Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Pan-cancer methylome analysis for cancer diagnosis and classification of cancer cell of origin

Abstract

The accurate and early diagnosis and classification of cancer origin from either tissue or liquid biopsy is crucial for selecting the appropriate treatment and reducing cancer-related mortality. Here, we established the CAncer Cell-of-Origin (CACO) methylation panel using the methylation data of the 28 types of cancer in The Cancer Genome Atlas (7950 patients and 707 normal controls) as well as healthy whole blood samples (95 subjects). We showed that the CACO methylation panel had high diagnostic potential with high sensitivity and specificity in the discovery (maximum AUC = 0.998) and validation (maximum AUC = 1.000) cohorts. Moreover, we confirmed that the CACO methylation panel could identify the cancer cell type of origin using the methylation profile from liquid as well as tissue biopsy, including primary, metastatic, and multiregional cancer samples and cancer of unknown primary, independent of the methylation analysis platform and specimen preparation method. Together, the CACO methylation panel can be a powerful tool for the classification and diagnosis of cancer.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: CACO (CAncer Cell-of Origin) methylation panel can differentiate different cancer types and healthy whole blood from each other.
Fig. 2: CACO methylation panel demonstrated high diagnostic potential.
Fig. 3: CACO methylation panels can identify the cancer tissue of origin among breast, colorectal, and gastric cancer using the methylation signature from the EPIC platform.
Fig. 4: CACO methylation panel can identify COADREAD using both primary and metastatic tumor methylation signatures, which were measured by whole-genome bisulfite sequencing using a TACS ligation-mediated post-bisulfite adaptor tagging (tPBAT) method.
Fig. 5: CACO methylation panels can identify colorectal cancer using any region of the tissue.
Fig. 6: CACO methylation panels can identify the cancer of origin using ctDNA methylation.

Similar content being viewed by others

Code availability

Custom R scripts used to analyze microarray and NGS data are available from the corresponding authors upon reasonable request.

Data availability

Microarray data (IDAT files) have been deposited into the DDBJ Genomic Expression Archive (GEA). GEA accession: E-GEAD-396. tPBAT data (fastq files) have been deposited into the DDBJ Sequence Read Archive (DRA). DRA accession: DRA010902. All other raw data that are not found in the supplementary information are available from the corresponding authors upon reasonable request.

References

  1. Fitzmaurice C, Akinyemiju TF, Al Lami FH, Alam T, Alizadeh-Navaei R, Allen C, et al. Global, regional, and national cancer incidence, mortality, years of life lost, years lived with disability, and disability-adjusted life-years for 29 cancer groups, 1990 to 2016 a systematic analysis for the global burden of disease study global burden of disease cancer collaboration. JAMA Oncol. 2018;4:1553–68.

    Article  PubMed  Google Scholar 

  2. Hoadley KA, Yau C, Hinoue T, Wolf DM, Lazar AJ, Drill E, et al. Cell-of-origin patterns dominate the molecular classification of 10,000 tumors from 33 types of cancer. Cell. 2018;173:291–304. e6

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Hoadley KA, Yau C, Wolf DM, Cherniack AD, Tamborero D, Ng S, et al. Multiplatform analysis of 12 cancer types reveals molecular classification within and across tissues of origin. Cell. 2014;158:929–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Han L, Yuan Y, Zheng S, Yang Y, Li J, Edgerton ME, et al. The Pan-cancer analysis of pseudogene expression reveals biologically and clinically relevant tumour subtypes. Nat Commun 2014;5. https://doi.org/10.1038/ncomms4963.

  5. Akbani R, Ng PKS, Werner HMJ, Shahmoradgoli M, Zhang F, Ju Z, et al. A pan-cancer proteomic perspective on the cancer genome atlas. Nat Commun 2014; 5. https://doi.org/10.1038/ncomms4887.

  6. Stroun M, Maurice P, Vasioukhin V, Lyautey J, Lederrey C, Lefort F, et al. The origin and mechanism of circulating DNA. Ann N Y Acad Sci. 2000;906:161–8.

    Article  CAS  PubMed  Google Scholar 

  7. Schwarzenbach H, Hoon DSB, Pantel K. Cell-free nucleic acids as biomarkers in cancer patients. Nat Rev Cancer 2011;11:426–37.

    Article  CAS  PubMed  Google Scholar 

  8. Francis G, Stein S. Circulating cell-free tumour DNA in the management of cancer. Int J Mol Sci 2015;16:14122–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Jaenisch R, Bird A. Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat Genet 2003;33:245–54.

    Article  CAS  PubMed  Google Scholar 

  10. Esteller M. Epigenetics in cancer. N. Engl J Med. 2008;358:1148–59.

    Article  CAS  PubMed  Google Scholar 

  11. Jones PA, Baylin SB. The epigenomics of cancer. Cell. 2007;128:683–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Baylin SB, Jones PA. Epigenetic determinants of cancer. Cold Spring Harb Perspect Biol. 2016;8:a019505.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Irizarry RA, Ladd-Acosta C, Wen B, Wu Z, Montano C, Onyango P, et al. The human colon cancer methylome shows similar hypo- and hypermethylation at conserved tissue-specific CpG island shores. Nat Genet. 2009;41:178–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Baylin SB, Jones PA. A decade of exploring the cancer epigenome-biological and translational implications. Nat Rev Cancer 2011;11:726–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Field AE, Robertson NA, Wang T, Havas A, Ideker T, Adams PD. DNA methylation clocks in aging: categories, causes, and consequences. Mol Cell 2018;71:882–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Hao X, Luo H, Krawczyk M, Wei W, Wang W, Wang J, et al. DNA methylation markers for diagnosis and prognosis of common cancers. Proc Natl Acad Sci USA. 2017;114:7414–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Witte T, Plass C, Gerhauser C. Pan-cancer patterns of DNA methylation. Genome Med 2014;6:66.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Ding W, Chen G, Shi T. Integrative analysis identifies potential DNA methylation biomarkers for pan-cancer diagnosis and prognosis. Epigenetics. 2019;14:67–80.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Liu B, Liu Y, Pan X, Li M, Yang S, Li SC. DNA methylation markers for pan-cancer prediction by deep learning. Genes (Basel). 2019;10:778.

    Article  CAS  Google Scholar 

  20. Guo S, Diep D, Plongthongkum N, Fung HL, Zhang K, Zhang K. Identification of methylation haplotype blocks AIDS in deconvolution of heterogeneous tissue samples and tumor tissue-of-origin mapping from plasma DNA. Nat Genet. 2017;49:635–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Xu RH, Wei W, Krawczyk M, Wang W, Luo H, Flagg K, et al. Circulating tumour DNA methylation markers for diagnosis and prognosis of hepatocellular carcinoma. Nat Mater. 2017;16:1155–62.

    Article  CAS  PubMed  Google Scholar 

  22. Luo H, Zhao Q, Wei W, Zheng L, Yi S, Li G, et al. Circulating tumor DNA methylation profiles enable early diagnosis, prognosis prediction, and screening for colorectal cancer. Sci Transl Med. 2020;12:eaax7533.

    Article  CAS  PubMed  Google Scholar 

  23. Nuzzo PV, Berchuck JE, Korthauer K, Spisak S, Nassar AH, Abou Alaiwi S, et al. Detection of renal cell carcinoma using plasma and urine cell-free DNA methylomes. Nat Med. 2020;26:1041–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. 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;563:579–83.

    Article  CAS  PubMed  Google Scholar 

  25. Liu L, Toung J, Jassowicz A, Vijayaraghavan R, Kang H, Zhang R, et al. Targeted methylation sequencing of plasma cell-free DNA for cancer detection and classification. Ann Oncol J Eur Soc Med Oncol. 2018;29:1445–53.

    Article  CAS  Google Scholar 

  26. Cokus SJ, Feng S, Zhang X, Chen Z, Merriman B, Haudenschild CD, et al. Shotgun bisulphite sequencing of the Arabidopsis genome reveals DNA methylation patterning. Nature. 2008;452:215–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Lister R, O’Malley RC, Tonti-Filippini J, Gregory BD, Berry CC, Millar AH, et al. Highly integrated single-base resolution maps of the epigenome in Arabidopsis. Cell. 2008;133:523–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Miura F, Enomoto Y, Dairiki R, Ito T. Amplification-free whole-genome bisulfite sequencing by post-bisulfite adaptor tagging. Nucleic Acids Res. 2012;40:e136.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Miura F, Shibata Y, Miura M, Sangatsuda Y, Hisano O, Araki H, et al. Highly efficient single-stranded DNA ligation technique improves low-input whole-genome bisulfite sequencing by post-bisulfite adaptor tagging. Nucleic Acids Res. 2019;47:e85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Lawson DA, Kessenbrock K, Davis RT, Pervolarakis N, Werb Z. Tumour heterogeneity and metastasis at single-cell resolution. Nat Cell Biol 2018;20:1349–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. McGranahan N, Swanton C. Clonal heterogeneity and tumor evolution: past, present, and the future. Cell. 2017;168:613–28.

    Article  CAS  PubMed  Google Scholar 

  32. Turajlic S, Sottoriva A, Graham T, Swanton C. Resolving genetic heterogeneity in cancer. Nat Rev Genet 2019;20:404–16.

    Article  CAS  PubMed  Google Scholar 

  33. Reiter JG, Baretti M, Gerold JM, Makohon-Moore AP, Daud A, Iacobuzio-Donahue CA, et al. An analysis of genetic heterogeneity in untreated cancers. Nat Rev Cancer. 2019;19:639–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Uchi R, Takahashi Y, Niida A, Shimamura T, Hirata H, Sugimachi K, et al. Integrated multiregional analysis proposing a new model of colorectal cancer evolution. PLOS Genet. 2016;12:e1005778.

    Article  PubMed  PubMed Central  Google Scholar 

  35. 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. https://doi.org/10.1038/s41467-018-07466-6.

  36. Varadhachary GR, Raber MN. Cancer of unknown primary site. N. Engl J Med. 2014;371:757–65.

    Article  CAS  PubMed  Google Scholar 

  37. Briasoulis E, Tolis C, Pavlidis N. ESMO minimum clinical recommendations for diagnosis, treatment and follow-up of cancers of unknown primary site (CUP). Ann Oncol. 2001;12:1057–8.

    Article  Google Scholar 

  38. Pavlidis N, Pentheroudakis G. Cancer of unknown primary site. Lancet. 2012;379:1428–35.

    Article  PubMed  Google Scholar 

  39. Moran S, Martinez-Cardús A, Boussios S, Esteller M. Precision medicine based on epigenomics: the paradigm of carcinoma of unknown primary. Nat Rev Clin Oncol 2017;14:682–94.

    Article  PubMed  Google Scholar 

  40. Moran S, Martínez-Cardús A, Sayols S, Musulén E, Balañá C, Estival-Gonzalez A, et al. Epigenetic profiling to classify cancer of unknown primary: a multicentre, retrospective analysis. Lancet Oncol. 2016;17:1386–95.

    Article  PubMed  Google Scholar 

  41. Zhu B, Poeta ML, Costantini M, Zhang T, Shi J, Sentinelli S, et al. The genomic and epigenomic evolutionary history of papillary renal cell carcinomas. Nat Commun 2020; 11. https://doi.org/10.1038/s41467-020-16546-5.

  42. Benson C, Miah AB. Uterine sarcoma - current perspectives. Int J Women’s Health 2017;9:597–606.

    Article  CAS  Google Scholar 

  43. Shen SY, Burgener JM, Bratman SV, De Carvalho DD. Preparation of cfMeDIP-seq libraries for methylome profiling of plasma cell-free DNA. Nat Protoc. 2019;14:2749–80.

    Article  CAS  PubMed  Google Scholar 

  44. Liu Y, Siejka-Zielińska P, Velikova G, Bi Y, Yuan F, Tomkova M, et al. Bisulfite-free direct detection of 5-methylcytosine and 5-hydroxymethylcytosine at base resolution. Nat Biotechnol. 2019;37:424–9.

    Article  CAS  PubMed  Google Scholar 

  45. C H. Statistical analysis for toxicity studies. J Toxicol Pathol. 2018;31:15–22.

    Article  Google Scholar 

Download references

Acknowledgements

The images in Fig. 1a and Supplementary Fig. S1b are from TogoTV (© 2016 DBCLS TogoTV) and Pintarest.

Funding

D.S. was supported by JSPS KAKENHI Grant Number 19K16868. K.M. received funding from the Platform Project for Supporting Drug Discovery, Life Science Research (Basis for Supporting Innovative Drug Discovery and Life Science Research (BINDS)) from AMED under Grant Number JP20am0101103 (support number 0958), P-CREATE from AMED (20cm0106475h0001(e-Rad ID: 20317791)), JSPS KAKENHI (20H05039, 19H03715, 19K09220), Grant-in-Aid for Scientific Research on Innovative Areas (15H05912), Priority Issue on Post-K computer (hp170227, hp160219), the Project for Cancer Research and Therapeutic Evolution (19cm0106504h0004), Research Grant of the Princess Takamatsu Cancer Research, and SRL, Miraka Research Institute and Takeda Science Foundation. K.T. was supported by the Kobayashi Foundation, Takeda Science Foundation and JSPS KAKENHI Grant Number (21H02758 and 21K19402).

Author information

Authors and Affiliations

Authors

Contributions

D.S. and K.T. conceived and designed the research. D.S. performed most of the bioinformatics analysis with assistance from K.T., Y.M., H.H., M.S., and A.N. M.F., K.S., Y.M., Y.K., M.S., and H.B. collected, analyzed, and interpreted the clinical data. K.T., S.T., and A.K. performed the microarray experiments. H.A., F.M., and T.I. performed the PBAT analysis. Y.K., A.K., Y.Y., K.S., T.S., S.I., T.M., M.S., H.B., N.A., and Y.K. provided guidance and scientific input. D.S., K.T., and K.M. wrote the paper.

Corresponding authors

Correspondence to Dai Shimizu or Kenzui Taniue.

Ethics declarations

Competing interests

K.T., S.T., and A.K. were employees of Genomedia Inc. The remaining authors declare no competing interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shimizu, D., Taniue, K., Matsui, Y. et al. Pan-cancer methylome analysis for cancer diagnosis and classification of cancer cell of origin. Cancer Gene Ther 29, 428–436 (2022). https://doi.org/10.1038/s41417-021-00401-w

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41417-021-00401-w

This article is cited by

Search

Quick links