切换至 "中华医学电子期刊资源库"

中华结直肠疾病电子杂志 ›› 2021, Vol. 10 ›› Issue (03) : 302 -305. doi: 10.3877/cma.j.issn.2095-3224.2021.03.014

综述

结直肠癌类器官生物样本库的建立和应用研究进展
王一1, 吴小倩2, 黄伟芳2, 裴斌2, 尚芳2, 孔德松3, 王小峰1, 朱勇1, 姚航1, 刘飞1, 樊志敏1,()   
  1. 1. 210001 南京中医药大学附属南京中医院肛肠科
    2. 南京中医药现代化与大数据研究中心;210029 南京中医药大学,医学院·整合医学学院
    3. 南京中医药现代化与大数据研究中心
  • 收稿日期:2020-08-17 出版日期:2021-06-25
  • 通信作者: 樊志敏
  • 基金资助:
    南京市部省共建临床医学研究中心培育计划(GCYJZX-2019); 南京市卫健委重点项目(ZKX19034)

Research progress on establishment and application of organoid biobank of colorectal cancer patients

Yi Wang1, Xiaoqian Wu2, Weifang Huang2, Bin Pei2, Fang Shang2, Desong Kong3, Xiaofeng Wang1, Yong Zhu1, Hang Yao1, Fei Liu1, Zhimin Fan1,()   

  1. 1. Colon and Rectal Surgery, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210001, China
    2. Chinese Medicine Modernization and Big Data Research Center, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210001, China; School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210029, China
    3. Chinese Medicine Modernization and Big Data Research Center, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210001, China
  • Received:2020-08-17 Published:2021-06-25
  • Corresponding author: Zhimin Fan
引用本文:

王一, 吴小倩, 黄伟芳, 裴斌, 尚芳, 孔德松, 王小峰, 朱勇, 姚航, 刘飞, 樊志敏. 结直肠癌类器官生物样本库的建立和应用研究进展[J]. 中华结直肠疾病电子杂志, 2021, 10(03): 302-305.

Yi Wang, Xiaoqian Wu, Weifang Huang, Bin Pei, Fang Shang, Desong Kong, Xiaofeng Wang, Yong Zhu, Hang Yao, Fei Liu, Zhimin Fan. Research progress on establishment and application of organoid biobank of colorectal cancer patients[J]. Chinese Journal of Colorectal Diseases(Electronic Edition), 2021, 10(03): 302-305.

结直肠癌是世界上最常见的恶性肿瘤之一,我国结直肠癌的发病率和死亡率仍处于上升趋势。以往结直肠癌的研究主要依靠细胞系和异种移植模型,但二者都有一定的局限性,且难以建立大规模生物样本库。类器官模型是一种新兴的体外模型,具有构建周期较短、易于保存以及保留供体异质性的特点,在结直肠癌的基础研究、新药开发、个体化治疗等方面都极具应用价值。

Colorectal cancer is one of the most common malignant tumors in the world. The incidence rate and mortality rate of colorectal cancer in China are still on the rise. In the past, research on colorectal cancer mainly relied on cell lines and xenograft models, but both have certain limitations and it is difficult to establish a large-scale bio bank. Organoid model is a new in vitro model, which has the characteristics of short construction period, easy storage and good preservation of donor heterogeneity. It has great application value in the research of colorectal cancer occurrence and development mechanism, new drug development, individual treatment and so on.

表1 目前结直肠癌类器官生物样本库建设情况
参考文献 样本类型及数量 培养成功率 组织形态学比较 基因组学研究 主要结论
Wetering et al., 201517

结直肠癌22例

配对正常肠组织19例

≈90% 与供体组织类似,“囊性”或“实心”样的特征得以保留 与供体组织拷贝数变异、突变谱相一致 证实类器官样本库可用于高通量药物筛选
Fujii et al., 201618

结直肠肿瘤55例

配对正常肠组织41例

100% 保留了供体肿瘤的组织病理学结构 能够反应供体组织的基因特性,并准确判断亚型 肿瘤发生过程中对干细胞微环境相关因子依赖性逐渐降低
Schutte et al., 201719 结直肠癌35例 ≈60% N/A 能一定程度地概括供体组织的基因和转录组特征 类器官样本库可用于探索新的生物标记物以指导临床的精准用药
Vlachogiannis et al., 201820 结直肠癌肝转移24例 70% 与供体肿瘤有着显著的组织病理学相似性 与供体组织的突变谱有96%的重合 类器官对靶向药物和化疗药有效性的预测有着88%的准确性
Ganesh et al., 201921

直肠癌65例

配对正常肠组织51例

77% 保留了供体肿瘤的组织病理学特征

与供体组织的突变谱相

一致

类器官可以用于化疗药物敏感性检测,实现个体化治疗
Ooft et al., 201922 结直肠癌40例 63% N/A N/A 临床上运用类器官对伊立替康的疗效进行预测是可行的
Yao et al., 202023 直肠癌80例 85.7% 与供体肿瘤的组织病理学结构相一致 与供体组织基因组特征相一致 类器官能够预测局部晚期直肠癌对放化疗的反应
Yan et al., 202024

结直肠癌20例

配对正常肠组织29例

≈82% N/A 结直肠癌类器官在长期培养过程中能够保持基因组稳定性 类器官相较传统的体外模型能够更好地揭示结直肠癌的基因组特征
1
Bray F, Ferlay J, Soerjomataram L, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries[J]. CA: A Cancer Journal for Clinicians, 2018, 68(6): 394-424.
2
Zhu J, Tan Z, Hollis-Hansen K, et al. Epidemiological trends in colorectal cancer in China: an ecological Study[J]. Dig Dis, 2017, 62(1): 235-243.
3
Arnold M, Sierra MS, Laversanne M, et al. Global patterns and trends in colorectal cancer incidence and mortality[J]. Gut, 2017, 66(4): 683-691.
4
Ganesh K, Stadler K, Cercek A, et al. Immunotherapy in colorectal cancer: rationale, challenges and potential[J]. Nature Reviews Gastroenterology & Hepatology, 2019, 16(6): 361-375.
5
Boland P, Ma W. Immunotherapy for colorectal cancer[J]. Cancers, 2017, 9(5): 50.
6
Sato T, Vrise R, Snippert H, et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche[J]. Nature, 2009, 459(7244): 262-265.
7
Lancaster M, Knoblich J. Organogenesis in a dish: Modeling development and disease using organoid technologies[J]. Science, 2014, 345(6194): 1247125.
8
Hunh M, Dorrell C, Boj S, et al. In vitro expansion of single Lgr5+ liver stem cells induced by Wnt-driven regeneration[J]. Nature, 2013, 494(7436): 247-250.
9
Barker N, Huch M, Kujala P, et al. Lgr5(+ve) stem cells drive self-renewal in the stomach and build long-lived gastric units in vitro[J]. Cell Stem Cell, 2010, 6(1): 25-36.
10
Lancater M, Renner M, Martin C, et al. Cerebral organoids model human brain development and microcephaly[J]. Nature, 2013, 501(7467): 373-379.
11
Xia Y, Nivet E, Martinez L, et al. Directed differentiation of human pluripotent cells to ureteric bud kidney progenitor-like cells[J]. Nat Cell Biol, 2013, 515(12): 1507-1515.
12
Nakano T, Ando S, Tkata N, et al. Self-formation of optic cups and storable stratified neural retina from human ESCs[J]. Cell Stem Cell, 2012, 10(6): 771-785.
13
Levy E, Delvin E, Menard D, et al. Functional development of human fetal gastrointestinal tract[J]. Methods Mol Biol, 2009, 550: 205-224.
14
Crespo M, Vilar E, Tsai S, et al. Colonic organoids derived from human induced pluripotent stem cells for modeling colorectal cancer and drug testing[J]. Nat Med, 2017, 23(7): 878-884.
15
Barretina J, Caponigro G, Stransky N, et al. The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensi- tivity[J]. Nature, 2012, 483(7391):603-607.
16
Jin K, Teng L, Shen Y, et al. Patient-derived human tumour tissue xenografts in immunodeficient mice: a systematic review[J]. Clin Transl Oncol, 2010, 12(7): 473-480.
17
Wetering M, Francies H, Francies J, et al. Prospective Derivation of a Living Organoid Biobank of Colorectal Cancer Patients[J]. Cell, 2015, 161(4): 933-945.
18
Fujii M, Shimokawa M, Date S, et al. A colorectal tumor organoid library demonstrates progressive loss of niche factor requirements during Tumorigenesis[J]. Cell Stem Cell, 2016, 18(6): 827-838.
