The Promise of Circulating Tumor DNA in Head and Neck Cancer

Sukhkaran S Aulakh, Dustin A Silverman, Kurtis Young, Steven K Dennis, Andrew C Birkeland, Sukhkaran S Aulakh, Dustin A Silverman, Kurtis Young, Steven K Dennis, Andrew C Birkeland

Abstract

As the seventh most common cancer globally, head and neck cancers (HNC) exert considerable disease burden, with an estimated 277,597 deaths worldwide in 2020 alone. Traditional risk factors for HNC include tobacco, alcohol, and betel nut; more recently, human papillomavirus has emerged as a distinct driver of disease. Currently, limitations of cancer screening and surveillance methods often lead to identifying HNC in more advanced stages, with associated poor outcomes. Liquid biopsies, in particular circulating tumor DNA (ctDNA), offer the potential for enhancing screening, early diagnosis, and surveillance in HNC patients, with potential improvements in HNC patient outcomes. In this review, we examine current methodologies for detecting ctDNA and highlight current research illustrating viral and non-viral ctDNA biomarker utilities in HNC screening, diagnosis, treatment response, and prognosis. We also summarize current challenges and future directions for ctDNA testing in HNC patients.

Keywords: circulating tumor DNA; exosomes; gene methylation; head and neck cancer; head and neck squamous cell carcinoma; liquid biopsy.

Conflict of interest statement

The authors declare no conflict of interest.

References

    1. Siegel R.L., Miller K.D., Fuchs H.E., Jemal A. Cancer statistics, 2022. CA Cancer J. Clin. 2022;72:7–33. doi: 10.3322/caac.21708.
    1. Posner M.R., Hershock D.M., Blajman C.R., Mickiewicz E., Winquist E., Gorbounova V., Tjulandin S., Shin D.M., Cullen K., Ervin T.J., et al. Cisplatin and fluorouracil alone or with docetaxel in head and neck cancer. N. Engl. J. Med. 2007;357:1705–1715. doi: 10.1056/NEJMoa070956.
    1. Pignon J.P., le Maître A., Maillard E., Bourhis J. Meta-analysis of chemotherapy in head and neck cancer (MACH-NC): An update on 93 randomised trials and 17,346 patients. Radiother. Oncol. 2009;92:4–14. doi: 10.1016/j.radonc.2009.04.014.
    1. Brockstein B., Haraf D.J., Rademaker A.W., Kies M.S., Stenson K.M., Rosen F., Mittal B.B., Pelzer H., Fung B.B., Witt M.E., et al. Patterns of failure, prognostic factors and survival in locoregionally advanced head and neck cancer treated with concomitant chemoradiotherapy: A 9-year, 337-patient, multi-institutional experience. Ann. Oncol. 2004;15:1179–1186. doi: 10.1093/annonc/mdh308.
    1. Guizard A.-V.N., Dejardin O.J., Launay L.C., Bara S., Lapôtre-Ledoux B.M., Babin E.B., Launoy G.D., Ligier K.A. Diagnosis and management of head and neck cancers in a high-incidence area in France: A population-based study. Medicine. 2017;96:e7285. doi: 10.1097/MD.0000000000007285.
    1. Thierry A.R., El Messaoudi S., Gahan P.B., Anker P., Stroun M. Origins, structures, and functions of circulating DNA in oncology. Cancer Metastasis Rev. 2016;35:347–376. doi: 10.1007/s10555-016-9629-x.
    1. Mandel P., Metais P. Nuclear Acids in Human Blood Plasma. C. R. Seances Soc. Biol. Fil. 1948;142:241–243.
    1. Schwarzenbach H., Hoon D.S.B., Pantel K. Cell-free nucleic acids as biomarkers in cancer patients. Nat. Rev. Cancer. 2011;11:426–437. doi: 10.1038/nrc3066.
    1. Alix-Panabières C., Pantel K. Clinical Applications of Circulating Tumor Cells and Circulating Tumor DNA as Liquid Biopsy. Cancer Discov. 2016;6:479–491. doi: 10.1158/-15-1483.
    1. Alix-Panabières C., Schwarzenbach H., Pantel K. Circulating tumor cells and circulating tumor DNA. Annu. Rev. Med. 2012;63:199–215. doi: 10.1146/annurev-med-062310-094219.
    1. Skvortsova T.E., Rykova E.Y., Tamkovich S.N., Bryzgunova O.E., Starikov A.V., Kuznetsova N.P., Vlassov V.V., Laktionov P.P. Cell-free and cell-bound circulating DNA in breast tumours: DNA quantification and analysis of tumour-related gene methylation. Br. J. Cancer. 2006;94:1492–1495. doi: 10.1038/sj.bjc.6603117.
    1. van Ginkel J.H., Huibers M.M.H., van Es R.J.J., de Bree R., Willems S.M. Droplet digital PCR for detection and quantification of circulating tumor DNA in plasma of head and neck cancer patients. BMC Cancer. 2017;17:428. doi: 10.1186/s12885-017-3424-0.
    1. Diehl F., Schmidt K., Choti M.A., Romans K., Goodman S., Li M., Thornton K., Agrawal N., Sokoll L., Szabo S.A., et al. Circulating mutant DNA to assess tumor dynamics. Nat. Med. 2008;14:985–990. doi: 10.1038/nm.1789.
    1. Leary R.J., Kinde I., Diehl F., Schmidt K., Clouser C., Duncan C., Antipova A., Lee C., McKernan K., De La Vega F.M., et al. Development of personalized tumor biomarkers using massively parallel sequencing. Sci. Transl. Med. 2010;2:20ra14. doi: 10.1126/scitranslmed.3000702.
    1. Dawson S.J., Tsui D.W., Murtaza M., Biggs H., Rueda O.M., Chin S.F., Dunning M.J., Gale D., Forshew T., Mahler-Araujo B., et al. Analysis of circulating tumor DNA to monitor metastatic breast cancer. N. Engl. J. Med. 2013;368:1199–1209. doi: 10.1056/NEJMoa1213261.
    1. Leary R.J., Sausen M., Kinde I., Papadopoulos N., Carpten J.D., Craig D., O’Shaughnessy J., Kinzler K.W., Parmigiani G., Vogelstein B., et al. Detection of chromosomal alterations in the circulation of cancer patients with whole-genome sequencing. Sci. Transl. Med. 2012;4:162ra154. doi: 10.1126/scitranslmed.3004742.
    1. Pinheiro L.B., Coleman V.A., Hindson C.M., Herrmann J., Hindson B.J., Bhat S., Emslie K.R. Evaluation of a Droplet Digital Polymerase Chain Reaction Format for DNA Copy Number Quantification. Anal. Chem. 2012;84:1003–1011. doi: 10.1021/ac202578x.
    1. Diehl F., Li M., He Y., Kinzler K.W., Vogelstein B., Dressman D. BEAMing: Single-molecule PCR on microparticles in water-in-oil emulsions. Nat. Methods. 2006;3:551–559. doi: 10.1038/nmeth898.
    1. Li M., Diehl F., Dressman D., Vogelstein B., Kinzler K.W. BEAMing up for detection and quantification of rare sequence variants. Nat. Methods. 2006;3:95–97. doi: 10.1038/nmeth850.
    1. O’Leary B., Hrebien S., Beaney M., Fribbens C., Garcia-Murillas I., Jiang J., Li Y., Huang Bartlett C., André F., Loibl S., et al. Comparison of BEAMing and Droplet Digital PCR for Circulating Tumor DNA Analysis. Clin. Chem. 2019;65:1405–1413. doi: 10.1373/clinchem.2019.305805.
    1. Denis J.A., Guillerm E., Coulet F., Larsen A.K., Lacorte J.-M. The Role of BEAMing and Digital PCR for Multiplexed Analysis in Molecular Oncology in the Era of Next-Generation Sequencing. Mol. Diagn. Ther. 2017;21:587–600. doi: 10.1007/s40291-017-0287-7.
    1. Ku J.-L., Jeon Y.-K., Park J.-G. Methylation-Specific PCR. In: Tollefsbol T.O., editor. Epigenetics Protocols. Humana Press; Totowa, NJ, USA: 2011. pp. 23–32.
    1. Mattox A.K., D’Souza G., Khan Z., Allen H., Henson S., Seiwert T.Y., Koch W., Pardoll D.M., Fakhry C. Comparison of next generation sequencing, droplet digital PCR, and quantitative real-time PCR for the earlier detection and quantification of HPV in HPV-positive oropharyngeal cancer. Oral. Oncol. 2022;128:105805. doi: 10.1016/j.oraloncology.2022.105805.
    1. Wang Y., Springer S., Mulvey C.L., Silliman N., Schaefer J., Sausen M., James N., Rettig E.M., Guo T., Pickering C.R., et al. Detection of somatic mutations and HPV in the saliva and plasma of patients with head and neck squamous cell carcinomas. Sci. Transl. Med. 2015;7:293ra104. doi: 10.1126/scitranslmed.aaa8507.
    1. Cao H., Banh A., Kwok S., Shi X., Wu S., Krakow T., Khong B., Bavan B., Bala R., Pinsky B.A., et al. Quantitation of human papillomavirus DNA in plasma of oropharyngeal carcinoma patients. Int. J. Radiat. Oncol. Biol. Phys. 2012;82:e351–e358. doi: 10.1016/j.ijrobp.2011.05.061.
    1. Chera B.S., Kumar S., Shen C., Amdur R., Dagan R., Green R., Goldman E., Weiss J., Grilley-Olson J., Patel S., et al. Plasma circulating tumor HPV DNA for the surveillance of cancer recurrence in HPV-associated oropharyngeal cancer. J. Clin. Oncol. 2020;38:1050–1058. doi: 10.1200/JCO.19.02444.
    1. Lee J.S., Hur J.Y., Kim I.A., Kim H.J., Choi C.M., Lee J.C., Kim W.S., Lee K.Y. Liquid biopsy using the supernatant of a pleural effusion for EGFR genotyping in pulmonary adenocarcinoma patients: A comparison between cell-free DNA and extracellular vesicle-derived DNA. BMC Cancer. 2018;18:1236. doi: 10.1186/s12885-018-5138-3.
    1. Satyal U., Srivastava A., Abbosh P.H. Urine Biopsy—Liquid Gold for Molecular Detection and Surveillance of Bladder Cancer. Front. Oncol. 2019;9:1266. doi: 10.3389/fonc.2019.01266.
    1. Ahn S.M., Chan J.Y.K., Zhang Z., Wang H., Khan Z., Bishop J.A., Westra W., Koch W.M., Califano J.A. Saliva and Plasma Quantitative Polymerase Chain Reaction–Based Detection and Surveillance of Human Papillomavirus–Related Head and Neck Cancer. JAMA Otolaryngol. Head Neck Surg. 2014;140:846–854. doi: 10.1001/jamaoto.2014.1338.
    1. Saraiya M., Unger E.R., Thompson T.D., Lynch C.F., Hernandez B.Y., Lyu C.W., Steinau M., Watson M., Wilkinson E.J., Hopenhayn C., et al. US assessment of HPV types in cancers: Implications for current and 9-valent HPV vaccines. J. Natl. Cancer Inst. 2015;107:djv086. doi: 10.1093/jnci/djv086.
    1. Schiffman M., Doorbar J., Wentzensen N., de Sanjosé S., Fakhry C., Monk B.J., Stanley M.A., Franceschi S. Carcinogenic human papillomavirus infection. Nat. Rev. Dis. Primers. 2016;2:16086. doi: 10.1038/nrdp.2016.86.
    1. Galloway T.J., Zhang Q.E., Nguyen-Tan P.F., Rosenthal D.I., Soulieres D., Fortin A., Silverman C.L., Daly M.E., Ridge J.A., Hammond J.A., et al. Prognostic Value of p16 Status on the Development of a Complete Response in Involved Oropharynx Cancer Neck Nodes After Cisplatin-Based Chemoradiation: A Secondary Analysis of NRG Oncology RTOG 0129. Int. J. Radiat. Oncol. Biol. Phys. 2016;96:362–371. doi: 10.1016/j.ijrobp.2016.05.026.
    1. Strohl M.P., Wai K.C., Ha P.K. De-intensification strategies in HPV-related oropharyngeal squamous cell carcinoma-a narrative review. Ann. Transl. Med. 2020;8:1601. doi: 10.21037/atm-20-2984.
    1. Jalaly J.B., Hosseini S.M., Shafique K., Baloch Z.W. Current Status of p16 Immunohistochemistry and HPV Testing in Fine Needle Aspiration Specimens of the Head and Neck. Acta Cytol. 2020;64:30–39. doi: 10.1159/000496158.
    1. Siravegna G., O’Boyle C.J., Varmeh S., Queenan N., Michel A., Stein J., Thierauf J., Sadow P.M., Faquin W.C., Perry S.K., et al. Cell-Free HPV DNA Provides an Accurate and Rapid Diagnosis of HPV-Associated Head and Neck Cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2022;28:719–727. doi: 10.1158/1078-0432.CCR-21-3151.
    1. Rettig E.M., Faden D.L., Sandhu S., Wong K., Faquin W.C., Warinner C., Stephens P., Kumar S., Kuperwasser C., Richmon J.D., et al. Detection of circulating tumor human papillomavirus DNA before diagnosis of HPV-positive head and neck cancer. Int. J. Cancer. 2022. early view .
    1. Tewari S.R., D’Souza G., Troy T., Wright H., Struijk L., Waterboer T., Fakhry C. Association of Plasma Circulating Tumor HPV DNA with HPV-Related Oropharynx Cancer. JAMA Otolaryngol. Head Neck Surg. 2022;148:488. doi: 10.1001/jamaoto.2022.0159.
    1. Tanaka H., Takemoto N., Horie M., Takai E., Fukusumi T., Suzuki M., Eguchi H., Komukai S., Tatsumi M., Isohashi F., et al. Circulating tumor HPV DNA complements PET-CT in guiding management after radiotherapy in HPV-related squamous cell carcinoma of the head and neck. Int. J. Cancer. 2021;148:995–1005. doi: 10.1002/ijc.33287.
    1. Chera B.S., Kumar S., Beaty B.T., Marron D., Jefferys S., Green R., Goldman E.C., Amdur R., Sheets N., Dagan R., et al. Rapid Clearance Profile of Plasma Circulating Tumor HPV Type 16 DNA during Chemoradiotherapy Correlates with Disease Control in HPV-Associated Oropharyngeal Cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2019;25:4682–4690. doi: 10.1158/1078-0432.CCR-19-0211.
    1. Haring C.T., Bhambhani C., Brummel C., Jewell B., Bellile E., Heft Neal M.E., Sandford E., Spengler R.M., Bhangale A., Spector M.E., et al. Human papilloma virus circulating tumor DNA assay predicts treatment response in recurrent/metastatic head and neck squamous cell carcinoma. Oncotarget. 2021;12:1214–1229. doi: 10.18632/oncotarget.27992.
    1. O’Boyle C.J., Siravegna G., Varmeh S., Queenan N., Michel A., Pang K.C.S., Stein J., Thierauf J.C., Sadow P.M., Faquin W.C., et al. Cell-free human papillomavirus DNA kinetics after surgery for human papillomavirus-associated oropharyngeal cancer. Cancer. 2022;128:2193–2204. doi: 10.1002/cncr.34109.
    1. Routman D.M., Kumar S., Chera B.S., Jethwa K.R., Van Abel K.M., Frechette K., DeWees T., Golafshar M., Garcia J.J., Price D.L., et al. Detectable Post-operative Circulating Tumor Human Papillomavirus (HPV) DNA And Association with Recurrence in Patients with HPV-Associated Oropharyngeal Squamous Cell Carcinoma. Int. J. Radiat. Oncol. Biol. Phys. 2022. in press .
    1. Kreimer A.R., Shiels M.S., Fakhry C., Johansson M., Pawlita M., Brennan P., Hildesheim A., Waterboer T. Screening for human papillomavirus-driven oropharyngeal cancer: Considerations for feasibility and strategies for research. Cancer. 2018;124:1859–1866. doi: 10.1002/cncr.31256.
    1. Reder H., Taferner V.F., Wittekindt C., Bräuninger A., Speel E.J.M., Gattenlöhner S., Wolf G., Klussmann J.P., Wuerdemann N., Wagner S. Plasma Cell-Free Human Papillomavirus Oncogene E6 and E7 DNA Predicts Outcome in Oropharyngeal Squamous Cell Carcinoma. J. Mol. Diagn. JMD. 2020;22:1333–1343. doi: 10.1016/j.jmoldx.2020.08.002.
    1. Akashi K., Sakai T., Fukuoka O., Saito Y., Yoshida M., Ando M., Ito T., Murakami Y., Yamasoba T. Usefulness of circulating tumor DNA by targeting human papilloma virus-derived sequences as a biomarker in p16-positive oropharyngeal cancer. Sci. Rep. 2022;12:572. doi: 10.1038/s41598-021-04307-3.
    1. Rulach R., Zhou S., Hendry F., Stobo D., James A., Dempsey M.F., Grose D., Lamb C., Schipani S., Rizwanullah M., et al. 12 week PET-CT has low positive predictive value for nodal residual disease in human papillomavirus-positive oropharyngeal cancers. Oral. Oncol. 2019;97:76–81. doi: 10.1016/j.oraloncology.2019.08.011.
    1. Vainshtein J.M., Spector M.E., Stenmark M.H., Bradford C.R., Wolf G.T., Worden F.P., Chepeha D.B., McHugh J.B., Carey T., Wong K.K., et al. Reliability of post-chemoradiotherapy F-18-FDG PET/CT for prediction of locoregional failure in human papillomavirus-associated oropharyngeal cancer. Oral. Oncol. 2014;50:234–239. doi: 10.1016/j.oraloncology.2013.12.003.
    1. Lee J.Y., Garcia-Murillas I., Cutts R.J., De Castro D.G., Grove L., Hurley T., Wang F., Nutting C., Newbold K., Harrington K., et al. Predicting response to radical (chemo)radiotherapy with circulating HPV DNA in locally advanced head and neck squamous carcinoma. Br. J. Cancer. 2017;117:876–883. doi: 10.1038/bjc.2017.258.
    1. Kim K.Y., Le Q.T., Yom S.S., Pinsky B.A., Bratman S.V., Ng R.H., El Mubarak H.S., Chan K.C., Sander M., Conley B.A. Current State of PCR-Based Epstein-Barr Virus DNA Testing for Nasopharyngeal Cancer. J. Natl. Cancer Inst. 2017;109:djx007. doi: 10.1093/jnci/djx007.
    1. Chen W.J., Xu W.N., Wang H.Y., Chen X.X., Li X.Q., Xie S.H., Lin D.F., Cao S.M. Plasma Epstein-Barr virus DNA and risk of nasopharyngeal carcinoma in a prospective seropositive population. BMC Cancer. 2021;21:651. doi: 10.1186/s12885-021-08408-0.
    1. Peng H., Li Z., Long Y., Li J., Liu Z., Zhou R. Clinical value of a plasma Epstein-Barr virus DNA assay in the diagnosis of recurrent or metastatic nasopharyngeal carcinoma: A meta-analysis. Biosci. Rep. 2019;39:BSR20190691. doi: 10.1042/BSR20190691.
    1. Lee A.W.M., Lee V.H.F., Ng W.T., Strojan P., Saba N.F., Rinaldo A., Willems S.M., Rodrigo J.P., Forastiere A.A., Ferlito A. A systematic review and recommendations on the use of plasma EBV DNA for nasopharyngeal carcinoma. Eur. J. Cancer. 2021;153:109–122. doi: 10.1016/j.ejca.2021.05.022.
    1. Lo Y.M., Chan L.Y., Lo K.W., Leung S.F., Zhang J., Chan A.T., Lee J.C., Hjelm N.M., Johnson P.J., Huang D.P. Quantitative analysis of cell-free Epstein-Barr virus DNA in plasma of patients with nasopharyngeal carcinoma. Cancer Res. 1999;59:1188–1191.
    1. Krishna S.M., James S., Kattoor J., Balaram P. Serum EBV DNA as a biomarker in primary nasopharyngeal carcinoma of Indian origin. Jpn. J. Clin. Oncol. 2004;34:307–311. doi: 10.1093/jjco/hyh055.
    1. Leung S.F., Tam J.S., Chan A.T., Zee B., Chan L.Y., Huang D.P., Van Hasselt A., Johnson P.J., Lo Y.M. Improved accuracy of detection of nasopharyngeal carcinoma by combined application of circulating Epstein-Barr virus DNA and anti-Epstein-Barr viral capsid antigen IgA antibody. Clin. Chem. 2004;50:339–345. doi: 10.1373/clinchem.2003.022426.
    1. Shao J.Y., Li Y.H., Gao H.Y., Wu Q.L., Cui N.J., Zhang L., Cheng G., Hu L.F., Ernberg I., Zeng Y.X. Comparison of plasma Epstein-Barr virus (EBV) DNA levels and serum EBV immunoglobulin A/virus capsid antigen antibody titers in patients with nasopharyngeal carcinoma. Cancer. 2004;100:1162–1170. doi: 10.1002/cncr.20099.
    1. Tay J.K., Siow C.H., Goh H.L., Lim C.M., Hsu P.P., Chan S.H., Loh K.S. A comparison of EBV serology and serum cell-free DNA as screening tools for nasopharyngeal cancer: Results of the Singapore NPC screening cohort. Int. J. Cancer. 2020;146:2923–2931. doi: 10.1002/ijc.32774.
    1. Ji M.F., Huang Q.H., Yu X., Liu Z., Li X., Zhang L.F., Wang P., Xie S.H., Rao H.L., Fang F., et al. Evaluation of plasma Epstein-Barr virus DNA load to distinguish nasopharyngeal carcinoma patients from healthy high-risk populations in Southern China. Cancer. 2014;120:1353–1360. doi: 10.1002/cncr.28564.
    1. Chan K.C.A., Woo J.K.S., King A., Zee B.C.Y., Lam W.K.J., Chan S.L., Chu S.W.I., Mak C., Tse I.O.L., Leung S.Y.M., et al. Analysis of Plasma Epstein–Barr Virus DNA to Screen for Nasopharyngeal Cancer. N. Engl. J. Med. 2017;377:513–522. doi: 10.1056/NEJMoa1701717.
    1. Miller J.A., Le Q.-T., Pinsky B.A., Wang H. Cost-Effectiveness of Nasopharyngeal Carcinoma Screening with Epstein-Barr Virus Polymerase Chain Reaction or Serology in High-Incidence Populations Worldwide. JNCI J. Natl. Cancer Inst. 2021;113:852–862. doi: 10.1093/jnci/djaa198.
    1. Trevisiol C., Gion M., Vaona A., Fabricio A.S.C., Roca E., Licitra L., Alfieri S., Bossi P. The appropriate use of circulating EBV-DNA in nasopharyngeal carcinoma: Comprehensive clinical practice guidelines evaluation. Oral. Oncol. 2021;114:105128. doi: 10.1016/j.oraloncology.2020.105128.
    1. Alfieri S., Iacovelli N.A., Marceglia S., Lasorsa I., Resteghini C., Taverna F., Mazzocchi A., Orlandi E., Guzzo M., Bianchi R., et al. Circulating pre-treatment Epstein-Barr virus DNA as prognostic factor in locally-advanced nasopharyngeal cancer in a non-endemic area. Oncotarget. 2017;8:47780–47789. doi: 10.18632/oncotarget.17822.
    1. Wei Z.G., Hu X.L., He Y., Guan H., Wang J.J., He L., Mu X.L., Liu Z.R., Li R.D., Peng X.C. Clinical and survival analysis of nasopharyngeal carcinoma with consistently negative Epstein-Barr virus DNA. Head Neck. 2021;43:1465–1475. doi: 10.1002/hed.26608.
    1. Huang C.L., Sun Z.Q., Guo R., Liu X., Mao Y.P., Peng H., Tian L., Lin A.H., Li L., Shao J.Y., et al. Plasma Epstein-Barr Virus DNA Load After Induction Chemotherapy Predicts Outcome in Locoregionally Advanced Nasopharyngeal Carcinoma. Int. J. Radiat. Oncol. Biol. Phys. 2019;104:355–361. doi: 10.1016/j.ijrobp.2019.01.007.
    1. Zhao F.P., Liu X., Chen X.M., Lu J., Yu B.L., Tian W.D., Wang L.U., Xu X., Huang H.R., Zhang M.W., et al. Levels of plasma Epstein-Barr virus DNA prior and subsequent to treatment predicts the prognosis of nasopharyngeal carcinoma. Oncol. Lett. 2015;10:2888–2894. doi: 10.3892/ol.2015.3628.
    1. Chen C., Xu T., Qiu X., Xie S., You Z., Hu Y., Zheng Y., Liang Z., Huang C., Chen T., et al. Selectively recommend 18 F-FDG PET/CT for patients with de novo nasopharyngeal carcinoma in endemic areas. Radiat. Oncol. 2021;16:229. doi: 10.1186/s13014-021-01954-8.
    1. Liu L.T., Liang Y.J., Guo S.S., Xie Y., Jia G.D., Wen D.X., Tang L.Q., Chen Q.Y., Mai H.Q. Identifying distinct risks of treatment failure in nasopharyngeal carcinoma: Study based on the dynamic changes in peripheral blood lymphocytes, monocytes, N classification, and plasma Epstein-Barr virus DNA. Head Neck. 2022;44:34–45. doi: 10.1002/hed.26897.
    1. Lee V.H.F., Kwong D.L.W., Leung T.W., Choi C.W., Lai V., Ng L., Lam K.O., Ng S.C.Y., Sze C.K., Tong C.C., et al. Prognostication of serial post-intensity-modulated radiation therapy undetectable plasma EBV DNA for nasopharyngeal carcinoma. Oncotarget. 2017;8:5292–5308. doi: 10.18632/ONCOTARGET.14137.
    1. Liu T.B., Zheng Z.H., Pan J., Pan L.L., Chen L.H. Prognostic role of plasma Epstein-Barr virus DNA load for nasopharyngeal carcinoma: A meta-analysis. Clin. Investig. Med. 2017;40:E1–E12. doi: 10.25011/cim.v40i1.28049.
    1. Lin J.C., Wang W.Y., Chen K.Y., Wei Y.H., Liang W.M., Jan J.S., Jiang R.S. Quantification of plasma Epstein-Barr virus DNA in patients with advanced nasopharyngeal carcinoma. N. Engl. J. Med. 2004;350:2461–2470. doi: 10.1056/NEJMoa032260.
    1. Pramanik R., Arora S., Sharma P., Biswas A., Nayak B., Thakar A., Sharma A., Ghose S. Cell-free EBV DNA as a biomarker during clinical management of nasopharyngeal carcinoma in a nonendemic region. J. Med. Virol. 2022;94:720–728. doi: 10.1002/jmv.27440.
    1. To E.W., Chan K.C., Leung S.F., Chan L.Y., To K.F., Chan A.T., Johnson P.J., Lo Y.M. Rapid clearance of plasma Epstein-Barr virus DNA after surgical treatment of nasopharyngeal carcinoma. Clin. Cancer Res. 2003;9:3254–3259.
    1. Lo Y.M., Chan L.Y., Chan A.T., Leung S.F., Lo K.W., Zhang J., Lee J.C., Hjelm N.M., Johnson P.J., Huang D.P. Quantitative and temporal correlation between circulating cell-free Epstein-Barr virus DNA and tumor recurrence in nasopharyngeal carcinoma. Cancer Res. 1999;59:5452–5455.
    1. Leung S.F., Chan A.T., Zee B., Ma B., Chan L.Y., Johnson P.J., Lo Y.M. Pretherapy quantitative measurement of circulating Epstein-Barr virus DNA is predictive of posttherapy distant failure in patients with early-stage nasopharyngeal carcinoma of undifferentiated type. Cancer. 2003;98:288–291. doi: 10.1002/cncr.11496.
    1. Chen M., Yin L., Wu J., Gu J.J., Jiang X.S., Wang D.J., Zong D., Guo C., Zhu H.F., Wu J.F., et al. Impact of plasma Epstein-Barr virus-DNA and tumor volume on prognosis of locally advanced nasopharyngeal carcinoma. BioMed Res. Int. 2015;2015:617949. doi: 10.1155/2015/617949.
    1. Ferrari D., Codeca C., Bertuzzi C., Broggio F., Crepaldi F., Luciani A., Floriani I., Ansarin M., Chiesa F., Alterio D., et al. Role of plasma EBV DNA levels in predicting recurrence of nasopharyngeal carcinoma in a Western population. BMC Cancer. 2012;12:208. doi: 10.1186/1471-2407-12-208.
    1. Leung S.F., Chan K.C., Ma B.B., Hui E.P., Mo F., Chow K.C., Leung L., Chu K.W., Zee B., Lo Y.M., et al. Plasma Epstein-Barr viral DNA load at midpoint of radiotherapy course predicts outcome in advanced-stage nasopharyngeal carcinoma. Ann. Oncol. 2014;25:1204–1208. doi: 10.1093/annonc/mdu117.
    1. Chan S.K., Chan S.Y., Choi H.C., Tong C.C., Lam K.O., Kwong D.L., Vardhanabhuti V., Leung T.W., Luk M.Y., Lee A.W., et al. Prognostication of Half-Life Clearance of Plasma EBV DNA in Previously Untreated Non-metastatic Nasopharyngeal Carcinoma Treated with Radical Intensity-Modulated Radiation Therapy. Front. Oncol. 2020;10:1417. doi: 10.3389/fonc.2020.01417.
    1. Hui E.P., Ma B.B.Y., Jacky Lam W.K., Allen Chan K.C., Mo F., Hemis Ai Q.Y., King A.D., Wong C.H., Wong K.C.W., Lam D.C.M., et al. Dynamic Changes of Post-Radiotherapy Plasma Epstein-Barr Virus DNA in a Randomized Trial of Adjuvant Chemotherapy Versus Observation in Nasopharyngeal Cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2021;27:2827–2836. doi: 10.1158/1078-0432.CCR-20-3519.
    1. Cancer Genome Atlas Network Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature. 2015;517:576–582. doi: 10.1038/nature14129.
    1. Zilberg C., Lee M.W., Yu B., Ashford B., Kraitsek S., Ranson M., Shannon K., Cowley M., Iyer N.G., Palme C.E., et al. Analysis of clinically relevant somatic mutations in high-risk head and neck cutaneous squamous cell carcinoma. Mod. Pathol. 2018;31:275–287. doi: 10.1038/modpathol.2017.128.
    1. Liu C.J., Liu T.Y., Kuo L.T., Cheng H.W., Chu T.H., Chang K.W., Lin S.C. Differential gene expression signature between primary and metastatic head and neck squamous cell carcinoma. J. Pathol. 2008;214:489–497. doi: 10.1002/path.2306.
    1. Liu C.J., Lin S.C., Chen Y.J., Chang K.M., Chang K.W. Array-comparative genomic hybridization to detect genomewide changes in microdissected primary and metastatic oral squamous cell carcinomas. Mol. Carcinog. 2006;45:721–731. doi: 10.1002/mc.20213.
    1. Lin S.C., Chen Y.J., Kao S.Y., Hsu M.T., Lin C.H., Yang S.C., Liu T.Y., Chang K.W. Chromosomal changes in betel-associated oral squamous cell carcinomas and their relationship to clinical parameters. Oral. Oncol. 2002;38:266–273. doi: 10.1016/S1368-8375(01)00054-9.
    1. Durinck S., Ho C., Wang N.J., Liao W., Jakkula L.R., Collisson E.A., Pons J., Chan S.W., Lam E.T., Chu C., et al. Temporal dissection of tumorigenesis in primary cancers. Cancer Discov. 2011;1:137–143. doi: 10.1158/-11-0028.
    1. Croce C.M. Oncogenes and cancer. N. Engl. J. Med. 2008;358:502–511. doi: 10.1056/NEJMra072367.
    1. Chang K.W., Sarraj S., Lin S.C., Tsai P.I., Solt D. P53 expression, p53 and Ha-ras mutation and telomerase activation during nitrosamine-mediated hamster pouch carcinogenesis. Carcinogenesis. 2000;21:1441–1451.
    1. Cancer Genome Atlas Research N., Weinstein J.N., Collisson E.A., Mills G.B., Shaw K.R., Ozenberger B.A., Ellrott K., Shmulevich I., Sander C., Stuart J.M. The Cancer Genome Atlas Pan-Cancer analysis project. Nat. Genet. 2013;45:1113–1120.
    1. Agrawal N., Frederick M.J., Pickering C.R., Bettegowda C., Chang K., Li R.J., Fakhry C., Xie T.X., Zhang J., Wang J., et al. Exome sequencing of head and neck squamous cell carcinoma reveals inactivating mutations in NOTCH1. Science. 2011;333:1154–1157. doi: 10.1126/science.1206923.
    1. Aung K.L., Siu L.L. Genomically personalized therapy in head and neck cancer. Cancers Head Neck. 2016;1:2. doi: 10.1186/s41199-016-0004-y.
    1. Coulet F., Blons H., Cabelguenne A., Lecomte T., Lacourreye O., Brasnu D., Beaune P., Zucman J., Laurent-Puig P. Detection of plasma tumor DNA in head and neck squamous cell carcinoma by microsatellite typing and p53 mutation analysis. Cancer Res. 2000;60:707–711.
    1. Hudeckova M., Koucky V., Rottenberg J., Gal B. Gene Mutations in Circulating Tumour DNA as a Diagnostic and Prognostic Marker in Head and Neck Cancer-A Systematic Review. Biomedicines. 2021;9:1548. doi: 10.3390/biomedicines9111548.
