Skip to main content

The Introduction and Clinical Application of Cell-Free Tumor DNA

  • Protocol
Computational Systems Biology

Part of the book series: Methods in Molecular Biology ((MIMB,volume 1754))

Abstract

Cell-free tumor DNA (ctDNA) is a kind of potential tumor biomarkers originated from cancer lesion in the circulating liquids. Liquid biopsy, as a minimally invasive or noninvasive manner, is a cutting-edge technology to detect ctDNA and other circulating biomarkers in the blood or other body fluids. ctDNA is mostly used for cancer patients to select targeted drugs in clinical application. In addition, ctDNA could also be applied to monitor tumor progression and recurrence. In conclusion, ctDNA is a very promising tumor biomarker for diagnosis and monitoring, which would increasingly become a routine clinical application in recent years.

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

Access this chapter

Protocol
USD 49.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 129.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 169.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 249.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Allard WJ, Matera J, Miller MC, Repollet M, Connelly MC, Rao C, Tibbe AG, Uhr JW, Terstappen LW (2004) Tumour cells circulate in the peripheral blood of all major carcinomas but not in healthy subjects or patients with nonmalignant diseases. Clin Cancer Res 10(20):6897–6904. https://doi.org/10.1158/1078-0432.CCR-04-0378

    Article  PubMed  Google Scholar 

  2. Hiraiwa K, Takeuchi H, Hasegawa H, Saikawa Y, Suda K, Ando T, Kumagai K, Irino T, Yoshikawa T, Matsuda S, Kitajima M, Kitagawa Y (2008) Clinical significance of circulating tumour cells in blood from patients with gastrointestinal cancers. Ann Surg Oncol 15(11):3092. https://doi.org/10.1245/s10434-008-0122-9

    Article  PubMed  Google Scholar 

  3. Rink M, Chun FK, Minner S, Friedrich M, Mauermann O, Heinzer H, Huland H, Fisch M, Pantel K, Riethdorf S (2007) Detection of circulating tumour cells in peripheral blood of patients with metastatic breast cancer: a validation study of the CellSearch system. Clin Cancer Res 13(3):920–928. https://doi.org/10.1111/j.1464-410X.2010.09562.x.

    Article  Google Scholar 

  4. Stott SL, Lee RJ, Nagrath S, Yu M, Miyamoto DT, Ulkus L, Inserra EJ, Ulman M, Springer S, Nakamura Z, Moore AL, Tsukrov DI, Kempner ME, Dahl DM, Wu CL, Iafrate AJ, Smith MR, Tompkins RG, Sequist LV, Toner M, Haber DA, Maheswaran S (2010) Isolation and characterization of circulating tumour cells from patients with localized and metastatic prostate cancer. Sci Transl Med 2(25):25ra23-25ra23. https://doi.org/10.1126/scitranslmed.3000403

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Cohen SJ, Alpaugh RK, Gross S, O'Hara SM, Smirnov DA, Terstappen LW, Allard WJ, Bilbee M, Cheng JD, Hoffman JP, Lewis NL, Pellegrino A, Rogatko A, Sigurdson E, Wang H, Watson JC, Weiner LM, Meropol NJ (2006) Isolation and characterization of circulating tumour cells in patients with metastatic colorectal cancer. Clin Colorectal Cancer 6(2):125–132. https://doi.org/10.3816/CCC.2006.n.029

    Article  CAS  PubMed  Google Scholar 

  6. Cristofanilli M, Budd GT, Ellis MJ, Stopeck A, Matera J, Miller MC, Reuben JM, Doyle GV, Allard WJ, Terstappen LW, Hayes DF (2004) Circulating tumour cells, disease progression, and survival in metastatic breast cancer. N Engl J Med 351(8):781–791. https://doi.org/10.1056/NEJMoa040766

    Article  CAS  PubMed  Google Scholar 

  7. Moreno JG, O’Hara SM, Gross S, Doyle G, Fritsche H, Gomella LG, Terstappen LW (2001) Changes in circulating carcinoma cells in patients with metastatic prostate cancer correlate with disease status. Urology 58(3):386–392. https://doi.org/10.1016/S0090-4295(01)01191-8

    Article  CAS  PubMed  Google Scholar 

  8. Diehl F, Schmidt K, Choti MA, Romans K, Goodman S, Li M, Thornton K, Agrawal N, Sokoll L, Szabo SA, Kinzler KW, Vogelstein B, Diaz LA Jr (2008) Circulating mutant DNA to assess tumour dynamics. Nat Med 14(9):985–990. https://doi.org/10.1038/nm.1789

    Article  CAS  PubMed  Google Scholar 

  9. Diehl F, Li M, Dressman D, He Y, Shen D, Szabo S, Diaz LA Jr, Goodman SN, David KA, Juhl H, Kinzler KW, Vogelstein B (2005) Detection and quantification of mutations in the plasma of patients with colorectal tumours. Proc Natl Acad Sci U S A 102(45):16368–16373. https://doi.org/10.1073/pnas.0507904102.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y, Agrawal N, Bartlett BR, Wang H, Luber B, Alani RM, Antonarakis ES, Azad NS, Bardelli A, Brem H, Cameron JL, Lee CC, Fecher LA, Gallia GL, Gibbs P, Le D, Giuntoli RL, Goggins M, Hogarty MD, Holdhoff M, Hong SM, Jiao Y, Juhl HH, Kim JJ, Siravegna G, Laheru DA, Lauricella C, Lim M, Lipson EJ, Marie SK, Netto GJ, Oliner KS, Olivi A, Olsson L, Riggins GJ, Sartore-Bianchi A, Schmidt K, Shih lM, Oba-Shinjo SM, Siena S, Theodorescu D, Tie J, Harkins TT, Veronese S, Wang TL, Weingart JD, Wolfgang CL, Wood LD, Xing D, Hruban RH, Wu J, Allen PJ, Schmidt CM, Choti MA, Velculescu VE, Kinzler KW, Vogelstein B, Papadopoulos N, Diaz LA Jr (2014) Detection of circulating tumour DNA in early-and late-stage human malignancies. Sci Transl Med 6(224):224ra224-224ra224. https://doi.org/10.1126/scitranslmed.3007094

    Article  CAS  Google Scholar 

  11. Bidard FC, Madic J, Mariani P, Piperno-Neumann S, Rampanou A, Servois V, Cassoux N, Desjardins L, Milder M, Vaucher I, Pierga JY, Lebofsky R, Stern MH, Lantz O (2014) Detection rate and prognostic value of circulating tumour cells and circulating tumour DNA in metastatic uveal melanoma. Int J Cancer 134(5):1207–1213. https://doi.org/10.1002/ijc.28436

    Article  CAS  PubMed  Google Scholar 

  12. Freidin MB, Freydina DV, Leung M, Fernandez AM, Nicholson AG, Lim E (2015) Circulating tumour DNA outperforms circulating tumour cells for KRAS mutation detection in thoracic malignancies. Clin Chem 61(10):1299–1304. https://doi.org/10.1373/clinchem.2015.242453

    Article  CAS  PubMed  Google Scholar 

  13. Matsuda K, Maruyama H, Guo F, Kleeff J, Itakura J, Matsumoto Y, Lander AD, Korc M (2001) Glypican-1 is overexpressed in human breast cancer and modulates the mitogenic effects of multiple heparin-binding growth factors in breast cancer cells. Cancer Res 61(14):5562–5569

    CAS  PubMed  Google Scholar 

  14. Kleeff J, Ishiwata T, Kumbasar A, Friess H, Büchler MW, Lander AD, Korc M (1998) The cell-surface heparan sulfate proteoglycan glypican-1 regulates growth factor action in pancreatic carcinoma cells and is overexpressed in human pancreatic cancer. J Clin Investig 102(9):1662. https://doi.org/10.1172/JCI4105

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Melo SA, Luecke LB, Kahlert C, Fernandez AF, Gammon ST, Kaye J, LeBleu VS, Mittendorf EA, Weitz J, Rahbari N, Reissfelder C, Pilarsky C, Fraga MF, Piwnica-Worms D, Kalluri R (2015) Glypican-1 identifies cancer exosomes and detects early pancreatic cancer. Nature 523(7559):177–182. https://doi.org/10.1038/nature14581

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Bujanda L, Sarasqueta C, Cosme A, Hijona E, Enríquez-Navascués JM, Placer C, Villarreal E, Herreros-Villanueva M, Giraldez MD, Gironella M, Balaguer F, Castells A (2013) Evaluation of alpha 1-antitrypsin and the levels of mRNA expression of matrix metalloproteinase 7, urokinase type plasminogen activator receptor and COX-2 for the diagnosis of colorectal cancer. PLoS One 8(1):e51810. https://doi.org/10.1371/journal.pone.0051810

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Lim SH, Spring KJ, de Souza P, MacKenzie S, Bokey L (2015) Circulating tumour cells and circulating nucleic acids as a measure of tumour dissemination in non-metastatic colorectal cancer surgery. Eur J Surg Oncol 41(3):309–314. https://doi.org/10.1016/j.ejso.2014.12.005

    Article  CAS  PubMed  Google Scholar 

  18. Rodia MT, Ugolini G, Mattei G, Montroni I, Zattoni D, Ghignone F, Veronese G, Marisi G, Lauriola M, Strippoli P, Solmi R (2016) Systematic large-scale meta-analysis identifies a panel of two mRNAs as blood biomarkers for colorectal cancer detection. Oncotarget 7(21):30295–30306. https://doi.org/10.18632/oncotarget.8108

    Article  PubMed  PubMed Central  Google Scholar 

  19. Allen-Mersh TG, McCullough TK, Patel H, Wharton RQ, Glover C, Jonas SK (2007) Role of circulating tumour cells in predicting recurrence after excision of primary colorectal carcinoma. Br J Surg 94(1):96–105. https://doi.org/10.1002/bjs.5526

    Article  CAS  PubMed  Google Scholar 

  20. Katsuno H, Zacharakis E, Aziz O, Rao C, Deeba S, Paraskeva P, Ziprin P, Athanasiou T, Darzi A (2008) Does the presence of circulating tumour cells in the venous drainage of curative colorectal cancer resections determine prognosis? A meta-analysis. Ann Surg Oncol 15(11):3083–3091. https://doi.org/10.1245/s10434-008-0131-8

    Article  PubMed  Google Scholar 

  21. El-Hefnawy T, Raja S, Kelly L, Bigbee WL, Kirkwood JM, Luketich JD, Godfrey TE (2004) Characterization of amplifiable, circulating RNA in plasma and its potential as a tool for cancer diagnostics. Clin Chem 50(3):564–573. https://doi.org/10.1373/clinchem.2003.028506

    Article  CAS  PubMed  Google Scholar 

  22. Bao Y, Ching B, Mouanoutoua M, Yu W, Keys D, Desai S, Stevens J (2012) Cancer biomarker research using castPCR technology. AACR

    Google Scholar 

  23. Wilkening S, Hemminki K, Thirumaran RK, Bermejo JL, Bonn S, Försti A, Kumar R (2005) Determination of allele frequency in pooled DNA: comparison of three PCR-based methods. BioTechniques 39(6):853–858

    Article  CAS  Google Scholar 

  24. Goto K, Ichinose Y, Ohe Y, Yamamoto N, Negoro S, Nishio K, Itoh Y, Jiang H, Duffield E, McCormack R, Saijo N, Mok T, Fukuoka M (2012) Epidermal growth factor receptor mutation status in circulating free DNA in serum: from IPASS, a phase III study of gefitinib or carboplatin/paclitaxel in non-small cell lung cancer. J Thorac Oncol 7(1):115–121. https://doi.org/10.1097/JTO.0b013e3182307f98

    Article  CAS  PubMed  Google Scholar 

  25. Karachaliou N, Mayo-de las Casas C, Queralt C, de Aguirre I, Melloni B, Cardenal F, Garcia-Gomez R, Massuti B, Sánchez JM, Porta R, Ponce-Aix S, Moran T, Carcereny E, Felip E, Bover I, Insa A, Reguart N, Isla D, Vergnenegre A, de Marinis F, Gervais R, Corre R, Paz-Ares L, Morales-Espinosa D, Viteri S, Drozdowskyj A, Jordana-Ariza N, Ramirez-Serrano JL, Molina-Vila MA, Rosell R, Spanish Lung Cancer Group (2015) Association of EGFR L858R mutation in circulating free DNA with survival in the EURTAC trial. JAMA Oncol 1(2):149–157. https://doi.org/10.1001/jamaoncol.2014.257

    Article  PubMed  Google Scholar 

  26. Hindson BJ, Ness KD, Masquelier DA, Belgrader P, Heredia NJ, Makarewicz AJ, Bright IJ, Lucero MY, Hiddessen AL, Legler TC, Kitano TK, Hodel MR, Petersen JF, Wyatt PW, Steenblock ER, Shah PH, Bousse LJ, Troup CB, Mellen JC, Wittmann DK, Erndt NG, Cauley TH, Koehler RT, So AP, Dube S, Rose KA, Montesclaros L, Wang S, Stumbo DP, Hodges SP, Romine S, Milanovich FP, White HE, Regan JF, Karlin-Neumann GA, Hindson CM, Saxonov S, Colston BW (2011) High-throughput droplet digital PCR system for absolute quantitation of DNA copy number. Anal Chem 83(22):8604–8610. https://doi.org/10.1021/ac202028g

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Taniguchi K, Uchida J, Nishino K, Kumagai T, Okuyama T, Okami J, Kato K (2011) Quantitative detection of EGFR mutations in circulating tumour DNA derived from lung adenocarcinomas. Clin Cancer Res 17(24):7808–7815. https://doi.org/10.1158/1078-0432.CCR-11-1712

    Article  CAS  PubMed  Google Scholar 

  28. Taniguchi K, Uchida J, Nishino K, Kumagai T, Okuyama T, Okami J, Higashiyama M, Kodama K, Imamura F, Kato K (2012) Noninvasive identification and monitoring of cancer mutations by targeted deep sequencing of plasma DNA. Sci Transl Med 4(136):136ra168-136ra168. https://doi.org/10.1158/1078-0432.CCR-11-1712

    Article  CAS  Google Scholar 

  29. Kinde I, Wu J, Papadopoulos N, Kinzler KW, Vogelstein B (2011) Detection and quantification of rare mutations with massively parallel sequencing. Proc Natl Acad Sci 108(23):9530–9535. https://doi.org/10.1073/pnas.1105422108

    Article  PubMed  PubMed Central  Google Scholar 

  30. Newman AM, Bratman SV, To J, Wynne JF, Eclov NC, Modlin LA, Liu CL, Neal JW, Wakelee HA, Merritt RE, Shrager JB, Loo BW Jr, Alizadeh AA, Diehn M (2014) An ultrasensitive method for quantitating circulating tumour DNA with broad patient coverage. Nat Med 20(5):548–554. https://doi.org/10.1038/nm.3519

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Griffin TJ, Smith LM (2000) Single-nucleotide polymorphism analysis by MALDI–TOF mass spectrometry. Trends Biotechnol 18(2):77–84

    Article  CAS  Google Scholar 

  32. Giannini R, Lupi C, Sensi E, Alì G, Proietti A, Boldrini L, Servadio A, Giordano M, Macerola E, Bruno R, Borrelli N, Chella A, Melfi F, Lucchi M, Ribechini A, Vasile E, Cappuzzo F, Mussi A, Fontanini G (2016) EGFR and KRAS mutational analysis in a large series of Italian non-small cell lung cancer patients: 2,387 cases from a single center. Oncol Rep 36(2):1166–1172. https://doi.org/10.3892/or.2016.4874

    Article  CAS  PubMed  Google Scholar 

  33. Finney Rutten LJ, Nelson DE (2004) Meissner HI. Examination of population-wide trends in barriers to cancer screening from a diffusion of innovation perspective (1987–2000). Prev Med 38:258–268. https://doi.org/10.1016/j.ypmed.2003.10.011

    Article  PubMed  Google Scholar 

  34. Lo SH, Waller J, Wardle J (2013) Comparing barriers to colorectal cancer screening with barriers to breast and cervical screening a population-based survey of screening age women in great Britain. J Med Screen 20(2):73–79

    Article  Google Scholar 

  35. Potter NT, Hurban P, White MN, Whitlock KD, Lofton-Day CE, Tetzner R, Koenig T, Quigley NB, Weiss G (2014) Validation of a real-time PCR-based qualitative assay for the detection of methylated SEPT9 DNA in human plasma. Clin Chem 60(9):1183–1191. https://doi.org/10.1373/clinchem.2013.221044

    Article  CAS  PubMed  Google Scholar 

  36. Romero R, Mahoney MJ (2015) Noninvasive prenatal testing and detection of maternal cancer. JAMA 314(2):131–133. https://doi.org/10.1001/jama.2015.7523

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Bianchi DW, Chudova D, Sehnert AJ (2015) Noninvasive prenatal testing and incidental detection of occult maternal malignancies. JAMA 314(2):162–169. https://doi.org/10.1001/jama.2015.7120

    Article  CAS  PubMed  Google Scholar 

  38. Snyder MW, Kircher M, Hill AJ (2016) Cell-free DNA comprises an in vivo nucleosome footprint that informs its tissues-of-origin. Cell 164(1):57–68. https://doi.org/10.1016/j.cell.2015.11.05.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Olsson E, Winter C, George A, Chen Y, Howlin J, Tang MH, Dahlgren M, Schulz R, Grabau D, van Westen D, Fernö M, Ingvar C, Rose C, Bendahl PO, Rydén L, Borg Å, Gruvberger-Saal SK, Jernström H, Saal LH (2015) Serial monitoring of circulating tumour DNA in patients with primary breast cancer for detection of occult metastatic disease. EMBO Mol Med 7(8):1034–1047. https://doi.org/10.15252/emmm.201404913

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Reinert T, Schøler LV, Thomsen R, Tobiasen H, Vang S, Nordentoft I, Lamy P, Kannerup AS, Mortensen FV, Stribolt K, Hamilton-Dutoit S, Nielsen HJ, Laurberg S, Pallisgaard N, Pedersen JS, Ørntoft TF, Andersen CL (2016) Analysis of circulating tumour DNA to monitor disease burden following colorectal cancer surgery. Gut 65:gutjnl-2014. https://doi.org/10.1136/gutjnl-2014-308859

    Article  CAS  Google Scholar 

  41. Tie J, Kinde I, Wang Y (2014) Circulating tumour DNA (ctDNA) as a marker of recurrence risk in stage II colon cancer (CC). ASCO annual meeting proceedings, Chicago

    Google Scholar 

  42. Quasar Collaborative Group, Gray R, Barnwell J, McConkey C, Hills RK, Williams NS, Kerr DJ (2007) Adjuvant chemotherapy versus observation in patients with colorectal cancer: a randomised study. Lancet 370:2020–2029. https://doi.org/10.1016/S0140-6736(07)61866-2

    Article  CAS  Google Scholar 

  43. www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm504488.htm.

  44. Kuang Y, Rogers A, Yeap BY, Wang L, Makrigiorgos M, Vetrand K, Thiede S, Distel RJ, Jänne PA (2009) Noninvasive detection of EGFR T790M in gefitinib or erlotinib resistant non–small cell lung cancer. Clin Cancer Res 15(8):2630–2636. https://doi.org/10.1158/1078-0432.CCR-08-2592

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Nakamura T, Sueoka-Aragane N, Iwanaga K, Sato A, Komiya K, Kobayashi N, Hayashi S, Hosomi T, Hirai M, Sueoka E, Kimura S (2012) Application of a highly sensitive detection system for epidermal growth factor receptor mutations in plasma DNA. J Thorac Oncol 7(9):1369–1381. https://doi.org/10.1097/JTO.0b013e31825f2821

    Article  CAS  PubMed  Google Scholar 

  46. Kimura H, Kasahara K, Kawaishi M, Kunitoh H, Tamura T, Holloway B, Nishio K (2006) 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 12(13):3915–3921. https://doi.org/10.1158/1078-0432.CCR-05-2324

    Article  CAS  PubMed  Google Scholar 

  47. Luo J, Shen L, Zheng D (2014) Diagnostic value of circulating free DNA for the detection of EGFR mutation status in NSCLC: a systematic review and meta-analysis. Sci Rep 4:6269. https://doi.org/10.1038/srep06269

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Qiu M, Wang J, Xu Y, Ding X, Li M, Jiang F, Xu L, Yin R (2014) Circulating tumour DNA is effective for the detection of EGFR mutation in non–small cell lung cancer: a meta-analysis. Cancer Epidemiol Prev Biomarkers 24(1):206–212. https://doi.org/10.1158/1055-9965.EPI-14-0895

    Article  CAS  Google Scholar 

  49. Mok T, Wu YL, Lee JS, Yu CJ, Sriuranpong V, Sandoval-Tan J, Ladrera G, Thongprasert S, Srimuninnimit V, Liao M, Zhu Y, Zhou C, Fuerte F, Margono B, Wen W, Tsai J, Truman M, Klughammer B, Shames DS, Wu L (2015) Detection and dynamic changes of EGFR mutations from circulating tumour DNA as a predictor of survival outcomes in NSCLC patients treated with first-line intercalated erlotinib and chemotherapy. Clin Cancer Res 21(14):3196–3203. https://doi.org/10.1158/1078-0432.CCR-14-2594

    Article  CAS  PubMed  Google Scholar 

  50. Weber B, Meldgaard P, Hager H, Wu L, Wei W, Tsai J, Khalil A, Nexo E, Sorensen BS (2014) Detection of EGFR mutations in plasma and biopsies from non-small cell lung cancer patients by allele-specific PCR assays. BMC Cancer 14(1):294. https://doi.org/10.1186/1471-2407-14-294.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Cross DA, Ashton SE, Ghiorghiu S, Eberlein C, Nebhan CA, Spitzler PJ, Orme JP, Finlay MR, Ward RA, Mellor MJ, Hughes G, Rahi A, Jacobs VN, Red Brewer M, Ichihara E, Sun J, Jin H, Ballard P, Al-Kadhimi K, Rowlinson R, Klinowska T, Richmond GH, Cantarini M, Kim DW, Ranson MR, Pao W (2014) AZD9291, an irreversible EGFR TKI, overcomes T790M-mediated resistance to EGFR inhibitors in lung cancer. Cancer Discov 4(9):1046–1061. https://doi.org/10.1158/2159-8290.CD-14-0337

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Zhou W, Ercan D, Chen L, Yun CH, Li D, Capelletti M, Cortot AB, Chirieac L, Iacob RE, Padera R, Engen JR, Wong KK, Eck MJ, Gray NS, Jänne PA (2009) Novel mutant-selective EGFR kinase inhibitors against EGFR T790M. Nature 462(7276):1070–1074. https://doi.org/10.1038/nature08622

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Oxnard GR, Thress KS, Alden RS, Lawrance R, Paweletz CP, Cantarini M, Yang JC, Barrett JC, Jänne PA (2016) Association between plasma genotyping and outcomes of treatment with osimertinib (AZD9291) in advanced non–small-cell lung cancer. J Clin Oncol 34(28):3375–3382. https://doi.org/10.1200/JCO.2016.66.7162

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. http://www.fda.gov/ucm/groups/fdagov-public/@fdagov-meddev-gen/documents/document/ucm510824.pdf

  55. Sequist LV, Yang JC, Yamamoto N, O'Byrne K, Hirsh V, Mok T, Geater SL, Orlov S, Tsai CM, Boyer M, Su WC, Bennouna J, Kato T, Gorbunova V, Lee KH, Shah R, Massey D, Zazulina V, Shahidi M, Schuler M (2013) Phase III study of afatinib or cisplatin plus pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations. J Clin Oncol 31(27):3327–3334. https://doi.org/10.1200/JCO.2012.44.2806

    Article  CAS  PubMed  Google Scholar 

  56. FDA http://www.accessdata.fda.gov/cdrh_docs/pdf12/P120022c.pdf

  57. Douillard J, Ostoros G, Cobo M, Ciuleanu T, McCormack R, Webster A, Milenkova T (2014) First-line gefitinib in Caucasian EGFR mutation-positive NSCLC patients: a phase-IV, open-label, single-arm study. Br J Cancer 110(1):55–62. https://doi.org/10.1038/bjc.2013.721

    Article  CAS  PubMed  Google Scholar 

  58. http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_Product_Information/human/001016/WC500036358.pdf

  59. http://www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm504540.htm

  60. Jenkins S, Yang J, Ramalingam S, Yu K, Patel S, Weston S, Lawrance R, Cantarini M, Jänne P, Mitsudomi T (2016) 134O_PR: plasma ctDNA analysis for detection of EGFR T790M mutation in patients (pts) with EGFR mutation-positive advanced non-small cell lung cancer (aNSCLC). J Thorac Oncol 11((4):S153–S154. https://doi.org/10.1016/S1556-0864(16)30327-6

    Article  Google Scholar 

  61. Lanman RB, Mortimer SA, Zill OA, Sebisanovic D, Lopez R, Blau S, Collisson EA, Divers SG, Hoon DS, Kopetz ES, Lee J, Nikolinakos PG, Baca AM, Kermani BG, Eltoukhy H, Talasaz A (2015) Analytical and clinical validation of a digital sequencing panel for quantitative, highly accurate evaluation of cell-free circulating tumour DNA. PLoS One 10(10):e0140712. https://doi.org/10.1371/journal.pone.0140712

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Zill OA, Mortimer S, Banks K (2016) Somatic genomic landscape of over 15,000 patients with advanced-stage cancer from clinical next-generation sequencing analysis of circulating tumour DNA. J Clin Oncol 34(suppl)

    Article  Google Scholar 

  63. Kwak EL, Bang YJ, Camidge DR, Shaw AT, Solomon B, Maki RG, Ou SH, Dezube BJ, Jänne PA, Costa DB, Varella-Garcia M, Kim WH, Lynch TJ, Fidias P, Stubbs H, Engelman JA, Sequist LV, Tan W, Gandhi L, Mino-Kenudson M, Wei GC, Shreeve SM, Ratain MJ, Settleman J, Christensen JG, Haber DA, Wilner K, Salgia R, Shapiro GI, Clark JW, Iafrate AJ (2010) Anaplastic lymphoma kinase inhibition in non–small-cell lung cancer. N Engl J Med 363(18):1693–1703. https://doi.org/10.1200/JCO.2016.34.15_suppl.LBA11501

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Koivunen JP, Kim J, Lee J, Rogers AM, Park JO, Zhao X, Naoki K, Okamoto I, Nakagawa K, Yeap BY, Meyerson M, Wong KK, Richards WG, Sugarbaker DJ, Johnson BE, Jänne PA (2008) Mutations in the LKB1 tumour suppressor are frequently detected in tumours from Caucasian but not Asian lung cancer patients. Br J Cancer 99(2):245–252. https://doi.org/10.1038/sj.bjc.6604469

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Shaw AT, Yeap BY, Mino-Kenudson M, Digumarthy SR, Costa DB, Heist RS, Solomon B, Stubbs H, Admane S, McDermott U, Settleman J, Kobayashi S, Mark EJ, Rodig SJ, Chirieac LR, Kwak EL, Lynch TJ, Iafrate AJ (2009) Clinical features and outcome of patients with non–small-cell lung cancer who harbor EML4-ALK. J Clin Oncol 27(26):4247–4253. https://doi.org/10.1200/JCO.2009.22.6993

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Inamura K, Takeuchi K, Togashi Y, Hatano S, Ninomiya H, Motoi N, Mun MY, Sakao Y, Okumura S, Nakagawa K, Soda M, Choi YL, Mano H, Ishikawa Y, Nakagawa K (2009) EML4-ALK lung cancers are characterized by rare other mutations, a TTF-1 cell lineage, an acinar histology, and young onset. Mod Pathol 22(4):508–515. https://doi.org/10.1038/modpathol.2009.2

    Article  CAS  PubMed  Google Scholar 

  67. Soda M, Takada S, Takeuchi K, Choi YL, Enomoto M, Ueno T, Haruta H, Hamada T, Yamashita Y, Ishikawa Y, Sugiyama Y, Mano H (2008) A mouse model for EML4-ALK-positive lung cancer. Proc Natl Acad Sci 105(50):19893–19897. https://doi.org/10.1073/pnas.0805381105

    Article  PubMed  PubMed Central  Google Scholar 

  68. Kim DW, Mehra R, Tan DS, Felip E, Chow LQ, Camidge DR, Vansteenkiste J, Sharma S, De Pas T, Riely GJ, Solomon BJ, Wolf J, Thomas M, Schuler M, Liu G, Santoro A, Sutradhar S, Li S, Szczudlo T, Yovine A, Shaw AT (2016) Activity and safety of ceritinib in patients with ALK-rearranged non-small-cell lung cancer (ASCEND-1): updated results from the multicentre, open-label, phase 1 trial. Lancet Oncol 17(4):452–463. https://doi.org/10.1016/S1470-2045(15)00614-2

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Ou SH, Ahn JS, De Petris L, Govindan R, Yang JC, Hughes B, Lena H, Moro-Sibilot D, Bearz A, Ramirez SV, Mekhail T, Spira A, Bordogna W, Balas B, Morcos PN, Monnet A, Zeaiter A, Kim DW (2015) Alectinib in crizotinib-refractory ALK-rearranged non–small-cell lung cancer: a phase II global study. J Clin Oncol 34(7):661–668. https://doi.org/10.1200/JCO.2015.63.9443.

