Page 141 - 南京医科大学学报自然科学版
P. 141

第44卷第1期                 夏经纬,沈 波. 肠道微生物在肺癌发生发展及治疗中的研究进展[J].
                  2024年1月                     南京医科大学学报(自然科学版),2024,44(01):130-137                       ·135 ·


                微环境,降低放疗疗效,而共生细菌却有着截然相                           [11] YAMAMURA K,IZUMI D,KANDIMALLA R,et al. In⁃
                反的作用,并且肠道益生菌具有保护肠黏膜,减轻                                 tratumoral Fusobacterium nucleatum levels predict thera⁃
                放疗不良反应的作用;肠道菌群与肺癌靶向治疗相                                 peutic response to neoadjuvant chemotherapy in esopha⁃
                关的研究仍然处于起步阶段;在肺癌免疫治疗方                                  geal squamous cell carcinoma[J]. Clin Cancer Res,
                                                                       2019,25(20):6170-6179
                面,肠道菌群的作用已经得到了广泛认可。目前多
                                                                 [12] GELLER L T,BARZILY⁃ROKNI M,DANINO T,et al.
                项研究表明,肠道微生物的多样性、特定类群及以
                                                                       Potential role of intratumor bacteria in mediating tumor
                SCFA 为主的代谢产物可以通过活化免疫细胞、改
                                                                       resistance to the chemotherapeutic drug gemcitabine[J].
                善肿瘤微环境等方式来影响 ICI 的治疗效果。因
                                                                       Science,2017,357(6356):1156-1160
                此,未来的研究需要更加深入地探讨菌群与肺癌治                           [13] ROUTY B,LE CHATELIER E,DEROSA L,et al. Gut mi⁃
                疗的关系,并开发出更加有效的治疗策略。我们坚                                 crobiome influences efficacy of PD⁃1⁃based immunothera⁃
                信,通过不断地研究和探索,肠道菌群将成为肺癌                                 py against epithelial tumors[J]. Science,2018,359
                治疗中不可或缺的一部分,为肺癌患者带来更好的                                (6371):91-97
                治疗效果和预后。                                         [14] ZHENG Y,FANG Z,XUE Y,et al. Specific gut micro⁃
                                                                       biome signature predicts the early⁃stage lung cancer[J].
               [参考文献]
                                                                       Gut Microbes,2020,11(4):1030-1042
               [1] SIEGEL R L,MILLER K D,WAGLE N S,et al. Cancer  [15] QIN X,BI L,YANG W,et al. Dysbiosis of the gut micro⁃
                    statistics,2023[J]. CA Cancer J Clin,2023,73(1):17-  biome is associated with histopathology of lung cancer
                    48
                                                                      [J]. Front Microbiol,2022,13:918823
               [2] YOSHIDA K,GOWERS K H C,LEE⁃SIX H,et al. Tobacco  [16] LIU F,LI J,GUAN Y,et al. Dysbiosis of the gut microbi⁃
                    smoking and somatic mutations in human bronchial   ome is associated with tumor biomarkers in lung cancer
                    epithelium[J]. Nature,2020,578(7794):266-272      [J]. Int J Biol Sci,2019,15(11):2381-2392
               [3] YANG J J,YU D X,SHU X O. Association of dietary fi⁃  [17] ALEXANDER J L,WILSON I D,TEARE J,et al. Gut mi⁃
                    ber and yogurt consumption with lung cancer risk:a  crobiota modulation of chemotherapy efficacy and toxicity
                    pooled analysis[J]. JAMA Oncol,2020,6(2):e194107  [J]. Nat Rev Gastroenterol Hepatol,2017,14(6):356-
               [4] HE Q Q,QU M Y,SHEN T Y,et al. Suppression of        365
                     VEGFD expression by S⁃nitrosylation promotes the deve⁃  [18] CURRÒ D. The role of gut microbiota in the modulation
                     lopment of lung adenocarcinoma[J]. J Exp Clin Cancer  of drug action:a focus on some clinically significant issues
                     Res,2022,41(1):239                               [J]. Expert Rev Clin Pharmacol,2018,11(2):171-183
               [5] SHREINER A B,KAO J Y,YOUNG V B. The gut micro⁃  [19] KOPPEL N,MAINI REKDAL V,BALSKUS E P. Chemi⁃
                    biome in health and in disease[J]. Curr Opin Gastroenterol,  cal transformation of xenobiotics by the human gut micro⁃

                    2015,31(1):69-75                                   biota[J]. Science,2017,356(6344):eaag2770
               [6] DE MARTEL C,FERLAY J,FRANCESCHI S,et al.      [20] ROUTY B,GOPALAKRISHNAN V,DAILLÈRE R,et al.
                     Global burden of cancers attributable to infections in  The gut microbiota influences anticancer immunosurveil⁃
                     2008:a review and synthetic analysis[J]. Lancet Oncol,  lance and general health[J]. Nat Rev Clin Oncol,2018,
                     2012,13(6):607-615                                15(6):382-396
               [7] FULBRIGHT L E,ELLERMANN M,ARTHUR J C. The     [21] NAJMI M,TRAN T,WITT R G,et al. Modulation of the
                    microbiome and the hallmarks of cancer[J]. PLoS Pat⁃  gut microbiome to enhance immunotherapy response in
                    hog,2017,13(9):e1006480                            metastatic melanoma patients:a clinical review[J]. Der⁃
               [8] LEE M H,LOZANO G. Regulation of the p53 ⁃ MDM2      matol Ther,2022,12(11):2489-2497
                     pathway by 14⁃3⁃3 sigma and other proteins[J]. Semin  [22] MATSON V,FESSLER J,BAO R Y,et al. The commen⁃
                     Cancer Biol,2006,16(3):225-234                    sal microbiome is associated with anti⁃PD⁃ 1 efficacy in
               [9] YANG H Y,WEN Y Y,CHEN C H,et al. 14⁃3⁃3 sigma       metastatic melanoma patients[J]. Science,2018,359
                    positively regulates p53 and suppresses tumor growth[J].  (6371):104-108
                    Mol Cell Biol,2003,23(20):7096-7107          [23] CULLIN N,AZEVEDO ANTUNES C,STRAUSSMAN R,
               [10] LAU H C H,YU J. Gut microbiome alters functions of mu⁃  et al. Microbiome and cancer[J]. Cancer Cell,2021,39
                    tant p53 to promote tumorigenesis[J]. Signal Transduct  (10):1317-1341
                    Target Ther,2020,5(1):232                    [24]DENK D,GRETEN F R. Inflammation:the incubator of
   136   137   138   139   140   141   142   143   144   145   146