文献检索文档翻译深度研究
Suppr Zotero 插件Zotero 插件
邀请有礼套餐&价格历史记录

新学期,新优惠

限时优惠:9月1日-9月22日

30天高级会员仅需29元

1天体验卡首发特惠仅需5.99元

了解详情
不再提醒
插件&应用
Suppr Zotero 插件Zotero 插件浏览器插件Mac 客户端Windows 客户端微信小程序
高级版
套餐订阅购买积分包
AI 工具
文献检索文档翻译深度研究
关于我们
关于 Suppr公司介绍联系我们用户协议隐私条款
关注我们

Suppr 超能文献

核心技术专利:CN118964589B侵权必究
粤ICP备2023148730 号-1Suppr @ 2025

HLA Ⅰ类基因敲除将同种异体原代 NK 细胞转化为“现货”免疫疗法的合适效应细胞。

HLA Class I Knockout Converts Allogeneic Primary NK Cells Into Suitable Effectors for "Off-the-Shelf" Immunotherapy.

机构信息

Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.

Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich-Heine University, Düsseldorf, Germany.

出版信息

Front Immunol. 2021 Jan 29;11:586168. doi: 10.3389/fimmu.2020.586168. eCollection 2020.


DOI:10.3389/fimmu.2020.586168
PMID:33584651
原文链接:https://pmc.ncbi.nlm.nih.gov/articles/PMC7878547/
Abstract

Cellular immunotherapy using chimeric antigen receptors (CARs) so far has almost exclusively used autologous peripheral blood-derived T cells as immune effector cells. However, harvesting sufficient numbers of T cells is often challenging in heavily pre-treated patients with malignancies and perturbed hematopoiesis and perturbed hematopoiesis. Also, such a CAR product will always be specific for the individual patient. In contrast, NK cell infusions can be performed in non-HLA-matched settings due to the absence of alloreactivity of these innate immune cells. Still, the infused NK cells are subject to recognition and rejection by the patient's immune system, thereby limiting their life-span and undermining the possibility for multiple infusions. Here, we designed genome editing and advanced lentiviral transduction protocols to render primary human NK cells unsusceptible/resistant to an allogeneic response by the recipient's CD8 T cells. After knocking-out surface expression of HLA class I molecules by targeting the B2M gene CRISPR/Cas9, we also co-expressed a single-chain HLA-E molecule, thereby preventing NK cell fratricide of B2M-knockout (KO) cells "missing self"-induced lysis. Importantly, these genetically engineered NK cells were functionally indistinguishable from their unmodified counterparts with regard to their phenotype and their natural cytotoxicity towards different AML cell lines. In co-culture assays, B2M-KO NK cells neither induced immune responses of allogeneic T cells nor re-activated allogeneic T cells which had been expanded/primed using irradiated PBMNCs of the respective NK cell donor. Our study demonstrates the feasibility of genome editing in primary allogeneic NK cells to diminish their recognition and killing by mismatched T cells and is an important prerequisite for using non-HLA-matched primary human NK cells as readily available, "off-the-shelf" immune effectors for a variety of immunotherapy indications in human cancer.

摘要

嵌合抗原受体(CAR)的细胞免疫疗法迄今为止几乎完全使用自体外周血来源的 T 细胞作为免疫效应细胞。然而,在恶性肿瘤和造血功能紊乱的重度预处理患者中,通常难以采集足够数量的 T 细胞。此外,此类 CAR 产品将始终针对个体患者。相比之下,由于这些先天免疫细胞不存在同种异体反应,NK 细胞输注可以在非 HLA 匹配的环境中进行。然而,输注的 NK 细胞仍会被患者的免疫系统识别和排斥,从而限制其寿命,并破坏多次输注的可能性。在这里,我们设计了基因组编辑和先进的慢病毒转导方案,使原代人 NK 细胞对受者的 CD8 T 细胞的同种异体反应具有抗性。通过靶向 B2M 基因的 CRISPR/Cas9 敲除表面表达的 HLA Ⅰ类分子后,我们还共同表达了单链 HLA-E 分子,从而防止 NK 细胞对 B2M 敲除(KO)细胞的同种异体反应杀伤 "自身缺失"诱导的裂解。重要的是,这些经过基因工程改造的 NK 细胞在表型和对不同 AML 细胞系的天然细胞毒性方面与未修饰的 NK 细胞没有功能区别。在共培养实验中,B2M-KO NK 细胞既不会诱导同种异体 T 细胞的免疫反应,也不会重新激活使用各自 NK 细胞供体的辐照 PBMC 扩增/启动的同种异体 T 细胞。我们的研究证明了在原代同种异体 NK 细胞中进行基因组编辑以减少其被错配 T 细胞识别和杀伤的可行性,这是使用非 HLA 匹配的原代人 NK 细胞作为各种人类癌症免疫治疗适应症的现成、"现成"免疫效应物的重要前提。

https://cdn.ncbi.nlm.nih.gov/pmc/blobs/2d24/7878547/2f27f87f2c27/fimmu-11-586168-g005.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/2d24/7878547/d43afcb88617/fimmu-11-586168-g001.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/2d24/7878547/3f0d5179942e/fimmu-11-586168-g002.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/2d24/7878547/9c8748959bf8/fimmu-11-586168-g003.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/2d24/7878547/2939c47e6826/fimmu-11-586168-g004.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/2d24/7878547/2f27f87f2c27/fimmu-11-586168-g005.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/2d24/7878547/d43afcb88617/fimmu-11-586168-g001.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/2d24/7878547/3f0d5179942e/fimmu-11-586168-g002.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/2d24/7878547/9c8748959bf8/fimmu-11-586168-g003.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/2d24/7878547/2939c47e6826/fimmu-11-586168-g004.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/2d24/7878547/2f27f87f2c27/fimmu-11-586168-g005.jpg

相似文献

[1]
HLA Class I Knockout Converts Allogeneic Primary NK Cells Into Suitable Effectors for "Off-the-Shelf" Immunotherapy.

