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.

  • Technical Report
  • Published:

An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage

Abstract

Circulating tumor DNA (ctDNA) is a promising biomarker for noninvasive assessment of cancer burden, but existing ctDNA detection methods have insufficient sensitivity or patient coverage for broad clinical applicability. Here we introduce cancer personalized profiling by deep sequencing (CAPP-Seq), an economical and ultrasensitive method for quantifying ctDNA. We implemented CAPP-Seq for non–small-cell lung cancer (NSCLC) with a design covering multiple classes of somatic alterations that identified mutations in >95% of tumors. We detected ctDNA in 100% of patients with stage II–IV NSCLC and in 50% of patients with stage I, with 96% specificity for mutant allele fractions down to 0.02%. Levels of ctDNA were highly correlated with tumor volume and distinguished between residual disease and treatment-related imaging changes, and measurement of ctDNA levels allowed for earlier response assessment than radiographic approaches. Finally, we evaluated biopsy-free tumor screening and genotyping with CAPP-Seq. We envision that CAPP-Seq could be routinely applied clinically to detect and monitor diverse malignancies, thus facilitating personalized cancer therapy.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: Development of CAPP-Seq.
Figure 2: Analytical performance.
Figure 3: Sensitivity and specificity analysis.
Figure 4: Noninvasive detection and monitoring of ctDNA.

Similar content being viewed by others

Accession codes

Primary accessions

Sequence Read Archive

References

  1. Taniguchi, K. et al. Quantitative detection of EGFR mutations in circulating tumor DNA derived from lung adenocarcinomas. Clin. Cancer Res. 17, 7808–7815 (2011).

    Article  CAS  Google Scholar 

  2. Rosell, R. et al. Screening for epidermal growth factor receptor mutations in lung cancer. N. Engl. J. Med. 361, 958–967 (2009).

    Article  CAS  Google Scholar 

  3. Kuang, Y. et al. Noninvasive detection of EGFR T790M in gefitinib or erlotinib resistant non-small cell lung cancer. Clin. Cancer Res. 15, 2630–2636 (2009).

    Article  CAS  Google Scholar 

  4. Gautschi, O. et al. Origin and prognostic value of circulating KRAS mutations in lung cancer patients. Cancer Lett. 254, 265–273 (2007).

    Article  CAS  Google Scholar 

  5. Leary, R.J. et al. Development of personalized tumor biomarkers using massively parallel sequencing. Sci. Transl. Med. 2, 20ra14 (2010).

    Article  Google Scholar 

  6. McBride, D.J. et al. Use of cancer-specific genomic rearrangements to quantify disease burden in plasma from patients with solid tumors. Genes Chromosom. Cancer 49, 1062–1069 (2010).

    Article  CAS  Google Scholar 

  7. He, J. et al. IgH gene rearrangements as plasma biomarkers in non-Hodgkin's lymphoma patients. Oncotarget 2, 178–185 (2011).

    Article  Google Scholar 

  8. Forshew, T. et al. Noninvasive identification and monitoring of cancer mutations by targeted deep sequencing of plasma DNA. Sci. Transl. Med. 4, 136ra168 (2012).

    Article  Google Scholar 

  9. Leary, R.J. et al. Detection of chromosomal alterations in the circulation of cancer patients with whole-genome sequencing. Sci. Transl. Med. 4, 162ra154 (2012).

    Article  Google Scholar 

  10. Narayan, A. et al. Ultrasensitive measurement of hotspot mutations in tumor DNA in blood using error-suppressed multiplexed deep sequencing. Cancer Res. 72, 3492–3498 (2012).

    Article  CAS  Google Scholar 

  11. Dawson, S.J. et al. Analysis of circulating tumor DNA to monitor metastatic breast cancer. N. Engl. J. Med. 368, 1199–1209 (2013).

    Article  CAS  Google Scholar 

  12. Murtaza, M. et al. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature 497, 108–112 (2013).

    Article  CAS  Google Scholar 

  13. Crowley, E., Di Nicolantonio, F., Loupakis, F. & Bardelli, A. Liquid biopsy: monitoring cancer-genetics in the blood. Nat. Rev. Clin. Oncol. 10, 472–484 (2013).

    Article  CAS  Google Scholar 

  14. Forbes, S.A. et al. COSMIC (the Catalogue of Somatic Mutations in Cancer): a resource to investigate acquired mutations in human cancer. Nucleic Acids Res. 38, D652–D657 (2010).

    Article  CAS  Google Scholar 

  15. Ding, L. et al. Somatic mutations affect key pathways in lung adenocarcinoma. Nature 455, 1069–1075 (2008).

    Article  CAS  Google Scholar 

  16. Youn, A. & Simon, R. Identifying cancer driver genes in tumor genome sequencing studies. Bioinformatics 27, 175–181 (2011).

    Article  CAS  Google Scholar 

  17. Bergethon, K. et al. ROS1 rearrangements define a unique molecular class of lung cancers. J. Clin. Oncol. 30, 863–870 (2012).

    Article  CAS  Google Scholar 

  18. Kwak, E.L. et al. Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. N. Engl. J. Med. 363, 1693–1703 (2010).

    Article  CAS  Google Scholar 

  19. Pao, W. & Hutchinson, K.E. Chipping away at the lung cancer genome. Nat. Med. 18, 349–351 (2012).

    Article  CAS  Google Scholar 

  20. Imielinski, M. et al. Mapping the hallmarks of lung adenocarcinoma with massively parallel sequencing. Cell 150, 1107–1120 (2012).

    Article  CAS  Google Scholar 

  21. Govindan, R. et al. Genomic landscape of non-small cell lung cancer in smokers and never-smokers. Cell 150, 1121–1134 (2012).

    Article  CAS  Google Scholar 

  22. Koivunen, J.P. et al. EML4-ALK fusion gene and efficacy of an ALK kinase inhibitor in lung cancer. Clin. Cancer Res. 14, 4275–4283 (2008).

    Article  CAS  Google Scholar 

  23. Rikova, K. et al. Global survey of phosphotyrosine signaling identifies oncogenic kinases in lung cancer. Cell 131, 1190–1203 (2007).

    Article  CAS  Google Scholar 

  24. Fan, H.C., Blumenfeld, Y.J., Chitkara, U., Hudgins, L. & Quake, S.R. Noninvasive diagnosis of fetal aneuploidy by shotgun sequencing DNA from maternal blood. Proc. Natl. Acad. Sci. USA 105, 16266–16271 (2008).

    Article  CAS  Google Scholar 

  25. Su, Z. et al. A platform for rapid detection of multiple oncogenic mutations with relevance to targeted therapy in non-small-cell lung cancer. J. Mol. Diagn. 13, 74–84 (2011).

    Article  CAS  Google Scholar 

  26. Kobayashi, S. et al. EGFR mutation and resistance of non-small-cell lung cancer to gefitinib. N. Engl. J. Med. 352, 786–792 (2005).

    Article  CAS  Google Scholar 

  27. Iyengar, P. & Timmerman, R.D. Stereotactic ablative radiotherapy for non-small cell lung cancer: rationale and outcomes. J. Natl. Compr. Canc. Netw. 10, 1514–1520 (2012).

    Article  Google Scholar 

  28. Nesbitt, J.C., Putnam, J.B. Jr., Walsh, G.L., Roth, J.A. & Mountain, C.F. Survival in early-stage non-small cell lung cancer. Ann. Thorac. Surg. 60, 466–472 (1995).

    Article  CAS  Google Scholar 

  29. Aberle, D.R. et al. Reduced lung-cancer mortality with low-dose computed tomographic screening. N. Engl. J. Med. 365, 395–409 (2011).

    Article  Google Scholar 

  30. Diehl, F. et al. Detection and quantification of mutations in the plasma of patients with colorectal tumors. Proc. Natl. Acad. Sci. USA 102, 16368–16373 (2005).

    Article  CAS  Google Scholar 

  31. Diehl, F. et al. Analysis of mutations in DNA isolated from plasma and stool of colorectal cancer patients. Gastroenterology 135, 489–498 (2008).

    Article  CAS  Google Scholar 

  32. Chan, K.C. et al. Cancer genome scanning in plasma: detection of tumor-associated copy number aberrations, single-nucleotide variants, and tumoral heterogeneity by massively parallel sequencing. Clin. Chem. 59, 211–224 (2013).

    Article  CAS  Google Scholar 

  33. Heitzer, E. et al. Tumor associated copy number changes in the circulation of patients with prostate cancer identified through whole-genome sequencing. Genome Med. 5, 30 (2013).

    Article  CAS  Google Scholar 

  34. Schmitt, M.W. et al. Detection of ultra-rare mutations by next-generation sequencing. Proc. Natl. Acad. Sci. USA 109, 14508–14513 (2012).

    Article  CAS  Google Scholar 

  35. Shiroguchi, K., Jia, T.Z., Sims, P.A. & Xie, X.S. Digital RNA sequencing minimizes sequence-dependent bias and amplification noise with optimized single-molecule barcodes. Proc. Natl. Acad. Sci. USA 109, 1347–1352 (2012).

    Article  CAS  Google Scholar 

  36. Quail, M.A. et al. Optimal enzymes for amplifying sequencing libraries. Nat. Methods 9, 10–11 (2012).

    Article  CAS  Google Scholar 

  37. Oyola, S.O. et al. Optimizing Illumina next-generation sequencing library preparation for extremely AT-biased genomes. BMC Genomics 13, 1 (2012).

    Article  CAS  Google Scholar 

  38. Fisher, S. et al. A scalable, fully automated process for construction of sequence-ready human exome targeted capture libraries. Genome Biol. 12, R1 (2011).

    Article  Google Scholar 

  39. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    Article  CAS  Google Scholar 

  40. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  Google Scholar 

  41. Quinlan, A.R. & Hall, I.M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).

    Article  CAS  Google Scholar 

  42. Koboldt, D.C. et al. VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res. 22, 568–576 (2012).

    Article  CAS  Google Scholar 

  43. Fisher, R.A. Statistical Methods for Research Workers (Oliver and Boyd, 1925).

Download references

Acknowledgements

We thank S. Quake and members of his lab for suggestions and N. Neff for technical assistance. This work was supported by the US Department of Defense (M.D., A.A.A., A.M.N.), the US National Institutes of Health Director's New Innovator Award Program (M.D.; 1-DP2-CA186569), the Ludwig Institute for Cancer Research (M.D., A.A.A.), the Radiological Society of North America (S.V.B.; #RR1221), an Association of American Cancer Institutes Translational Cancer Research Fellowship (S.V.B.) and a grant from both the Siebel Stem Cell Institute and the Thomas and Stacey Siebel Foundation (A.M.N.). A.A.A. and M.D. are supported by Doris Duke Clinical Scientist Development Awards.

Author information

Authors and Affiliations

Authors

Contributions

A.M.N., S.V.B., A.A.A. and M.D. developed the concept, designed the experiments, analyzed the data and wrote the manuscript. S.V.B. performed the molecular biology experiments, and A.M.N. performed the bioinformatics analyses. C.L.L. helped develop analytical pipeline software. S.V.B., J.T., J.F.W., N.C.W.E., L.A.M., J.W.N., H.A.W., R.E.M., J.B.S., B.W.L. Jr. and M.D. provided patient specimens. A.A.A. and M.D. contributed equally as senior authors. All authors commented on the manuscript at all stages.

Corresponding authors

Correspondence to Ash A Alizadeh or Maximilian Diehn.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 and Supplementary Methods (PDF 2806 kb)

Supplementary Table 1

NSCLC selector design and coordinates (XLSX 115 kb)

Supplementary Table 2

Quality control metrics of NGS libraries (XLSX 69 kb)

Supplementary Table 3

Clinical history of patients in this study and somatic variants discovered by CAPP-Seq (XLSX 31 kb)

Supplementary Table 4

Detection of somatic variants in plasma DNA from patients with NSCLC (XLSX 78 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Cite this article

Newman, A., Bratman, S., To, J. et al. An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage. Nat Med 20, 548–554 (2014). https://doi.org/10.1038/nm.3519

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.3519

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer