陈锦锋, 马瑜婷. 砷剂调控肿瘤细胞命运的机制及激活抗肿瘤免疫的潜力[J]. 中国肿瘤临床, 2021, 48(21): 1113-1119. DOI: 10.12354/j.issn.1000-8179.2021.20210903
引用本文: 陈锦锋, 马瑜婷. 砷剂调控肿瘤细胞命运的机制及激活抗肿瘤免疫的潜力[J]. 中国肿瘤临床, 2021, 48(21): 1113-1119. DOI: 10.12354/j.issn.1000-8179.2021.20210903
Jinfeng Chen, Yuting Ma. Cell fate regulatory mechanisms of arsenicals and their potential in triggering anti-tumor immunity[J]. CHINESE JOURNAL OF CLINICAL ONCOLOGY, 2021, 48(21): 1113-1119. DOI: 10.12354/j.issn.1000-8179.2021.20210903
Citation: Jinfeng Chen, Yuting Ma. Cell fate regulatory mechanisms of arsenicals and their potential in triggering anti-tumor immunity[J]. CHINESE JOURNAL OF CLINICAL ONCOLOGY, 2021, 48(21): 1113-1119. DOI: 10.12354/j.issn.1000-8179.2021.20210903

砷剂调控肿瘤细胞命运的机制及激活抗肿瘤免疫的潜力

Cell fate regulatory mechanisms of arsenicals and their potential in triggering anti-tumor immunity

  • 摘要: 中药砷剂的主要活性成分是三氧化二砷(arsenic trioxide,ATO),对急性早幼粒细胞白血病(acute promyelocytic leukemia,APL)等血液肿瘤显示出良好的临床疗效。尽管ATO在体外实验中能高效杀伤多种癌细胞,其单药治疗实体肿瘤的临床获益却并不理想,而联合用药以及药物靶向递送等策略也尚未取得突破。ATO药物作用机理复杂,可诱导活性氧的生成、抑制谷胱甘肽过氧化物酶及多种代谢限速酶的活性、引发细胞氧化应激、激活线粒体介导的细胞凋亡通路等。长期以来,ATO的细胞毒性及其诱导肿瘤细胞死亡的机制一直是研究的重点,ATO能否增强肿瘤细胞的免疫原性、调控抗肿瘤免疫应答尚存在认知空白。本文总结ATO的临床应用现状,归纳了其对多种细胞死亡方式的影响,分析了相关分子机制。此外,基于诱导免疫原性细胞死亡的理念,本文初步探讨了利用砷剂制备预防型及治疗型肿瘤全细胞疫苗的可能,并展望了该疫苗联合多种免疫治疗策略提升实体瘤疗效的思路和可能性。

     

    Abstract: As a principal bioactive member of arsenic compounds, arsenic trioxide (ATO) is highly effective in treating hematologic malignancies, particularly acute promyelocytic leukemia (APL). Although ATO has strong tumoricidal activities in vitro, the clinical benefits of ATO monotherapy against solid tumors are often limited. Strategic exploration of novel ATO-based anti-cancer therapies have recently been promoted because of various drug combinations and targeted drug delivery approaches. However, breakthroughs in therapeutic outcomes have rarely been achieved. To that end, multiple mechanisms of action have been documented for ATO, including triggering reactive oxygen species generation, inhibiting the activity of glutathione peroxidase and several rate-limiting metabolic enzymes, causing oxidative stress, and initiating the mitochondrial apoptotic pathway. In particular, researches have long been focused on ATO cytotoxicity and the molecular mechanisms underlying ATO-induced cell death. However, whether ATO can boost tumor immunogenicity and influence anti-tumor immunity remains largely unknown. Therefore, we intend to summarize the current clinical applications of ATO, recapitulate its impacts on various cell death modalities, and analyze molecular associations among different cell death pathways. This review also preliminarily explores the possibility of whole-cell tumor vaccine development by maximizing ATO-triggered immunogenic cell death. Furthermore, the combination of immunotherapies with ATO-based tumor vaccine may provide novel possibilities for cancer treatment.

     

/

返回文章
返回