We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Multimodal analysis of ctDNA methylation and fragmentomic profiles enhances detection of nonmetastatic colorectal cancer

    Huu Thinh Nguyen

    University Medical Center, Ho Chi Minh City, Vietnam

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Le Anh Khoa Huynh

    Medical Genetics Institute, Ho Chi Minh City, Vietnam

    Department of Biostatistics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Trieu Vu Nguyen

    Thu Duc City Hospital, Ho Chi Minh City, Vietnam

    ,
    Duc Huy Tran

    University Medical Center, Ho Chi Minh City, Vietnam

    ,
    Thuy Thi Thu Tran

    Medical Genetics Institute, Ho Chi Minh City, Vietnam

    Gene Solutions, Ho Chi Minh City, Vietnam

    ,
    Nguyen Duy Khang Le

    Medical Genetics Institute, Ho Chi Minh City, Vietnam

    Gene Solutions, Ho Chi Minh City, Vietnam

    ,
    Ngoc-An Trinh Le

    University Medical Center, Ho Chi Minh City, Vietnam

    ,
    Truong-Vinh Ngoc Pham

    University Medical Center, Ho Chi Minh City, Vietnam

    ,
    Minh-Triet Le

    University Medical Center, Ho Chi Minh City, Vietnam

    ,
    Thi Mong Quynh Pham

    Medical Genetics Institute, Ho Chi Minh City, Vietnam

    Gene Solutions, Ho Chi Minh City, Vietnam

    ,
    Trong Hieu Nguyen

    Medical Genetics Institute, Ho Chi Minh City, Vietnam

    Gene Solutions, Ho Chi Minh City, Vietnam

    ,
    Thien Chi Van Nguyen

    Medical Genetics Institute, Ho Chi Minh City, Vietnam

    Gene Solutions, Ho Chi Minh City, Vietnam

    ,
    Thanh Dat Nguyen

    Medical Genetics Institute, Ho Chi Minh City, Vietnam

    Gene Solutions, Ho Chi Minh City, Vietnam

    ,
    Bui Que Tran Nguyen

    Medical Genetics Institute, Ho Chi Minh City, Vietnam

    Gene Solutions, Ho Chi Minh City, Vietnam

    ,
    Minh-Duy Phan

    Medical Genetics Institute, Ho Chi Minh City, Vietnam

    Gene Solutions, Ho Chi Minh City, Vietnam

    ,
    Hoa Giang

    Medical Genetics Institute, Ho Chi Minh City, Vietnam

    Gene Solutions, Ho Chi Minh City, Vietnam

    &
    Le Son Tran

    *Author for correspondence: Tel.: +84 70 519 6257;

    E-mail Address: leson1808@gmail.com

    Medical Genetics Institute, Ho Chi Minh City, Vietnam

    Gene Solutions, Ho Chi Minh City, Vietnam

    Published Online:https://doi.org/10.2217/fon-2022-1041

    Aims: Early detection of colorectal cancer (CRC) provides substantially better survival rates. This study aimed to develop a blood-based screening assay named SPOT-MAS (‘screen for the presence of tumor by DNA methylation and size’) for early CRC detection with high accuracy. Methods: Plasma cell-free DNA samples from 159 patients with nonmetastatic CRC and 158 healthy controls were simultaneously analyzed for fragment length and methylation profiles. We then employed a deep neural network with fragment length and methylation signatures to build a classification model. Results: The model achieved an area under the curve of 0.989 and a sensitivity of 96.8% at 97% specificity in detecting CRC. External validation of our model showed comparable performance, with an area under the curve of 0.96. Conclusion: SPOT-MAS based on integration of cancer-specific methylation and fragmentomic signatures could provide high accuracy for early-stage CRC detection.

    Plain language summary

    A novel blood test for early detection of colorectal cancer. Colorectal cancer is a cancer of the colon or rectum, located at the lower end of the digestive tract. The early detection of colorectal cancer can help people with the disease have a higher chance of survival and a better quality of life. Current screening methods can be invasive, cause discomfort or have low accuracy; therefore newer screening methods are needed. In this study we developed a new screening method, called SPOT-MAS, which works by measuring the signals of cancer DNA in the blood. By combining different characteristics of cancer DNA, SPOT-MAS could distinguish blood samples of people with colorectal cancer from those of healthy individuals with high accuracy.

    Tweetable abstract

    SPOT-MAS technology combines methylation and fragmentomic signatures of blood-based circulating tumor DNA in a multimodal deep-learning analysis to enable early detection of colorectal cancer with high accuracy.

    Papers of special note have been highlighted as: • of interest

    References

    • 1. Ferlay J, Colombet M, Soerjomataram I et al. Cancer statistics for the year 2020: an overview. Int. J. Cancer doi:10.1002/ijc.33588 (2021) (Epub ahead of print).
    • 2. 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. CA Cancer J. Clin. 71(3), 209–249 (2021).
    • 3. Bluethmann SM, Mariotto AB, Rowland JH. Anticipating the ‘silver tsunami’: prevalence trajectories and comorbidity burden among older cancer survivors in the United States. Cancer Epidemiol. Biomarkers Prev. 25(7), 1029–1036 (2016).
    • 4. Mariotto AB, Robin Yabroff K, Shao Y, Feuer EJ, Brown ML. Projections of the cost of cancer care in the United States: 2010–2020. J. Natl Cancer Inst. 103(2), 117–128 (2011).
    • 5. Mariotto AB, Enewold L, Zhao J, Zeruto CA, Yabroff KR. Medical care costs associated with cancer survivorship in the United States. Cancer Epidemiol. Biomarkers Prev. 29(7), 1304–1312 (2020).
    • 6. Clarke CA, Hubbell E, Kurian AW, Colditz GA, Hartman A-R, Gomez SL. Projected reductions in absolute cancer-related deaths from diagnosing cancers before metastasis, 2006–2015. Cancer Epidemiol. Biomarkers Prev. 29(5), 895–902 (2020).
    • 7. Siegel R, DeSantis C, Jemal A. Colorectal cancer statistics, 2014. CA Cancer J. Clin. 64(2), 104–117 (2014).
    • 8. Miller KD, Nogueira L, Mariotto AB et al. Cancer treatment and survivorship statistics, 2019. CA Cancer J. Clin. 69(5), 363–385 (2019).
    • 9. Centers for Disease Control and Prevention. What is colorectal cancer screening? (2022). www.cdc.gov/cancer/colorectal/basic_info/screening/index.htm
    • 10. Winawer S, Classen M, Lambert R et al. Colorectal cancer screening: World Gastroenterology Organisation/International Digestive Cancer Alliance practice guidelines. South Afr. Gastroenterol. Rev. 6(1), 13–20 (2008).
    • 11. Ramón y Cajal S, Sesé M, Capdevila C et al. Clinical implications of intratumor heterogeneity: challenges and opportunities. J. Mol. Med. 98(2), 161–177 (2020).
    • 12. Dagogo-Jack I, Shaw AT. Tumour heterogeneity and resistance to cancer therapies. Nat. Rev. Clin. Oncol. 15(2), 81–94 (2018).
    • 13. Marrugo-Ramírez J, Mir M, Samitier J. Blood-based cancer biomarkers in liquid biopsy: a promising non-invasive alternative to tissue biopsy. Int. J. Mol. Sci. 19(10), 2877 (2018).
    • 14. Coto-Llerena M, Terracciano LM, von Flüe M, Ng CKY, Piscuoglio S. Chapter 19: Cell-free DNA in hepatocellular carcinoma. In: Precision Medicine for Investigators, Practitioners and Providers. Faintuch JFaintuch S (Eds). Academic Press, Cambridge, MA, 199–209 (2020). www.sciencedirect.com/science/article/pii/B9780128191781000198
    • 15. Sorenson GD, Pribish DM, Valone FH, Memoli VA, Bzik DJ, Yao SL. Soluble normal and mutated DNA sequences from single-copy genes in human blood. Cancer Epidemiol. Biomarkers Prev. 3(1), 67–71 (1994).
    • 16. Mandel P, Metais P. Nuclear acids in human blood plasma. C. R. Seances Soc. Biol. Fil. 142(3–4), 241–243 (1948).
    • 17. 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 46(5), 318–322 (1989).
    • 18. Thierry AR, El Messaoudi S, Gahan PB, Anker P, Stroun M. Origins, structures, and functions of circulating DNA in oncology. Cancer Metastasis Rev. 35(3), 347–376 (2016).
    • 19. Newman AM, Bratman SV, To J et al. An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage. Nat. Med. 20(5), 548–554 (2014).
    • 20. Forshew T, Murtaza M, Parkinson C et al. Noninvasive identification and monitoring of cancer mutations by targeted deep sequencing of plasma DNA. Sci. Transl. Med. 4(136), 136ra68 (2012).
    • 21. Kinde I, Wu J, Papadopoulos N, Kinzler KW, Vogelstein B. Detection and quantification of rare mutations with massively parallel sequencing. Proc. Natl Acad. Sci. USA 108(23), 9530–9535 (2011).
    • 22. Nguyen HT, Tran DH, Ngo QD et al. Evaluation of a liquid biopsy protocol using ultra-deep massive parallel sequencing for detecting and quantifying circulation tumor DNA in colorectal cancer patients. Cancer Invest. 38(2), 85–93 (2020).
    • 23. Nguyen H-T, Luong BA, Tran D-H et al. Ultra-deep sequencing of plasma-circulating DNA for the detection of tumor- derived mutations in patients with nonmetastatic colorectal cancer. Cancer Invest. 0(0), 1–12 (2021).
    • 24. Razavi P, Li BT, Brown DN et al. High-intensity sequencing reveals the sources of plasma circulating cell-free DNA variants. Nat Med. 25(12), 1928–1937 (2019).
    • 25. Lo YMD, Han DSC, Jiang P, Chiu RWK. Epigenetics, fragmentomics, and topology of cell-free DNA in liquid biopsies. Science 372(6538), eaaw3616 (2021).
    • 26. Xu R-H, Wei W, Krawczyk M et al. Circulating tumour DNA methylation markers for diagnosis and prognosis of hepatocellular carcinoma. Nat. Mater. 16(11), 1155–1161 (2017).
    • 27. Chan KCA, Jiang P, Chan CWM et al. Noninvasive detection of cancer-associated genome-wide hypomethylation and copy number aberrations by plasma DNA bisulfite sequencing. Proc. Natl Acad. Sci. USA 110(47), 18761–18768 (2013). • Demonstrated the use of genome-wide methylation and copy number aberrations in the detection of multiple types of cancer. Analysis with reduced sequencing depth still achieved high diagnostic performance
    • 28. Jin C, Liu X, Zheng W et al. Characterization of fragment sizes, copy number aberrations and 4-mer end motifs in cell-free DNA of hepatocellular carcinoma for enhanced liquid biopsy-based cancer detection. Mol. Oncol. 15(9), 2377–2389 (2021). • Certain 4-mer end motifs of cell-free DNA (cfDNA) associated with fragment size. End motif, fragment size and copy number variation have potential for distinguishing patients with hepatocellular carcinoma
    • 29. Cristiano S, Leal A, Phallen J et al. Genome-wide cell-free DNA fragmentation in patients with cancer. Nature 570(7761), 385–389 (2019). • Fragment length profiles of cfDNA are a promising marker in distinguishing cancer patients from controls and could be used to identify the tissue of origin of the cancers
    • 30. Chen X, Gole J, Gore A et al. Non-invasive early detection of cancer four years before conventional diagnosis using a blood test. Nat. Commun. 11(1), 3475 (2020).
    • 31. Nguyen H-N, Cao N-PT, Van Nguyen T-C et al. Liquid biopsy uncovers distinct patterns of DNA methylation and copy number changes in NSCLC patients with different EGFR-TKI resistant mutations. Sci. Rep. 11(1), 16436 (2021).
    • 32. Andrews S. FastQC: A Quality Control Tool for High Throughput Sequence Data. [Online]. Available at: http://www.bioinformatics.babraham.ac.uk/projects/fastqc/ (2010).
    • 33. Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30(15), 2114–2120 (2014).
    • 34. Krueger F, Andrews SR. Bismark: a flexible aligner and methylation caller for bisulfite-seq applications. Bioinformatics 27(11), 1571–1572 (2011).
    • 35. Li H, Handsaker B, Wysoker A et al. The sequence alignment/map format and SAMtools. Bioinformatics 25(16), 2078–2079 (2009).
    • 36. Quinlan AR, Hall IM. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26(6), 841–842 (2010).
    • 37. Jiang P, Sun K, Lun FMF et al. Methy-pipe: an integrated bioinformatics pipeline for whole genome bisulfite sequencing data analysis. PLOS ONE 9(6), e100360 (2014). • Useful pipeline to process whole-genome bisufite sequencing data