19
Schutte M, Risch T, Azar N, et al. Molecular dissection of colorectal cancer in pre-clinical models identifies biomarkers predicting sensitivity to EGFR inhibitors[J]. Nat Commun, 2017, 8: 14262.
20
Vlachogiannis G, Hedayat S, al Vatsiouet A. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers[J]. Science, 2018, 359(6378): 920-926.
21
Ganesh K, Wu C, O'Rourke KP, et al. A rectal cancer organoid platform to study individual responses to chemoradiation[J]. Nature Medicine, 2019, 25(10):1607-1614.
22
Ooft S, Weeber F, Dijkstra K, et al. Patient-derived organoids can predict response to chemotherapy in metastatic colorectal cancer patients[J]. Sci Transl Med, 2019, 11(513):eaay2574.
23
Yao Y, Xu X, Yang L, et al. Patient-derived organoids predict chemoradiation responses of locally advanced rectal cancer[J]. Cell Stem Cell, 2020, 26(1): 17-26.
24
Yan H, Siu H, Ho S, et al. Organoid cultures of early-onset colorectal cancers reveal distinct and rare genetic profiles[J]. Cut, 2020, 0: 1-15.
25
Weeber F, Ooft S, Dijkstra J, et al. Tumor organoids as a pre-clinical cancer model for drug discovery[J]. Cell Chem Biol, 2017, 24(9): 1092-1100.
26
Narasimhan V, Wright J, Churchill M, et al. Medium-throughput drug screening of patient-derived organoids from colorectal peritoneal metastases to direct personalized therapy[J]. Clin Cancer Res, 2020, 26(14): 3662-3670.
27
Drost J, Jaarsveld R, Ponsioen B, et al. Sequential cancer mutations in cultured human intestinal stem cells[J]. Nature, 2015, 521(7550): 43-47.
28
Takeda H, Kataoka S, Nakayama M, et al. CRISPR-Cas9-mediated gene knockout in intestinal tumor organoids provides functional validation for colorectal cancer driver genes[J]. Proc Natl Acad Sci USA, 2019, 116(31):15635-15644.
[1] 康夏, 田浩, 钱进, 高源, 缪洪明, 齐晓伟. 骨织素抑制破骨细胞分化改善肿瘤骨转移中骨溶解的机制研究[J]. 中华乳腺病杂志(电子版), 2023, 17(06): 329-339.
[2] 代莉, 邓恢伟, 郭华静, 黄芙蓉. 术中持续输注艾司氯胺酮对腹腔镜结直肠癌手术患者术后睡眠质量的影响[J]. 中华普通外科学文献(电子版), 2023, 17(06): 408-412.
[3] 王得晨, 杨康, 杨自杰, 归明彬, 屈莲平, 张小凤, 高峰. 结直肠癌微卫星稳定状态和程序性死亡、吲哚胺2,3-双加氧酶关系的研究进展[J]. 中华普通外科学文献(电子版), 2023, 17(06): 462-465.
[4] 唐旭, 韩冰, 刘威, 陈茹星. 结直肠癌根治术后隐匿性肝转移危险因素分析及预测模型构建[J]. 中华普外科手术学杂志(电子版), 2024, 18(01): 16-20.
[5] 张生军, 赵阿静, 李守博, 郝祥宏, 刘敏丽. 高糖通过HGF/c-met通路促进结直肠癌侵袭和迁移的实验研究[J]. 中华普外科手术学杂志(电子版), 2024, 18(01): 21-24.
[6] 李婷, 张琳. 血清脂肪酸代谢物及维生素D水平与结直肠癌发生的关系研究[J]. 中华普外科手术学杂志(电子版), 2023, 17(06): 661-665.
[7] 倪文凯, 齐翀, 许小丹, 周燮程, 殷庆章, 蔡元坤. 结直肠癌患者术后发生延迟性肠麻痹的影响因素分析[J]. 中华结直肠疾病电子杂志, 2023, 12(06): 484-489.
[8] 范小彧, 孙司正, 鄂一民, 喻春钊. 梗阻性左半结肠癌不同手术治疗方案的选择应用[J]. 中华结直肠疾病电子杂志, 2023, 12(06): 500-504.
[9] 蓝冰, 王怀明, 王辉, 马波. 局部晚期结肠癌膀胱浸润的研究进展[J]. 中华结直肠疾病电子杂志, 2023, 12(06): 505-511.
[10] 关旭, 王锡山. 基于外科与免疫视角思考结直肠癌区域淋巴结处理的功与过[J]. 中华结直肠疾病电子杂志, 2023, 12(06): 448-452.
[11] 顾睿祈, 方洪生, 蔡国响. 循环肿瘤DNA检测在结直肠癌诊治中的应用与进展[J]. 中华结直肠疾病电子杂志, 2023, 12(06): 453-459.
[12] 黄怡诚, 陆晨, 孙司正, 喻春钊. 肝特异性转录因子FOXA2在人结直肠癌肝转移阶梯模型中的表达变化及其意义[J]. 中华结直肠疾病电子杂志, 2023, 12(05): 396-403.
[13] 刘祺, 张凯, 李建男, 刘铜军. 结直肠癌肝转移生物治疗的现状及进展[J]. 中华结直肠疾病电子杂志, 2023, 12(05): 415-419.
[14] 吴寅, 陈智琴, 高勇, 权明. Her-2阳性结直肠癌的诊治进展[J]. 中华结直肠疾病电子杂志, 2023, 12(05): 420-425.
[15] 孙昕, 程海波, 沈卫星. 基于全转录组学探讨仙连解毒方治疗Ⅲ期结直肠癌患者的疗效机制[J]. 中华消化病与影像杂志(电子版), 2023, 13(05): 277-283.
阅读次数
全文


摘要