    1. Perdomo S., Avogbe P.H., Foll M., Abedi-Ardekani B., Facciolla V.L., Anantharaman D., Chopard P., Calvez-Kelm F.L., Vilensky M., Polesel J., et al. Circulating tumor DNA detection in head and neck cancer: Evaluation of two different detection approaches. Oncotarget. 2017;8:72621–72632. doi: 10.18632/oncotarget.20004.
    1. Galot R., van Marcke C., Helaers R., Mendola A., Goebbels R.M., Caignet X., Ambroise J., Wittouck K., Vikkula M., Limaye N., et al. Liquid biopsy for mutational profiling of locoregional recurrent and/or metastatic head and neck squamous cell carcinoma. Oral. Oncol. 2020;104:104631. doi: 10.1016/j.oraloncology.2020.104631.
    1. Mes S.W., Brink A., Sistermans E.A., Straver R., Oudejans C.B.M., Poell J.B., Leemans C.R., Brakenhoff R.H. Comprehensive multiparameter genetic analysis improves circulating tumor DNA detection in head and neck cancer patients. Oral. Oncol. 2020;109:104852. doi: 10.1016/j.oraloncology.2020.104852.
    1. Kogo R., Manako T., Iwaya T., Nishizuka S., Hiraki H., Sasaki Y., Idogawa M., Tokino T., Koide A., Komune N., et al. Individualized circulating tumor DNA monitoring in head and neck squamous cell carcinoma. Cancer Med. 2022. early view .
    1. Schmidt H., Kulasinghe A., Allcock R.J.N., Tan L.Y., Mokany E., Kenny L., Punyadeera C. A Pilot Study to Non-Invasively Track PIK3CA Mutation in Head and Neck Cancer. Diagnostics. 2018;8:79. doi: 10.3390/diagnostics8040079.
    1. Bidard F.C., Madic J., Mariani P., Piperno-Neumann S., Rampanou A., Servois V., Cassoux N., Desjardins L., Milder M., Vaucher I., et al. Detection rate and prognostic value of circulating tumor cells and circulating tumor DNA in metastatic uveal melanoma. Int. J. Cancer. 2014;134:1207–1213. doi: 10.1002/ijc.28436.
    1. Schwaederle M., Husain H., Fanta P.T., Piccioni D.E., Kesari S., Schwab R.B., Patel S.P., Harismendy O., Ikeda M., Parker B.A., et al. Use of Liquid Biopsies in Clinical Oncology: Pilot Experience in 168 Patients. Clin. Cancer Res. 2016;22:5497–5505. doi: 10.1158/1078-0432.CCR-16-0318.
    1. Yang X., Zhuo M., Ye X., Bai H., Wang Z., Sun Y., Zhao J., An T., Duan J., Wu M., et al. Quantification of mutant alleles in circulating tumor DNA can predict survival in lung cancer. Oncotarget. 2016;7:20810–20824. doi: 10.18632/oncotarget.8021.
    1. Tie J., Wang Y., Tomasetti C., Li L., Springer S., Kinde I., Silliman N., Tacey M., Wong H.L., Christie M., et al. Circulating tumor DNA analysis detects minimal residual disease and predicts recurrence in patients with stage II colon cancer. Sci. Transl. Med. 2016;8:346ra392. doi: 10.1126/scitranslmed.aaf6219.
    1. Sausen M., Phallen J., Adleff V., Jones S., Leary R.J., Barrett M.T., Anagnostou V., Parpart-Li S., Murphy D., Kay Li Q., et al. Clinical implications of genomic alterations in the tumour and circulation of pancreatic cancer patients. Nat. Commun. 2015;6:7686. doi: 10.1038/ncomms8686.
    1. Beaver J.A., Jelovac D., Balukrishna S., Cochran R., Croessmann S., Zabransky D.J., Wong H.Y., Toro P.V., Cidado J., Blair B.G., et al. Detection of cancer DNA in plasma of patients with early-stage breast cancer. Clin. Cancer Res. 2014;20:2643–2650. doi: 10.1158/1078-0432.CCR-13-2933.
    1. Porter A., Natsuhara M., Daniels G.A., Patel S.P., Sacco A.G., Bykowski J., Banks K.C., Cohen E.E.W. Next generation sequencing of cell free circulating tumor DNA in blood samples of recurrent and metastatic head and neck cancer patients. Transl. Cancer Res. 2020;9:203–209. doi: 10.21037/tcr.2019.12.70.
    1. Pall A.H., Jakobsen K.K., Grønhøj C., von Buchwald C. Circulating tumour DNA alterations as biomarkers for head and neck cancer: A systematic review. Acta Oncol. 2020;59:845–850. doi: 10.1080/0284186X.2020.1742930.
    1. Wong T.S., Man M.W., Lam A.K., Wei W.I., Kwong Y.L., Yuen A.P. The study of p16 and p15 gene methylation in head and neck squamous cell carcinoma and their quantitative evaluation in plasma by real-time PCR. Eur. J. Cancer. 2003;39:1881–1887. doi: 10.1016/S0959-8049(03)00428-3.
    1. Mydlarz W.K., Hennessey P.T., Wang H., Carvalho A.L., Califano J.A. Serum biomarkers for detection of head and neck squamous cell carcinoma. Head Neck. 2016;38:9–14. doi: 10.1002/hed.23842.
    1. Ovchinnikov D.A., Cooper M.A., Pandit P., Coman W.B., Cooper-White J.J., Keith P., Wolvetang E.J., Slowey P.D., Punyadeera C. Tumor-suppressor Gene Promoter Hypermethylation in Saliva of Head and Neck Cancer Patients. Transl. Oncol. 2012;5:321–326. doi: 10.1593/tlo.12232.
    1. Righini C.A., de Fraipont F., Timsit J.F., Faure C., Brambilla E., Reyt E., Favrot M.C. Tumor-specific methylation in saliva: A promising biomarker for early detection of head and neck cancer recurrence. Clin. Cancer Res. 2007;13:1179–1185. doi: 10.1158/1078-0432.CCR-06-2027.
    1. Tian F., Yip S.P., Kwong D.L., Lin Z., Yang Z., Wu V.W. Promoter hypermethylation of tumor suppressor genes in serum as potential biomarker for the diagnosis of nasopharyngeal carcinoma. Cancer Epidemiol. 2013;37:708–713. doi: 10.1016/j.canep.2013.05.012.
    1. Yang X., Dai W., Kwong D.L., Szeto C.Y., Wong E.H., Ng W.T., Lee A.W., Ngan R.K., Yau C.C., Tung S.Y., et al. Epigenetic markers for noninvasive early detection of nasopharyngeal carcinoma by methylation-sensitive high resolution melting. Int. J. Cancer. 2015;136:E127–E135. doi: 10.1002/ijc.29192.
    1. Thakur B.K., Zhang H., Becker A., Matei I., Huang Y., Costa-Silva B., Zheng Y., Hoshino A., Brazier H., Xiang J., et al. Double-stranded DNA in exosomes: A novel biomarker in cancer detection. Cell Res. 2014;24:766–769. doi: 10.1038/cr.2014.44.
    1. Li S., Yi M., Dong B., Tan X., Luo S., Wu K. The role of exosomes in liquid biopsy for cancer diagnosis and prognosis prediction. Int. J. Cancer. 2021;148:2640–2651. doi: 10.1002/ijc.33386.
    1. Ludwig S., Sharma P., Theodoraki M.N., Pietrowska M., Yerneni S.S., Lang S., Ferrone S., Whiteside T.L. Molecular and Functional Profiles of Exosomes From HPV(+) and HPV(−) Head and Neck Cancer Cell Lines. Front. Oncol. 2018;8:445. doi: 10.3389/fonc.2018.00445.
    1. Ludwig N., Yerneni S.S., Razzo B.M., Whiteside T.L. Exosomes from HNSCC Promote Angiogenesis through Reprogramming of Endothelial Cells. Mol. Cancer Res. MCR. 2018;16:1798–1808. doi: 10.1158/1541-7786.MCR-18-0358.
    1. Muller L., Hong C.S., Stolz D.B., Watkins S.C., Whiteside T.L. Isolation of biologically-active exosomes from human plasma. J. Immunol. Methods. 2014;411:55–65. doi: 10.1016/j.jim.2014.06.007.
    1. Ferris R.L. Immunology and Immunotherapy of Head and Neck Cancer. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2015;33:3293–3304. doi: 10.1200/JCO.2015.61.1509.
    1. Gavrielatou N., Doumas S., Economopoulou P., Foukas P.G., Psyrri A. Biomarkers for immunotherapy response in head and neck cancer. Cancer Treat. Rev. 2020;84:101977. doi: 10.1016/j.ctrv.2020.101977.
    1. Theodoraki M.N., Yerneni S.S., Hoffmann T.K., Gooding W.E., Whiteside T.L. Clinical Significance of PD-L1(+) Exosomes in Plasma of Head and Neck Cancer Patients. Clin. Cancer Res. 2018;24:896–905. doi: 10.1158/1078-0432.CCR-17-2664.
    1. Koster H.J., Rojalin T., Powell A., Pham D., Mizenko R.R., Birkeland A.C., Carney R.P. Surface enhanced Raman scattering of extracellular vesicles for cancer diagnostics despite isolation dependent lipoprotein contamination. Nanoscale. 2021;13:14760–14776. doi: 10.1039/D1NR03334D.
    1. Faur C.I., Rotaru H., Osan C., Jurj A., Roman R.C., Moldovan M., Chirila M., Hedesiu M. Salivary exosomal microRNAs as biomarkers for head and neck cancer detection-a literature review. Maxillofac. Plast. Reconstr. Surg. 2021;43:19. doi: 10.1186/s40902-021-00303-9.
    1. He L., Ping F., Fan Z., Zhang C., Deng M., Cheng B., Xia J. Salivary exosomal miR-24-3p serves as a potential detective biomarker for oral squamous cell carcinoma screening. Biomed. Pharmacother. 2020;121:109553. doi: 10.1016/j.biopha.2019.109553.
    1. Cristaldi M., Mauceri R., Di Fede O., Giuliana G., Campisi G., Panzarella V. Salivary Biomarkers for Oral Squamous Cell Carcinoma Diagnosis and Follow-Up: Current Status and Perspectives. Front. Physiol. 2019;10:1476. doi: 10.3389/fphys.2019.01476.
    1. Langevin S., Kuhnell D., Parry T., Biesiada J., Huang S., Wise-Draper T., Casper K., Zhang X., Medvedovic M., Kasper S. Comprehensive microRNA-sequencing of exosomes derived from head and neck carcinoma cells in vitro reveals common secretion profiles and potential utility as salivary biomarkers. Oncotarget. 2017;8:82459–82474. doi: 10.18632/oncotarget.19614.
    1. Salazar C., Nagadia R., Pandit P., Cooper-White J., Banerjee N., Dimitrova N., Coman W.B., Punyadeera C. A novel saliva-based microRNA biomarker panel to detect head and neck cancers. Cell. Oncol. 2014;37:331–338. doi: 10.1007/s13402-014-0188-2.
    1. Liu C.J., Kao S.Y., Tu H.F., Tsai M.M., Chang K.W., Lin S.C. Increase of microRNA miR-31 level in plasma could be a potential marker of oral cancer. Oral. Dis. 2010;16:360–364. doi: 10.1111/j.1601-0825.2009.01646.x.
    1. Wang H.M., Wu M.H., Chang P.H., Lin H.C., Liao C.D., Wu S.M., Hung T.M., Lin C.Y., Chang T.C., Tzu-Tsen Y., et al. The change in circulating tumor cells before and during concurrent chemoradiotherapy is associated with survival in patients with locally advanced head and neck cancer. Head Neck. 2019;41:2676–2687. doi: 10.1002/hed.25744.
    1. Hristozova T., Konschak R., Stromberger C., Fusi A., Liu Z., Weichert W., Stenzinger A., Budach V., Keilholz U., Tinhofer I. The presence of circulating tumor cells (CTCs) correlates with lymph node metastasis in nonresectable squamous cell carcinoma of the head and neck region (SCCHN) Ann. Oncol. 2011;22:1878–1885. doi: 10.1093/annonc/mdr130.
    1. Jatana K.R., Balasubramanian P., Lang J.C., Yang L., Jatana C.A., White E., Agrawal A., Ozer E., Schuller D.E., Teknos T.N., et al. Significance of circulating tumor cells in patients with squamous cell carcinoma of the head and neck: Initial results. Arch. Otolaryngol. Head Neck Surg. 2010;136:1274–1279. doi: 10.1001/archoto.2010.223.
    1. Tinhofer I., Konschak R., Stromberger C., Raguse J.D., Dreyer J.H., Jöhrens K., Keilholz U., Budach V. Detection of circulating tumor cells for prediction of recurrence after adjuvant chemoradiation in locally advanced squamous cell carcinoma of the head and neck. Ann. Oncol. 2014;25:2042–2047. doi: 10.1093/annonc/mdu271.
    1. Liu H.E., Triboulet M., Zia A., Vuppalapaty M., Kidess-Sigal E., Coller J., Natu V.S., Shokoohi V., Che J., Renier C., et al. Workflow optimization of whole genome amplification and targeted panel sequencing for CTC mutation detection. NPJ Genom. Med. 2017;2:34. doi: 10.1038/s41525-017-0034-3.
    1. Onidani K., Shoji H., Kakizaki T., Yoshimoto S., Okaya S., Miura N., Sekikawa S., Furuta K., Lim C.T., Shibahara T., et al. Monitoring of cancer patients via next-generation sequencing of patient-derived circulating tumor cells and tumor DNA. Cancer Sci. 2019;110:2590–2599. doi: 10.1111/cas.14092.
