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

局部放射治疗原位递送 iPSC 来源的树突状细胞可在临床前免疫原性低的肿瘤模型中产生全身抗肿瘤免疫并增强 PD-L1 阻断作用。

In situ delivery of iPSC-derived dendritic cells with local radiotherapy generates systemic antitumor immunity and potentiates PD-L1 blockade in preclinical poorly immunogenic tumor models.

机构信息

Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA.

Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University, Matsumoto, Nagano, Japan.

出版信息

J Immunother Cancer. 2021 May;9(5). doi: 10.1136/jitc-2021-002432.


DOI:10.1136/jitc-2021-002432
PMID:34049930
原文链接:https://pmc.ncbi.nlm.nih.gov/articles/PMC8166607/
Abstract

BACKGROUND: Dendritic cells (DCs) are a promising therapeutic target in cancer immunotherapy given their ability to prime antigen-specific T cells, and initiate antitumor immune response. A major obstacle for DC-based immunotherapy is the difficulty to obtain a sufficient number of functional DCs. Theoretically, this limitation can be overcome by using induced pluripotent stem cells (iPSCs); however, therapeutic strategies to engage iPSC-derived DCs (iPSC-DCs) into cancer immunotherapy remain to be elucidated. Accumulating evidence showing that induction of tumor-residing DCs enhances immunomodulatory effect of radiotherapy (RT) prompted us to investigate antitumor efficacy of combining intratumoral administration of iPSC-DCs with local RT. METHODS: Mouse iPSCs were differentiated to iPSC-DCs on OP9 stromal cells expressing the notch ligand delta-like 1 in the presence of granulocyte macrophage colony-stimulating factor. Phenotype and the capacities of iPSC-DCs to traffic tumor-draining lymph nodes (TdLNs) and prime antigen-specific T cells were evaluated by flow cytometry and imaging flow cytometry. Antitumor efficacy of intratumoral injection of iPSC-DCs and RT was tested in syngeneic orthotopic mouse tumor models resistant to anti-PD-1 ligand 1 (PD-L1) therapy. RESULTS: Mouse iPSC-DCs phenotypically resembled conventional type 2 DCs, and had a capacity to promote activation, proliferation and effector differentiation of antigen-specific CD8 T cells in the presence of the cognate antigen in vitro. Combination of in situ administration of iPSC-DCs and RT facilitated the priming of tumor-specific CD8 T cells, and synergistically delayed the growth of not only the treated tumor but also the distant non-irradiated tumors. Mechanistically, RT enhanced trafficking of intratumorally injected iPSC-DCs to the TdLN, upregulated CD40 expression, and increased the frequency of DC/CD8 T cell aggregates. Phenotypic analysis of tumor-infiltrating CD8 T cells and myeloid cells revealed an increase of stem-like Slamf6 TIM3 CD8 T cells and PD-L1 expression in tumor-associated macrophages and DCs. Consequently, combined therapy rendered poorly immunogenic tumors responsive to anti-PD-L1 therapy along with the development of tumor-specific immunological memory. CONCLUSIONS: Our findings illustrate the translational potential of iPSC-DCs, and identify the therapeutic efficacy of a combinatorial platform to engage them for overcoming resistance to anti-PD-L1 therapy in poorly immunogenic tumors.

摘要

背景:树突状细胞(DCs)在癌症免疫治疗中具有很大的应用前景,因为它们能够刺激抗原特异性 T 细胞,引发抗肿瘤免疫反应。然而,基于 DC 的免疫治疗面临的一个主要障碍是难以获得足够数量的功能性 DCs。理论上,这一局限性可以通过诱导多能干细胞(iPSCs)来克服;然而,将 iPSC 来源的 DC(iPSC-DCs)用于癌症免疫治疗的治疗策略仍有待阐明。越来越多的证据表明,诱导肿瘤驻留 DC 可增强放射治疗(RT)的免疫调节作用,这促使我们研究联合肿瘤内注射 iPSC-DCs 和局部 RT 的抗肿瘤疗效。

方法:在存在粒细胞巨噬细胞集落刺激因子的情况下,将小鼠 iPSCs 在表达 notch 配体 delta-like 1 的 OP9 基质细胞上分化为 iPSC-DCs。通过流式细胞术和成像流式细胞术评估 iPSC-DCs 的表型和迁移肿瘤引流淋巴结(TdLNs)和刺激抗原特异性 T 细胞的能力。在对 PD-L1 配体 1(PD-L1)治疗耐药的同种异体原位小鼠肿瘤模型中,测试了肿瘤内注射 iPSC-DCs 和 RT 的抗肿瘤疗效。

结果:小鼠 iPSC-DCs 的表型与传统的 2 型 DC 相似,并且在体外存在同源抗原的情况下,具有促进抗原特异性 CD8 T 细胞激活、增殖和效应分化的能力。原位给予 iPSC-DCs 和 RT 的联合治疗促进了肿瘤特异性 CD8 T 细胞的启动,并协同延缓了不仅是治疗肿瘤而且是远处未照射肿瘤的生长。机制上,RT 增强了肿瘤内注射的 iPSC-DCs 向 TdLN 的迁移,上调了 CD40 的表达,并增加了 DC/CD8 T 细胞聚集的频率。对肿瘤浸润性 CD8 T 细胞和髓样细胞的表型分析显示,肿瘤相关巨噬细胞和 DC 中干细胞样 Slamf6 TIM3 CD8 T 细胞和 PD-L1 的表达增加。因此,联合治疗使免疫原性差的肿瘤对抗 PD-L1 治疗有反应,并产生肿瘤特异性免疫记忆。

结论:我们的研究结果说明了 iPSC-DCs 的转化潜力,并确定了一种组合平台的治疗效果,该平台可用于使其参与克服免疫原性差的肿瘤对抗 PD-L1 治疗的耐药性。

https://cdn.ncbi.nlm.nih.gov/pmc/blobs/ed56/8166607/af456d376cd3/jitc-2021-002432f08.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/ed56/8166607/e3c76b316277/jitc-2021-002432f01.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/ed56/8166607/10a256f2db06/jitc-2021-002432f02.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/ed56/8166607/976633b2735f/jitc-2021-002432f03.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/ed56/8166607/978f104528c6/jitc-2021-002432f04.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/ed56/8166607/318b93a6e08f/jitc-2021-002432f05.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/ed56/8166607/1edb397f5cee/jitc-2021-002432f06.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/ed56/8166607/f96ec004370d/jitc-2021-002432f07.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/ed56/8166607/af456d376cd3/jitc-2021-002432f08.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/ed56/8166607/e3c76b316277/jitc-2021-002432f01.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/ed56/8166607/10a256f2db06/jitc-2021-002432f02.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/ed56/8166607/976633b2735f/jitc-2021-002432f03.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/ed56/8166607/978f104528c6/jitc-2021-002432f04.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/ed56/8166607/318b93a6e08f/jitc-2021-002432f05.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/ed56/8166607/1edb397f5cee/jitc-2021-002432f06.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/ed56/8166607/f96ec004370d/jitc-2021-002432f07.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/ed56/8166607/af456d376cd3/jitc-2021-002432f08.jpg

相似文献

[1]
In situ delivery of iPSC-derived dendritic cells with local radiotherapy generates systemic antitumor immunity and potentiates PD-L1 blockade in preclinical poorly immunogenic tumor models.

J Immunother Cancer. 2021-5

[2]
Releasing the brakes of tumor immunity with anti-PD-L1 and pushing its accelerator with L19-IL2 cures poorly immunogenic tumors when combined with radiotherapy.

J Immunother Cancer. 2021-3

[3]
IFNβ Is a Potent Adjuvant for Cancer Vaccination Strategies.

Front Immunol. 2021

[4]
Case Report: Vaccination by Autologous CD16 Dendritic Cells and Anti-PD-L 1 Antibody Synergized With Radiotherapy To Boost T Cells-Mediated Antitumor Efficacy In A Psoriatic Patient With Cutaneous Squamous Cell Carcinoma.