    Article  PubMed  Google Scholar 

  70. Shaw AT, Gandhi L, Gadgeel S, Riely GJ, Cetnar J, West H, Camidge DR, Socinski MA, Chiappori A, Mekhail T, Chao BH, Borghaei H, Gold KA, Zeaiter A, Bordogna W, Balas B, Puig O, Henschel V, Ou SH (2016) Alectinib in ALK-positive, crizotinib-resistant, non-small-cell lung cancer: a single-group, multicentre, phase 2 trial. Lancet Oncol 17(2):234–242. https://doi.org/10.1016/S1470-2045(15)00488-X

    Article  CAS  PubMed  Google Scholar 

  71. Shaw AT, Kim DW, Mehra R, Tan DS, Felip E, Chow LQ, Camidge DR, Vansteenkiste J, Sharma S, De Pas T, Riely GJ, Solomon BJ, Wolf J, Thomas M, Schuler M, Liu G, Santoro A, Lau YY, Goldwasser M, Boral AL, Engelman JA (2014) Ceritinib in ALK-rearranged non–small-cell lung cancer. N Engl J Med 370(13):1189–1197. https://doi.org/10.1056/NEJMoa1311107.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Shaw AT, Kim DW, Nakagawa K, Seto T, Crinó L, Ahn MJ, De Pas T, Besse B, Solomon BJ, Blackhall F, Wu YL, Thomas M, O'Byrne KJ, Moro-Sibilot D, Camidge DR, Mok T, Hirsh V, Riely GJ, Iyer S, Tassell V, Polli A, Wilner KD, Jänne PA (2013) Crizotinib versus chemotherapy in advanced ALK-positive lung cancer. N Engl J Med 368(25):2385–2394. https://doi.org/10.1056/NEJMoa1214886

    Article  CAS  PubMed  Google Scholar 

  73. Solomon BJ, Mok T, Kim DW, Wu YL, Nakagawa K, Mekhail T, Felip E, Cappuzzo F, Paolini J, Usari T, Iyer S, Reisman A, Wilner KD, Tursi J, Blackhall F, PROFILE 1014 Investigators (2014) First-line crizotinib versus chemotherapy in ALK-positive lung cancer. N Engl J Med 371(23):2167–2177. https://doi.org/10.1056/NEJMoa1408440

    Article  CAS  PubMed  Google Scholar 

  74. Zou HY, Li Q, Lee JH, Arango ME, McDonnell SR, Yamazaki S, Koudriakova TB, Alton G, Cui JJ, Kung PP, Nambu MD, Los G, Bender SL, Mroczkowski B, Christensen JG (2007) An orally available small-molecule inhibitor of c-met, PF-2341066, exhibits cytoreductive antitumor efficacy through antiproliferative and antiangiogenic mechanisms. Cancer Res 67(9):4408–4417. https://doi.org/10.1158/0008-5472.CAN-06-4443

    Article  CAS  PubMed  Google Scholar 

  75. Gettinger SN, Bazhenova LA, Langer CJ, Salgia R, Gold KA, Rosell R, Shaw AT, Weiss GJ, Tugnait M, Narasimhan NI, Dorer DJ, Kerstein D, Rivera VM, Clackson T, Haluska FG, Camidge DR (2016) Activity and safety of brigatinib in ALK-rearranged non-small-cell lung cancer and other malignancies: a single-arm, open-label, phase 1/2 trial. Lancet Oncol 17(12):1683–1696. https://doi.org/10.1016/S1470-2045(16)30392-8

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Mingyan Xu .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2018 Springer Science+Business Media, LLC, part of Springer Nature

About this protocol

Cite this protocol

Li, J., Liu, R., Huang, C., Chen, S., Xu, M. (2018). The Introduction and Clinical Application of Cell-Free Tumor DNA. In: Huang, T. (eds) Computational Systems Biology. Methods in Molecular Biology, vol 1754. Humana Press, New York, NY. https://doi.org/10.1007/978-1-4939-7717-8_4

Download citation

  • DOI: https://doi.org/10.1007/978-1-4939-7717-8_4

  • Publisher Name: Humana Press, New York, NY

  • Print ISBN: 978-1-4939-7716-1

  • Online ISBN: 978-1-4939-7717-8

  • eBook Packages: Springer Protocols

Publish with us

Policies and ethics