Skip to main content
Top
Published in: Investigational New Drugs 5/2022

Open Access 21-06-2022 | Respiratory Microbiota | Preclinical Studies

A comprehensive analysis of the microbiota composition and host driver gene mutations in colorectal cancer

Authors: Danping Yuan, Yong Tao, Haoyi Wang, Jiawei Wang, Yuepeng Cao, Wen Cao, Shou Pan, Zhaonan Yu

Published in: Investigational New Drugs | Issue 5/2022

Login to get access

Abstract

Studies of both, microbiota and target therapy associated with gene mutations in colorectal cancer, (CRC) have attracted increasing attention. However, only a few of them analyzed the combined effects on CRC. we analyzed differences in intestinal microbiota of 44 colorectal cancer patients and 20 healthy controls (HC) using 16S rRNA gene sequencing of fecal samples. For 39 of the CRC patients, targeted Next Generation Sequencing (NGS) was carried out at formalin fixed paraffin embedded (FFPE) samples to identify somatic mutation profiles. Compared to the HC group, the microbial diversity of CRC patients was significantly lower. In the CRC group, we found a microbiome that was significantly enriched for strains of Bifidobacterium, Bacteroides, and Megasphaera whereas in the HC group the abundance of Collinsella, Faecalibacterium, and Agathobacter strains was higher. Among the mutations detected in the CRC group, the APC gene had the highest mutation rate (77%, 30/39). We found that the KRAS mutant type was closely associated with Faecalibacterium, Roseburia, Megamonas, Lachnoclostridium, and Harryflintia. Notably, Spearman correlation analysis showed that KRAS mutations were negatively correlated with the existence of Bifidobacterium and positively correlated with Faecalibacterium. By employing 16S rRNA gene sequencing, we identified more unique features of microbiota profiles in CRC patients. For the first time, our study showed that gene mutations could directly be linked to the microbiota composition of CRC patients. We hypothesize that the effect of a targeted colorectal cancer therapy is also closely related to the colorectal flora, however, this requires further investigation.
Literature
1.
go back to reference Datta S, Chowdhury S, Roy HK (2017) Metabolism, microbiome and colorectal cancer. Aging (Albany NY) 9(4):1086CrossRef Datta S, Chowdhury S, Roy HK (2017) Metabolism, microbiome and colorectal cancer. Aging (Albany NY) 9(4):1086CrossRef
2.
go back to reference Huang D, Sun W, Zhou Y et al (2018) Mutations of key driver genes in colorectal cancer progression and metastasis. Cancer Metastasis Rev 37(1):173–187CrossRef Huang D, Sun W, Zhou Y et al (2018) Mutations of key driver genes in colorectal cancer progression and metastasis. Cancer Metastasis Rev 37(1):173–187CrossRef
3.
go back to reference Irrazábal T, Belcheva A, Girardin SE et al (2014) The multifaceted role of the intestinal microbiota in colon cancer. Mol Cell 54(2):309–320CrossRef Irrazábal T, Belcheva A, Girardin SE et al (2014) The multifaceted role of the intestinal microbiota in colon cancer. Mol Cell 54(2):309–320CrossRef
4.
go back to reference Yachida S, Mizutani S, Shiroma H et al (2019) Metagenomic and metabolomic analyses reveal distinct stage-specific phenotypes of the gut microbiota in colorectal cancer. Nat Med 25(6):968–976CrossRef Yachida S, Mizutani S, Shiroma H et al (2019) Metagenomic and metabolomic analyses reveal distinct stage-specific phenotypes of the gut microbiota in colorectal cancer. Nat Med 25(6):968–976CrossRef
5.
go back to reference Thomas AM, Manghi P, Asnicar F et al (2019) Metagenomic analysis of colorectal cancer datasets identifies cross-cohort microbial diagnostic signatures and a link with choline degradation. Nature medicine 25(4):667–678 Thomas AM, Manghi P, Asnicar F et al (2019) Metagenomic analysis of colorectal cancer datasets identifies cross-cohort microbial diagnostic signatures and a link with choline degradation. Nature medicine 25(4):667–678
6.
go back to reference Liang JQ, Li T, Nakatsu G et al (2020) A novel faecal Lachnoclostridium marker for the non-invasive diagnosis of colorectal adenoma and cancer. Gut 69(7):1248–1257CrossRef Liang JQ, Li T, Nakatsu G et al (2020) A novel faecal Lachnoclostridium marker for the non-invasive diagnosis of colorectal adenoma and cancer. Gut 69(7):1248–1257CrossRef
7.
go back to reference Wong SH, Yu J (2019) Gut microbiota in colorectal cancer: mechanisms of action and clinical applications. Nat Rev Gastroenterol Hepatol 16(11):690–704CrossRef Wong SH, Yu J (2019) Gut microbiota in colorectal cancer: mechanisms of action and clinical applications. Nat Rev Gastroenterol Hepatol 16(11):690–704CrossRef