Front Immunol. 2021

[5]
Modulation of lactate-lysosome axis in dendritic cells by clotrimazole potentiates antitumor immunity.

J Immunother Cancer. 2021-5

[6]
STING agonist-based treatment promotes vascular normalization and tertiary lymphoid structure formation in the therapeutic melanoma microenvironment.

J Immunother Cancer. 2021-2

[7]
Skin dendritic cells in melanoma are key for successful checkpoint blockade therapy.

J Immunother Cancer. 2021-1

[8]
Neoadjuvant Immunomodulation Enhances Systemic Antitumor Immunity against Highly Metastatic Tumors.

Cancer Res. 2021-12-15

[9]
DNGR-1 limits Flt3L-mediated antitumor immunity by restraining tumor-infiltrating type I conventional dendritic cells.

J Immunother Cancer. 2021-5

[10]
Blockade of programmed death ligand 1 enhances the therapeutic efficacy of combination immunotherapy against melanoma.

J Immunol. 2010-3-1

引用本文的文献

[1]
Induced Pluripotent Stem Cell-Based Cancer Immunotherapy: Strategies and Perspectives.

Biomedicines. 2025-8-19

[2]
Local CpG- siRNA treatment improves antitumor effects of immune checkpoint inhibitors.

Mol Ther Nucleic Acids. 2024-10-9

[3]
Building a human lung from pluripotent stem cells to model respiratory viral infections.

Respir Res. 2024-7-15

[4]
Neoantigen-augmented iPSC cancer vaccine combined with radiotherapy promotes antitumor immunity in poorly immunogenic cancers.

NPJ Vaccines. 2024-5-31

[5]
Intratumoral delivery of immunotherapy to treat breast cancer: current development in clinical and preclinical studies.

Front Immunol. 2024

[6]
PD-L1 macrophages suppress T cell-mediated anticancer immunity.

Oncoimmunology. 2024

[7]
Exploring the promising potential of induced pluripotent stem cells in cancer research and therapy.

Mol Cancer. 2023-11-28

[8]
Dendritic Cell Vaccination in Non-Small Cell Lung Cancer: Remodeling the Tumor Immune Microenvironment.

Cells. 2023-10-4

[9]
Application of Induced Pluripotent Stem Cells in Malignant Solid Tumors.

Stem Cell Rev Rep. 2023-11

[10]
Eribulin promotes proliferation of CD8 T cells and potentiates T cell-mediated anti-tumor activity against triple-negative breast cancer cells.

Breast Cancer Res Treat. 2024-1

本文引用的文献

[1]
Overcoming primary and acquired resistance to anti-PD-L1 therapy by induction and activation of tumor-residing cDC1s.

Nat Commun. 2020-10-27

[2]
Hematopoietic stem cells from pluripotent stem cells: Clinical potential, challenges, and future perspectives.

Stem Cells Transl Med. 2020-12

[3]
Engineering dendritic cell vaccines to improve cancer immunotherapy.

Nat Commun. 2019-11-27

[4]
Type I Interferon Delivery by iPSC-Derived Myeloid Cells Elicits Antitumor Immunity via XCR1 Dendritic Cells.

Cell Rep. 2019-10-1

[5]
Dendritic cells in cancer immunology and immunotherapy.

Nat Rev Immunol. 2019-8-29

[6]
Systemic clinical tumor regressions and potentiation of PD1 blockade with in situ vaccination.

Nat Med. 2019-4-8

[7]
Subsets of exhausted CD8 T cells differentially mediate tumor control and respond to checkpoint blockade.

Nat Immunol. 2019-2-18

[8]
Intratumoral Tcf1PD-1CD8 T Cells with Stem-like Properties Promote Tumor Control in Response to Vaccination and Checkpoint Blockade Immunotherapy.

Immunity. 2019-1-8

[9]
Towards superior dendritic-cell vaccines for cancer therapy.

Nat Biomed Eng. 2018-6

[10]
Large-Scale Human Dendritic Cell Differentiation Revealing Notch-Dependent Lineage Bifurcation and Heterogeneity.

Cell Rep. 2018-8-14

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