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

Akt 抑制增强了具有记忆细胞特征的强效肿瘤特异性淋巴细胞的扩增。

Akt inhibition enhances expansion of potent tumor-specific lymphocytes with memory cell characteristics.

机构信息

National Cancer Institute (NCI), NIH, Bethesda, Maryland. Department of Surgery, University of California Los Angeles, Los Angeles, California. Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom.

National Cancer Institute (NCI), NIH, Bethesda, Maryland.

出版信息

Cancer Res. 2015 Jan 15;75(2):296-305. doi: 10.1158/0008-5472.CAN-14-2277. Epub 2014 Nov 28.


DOI:10.1158/0008-5472.CAN-14-2277
PMID:25432172
原文链接:https://pmc.ncbi.nlm.nih.gov/articles/PMC4384335/
Abstract

Adoptive cell therapy (ACT) using autologous tumor-infiltrating lymphocytes (TIL) results in complete regression of advanced cancer in some patients, but the efficacy of this potentially curative therapy may be limited by poor persistence of TIL after adoptive transfer. Pharmacologic inhibition of the serine/threonine kinase Akt has recently been shown to promote immunologic memory in virus-specific murine models, but whether this approach enhances features of memory (e.g., long-term persistence) in TIL that are characteristically exhausted and senescent is not established. Here, we show that pharmacologic inhibition of Akt enables expansion of TIL with the transcriptional, metabolic, and functional properties characteristic of memory T cells. Consequently, Akt inhibition results in enhanced persistence of TIL after adoptive transfer into an immunodeficient animal model and augments antitumor immunity of CD8 T cells in a mouse model of cell-based immunotherapy. Pharmacologic inhibition of Akt represents a novel immunometabolomic approach to enhance the persistence of antitumor T cells and improve the efficacy of cell-based immunotherapy for metastatic cancer.

摘要

过继细胞疗法(ACT)使用自体肿瘤浸润淋巴细胞(TIL),可使部分晚期癌症患者完全消退,但这种潜在的治愈疗法的疗效可能受到 TIL 过继转移后持续时间短的限制。最近的研究表明,丝氨酸/苏氨酸激酶 Akt 的药理学抑制作用可促进病毒特异性小鼠模型中的免疫记忆,但这种方法是否能增强 TIL 作为特征性衰竭和衰老的记忆特征(如长期持久性)尚不清楚。在这里,我们发现 Akt 的药理学抑制作用可使 TIL 扩增,并具有记忆 T 细胞的转录、代谢和功能特性。因此,Akt 抑制作用可增强 TIL 在免疫缺陷动物模型中的过继转移后的持久性,并增强细胞免疫治疗小鼠模型中 CD8 T 细胞的抗肿瘤免疫。Akt 的药理学抑制作用代表了一种增强抗肿瘤 T 细胞持久性和提高转移性癌症细胞免疫治疗疗效的新型免疫代谢组学方法。

相似文献

[1]
Akt inhibition enhances expansion of potent tumor-specific lymphocytes with memory cell characteristics.

Cancer Res. 2014-11-28

[2]
Co-stimulation through 4-1BB/CD137 improves the expansion and function of CD8(+) melanoma tumor-infiltrating lymphocytes for adoptive T-cell therapy.

PLoS One. 2013-4-1

[3]
TGF-beta1 induces preferential rapid expansion and persistence of tumor antigen-specific CD8+ T cells for adoptive immunotherapy.

J Immunother. 2010-5

[4]
Tumor-specific CD8 T cells from the bone marrow resist exhaustion and exhibit increased persistence in tumor-bearing hosts as compared with tumor-infiltrating lymphocytes.

J Immunother Cancer. 2025-2-25

[5]
Specific antitumor activity of tumor-infiltrating lymphocytes expanded first in a culture with both anti-CD3 monoclonal antibody and activated B cells and then in a culture with interleukin-2.

Cancer Immunol Immunother. 1995-12

[6]
Costimulation through the CD137/4-1BB pathway protects human melanoma tumor-infiltrating lymphocytes from activation-induced cell death and enhances antitumor effector function.

J Immunother. 2011-4

[7]
Optimizing TIL therapy for uveal melanoma: lessons learned and unlearned from cutaneous melanoma.

Immunotherapy. 2025-3

[8]
Enhanced local and systemic anti-melanoma CD8+ T cell responses after memory T cell-based adoptive immunotherapy in mice.

Cancer Immunol Immunother. 2016-5

[9]
Antiangiogenic agents can increase lymphocyte infiltration into tumor and enhance the effectiveness of adoptive immunotherapy of cancer.

Cancer Res. 2010-7-14

[10]
BRAF inhibitor vemurafenib improves the antitumor activity of adoptive cell immunotherapy.

Cancer Res. 2012-6-12

引用本文的文献

[1]
Ensemble quantitation of absolute metabolite concentrations in T cells reveals conserved features of immunometabolism.

bioRxiv. 2025-6-12

[2]
Recent advances and challenges of cellular immunotherapies in lung cancer treatment.

Exp Hematol Oncol. 2025-7-7

[3]
Rapid enrichment of progenitor exhausted neoantigen-specific CD8 T cells from peripheral blood.

bioRxiv. 2025-5-15

[4]
Beyond Base Camp: Promise and Pitfalls of PI3K/mTOR Inhibition in Pediatric High- Grade Gliomas.

Res Sq. 2025-5-5

[5]
Recent Advances in Immune Checkpoint Inhibitors for Triple-Negative Breast Cancer.

Immunotargets Ther. 2025-4-3

[6]
Aspirin prevents metastasis by limiting platelet TXA suppression of T cell immunity.

Nature. 2025-4

[7]
CaMKK2 Regulates Macrophage Polarization Induced by Matrix Stiffness: Implications for Shaping the Immune Response in Stiffened Tissues.

Adv Sci (Weinh). 2025-4

[8]
Divide and Conquer-Targeted Therapy for Triple-Negative Breast Cancer.

Int J Mol Sci. 2025-2-7

[9]
Mitigating T-cell mitochondrial dysfunction in CLL to augment CAR T-cell therapy: evaluation in an immunocompetent model.

Blood Adv. 2025-5-27

[10]
Novel strategies to manage CAR-T cell toxicity.

Nat Rev Drug Discov. 2025-5

本文引用的文献

[1]
Serial transfer of single-cell-derived immunocompetence reveals stemness of CD8(+) central memory T cells.

Immunity. 2014-7-17

[2]
Adoptive transfer of MART-1 T-cell receptor transgenic lymphocytes and dendritic cell vaccination in patients with metastatic melanoma.

Clin Cancer Res. 2014-5-1

[3]
Uncoupling T-cell expansion from effector differentiation in cell-based immunotherapy.

Immunol Rev. 2014-1

[4]
Inhibiting glycolytic metabolism enhances CD8+ T cell memory and antitumor function.

J Clin Invest. 2013-9-16

[5]
CD8 memory T cells have a bioenergetic advantage that underlies their rapid recall ability.

Proc Natl Acad Sci U S A. 2013-8-12

[6]
mTORC1 regulates CD8+ T-cell glucose metabolism and function independently of PI3K and PKB.

Biochem Soc Trans. 2013-4

[7]
Role of PI3K/Akt signaling in memory CD8 T cell differentiation.

Front Immunol. 2013-2-1

[8]
Transcriptional control of effector and memory CD8+ T cell differentiation.

Nat Rev Immunol. 2012-10-19

[9]
mTOR signaling in growth control and disease.

Cell. 2012-4-13

[10]
Signal integration by Akt regulates CD8 T cell effector and memory differentiation.

J Immunol. 2012-3-30

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