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

获得性癌症免疫疗法耐药性:肿瘤介导免疫抑制的作用。

Acquired resistance to cancer immunotherapy: Role of tumor-mediated immunosuppression.

机构信息

Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar.

Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar; Institute of Cancer Sciences, University of Manchester, Manchester, United Kingdom.

出版信息

Semin Cancer Biol. 2020 Oct;65:13-27. doi: 10.1016/j.semcancer.2019.07.017. Epub 2019 Jul 27.


DOI:10.1016/j.semcancer.2019.07.017
PMID:31362073
Abstract

In the tumor microenvironment (TME), tumor cells are constantly evolving to reduce neoantigen generation and the mutational burden to escape the anti-tumor response. This will lower tumor reactivity to the adaptive immune response and give rise to tumor intrinsic factors, such as altered expression of immune regulatory molecules on tumor cells. Tumor-extrinsic factors, such as immunosuppressive cells, soluble suppressive molecules or inhibitory receptors expressed by immune cells will alter the composition and activity of tumor-infiltrating lymphocytes (TILs) (by increasing T regulatory cells:T effector cells ratio and inhibiting T effector cell function) and promote tumor growth and metastasis. Together, these factors limit the response rates and clinical outcomes to a particular cancer therapy. Within the TME, the cross-talks between immune and non-immune cells result in the generation of positive feedback loops, which augment immunosuppression and support tumor growth and survival (termed as tumor-mediated immunosuppression). Cancer immunotherapies, such as immune checkpoint inhibitors (ICIs) and adoptive cell transfer (ACT), have shown therapeutic efficacy in hematologic cancers and different types of solid tumors. However, achieving durable response rates in some cancer patients remains a challenge as a result of acquired resistance and tumor immune evasion. This could be driven by the cellular and molecular suppressive network within the TME or due to the loss of tumor antigens. In this review, we describe the contribution of the immunosuppressive cellular and molecular tumor network to the development of acquired resistance against cancer immunotherapies. We also discuss potential combined therapeutic strategies which could help to overcome such resistance against cancer immunotherapies, and to enhance anti-tumor immune responses and improve clinical outcomes in patients.

摘要

在肿瘤微环境 (TME) 中,肿瘤细胞不断进化以减少新抗原生成和突变负担,从而逃避抗肿瘤反应。这将降低肿瘤对适应性免疫反应的反应性,并导致肿瘤内在因素,例如肿瘤细胞上免疫调节分子的表达改变。肿瘤外在因素,如免疫抑制细胞、可溶性抑制分子或免疫细胞表达的抑制性受体,将改变肿瘤浸润淋巴细胞 (TIL) 的组成和活性(通过增加调节性 T 细胞:效应 T 细胞的比例并抑制效应 T 细胞功能),并促进肿瘤生长和转移。这些因素共同限制了特定癌症治疗的反应率和临床结果。在 TME 中,免疫和非免疫细胞之间的串扰导致正反馈环的产生,增强了免疫抑制作用,并支持肿瘤生长和存活(称为肿瘤介导的免疫抑制)。癌症免疫疗法,如免疫检查点抑制剂 (ICI) 和过继细胞转移 (ACT),已在血液系统癌症和不同类型的实体瘤中显示出治疗效果。然而,由于获得性耐药和肿瘤免疫逃逸,一些癌症患者仍难以实现持久的反应率。这可能是由于 TME 中的细胞和分子抑制性网络驱动的,也可能是由于肿瘤抗原的丢失。在这篇综述中,我们描述了免疫抑制性细胞和分子肿瘤网络对癌症免疫疗法获得性耐药的发展的贡献。我们还讨论了潜在的联合治疗策略,这些策略可以帮助克服癌症免疫疗法的这种耐药性,并增强抗肿瘤免疫反应,改善患者的临床结果。

相似文献

[1]
Acquired resistance to cancer immunotherapy: Role of tumor-mediated immunosuppression.

Semin Cancer Biol. 2020-10

[2]
Treg-mediated acquired resistance to immune checkpoint inhibitors.

Cancer Lett. 2019-5-9

[3]
FoxP3 T regulatory cells in cancer: Prognostic biomarkers and therapeutic targets.

Cancer Lett. 2020-10-10

[4]
Overcoming tumor-mediated immunosuppression.

Immunotherapy. 2014

[5]
Hijacked Immune Cells in the Tumor Microenvironment: Molecular Mechanisms of Immunosuppression and Cues to Improve T Cell-Based Immunotherapy of Solid Tumors.

Int J Mol Sci. 2021-5-27

[6]
Combining epigenetic and immune therapy to overcome cancer resistance.

Semin Cancer Biol. 2020-10

[7]
Reduction of immunosuppressive tumor microenvironment in cholangiocarcinoma by ex vivo targeting immune checkpoint molecules.

J Hepatol. 2019-6-11

[8]
Expression of costimulatory and inhibitory receptors in FoxP3 regulatory T cells within the tumor microenvironment: Implications for combination immunotherapy approaches.

Adv Cancer Res. 2019-6-6

[9]
Common phenotypic dynamics of tumor-infiltrating lymphocytes across different histologies upon checkpoint inhibition: impact on clinical outcome.

Cytotherapy. 2020-3-20

[10]
CD8 cytotoxic T lymphocytes in cancer immunotherapy: A review.

J Cell Physiol. 2018-11-22

引用本文的文献

[1]
Multifunctional Superparamagnetic Copper Iron Oxide Nanoparticles for Synergistic Cancer Therapy via Magnetic Hyperthermia, Oxidative Stress and Immune Reprogramming.

Adv Funct Mater. 2025-3-28

[2]
Pareto task inference analysis reveals cellular trade-offs in diffuse large B-Cell lymphoma transcriptomic data.

Front Syst Biol. 2024-3-1

[3]
Targeting tumor-associated macrophages to overcome immune checkpoint inhibitor resistance in hepatocellular carcinoma.

J Exp Clin Cancer Res. 2025-8-5

[4]
Neoantigen-driven personalized tumor therapy: An update from discovery to clinical application.

Chin Med J (Engl). 2025-9-5

[5]
Overcoming resistant cancerous tumors through combined photodynamic and immunotherapy (photoimmunotherapy).

Front Immunol. 2025-7-17

[6]
Nanoliposomal PD-1 antagonist target tumor-draining lymph nodes to revitalize T cells and improve anti-tumor effect in hepatocellular carcinoma.

J Nanobiotechnology. 2025-7-30

[7]
In Situ Vaccination with a Vpr-Derived Peptide Elicits Systemic Antitumor Immunity by Improving Tumor Immunogenicity.

Vaccines (Basel). 2025-6-30

[8]
Identification of anoikis-related subtypes and a risk score prognosis model, the association with TME landscapes and therapeutic responses in hepatocellular carcinoma.

Front Immunol. 2025-6-17

[9]
Nanotechnology-driven strategies in postoperative cancer treatment: innovations in drug delivery systems.

Front Pharmacol. 2025-4-30

[10]
Cancer immunotherapy by silencing transcription factor c-Rel using peptide-based nanoparticles.

Front Immunol. 2025-3-11

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