Cancer Res Treat.  2023 Apr;55(2):351-366. 10.4143/crt.2022.1026.

Clinical Circulating Tumor DNA Testing for Precision Oncology

Affiliations
  • 1Department of Laboratory Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea

Abstract

Circulating tumor DNA (ctDNA) is the portion of the cell-free DNA in the blood of cancer patients released from tumor cells via apoptosis, necrosis, or active release. From 10 mL of blood, the 4-5 mL of plasma obtained from a cancer patient contains 5-10 ng/mL of ctDNA. The plasma contains not only ctDNA of tumor origin, but also DNA from normal cells or clonal hematopoiesis. Another characteristic of ctDNA is its rapid clearance from circulation; it has a half-life of 16 minutes to 2.5 hours. Obtaining reliable results from ctDNA requires the application and approval of standardized clinical validation guidelines; however, the status of numerous ctDNA tests currently varies. The clinical use of ctDNA testing should be carefully considered based on the test’s specific needs and characteristics. Here we provide the different characteristics of ctDNA tests and information regarding their validation and approval status.

Keyword

Circulating tumor DNA; Clinical validation; Clinical utility; Precision oncology

Figure

  • Fig. 1 The number of publications searched on the PubMed database (https://pubmed.ncbi.nlm.nih.gov/) from 1974 to 2021 using the following keywords: cell-free DNA, cfDNA; circulating tumor DNA, ctDNA; and liquid biopsy. Search was limited to exact match for each term, and the species limited to human. The liquid biopsy was first reported in 1974, and cell-free DNA was first reported in 1986. Since 2014, the number of publications for all search terms, including circulating tumor DNA has increased exponentially. The rapid increase in publications resulted in fragmented use of similar nomenclatures, and the number of search results using terms with the same meaning are displayed differently.

  • Fig. 2 Defining a systematic nomenclature for confounding terms regarding circulating tumor DNA (ctDNA). The ctDNA is a concept that belongs to cell-free nucleic acid in a broad sense and cell-free DNA in a narrow sense. The cell-free nucleic acids are biologically and structurally diverse. Depending on the origin of the nucleic acids, the nucleic acids are mixed, such as nucleic acids derived from various cells or microorganisms. For systematic nomenclature, a top-down approach from cell-free nucleic acid into subclasses are necessary. First, nucleic acids can be classified according to its presence in circulation such as blood and lymphatics, or in non-circulatory fluids, such as urine, saliva, and cerebrospinal fluid. As the use of term cell-free nucleic acids is not limited to cancer, but also applied in prenatal testing and post-transplant surveillance, the terms should be organized by their classifications. Figures were created with BioRender [23].

  • Fig. 3 Different features of circulating tumor DNA (ctDNA) in the plasma. Different tumor-related clinical information can be obtained depending on the structural features of the ctDNA detected during the examination. In ctDNA, DNA abnormalities related to cancer are identified as somatic mutations, copy number aberrations, and structural abnormalities of chromosomes such as inversions, translocations, insertions, and deletions. In addition, useful information can be obtained through epigenetic aberrations such as methylation patterns and DNA fragment size distributions. Figures were created with BioRender [23].

  • Fig. 4 A brief summary of the FDA-approved assays in chronological order. Assays are presented in the order of FDA approval date (circle) placed within the timeline (red line). The assays listed above are categorized as in vitro diagnostic devices and assays provided as laboratory services are listed under the timeline. ALK, anaplastic lymphoma kinase; AS-PCR, allele-specific PCR; BCT, blood collection tube; EGFR, epidermal growth factor receptor; cfDNA, cell-free DNA; FDA, Food and Drug Administration; FFPE, formalin-fixed paraffin-embedded; N/A, not applicable; NGS, next-generation sequencing; RT-PCR, real time-polymerase chain reaction.

  • Fig. 5 Current variances present among circulating tumor DNA (ctDNA) testing used in clinical trials. The following critical testing conditions were identified from the clinical trials using ctDNA provided from ClinicalTrials.gov when available; blood collection tube (A), whole blood volume (B), time to sample processing (C), 1st centrifugations (D), 2nd centrifugations (E), DNA extraction method (F), input DNA volume (G), and use of Food and Drug Administration (FDA)–approved test (H). The degree of agreement in factors of whole blood volume, sample processing methods used for ctDNA acquisition, and utilization of FDA-approved assay was low.


Reference

References

1. Mandel P, Metais P. Nuclear acids in human blood plasma. C R Seances Soc Biol Fil. 1948; 142:241–3.
2. 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. 1994; 3:67–71.
3. Nawroz H, Koch W, Anker P, Stroun M, Sidransky D. Microsatellite alterations in serum DNA of head and neck cancer patients. Nat Med. 1996; 2:1035–7.
Article
4. Stroun M, Lyautey J, Lederrey C, Olson-Sand A, Anker P. About the possible origin and mechanism of circulating DNA apoptosis and active DNA release. Clin Chim Acta. 2001; 313:139–42.
5. Diaz LA Jr, Bardelli A. Liquid biopsies: genotyping circulating tumor DNA. J Clin Oncol. 2014; 32:579–86.
Article
6. Pantel K, Alix-Panabieres C. Circulating tumour cells in cancer patients: challenges and perspectives. Trends Mol Med. 2010; 16:398–406.
Article
7. Diehl F, Li M, Dressman D, He Y, Shen D, Szabo S, et al. Detection and quantification of mutations in the plasma of patients with colorectal tumors. Proc Natl Acad Sci U S A. 2005; 102:16368–73.
Article
8. Jahr S, Hentze H, Englisch S, Hardt D, Fackelmayer FO, Hesch RD, et al. DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res. 2001; 61:1659–65.
9. Sacher AG, Paweletz C, Dahlberg SE, Alden RS, O’Connell A, Feeney N, et al. Prospective validation of rapid plasma genotyping for the detection of EGFR and KRAS mutations in advanced lung cancer. JAMA Oncol. 2016; 2:1014–22.
Article
10. Diehl F, Schmidt K, Choti MA, Romans K, Goodman S, Li M, et al. Circulating mutant DNA to assess tumor dynamics. Nat Med. 2008; 14:985–90.
Article
11. Umetani N, Giuliano AE, Hiramatsu SH, Amersi F, Nakagawa T, Martino S, et al. Prediction of breast tumor progression by integrity of free circulating DNA in serum. J Clin Oncol. 2006; 24:4270–6.
Article
12. Li L, Hann HW, Wan S, Hann RS, Wang C, Lai Y, et al. Cell-free circulating mitochondrial DNA content and risk of hepatocellular carcinoma in patients with chronic HBV infection. Sci Rep. 2016; 6:23992.
Article
13. Lehmann-Werman R, Neiman D, Zemmour H, Moss J, Magenheim J, Vaknin-Dembinsky A, et al. Identification of tissue-specific cell death using methylation patterns of circulating DNA. Proc Natl Acad Sci U S A. 2016; 113:E1826–34.
Article
14. Schwarzenbach H, Hoon DS, Pantel K. Cell-free nucleic acids as biomarkers in cancer patients. Nat Rev Cancer. 2011; 11:426–37.
Article
15. Xie M, Lu C, Wang J, McLellan MD, Johnson KJ, Wendl MC, et al. Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nat Med. 2014; 20:1472–8.
Article
16. Genovese G, Kahler AK, Handsaker RE, Lindberg J, Rose SA, Bakhoum SF, et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N Engl J Med. 2014; 371:2477–87.
Article
17. Jaiswal S, Fontanillas P, Flannick J, Manning A, Grauman PV, Mar BG, et al. Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med. 2014; 371:2488–98.
Article
18. Wan JC, Massie C, Garcia-Corbacho J, Mouliere F, Brenton JD, Caldas C, et al. Liquid biopsies come of age: towards implementation of circulating tumour DNA. Nat Rev Cancer. 2017; 17:223–38.
Article
19. Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y, Agrawal N, et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med. 2014; 6:224ra24.
20. Cisneros-Villanueva M, Hidalgo-Perez L, Rios-Romero M, Cedro-Tanda A, Ruiz-Villavicencio CA, Page K, et al. Cell-free DNA analysis in current cancer clinical trials: a review. Br J Cancer. 2022; 126:391–400.
Article
21. Pantel K, Alix-Panabieres C. Real-time liquid biopsy in cancer patients: fact or fiction? Cancer Res. 2013; 73:6384–8.
Article
22. Bronkhorst AJ, Ungerer V, Diehl F, Anker P, Dor Y, Fleischhacker M, et al. Towards systematic nomenclature for cell-free DNA. Hum Genet. 2021; 140:565–78.
Article
23. BioRender. Create professional science figures in minutes [Internet]. Toronto, ON: BioRender;2022. [cited 2022 Oct 10]. Available from: https://biorender.com/.
24. Keller L, Belloum Y, Wikman H, Pantel K. Clinical relevance of blood-based ctDNA analysis: mutation detection and beyond. Br J Cancer. 2021; 124:345–58.
Article
25. Gray ES, Witkowski T, Pereira M, Calapre L, Herron K, Irwin D, et al. Genomic analysis of circulating tumor DNA using a melanoma-specific UltraSEEK Oncogene Panel. J Mol Diagn. 2019; 21:418–26.
26. Kohn L, Johansson M, Grankvist K, Nilsson J. Liquid biopsies in lung cancer-time to implement research technologies in routine care? Ann Transl Med. 2017; 5:278.
Article
27. Diehl F, Li M, He Y, Kinzler KW, Vogelstein B, Dressman D. BEAMing: single-molecule PCR on microparticles in water-in-oil emulsions. Nat Methods. 2006; 3:551–9.
Article
28. Sorber L, Zwaenepoel K, Deschoolmeester V, Van Schil PE, Van Meerbeeck J, Lardon F, et al. Circulating cell-free nucleic acids and platelets as a liquid biopsy in the provision of personalized therapy for lung cancer patients. Lung Cancer. 2017; 107:100–7.
Article
29. Santis G, Angell R, Nickless G, Quinn A, Herbert A, Cane P, et al. Screening for EGFR and KRAS mutations in endobronchial ultrasound derived transbronchial needle aspirates in non-small cell lung cancer using COLD-PCR. PLoS One. 2011; 6:e25191.
Article
30. Castellanos-Rizaldos E, Liu P, Milbury CA, Guha M, Brisci A, Cremonesi L, et al. Temperature-tolerant COLD-PCR reduces temperature stringency and enables robust mutation enrichment. Clin Chem. 2012; 58:1130–8.
Article
31. Narayan A, Carriero NJ, Gettinger SN, Kluytenaar J, Kozak KR, Yock TI, et al. Ultrasensitive measurement of hotspot mutations in tumor DNA in blood using error-suppressed multiplexed deep sequencing. Cancer Res. 2012; 72:3492–8.
Article
32. Newman AM, Lovejoy AF, Klass DM, Kurtz DM, Chabon JJ, Scherer F, et al. Integrated digital error suppression for improved detection of circulating tumor DNA. Nat Biotechnol. 2016; 34:547–55.
Article
33. Dressman D, Yan H, Traverso G, Kinzler KW, Vogelstein B. Transforming single DNA molecules into fluorescent magnetic particles for detection and enumeration of genetic variations. Proc Natl Acad Sci U S A. 2003; 100:8817–22.
Article
34. Dang DK, Park BH. Circulating tumor DNA: current challenges for clinical utility. J Clin Invest. 2022; 132:e154941.
Article
35. Chen M, Zhao H. Next-generation sequencing in liquid biopsy: cancer screening and early detection. Hum Genomics. 2019; 13:34.
Article
36. Mosko MJ, Nakorchevsky AA, Flores E, Metzler H, Ehrich M, van den Boom DJ, et al. Ultrasensitive detection of multiplexed somatic mutations using MALDI-TOF mass spectrometry. J Mol Diagn. 2016; 18:23–31.
Article
37. Arisi MF, Dotan E, Fernandez SV. Circulating tumor DNA in precision oncology and its applications in colorectal cancer. Int J Mol Sci. 2022; 23:4441.
Article
38. Baer C, Kern W, Koch S, Nadarajah N, Schindela S, Meggendorfer M, et al. Ultra-deep sequencing leads to earlier and more sensitive detection of the tyrosine kinase inhibitor resistance mutation T315I in chronic myeloid leukemia. Haematologica. 2016; 101:830–8.
Article
39. Soda N, Clack K, Shiddiky MJ. Recent advances in liquid biopsy technologies for cancer biomarker detection. Sens Diagn. 2022; 1:343–75.
Article
40. Underhill HR, Kitzman JO, Hellwig S, Welker NC, Daza R, Baker DN, et al. Fragment length of circulating tumor DNA. PLoS Genet. 2016; 12:e1006162.
Article
41. Chen A, Li J, Wang L, Huang Q, Zhu J, Wen S, et al. Comparison of paired cerebrospinal fluid and serum cell-free mitochondrial and nuclear DNA with copy number and fragment length. J Clin Lab Anal. 2020; 34:e23238.
Article
42. Cristiano S, Leal A, Phallen J, Fiksel J, Adleff V, Bruhm DC, et al. Genome-wide cell-free DNA fragmentation in patients with cancer. Nature. 2019; 570:385–9.
Article
43. Cai Z, Wang Z, Liu C, Shi D, Li D, Zheng M, et al. Detection of microsatellite instability from circulating tumor DNA by targeted deep sequencing. J Mol Diagn. 2020; 22:860–70.
44. Arzimanoglou II, Gilbert F, Barber HR. Microsatellite instability in human solid tumors. Cancer. 1998; 82:1808–20.
Article
45. Cortes-Ciriano I, Lee S, Park WY, Kim TM, Park PJ. A molecular portrait of microsatellite instability across multiple cancers. Nat Commun. 2017; 8:15180.
Article
46. Boyiadzis MM, Kirkwood JM, Marshall JL, Pritchard CC, Azad NS, Gulley JL. Significance and implications of FDA approval of pembrolizumab for biomarker-defined disease. J Immunother Cancer. 2018; 6:35.
Article
47. Niu B, Ye K, Zhang Q, Lu C, Xie M, McLellan MD, et al. MSIsensor: microsatellite instability detection using paired tumor-normal sequence data. Bioinformatics. 2014; 30:1015–6.
Article
48. Salipante SJ, Scroggins SM, Hampel HL, Turner EH, Pritchard CC. Microsatellite instability detection by next generation sequencing. Clin Chem. 2014; 60:1192–9.
Article
49. Kautto EA, Bonneville R, Miya J, Yu L, Krook MA, Reeser JW, et al. Performance evaluation for rapid detection of pan-cancer microsatellite instability with MANTIS. Oncotarget. 2017; 8:7452–63.
Article
50. Suva ML, Riggi N, Bernstein BE. Epigenetic reprogramming in cancer. Science. 2013; 339:1567–70.
Article
51. 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
52. Ofman JJ, Hall MP, Aravanis AM. GRAIL and the quest for earlier multi-cancer detection [Internet]. Berlin: Springer Nature;2018. [cited 2022 Oct 10]. Available from: https://www.nature.com/articles/d42473-020-00079-y.
53. Koch A, Joosten SC, Feng Z, de Ruijter TC, Draht MX, Melotte V, et al. Analysis of DNA methylation in cancer: location revisited. Nat Rev Clin Oncol. 2018; 15:459–66.
Article
54. Daniunaite K, Jarmalaite S, Kriukiene E. Epigenomic technologies for deciphering circulating tumor DNA. Curr Opin Biotechnol. 2019; 55:23–9.
Article
55. Colella S, Shen L, Baggerly KA, Issa JP, Krahe R. Sensitive and quantitative universal pyrosequencing methylation analysis of CpG sites. Biotechniques. 2003; 35:146–50.
Article
56. Tost J, Dunker J, Gut IG. Analysis and quantification of multiple methylation variable positions in CpG islands by pyrosequencing. Biotechniques. 2003; 35:152–6.
Article
57. Wong IH, Lo YM, Zhang J, Liew CT, Ng MH, Wong N, et al. Detection of aberrant p16 methylation in the plasma and serum of liver cancer patients. Cancer Res. 1999; 59:71–3.
58. Wojdacz TK, Dobrovic A, Hansen LL. Methylation-sensitive high-resolution melting. Nat Protoc. 2008; 3:1903–8.
Article
59. Xia Y, Tang W, Qian X, Li X, Cheng F, Wang K, et al. Efficacy and safety of camrelizumab plus apatinib during the perioperative period in resectable hepatocellular carcinoma: a single-arm, open label, phase II clinical trial. J Immunother Cancer. 2022; 10:e004656.
Article
60. Openshaw MR, Mohamed AA, Ottolini B, Fernandez-Garcia D, Richards CJ, Page K, et al. Longitudinal monitoring of circulating tumour DNA improves prognostication and relapse detection in gastroesophageal adenocarcinoma. Br J Cancer. 2020; 123:1271–9.
Article
61. Martinez-Saez O, Pascual T, Braso-Maristany F, Chic N, Gonzalez-Farre B, Sanfeliu E, et al. Circulating tumor DNA dynamics in advanced breast cancer treated with CDK4/6 inhibition and endocrine therapy. NPJ Breast Cancer. 2021; 7:8.
Article
62. Engel T, Ben-Horin S, Beer-Gabel M. Autonomic dysfunction correlates with clinical and inflammatory activity in patients with Crohn’s disease. Inflamm Bowel Dis. 2015; 21:2320–6.
Article
63. Wang X, Liu H, Zhao C, Li W, Xu H, Chen Y. The DEAD-box RNA helicase 51 controls non-small cell lung cancer proliferation by regulating cell cycle progression via multiple pathways. Sci Rep. 2016; 6:26108.
Article
64. Merker JD, Oxnard GR, Compton C, Diehn M, Hurley P, Lazar AJ, et al. Circulating tumor DNA analysis in patients with cancer: American Society of Clinical Oncology and College of American Pathologists Joint Review. Arch Pathol Lab Med. 2018; 142:1242–53.
Article
65. El Messaoudi S, Rolet F, Mouliere F, Thierry AR. Circulating cell free DNA: preanalytical considerations. Clin Chim Acta. 2013; 424:222–30.
Article
66. Norton SE, Lechner JM, Williams T, Fernando MR. A stabilizing reagent prevents cell-free DNA contamination by cellular DNA in plasma during blood sample storage and shipping as determined by digital PCR. Clin Biochem. 2013; 46:1561–5.
Article
67. Toro PV, Erlanger B, Beaver JA, Cochran RL, VanDenBerg DA, Yakim E, et al. Comparison of cell stabilizing blood collection tubes for circulating plasma tumor DNA. Clin Biochem. 2015; 48:993–8.
Article
68. Barra GB, Santa Rita TH, de Almeida Vasques J, Chianca CF, Nery LF, Santana Soares Costa S. EDTA-mediated inhibition of DNases protects circulating cell-free DNA from ex vivo degradation in blood samples. Clin Biochem. 2015; 48:976–81.
Article
69. Li D, Kusko R, Ning B, Tong W, Johann DJ Jr, Xu J. FDA-led consortium studies advance quality control of targeted next generation sequencing assays for precision oncology. Precis Cancer Med. 2021; 4:32.
Article
70. Gong B, Li D, Kusko R, Novoradovskaya N, Zhang Y, Wang S, et al. Cross-oncopanel study reveals high sensitivity and accuracy with overall analytical performance depending on genomic regions. Genome Biol. 2021; 22:109.
71. Deveson IW, Gong B, Lai K, LoCoco JS, Richmond TA, Schageman J, et al. Evaluating the analytical validity of circulating tumor DNA sequencing assays for precision oncology. Nat Biotechnol. 2021; 39:1115–28.
Article
72. Zhang Y, Blomquist TM, Kusko R, Stetson D, Zhang Z, Yin L, et al. Deep oncopanel sequencing reveals within block position-dependent quality degradation in FFPE processed samples. Genome Biol. 2022; 23:141.
Article
73. Willey JC, Morrison TB, Austermiller B, Crawford EL, Craig DJ, Blomquist TM, et al. Advancing NGS quality control to enable measurement of actionable mutations in circulating tumor DNA. Cell Rep Methods. 2021; 1:100106.
Article
74. U.S. Food Drug Administration. Drug Approvals and database [Internet]. Silver Spring MD: U.S. Food and Drug Administration;2020. [cited 2020 Oct 10]. Available from: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-liquid-biopsy-ngs-companion-diagnostic-test-multiple-cancers-and-biomarkers.
75. Duffy MJ, Crown J. Use of circulating tumour DNA (ctDNA) for measurement of therapy predictive biomarkers in patients with cancer. J Pers Med. 2022; 12:99.
Article
76. Ettinger DS, Wood DE, Aisner DL, Akerley W, Bauman JR, Bharat A, et al. NCCN guidelines insights: non-small cell lung cancer, version 2.2021. J Natl Compr Canc Netw. 2021; 19:254–66.
77. Corcoran RB. Liquid biopsy versus tumor biopsy for clinical-trial recruitment. Nat Med. 2020; 26:1815–6.
Article
78. Ghosh RK, Pandey T, Dey P. Liquid biopsy: a new avenue in pathology. Cytopathology. 2019; 30:138–43.
Article
79. Marrugo-Ramirez J, Mir M, Samitier J. Blood-based cancer biomarkers in liquid biopsy: a promising non-invasive alternative to tissue biopsy. Int J Mol Sci. 2018; 19:2877.
Article
80. Chae YK, Oh MS. Detection of minimal residual disease using ctDNA in lung cancer: current evidence and future directions. J Thorac Oncol. 2019; 14:16–24.
Article
81. Moding EJ, Nabet BY, Alizadeh AA, Diehn M. Detecting liquid remnants of solid tumors: circulating tumor DNA minimal residual disease. Cancer Discov. 2021; 11:2968–86.
Article
82. Pellini B, Chaudhuri AA. Circulating tumor DNA minimal residual disease detection of non-small-cell lung cancer treated with curative intent. J Clin Oncol. 2022; 40:567–75.
Article
83. Parikh AR, Van Seventer EE, Siravegna G, Hartwig AV, Jaimovich A, He Y, et al. Minimal residual disease detection using a plasma-only circulating tumor DNA assay in patients with colorectal cancer. Clin Cancer Res. 2021; 27:5586–94.
84. Menikoff J, Kaneshiro J, Pritchard I. The common rule, updated. N Engl J Med. 2017; 376:613–5.
Article
85. Dresser R. Research ethics. Aligning regulations and ethics in human research. Science. 2012; 337:527–8.
86. Detsky AS. Sources of bias for authors of clinical practice guidelines. CMAJ. 2006; 175:1033–5.
Article
87. Sturgeon CM, Diamandis E. Laboratory medicine practice guidelines. Use of tumor markers in clinical practice: quality requirements. Washington, DC: National Academy of Clinical Biochemistry;2009.
Full Text Links
  • CRT
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr