您的位置:医药网首页 > 医药资讯 > 医疗器械行业 > 《JAMA·肿瘤学》:全方位参与癌症叙事 肠道微生物有望重塑癌症治疗范式!

《JAMA·肿瘤学》:全方位参与癌症叙事 肠道微生物有望重塑癌症治疗范式!

目前正在进行多项临床试验,涵盖了我们前面介绍的各个方向,可能会彻底改变未来癌症治疗的范式。

然而,靶向肠道微生物疗法的应用仍存在许多局限和不足。

首先,研究结果的异质性可能是由于测定微生物组成时取样样品和分析方法的不一致造成的。

其次,即使在健康人群中,肠道微生物的组成也存在很大差异。此外,我们目前对许多肠道微生物的功能缺乏了解,无法定义理想的菌群。因此,癌症患者如果想用益生菌治疗,需要谨慎。

再次,人的肠道微生物组成更加复杂,从动物研究到临床研究的转化不稳定。例如,在临床试验中,补充鼠李糖乳杆菌GG未能显著改善HSCT后患者的肠道微生物组成,与对照组相比,发生移植物抗宿主病的比例更高[26]。

另外,就像我们之前对抗生素的关注一样,饮食作为一个对肠道微生物组成也有很大影响的因素,也需要给予更多的关注。因为肿瘤也需要来自饮食的营养,所以饮食辅助治疗也是研究方向之一,比如热量限制、生酮饮食,或者限制氨基酸含量的饮食等。饮食不仅影响肿瘤,还影响肠道微生物组成。这些影响是重叠的还是冲突的?如何更有效的调节肠道微生物和抗肿瘤效果?这些都是亟待解决的重要问题。

众所周知,癌症的治疗不是单一疗法就能成功的,而是需要多方面的综合治疗。因此,仍有许多可能性值得探索,例如,将手术、免疫治疗与饮食和益生菌/代谢物治疗结合起来,成为一种新的治疗范式,可以增强抗肿瘤效果,降低全身毒性,提高癌症的全程管理。

参考资料:

[1]刘L,Shah K .肠道微生物群重塑肿瘤治疗典范的潜力: A综述[J].JAMA肿瘤学,2022。

[2] Murata-Kamiya N,Kurashima Y,Teishikata Y,等.幽门螺杆菌CagA与E-cadherin相互作用并解除对促进胃上皮细胞肠转分化的-catenin信号的调控[J].癌基因,2007,26(32): 4617-4626。

[3]刘志军,蒋岩,等.人肠道细菌基因毒素大肠杆菌烷化DNA的研究[J].理科,2019,363(6428): eaar7785。

[4] Drewes J L,Chen J,马卡姆N O,等.人结肠癌来源的艰难梭菌菌株驱动小鼠结肠肿瘤的发生[J].癌症发现,2022: candisc。1273.2021-10-4 15: 58: 09.930.

[5] Goodwin A C,Shields C E D,Wu S,等.多胺分解代谢促进产毒性脆弱类杆菌诱导的结肠肿瘤发生[J].美国国家科学院学报,2011,108(37): 15354-15359。

[6] Bergounioux J,Elisee R,Prunier A L,等.福氏志贺氏菌效应病毒激活钙蛋白酶调节细菌上皮小生境形成和寿命的关键步骤[J].细胞宿主微生物,2012,11(3): 240-252。

[7] Gur C,Ibrahim Y,Isaacson B,等.具核梭杆菌Fap2蛋白与人抑制性受体TIGIT的结合保护肿瘤免受免疫细胞攻击[J].豁免权,2015,42(2): 344-355。

[8]科斯蒂克公元,春娥,袍

rtson L, et al. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment[J]. Cell host microbe, 2013, 14(2): 207-215.

[9] V tizou M, Pitt J M, Daill re R, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota[J]. Science, 2015, 350(6264): 1079-1084.

[10] Sivan A, Corrales L, Hubert N, et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti PD-L1 efficacy[J]. Science, 2015, 350(6264): 1084-1089.

[11] Si W, Liang H, Bugno J, et al. Lactobacillus rhamnosus GG induces cGAS/STING-dependent type I interferon and improves response to immune checkpoint blockade[J]. Gut, 2022, 71(3): 521-533.