Front Immunol. 2020

[2]
Mutant B2M-HLA-E and B2M-HLA-G fusion proteins protects universal chimeric antigen receptor-modified T cells from allogeneic NK cell-mediated lysis.

Eur J Immunol. 2021-10

[3]
[Allogeneic CAR-NK cells: A promising alternative to autologous CAR-T cells - State of the art, sources of NK cells, limits and perspectives].

Bull Cancer. 2021-10

[4]
[Development of allogeneic CAR T-cells].

Bull Cancer. 2021-10

[5]
Enhanced cytotoxicity of allogeneic NK cells with killer immunoglobulin-like receptor ligand incompatibility against melanoma and renal cell carcinoma cells.

Blood. 2004-7-1

[6]
Current Perspectives on "Off-The-Shelf" Allogeneic NK and CAR-NK Cell Therapies.

Front Immunol. 2021

[7]
Genetic Engineering and Enrichment of Human NK Cells for CAR-Enhanced Immunotherapy of Hematological Malignancies.

Front Immunol. 2022

[8]
HLA reduction of human T cells facilitates generation of immunologically multicompatible cellular products.

Blood Adv. 2024-7-9

[9]
Use of Cell and Genome Modification Technologies to Generate Improved "Off-the-Shelf" CAR T and CAR NK Cells.

Front Immunol. 2020

[10]
T cell receptor engineering of primary NK cells to therapeutically target tumors and tumor immune evasion.

J Immunother Cancer. 2022-3

引用本文的文献

[1]
From bench to bedside: translating mesenchymal stem cell therapies through preclinical and clinical evidence.

Front Bioeng Biotechnol. 2025-7-30

[2]
Overcoming Immune Barriers in Allogeneic CAR-NK Therapy: From Multiplex Gene Editing to AI-Driven Precision Design.

Biomolecules. 2025-6-26

[3]
Cancer-on-a-chip for precision cancer medicine.

Lab Chip. 2025-5-16

[4]
Advancing Allogeneic NK Cell Immunotherapy through Microfluidic Gene Delivery.

Adv Sci (Weinh). 2025-5

[5]
Peptide-enabled ribonucleoprotein delivery for CRISPR engineering (PERC) in primary human immune cells and hematopoietic stem cells.

Nat Protoc. 2025-3-3

[6]
Novel gene manipulation approaches to unlock the existing bottlenecks of CAR-NK cell therapy.

Front Cell Dev Biol. 2025-2-11

[7]
Unlocking the potential of engineered immune cell therapy for solid tumors.

Nat Commun. 2025-1-29

[8]
Advances in CRISPR-Cas technology and its applications: revolutionising precision medicine.

Front Genome Ed. 2024-12-12

[9]
Hypoimmunogenic CD19 CAR-NK cells derived from embryonic stem cells suppress the progression of human B-cell malignancies in xenograft animals.

Front Immunol. 2024-11-27

[10]
CRISPR-Cas9 in basic and translational aspects of cancer therapy.

Bioimpacts. 2024

本文引用的文献

[1]
Directed Differentiation of Mobilized Hematopoietic Stem and Progenitor Cells into Functional NK cells with Enhanced Antitumor Activity.

Cells. 2020-3-27

[2]
High Cytotoxic Efficiency of Lentivirally and Alpharetrovirally Engineered CD19-Specific Chimeric Antigen Receptor Natural Killer Cells Against Acute Lymphoblastic Leukemia.

Front Immunol. 2020-1-24

[3]
"UniCAR"-modified off-the-shelf NK-92 cells for targeting of GD2-expressing tumour cells.

Sci Rep. 2020-2-7

[4]
Use of CAR-Transduced Natural Killer Cells in CD19-Positive Lymphoid Tumors.

N Engl J Med. 2020-2-6

[5]
Efficient and Robust NK-Cell Transduction With Baboon Envelope Pseudotyped Lentivector.

Front Immunol. 2019-12-16

[6]
A simple method for in vitro preparation of natural killer cells from cord blood.

BMC Biotechnol. 2019-11-21

[7]
Efficacy of third-party chimeric antigen receptor modified peripheral blood natural killer cells for adoptive cell therapy of B-cell precursor acute lymphoblastic leukemia.

Leukemia. 2019-11-19

[8]
Knockout of beta-2 microglobulin reduces stem cell-induced immune rejection and enhances ischaemic hindlimb repair via exosome/miR-24/Bim pathway.

J Cell Mol Med. 2020-1

[9]
A Distinct Subset of Highly Proliferative and Lentiviral Vector (LV)-Transducible NK Cells Define a Readily Engineered Subset for Adoptive Cellular Therapy.

Front Immunol. 2019-8-22

[10]
A strategy to protect off-the-shelf cell therapy products using virus-specific T-cells engineered to eliminate alloreactive T-cells.

J Transl Med. 2019-7-24

文献AI研究员

20分钟写一篇综述,助力文献阅读效率提升50倍

立即体验

用中文搜PubMed

大模型驱动的PubMed中文搜索引擎

马上搜索

推荐工具

医学文档翻译智能文献检索