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

基因工程改造的T细胞与免疫检查点阻断联合用于癌症治疗。

Combination of genetically engineered T cells and immune checkpoint blockade for the treatment of cancer.

作者信息

Rossetti Rafaela, Brand Heloísa, Lima Sarah Caroline Gomes, Furtado Izadora Peter, Silveira Roberta Maraninchi, Fantacini Daianne Maciely Carvalho, Covas Dimas Tadeu, de Souza Lucas Eduardo Botelho

机构信息

Center for Cell-Based Therapy, Blood Center of Ribeirão Preto, University of São Paulo, São Paulo, Brazil.

Biotechnology Nucleus - Blood Center of Ribeirão Preto, Butantan Institute, São Paulo, Brazil.

出版信息

Immunother Adv. 2022 Jan 25;2(1):ltac005. doi: 10.1093/immadv/ltac005. eCollection 2022.


DOI:10.1093/immadv/ltac005
PMID:35919489
原文链接:https://pmc.ncbi.nlm.nih.gov/articles/PMC9327125/
Abstract

Immune checkpoint (IC) blockade using monoclonal antibodies is currently one of the most successful immunotherapeutic interventions to treat cancer. By reinvigorating antitumor exhausted T cells, this approach can lead to durable clinical responses. However, the majority of patients either do not respond or present a short-lived response to IC blockade, in part due to a scarcity of tumor-specific T cells within the tumor microenvironment. Adoptive transfer of T cells genetically engineered to express chimeric antigen receptors (CARs) or engineered T-cell receptors (TCRs) provide the necessary tumor-specific immune cell population to target cancer cells. However, this therapy has been considerably ineffective against solid tumors in part due to IC-mediated immunosuppressive effects within the tumor microenvironment. These limitations could be overcome by associating adoptive cell transfer of genetically engineered T cells and IC blockade. In this comprehensive review, we highlight the strategies and outcomes of preclinical and clinical attempts to disrupt IC signaling in adoptive T-cell transfer against cancer. These strategies include combined administration of genetically engineered T cells and IC inhibitors, engineered T cells with intrinsic modifications to disrupt IC signaling, and the design of CARs against IC molecules. The current landscape indicates that the synergy of the fast-paced refinements of gene-editing technologies and synthetic biology and the increased comprehension of IC signaling will certainly translate into a novel and more effective immunotherapeutic approaches to treat patients with cancer.

摘要

使用单克隆抗体的免疫检查点(IC)阻断是目前治疗癌症最成功的免疫治疗干预措施之一。通过重振抗肿瘤耗竭性T细胞,这种方法可带来持久的临床反应。然而,大多数患者对IC阻断要么无反应,要么反应短暂,部分原因是肿瘤微环境中缺乏肿瘤特异性T细胞。过继转移经基因工程改造以表达嵌合抗原受体(CAR)或工程化T细胞受体(TCR)的T细胞,可提供靶向癌细胞所需的肿瘤特异性免疫细胞群体。然而,这种疗法对实体瘤相当无效,部分原因是肿瘤微环境中IC介导的免疫抑制作用。将基因工程改造T细胞的过继性细胞转移与IC阻断相结合可克服这些限制。在这篇综述中,我们重点介绍了在过继性T细胞转移治疗癌症中破坏IC信号传导的临床前和临床尝试的策略及结果。这些策略包括联合给予基因工程改造T细胞和IC抑制剂、对工程化T细胞进行内在修饰以破坏IC信号传导,以及设计针对IC分子的CAR。当前的情况表明,基因编辑技术和合成生物学的快速改进与对IC信号传导的深入理解相结合,必将转化为治疗癌症患者的新颖且更有效的免疫治疗方法。

https://cdn.ncbi.nlm.nih.gov/pmc/blobs/9779/9327125/afd68dd10986/ltac005_fig3.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/9779/9327125/7dac9c665471/ltac005_fig1.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/9779/9327125/2341e07917f7/ltac005_fig2.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/9779/9327125/afd68dd10986/ltac005_fig3.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/9779/9327125/7dac9c665471/ltac005_fig1.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/9779/9327125/2341e07917f7/ltac005_fig2.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/9779/9327125/afd68dd10986/ltac005_fig3.jpg

相似文献

[1]
Combination of genetically engineered T cells and immune checkpoint blockade for the treatment of cancer.

Immunother Adv. 2022-1-25

[2]
Insufficiency of compound immune checkpoint blockade to overcome engineered T cell exhaustion in pancreatic cancer.

