因编辑 ELANE 了解和治疗严重先天性中性粒细胞减少症
- 批准号:
10338097 - 财政年份:2020
- 资助金额:
$ 80.77万 - 项目类别:
Rectifying splicing mutations in blood disorders by gene editing
通过基因编辑纠正血液疾病中的剪接突变
- 批准号:
10531577 - 财政年份:2019
- 资助金额:
$ 80.77万 - 项目类别:
相似国自然基金
海洋缺氧对持久性有机污染物入海后降解行为的影响
- 批准号:42377396
- 批准年份:2023
- 资助金额:49 万元
- 项目类别:面上项目
氮磷的可获得性对拟柱孢藻水华毒性的影响和调控机制
- 批准号:32371616
- 批准年份:2023
- 资助金额:50 万元
- 项目类别:面上项目
还原条件下铜基催化剂表面供-受电子作用表征及其对CO2电催化反应的影响
- 批准号:22379027
- 批准年份:2023
- 资助金额:50 万元
- 项目类别:面上项目
CCT2分泌与内吞的机制及其对毒性蛋白聚集体传递的影响
- 批准号:32300624
- 批准年份:2023
- 资助金额:10 万元
- 项目类别:青年科学基金项目
在轨扰动影响下空间燃料电池系统的流动沸腾传质机理与抗扰控制研究
- 批准号:52377215
- 批准年份:2023
- 资助金额:50 万元
- 项目类别:面上项目
相似海外基金
Prognostic implications of mitochondrial inheritance in myelodysplastic syndromes after stem-cell transplantation
干细胞移植后骨髓增生异常综合征线粒体遗传的预后意义
- 批准号:
10662946 - 财政年份:2023
- 资助金额:
$ 80.77万 - 项目类别:
ADEPT-STAR therapy for high risk neuroblastoma
ADEPT-STAR 治疗高危神经母细胞瘤
- 批准号:
10760738 - 财政年份:2023
- 资助金额:
$ 80.77万 - 项目类别:
Targeting Dystroglycanopathies using Pluripotent-derived Myogenic Progenitors
使用多能源性肌源性祖细胞靶向肌营养不良症
- 批准号:
10561375 - 财政年份:2023
- 资助金额:
$ 80.77万 - 项目类别:
Developing novel therapies to improve blood stem cell transplantation outcomes
开发新疗法以改善造血干细胞移植结果
- 批准号:
10830194 - 财政年份:2023
- 资助金额:
$ 80.77万 - 项目类别:
Allogeneic BAFF Ligand Based CAR T-Cells as a Novel Therapy for Systemic Lupus Erythematous
基于同种异体 BAFF 配体的 CAR T 细胞作为系统性红斑狼疮的新疗法
- 批准号:
10761003 - 财政年份:2023
- 资助金额:
$ 80.77万 - 项目类别: