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 ›› 2023, Vol. 12 ›› Issue (04) : 331 -336. doi: 10.3877/cma.j.issn.2095-3224.2023.04.010

综述

结直肠癌粪便筛查的现状与展望
鄂一民, 孙司正, 范小彧, 喻春钊()   
  1. 210011 南京医科大学第二附属医院普外科
    210011 南京医科大学第二附属医院普外科;211112 南京医科大学附属逸夫医院普外科
  • 收稿日期:2022-11-25 出版日期:2023-08-25
  • 通信作者: 喻春钊
  • 基金资助:
    国家重点研发计划(2018YFE0127300); 江苏省“333工程”培养资金(BRA2020091); 江苏省科技计划重点项目(BE2019759)

Present status and prospect of fecal screening for colorectal cancer

Yimin E, Sizheng Sun, Xiaoyu Fan, Chunzhao Yu()   

  1. Department of General Surgery, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
    Department of General Surgery, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China; Department of General Surgery, Sir Run Run Hospital Nanjing Medical University, Nanjing 211112, China
  • Received:2022-11-25 Published:2023-08-25
  • Corresponding author: Chunzhao Yu
引用本文:

鄂一民, 孙司正, 范小彧, 喻春钊. 结直肠癌粪便筛查的现状与展望[J]. 中华结直肠疾病电子杂志, 2023, 12(04): 331-336.

Yimin E, Sizheng Sun, Xiaoyu Fan, Chunzhao Yu. Present status and prospect of fecal screening for colorectal cancer[J]. Chinese Journal of Colorectal Diseases(Electronic Edition), 2023, 12(04): 331-336.

结直肠癌是世界范围内常见的消化系统恶性肿瘤之一,常因早期症状不易察觉或不典型而被忽视或延误诊治。近年来,以粪便为标本的非侵入性结直肠癌筛查方法因其简便、无创、经济等因素在基层及临床广泛开展。粪便隐血试验是目前结直肠癌非侵入性筛查中最重要的手段之一,其他粪便筛查方法仍待进一步研究和验证。多种方式的粪便筛查联合检测较单一方法表现出了更好的灵敏度,这提示可以联合多种非侵入性筛查方法构建结直肠癌筛查模型,以精确更适合行结肠镜检查的目标人群。本文就结直肠癌粪便筛查方法的现状及未来进行综述。

Colorectal cancer is one of the most common malignant tumors of the digestive system in the world. Due to the unnoticeable or atypical early symptoms, colorectal cancer is often ignored or delayed in diagnosis and treatment. In recent years, non-invasive colorectal cancer screening methods based on stool specimens have been widely used at the grassroots and clinical level due to their simplicity, non-invasiveness and economic factors. Fecal occult blood test is the most important method for non-invasive screening of colorectal cancer. Other fecal screening methods still need to be further studied and validated. The combination of multiple methods of fecal screening showed better sensitivity than single methods, suggesting that multiple non-invasive screening methods can be combined to construct colorectal cancer screening models that are more accurately suited to the target population undergoing colonoscopy. This article reviews the current status and future of fecal screening methods for colorectal cancer.

[1]
Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries[J]. CA: A Cancer Journal for Clinicians, 2021, 71(3): 209-249.
[2]
Zheng RS, Zhang SW, Zeng HM, et al. Cancer incidence and mortality in China, 2016[J]. J National Cancer Center, 2022, 2(1): 1-9.
[3]
Scheer MGW, Sloots CEJ, van der Wilt GJ, et al. Management of patients with asymptomatic colorectal cancer and synchronous irresectable metastases[J]. Ann Oncol, 2008, 19(11): 1829-1835.
[4]
Ng K, May FP, Schrag D. US preventive services task force recommendations for colorectal cancer screening: forty-five is the new fifty[J]. JAMA-J Am Med Assoc 2021, 325(19): 1943-1945.
[5]
国家癌症中心中国结直肠癌筛查与早诊早治指南制定专家组. 中国结直肠癌筛查与早诊早治指南(2020, 北京)[J]. 中华肿瘤杂志, 2021, 43(1): 16-38.
[6]
马志刚, 朱晓麟, 马丽红, 等. 基于多靶点粪便FIT-DNA联合检测技术的结直肠癌早期筛查结果分析[J/CD]. 中华结直肠疾病电子杂志, 2019, 8(6): 616-621.
[7]
Tinmouth J, Lansdorp-Vogelaar I, Allison JE. Faecal immunochemical tests versus guaiac faecal occult blood tests: what clinicians and colorectal cancer screening programme organisers need to know[J]. Gut, 2015, 64(8): 1327-1337.
[8]
Li JN, Yuan SY. Fecal occult blood test in colorectal cancer screening[J]. J Dig Dis, 2019, 20(2): 62-64.
[9]
Clark G, Strachan JA, Carey FA, et al. Transition to quantitative faecal immunochemical testing from guaiac faecal occult blood testing in a fully rolled-out population-based national bowel screening programme[J]. Gut, 2021, 70(1): 106-113.
[10]
Zackular JP, Rogers MAM, Ruffin MT, et al. The human gut microbiome as a screening tool for colorectal cancer[J]. Cancer Prev Res, 2014, 7(11): 1112-1121.
[11]
Zou J, Xiao Z, Wu Y, et al. Noninvasive fecal testing for colorectal cancer[J]. Clin Chim Acta, 2022, 524: 123-131.
[12]
Peng SM, Hsu WF, Wang YW, et al. Faecal immunochemical test after negative colonoscopy may reduce the risk of incident colorectal cancer in a population-based screening programme[J]. Gut, 2021, 70(7): 1318-1324.
[13]
Nicholson BD, James T, Paddon M, et al. Faecal immunochemical testing for adults with symptoms of colorectal cancer attending English primary care: a retrospective cohort study of 14 487 consecutive test requests[J]. Aliment Pharmacol Ther, 2020, 52(6): 1031-1041.
[14]
马晨曦, 关旭, 王松, 等. 粪便DNA检测技术在结直肠癌筛查中的应用现状及展望[J]. 中华胃肠外科杂志, 2019, 22(5): 491-494.
[15]
Carethers JM. Fecal DNA testing for colorectal cancer screening[J]. Annu Rev Med, 2020, 71: 59-69.
[16]
Sidransky D, Tokino T, Hamilton SR, et al. Identification of ras oncogene mutations in the stool of patients with curable colorectal tumors[J]. Science (New York, NY), 1992, 256(5053): 102-105.
[17]
Ahlquist DA, Skoletsky JE, Boynton KA, et al. Colorectal cancer screening by detection of altered human DNA in stool: Feasibility of a multitarget assay panel[J]. Gastroenterology, 2000, 119(5): 1219-1227.
[18]
Imperiale TF, Ransohoff DF, Itzkowitz SH, et al. Multitarget stool DNA testing for colorectal-cancer screening[J]. N Engl J Med, 2014, 370(14): 1287-1297.
[19]
Redwood DG, Asay ED, Blake ID, et al. Stool DNA testing for screening detection of colorectal neoplasia in alaska native people[J]. Mayo Clin Proc, 2016, 91(1): 61-70.
[20]
Redwood DG, Dinh TA, Kisiel JB, et al. Cost-effectiveness of multitarget stool dna testing vs colonoscopy or fecal immunochemical testing for colorectal cancer screening in alaska native people[J]. Mayo Clin Proc, 2021, 96(5): 1203-1217.
[21]
Ladabaum U, Mannalithara A. Comparative effectiveness and cost effectiveness of a multitarget stool DNA test to screen for colorectal neoplasia[J]. Gastroenterology, 2016, 151(3): 427-439.
[22]
Yang TW, Lee WH, Tu SJ, et al. Enterotype-based analysis of gut microbiota along the conventional adenoma-carcinoma colorectal cancer pathway[J]. Sci Rep, 2019, 9(1): 10923.
[23]
Yachida S, Mizutani S, Shiroma H, et al. Metagenomic and metabolomic analyses reveal distinct stage-specific phenotypes of the gut microbiota in colorectal cancer[J]. Nat Med, 2019, 25(6): 968-976.
[24]
Wong SH, Kwong TNY, Chow TC, et al. Quantitation of faecal fusobacterium improves faecal immunochemical test in detecting advanced colorectal neoplasia[J]. Gut, 2017, 66(8): 1441-1448.
[25]
Wong SH, Yu J. Gut microbiota in colorectal cancer: mechanisms of action and clinical applications[J]. Nat Rev Gastroenterol Hepatol, 2019, 16(11): 690-704.
[26]
Wu YQ, Jiao N, Zhu RX, et al. Identification of microbial markers across populations in early detection of colorectal cancer[J]. Nature Communications, 2021, 12(1): 13.
[27]
Coker OO, Liu C, Wu WKK, et al. Altered gut metabolites and microbiota interactions are implicated in colorectal carcinogenesis and can be non-invasive diagnostic biomarkers[J]. Microbiome, 2022, 10(1): 12.
[28]
Jain S, Maque J, Galoosian A, et al. Optimal strategies for colorectal cancer screening[J]. Curr Treat Options Oncol, 2022, 23(4): 474-493.
[29]
Slaby O. Non-coding RNAs as biomarkers for colorectal cancer screening and early detection[J]. Adv Exp Med Biol, 2016, 937: 153-170.
[30]
Wu CW, Ng SC, Dong YJ, et al. Identification of microRNA-135b in stool as a potential noninvasive biomarker for colorectal cancer and adenoma[J]. Clin Cancer Res, 2014, 20(11): 2994-3002.
[31]
Duran-Sanchon S, Moreno L, Auge JM, et al. Identification and validation of microRNA profiles in fecal samples for detection of colorectal cancer[J]. Gastroenterology, 2020, 158(4): 947-957.
[32]
Uppara M, Adaba F, Askari A, et al. A systematic review and meta-analysis of the diagnostic accuracy of pyruvate kinase M2 isoenzymatic assay in diagnosing colorectal cancer[J]. World J Surg Oncol, 2015, 13: 48.
[33]
Lue A, Hijos G, Sostres C, et al. The combination of quantitative faecal occult blood test and faecal calprotectin is a cost-effective strategy to avoid colonoscopies in symptomatic patients without relevant pathology[J]. Ther Adv Gastroenterol, 2020, 13: 1756284820920786.
[34]
Jukic A, Bakiri L, Wagner EF, et al. Calprotectin: from biomarker to biological function[J]. Gut, 2021, 70(10): 1978-1988.
[35]
Komor MA, Bosch LJW, Coupe VMH, et al. Proteins in stool as biomarkers for non-invasive detection of colorectal adenomas with high risk of progression[J]. J Pathol, 2020, 250(3): 288-298.
[36]
Shah MS, DeSantis TZ, Weinmaier T, et al. Leveraging sequence-based faecal microbial community survey data to identify a composite biomarker for colorectal cancer[J]. Gut, 2018, 67(5): 882-891.
[37]
Liang JQ, Li T, Nakatsu G, et al. A novel faecal Lachnoclostridium marker for the non-invasive diagnosis of colorectal adenoma and cancer[J]. Gut, 2020, 69(7): 1248-1257.
[38]
Koga Y, Yamazaki N, Yamamoto Y, et al. Fecal miR-106a is a useful marker for colorectal cancer patients with false-negative results in immunochemical fecal occult blood test[J]. Cancer Epidemiol Biomarkers Prev, 2013, 22(10): 1844-1852.
[39]
Duran-Sanchon S, Moreno L, Gomez-Matas J, et al. Fecal microRNA-based algorithm increases effectiveness of fecal immunochemical test-based screening for colorectal cancer[J]. Clin Gastroenterol Hepatol, 2021, 19(2): 323-330.
[40]
Wong MCS, Huang JJ, Wong YY, et al. The use of a non-invasive biomarker for colorectal cancer screening: a comparative cost-effectiveness modeling study[J]. Cancers, 2023, 15(3): 14.
[41]
Zhou H, Zhu LY, Song J, et al. Liquid biopsy at the frontier of detection, prognosis and progression monitoring in colorectal cancer[J]. Molecular Cancer, 2022, 21(1): 21.
[42]
Zhou BT, Xu KL, Zheng X, et al. Application of exosomes as liquid biopsy in clinical diagnosis[J]. Signal Transduct Target Ther, 2020, 5(1): 14.
[43]
Young GP, Symonds EL, Allison JE, et al. Advances in fecal occult blood tests: the FIT revolution[J]. Dig Dis Sci, 2015, 60(3): 609-622.
[44]
Nikolaou S, Qiu SY, Fiorentino F, et al. Systematic review of blood diagnostic markers in colorectal cancer[J]. Tech Coloproctology, 2018, 22(7): 481-498.
[45]
Marcuello M, Duran-Sanchon S, Moreno L, et al. Analysis of A 6-mirna signature in serum from colorectal cancer screening participants as non-invasive biomarkers for advanced adenoma and colorectal cancer detection[J]. Cancers, 2019, 11(10): 12.
[46]
Gao HL, Lv LB, Zhao WF, et al. Diagnostic accuracy of the multi-target stool DNA test in detecting colorectal cancer: A hospital-based study[J]. World J Gastrointest Oncol, 2023, 15(1): 102-111.
[47]
Zhong GC, Sun WP, Wan L, et al. Efficacy and cost-effectiveness of fecal immunochemical test versus colonoscopy in colorectal cancer screening: a systematic review and meta-analysis[J]. Gastrointestinal Endoscopy, 2020, 91(3): 684-697.
[48]
Peterse EFP, Meester RGS, de Jonge L, et al. Comparing the cost-effectiveness of innovative colorectal cancer screening tests[J]. JNCI-J Natl Cancer Inst, 2021, 113(2): 154-161.
[49]
Cruz A, Carvalho CM, Cunha A, et al. Faecal diagnostic biomarkers for colorectal cancer[J]. Cancers, 2021, 13(21): 10.
[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] 付佳, 肖海敏, 武曦, 冯涛, 师帅. 年龄校正查尔森合并症指数对腹腔镜结直肠癌围手术期并发症的预测价值[J]. 中华普通外科学文献(电子版), 2023, 17(05): 336-341.
[5] 薛永婷, 高峰, 王雅楠, 屈莲平. 溶瘤病毒治疗在结直肠癌中的应用[J]. 中华普通外科学文献(电子版), 2023, 17(05): 380-384.
[6] 武慧铭, 郭仁凯, 李辉宇. 机器人辅助下经自然腔道取标本手术治疗结直肠癌安全性和有效性的Meta分析[J]. 中华普通外科学文献(电子版), 2023, 17(05): 395-400.
[7] 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.
[8] 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.
[9] 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.
[10] 常剑, 邱峰, 毛郁琪. 摄食抑制因子-1与腹腔镜结直肠癌根治术后肝转移的关系分析[J]. 中华普外科手术学杂志(电子版), 2023, 17(05): 502-505.
[11] 王晓燕, 肖佑, 肖戈, 王真权. 老年结直肠癌肺转移CT特征及高危因素研究[J]. 中华普外科手术学杂志(电子版), 2023, 17(05): 506-509.
[12] 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.
[13] 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.
[14] 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.
[15] 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.
阅读次数
全文


摘要