[12] Routy B, Le Chatelier E, Derosa L, et al. Gut microbiome influences efficacy of PD-1 based immunotherapy against epithelial tumors[J]. Science, 2018, 359(6371): 91-97.

[13] Derosa L, Hellmann M D, Spaziano M, et al. Negative association of antibiotics on clinical activity of immune checkpoint inhibitors in patients with advanced renal cell and non-small-cell lung cancer[J]. Annals of Oncology, 2018, 29(6): 1437-1444.

[14] Viaud S, Saccheri F, Mignot G, et al. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide[J]. science, 2013, 342(6161): 971-976.

[15] Daill re R, V tizou M, Waldschmitt N, et al. Enterococcus hirae and Barnesiella intestinihominis facilitate cyclophosphamide-induced therapeutic immunomodulatory effects[J]. Immunity, 2016, 45(4): 931-943.

[16] Yu T C, Guo F, Yu Y, et al. Fusobacterium nucleatum promotes chemoresistance to colorectal cancer by modulating autophagy[J]. Cell, 2017, 170(3): 548-563. e16.

[17] Su W, Chen Y, Cao P, et al. Fusobacterium nucleatum promotes the development of ulcerative colitis by inducing the autophagic cell death of intestinal epithelial[J]. Frontiers in cellular and infection microbiology, 2020, 10: 594806.

[18] Liu Y, Baba Y, Ishimoto T, et al. Fusobacterium nucleatum confers chemoresistance by modulating autophagy in oesophageal squamous cell carcinoma[J]. British journal of cancer, 2021, 124(5): 963-974.

[19] Wallace B D, Wang H, Lane K T, et al. Alleviating cancer drug toxicity by inhibiting a bacterial enzyme[J]. Science, 2010, 330(6005): 831-835.

[20] Taur Y, Jenq R R, Perales M A, et al. The effects of intestinal tract bacterial diversity on mortality following allogeneic hematopoietic stem cell transplantation[J]. Blood, The Journal of the American Society of Hematology, 2014, 124(7): 1174-1182.

[21] Jenq R R, Taur Y, Devlin S M, et al. Intestinal Blautia is associated with reduced death from graft-versus-host disease[J]. Biology of Blood and Marrow Transplantation, 2015, 21(8): 1373-1383.

[22] Peled J U, Devlin S M, Staffas A, et al. Intestinal microbiota and relapse after hematopoietic-cell transplantation[J]. Journal of Clinical Oncology, 2017, 35(15): 1650.

[23] Wang Q, Fu Y W, Wang Y Q, et al. Fecal microbiota transplantation for patients with refractory diarrhea after allogeneic hematopoietic stem cell transplantation[J]. Zhonghua xue ye xue za zhi= Zhonghua Xueyexue Zazhi, 2019, 40(10): 853-855.

[24] van Praagh J B, de Goffau M C, Bakker I S, et al. Mucus microbiome of anastomotic tissue during surgery has predictive value for colorectal anastomotic leakage[J]. Annals of Surgery, 2019, 269(5): 911-916.

[25] Valdez J C, Peral M C, Rachid M, et al. Interference of Lactobacillus plantarum with Pseudomonas aeruginosa in vitro and in infected burns: the potential use of probiotics in wound treatment[J]. Clinical microbiology and infection, 2005, 11(6): 472-479.

[26] Gorshein E, Wei C, Ambrosy S, et al. Lactobacillus rhamnosus GG probiotic enteric regimen does not appreciably alter the gut microbiome or provide protection against GVHD after allogeneic hematopoietic stem cell transplantation[J]. Clinical Transplantation, 2017, 31(5): e12947.

医药网新闻
返回顶部】【打印】【关闭
扫描100医药网微信二维码
视频新闻
图片新闻
医药网免责声明:
  • 本公司对医药网上刊登之所有信息不声明或保证其内容之正确性或可靠性;您于此接受并承认信赖任何信息所生之风险应自行承担。本公司,有权但无此义务,改善或更正所刊登信息任何部分之错误或疏失。
  • 凡本网注明"来源:XXX(非医药网)"的作品,均转载自其它媒体,转载目的在于传递更多信息,并不代表本网赞同其观点和对其真实性负责。本网转载其他媒体之稿件,意在为公众提供免费服务。如稿件版权单位或个人不想在本网发布,可与本网联系,本网视情况可立即将其撤除。联系QQ:896150040