J Immunother Cancer. 2022-2

[3]
Prospects for personalized combination immunotherapy for solid tumors based on adoptive cell therapies and immune checkpoint blockade therapies.

Nihon Rinsho Meneki Gakkai Kaishi. 2017

[4]
Perspectives on Chimeric Antigen Receptor T-Cell Immunotherapy for Solid Tumors.

Front Immunol. 2018-5-22

[5]
Incorporation of Immune Checkpoint Blockade into Chimeric Antigen Receptor T Cells (CAR-Ts): Combination or Built-In CAR-T.

Int J Mol Sci. 2018-1-24

[6]
The Complement Receptors C3aR and C5aR Are a New Class of Immune Checkpoint Receptor in Cancer Immunotherapy.

Front Immunol. 2019-7-19

[7]
Genetically engineered T cells for cancer immunotherapy.

Signal Transduct Target Ther. 2019

[8]
CRISPR/Cas systems to overcome challenges in developing the next generation of T cells for cancer therapy.

Adv Drug Deliv Rev. 2020

[9]
FCγ Chimeric Receptor-Engineered T Cells: Methodology, Advantages, Limitations, and Clinical Relevance.

Front Immunol. 2017-4-27

[10]
Current Headway in Cancer Immunotherapy Emphasizing the Practice of Genetically Engineered T Cells to Target Selected Tumor Antigens.

Crit Rev Immunol. 2021

引用本文的文献

[1]
Unconventional Immunotherapies in Cancer: Opportunities and Challenges.

Pharmaceuticals (Basel). 2025-8-4

[2]
Modulating CD226 and PD-(L)1 pathways improves CMV-specific CD8+T cell responses in the absence of IL-2.

BMB Rep. 2025-7

[3]
Neoantigen-based immunotherapy: advancing precision medicine in cancer and glioblastoma treatment through discovery and innovation.

Explor Target Antitumor Ther. 2025-4-27

[4]
A Proteomics Outlook on the Molecular Effectors of CAR-T Cell Therapy in Cancer Management.

J Proteome Res. 2025-6-6

[5]
Adoptive T Cell Therapy Targeting MAGE-A4.

Cancers (Basel). 2025-1-26

[6]
Agent-Based Modeling of Virtual Tumors Reveals the Critical Influence of Microenvironmental Complexity on Immunotherapy Efficacy.

Cancers (Basel). 2024-8-23

[7]
Applications of cell therapy in the treatment of virus-associated cancers.

Nat Rev Clin Oncol. 2024-10

[8]
Contemporary Approaches to Immunotherapy of Solid Tumors.

Cancers (Basel). 2024-6-19

[9]
Overcoming the challenges of primary resistance and relapse after CAR-T cell therapy.

Expert Rev Clin Immunol. 2024-7

[10]
CD8 T cell-based cancer immunotherapy.

J Transl Med. 2024-4-29

本文引用的文献

[1]
PD-1 and TIGIT downregulation distinctly affect the effector and early memory phenotypes of CD19-targeting CAR T cells.

Mol Ther. 2022-2-2

[2]
Effective Killing of Acute Myeloid Leukemia by TIM-3 Targeted Chimeric Antigen Receptor T Cells.

Mol Cancer Ther. 2021-9

[3]
CRISPR/Cas9 mediated deletion of the adenosine A2A receptor enhances CAR T cell efficacy.

Nat Commun. 2021-5-28

[4]
Strategies to Enhance the Therapeutic Efficacy, Applicability, and Safety of Genetically Engineered Immune Cells.

Crit Rev Immunol. 2021

[5]
A novel dominant-negative PD-1 armored anti-CD19 CAR T cell is safe and effective against refractory/relapsed B cell lymphoma.

Transl Oncol. 2021-7

[6]
Transient rest restores functionality in exhausted CAR-T cells through epigenetic remodeling.

Science. 2021-4-2

[7]
First immunotherapeutic CAR-T cells against the immune checkpoint protein HLA-G.

J Immunother Cancer. 2021-3

[8]
Five-Year Outcomes for Refractory B-Cell Lymphomas with CAR T-Cell Therapy.

N Engl J Med. 2021-2-18

[9]
Addition of anti-TIM3 or anti-TIGIT Antibodies to anti-PD1 Blockade Augments Human T cell Adoptive Cell Transfer.

Oncoimmunology. 2021-1-20

[10]
CD19-specific CAR T Cells that Express a PD-1/CD28 Chimeric Switch-Receptor are Effective in Patients with PD-L1-positive B-Cell Lymphoma.

Clin Cancer Res. 2021-1-15

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