    1. Bardelli A., Pantel K. Liquid Biopsies, What We Do Not Know (Yet) Cancer Cell. 2017;31:172–179. doi: 10.1016/j.ccell.2017.01.002.
    1. Liebs S., Eder T., Klauschen F., Schütte M., Yaspo M.-L., Keilholz U., Tinhofer I., Kidess-Sigal E., Braunholz D. Applicability of liquid biopsies to represent the mutational profile of tumor tissue from different cancer entities. Oncogene. 2021;40:5204–5212. doi: 10.1038/s41388-021-01928-w.
    1. Russano M., Napolitano A., Ribelli G., Iuliani M., Simonetti S., Citarella F., Pantano F., Dell’Aquila E., Anesi C., Silvestris N., et al. Liquid biopsy and tumor heterogeneity in metastatic solid tumors: The potentiality of blood samples. J. Exp. Clin. Cancer Res. CR. 2020;39:95. doi: 10.1186/s13046-020-01601-2.
    1. Chu D., Paoletti C., Gersch C., VanDenBerg D.A., Zabransky D.J., Cochran R.L., Wong H.Y., Toro P.V., Cidado J., Croessmann S., et al. ESR1 Mutations in Circulating Plasma Tumor DNA from Metastatic Breast Cancer Patients. Clin. Cancer Res. 2016;22:993–999. doi: 10.1158/1078-0432.CCR-15-0943.
    1. Chin R.I., Chen K., Usmani A., Chua C., Harris P.K., Binkley M.S., Azad T.D., Dudley J.C., Chaudhuri A.A. Detection of Solid Tumor Molecular Residual Disease (MRD) Using Circulating Tumor DNA (ctDNA) Mol. Diagn. Ther. 2019;23:311–331. doi: 10.1007/s40291-019-00390-5.
    1. Radovich M., Jiang G., Hancock B.A., Chitambar C., Nanda R., Falkson C., Lynce F.C., Gallagher C., Isaacs C., Blaya M., et al. Association of Circulating Tumor DNA and Circulating Tumor Cells After Neoadjuvant Chemotherapy with Disease Recurrence in Patients with Triple-Negative Breast Cancer: Preplanned Secondary Analysis of the BRE12-158 Randomized Clinical Trial. JAMA Oncol. 2020;6:1410–1415. doi: 10.1001/jamaoncol.2020.2295.
    1. Bu J., Lee T.H., Poellmann M.J., Rawding P.A., Jeong W.-J., Hong R.S., Hyun S.H., Eun H.S., Hong S. Tri-modal liquid biopsy: Combinational analysis of circulating tumor cells, exosomes, and cell-free DNA using machine learning algorithm. Clin. Transl. Med. 2021;11:e499. doi: 10.1002/ctm2.499.
    1. Egyud M., Sridhar P., Devaiah A., Yamada E., Saunders S., Ståhlberg A., Filges S., Krzyzanowski P.M., Kalatskaya I., Jiao W., et al. Plasma circulating tumor DNA as a potential tool for disease monitoring in head and neck cancer. Head Neck. 2019;41:1351–1358. doi: 10.1002/hed.25563.
    1. Hamana K., Uzawa K., Ogawara K., Shiiba M., Bukawa H., Yokoe H., Tanzawa H. Monitoring of circulating tumour-associated DNA as a prognostic tool for oral squamous cell carcinoma. Br. J. Cancer. 2005;92:2181–2184. doi: 10.1038/sj.bjc.6602635.
    1. van Ginkel J.H., Huibers M.M.H., Noorlag R., de Bree R., van Es R.J.J., Willems S.M. Liquid Biopsy: A Future Tool for Posttreatment Surveillance in Head and Neck Cancer? Pathobiol. J. Immunopathol. Mol. Cell. Biol. 2017;84:115–120. doi: 10.1159/000452861.
    1. Memorial Sloan Kettering Cancer Center A Study on Using Cell-Free Tumor DNA (ctDNA) Testing to Decide When to StartRoutine Treatment in People with Human Papilloma Virus (HPV)-Associated Oropharynx Cancer (OPC). Identifier: NCT05307939. Updated April 11, 2022. [(accessed on 6 June 2022)]; Available online: .
    1. von Buchwald C. Cell-free Tumor DNA in Head and Neck Cancer Patients. Identifier: NCT03942380. Updated September 21, 2021. [(accessed on 6 June 2022)]; Available online: .
    1. Schoenfeld J.D. Risk-Adapted Therapy in Low-Risk HPV+ Oropharyngeal Cancer Using Circulating Tumor (ct)HPV DNA Profile—The ReACT Study. Identifier: NCT04900623. Updated July 7, 2021. [(accessed on 6 June 2022)]; Available online: .
    1. Chaudhuri A.A. Circulating Tumor DNA (ctDNA) for Early Treatment Response Assessment of Solid Tumors. Identifier: NCT04354064. Updated April 12, 2022. [(accessed on 6 June 2022)]; Available online: .
    1. Berger B., Hanna G.J., Posner M., Genden E., Del Vecchio Fitz C., Naber S.P., Kuperwasser C. Detection of Occult Recurrence Using Circulating HPV Tumor DNA Among Patients Treated for HPV-driven Oropharyngeal Squamous Cell Carcinoma. Int. J. Radiat. Oncol. Biol. Phys. 2022;112:e4. doi: 10.1016/j.ijrobp.2021.12.016.
    1. Pritchett M.A., Camidge D.R., Patel M., Khatri J., Boniol S., Friedman E.K., Khomani A., Dalia S., Baker-Neblett K., Plagnol V., et al. Prospective Clinical Validation of the InVisionFirst-Lung Circulating Tumor DNA Assay for Molecular Profiling of Patients with Advanced Nonsquamous Non-Small-Cell Lung Cancer. JCO Precis. Oncol. 2019;3:1–15. doi: 10.1200/PO.18.00299.
    1. Thierry A.R., Mouliere F., El Messaoudi S., Mollevi C., Lopez-Crapez E., Rolet F., Gillet B., Gongora C., Dechelotte P., Robert B., et al. Clinical validation of the detection of KRAS and BRAF mutations from circulating tumor DNA. Nat. Med. 2014;20:430–435. doi: 10.1038/nm.3511.
    1. Groot V.P., Mosier S., Javed A.A., Teinor J.A., Gemenetzis G., Ding D., Haley L.M., Yu J., Burkhart R.A., Hasanain A., et al. Circulating Tumor DNA as a Clinical Test in Resected Pancreatic Cancer. Clin. Cancer Res. 2019;25:4973–4984. doi: 10.1158/1078-0432.CCR-19-0197.
    1. Merker J.D., Oxnard G.R., Compton C., Diehn M., Hurley P., Lazar A.J., Lindeman N., Lockwood C.M., Rai A.J., Schilsky R.L., 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;36:1631–1641. doi: 10.1200/JCO.2017.76.8671.

Source: PubMed

3
订阅