    • 38. Jette P. Entropy, information gain, and data exploration in R (2019). https://rstudio-pubs-static.s3.amazonaws.com/455435_30729e265f7a4d049400d03a18e218db.html
    • 39. Momparler RL, Bovenzi V. DNA methylation and cancer. J. Cell. Physiol. 183(2), 145–154 (2000).
    • 40. Luczak MW, Jagodziński PP. The role of DNA methylation in cancer development. Folia Histochem. Cytobiol. 44(3), 143–154 (2006).
    • 41. Belinsky SA. Gene-promoter hypermethylation as a biomarker in lung cancer. Nat. Rev. Cancer 4(9), 707–717 (2004).
    • 42. Das PM, Singal R. DNA methylation and cancer. J. Clin. Oncol. 22(22), 4632–4642 (2004).
    • 43. Ehrlich M. DNA methylation in cancer: too much, but also too little. Oncogene 21(35), 5400–5413 (2002).
    • 44. Eckert D, Buhl S, Weber S, Jäger R, Schorle H. The AP-2 family of transcription factors. Genome Biol. 6(13), 246 (2005).
    • 45. Ropponen K, Kellokoski J, Pirinen R et al. Expression of transcription factor AP-2 in colorectal adenomas and adenocarcinomas; comparison of immunohistochemistry and in situ hybridisation. J. Clin. Pathol. 54(7), 533–538 (2001).
    • 46. Beck AC, Cho E, White JR et al. AP-2α regulates S-phase and is a marker for sensitivity to PI3K inhibitor buparlisib in colon cancer. Mol. Cancer Res. 19(7), 1156–1167 (2021).
    • 47. Bogachek MV, Park JM, De Andrade JP et al. Inhibiting the SUMO pathway represses the cancer stem cell population in breast and colorectal carcinomas. Stem Cell Rep. 7(6), 1140–1151 (2016).
    • 48. Wang X, Sun D, Tai J et al. TFAP2C promotes stemness and chemotherapeutic resistance in colorectal cancer via inactivating Hippo signaling pathway. J. Exp. Clin. Cancer Res. 37(1), 27 (2018).
    • 49. Kamachi Y, Kondoh H. Sox proteins: regulators of cell fate specification and differentiation. Development 140(20), 4129–4144 (2013).
    • 50. Huang J, Tan Z-R, Yu J et al. DNA hypermethylated status and gene expression of PAX1/SOX1 in patients with colorectal carcinoma. Onco Targets Ther. 10, 4739–4751 (2017).
    • 51. Chung H-H, Kuo C-C, Hsiao C-W et al. A novel prognostic DNA methylation panel for colorectal cancer. Int. J. Mol. Sci. 20(19), 4672 (2019).
    • 52. Zhang Y, Huang S, Dong W et al. SOX7, down-regulated in colorectal cancer, induces apoptosis and inhibits proliferation of colorectal cancer cells. Cancer Lett. 277(1), 29–37 (2009).
    • 53. Zhang W, Glöckner SC, Guo M et al. Epigenetic inactivation of the canonical Wnt antagonist SRY-box containing gene 17 in colorectal cancer. Cancer Res. 68(8), 2764–2772 (2008).
    • 54. Hoffmann MJ, Schulz WA. Causes and consequences of DNA hypomethylation in human cancer. Biochem. Cell Biol. 83(3), 296–321 (2005).
    • 55. Raizis AM, Schmitt F, Jost JP. A bisulfite method of 5-methylcytosine mapping that minimizes template degradation. Anal. Biochem. 226(1), 161–166 (1995).
    • 56. Tanaka K, Okamoto A. Degradation of DNA by bisulfite treatment. Bioorg. Med. Chem. Lett. 17(7), 1912–1915 (2007).
    • 57. Kint S, De Spiegelaere W, De Kesel J, Vandekerckhove L, Van Criekinge W. Evaluation of bisulfite kits for DNA methylation profiling in terms of DNA fragmentation and DNA recovery using digital PCR. PLOS ONE 13(6), e0199091 (2018).
    • 58. Ehrich M, Zoll S, Sur S, van den Boom D. A new method for accurate assessment of DNA quality after bisulfite treatment. Nucleic Acids Res. 35(5), e29 (2007).
    • 59. Werner B, Yuwono NL, Henry C et al. Circulating cell-free DNA from plasma undergoes less fragmentation during bisulfite treatment than genomic DNA due to low molecular weight. PLOS ONE 14(10), e0224338 (2019).
    • 60. Jiang P, Chan CWM, Chan KCA et al. Lengthening and shortening of plasma DNA in hepatocellular carcinoma patients. Proc. Natl Acad. Sci. USA 112(11), E1317–E1325 (2015).
    • 61. Ding SC, Lo YMD. Cell-free DNA fragmentomics in liquid biopsy. Diagnostics (Basel) 12(4), 978 (2022). • Review of different characteristics of cfDNA currently being used for prenatal testing, cancer detection and transplant monitoring
    • 62. Rawla P, Sunkara T, Barsouk A. Epidemiology of colorectal cancer: incidence, mortality, survival, and risk factors. Prz. Gastroenterol. 14(2), 89–103 (2019). • Review on different risk factors for colorectal cancer
    • 63. Kim S-E, Paik HY, Yoon H, Lee JE, Kim N, Sung M-K. Sex- and gender-specific disparities in colorectal cancer risk. World J. Gastroenterol. 21(17), 5167–5175 (2015).
    • 64. Li LY, Luo X, Wang X. Endonuclease G is an apoptotic DNase when released from mitochondria. Nature 412(6842), 95–99 (2001).
    • 65. Ivanov M, Baranova A, Butler T, Spellman P, Mileyko V. Non-random fragmentation patterns in circulating cell-free DNA reflect epigenetic regulation. BMC Genomics 16(Suppl. 13), S1 (2015).
    • 66. Clark DJ. Nucleosome positioning, nucleosome spacing and the nucleosome code. J. Biomol. Struct. Dyn. 27(6), 781–793 (2010).
    • 67. Chodavarapu RK, Feng S, Bernatavichute YV et al. Relationship between nucleosome positioning and DNA methylation. Nature 466(7304), 388–392 (2010).
    • 68. Kang M, Ko E, Mersha TB. A roadmap for multi-omics data integration using deep learning. Brief. Bioinform. 23(1), bbab454 (2022).
    • 69. Chen Y, Li Y, Narayan R, Subramanian A, Xie X. Gene expression inference with deep learning. Bioinformatics 32(12), 1832–1839 (2016).
    • 70. Alipanahi B, Delong A, Weirauch MT, Frey BJ. Predicting the sequence specificities of DNA- and RNA-binding proteins by deep learning. Nat. Biotechnol. 33(8), 831–838 (2015).
    • 71. Poplin R, Chang P-C, Alexander D et al. A universal SNP and small-indel variant caller using deep neural networks. Nat. Biotechnol. 36(10), 983–987 (2018).
    • 72. Lofton-Day C, Model F, Devos T et al. DNA methylation biomarkers for blood-based colorectal cancer screening. Clin. Chem. 54(2), 414–423 (2008).
    • 73. deVos T, Tetzner R, Model F et al. Circulating methylated SEPT9 DNA in plasma is a biomarker for colorectal cancer. Clin. Chem. 55(7), 1337–1346 (2009).
    • 74. Elliott HR, Burrows K, Min JL et al. Characterisation of ethnic differences in DNA methylation between UK-resident South Asians and Europeans. Clin. Epigenetics 14(1), 130 (2022).
    • 75. Galanter JM, Gignoux CR, Oh SS et al. Differential methylation between ethnic sub-groups reflects the effect of genetic ancestry and environmental exposures. Elife 6, e20532 (2017).
    • 76. Chen M, Zhao H. Next-generation sequencing in liquid biopsy: cancer screening and early detection. Hum. Genomics 13(1), 34 (2019).
    • 77. Xiao B, Ye X, Wang L et al. Whole genome low-coverage sequencing concurrently detecting copy number variations and their underlying complex chromosomal rearrangements by systematic breakpoint mapping in intellectual deficiency/developmental delay patients. Front. Genet. 11, 616 (2020).
    • 78. Bouzidi A, Labreche K, Baron M et al. Low-coverage whole genome sequencing of cell-free DNA from immunosuppressed cancer patients enables tumor fraction determination and reveals relevant copy number alterations. Front. Cell Dev. Biol. 9, 661272 (2021).
    • 79. Yang S, Zhihua P, Yu J et al. Early cancer detection using low-coverage whole-genome sequencing of cell-free DNA fragments. J. Clin. Oncol. 39(Suppl. 15), e22510–e22510 (2021).
    • 80. Mouliere F, Smith CG, Heider K et al. Fragmentation patterns and personalized sequencing of cell-free DNA in urine and plasma of glioma patients. EMBO Mol. Med. 13(8), e12881 (2021).