Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine (DGSOM), University of California, Los Angeles (UCLA), Los Angeles, California.
Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California.
Clin Cancer Res. 2019 Feb 1;25(3):1000-1011. doi: 10.1158/1078-0432.CCR-18-0963. Epub 2018 Nov 8.
PURPOSE: To improve persistence of adoptively transferred T-cell receptor (TCR)-engineered T cells and durable clinical responses, we designed a clinical trial to transplant genetically-modified hematopoietic stem cells (HSCs) together with adoptive cell transfer of T cells both engineered to express an NY-ESO-1 TCR. Here, we report the preclinical studies performed to enable an investigational new drug (IND) application. EXPERIMENTAL DESIGN: HSCs transduced with a lentiviral vector expressing NY-ESO-1 TCR and the PET reporter/suicide gene HSV1-sr39TK and T cells transduced with a retroviral vector expressing NY-ESO-1 TCR were coadministered to myelodepleted HLA-A2/K mice within a formal Good Laboratory Practice (GLP)-compliant study to demonstrate safety, persistence, and HSC differentiation into all blood lineages. Non-GLP experiments included assessment of transgene immunogenicity and viral insertion safety studies. Furthermore, Good Manufacturing Practice (GMP)-compliant cell production qualification runs were performed to establish the manufacturing protocols for clinical use. RESULTS: TCR genetically modified and -cultured HSCs differentiated into all blood subsets after HSC transplantation, and coadministration of TCR-transduced T cells did not result in increased toxicity. The expression of NY-ESO-1 TCR and sr39TK transgenes did not have a detrimental effect on gene-modified HSC's differentiation to all blood cell lineages. There was no evidence of genotoxicity induced by the lentiviral vector. GMP batches of clinical-grade transgenic cells produced during qualification runs had adequate stability and functionality. CONCLUSIONS: Coadministration of HSCs and T cells expressing an NY-ESO-1 TCR is safe in preclinical models. The results presented in this article led to the FDA approval of IND 17471.
目的:为了提高过继性 T 细胞受体(TCR)工程化 T 细胞的持久性和持久的临床反应,我们设计了一项临床试验,即将工程化表达 NY-ESO-1 TCR 的基因修饰造血干细胞(HSCs)与过继性 T 细胞转移联合移植。在此,我们报告了进行临床前研究以获得新药临床试验(IND)申请的情况。
实验设计:在符合良好实验室规范(GLP)的研究中,将表达 NY-ESO-1 TCR 的慢病毒载体转导的 HSCs 和表达 NY-ESO-1 TCR 的逆转录病毒载体转导的 T 细胞共同给予骨髓清除 HLA-A2/K 小鼠,以证明安全性、持久性和 HSC 分化为所有血液谱系。非 GLP 实验包括转基因免疫原性和病毒插入安全性研究的评估。此外,进行了符合良好生产规范(GMP)的细胞生产资格运行,以建立用于临床使用的制造方案。
结果:TCR 基因修饰和培养的 HSCs 在 HSC 移植后分化为所有血液亚群,并且共给予 TCR 转导的 T 细胞不会导致毒性增加。NY-ESO-1 TCR 和 sr39TK 转基因的表达对基因修饰 HSC 向所有血细胞谱系的分化没有不利影响。慢病毒载体没有引起遗传毒性的证据。在资格运行期间生产的 GMP 批次临床级转基因细胞具有足够的稳定性和功能。
结论:在临床前模型中,共给予表达 NY-ESO-1 TCR 的 HSCs 和 T 细胞是安全的。本文介绍的结果导致 FDA 批准 IND 17471。
Cancer Immunol Res. 2018-3-27
J Immunother Cancer. 2019-10-24
Nat Commun. 2025-7-1
Signal Transduct Target Ther. 2021-2-8
Nat Rev Drug Discov. 2020-4-6
Cancers (Basel). 2020-5-23
Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2019-4-10
N Engl J Med. 2017-10-26
N Engl J Med. 2017-3-2
Sci Transl Med. 2016-4-20
J Immunother. 2016-1