Liu Qianwen, Li Jingfeng, Sun Xiuqiao, Lin Jiayu, Yu Zhengwei, Xiao Yue, Li Dan, Sun Baofa, Bao Haili, Liu Yihao
Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
State Key Laboratory of Systems Medicine for Cancer, Shanghai, 200025, China.
J Hematol Oncol. 2025 Aug 22;18(1):81. doi: 10.1186/s13045-025-01735-w.
Immunosenescence, the age-related decline in immune function, profoundly impacts cancer progression and therapeutic outcomes by fostering a tumor-promoting microenvironment and impairing immune surveillance. This review delineates eleven molecular hallmarks of immunosenescence, including genomic instability, telomere attrition, epigenetic dysregulation, mitochondrial dysfunction, and chronic inflammation, which collectively drive immune cell dysfunction and systemic immunosuppression. Aging reshapes the tumor microenvironment (TME) through recruitment of immunosuppressive cells, senescence-associated secretory phenotypes (SASP), and metabolic reprogramming, contributing to therapy resistance and poor prognosis in elderly patients. While immunotherapies such as immune checkpoint inhibitors (ICIs) and chimeric antigen receptor T-cell immunotherapy (CAR-T) cells show promise, their efficacy in aging populations is limited by T cell exhaustion, myeloid bias, and altered intercellular communication. Emerging strategies-including senolytics, epigenetic modulators (e.g., histone deacetylase (HDAC) inhibitor), and metabolic interventions (e.g., spermidine, nicotinamide mononucleotide (NMN))-highlight potential avenues to rejuvenate aged immunity. Single-cell multi-omics (single cell RNA-seq, single cell ATAC-seq) further unravel immune cell heterogeneity, revealing tissue-specific chromatin accessibility dynamics and novel targets like interleukin-34 (IL-34) for microglia-mediated neuroinflammation. However, challenges persist in translating preclinical findings to clinical practice, necessitating age-tailored trials and biomarker-driven approaches. By integrating mechanistic insights with translational innovations, this review underscores the urgency of addressing immunosenescence to optimize cancer immunotherapy for aging populations, ultimately bridging the gap between aging biology and precision oncology.
免疫衰老,即与年龄相关的免疫功能衰退,通过促进肿瘤微环境的形成和损害免疫监视,深刻影响癌症进展和治疗效果。本综述阐述了免疫衰老的11个分子特征,包括基因组不稳定、端粒磨损、表观遗传失调、线粒体功能障碍和慢性炎症,这些特征共同导致免疫细胞功能障碍和全身免疫抑制。衰老通过招募免疫抑制细胞、衰老相关分泌表型(SASP)和代谢重编程重塑肿瘤微环境(TME),导致老年患者产生治疗抵抗和预后不良。虽然免疫检查点抑制剂(ICI)和嵌合抗原受体T细胞免疫疗法(CAR-T)等免疫疗法显示出前景,但其在老年人群中的疗效受到T细胞耗竭、髓系偏向和细胞间通讯改变的限制。新兴策略,包括衰老细胞溶解剂、表观遗传调节剂(如组蛋白脱乙酰酶(HDAC)抑制剂)和代谢干预(如亚精胺、烟酰胺单核苷酸(NMN)),突出了恢复衰老免疫力的潜在途径。单细胞多组学(单细胞RNA测序、单细胞ATAC测序)进一步揭示了免疫细胞的异质性,揭示了组织特异性染色质可及性动态以及小胶质细胞介导的神经炎症的新靶点,如白细胞介素-34(IL-34)。然而,将临床前研究结果转化为临床实践仍存在挑战,需要进行针对年龄的试验和生物标志物驱动的方法。通过将机制见解与转化创新相结合,本综述强调了解决免疫衰老以优化老年人群癌症免疫治疗的紧迫性,最终弥合衰老生物学与精准肿瘤学之间的差距。
Cancer Biol Med. 2025-6-10
Mol Cancer. 2025-7-26
Med Oncol. 2025-6-8
J Exp Clin Cancer Res. 2025-8-23
Signal Transduct Target Ther. 2025-7-31
Clin Epigenetics. 2025-3-21