肿瘤微环境多胺抑制T细胞抗肿瘤活性
DOI:
CSTR:
作者:
作者单位:

湖北工业大学生命科学与健康工程学院

作者简介:

通讯作者:

中图分类号:

基金项目:

国家自然科学基金(31971150),湖北省创新群体项目(2024AFA014),湖北省杰出青年基金(2019CFA069)和细胞调控与分子药物“111”引智基地青年学者国际合作研究基金(XBTK-2024010)资助


Tumor Microenvironment Polyamines Inhibit T Cell Antitumor Activity
Author:
Affiliation:

School of Life and Health Sciences, Hubei University of Technology

Fund Project:

The National Natural Science Foundation of China (31971150) , Creative Research Groups grant of Hubei Province (2024AFA014), The Project of Hubei Province Fund for Distinguished Young Scholars (2019CFA069) and Funded by the Collaborative Grant-in-Aid of the HBUT National “111” Center for Cellular Regulation and Molecular Pharmaceutics (XBTK-2024010)

  • 摘要
  • |
  • 图/表
  • |
  • 访问统计
  • |
  • 参考文献
  • |
  • 相似文献
  • |
  • 引证文献
  • |
  • 资源附件
  • |
  • 文章评论
    摘要:

    肿瘤免疫治疗是继手术、放疗、化疗之后的第四大肿瘤治疗技术,克服肿瘤免疫微环境(Tumor microenvironment, TME)的免疫抑制作用已成为当前研究的核心问题。多胺作为重要的免疫调节因子,在TME中常异常积聚,对肿瘤浸润性T细胞具有严重的抑制作用。因此,系统探讨多胺对T细胞功能的调控,对提升免疫治疗效果和解决免疫耐药问题具有重要意义。多胺阻断治疗(Polyamine blocking therapy, PBT)作为新型辅助肿瘤免疫治疗策略,通过降低TME多胺水平恢复T细胞功能,并且联合免疫检查点抑制剂(Immune checkpoint inhibitors, ICIs)治疗时展现出克服耐药的潜力。尽管已有研究揭示多胺对T细胞免疫功能具有抑制作用,但其中的调控机制仍有待进一步阐明。此外,考虑到肿瘤细胞存在多胺代偿机制,PBT应采用多重机制抑制策略,以提高治疗的有效性和安全性。未来,PBT的临床转化可联合多组学技术,以及结合纳米递药系统,提升PBT在肿瘤免疫治疗中的应用潜力。本文阐述了TME中多胺对T细胞免疫功能的调控作用,旨在为肿瘤免疫治疗提供参考。

    Abstract:

    Tumor immunotherapy has emerged as the fourth major therapeutic modality, following surgery, radiotherapy, and chemotherapy. Unlike traditional treatments that primarily target tumor cells directly, immunotherapy harnesses the body's immune system to recognize and eliminate cancer cells. Over the past decade, various immunotherapeutic strategies have been developed, including immune checkpoint inhibitors (ICIs), chimeric antigen receptor (CAR) T cell therapy, cancer vaccines, and cytokine-based therapies. However, the immunosuppressive tumor microenvironment (TME) poses a significant obstacle to the effectiveness of these treatments. Polyamines, including putrescine, spermidine, and spermine, are polycationic metabolites that often accumulate abnormally in the TME and serve as critical immunoregulatory molecules. T cells play a central role in antitumor immunity, yet their function is often influenced by immunoregulatory factors within the TME. Elevated polyamine levels in the TME have been implicated in dampening antitumor T cell responses, thereby facilitating tumor immune evasion. Polyamines in the TME originate from both tumor cells and tumor-associated immune cells. Tumor cells often overexpress the oncogene MYC, which drives the upregulation of polyamine biosynthetic enzymes, resulting in excessive intracellular polyamine production. Additionally, M2-polarized tumor-associated macrophages (M2-TAMs) contribute to polyamine accumulation by upregulating arginase-I (Arg-Ⅰ), an enzyme that catalyzes the conversion of arginine into ornithine—a key precursor in the polyamine biosynthetic pathway. These combined sources lead to sustained polyamine enrichment in the TME, contributing to immune dysfunction and supporting tumor progression. Moreover, polyamines indirectly affect T cell activity by modulating macrophage polarization and directly suppress tumor cell apoptosis, further promoting an immunosuppressive environment. This review highlights the multifaceted roles of polyamines in modulating tumor-infiltrating T cell function, with a particular focus on their influence on CD4? T cell differentiation, CD8? T cell cytotoxicity, and immune checkpoint molecule expression. Recent studies suggest that polyamines suppress CD4? T cell activation and differentiation by modulating the MAPK/ERK signaling pathway. Additionally, polyamines can impair T cell receptor (TCR) signaling and promote immune evasion through the upregulation of PD-L1 expression on tumor cells. These effects collectively contribute to weakened antitumor T cell responses. Polyamine blocking therapy (PBT), which primarily targets polyamine biosynthesis and transport, has emerged as a novel adjunctive immunotherapeutic strategy in cancer treatment. By reducing polyamine levels in the TME, PBT restores T cell effector functions and helps alleviate immunosuppression. Notably, studies have demonstrated that combining PBT with ICIs produces synergistic antitumor effects and may overcome resistance to ICI monotherapy. Although studies have revealed the inhibitory effects of polyamines on T cell immune function, the underlying regulatory mechanisms remain to be fully elucidated. Moreover, due to compensatory mechanisms employed by tumor cells to maintain polyamine homeostasis, multi-targeted approaches may be necessary to achieve safe and effective therapeutic outcomes. Future PBT strategies may benefit from the integration of multi-omics technologies and the development of nanocarrier-based drug delivery systems, which could collectively enhance their specificity, efficacy, and applicability in cancer immunotherapy. This review systematically elucidates the immunomodulatory effects of polyamines on T cell function within the TME and provides theoretical support and novel insights for the advancement of tumor immunotherapeutic strategies.

    参考文献
    相似文献
    引证文献
引用本文

艾元宝,黄雪梅,刘森.肿瘤微环境多胺抑制T细胞抗肿瘤活性[J].生物化学与生物物理进展,,():

复制
分享
文章指标
  • 点击次数:
  • 下载次数:
  • HTML阅读次数:
  • 引用次数:
历史
  • 收稿日期:2025-01-15
  • 最后修改日期:2025-05-14
  • 接受日期:2025-05-14
  • 在线发布日期:
  • 出版日期: