COL25A1 and METAP1D DNA methylation are promising liquid biopsy epigenetic biomarkers of colorectal cancer using digital PCR.


Journal

Clinical epigenetics
ISSN: 1868-7083
Titre abrégé: Clin Epigenetics
Pays: Germany
ID NLM: 101516977

Informations de publication

Date de publication:
18 Oct 2024
Historique:
received: 08 07 2024
accepted: 16 09 2024
medline: 19 10 2024
pubmed: 19 10 2024
entrez: 18 10 2024
Statut: epublish

Résumé

Colorectal cancer is a public health issue and was the third leading cause of cancer-related death worldwide in 2022. Early diagnosis can improve prognosis, making screening a central part of colorectal cancer management. Blood-based screening, diagnosis and follow-up of colorectal cancer patients are possible with the study of cell-free circulating tumor DNA. This study aimed to identify novel DNA methylation biomarkers of colorectal cancer that can be used for the follow-up of patients with colorectal cancer. A DNA methylation profile was established in the Gene Expression Omnibus (GEO) database (n = 507) using bioinformatics analysis and subsequently confirmed using The Cancer Genome Atlas (TCGA) database (n = 348). The in silico profile was then validated on local tissue and cell-free DNA samples using methylation-specific digital PCR in colorectal cancer patients (n = 35) and healthy donors (n = 35). The DNA methylation of COL25A1 and METAP1D was predicted to be a colorectal cancer biomarker by bioinformatics analysis (ROC AUC = 1, 95% CI [0.999-1]). The two biomarkers were confirmed with tissue samples, and the combination of COL25A1 and METAP1D yielded 49% sensitivity and 100% specificity for cell-free DNA. Bioinformatics analysis of public databases revealed COL25A1 and METAP1D DNA methylation as clinically applicable liquid biopsies DNA methylation biomarkers. The specificity implies an excellent positive predictive value for follow-up, and the high sensitivity and relative noninvasiveness of a blood-based test make these biomarkers compatible with colorectal cancer screening. However, the clinical impact of these biomarkers in colorectal cancer screening and follow-up needs to be established in further prospective studies.

Sections du résumé

BACKGROUND BACKGROUND
Colorectal cancer is a public health issue and was the third leading cause of cancer-related death worldwide in 2022. Early diagnosis can improve prognosis, making screening a central part of colorectal cancer management. Blood-based screening, diagnosis and follow-up of colorectal cancer patients are possible with the study of cell-free circulating tumor DNA. This study aimed to identify novel DNA methylation biomarkers of colorectal cancer that can be used for the follow-up of patients with colorectal cancer.
METHODS METHODS
A DNA methylation profile was established in the Gene Expression Omnibus (GEO) database (n = 507) using bioinformatics analysis and subsequently confirmed using The Cancer Genome Atlas (TCGA) database (n = 348). The in silico profile was then validated on local tissue and cell-free DNA samples using methylation-specific digital PCR in colorectal cancer patients (n = 35) and healthy donors (n = 35).
RESULTS RESULTS
The DNA methylation of COL25A1 and METAP1D was predicted to be a colorectal cancer biomarker by bioinformatics analysis (ROC AUC = 1, 95% CI [0.999-1]). The two biomarkers were confirmed with tissue samples, and the combination of COL25A1 and METAP1D yielded 49% sensitivity and 100% specificity for cell-free DNA.
CONCLUSION CONCLUSIONS
Bioinformatics analysis of public databases revealed COL25A1 and METAP1D DNA methylation as clinically applicable liquid biopsies DNA methylation biomarkers. The specificity implies an excellent positive predictive value for follow-up, and the high sensitivity and relative noninvasiveness of a blood-based test make these biomarkers compatible with colorectal cancer screening. However, the clinical impact of these biomarkers in colorectal cancer screening and follow-up needs to be established in further prospective studies.

Identifiants

pubmed: 39425144
doi: 10.1186/s13148-024-01748-1
pii: 10.1186/s13148-024-01748-1
doi:

Substances chimiques

Biomarkers, Tumor 0
Circulating Tumor DNA 0

Types de publication

Journal Article

Langues

eng

Sous-ensembles de citation

IM

Pagination

146

Informations de copyright

© 2024. The Author(s).

Références

Ferlay J, Ervik M, Lam F, Laversanne M, Colombet M, Mery L, Piñeros M, Znaor A, Soerjomataram I, Bray F. Global cancer observatory: cancer today. Lyon: International Agency for Research on Cancer; 2024. Available from: https://gco.iarc.who.int/today , Accessed 06 March 2024.
Dekker E, Tanis PJ, Vleugels JLA, Kasi PM, Wallace MB. Colorectal cancer. Lancet. 2019;394:1467–80.
doi: 10.1016/S0140-6736(19)32319-0 pubmed: 31631858
Winawer SJ, Zauber AG. The advanced adenoma as the primary target of screening. Gastrointest Endosc Clin N Am. 2002;12(1–9):v.
pubmed: 11916153
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. Gut. 2015;64:1327–37.
doi: 10.1136/gutjnl-2014-308074 pubmed: 26041750
Malla M, Loree JM, Kasi PM, Parikh AR. Using circulating tumor DNA in colorectal cancer: current and evolving practices. J Clin Oncol. 2022;40:2846–57.
doi: 10.1200/JCO.21.02615 pubmed: 35839443 pmcid: 9390824
Morris VK, Kennedy EB, Baxter NN, Benson AB, Cercek A, Cho M, et al. Treatment of metastatic colorectal cancer: ASCO guideline. J Clin Oncol. 2023;41:678–700.
doi: 10.1200/JCO.22.01690 pubmed: 36252154
Levin-Sparenberg E, Bylsma LC, Lowe K, Sangare L, Fryzek JP, Alexander DD. A systematic literature review and meta-analysis describing the prevalence of KRAS, NRAS, and BRAF gene mutations in metastatic colorectal cancer. Gastroenterol Res. 2020;13:184–98.
doi: 10.14740/gr1167
You JS, Jones PA. Cancer genetics and epigenetics: Two sides of the same coin? Cancer Cell. 2012;22:9–20.
doi: 10.1016/j.ccr.2012.06.008 pubmed: 22789535 pmcid: 3396881
Weinstein JN, Collisson EA, Mills GB, Shaw KM, Ozenberger BA, Ellrott K, et al. The cancer genome atlas pan-cancer analysis project. Nat Genet. 2013;45:1113–20.
doi: 10.1038/ng.2764 pubmed: 24071849 pmcid: 3919969
Müller D, Győrffy B. DNA methylation-based diagnostic, prognostic, and predictive biomarkers in colorectal cancer. Biochim Biophys Acta Rev Cancer. 2022;1877:188722.
doi: 10.1016/j.bbcan.2022.188722 pubmed: 35307512
Chung DC, Gray DM, Singh H, Issaka RB, Raymond VM, Eagle C, et al. A cell-free DNA blood-based test for colorectal cancer screening. N Engl J Med. 2024;390:973–83.
doi: 10.1056/NEJMoa2304714 pubmed: 38477985
Diaz LA, Bardelli A. Liquid biopsies: genotyping circulating tumor DNA. J Clin Oncol. 2014;32:579–86.
doi: 10.1200/JCO.2012.45.2011 pubmed: 24449238 pmcid: 4820760
R Core Team (2020) R: A language and environment for statistical computing. R Foundation for Statistical Computing, Vienna, Austria. https://www.R-project.org/ . [Internet]. [cited 2021 Jan 22]. Available from: https://www.r-project.org/
Robin X, Turck N, Hainard A, Tiberti N, Lisacek F, Sanchez J-C, et al. pROC: an open-source package for R and S+ to analyze and compare ROC curves. BMC Bioinform. 2011;12:77.
doi: 10.1186/1471-2105-12-77
Davis S, Meltzer PS. GEOquery: a bridge between the Gene Expression Omnibus (GEO) and BioConductor. Bioinformatics. 2007;23:1846–7.
doi: 10.1093/bioinformatics/btm254 pubmed: 17496320
Colaprico A, Silva TC, Olsen C, Garofano L, Cava C, Garolini D, et al. TCGAbiolinks: an R/Bioconductor package for integrative analysis of TCGA data. Nucleic Acids Res. 2016;44:e71–e71.
doi: 10.1093/nar/gkv1507 pubmed: 26704973
Du P, Zhang X, Huang C-C, Jafari N, Kibbe WA, Hou L, et al. Comparison of Beta-value and M-value methods for quantifying methylation levels by microarray analysis. BMC Bioinform. 2010;11:587.
doi: 10.1186/1471-2105-11-587
DeLong ER, DeLong DM, Clarke-Pearson DL. Comparing the areas under two or more correlated receiver operating characteristic curves: a nonparametric approach. Biometrics. 1988;44:837–45.
doi: 10.2307/2531595 pubmed: 3203132
Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15:550.
doi: 10.1186/s13059-014-0550-8 pubmed: 25516281 pmcid: 4302049
Stevenson M, Sergeant E, Heuer C, Nunes T, Heuer C, Marshall J, et al. epiR: Tools for the Analysis of Epidemiological Data [Internet]. 2024 [cited 2024 Aug 25]. Available from: https://cran.r-project.org/web/packages/epiR/index.html
Li L-C, Dahiya R. MethPrimer: designing primers for methylation PCRs. Bioinformatics. 2002;18:1427–31.
doi: 10.1093/bioinformatics/18.11.1427 pubmed: 12424112
Tusnády GE, Simon I, Váradi A, Arányi T. BiSearch: primer-design and search tool for PCR on bisulfite-treated genomes. Nucleic Acids Res. 2005;33:e9.
doi: 10.1093/nar/gni012 pubmed: 15653630 pmcid: 546182
Weisenberger DJ, Trinh BN, Campan M, Sharma S, Long TI, Ananthnarayan S, et al. DNA methylation analysis by digital bisulfite genomic sequencing and digital MethyLight. Nucleic Acids Res. 2008;36:4689–98.
doi: 10.1093/nar/gkn455 pubmed: 18628296 pmcid: 2504308
Singh RR. Next-generation sequencing in high-sensitive detection of mutations in tumors: challenges, advances, and applications. J Mol Diagn. 2020;22:994–1007.
doi: 10.1016/j.jmoldx.2020.04.213 pubmed: 32480002
BlueBooksOnline [Internet]. [cited 2024 Sep 5]. Available from: https://tumourclassification.iarc.who.int/chaptercontent/31/280 .
Li H, Du Y, Zhang D, Wang L-N, Yang C, Liu B, et al. Identification of novel DNA methylation markers in colorectal cancer using MIRA-based microarrays. Oncol Rep. 2012;28:99–104.
pubmed: 22552777
Naumov VA, Generozov EV, Zaharjevskaya NB, Matushkina DS, Larin AK, Chernyshov SV, et al. Genome-scale analysis of DNA methylation in colorectal cancer using Infinium HumanMethylation450 BeadChips. Epigenetics. 2013;8:921–34.
doi: 10.4161/epi.25577 pubmed: 23867710 pmcid: 3883769
Nakayama F, Umeda S, Yasuda T, Fujita M, Asada M, Meineke V, et al. Cellular internalization of fibroblast growth factor-12 exerts radioprotective effects on intestinal radiation damage independently of FGFR signaling. Int J Radiat Oncol Biol Phys. 2014;88:377–84.
doi: 10.1016/j.ijrobp.2013.10.035 pubmed: 24315567
Pan Y, Tong JHM, Kang W, Lung RWM, Chak WP, Chung LY, et al. EXOSC4 functions as a potential oncogene in development and progression of colorectal cancer. Mol Carcinog. 2018;57:1780–91.
doi: 10.1002/mc.22896 pubmed: 30155936
Lendvai Á, Johannes F, Grimm C, Eijsink JJH, Wardenaar R, Volders HH, et al. Genome-wide methylation profiling identifies hypermethylated biomarkers in high-grade cervical intraepithelial neoplasia. Epigenetics. 2012;7:1268–78.
doi: 10.4161/epi.22301 pubmed: 23018867 pmcid: 3499328
Rappaport N, Fishilevich S, Nudel R, Twik M, Belinky F, Plaschkes I, et al. Rational confederation of genes and diseases: NGS interpretation via GeneCards. MalaCards VarElect Biomed Eng Online. 2017;16:72.
doi: 10.1186/s12938-017-0359-2 pubmed: 28830434
Lee Y, Kim H, Lee E, Hahn H, Heo Y, Jang DM, et al. Structural insights into N-terminal methionine cleavage by the human mitochondrial methionine aminopeptidase, MetAP1D. Sci Rep. 2023;13:22326.
doi: 10.1038/s41598-023-49332-6 pubmed: 38102161 pmcid: 10724148
Hu X, Addlagatta A, Lu J, Matthews BW, Liu JO. Elucidation of the function of type 1 human methionine aminopeptidase during cell cycle progression. Proc Natl Acad Sci. 2006;103:18148–53.
doi: 10.1073/pnas.0608389103 pubmed: 17114291 pmcid: 1838721
Randhawa H, Chikara S, Gehring D, Yildirim T, Menon J, Reindl KM. Overexpression of peptide deformylase in breast, colon, and lung cancers. BMC Cancer. 2013;13:321.
doi: 10.1186/1471-2407-13-321 pubmed: 23815882 pmcid: 3722014
Leszczyniecka M, Bhatia U, Cueto M, Nirmala NR, Towbin H, Vattay A, et al. MAP1D, a novel methionine aminopeptidase family member is overexpressed in colon cancer. Oncogene. 2006;25:3471–8.
doi: 10.1038/sj.onc.1209383 pubmed: 16568094
Church TR, Wandell M, Lofton-Day C, Mongin SJ, Burger M, Payne SR, et al. Prospective evaluation of methylated SEPT9 in plasma for detection of asymptomatic colorectal cancer. Gut. 2014;63:317–25.
doi: 10.1136/gutjnl-2012-304149 pubmed: 23408352
Petit J, Carroll G, Williams H, Pockney P, Scott RJ. Evaluation of a multi-gene methylation blood-test for the detection of colorectal cancer. Med Sci (Basel). 2023;11:60.
pubmed: 37755164
Li D, Zhang L, Fu J, Huang H, Liu Y, Zhu L, et al. Discovery and validation of tissue-specific DNA methylation as noninvasive diagnostic markers for colorectal cancer. Clin Epigenet. 2022;14:102.
doi: 10.1186/s13148-022-01312-9
Jensen SØ, Øgaard N, Ørntoft M-BW, Rasmussen MH, Bramsen JB, Kristensen H, et al. Novel DNA methylation biomarkers show high sensitivity and specificity for blood-based detection of colorectal cancer—a clinical biomarker discovery and validation study. Clin Epigenet. 2019;11:158.
doi: 10.1186/s13148-019-0757-3
Strijker M, Soer EC, de Pastena M, Creemers A, Balduzzi A, Beagan JJ, et al. Circulating tumor DNA quantity is related to tumor volume and both predict survival in metastatic pancreatic ductal adenocarcinoma. Int J Cancer. 2020;146:1445–56.
doi: 10.1002/ijc.32586 pubmed: 31340061
Avanzini S, Kurtz DM, Chabon JJ, Moding EJ, Hori SS, Gambhir SS, et al. A mathematical model of ctDNA shedding predicts tumor detection size. Sci Adv. 2020;6:4308.
doi: 10.1126/sciadv.abc4308

Auteurs

Alexis Overs (A)

Department of Oncobiology, University Hospital of Besançon, 3 Boulevard Alexandre Fleming, 25000, Besançon, France. aovers@chu-besancon.fr.
UMR1098, INSERM, University of Bourgogne Franche-Comté, Besançon, France. aovers@chu-besancon.fr.

Paul Peixoto (P)

UMR1098, INSERM, University of Bourgogne Franche-Comté, Besançon, France.

Eric Hervouet (E)

UMR1098, INSERM, University of Bourgogne Franche-Comté, Besançon, France.

Chloé Molimard (C)

Department of Pathology, University Hospital of Besançon, 25000, Besancon, France.

Franck Monnien (F)

Department of Pathology, University Hospital of Besançon, 25000, Besancon, France.

Jules Durand (J)

UMR1098, INSERM, University of Bourgogne Franche-Comté, Besançon, France.

Michael Guittaut (M)

UMR1098, INSERM, University of Bourgogne Franche-Comté, Besançon, France.

Angélique Vienot (A)

Department of Oncology, University Hospital of Besançon, 25000, Besancon, France.

Julien Viot (J)

Department of Oncology, University Hospital of Besançon, 25000, Besancon, France.

Michael Herfs (M)

Laboratory of Experimental Pathology, GIGA-Cancer, University of Liege, Liège, Belgium.

Christophe Borg (C)

UMR1098, INSERM, University of Bourgogne Franche-Comté, Besançon, France.
Department of Oncology, University Hospital of Besançon, 25000, Besancon, France.

Jean-Paul Feugeas (JP)

Department of Oncobiology, University Hospital of Besançon, 3 Boulevard Alexandre Fleming, 25000, Besançon, France.
UMR1098, INSERM, University of Bourgogne Franche-Comté, Besançon, France.

Zohair Selmani (Z)

Department of Oncobiology, University Hospital of Besançon, 3 Boulevard Alexandre Fleming, 25000, Besançon, France.
UMR1098, INSERM, University of Bourgogne Franche-Comté, Besançon, France.

Articles similaires

[Redispensing of expensive oral anticancer medicines: a practical application].

Lisanne N van Merendonk, Kübra Akgöl, Bastiaan Nuijen
1.00
Humans Antineoplastic Agents Administration, Oral Drug Costs Counterfeit Drugs

Smoking Cessation and Incident Cardiovascular Disease.

Jun Hwan Cho, Seung Yong Shin, Hoseob Kim et al.
1.00
Humans Male Smoking Cessation Cardiovascular Diseases Female
Humans United States Aged Cross-Sectional Studies Medicare Part C
1.00
Humans Yoga Low Back Pain Female Male

Classifications MeSH