Novel DNA methylation biomarkers show high sensitivity and specificity for blood-based detection of colorectal cancer-a clinical biomarker discovery and validation study

Sarah Østrup Jensen, Nadia Øgaard, Mai-Britt Worm Ørntoft, Mads Heilskov Rasmussen, Jesper Bertram Bramsen, Helle Kristensen, Peter Mouritzen, Mogens Rørbæk Madsen, Anders Husted Madsen, Kåre Gotschalck Sunesen, Lene Hjerrild Iversen, Søren Laurberg, Ib Jarle Christensen, Hans Jørgen Nielsen, Claus Lindbjerg Andersen, Sarah Østrup Jensen, Nadia Øgaard, Mai-Britt Worm Ørntoft, Mads Heilskov Rasmussen, Jesper Bertram Bramsen, Helle Kristensen, Peter Mouritzen, Mogens Rørbæk Madsen, Anders Husted Madsen, Kåre Gotschalck Sunesen, Lene Hjerrild Iversen, Søren Laurberg, Ib Jarle Christensen, Hans Jørgen Nielsen, Claus Lindbjerg Andersen

Abstract

Background: Early detection plays an essential role to reduce colorectal cancer (CRC) mortality. While current screening methods suffer from poor compliance, liquid biopsy-based strategies for cancer detection is rapidly gaining promise. Here, we describe the development of TriMeth, a minimal-invasive blood-based test for detection of early-stage colorectal cancer. The test is based on assessment of three tumour-specific DNA methylation markers in circulating cell-free DNA.

Results: A thorough multi-step biomarker discovery study based on DNA methylation profiles of more than 5000 tumours and blood cell populations identified CRC-specific DNA methylation markers. The DNA methylation patterns of biomarker candidates were validated by bisulfite sequencing and methylation-specific droplet digital PCR in CRC tumour tissue and peripheral blood leucocytes. The three best performing markers were first applied to plasma from 113 primarily early-stage CRC patients and 87 age- and gender-matched colonoscopy-verified controls. Based on this, the test scoring algorithm was locked, and then TriMeth was validated in an independent cohort comprising 143 CRC patients and 91 controls. Three DNA methylation markers, C9orf50, KCNQ5, and CLIP4, were identified, each capable of discriminating plasma from colorectal cancer patients and healthy individuals (areas under the curve 0.86, 0.91, and 0.88). When combined in the TriMeth test, an average sensitivity of 85% (218/256) was observed (stage I: 80% (33/41), stage II: 85% (121/143), stage III: 89% (49/55), and stage IV: 88% (15/17)) at 99% (176/178) specificity in two independent plasma cohorts.

Conclusion: TriMeth enables detection of early-stage colorectal cancer with high sensitivity and specificity. The reported results underline the potential utility of DNA methylation-based detection of circulating tumour DNA in the clinical management of colorectal cancer.

Keywords: Cancer; Circulating tumour DNA; Colorectal cancer; DNA methylation; Early detection; Epigenetic biomarkers; Liquid biopsy.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
Overview of study workflow. Infinium Human Methylation450K BeadChip® array data from > 4000 samples including CRC, PBL, normal colorectal mucosa, and other cancer types were used to identify CRC-specific DNA methylation biomarkers. The methylation pattern of candidate marker regions was confirmed by bisulfite Sanger sequencing of paired CRC tissue, normal colorectal mucosa, and PBLs. Methylation-specific ddPCR assays targeting candidate regions were designed and optimized, and clinical validation was performed by evaluating assays in CRC tumour tissue (n = 36) and PBL from blood donors (n = 27). The top three candidates were analysed in two independent cohorts consisting of plasma from CRC patients and controls. CRC colorectal cancer, PBLs peripheral blood leucocytes, ddPCR droplet digital PCR
Fig. 2
Fig. 2
Schematic representation of biomarker discovery and validation pipeline. Infinium HumanMethylation450K Beadchip® array data were used to evaluate the methylation status of CpG sites in CRC, PBL, normal mucosa, and other cancer types (left panel). We excluded CpG sites that were methylated in blood cells and CpG sites with low methylation in CRC. The remaining 6700 CpG sites were ranked according to CRC sensitivity and specificity against other common cancers. To confirm uniform methylation in genomic regions of candidate CpG sites, bisulfite Sanger sequencing was performed on paired samples of CRC, PBL, and normal colorectal mucosa. Twenty-nine of the top 50 CpG sites were located in regions compatible with successful methylation-specific ddPCR assay design. A total of 58 methylation-specific ddPCR assays were designed for the 29 CpG sites (markers) and tested in a sequence of validation steps (right panel). Assays were excluded if their performance was suboptimal in methylated and unmethylated control DNA, PBLs, and CRC tissue or in plasma from CRC patients. Three markers passed all selection criteria. CRC colorectal cancer, PBL peripheral blood leukocytes, ddPCR droplet digital PCR
Fig. 3
Fig. 3
DNA methylation of C9orf50, KCNQ5, and CLIP4. a DNA methylation levels (Infinium HumanMethylation450K BeadChip® array data) of the three markers C9orf50, KCNQ5, and CLIP4 in 571 individual CRC tumours, 556 PBL samples, and 4111 samples from other cancer types. Each of the three markers are hypermethylated (β-value > 0.35) in > 97% of CRC tumours. b Correlation of DNA methylation levels in CRC tissue of the C9orf50, KCNQ5, and CLIP4 markers. HNSCC head and neck squamous cell carcinoma, BCL B-Cell lymphoma, AML acute myeloid leukaemia
Fig. 4
Fig. 4
Detection of C9orf50, KCNQ5, and CLIP4 markers in plasma (test cohort). a Methylation-specific ddPCR was performed on 4500 copies of bisulfite-converted cfDNA to detect methylated C9orf50, KCNQ5, and CLIP4 in plasma samples from CRC patients and controls in the test cohort. The total number of methylated DNA copies (sum of the three markers) are recorded on the y-axis and CRC samples are arranged by UICC stage. bd ROC curves (right) from test of C9orf50, KCNQ5, and CLIP4 individual markers in plasma. The red dots and dashed lines indicate the sensitivity and specificity when calling samples positive if they contained any methylated DNA. e Sensitivity and specificity by the TriMeth test in plasma. f UICC stage-stratified sensitivity of TriMeth in plasm. Error bars represent 95% CI. ddPCR droplet digital PCR, CRC colorectal cancer, UICC Union for International Cancer Control, ROC receiver operating characteristics
Fig. 5
Fig. 5
Independent validation of TriMeth in plasma. a Methylation-specific ddPCR was performed on cfDNA purified from plasma of CRC patients and controls (validation cohort) to detect methylated C9orf50, KCNQ5, and CLIP4. b Sensitivity and specificity of the TriMeth tes. c UICC stage-stratified sensitivity of TriMeth. Error bars represent 95% CI. ddPCR droplet digital PCR, CRC colorectal cancer, UICC Union for International Cancer Control

References

    1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424. doi: 10.3322/caac.21492.
    1. GLOBOCAN: World Health Organization; 2019 [Available from: .
    1. Zorzi M, Fedeli U, Schievano E, Bovo E, Guzzinati S, Baracco S, et al. Impact on colorectal cancer mortality of screening programmes based on the faecal immunochemical test. Gut. 2015;64(5):784–790. doi: 10.1136/gutjnl-2014-307508.
    1. Brenner H, Stock C, Hoffmeister M. Effect of screening sigmoidoscopy and screening colonoscopy on colorectal cancer incidence and mortality: systematic review and meta-analysis of randomised controlled trials and observational studies. BMJ. 2014;348:g2467. doi: 10.1136/bmj.g2467.
    1. Navarro M, Nicolas A, Ferrandez A, Lanas A. Colorectal cancer population screening programs worldwide in 2016: an update. World J Gastroenterol. 2017;23(20):3632–3642. doi: 10.3748/wjg.v23.i20.3632.
    1. Osborne JM, Wilson C, Moore V, Gregory T, Flight I, Young GP. Sample preference for colorectal cancer screening tests: blood or stool? OJPM. 2012;02(03):326–331. doi: 10.4236/ojpm.2012.23047.
    1. Adler A, Geiger S, Keil A, Bias H, Schatz P, de Vos T, et al. Improving compliance to colorectal cancer screening using blood and stool based tests in patients refusing screening colonoscopy in Germany. BMC Gastroenterol. 2014;14:183. doi: 10.1186/1471-230X-14-183.
    1. Stroun M, Anker P, Maurice P, Lyautey J, Lederrey C, Beljanski M. Neoplastic characteristics of the DNA found in the plasma of cancer patients. Oncology. 1989;46(5):318–322. doi: 10.1159/000226740.
    1. Thierry AR, El Messaoudi S, Gahan PB, Anker P, Stroun M. Origins, structures, and functions of circulating DNA in oncology. Cancer Metastasis Rev. 2016;35(3):347–376. doi: 10.1007/s10555-016-9629-x.
    1. Reinert T, Schøler LV, Thomsen R, Tobiasen H, Vang S, Nordentoft I, et al. Analysis of circulating tumour DNA to monitor disease burden following colorectal cancer surgery. Gut. 2016;65(4):625–634. doi: 10.1136/gutjnl-2014-308859.
    1. Scholer LV, Reinert T, Orntoft MW, Kassentoft CG, Arnadottir SS, Vang S, et al. Clinical implications of monitoring circulating tumor DNA in patients with colorectal Cancer. Clin Cancer Res. 2017;23(18):5437–5445. doi: 10.1158/1078-0432.CCR-17-0510.
    1. Cohen JD, Li L, Wang Y, Thoburn C, Afsari B, Danilova L, et al. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science. 2018;359(6378):926–930. doi: 10.1126/science.aar3247.
    1. Phallen Jillian, Sausen Mark, Adleff Vilmos, Leal Alessandro, Hruban Carolyn, White James, Anagnostou Valsamo, Fiksel Jacob, Cristiano Stephen, Papp Eniko, Speir Savannah, Reinert Thomas, Orntoft Mai-Britt Worm, Woodward Brian D., Murphy Derek, Parpart-Li Sonya, Riley David, Nesselbush Monica, Sengamalay Naomi, Georgiadis Andrew, Li Qing Kay, Madsen Mogens Rørbæk, Mortensen Frank Viborg, Huiskens Joost, Punt Cornelis, van Grieken Nicole, Fijneman Remond, Meijer Gerrit, Husain Hatim, Scharpf Robert B., Diaz Luis A., Jones Siân, Angiuoli Sam, Ørntoft Torben, Nielsen Hans Jørgen, Andersen Claus Lindbjerg, Velculescu Victor E. Direct detection of early-stage cancers using circulating tumor DNA. Science Translational Medicine. 2017;9(403):eaan2415. doi: 10.1126/scitranslmed.aan2415.
    1. Shen SY, Singhania R, Fehringer G, Chakravarthy A, Roehrl MHA, Chadwick D, et al. Sensitive tumour detection and classification using plasma cell-free DNA methylomes. Nature. 2018;563(7732):579–583. doi: 10.1038/s41586-018-0703-0.
    1. Oster B, Thorsen K, Lamy P, Wojdacz TK, Hansen LL, Birkenkamp-Demtroder K, et al. Identification and validation of highly frequent CpG island hypermethylation in colorectal adenomas and carcinomas. Int J Cancer. 2011;129(12):2855–2866. doi: 10.1002/ijc.25951.
    1. Pedersen SK, Symonds EL, Baker RT, Murray DH, McEvoy A, Van Doorn SC, et al. Evaluation of an assay for methylated BCAT1 and IKZF1 in plasma for detection of colorectal neoplasia. BMC Cancer. 2015;15:654. doi: 10.1186/s12885-015-1674-2.
    1. Grützmann R, Molnar B, Pilarsky C, Habermann JK, Schlag PM, Saeger HD, et al. Sensitive detection of colorectal cancer in peripheral blood by septin 9 DNA methylation assay. PLoS One. 2008;3(11):e3759. doi: 10.1371/journal.pone.0003759.
    1. 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(2):317–325. doi: 10.1136/gutjnl-2012-304149.
    1. Worm Ørntoft M-B. Review of blood-based colorectal cancer screening: how far are circulating cell-free DNA methylation markers from clinical implementation? Clin Colorectal Cancer. 2018;17(2):e415–ee33. doi: 10.1016/j.clcc.2018.02.012.
    1. Moss J, Magenheim J, Neiman D, Zemmour H, Loyfer N, Korach A, et al. Comprehensive human cell-type methylation atlas reveals origins of circulating cell-free DNA in health and disease. Nat Commun. 2018;9(1):5068. doi: 10.1038/s41467-018-07466-6.
    1. Lee JK, Liles EG, Bent S, Levin TR, Corley DA. Accuracy of fecal immunochemical tests for colorectal cancer: systematic review and meta-analysis. Ann Intern Med. 2014;160(3):171. doi: 10.7326/M13-1484.
    1. Mertz-Petersen M, Piper TB, Kleif J, Ferm L, Christensen IJ, Nielsen HJ. Triage for selection to colonoscopy? Eur J Surg Oncol. 2018;44(10):1539–1541. doi: 10.1016/j.ejso.2018.06.013.
    1. Rasmussen L, Wilhelmsen M, Christensen IJ, Andersen J, Jørgensen LN, Rasmussen M, et al. Protocol outlines for parts 1 and 2 of the prospective endoscopy III study for the early detection of colorectal cancer: validation of a concept based on blood biomarkers. JMIR Res Protoc. 2016;5(3):e182. doi: 10.2196/resprot.6346.
    1. Bramsen JB, Rasmussen MH, Ongen H, Mattesen TB, Ørntoft M-BW, Árnadóttir SS, et al. Molecular-subtype-specific biomarkers improve prediction of prognosis in colorectal Cancer. Cell Rep. 2017;19(6):1268–1280. doi: 10.1016/j.celrep.2017.04.045.
    1. Ongen H, Andersen CL, Bramsen JB, Oster B, Rasmussen MH, Ferreira PG, et al. Putative cis-regulatory drivers in colorectal cancer. Nature. 2014;512(7512):87–90. doi: 10.1038/nature13602.
    1. Lowe R, Rakyan VK. Marmal-aid--a database for Infinium HumanMethylation450. BMC Bioinformatics. 2013;14:359. doi: 10.1186/1471-2105-14-359.
    1. Morris TJ, Butcher LM, Feber A, Teschendorff AE, Chakravarthy AR, Wojdacz TK, et al. ChAMP: 450k Chip analysis methylation pipeline. Bioinformatics. 2014;30(3):428–430. doi: 10.1093/bioinformatics/btt684.
    1. Aranyi T, Varadi A, Simon I, Tusnady GE. The BiSearch web server. BMC Bioinformatics. 2006;7:431. doi: 10.1186/1471-2105-7-431.
    1. Gustafson KS. Locked nucleic acids can enhance the analytical performance of quantitative methylation-specific polymerase chain reaction. J Mol Diagn. 2008;10(1):33–42. doi: 10.2353/jmoldx.2008.070076.
    1. Huggett JF, Foy CA, Benes V, Emslie K, Garson JA, Haynes R, et al. The digital MIQE guidelines: minimum information for publication of quantitative digital PCR experiments. Clin Chem. 2013;59(6):892–902. doi: 10.1373/clinchem.2013.206375.
    1. Dube S, Qin J, Ramakrishnan R. Mathematical analysis of copy number variation in a DNA sample using digital PCR on a nanofluidic device. PLoS One. 2008;3(8):e2876. doi: 10.1371/journal.pone.0002876.

Source: PubMed

3
Abonner