Skip to main content
Log in

Detection of Solid Tumor Molecular Residual Disease (MRD) Using Circulating Tumor DNA (ctDNA)

  • Review Article
  • Published:
Molecular Diagnosis & Therapy Aims and scope Submit manuscript

Abstract

Circulating tumor DNA (ctDNA) is a component of cell-free DNA that is shed by malignant tumors into the bloodstream and other bodily fluids. Levels of ctDNA are typically low, particularly in patients with localized disease, requiring highly sophisticated methods for detection and quantification. Multiple liquid biopsy methods have been developed for ctDNA analysis in solid tumor malignancies and are now enabling detection and assessment of earlier stages of disease, post-treatment molecular residual disease (MRD), resistance to targeted systemic therapy, and tumor mutational burden. Understanding ctDNA biology, mechanisms of release, and clearance and size characteristics, in conjunction with the application of molecular barcoding and targeted error correction, have increased the sensitivity and specificity of ctDNA detection techniques. Combinatorial approaches including integration of ctDNA data with circulating protein biomarkers may further improve assay sensitivity and broaden the scope of ctDNA applications. Circulating viral DNA may be utilized to monitor disease in some virally induced malignancies. In spite of increasingly accurate methods of ctDNA detection, results need to be interpreted with caution given that somatic mosaicisms such as clonal hematopoiesis of indeterminate potential (CHIP) may give rise to genetic variants in the bloodstream unrelated to solid tumors, and the limited concordance observed between different commercial platforms. Overall, highly precise ctDNA detection and quantification methods have the potential to transform clinical practice via non-invasive monitoring of solid tumor malignancies, residual disease detection at earlier timepoints than standard clinical and/or imaging surveillance, and treatment personalization based on real-time assessment of the tumor genomic landscape.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1: ctDNA release into the bloodstream from solid tumors.
Fig. 2: Potential diagnostic and therapeutic approaches for solid tumors based on ctDNA detection and oncogenomic profiling.
Fig. 3: Trade-offs between sequencing breadth, depth, cost, and limit of detection.
Fig. 4: ctDNA MRD as a potential tool for adjuvant therapy clinical decision-making.

Similar content being viewed by others

References

  1. Mandel P, Métais P. Les acides nucléiques du plasma sanguin chez l’ homme. C R Seances Soc Biol Fil. 1948;142:241–3.

    CAS  PubMed  Google Scholar 

  2. Vietsch EE, Wellstein A. Circulating DNA in cancer diagnosis and prognosis. In: Dammacco F, Silvestris F, editors. Oncogenomics. London: Elsevier Inc.; 2019.

    Google Scholar 

  3. Botezatu I, Serdyuk O, Potapova G, et al. Genetic analysis of DNA excreted in urine: a new approach for detecting specific genomic DNA sequences from cells dying in an organism. Clin Chem. 2000;46:1078–84.

    CAS  PubMed  Google Scholar 

  4. Chan KCA, Leung SF, Yeung SW, et al. Quantitative analysis of the transrenal excretion of circulating EBV DNA in nasopharyngeal carcinoma patients. Clin Cancer Res. 2008;14:4809–13.

    Article  CAS  PubMed  Google Scholar 

  5. Burnham P, Dadhania D, Heyang M, et al. Urinary cell-free DNA is a versatile analyte for monitoring infections of the urinary tract. Nat Commun. 2018;9:1–10.

    Article  CAS  Google Scholar 

  6. Dudley JC, Schroers-Martin J, Lazzareschi DV, et al. Detection and surveillance of bladder cancer using urine tumor DNA. Cancer Discov. 2018. https://doi.org/10.1158/2159-8290.CD-18-0825.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Springer SU, Chen C-H, Rodriguez Pena MDC, et al. Non-invasive detection of urothelial cancer through the analysis of driver gene mutations and aneuploidy. Elife. 2018;7:1–27.

    Google Scholar 

  8. Lu T, Li J. Clinical applications of urinary cell-free DNA in cancer: current insights and promising future. Am J Cancer Res. 2017;7:2318–32.

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Wang Y, Springer S, Zhang M, et al. Detection of tumor-derived DNA in cerebrospinal fluid of patients with primary tumors of the brain and spinal cord. Proc Natl Acad Sci USA. 2015;112:9704–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Pan W, Gu W, Nagpal S, et al. Brain tumor mutations detected in cerebral spinal fluid. Clin Chem. 2015;61:514–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. De Mattos-Arruda L, Mayor R, Ng CKY, et al. Cerebrospinal fluid-derived circulating tumour DNA better represents the genomic alterations of brain tumours than plasma. Nat Commun. 2015;6:1–6.

    Google Scholar 

  12. Sriram KB, Relan V, Clarke BE, et al. Pleural fluid cell-free DNA integrity index to identify cytologically negative malignant pleural effusions including mesotheliomas. BMC Cancer. 2012;12:1–12.

    Article  CAS  Google Scholar 

  13. Soh J, Toyooka S, Aoe K, et al. Usefulness of EGFR mutation screening in pleural fluid to predict the clinical outcome of gefitinib treated patients with lung cancer. Int J Cancer. 2006;119:2353–8.

    Article  CAS  PubMed  Google Scholar 

  14. Husain H, Nykin D, Bui N, et al. Cell-free DNA from ascites and pleural effusions: molecular insights into genomic aberrations and disease biology. Mol Cancer Ther. 2017;16:948–55.

    Article  CAS  PubMed  Google Scholar 

  15. Mithani SK, Smith IM, Zhou S, et al. Mitochondrial resequencing arrays detect tumor-specific mutations in salivary rinses of patients with head and neck cancer. Clin Cancer Res. 2007;13:7335–40.

    Article  CAS  PubMed  Google Scholar 

  16. Wei F, Lin C-C, Joon A, et al. Noninvasive saliva-based EGFR gene mutation detection in patients with lung cancer. Am J Respir Crit Care Med. 2014;190:1117–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Jahr S, Hentze H, Englisch S, 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.

    CAS  PubMed  Google Scholar 

  18. Choi JJ, Reich CF, Pisetsky DS. The role of macrophages in the in vitro generation of extracellular DNA from apoptotic and necrotic cells. Immunology. 2005;115:55–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Chaudhuri AA, Binkley MS, Osmundson EC, et al. Predicting radiotherapy responses and treatment outcomes through analysis of circulating tumor DNA. Semin Radiat Oncol. 2015;25:305–12.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Thierry AR, El Messaoudi S, Gahan PB, et al. Origins, structures, and functions of circulating DNA in oncology. Cancer Metastasis Rev. 2016;35:347–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Stroun M, Lyautey J, Lederrey C, et al. About the possible origin and mechanism of circulating DNA apoptosis and active DNA release. Ann N Y Acad Sci. 2006;906:161–8.

    Article  Google Scholar 

  22. Anker P, Stroun M, Maurice PA. Spontaneous extracellular synthesis of DNA released by human blood lymphocytes. Cancer Res. 1976;36:2832–9.

    CAS  PubMed  Google Scholar 

  23. Bronkhorst AJ, Wentzel JF, Aucamp J, et al. Characterization of the cell-free DNA released by cultured cancer cells. Biochim Biophys Acta. 2016;1863:157–65.

    Article  CAS  PubMed  Google Scholar 

  24. Tsumita T, Iwanaga M. Fate of injected deoxyribonucleic acid in mice. Nature. 1963;198:1088–9.

    Article  CAS  PubMed  Google Scholar 

  25. Korabecna M, Opatrna S, Wirth J, et al. Cell-free plasma DNA during peritoneal dialysis and hemodialysis and in patients with chronic kidney disease. Ann N Y Acad Sci. 2008;1137:296–301.

    Article  CAS  PubMed  Google Scholar 

  26. Lo YM, Zhang J, Leung TN, et al. Rapid clearance of fetal DNA from maternal plasma. Am J Hum Genet. 1999;64:218–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Tamkovich SN, Cherepanova AV, Kolesnikova EV, et al. Circulating DNA and DNase activity in human blood. Ann N Y Acad Sci. 2006;1075:191–6.

    Article  CAS  PubMed  Google Scholar 

  28. Yao W, Mei C, Nan X, et al. Evaluation and comparison of in vitro degradation kinetics of DNA in serum, urine and saliva: a qualitative study. Gene. 2016;590:142–8.

    Article  CAS  PubMed  Google Scholar 

  29. Elshimali YI, Khaddour H, Sarkissyan M, et al. The clinical utilization of circulating cell free DNA (CCFDNA) in blood of cancer patients. Int J Mol Sci. 2013;14:18925–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Lui YYN, Chik KW, Chiu RWK, et al. Predominant hematopoietic origin of cell-free DNA in plasma and serum after sex-mismatched bone marrow transplantation. Clin Chem. 2002;48:421–7.

    CAS  PubMed  Google Scholar 

  31. Snyder MW, Kircher M, Hill AJ, et al. Cell-free DNA comprises an in vivo nucleosome footprint that informs its tissues-of-origin. Cell. 2016;164:57–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. 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 

  33. Lehmann-Werman R, Neiman D, Zemmour H, et al. Identification of tissue-specific cell death using methylation patterns of circulating DNA. Proc Natl Acad Sci USA. 2016;113:E1826–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Chang CPY, Chia RH, Wu TL, et al. Elevated cell-free serum DNA detected in patients with myocardial infarction. Clin Chim Acta. 2003;327:95–101.

    Article  CAS  PubMed  Google Scholar 

  35. Bustamante A, Mancha F, Macher HC, et al. Circulating cell-free DNA is a predictor of short-term neurological outcome in stroke patients treated with intravenous thrombolysis. J Circ Biomarkers. 2016;5:1–6.

    Article  CAS  Google Scholar 

  36. O’Connell GC, Petrone AB, Tennant CS, et al. Circulating extracellular DNA levels are acutely elevated in ischaemic stroke and associated with innate immune system activation. Brain Inj. 2017;31:1369–75.

    Article  PubMed  Google Scholar 

  37. De Vlaminck I, Martin L, Kertesz M, et al. Noninvasive monitoring of infection and rejection after lung transplantation. Proc Natl Acad Sci USA. 2015;112:13336–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Lo YMD, Rainer TH, Chan LYS, et al. Plasma DNA as a prognostic marker in trauma patients. Clin Chem. 2000;46:319–23.

    CAS  PubMed  Google Scholar 

  39. Weerakoon KG, McManus DP. Cell-Free DNA as a diagnostic tool for human parasitic infections. Trends Parasitol. 2016;32:378–91.

    Article  CAS  PubMed  Google Scholar 

  40. Margraf S, Lögters T, Reipen J, et al. Neutrophil-derived circulating free DNA (CF-DNA/NETs): a potential prognostic marker for posttraumatic development of inflammatory second hit and sepsis. Shock. 2008;30:352–8.

    Article  CAS  PubMed  Google Scholar 

  41. Zhong XY, Von Mühlenen I, Li Y, et al. Increased concentrations of antibody-bound circulatory cell-free DNA in rheumatoid arthritis. Clin Chem. 2007;53:1609–14.

    Article  CAS  PubMed  Google Scholar 

  42. Tug S, Helmig S, Menke J, et al. Correlation between cell free DNA levels and medical evaluation of disease progression in systemic lupus erythematosus patients. Cell Immunol. 2014;292:32–9.

    Article  CAS  PubMed  Google Scholar 

  43. Lo D, Sargent IL, Redman CW, et al. Presence of fetal DNA in maternal plasma and serum. Lancet. 1997;350:485–7.

    Article  CAS  PubMed  Google Scholar 

  44. Tug S, Helmig S, Deichmann ER, et al. Exercise-induced increases in cell free DNA in human plasma originate predominantly from cells of the haematopoietic lineage. Exerc Immunol Rev. 2015;21:164–73.

    PubMed  Google Scholar 

  45. Atamaniuk J, Vidotto C, Tschan H, et al. Increased concentrations of cell-free plasma DNA after exhaustive exercise. Clin Chem. 2004;50:1664–8.

    Article  CAS  Google Scholar 

  46. Breitbach S, Tug S, Simon P. Circulating cell-free DNA: an up-coming molecular marker in exercise physiology. Sport Med. 2012;42:565–86.

    Article  Google Scholar 

  47. Corcoran RB, Chabner BA. Application of cell-free DNA analysis to cancer treatment. N Engl J Med. 2018;379:1754–65.

    Article  CAS  PubMed  Google Scholar 

  48. Lo YMD, Chan KCA, Sun H, et al. Maternal plasma DNA sequencing reveals the genome-wide genetic and mutational profile of the fetus. Sci Transl Med. 2010;2:1–13.

    Article  CAS  Google Scholar 

  49. Chandrananda D, Thorne NP, Bahlo M. High-resolution characterization of sequence signatures due to non-random cleavage of cell-free DNA. BMC Med Genom. 2015;8:1–19.

    Article  CAS  Google Scholar 

  50. Jiang P, Lo YMD. The long and short of circulating cell-free DNA and the ins and outs of molecular diagnostics. Trends Genet. 2016;32:360–71.

    Article  CAS  PubMed  Google Scholar 

  51. Underhill HR, Kitzman JO, Hellwig S, et al. Fragment length of circulating tumor DNA. PLoS Genet. 2016;12:1–24.

    Article  CAS  Google Scholar 

  52. Mouliere F, El Messaoudi S, Gongora C, et al. Circulating cell-free DNA from colorectal cancer patients may reveal high KRAS or BRAF mutation load. Transl Oncol. 2013;6:319–28.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Mouliere F, Robert B, Peyrotte E, et al. High fragmentation characterizes tumour-derived circulating DNA. PLoS One. 2011;6:e23418.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Mouliere F, Chandrananda D, Piskorz AM, et al. Enhanced detection of circulating tumor DNA by fragment size analysis. Sci Transl Med. 2018;10:1–14.

    Article  CAS  Google Scholar 

  55. Yu SCY, Chan KCA, Zheng YWL, et al. Size-based molecular diagnostics using plasma DNA for noninvasive prenatal testing. Proc Natl Acad Sci USA. 2014;111:8583–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Lun FM, Tsui NB, Chan KA, et al. Noninvasive prenatal diagnosis of monogenic diseases by digital size selection and relative mutation dosage on DNA in maternal plasma. Proc. Natl. Acad. Sci. 2008;105:19920–19925.

    Article  PubMed  PubMed Central  Google Scholar 

  57. Minarik G, Repiska G, Hyblova M, et al. Utilization of benchtop next generation sequencing platforms ion torrent PGM and miseq in noninvasive prenatal testing for chromosome 21 trisomy and testing of impact of in silico and physical size selection on its analytical performance. PLoS One. 2015;10:1–12.

    Google Scholar 

  58. Ulz P, Thallinger GG, Auer M, et al. Inferring expressed genes by whole-genome sequencing of plasma DNA. Nat Genet. 2016;48:1273–8.

    Article  CAS  PubMed  Google Scholar 

  59. Stroun M, Anker P, Lyautey J, et al. Isolation and characterization of DNA from the plasma of cancer patients. Eur J Cancer Clin Oncol. 1987;23:707–12.

    Article  CAS  PubMed  Google Scholar 

  60. Wan JCM, Massie C, Garcia-Corbacho J, et al. Liquid biopsies come of age: Towards implementation of circulating tumour DNA. Nat Rev Cancer. 2017;17:223–38.

    Article  CAS  PubMed  Google Scholar 

  61. Heitzer E, Haque IS, Roberts CES, et al. Current and future perspectives of liquid biopsies in genomics-driven oncology. Nat Rev Genet. 2019;20(2):71–88.

    Article  CAS  PubMed  Google Scholar 

  62. Sirera R, Bremnes RM, Cabrera A, et al. Circulating DNA is a useful prognostic factor in patients with advanced non-small cell lung cancer. J Thorac Oncol. 2011;6:286–90.

    Article  PubMed  Google Scholar 

  63. Chaudhuri AA, Chabon JJ, Lovejoy AF, et al. Early detection of molecular residual disease in localized lung cancer by circulating tumor DNA profiling. Cancer Discov. 2017;7:1394–403.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Abbosh C, Birkbak NJ, Wilson GA, et al. Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution. Nature. 2017;545:446–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Nong J, Gong Y, Guan Y, et al. Circulating tumor DNA analysis depicts subclonal architecture and genomic evolution of small cell lung cancer. Nat Commun. 2018;9:1–8.

    Article  CAS  Google Scholar 

  66. Diehl F, Schmidt K, Choti MA, et al. Circulating mutant DNA to assess tumor dynamics. Nat Med. 2008;14:985–90.

    Article  CAS  PubMed  Google Scholar 

  67. To EWH, Chan KCA, Leung S, et al. Rapid clearance of plasma Epstein-Barr virus DNA after surgical treatment of nasopharyngeal carcinoma. Clin Cancer Res. 2003;9:3254–9.

    CAS  PubMed  Google Scholar 

  68. O’Leary B, Cutts RJ, Liu Y, et al. The genetic landscape and clonal evolution of breast cancer resistance to palbociclib plus fulvestrant in the PALOMA-3 trial. Cancer Discov. 2018;8:1390–403.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Bettegowda C, Sausen M, Leary RJ, et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med. 2014;6:224ra24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Newman AM, Bratman SV, To J, et al. An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage. Nat Med. 2014;20:548–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Chung JH, Pavlick D, Hartmaier R, et al. Hybrid capture-based genomic profiling of circulating tumor DNA from patients with estrogen receptor-positive metastatic breast cancer. Ann Oncol. 2017;28:2866–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Scherer F, Kurtz DM, Newman AM, et al. Distinct biological subtypes and patterns of genome evolution in lymphoma revealed by circulating tumor DNA. Sci Transl Med. 2016;8:1–11.

    Article  CAS  Google Scholar 

  73. Phallen J, Sausen M, Adleff V, et al. Direct detection of early-stage cancers using circulating tumor DNA. Sci Transl Med. 2017;9:1–12.

    Article  CAS  Google Scholar 

  74. Cohen JD, Javed AA, Thoburn C, et al. Combined circulating tumor DNA and protein biomarker-based liquid biopsy for the earlier detection of pancreatic cancers. Proc Natl Acad Sci USA. 2017;114:10203–7.

    Google Scholar 

  75. Chabon JJ, Simmons AD, Lovejoy AF, et al. Circulating tumour DNA profiling reveals heterogeneity of EGFR inhibitor resistance mechanisms in lung cancer patients. Nat Commun. 2016;7:1–14.

    Google Scholar 

  76. Khan KH, Cunningham D, Werner B, et al. Longitudinal liquid biopsy and mathematical modeling of clonal evolution forecast time to treatment failure in the PROSPECT-C phase II colorectal cancer clinical trial. Cancer Discov. 2018;8(10):1270–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Oxnard GR, Thress KS, Alden RS, et al. Association between plasma genotyping and outcomes of treatment with osimertinib (AZD9291) in advanced non-small-cell lung cancer. J Clin Oncol. 2016;34:3375–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Thress KS, Paweletz CP, Felip E, et al. Acquired EGFR C797S mutation mediates resistance to AZD9291 in non-small cell lung cancer harboring EGFR T790M. Nat Med. 2015;21:560–2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Murtaza M, Dawson SJ, Tsui DWY, et al. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature. 2013;497:108–12.

    Article  CAS  PubMed  Google Scholar 

  80. Taniguchi K, Uchida J, Nishino K, et al. Quantitative detection of EGFR mutations in circulating tumor DNA derived from lung adenocarcinomas. Clin Cancer Res. 2011;17:7808–15.

    Article  CAS  PubMed  Google Scholar 

  81. Tie J, Cohen JD, Wang Y, et al. Serial circulating tumour DNA analysis during multimodality treatment of locally advanced rectal cancer: a prospective biomarker study. Gut. 2019;68(4):663–71. https://doi.org/10.1136/gutjnl-2017-315852.

    Article  CAS  PubMed  Google Scholar 

  82. Tie J, Wang Y, Tomasetti C, et al. Circulating tumor DNA analysis detects minimal residual disease and predicts recurrence in patients with stage II colon cancer. Sci Transl Med. 2016;8:1–10.

    Article  CAS  Google Scholar 

  83. Garcia-Murillas I, Schiavon G, Weigelt B, et al. Mutation tracking in circulating tumor DNA predicts relapse in early breast cancer. Sci Transl Med. 2015;7:1–7.

    Article  Google Scholar 

  84. Newman AM, Lovejoy AF, Klass DM, et al. Integrated digital error suppression for improved detection of circulating tumor DNA. Nat Biotechnol. 2016;34:547–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Dawson S-J, Tsui DWY, Murtaza M, et al. Analysis of circulating tumor DNA to monitor metastatic breast cancer. N Engl J Med. 2013;368:1199–209.

    Article  CAS  PubMed  Google Scholar 

  86. Kurtz DM, Scherer F, Jin MC, et al. Circulating tumor DNA measurements as early outcome predictors in diffuse large B-cell lymphoma. J Clin Oncol. 2018;36(28):2845–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Sausen M, Phallen J, Adleff V, et al. Clinical implications of genomic alterations in the tumour and circulation of pancreatic cancer patients. Nat Commun. 2015;6:1–6.

    Article  Google Scholar 

  88. Endesfelder D, Math D, Gronroos E, et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med. 2012;366:883–92.

    Article  PubMed  PubMed Central  Google Scholar 

  89. De Mattos-Arruda L, Weigelt B, Cortes J, et al. Capturing intra-tumor genetic heterogeneity by de novo mutation profiling of circulating cell-free tumor DNA: a proof-of-principle. Ann Oncol. 2014;25:1729–35.

    Article  PubMed  PubMed Central  Google Scholar 

  90. Jamal-Hanjani M, Wilson GA, McGranahan N, et al. Tracking the evolution of non–small-cell lung cancer. N Engl J Med. 2017;376:2109–21.

    Article  CAS  PubMed  Google Scholar 

  91. Abbosh C, Birkbak NJ, Swanton C. Early stage NSCLC: challenges to implementing ctDNA-based screening and MRD detection. Nat Rev Clin Oncol. 2018;15(9):577–86.

    Article  CAS  PubMed  Google Scholar 

  92. Swanton C, Venn O, Aravanis A, et al. Prevalence of clonal hematopoiesis of indeterminate potential (CHIP) measured by an ultra-sensitive sequencing assay: exploratory analysis of the Circulating Cancer Genome Atlas (CCGA) study. J Clin Oncol. 2018;36:12003.

    Article  Google Scholar 

  93. Hu Y, Ulrich BC, Supplee J, et al. False-positive plasma genotyping due to clonal hematopoiesis. Clin Cancer Res. 2018;24:4437–43.

    Article  CAS  PubMed  Google Scholar 

  94. Genovese G, Kähler AK, Handsaker RE, et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N Engl J Med. 2014;371:2477–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Jaiswal S, Fontanillas P, Flannick J, et al. Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med. 2014;371:2488–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Jaiswal S, Ebert BL. MDS is a stem cell disorder after all. Cancer Cell. 2014;25:713–4.

    Article  CAS  PubMed  Google Scholar 

  97. Potter NE, Greaves M. Cancer: persistence of leukaemic ancestors. Nature. 2014;506:300–1.

    Article  CAS  PubMed  Google Scholar 

  98. Damm F, Mylonas E, Cosson A, et al. Acquired initiating mutations in early hematopoietic cells of CLL patients. Cancer Discov. 2014;4:1088–101.

    Article  CAS  PubMed  Google Scholar 

  99. Chung SS, Kim E, Park JH, et al. BRAFV600E mutations in hematopoietic stem cells of hairy cell leukemia patients suggests a stem cell origin. Sci Transl Med. 2014;99:404–5.

    Google Scholar 

  100. Steensma DP, Bejar R, Jaiswal S, et al. Perspective: clonal hematopoiesis of indeterminate potential and its distinction from myelodysplastic syndromes. Blood. 2018;126:9–17.

    Article  CAS  Google Scholar 

  101. Acuna-Hidalgo R, Sengul H, Steehouwer M, et al. Ultra-sensitive sequencing identifies high prevalence of clonal hematopoiesis-associated mutations throughout adult life. Am J Hum Genet. 2017;101:50–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Zink F, Stacey SN, Norddahl GL, et al. Clonal hematopoiesis, with and without candidate driver mutations, is common in the elderly. Blood. 2017;130:742–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Young AL, Challen GA, Birmann BM, et al. Clonal haematopoiesis harbouring AML-associated mutations is ubiquitous in healthy adults. Nat Commun. 2016;22(7):12484.

    Article  CAS  Google Scholar 

  104. Xie M, Lu C, Wang J, et al. Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nat Med. 2014;20:1472–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. McKerrell T, Park N, Moreno T, et al. Leukemia-associated somatic mutations drive distinct patterns of age-related clonal hemopoiesis. Cell Rep. 2015;10:1239–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Coombs CC, Zehir A, Devlin SM, et al. Therapy-related clonal hematopoiesis in patients with non-hematologic cancers is common and associated with adverse clinical outcomes. Cell Stem Cell. 2017;21(374–382):e4.

    Google Scholar 

  107. Bailey MH, Tokheim C, Porta-Pardo E, et al. Comprehensive characterization of cancer driver genes and mutations. Cell. 2018;173(371–385):e18.

    Google Scholar 

  108. Zill OA, Banks KC, Fairclough SR, et al. The landscape of actionable genomic alterations in cell-free circulating tumor DNA from 21,807 advanced cancer patients. Clin Cancer Res. 2018;24:3528–38.

    Article  CAS  PubMed  Google Scholar 

  109. McCulloch SD, Kunkel TA. The fidelity of DNA synthesis by eukaryotic replicative and translesion synthesis polymerases. Cell Res. 2008;18:148–61.

    Article  CAS  PubMed  Google Scholar 

  110. Lynch M. Rate, molecular spectrum, and consequences of human mutation. Proc Natl Acad Sci USA. 2010;107:961–8.

    Article  PubMed  PubMed Central  Google Scholar 

  111. Kinde I, Wu J, Papadopoulos N, et al. Detection and quantification of rare mutations with massively parallel sequencing. Proc Natl Acad Sci USA. 2011;108:9530–5.

    Article  PubMed  PubMed Central  Google Scholar 

  112. Newton CR, Graham A, Heptinstall LE, et al. Analysis of any point mutation in DNA. The amplification refractory mutation system (ARMS). Nucleic Acids Res. 1989;17:2503–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Wu DY, Ugozzoli L, Pal BK, et al. Allele-specific enzymatic amplification of beta-globin genomic DNA for diagnosis of sickle cell anemia. Proc Natl Acad Sci USA. 1989;86:2757–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Kimura H, Kasahara K, Kawaishi M, et al. Detection of epidermal growth factor receptor mutations in serum as a predictor of the response to gefitinib in patients with non-small-cell lung cancer. Clin Cancer Res. 2006;12:3915–21.

    Article  CAS  PubMed  Google Scholar 

  115. Khakoo S, Georgiou A, Gerlinger M, et al. Circulating tumour DNA, a promising biomarker for the management of colorectal cancer. Crit Rev Oncol Hematol. 2018;122:72–82.

    Article  PubMed  Google Scholar 

  116. Milbury C, Li J, Makrigiorgos G. PCR-based methods for the enrichment of minority alleles and mutations. Clin Chem. 2009;55:632–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Vogelstein B, Kinzler KW. Digital PCR. Genetics. 1999;96:9236–41.

    CAS  Google Scholar 

  118. Gerdes L, Iwobi A, Busch U, et al. Optimization of digital droplet polymerase chain reaction for quantification of genetically modified organisms. Biomol Detect Quantif. 2016;7:9–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Hindson BJ, Ness KD, Masquelier DA, et al. High-throughput droplet digital PCR system for absolute quantitation of DNA copy number. Anal Chem. 2011;83:8604–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Karlovich C, Goldman JW, Sun JM, et al. Assessment of EGFR mutation status in matched plasma and tumor tissue of NSCLC patients from a phase I study of rociletinib (CO-1686). Clin Cancer Res. 2016;22:2386–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Liu J, Huang S, Sun M, et al. An improved allele-specific PCR primer design method for SNP marker analysis and its application. Plant Methods. 2012;8:1–9.

    Article  CAS  Google Scholar 

  122. Kwok S, Kellogg DE, Mckinney N, et al. Effects of primer-template mismatches on the polymerase chain reaction: human immunodeficiency virus type 1 model studies. Nucleic Acids Res. 1990;18:999–1005.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Cha RS, Zarbl H, Keohavong P, et al. Mismatch amplification mutation assay (MAMA): application to the c-H-ras gene. Genome Res. 1992;2:14–20.

    Article  CAS  Google Scholar 

  124. Huggett JF, Whale A. Digital PCR as a novel technology and its potential implications for molecular diagnostics. Clin Chem. 2013;59:1691–3.

    Article  CAS  PubMed  Google Scholar 

  125. Taly V, Pekin D, El Abed A, et al. Detecting biomarkers with microdroplet technology. Trends Mol Med. 2012;18:405–16.

    Article  CAS  PubMed  Google Scholar 

  126. Dressman D, Yan H, Traverso G, et al. Transforming single DNA molecules into fluorescent magnetic particles for detection and enumeration of genetic variations. Proc Natl Acad Sci USA. 2003;100:8817–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Nakano M, Komatsu J, Matsuura SI, et al. Single-molecule PCR using water-in-oil emulsion. J Biotechnol. 2003;102:117–24.

    Article  CAS  PubMed  Google Scholar 

  128. Diehl F, Li M, He Y, et al. BEAMing: single-molecule PCR on microparticles in water-in-oil emulsions. Nat Methods. 2006;3:551–9.

    Article  CAS  PubMed  Google Scholar 

  129. Taly V, Pekin D, Benhaim L, et al. Multiplex picodroplet digital PCR to detect KRAS mutations in circulating DNA from the plasma of colorectal cancer patients. Clin Chem. 2013;59:1722–31.

    Article  CAS  PubMed  Google Scholar 

  130. Zhu G, Ye X, Dong Z, et al. Highly sensitive droplet digital PCR method for detection of EGFR-activating mutations in plasma cell-free DNA from patients with advanced non-small cell lung cancer. J Mol Diagnostics. 2015;17:265–72.

    Article  CAS  Google Scholar 

  131. Yung TKF, Chan KCA, Mok TSK, et al. Single-molecule detection of epidermal growth factor receptor mutations in plasma by microfluidics digital PCR in non-small cell lung cancer patients. Clin Cancer Res. 2009;15:2076–84.

    Article  CAS  PubMed  Google Scholar 

  132. Sacher AG, Paweletz C, Dahlberg SE, et al. Prospective validation of rapid plasma genotyping as a sensitive and specific tool for guiding lung cancer care. JAMA Oncol. 2016;2:1014–22.

    Article  PubMed  PubMed Central  Google Scholar 

  133. Diehl F, Li M, Dressman D, et al. Detection and quantification of mutations in the plasma of patients with colorectal tumors. Proc Natl Acad Sci USA. 2005;102:16368–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Schiavon G, Hrebien S, Garcia-Murillas I, et al. Analysis of ESR1 mutation in circulating tumor DNA demonstrates evolution during therapy for metastatic breast cancer. Sci Transl Med. 2015;7:148–57.

    Article  CAS  Google Scholar 

  135. Sanmamed MF, Fernandez-Landazuri S, Rodriguez C, et al. Quantitative cell-free circulating BRAFV600E mutation analysis by use of droplet digital PCR in the follow-up of patients with melanoma being treated with BRAF inhibitors. Clin Chem. 2015;61:297–304.

    Article  CAS  PubMed  Google Scholar 

  136. Birkenkamp-Demtröder K, Nordentoft I, Christensen E, et al. Genomic alterations in liquid biopsies from patients with bladder cancer. Eur Urol. 2016;70:75–82.

    Article  CAS  PubMed  Google Scholar 

  137. Christensen E, Nordentoft I, Vang S, et al. Optimized targeted sequencing of cell-free plasma DNA from bladder cancer patients. Sci Rep. 2018;8:1–11.

    Article  CAS  Google Scholar 

  138. Alizadeh AA, Aranda V, Bardelli A, et al. Toward understanding and exploiting tumor heterogeneity. Nat Med. 2015;21:846–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Heitzer E, Ulz P, Belic J, et al. Tumor-associated copy number changes in the circulation of patients with prostate cancer identified through whole-genome sequencing. Genome Med. 2013;5:1–16.

    Article  CAS  Google Scholar 

  140. Farris C, Trimarchi JM. Plasma-seq: a novel strategy for metastatic prostate cancer analysis. Genome Med. 2013;5:35.

    Article  PubMed  PubMed Central  Google Scholar 

  141. Kinde I, Papadopoulos N, Kinzler KW, et al. FAST-SeqS: a simple and efficient method for the detection of aneuploidy by massively parallel sequencing. PLoS One. 2012;7:1–8.

    Article  CAS  Google Scholar 

  142. Douville C, Springer S, Kinde I, et al. Detection of aneuploidy in patients with cancer through amplification of long interspersed nucleotide elements (LINEs). Proc Natl Acad Sci USA. 2018;115:1871–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Chan KCA, Jiang P, Zheng YWL, 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. 2013;59:211–24.

    Article  CAS  PubMed  Google Scholar 

  144. Welch JS, Larson DE, Wallis J, et al. Use of whole-genome sequencing to diagnose a cryptic fusion oncogene. JAMA. 2011;305:1577–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Leary RJ, Kinde I, Diehl F, et al. Development of personalized tumor biomarkers using massively parallel sequencing. Sci Transl Med. 2010;2:1–15.

    Article  CAS  Google Scholar 

  146. McKernan KJ, Peckham HE, Costa G, et al. Sequence and structural variation in a human genome uncovered by short-read, massively parallel ligation sequencing using two-base encoding. Genome Res. 2009;19:1527–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. McBride DJ, Orpana AK, Sotiriou C, et al. Use of cancer-specific genomic rearrangements to quantify disease burden in plasma from patients with solid tumors. Genes. Chromosomes Cancer. 2010;49:1062–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Kirkizlar E, Zimmermann B, Constantin T, et al. Detection of clonal and subclonal copy-number variants in cell-free DNA from patients with breast cancer using a massively multiplexed PCR methodology. Transl Oncol. 2015;8:407–16.

    Article  PubMed  PubMed Central  Google Scholar 

  149. Przybyl J, Chabon JJ, Spans L, et al. Combination approach for detecting different types of alterations in circulating tumor DNA in leiomyosarcoma. Clin Cancer Res. 2018;24:2688–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Leary RJ, Sausen M, Kinde I, et al. Detection of chromosomal alterations in the circulation of cancer patients with whole-genome sequencing. Sci Transl Med. 2012;4:162ra154.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Heitzer E, Auer M, Hoffmann EM, et al. Establishment of tumor-specific copy number alterations from plasma DNA of patients with cancer. Int J Cancer. 2013;133:346–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Belic J, Koch M, Ulz P, et al. Rapid identification of plasma DNA samples with increased ctDNA levels by a modified FAST-SeqS approach. Clin Chem. 2015;61:838–49.

    Article  CAS  PubMed  Google Scholar 

  153. Han X, Wang J, Sun Y. Circulating tumor DNA as biomarkers for cancer detection. Genom Proteom Bioinform. 2017;15:59–72.

    Article  Google Scholar 

  154. Choi M, Scholl UI, Ji W, et al. Genetic diagnosis by whole exome capture and massively parallel DNA sequencing. Proc Natl Acad Sci USA. 2009;106:19096–101.

    Article  PubMed  PubMed Central  Google Scholar 

  155. 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. 2012;4:136ra68.

    Article  CAS  PubMed  Google Scholar 

  156. Gale D, Plagnol V, Lawson A, et al. Analytical performance and validation of an enhanced TAm-Seq circulating tumor DNA sequencing assay [abstract]. In: AACR 107th annual meeting, 16–20 Apr 2016, New Orleans.

  157. Jamal-Hanjani M, Hackshaw A, Ngai Y, et al. Tracking genomic cancer evolution for precision medicine: the Lung TRACERx study. PLoS Biol. 2014;12:1–7.

    Article  CAS  Google Scholar 

  158. Gagan J, Van Allen EM. Next-generation sequencing to guide cancer therapy. Genome Med. 2015;7:1–10.

    Article  Google Scholar 

  159. Newman AM, Bratman SV, Stehr H, et al. FACTERA: a practical method for the discovery of genomic rearrangements at breakpoint resolution. Bioinformatics. 2014;30:3390–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Bratman SV, Newman AM, Alizadeh AA, et al. Potential clinical utility of ultrasensitive circulating tumor DNA detection with CAPP-Seq. Expert Rev Mol Diagn. 2015;15:715–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Lanman RB, Mortimer SA, Zill OA, et al. Analytical and clinical validation of a digital sequencing panel for quantitative, highly accurate evaluation of cell-free circulating tumor DNA. PLoS One. 2015;10:1–27.

    Article  CAS  Google Scholar 

  162. Odegaard JI, Vincent JJ, Mortimer S, et al. Validation of a plasma-based comprehensive cancer genotyping assay utilizing orthogonal tissue- and plasma-based methodologies. Clin Cancer Res. 2018;24:3539–49.

    Article  CAS  PubMed  Google Scholar 

  163. FoundationOne® Liquid. Foundation Medicing Inc.; 2018. https://www.foundationmedicine.com/genomic-testing/foundation-one-liquid.

  164. Torga G, Pienta KJ. Patient-paired sample congruence between 2 commercial liquid biopsy tests. JAMA Oncol. 2018;4:868–70.

    Article  PubMed  Google Scholar 

  165. Alexandrov LB, Nik-zainal S, Wedge DC, et al. Signatures of mutational processes in human cancer. Nature. 2013;500:415–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Amant F, Verheecke M, Wlodarska I, et al. Presymptomatic identification of cancers in pregnant women during noninvasive prenatal testing. JAMA Oncol. 2015;1:814–9.

    Article  PubMed  Google Scholar 

  167. Cristofanilli M, Turner NC, Bondarenko I, Ro J, Im S-A, Masuda N, Colleoni M, DeMichele A, Loi S, Verma S, Iwata H, Harbeck N, Zhang K, Theall KP, Jiang Y, Bartlett CH, Koehler M. Slamon D (2016) Fulvestrant plus palbociclib versus fulvestrant plus placebo for treatment of hormone-receptor-positive, HER2-negative metastatic breast cancer that progressed on previous endocrine therapy (PALOMA-3): final analysis of the multicentre, double-blind, phase 3 randomised controlled trial. The Lancet Oncology. 2016;17:425–39.

    Article  CAS  PubMed  Google Scholar 

  168. Roth A, Khattra J, Yap D, et al. PyClone: statistical inference of clonal population structure in cancer. Nat Methods. 2014;11:396–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Cohen JD, Li L, Wang Y, et al. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science. 2018;359:926–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Ettinger DS, Wood DE, Aisner DL, et al. Non-small cell lung cancer, version 5.2017, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Cancer Netw. 2017;15:504–35.

    Article  Google Scholar 

  171. Billè A, Ahmad U, Woo KM, et al. Detection of recurrence patterns after wedge resection for early stage lung cancer: rationale for radiologic follow-up. Ann Thorac Surg. 2016;102:1067–73.

    Article  PubMed  PubMed Central  Google Scholar 

  172. Kocak Z, Evans ES, Zhou SM, et al. Challenges in defining radiation pneumonitis in patients with lung cancer. Int J Radiat Oncol Biol Phys. 2005;62:635–8.

    Article  PubMed  Google Scholar 

  173. Huang K, Dahele M, Senan S, et al. Radiographic changes after lung stereotactic ablative radiotherapy (SABR)—can we distinguish recurrence from fibrosis? A systematic review of the literature. Radiother Oncol. 2012;102:335–42.

    Article  PubMed  Google Scholar 

  174. Hung JJ, Hsu WH, Hsieh CC, et al. Post-recurrence survival in completely resected stage I non-small cell lung cancer with local recurrence. Thorax. 2009;64:192–6.

    Article  PubMed  Google Scholar 

  175. Shepherd F, Pereira J, Ciuleanu T, et al. Erlotinib in previously treated non-small-cell lung cancer. N Engl J Med. 2005;353:123–32.

    Article  CAS  PubMed  Google Scholar 

  176. Pérez-Soler R, Chachoua A, Hammond LA, et al. Determinants of tumor response and survival with erlotinib in patients with non-small-cell lung cancer. J Clin Oncol. 2004;22:3238–47.

    Article  CAS  PubMed  Google Scholar 

  177. Gandara DR, Paul SM, Kowanetz M, et al. Blood-based tumor mutational burden as a predictor of clinical benefit in non-small-cell lung cancer patients treated with atezolizumab. Nat Med. 2018;24(9):1441–8.

    Article  CAS  PubMed  Google Scholar 

  178. Frampton GM, Fichtenholtz A, Otto GA, et al. Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nat Biotechnol. 2013;31:1023–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Fehrenbacher L, Spira A, Ballinger M, Kowanetz M, Vansteenkiste J, Mazieres J, Park K, Smith D, Artal-Cortes A, Lewanski C, Braiteh F, Waterkamp D, He P, Zou W, Chen DS, Yi J, Sandler A, Rittmeyer A. Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): a multicentre, open-label, phase 2 randomised controlled trial. The Lancet. 2016;387:1837–46.

    Article  CAS  Google Scholar 

  180. Rittmeyer A, Barlesi F, Waterkamp D, Park K, Ciardiello F, von Pawel J, Gadgeel SM, Hida T, Kowalski DM, Dols MC, Cortinovis DL, Leach J, Polikoff J, Barrios C, Kabbinavar F, Frontera OA, De Marinis F, Turna H, Lee J-S, Ballinger M, Kowanetz M, He P, Chen DS, Sandler Gandara DR. Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial. The Lancet. 2017;389:255–65.

    Article  Google Scholar 

  181. Goodman AM, Kato S, Bazhenova L, et al. Tumor mutational burden as an independent predictor of response to immunotherapy in diverse cancers. Mol Cancer Ther. 2017;16(11):2598–608.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Rizvi NA, Hellmann MD, Snyder A, et al. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348:124–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Hellmann MD, Ciuleanu T-E, Pluzanski A, et al. Nivolumab plus ipilimumab in lung cancer with a high tumor mutational burden. N Engl J Med. 2018;378:2093–104.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Hoeben KWJ, van Steenbergen LN, van de Wouw AJ, et al. Treatment and complications in elderly stage III colon cancer patients in the Netherlands. Ann Oncol. 2013;24:974–9.

    Article  CAS  PubMed  Google Scholar 

  185. Benson AB, Venook AP, Al-Hawary MM, et al. NCCN guidelines insights: colon cancer, version 2.2018. J Natl Compr Cancer Netw. 2018;16:359–69.

    Article  Google Scholar 

  186. Wirtzfeld DA, Mikula L, Gryfe R, et al. Concordance with clinical practice guidelines for adjuvant chemotherapy in patients with stage I–III colon cancer: experience in 2 Canadian provinces. Can J Surg. 2009;52:92–7.

    PubMed  PubMed Central  Google Scholar 

  187. Perini MV, Montagnini AL, Jukemura J, et al. Clinical and pathologic prognostic factors for curative resection for pancreatic cancer. HPB (Oxford). 2008;10:356–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Walboomers JMM, Jacobs MV, Manos MM, et al. Human papillomavirus is a necessary cause of invasive cervical cancer worldwide. J Pathol. 1999;189:12–9.

    Article  CAS  PubMed  Google Scholar 

  189. Muñoz N, Bosch FX, de Sanjosé S, et al. Epidemiologic classification of human papillomavirus types associated with cervical cancer. N Engl J Med. 2003;348:518–27.

    Article  PubMed  Google Scholar 

  190. Frisch M, Glimelius B, van den Brule AJC, et al. Sexually transmitted infection as a cause of anal cancer. N Engl J Med. 1997;337:1350–8.

    Article  CAS  PubMed  Google Scholar 

  191. Chaturvedi AK, Engels EA, Pfeiffer RM, et al. Human papillomavirus and rising oropharyngeal cancer incidence in the United States. J Clin Oncol. 2011;29:4294–301.

    Article  PubMed  PubMed Central  Google Scholar 

  192. zur Hausen H, Schulte-Holthausen H, Klein G, et al. EBV DNA in biopsies of Burkitt tumours and anaplastic carcinomas of the nasopharynx. Nature. 1970;228:1056–8.

    Article  CAS  PubMed  Google Scholar 

  193. Kim C-M, Koike K, Saito I, et al. HBx gene of hepatitis B virus induces liver cancer in transgenic mice. Nature. 1988;351:317–20.

    Article  Google Scholar 

  194. Beasley RP. Hepatitis B virus. The major etiology of hepatocellular carcinoma. Cancer. 2016;61:1942–56.

    Article  Google Scholar 

  195. Saito I, Miyamura T, Ohbayashi A, et al. Hepatitis C virus infection is associated with the development of hepatocellular carcinoma. Proc Natl Acad Sci USA. 1990;87:6547–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Moriya K, Fujie H, Shintani Y, et al. The core protein of hepatitis C virus induces hepatocellular carcinoma in transgenic mice. Nat Med. 1998;4:1065–7.

    Article  CAS  PubMed  Google Scholar 

  197. Peitsaro P, Johansson B, Syrjänen S. Integrated human papillomavirus type 16 is frequently found in cervical cancer precursors as demonstrated by a novel quantitative real-time PCR technique. J Clin Microbiol. 2002;40:886–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Wiest T, Schwarz E, Enders C, Flechtenmacher C, Bosch FX. Involvement of intact HPV16 E6/E7 gene expression in head and neck cancers with unaltered p53 status and perturbed pRb cell cycle control. Oncogene. 2002;21:1510–7.

    Article  CAS  PubMed  Google Scholar 

  199. Raab-Traub N. Epstein–Barr virus in the pathogenesis of NPC. Semin Cancer Biol. 2002;14:423–30.

    Google Scholar 

  200. Mayrand M-H, Duarte-Franco E, Rodrigues I, et al. Human papillomavirus DNA versus Papanicolaou screening tests for cervical cancer. New Engl. J Med. 2010;357:1579–1588.

    Article  Google Scholar 

  201. Manos MM, Kinney WK, Hurley LB, et al. Identifying women with cervical neoplasia using human papillomavirus DNA testing for equivocal Papanicolaou results. JAMA. 1999;281:1605–10.

    Article  CAS  PubMed  Google Scholar 

  202. Ndiaye C, Mena M, Alemany L, et al. HPV DNA, E6/E7 mRNA, and p16INK4a detection in head and neck cancers: a systematic review and meta-analysis. Lancet Oncol. 2014;15:1319–31.

    Article  CAS  PubMed  Google Scholar 

  203. Reimers N, Kasper HU, Weissenborn SJ, et al. Combined analysis of HPV-DNA, p16 and EGFR expression to predict prognosis in oropharyngeal cancer. Int J Cancer. 2007;120:1731–8.

    Article  CAS  PubMed  Google Scholar 

  204. Feinmesser R, Miyazaki I, Cheung R, et al. Diagnosis of nasopharyngeal carcinoma by DNA amplification of tissue obtained by fine-needle aspiration. N Engl J Med. 2015;326:17–21.

    Article  Google Scholar 

  205. Chan KCA, Hung ECW, Woo JKS, et al. Early detection of nasopharyngeal carcinoma by plasma Epstein–Barr virus DNA analysis in a surveillance program. Cancer. 2013;119:1838–44.

    Article  CAS  PubMed  Google Scholar 

  206. Chan KCA, Woo JKS, King A, et al. Analysis of plasma Epstein–Barr virus DNA to screen for nasopharyngeal cancer. N Engl J Med. 2017;377:513–22.

    Article  CAS  PubMed  Google Scholar 

  207. Lin J-C, Wang W-Y, Chen KY, et al. Quantification of plasma Epstein–Barr virus DNA in patients with advanced nasopharyngeal carcinoma. N Engl J Med. 2004;350:2461–70.

    Article  CAS  PubMed  Google Scholar 

  208. Campitelli M, Jeannot E, Peter M, et al. Human papillomavirus mutational insertion: specific marker of circulating tumor DNA in cervical cancer patients. PLoS One. 2012;7:e43393.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. Jeannot E, Becette V, Campitelli M, et al. Circulating human papillomavirus DNA detected using droplet digital PCR in the serum of patients diagnosed with early stage human papillomavirus-associated invasive carcinoma. J Pathol Clin Res. 2016;2:201–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Lo YMD, Leung S, Chan LYS, et al. Kinetics of plasma Epstein–Barr virus DNA during radiation therapy for nasopharyngeal carcinoma. J Cancer Res. 2000;60:2351–5.

    CAS  Google Scholar 

  211. Wang W-Y, Twu C-W, Chen H-H, et al. Long-term survival analysis of nasopharyngeal carcinoma by plasma Epstein–Barr virus DNA levels. Cancer. 2013;119:963–70.

    Article  CAS  PubMed  Google Scholar 

  212. Le Q, Zhang Q, Cao H, et al. An international collaboration to harmonize the quantitative plasma Epstein–Barr Virus (EBV) DNA assay for future biomarker-guided trials in nasopharyngeal carcinoma. Clin Cancer Res. 2013;19:2208–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  213. Leung SF, Zee B, Ma BB, et al. Plasma Epstein–Barr viral deoxyribonucleic acid quantitation complements tumor-node-metastasis staging prognostication in nasopharyngeal carcinoma. J Clin Oncol. 2006;24:5414–8.

    Article  CAS  PubMed  Google Scholar 

  214. Chua MLK, Wee JTS, Hui EP, et al. Nasopharyngeal carcinoma. Lancet. 2016;387:1012–24.

    Article  PubMed  Google Scholar 

  215. Fakhry C, Westra WH, Li S, et al. Improved survival of patients with human papillomavirus-positive head and neck squamous cell carcinoma in a prospective clinical trial. J Natl Cancer Inst. 2008;100:261–9.

    Article  CAS  PubMed  Google Scholar 

  216. Ang KK, Harris J, Wheeler R, et al. Human papillomavirus and survival of patients with oropharyngeal cancer. N Engl J Med. 2010;363:24–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  217. Dahlstrom KR, Li G, Hussey CS, et al. Circulating human papillomavirus DNA as a marker for disease extent and recurrence among patients with oropharyngeal cancer. Cancer. 2015;121:3455–64.

    Article  CAS  PubMed  Google Scholar 

  218. Hanna GJ, Supplee JG, Kuang Y, et al. Plasma HPV cell-free DNA monitoring in advanced HPV-associated oropharyngeal cancer. Ann Oncol. 2018;29:1980–6.

    Article  CAS  PubMed  Google Scholar 

  219. Gupta GP, Kumar S, Marron D, et al. Circulating tumor HPV16 DNA as a biomarker of tumor genomics and disease control in HPV-associated oropharyngeal squamous cell carcinoma [abstract]. Radiat Oncol. 2018;100(5):1310.

    Google Scholar 

  220. Koliopoulos G, Nyaga VN, Santesso N, et al. Cytology versus HPV testing for cervical cancer screening in the general population. Cochrane Database Syst Rev. 2017;8:CD008587.

    PubMed  Google Scholar 

  221. Koss LG. The Papanicolaou test for cervical cancer detection: a triumph and a tragedy. JAMA. 1989;261:737–43.

    Article  CAS  PubMed  Google Scholar 

  222. Han K, Leung E, Barbera L, et al. Circulating human papillomavirus DNA as a biomarker of response in patients with locally advanced cervical cancer treated with definitive chemoradiation. JCO Precis Oncol. 2018;2:1–8.

    Article  PubMed  Google Scholar 

  223. El-Serag HB, Rudolph KL. Hepatocellular carcinoma: epidemiology and molecular carcinogenesis. Gastroenterology. 2007;132:2557–76.

    Article  CAS  PubMed  Google Scholar 

  224. Sun Yat-sen University. ctDNA for prediction of relapse in gastric cancer [ClinicalTrials.gov identifier NCT02887612]. National Institutes of Health, ClinicalTrials.gov. https://clinicaltrials.gov. Accessed 11 Feb 2019.

  225. Johns Hopkins University and the Walter and Eliza Hall Institute of Medical Research. Circulating tumour DNA as a marker of residual disease and response to adjuvant chemotherapy in stage I–IV ovarian cancer [ClinicalTrials.gov identifier NCT03691012]. National Institutes of Health, ClinicalTrials.gov. https://clinicaltrials.gov. Accessed 11 Feb 2019.

  226. University of California, San Francisco. Biomarkers for risk stratification in lung cancer [ClinicalTrials.gov identifier NCT03774758]. National Institutes of Health, ClinicalTrials.gov. https://clinicaltrials.gov. Accessed 11 Feb 2019.

  227. Merker JD, Oxnard GR, Compton C, et al. Circulating tumor DNA analysis in patients with cancer: American Society of Clinical Oncology and College of American Pathologists joint review. J Clin Oncol. 2018;36:1631–41.

    Article  CAS  PubMed  Google Scholar 

  228. Xu T, Kang X, You X, et al. Cross-platform comparison of four leading technologies for detecting EGFR mutations in circulating tumor DNA from non-small cell lung carcinoma patient plasma. Theranostics. 2017;7:1437–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  229. Thress KS, Brant R, Carr TH, et al. EGFR mutation detection in ctDNA from NSCLC patient plasma: a cross-platform comparison of leading technologies to support the clinical development of AZD9291. Lung Cancer. 2015;90:509–15.

    Article  PubMed  Google Scholar 

  230. Institute of Cancer Research, United Kingdom. A trial using ctDNA blood tests to detect cancer cells after standard treatment to trigger additional treatment in early stage triple negative breast cancer patients (c-TRAK-TN) [ClinicalTrials.gov identifier NCT03145961]. National Institutes of Health, ClinicalTrials.gov. https://clinicaltrials.gov. Accessed 11 Feb 2019.

  231. Pao W, Miller VA, Politi KA, et al. Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain. PLoS Med. 2005;2:0225–35.

    Article  CAS  Google Scholar 

  232. Kobayashi S, Boggon T, Dayaram T, et al. EGFR mutation and resistance of non-small-cell lung cancer to gefitinib. N Engl J Med. 2005;352:786–92.

    Article  CAS  PubMed  Google Scholar 

  233. Soria J-S, Ohe Y, Vansteenkiste J, et al. Osimertinib in untreated EGFR-mutated advanced non–small-cell lung cancer. NEJM. 2018;378:113–25.

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Aadel A. Chaudhuri.

Ethics declarations

Funding

This work was funded by the NCI under award number K08CA238711 (A. A. Chaudhuri), a Cancer Research Foundation Young Investigator Award (A. A. Chaudhuri), the Washington University SPORE in Pancreatic Cancer Career Enhancement Program (A. A. Chaudhuri), and the Conquer Cancer Foundation ASCO Young Investigator Award (A. A. Chaudhuri), supported by Takeda Pharmaceuticals. Any opinions, findings and conclusions expressed in this material are those of the authors and do not necessarily reflect those of the American Society of Clinical Oncology®, Conquer Cancer®, or Takeda®.

Conflict of interest

Aadel A. Chaudhuri has received speaker honoraria and travel support from Varian Medical Systems, Roche Sequencing Solutions, and Foundation Medicine, Inc., has research support from Roche Sequencing Solutions, has served as a consultant for Tempus Labs and for Oscar Health, and is a scientific advisor for Roche Sequencing Solutions and for Geneoscopy, LLC. Jonathan C. Dudley has served as a consultant for Merck. Re-I Chin, Kevin Chen, Abul Usmani, Chanelle Chua, Peter K. Harris, Michael S. Binkley, and Tej D. Azad declare that they have no conflicts of interest that might be relevant to the contents of this review.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chin, RI., Chen, K., Usmani, A. et al. Detection of Solid Tumor Molecular Residual Disease (MRD) Using Circulating Tumor DNA (ctDNA). Mol Diagn Ther 23, 311–331 (2019). https://doi.org/10.1007/s40291-019-00390-5

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40291-019-00390-5

Navigation