Suppr超能文献

基于纳米抗体的嵌合抗原受体自然杀伤细胞用于间皮素肿瘤的潜在免疫治疗

Nanobody-Based CAR NK Cells for Possible Immunotherapy of Mesothelin Tumors.

作者信息

Jung Dana, Choi Eunjeong, Jeoung Young-Hee, Lee Juheon, Gong Eun-Yeung, Jo Seo-Gyeong, Noh Kyunghee, Ha Kyungsoo, Wee Gabbine, Kim Hyeran, Jung Juyeon, Kim Seokho

机构信息

Department of Health Science, Graduate School of Dong-A University, Busan 49315, Korea.

Department of Medicinal Biotechnology, College of Health Science, Dong-A University, Busan 49315, Korea.

出版信息

Immune Netw. 2025 Jun 13;25(3):e23. doi: 10.4110/in.2025.25.e23. eCollection 2025 Jun.

Abstract

Chimeric Ag receptor (CAR)-engineered immune cells have demonstrated remarkable clinical efficacy, particularly in hematologic malignancies. Central to their success is the Ag-binding domain of the CAR, which governs both target specificity and therapeutic efficacy. Nanobodies (Nbs) possess a single-domain architecture and smaller molecular size, making them particularly amenable to the construction of tandem CARs that can co-target multiple Ags. This structural flexibility is advantageous for addressing tumor heterogeneity and reducing the risk of Ag escape in solid malignancies. Here, we developed mesothelin (MSLN)-specific nanobody-based chimeric Ag receptor-NK (Nb CAR-NK) cells using a synthetic nanobody identified from a phage display VHH library. The nanobody was selected after three rounds of biopanning and validated for high affinity and specificity using surface plasmon resonance and flow cytometry. The selected nanobody-based chimeric Ag receptor (Nb-CAR) construct was introduced into ex vivo expanded umbilical cord blood-derived NK cells via third-generation lentiviral transduction, resulting in stable expression and functional CAR-NK cells. The Nb CAR-NK cells exhibited potent cytotoxicity against MSLN-positive pancreatic cancer cells in vitro and significantly suppressed tumor growth in xenograft models. These findings support the clinical potential of Nb CAR-NK cells and highlight the value of Nb-CAR designs for targeting cell-surface Ags in solid tumors.

摘要

嵌合抗原受体(CAR)工程化免疫细胞已显示出显著的临床疗效,尤其是在血液系统恶性肿瘤中。其成功的关键在于CAR的抗原结合域,该域决定了靶点特异性和治疗效果。纳米抗体(Nb)具有单域结构且分子尺寸较小,这使得它们特别适合构建可同时靶向多种抗原的串联CAR。这种结构灵活性有利于应对实体恶性肿瘤中的肿瘤异质性并降低抗原逃逸风险。在此,我们利用从噬菌体展示VHH文库中鉴定出的一种合成纳米抗体,开发了基于间皮素(MSLN)特异性纳米抗体的嵌合抗原受体-NK(Nb CAR-NK)细胞。经过三轮生物淘选后筛选出该纳米抗体,并使用表面等离子体共振和流式细胞术验证了其高亲和力和特异性。通过第三代慢病毒转导将所选的基于纳米抗体的嵌合抗原受体(Nb-CAR)构建体引入体外扩增的脐带血来源的NK细胞中,从而产生稳定表达且具有功能的CAR-NK细胞。Nb CAR-NK细胞在体外对MSLN阳性胰腺癌细胞表现出强大的细胞毒性,并在异种移植模型中显著抑制肿瘤生长。这些发现支持了Nb CAR-NK细胞的临床潜力,并突出了Nb-CAR设计在靶向实体瘤细胞表面抗原方面的价值。

https://cdn.ncbi.nlm.nih.gov/pmc/blobs/3e2a/12226252/cd51385a57d3/in-25-e23-g001.jpg

相似文献

1
Nanobody-Based CAR NK Cells for Possible Immunotherapy of Mesothelin Tumors.
Immune Netw. 2025 Jun 13;25(3):e23. doi: 10.4110/in.2025.25.e23. eCollection 2025 Jun.
2
Precision targeting of rhabdomyosarcoma by combining primary CAR NK cells and radiotherapy.
J Immunother Cancer. 2025 Jul 7;13(7):e011330. doi: 10.1136/jitc-2024-011330.
3
B cell antigens: A key to optimizing CAR-T cell therapy.
Int Rev Immunol. 2025 Jun 19:1-28. doi: 10.1080/08830185.2025.2515839.
7
Engineering of potent CAR NK cells using non-viral Sleeping Beauty transposition from minimalistic DNA vectors.
Mol Ther. 2024 Jul 3;32(7):2357-2372. doi: 10.1016/j.ymthe.2024.05.022. Epub 2024 May 14.
9
Synthetic NKG2D receptor (SNR) armored CAR-T cells overcome antigen heterogeneity of solid tumor.
Cell Oncol (Dordr). 2025 Jun 19. doi: 10.1007/s13402-025-01066-5.
10
Chimeric antigen receptor therapies: Development, design, and implementation.
J Allergy Clin Immunol. 2025 Jul;156(1):70-80. doi: 10.1016/j.jaci.2025.04.005. Epub 2025 Apr 10.

本文引用的文献

1
Advancing pancreatic cancer therapy by mesothelin-specific nanobody conjugation.
Mol Cancer. 2025 Apr 24;24(1):124. doi: 10.1186/s12943-025-02325-7.
2
Novel and multiple targets for chimeric antigen receptor-based therapies in lymphoma.
Front Oncol. 2024 Apr 22;14:1396395. doi: 10.3389/fonc.2024.1396395. eCollection 2024.
4
Camel nanobody-based B7-H3 CAR-T cells show high efficacy against large solid tumours.
Nat Commun. 2023 Sep 22;14(1):5920. doi: 10.1038/s41467-023-41631-w.
5
From bench to bedside: the history and progress of CAR T cell therapy.
Front Immunol. 2023 May 15;14:1188049. doi: 10.3389/fimmu.2023.1188049. eCollection 2023.
6
Cytotoxicity of CD19-CAR-NK92 cells is primarily mediated via perforin/granzyme pathway.
Cancer Immunol Immunother. 2023 Aug;72(8):2573-2583. doi: 10.1007/s00262-023-03443-1. Epub 2023 Apr 13.
7
Tonic-signaling chimeric antigen receptors drive human regulatory T cell exhaustion.
Proc Natl Acad Sci U S A. 2023 Apr 4;120(14):e2219086120. doi: 10.1073/pnas.2219086120. Epub 2023 Mar 27.
8
Nanobody-based CAR-T cells for cancer immunotherapy.
Biomark Res. 2022 Apr 25;10(1):24. doi: 10.1186/s40364-022-00371-7.
9
CAR race to cancer immunotherapy: from CAR T, CAR NK to CAR macrophage therapy.
J Exp Clin Cancer Res. 2022 Mar 31;41(1):119. doi: 10.1186/s13046-022-02327-z.
10
Off-the-Shelf Prostate Stem Cell Antigen-Directed Chimeric Antigen Receptor Natural Killer Cell Therapy to Treat Pancreatic Cancer.
Gastroenterology. 2022 Apr;162(4):1319-1333. doi: 10.1053/j.gastro.2021.12.281. Epub 2022 Jan 6.

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

文档翻译

学术文献翻译模型,支持多种主流文档格式。

立即体验