8.
go back to reference Tjalsma H, Boleij A, Marchesi JR et al (2012) A bacterial driver–passenger model for colorectal cancer: beyond the usual suspects. Nat Rev Microbiol 10(8):575–582CrossRef Tjalsma H, Boleij A, Marchesi JR et al (2012) A bacterial driver–passenger model for colorectal cancer: beyond the usual suspects. Nat Rev Microbiol 10(8):575–582CrossRef
9.
go back to reference Rhee KJ, Wu S, Wu XQ et al (2009) Induction of persistent colitis by a human commensal, enterotoxigenic Bacteroides fragilis, in wild-type C57BL/6 mice. Infect Immun 77(4):1708–1718CrossRef Rhee KJ, Wu S, Wu XQ et al (2009) Induction of persistent colitis by a human commensal, enterotoxigenic Bacteroides fragilis, in wild-type C57BL/6 mice. Infect Immun 77(4):1708–1718CrossRef
10.
go back to reference Wang Y, Zhang C, Hou S et al (2020) Analyses of potential driver and passenger bacteria in human colorectal cancer. Cancer Manag Res 12:11553CrossRef Wang Y, Zhang C, Hou S et al (2020) Analyses of potential driver and passenger bacteria in human colorectal cancer. Cancer Manag Res 12:11553CrossRef
11.
go back to reference Li H, Handsaker B, Wysoker A et al (2009) The sequence alignment/map format and SAMtools. Bioinformatics 25(16):2078–2079CrossRef Li H, Handsaker B, Wysoker A et al (2009) The sequence alignment/map format and SAMtools. Bioinformatics 25(16):2078–2079CrossRef
12.
go back to reference Cibulskis K, Lawrence MS, Carter SL et al (2013) Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat Biotechnol 31(3):213–219CrossRef Cibulskis K, Lawrence MS, Carter SL et al (2013) Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat Biotechnol 31(3):213–219CrossRef
13.
go back to reference Bray F, Ferlay J, Soerjomataram I et al (2018) Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 68(6):394–424CrossRef Bray F, Ferlay J, Soerjomataram I et al (2018) Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 68(6):394–424CrossRef
15.
go back to reference Gurjao C, Zhong R, Haruki K et al (2021) Discovery and features of an alkylating signature in colorectal cancer. Cancer Discov 11(10):2446–2455CrossRef Gurjao C, Zhong R, Haruki K et al (2021) Discovery and features of an alkylating signature in colorectal cancer. Cancer Discov 11(10):2446–2455CrossRef
16.
go back to reference Flanagan DJ, Pentinmikko N, Luopajärvi K et al (2021) NOTUM from Apc-mutant cells biases clonal competition to initiate cancer. Nature 594(7863):430–435CrossRef Flanagan DJ, Pentinmikko N, Luopajärvi K et al (2021) NOTUM from Apc-mutant cells biases clonal competition to initiate cancer. Nature 594(7863):430–435CrossRef
17.
go back to reference Brandt R, Sell T, Lüthen M et al (2019) Cell type-dependent differential activation of ERK by oncogenic KRAS in colon cancer and intestinal epithelium. Nat Commun 10(1):2919CrossRef Brandt R, Sell T, Lüthen M et al (2019) Cell type-dependent differential activation of ERK by oncogenic KRAS in colon cancer and intestinal epithelium. Nat Commun 10(1):2919CrossRef
18.
go back to reference Song M, Chan AT (2019) Environmental factors, gut microbiota, and colorectal cancer prevention. Clin Gastroenterol Hepatol 17(2):275–289CrossRef Song M, Chan AT (2019) Environmental factors, gut microbiota, and colorectal cancer prevention. Clin Gastroenterol Hepatol 17(2):275–289CrossRef
19.
go back to reference Dejea CM, Fathi P, Craig JM et al (2018) Patients with familial adenomatous polyposis harbor colonic biofilms containing tumorigenic bacteria. Science 359(6375):592–597CrossRef Dejea CM, Fathi P, Craig JM et al (2018) Patients with familial adenomatous polyposis harbor colonic biofilms containing tumorigenic bacteria. Science 359(6375):592–597CrossRef
20.
go back to reference Wong SH, Yu J (2019) Gut microbiota in colorectal cancer: mechanisms of action and clinical applications. Nat Rev Gastroenterol Hepatol 16(11):690–704CrossRef Wong SH, Yu J (2019) Gut microbiota in colorectal cancer: mechanisms of action and clinical applications. Nat Rev Gastroenterol Hepatol 16(11):690–704CrossRef
21.
go back to reference Wirbel J, Pyl PT, Kartal E et al (2019) Meta-analysis of fecal metagenomes reveals global microbial signatures that are specific for colorectal cancer. Nat Med 25(4):679–689CrossRef Wirbel J, Pyl PT, Kartal E et al (2019) Meta-analysis of fecal metagenomes reveals global microbial signatures that are specific for colorectal cancer. Nat Med 25(4):679–689CrossRef
22.
