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

个体化肿瘤 RNA 负载脂质纳米颗粒为癌症免疫治疗的全身和肿瘤内环境做出反应做好准备。

Personalized Tumor RNA Loaded Lipid-Nanoparticles Prime the Systemic and Intratumoral Milieu for Response to Cancer Immunotherapy.

机构信息

Preston A. Wells Jr. Center for Brain Tumor Therapy, UF Brain Tumor Immunotherapy Program, McKnight Brain Institute, Lillian S. Wells Department of Neurosurgery, College of Medicine , University of Florida , Gainesville , Florida 32611 , United States.

Department of Pathology, College of Medicine , University of Florida , Gainesville , Florida 32611 , United States.

出版信息

Nano Lett. 2018 Oct 10;18(10):6195-6206. doi: 10.1021/acs.nanolett.8b02179. Epub 2018 Sep 27.


DOI:10.1021/acs.nanolett.8b02179
PMID:30259750
原文链接:https://pmc.ncbi.nlm.nih.gov/articles/PMC6597257/
Abstract

Translation of nanoparticles (NPs) into human clinical trials for patients with refractory cancers has lagged due to unknown biologic reactivities of novel NP designs. To overcome these limitations, simple well-characterized mRNA lipid-NPs have been developed as cancer immunotherapeutic vaccines. While the preponderance of RNA lipid-NPs encoding for tumor-associated antigens or neoepitopes have been designed to target lymphoid organs, they remain encumbered by the profound intratumoral and systemic immunosuppression that may stymie an activated T cell response. Herein, we show that systemic localization of untargeted tumor RNA (derived from whole transcriptome) encapsulated in lipid-NPs, with excess positive charge, primes the peripheral and intratumoral milieu for response to immunotherapy. In immunologically resistant tumor models, these RNA-NPs activate the preponderance of systemic and intratumoral myeloid cells (characterized by coexpression of PD-L1 and CD86). Addition of immune checkpoint inhibitors (ICIs) (to animals primed with RNA-NPs) augments peripheral/intratumoral PD-1CD8 cells and mediates synergistic antitumor efficacy in settings where ICIs alone do not confer therapeutic benefit. These synergistic effects are mediated by type I interferon released from plasmacytoid dendritic cells (pDCs). In translational studies, personalized mRNA-NPs were safe and active in a client-owned canine with a spontaneous malignant glioma. In summary, we demonstrate widespread immune activation from tumor loaded RNA-NPs concomitant with inducible PD-L1 expression that can be therapeutically exploited. While immunotherapy remains effective for only a subset of cancer patients, combination therapy with systemic immunomodulating RNA-NPs may broaden its therapeutic potency.

摘要

由于新型 NP 设计的未知生物学反应性,将纳米颗粒 (NPs) 转化为难治性癌症患者的人体临床试验一直滞后。为了克服这些限制,已经开发了简单的、特征明确的 mRNA 脂质-NP 作为癌症免疫治疗疫苗。虽然大多数编码肿瘤相关抗原或新表位的 RNA 脂质-NP 旨在靶向淋巴器官,但它们仍然受到深刻的肿瘤内和全身免疫抑制的阻碍,这可能会阻碍激活的 T 细胞反应。在这里,我们表明,封装在脂质-NP 中的未靶向肿瘤 RNA(来自全转录组)的系统定位,带有过多的正电荷,为免疫治疗的反应在周围和肿瘤内环境中做好准备。在免疫抵抗性肿瘤模型中,这些 RNA-NP 激活了占优势的全身和肿瘤内髓样细胞(其特征是 PD-L1 和 CD86 的共表达)。添加免疫检查点抑制剂 (ICI)(对用 RNA-NP 进行预处理的动物)可增加外周/肿瘤内 PD-1CD8 细胞,并在 ICI 单独治疗没有带来治疗益处的情况下介导协同的抗肿瘤疗效。这些协同效应是由浆细胞样树突状细胞 (pDC) 释放的 I 型干扰素介导的。在转化研究中,个性化的 mRNA-NP 在一只患有自发性恶性脑肿瘤的客户拥有的犬中是安全且有效的。总之,我们证明了负载肿瘤的 RNA-NP 可引起广泛的免疫激活,同时诱导 PD-L1 表达,这可被治疗性利用。虽然免疫疗法仍然对少数癌症患者有效,但与全身性免疫调节 RNA-NP 的联合治疗可能会扩大其治疗效力。

相似文献

[1]
Personalized Tumor RNA Loaded Lipid-Nanoparticles Prime the Systemic and Intratumoral Milieu for Response to Cancer Immunotherapy.

Nano Lett. 2018-9-27

[2]
Immunotherapy of Tumor RNA-Loaded Lipid Nanoparticles Against Hepatocellular Carcinoma.

Int J Nanomedicine. 2021

[3]
Phase I Trial of Intratumoral Injection of Gene-Modified Dendritic Cells in Lung Cancer Elicits Tumor-Specific Immune Responses and CD8 T-cell Infiltration.

Clin Cancer Res. 2017-8-15

[4]
Blockage of immune checkpoint molecules increases T-cell priming potential of dendritic cell vaccine.

Immunology. 2019-10-24

[5]
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

[6]
A TLR3-Specific Adjuvant Relieves Innate Resistance to PD-L1 Blockade without Cytokine Toxicity in Tumor Vaccine Immunotherapy.

Cell Rep. 2017-5-30

[7]
Reducing PD-L1 expression with a self-assembled nanodrug: an alternative to PD-L1 antibody for enhanced chemo-immunotherapy.

Theranostics. 2021

[8]
Synthetic High-density Lipoprotein Nanodiscs for Personalized Immunotherapy Against Gliomas.

Clin Cancer Res. 2020-8-15

[9]
Utilizing chemotherapy-induced tumor RNA nanoparticles to improve cancer chemoimmunotherapy.

Acta Biomater. 2023-3-1

[10]
Highly Enhanced Antitumor Immunity by a Three-Barreled Strategy of the l-Arginine-Promoted Nanovaccine and Gene-Mediated PD-L1 Blockade.

ACS Appl Mater Interfaces. 2020-9-16

引用本文的文献

[1]
Nanotechnology for immuno-oncology.

Nat Cancer. 2025-8-7

[2]
Sensitization of tumours to immunotherapy by boosting early type-I interferon responses enables epitope spreading.

Nat Biomed Eng. 2025-7-18

[3]
Immunotherapy for High-Grade Gliomas.

Cancers (Basel). 2025-5-31

[4]
Immunomodulatory nanoplatforms with multiple mechanisms of action in cancer treatment.

Nanomedicine (Lond). 2025-6

[5]
Peptide-Coated Nanoparticles for Noninvasive Biomedical Imaging.

Methods Mol Biol. 2025

[6]
Biomimetic Nanoparticle Based Targeted mRNA Vaccine Delivery as a Novel Therapy for Glioblastoma Multiforme.

AAPS PharmSciTech. 2025-2-21

[7]
Multi-Step Assembly of an RNA-Liposome Nanoparticle Formulation Revealed by Real-Time, Single-Particle Quantitative Imaging.

Adv Sci (Weinh). 2025-3

[8]
Immunotherapy for glioblastoma: current state, challenges, and future perspectives.

Cell Mol Immunol. 2024-12

[9]
Personalized mRNA vaccines in glioblastoma therapy: from rational design to clinical trials.

J Nanobiotechnology. 2024-10-4

[10]
Bidirectional crosstalk between therapeutic cancer vaccines and the tumor microenvironment: Beyond tumor antigens.

Fundam Res. 2022-3-26

本文引用的文献

[1]
PD-L1 on host cells is essential for PD-L1 blockade-mediated tumor regression.

J Clin Invest. 2018-1-16

[2]
Recurrent glioma clinical trial, CheckMate-143: the game is not over yet.

Oncotarget. 2017-10-6

[3]
Rindopepimut with temozolomide for patients with newly diagnosed, EGFRvIII-expressing glioblastoma (ACT IV): a randomised, double-blind, international phase 3 trial.

Lancet Oncol. 2017-8-23

[4]
Lipid Nanoparticle Assisted mRNA Delivery for Potent Cancer Immunotherapy.

Nano Lett. 2017-3-8

[5]
Systemic activation of antigen-presenting cells via RNA-loaded nanoparticles.

Oncoimmunology. 2016-11-18

[6]
Mutations Associated with Acquired Resistance to PD-1 Blockade in Melanoma.

N Engl J Med. 2016-9-1

[7]
Particle-mediated Intravenous Delivery of Antigen mRNA Results in Strong Antigen-specific T-cell Responses Despite the Induction of Type I Interferon.

Mol Ther Nucleic Acids. 2016-6-21

[8]
Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy.

Nature. 2016-6-1

[9]
Vaccines for established cancer: overcoming the challenges posed by immune evasion.

Nat Rev Cancer. 2016-3-11

[10]
Prospective identification of neoantigen-specific lymphocytes in the peripheral blood of melanoma patients.

Nat Med. 2016-4

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