高级搜索
曹广文. “癌症进化发育学”理论进展及其在肝细胞癌靶向/免疫治疗中的作用[J]. 肿瘤防治研究, 2022, 49(8): 747-755. DOI: 10.3971/j.issn.1000-8578.2022.22.0377
引用本文: 曹广文. “癌症进化发育学”理论进展及其在肝细胞癌靶向/免疫治疗中的作用[J]. 肿瘤防治研究, 2022, 49(8): 747-755. DOI: 10.3971/j.issn.1000-8578.2022.22.0377
CAO Guangwen. Theoretical Update of Cancer Evo-Dev and Its Role in Targeted Immunotherapy for Hepatocellular Carcinoma[J]. Cancer Research on Prevention and Treatment, 2022, 49(8): 747-755. DOI: 10.3971/j.issn.1000-8578.2022.22.0377
Citation: CAO Guangwen. Theoretical Update of Cancer Evo-Dev and Its Role in Targeted Immunotherapy for Hepatocellular Carcinoma[J]. Cancer Research on Prevention and Treatment, 2022, 49(8): 747-755. DOI: 10.3971/j.issn.1000-8578.2022.22.0377

“癌症进化发育学”理论进展及其在肝细胞癌靶向/免疫治疗中的作用

Theoretical Update of Cancer Evo-Dev and Its Role in Targeted Immunotherapy for Hepatocellular Carcinoma

  • 摘要: 原发性肝癌居我国癌症非成熟死亡原因的首位,其中肝细胞癌占93.0%,84.4%是乙肝病毒(HBV)慢性感染所致。在HBV致癌过程中,非可控性炎性反应通过IL-6上调胞苷脱氨酶AID/APOBEC3s家族成员以促病毒和宿主基因变异、下调尿嘧啶DNA糖基化酶(UNG)等降低变异修复,炎性微环境为HBV和人肝细胞的“变异-选择-适应”进化过程提供了必要条件并促进变异细胞逆向分化。该进化过程需满足体细胞驱动突变和免疫抑制微环境这两个前提条件。促癌基因组不稳定和HBV变异通过cGAS-STING抑制Ⅰ型干扰素信号,诱导免疫抑制性炎性反应,招募抑制性免疫细胞如TAMs、Treg、MDSC等构建肿瘤微环境。微环境中肿瘤细胞迅速生长所致的缺氧通过诱导免疫抑制性炎性反应因子激活肿瘤犬尿氨酸代谢,增强Treg的PD-1表达以加强免疫耐受、抑制CD8+T细胞和NK细胞毒性、促进新血管生成。以上“癌症进化发育学”理论为癌症的防治指明了方向。针对新血管生成的靶向治疗能显著遏制肝癌生长、提高抗肿瘤免疫,加强免疫治疗效果。靶向-免疫联合治疗应成为晚期肝癌的主要治疗措施。

     

    Abstract: Primary liver cancer (PLC) is the first leading cause of immature cancer death in China. Hepatocellular carcinoma (HCC) in China accounts for 93.0% of PLC. Chronic infection with hepatitis B virus (HBV) HCC contributes to 84.4% of HCC. During HBV-induced hepatocarcinogenesis, non-resolving inflammation facilitates viral and host genome mutations via the up-regulated expression of activation-induced cytidine deaminase/APOBEC3s and decreases mutation repair via the down-regulated expression of uracil DNA glycosylase by proinflammatory cytokines such as interleukin-6. The proinflammatory tumor microenvironment (TME) provides necessary conditions for the "mutation-selection-adaptation" evolutionary process of HBV and human hepatocytes and facilitates the retrodifferentiation of mutated hepatic cells. This evolutionary process has two prerequisites: driving somatic mutations and immune-suppressive TME. Cancer-promoting chromosome instability and viral mutations inhibit type I interferon signaling via activating the cyclic GMP-AMP synthase stimulator of interferon genes, induce immune-suppressive inflammation, and recruit immune-suppressive immune cells, including tumor-associated macrophages, regulatory T cells, and myeloid-derived suppressor cells, to build immunosuppressive TME. In the TME, the rapid growth of tumor cell-caused hypoxia activates kynurenine metabolism in the HCC tissues by inducing the expression of immune-suppressive inflammatory factors to promote the expression of PD-1 on regulatory T cells for enhanced immunosuppression; it also inhibits the cytotoxicities of CD8+T and natural killer cells and facilitates angiogenesis. The theoretical update of "Cancer Evo-Dev" highlights the direction of cancer prophylaxis and treatment. The treatments targeting angiogenesis significantly inhibit the growth of HCC, increase anti-tumoral immunity, and enhance the efficacy of immunotherapy. The combination of targeted therapy and immunotherapy should be the major therapeutic option for the treatment of advanced liver cancer.

     

/

返回文章
返回