go back to reference Jin Y, Liu Y, Zhao L et al (2019) Gut microbiota in patients after surgical treatment for colorectal cancer. Environ Microbiol 21(2):772–783CrossRef Jin Y, Liu Y, Zhao L et al (2019) Gut microbiota in patients after surgical treatment for colorectal cancer. Environ Microbiol 21(2):772–783CrossRef
23.
go back to reference Flemer B, Lynch DB, Brown JMR et al (2017) Tumour-associated and non-tumour-associated microbiota in colorectal cancer. Gut 66(4):633–643CrossRef Flemer B, Lynch DB, Brown JMR et al (2017) Tumour-associated and non-tumour-associated microbiota in colorectal cancer. Gut 66(4):633–643CrossRef
24.
go back to reference Russo E, Bacci G, Chiellini C et al (2018) Preliminary comparison of oral and intestinal human microbiota in patients with colorectal cancer: A pilot study. Front Microbiol 8:2699CrossRef Russo E, Bacci G, Chiellini C et al (2018) Preliminary comparison of oral and intestinal human microbiota in patients with colorectal cancer: A pilot study. Front Microbiol 8:2699CrossRef
25.
go back to reference Yeoh YK, Chen Z, Wong MCS et al (2020) Southern Chinese populations harbour non-nucleatum Fusobacteria possessing homologues of the colorectal cancer-associated FadA virulence factor. Gut 69(11):1998–2007CrossRef Yeoh YK, Chen Z, Wong MCS et al (2020) Southern Chinese populations harbour non-nucleatum Fusobacteria possessing homologues of the colorectal cancer-associated FadA virulence factor. Gut 69(11):1998–2007CrossRef
26.
go back to reference Ma H, Yu Y, Wang M et al (2019) Correlation between microbes and colorectal cancer: tumor apoptosis is induced by sitosterols through promoting gut microbiota to produce short-chain fatty acids. Apoptosis 24(1–2):168–183CrossRef Ma H, Yu Y, Wang M et al (2019) Correlation between microbes and colorectal cancer: tumor apoptosis is induced by sitosterols through promoting gut microbiota to produce short-chain fatty acids. Apoptosis 24(1–2):168–183CrossRef
27.
go back to reference Kang X, Zhang R, Kwong TN et al (2021) Serrated neoplasia in the colorectum: gut microbiota and molecular pathways. Gut Microbes 13(1):1–12PubMed Kang X, Zhang R, Kwong TN et al (2021) Serrated neoplasia in the colorectum: gut microbiota and molecular pathways. Gut Microbes 13(1):1–12PubMed
28.
go back to reference Wei Z, Cao S, Liu S et al (2016) Could gut microbiota serve as prognostic biomarker associated with colorectal cancer patients’ survival? A pilot study on relevant mechanism. Oncotarget 7(29):46158–46172CrossRef Wei Z, Cao S, Liu S et al (2016) Could gut microbiota serve as prognostic biomarker associated with colorectal cancer patients’ survival? A pilot study on relevant mechanism. Oncotarget 7(29):46158–46172CrossRef
29.
go back to reference Sokol H, Pigneur B, Watterlot L et al (2008) Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl Acad Sci 105(43):16731–16736CrossRef Sokol H, Pigneur B, Watterlot L et al (2008) Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl Acad Sci 105(43):16731–16736CrossRef
30.
go back to reference Phipps AI, Limburg PJ, Baron JA et al (2015) Association between molecular subtypes of colorectal cancer and patient survival. Gastroenterology 148(1):77–87CrossRef Phipps AI, Limburg PJ, Baron JA et al (2015) Association between molecular subtypes of colorectal cancer and patient survival. Gastroenterology 148(1):77–87CrossRef
31.
go back to reference Jass JR (2007) Classification of colorectal cancer based on correlation of clinical, morphological and molecular features. Histopathology 50(1):113–130CrossRef Jass JR (2007) Classification of colorectal cancer based on correlation of clinical, morphological and molecular features. Histopathology 50(1):113–130CrossRef
32.
go back to reference An JJ, Seok H, Ha EM (2021) GABA-producing Lactobacillus plantarum inhibits metastatic properties and induces apoptosis of 5-FU-resistant colorectal cancer cells via GABA B receptor signaling. J Microbiol 59(2):202–216CrossRef An JJ, Seok H, Ha EM (2021) GABA-producing Lactobacillus plantarum inhibits metastatic properties and induces apoptosis of 5-FU-resistant colorectal cancer cells via GABA B receptor signaling. J Microbiol 59(2):202–216CrossRef
Metadata
Title
A comprehensive analysis of the microbiota composition and host driver gene mutations in colorectal cancer
Authors
Danping Yuan
Yong Tao
Haoyi Wang
Jiawei Wang
Yuepeng Cao
Wen Cao
Shou Pan
Zhaonan Yu
Publication date
21-06-2022
Publisher
Springer US
Published in
Investigational New Drugs / Issue 5/2022
Print ISSN: 0167-6997
Electronic ISSN: 1573-0646
DOI
https://doi.org/10.1007/s10637-022-01263-1

Other articles of this Issue 5/2022

Investigational New Drugs 5/2022 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine