Liquid biopsy in hepatocellular carcinoma: circulating tumor cells and circulating tumor DNA

Qianwei Ye, Sunbin Ling, Shusen Zheng, Xiao Xu, Qianwei Ye, Sunbin Ling, Shusen Zheng, Xiao Xu

Abstract

Hepatocellular carcinoma (HCC) is one of the most common cancers and a leading cause of death worldwide. Due to latent liver disease, late diagnosis, and nonresponse to systemic treatments, surgical resection and/or biopsy specimens are still generally considered as the gold standard by clinicians for clinical decision-making until now. Since the conventional tissue biopsy is invasive and contains small tissue samples, it is unable to represent tumor heterogeneity or monitor dynamic tumor progression. Therefore, it is imperative to find a new less invasive or noninvasive diagnostic strategy to detect HCC at an early stage and to monitor HCC recurrence. Over the past years, a new diagnostic concept known as "liquid biopsy" has emerged with substantial attention. Liquid biopsy is noninvasive and allows repeated analyses to monitor tumor recurrence, metastasis or treatment responses in real time. With the advanced development of new molecular techniques, HCC circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA) detection have achieved interesting and encouraging results. In this review, we focus on the clinical applications of CTCs and ctDNA as key components of liquid biopsy in HCC patients.

Keywords: Circulating tumor DNA; Circulating tumor cells (CTCs); Clinical application; Hepatocellular carcinoma; Liquid biopsy.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
CTCs and ctDNA in the peripheral blood. CTCs and ctDNA are the essential elements and are widely believed to be the cornerstones of the liquid biopsy. CTCs are cancer cells that circulate in the bloodstream after being naturally shed from the original or metastatic tumors, they are “seeds” of tumors and can lead to a new fatal metastasis; ctDNA is derived from apoptotic and necrotic tumor cells that release their fragmented DNA into the circulation and contain genetic defects identical to the original tumor cells
Fig. 2
Fig. 2
Phantom drawing of clinical applications of liquid biopsy in HCC patients. Monitoring the response and relapse of HCC patients using liquid biopsy, the levels of ctDNA and CTCs correlate well with HCC progression as well as various therapies, including surgical resection, TACE, radiofrequency ablation (RFA) and targeted molecular therapy

References

    1. Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, et al. Cancer statistics in China, 2015. CA Cancer J Clin. 2016;66(2):115–132. doi: 10.3322/caac.21338.
    1. European Association For The Study Of The L, European Organisation For R, Treatment Of C EASL-EORTC clinical practice guidelines: management of hepatocellular carcinoma. J Hepatol. 2012;56(4):908–943. doi: 10.1016/j.jhep.2011.12.001.
    1. Colombo F, Baldan F, Mazzucchelli S, Martin-Padura I, Marighetti P, Cattaneo A, et al. Evidence of distinct tumour-propagating cell populations with different properties in primary human hepatocellular carcinoma. PLoS One. 2011;6(6):e21369. doi: 10.1371/journal.pone.0021369.
    1. Zhou L, Liu J, Luo F. Serum tumor markers for detection of hepatocellular carcinoma. World J Gastroenterol. 2006;12(8):1175–1181. doi: 10.3748/wjg.v12.i8.1175.
    1. Diaz LA, Jr, Bardelli A. Liquid biopsies: genotyping circulating tumor DNA. J Clin Oncol. 2014;32(6):579–586. doi: 10.1200/JCO.2012.45.2011.
    1. Alix-Panabieres C, Pantel K. Clinical applications of circulating tumor cells and circulating tumor DNA as liquid biopsy. Cancer Discov. 2016;6(5):479–491. doi: 10.1158/-15-1483.
    1. Corcoran RB, Chabner BA. Application of cell-free DNA analysis to Cancer treatment. N Engl J Med. 2018;379(18):1754–1765. doi: 10.1056/NEJMra1706174.
    1. Crowley E, Di Nicolantonio F, Loupakis F, Bardelli A. Liquid biopsy: monitoring cancer-genetics in the blood. Nat Rev Clin Oncol. 2013;10(8):472–484. doi: 10.1038/nrclinonc.2013.110.
    1. Ashworth TR. A case of cancer in which cells similar to those in the tumours were seen in the blood after death. Aust Med J. 1869;14:146–149.
    1. van de Stolpe A, Pantel K, Sleijfer S, Terstappen LW, den Toonder JM. Circulating tumor cell isolation and diagnostics: toward routine clinical use. Cancer Res. 2011;71(18):5955–5960. doi: 10.1158/0008-5472.CAN-11-1254.
    1. Meng S, Tripathy D, Frenkel EP, Shete S, Naftalis EZ, Huth JF, et al. Circulating tumor cells in patients with breast cancer dormancy. Clin Cancer Res. 2004;10(24):8152–8162. doi: 10.1158/1078-0432.CCR-04-1110.
    1. Strilic B, Offermanns S. Intravascular survival and extravasation of tumor cells. Cancer Cell. 2017;32(3):282–293. doi: 10.1016/j.ccell.2017.07.001.
    1. Okajima W, Komatsu S, Ichikawa D, Miyamae M, Ohashi T, Imamura T, et al. Liquid biopsy in patients with hepatocellular carcinoma: circulating tumor cells and cell-free nucleic acids. World J Gastroenterol. 2017;23(31):5650–5668. doi: 10.3748/wjg.v23.i31.5650.
    1. van der Vaart M, Pretorius PJ. The origin of circulating free DNA. Clin Chem. 2007;53(12):2215. doi: 10.1373/clinchem.2007.092734.
    1. Stroun M, Lyautey J, Lederrey C, Olson-Sand A, Anker P. About the possible origin and mechanism of circulating DNA apoptosis and active DNA release. Clin Chim Acta. 2001;313(1–2):139–142. doi: 10.1016/S0009-8981(01)00665-9.
    1. Anker P, Stroun M, Maurice PA. Spontaneous release of DNA by human blood lymphocytes as shown in an in vitro system. Cancer Res. 1975;35(9):2375–2382.
    1. Pantel K, Alix-Panabieres C. Real-time liquid biopsy in cancer patients: fact or fiction? Cancer Res. 2013;73(21):6384–6388. doi: 10.1158/0008-5472.CAN-13-2030.
    1. Underhill HR, Kitzman JO, Hellwig S, Welker NC, Daza R, Baker DN, et al. Fragment length of circulating tumor DNA. PLoS Genet. 2016;12(7):e1006162. doi: 10.1371/journal.pgen.1006162.
    1. Mouliere Florent, Chandrananda Dineika, Piskorz Anna M., Moore Elizabeth K., Morris James, Ahlborn Lise Barlebo, Mair Richard, Goranova Teodora, Marass Francesco, Heider Katrin, Wan Jonathan C. M., Supernat Anna, Hudecova Irena, Gounaris Ioannis, Ros Susana, Jimenez-Linan Mercedes, Garcia-Corbacho Javier, Patel Keval, Østrup Olga, Murphy Suzanne, Eldridge Matthew D., Gale Davina, Stewart Grant D., Burge Johanna, Cooper Wendy N., van der Heijden Michiel S., Massie Charles E., Watts Colin, Corrie Pippa, Pacey Simon, Brindle Kevin M., Baird Richard D., Mau-Sørensen Morten, Parkinson Christine A., Smith Christopher G., Brenton James D., Rosenfeld Nitzan. Enhanced detection of circulating tumor DNA by fragment size analysis. Science Translational Medicine. 2018;10(466):eaat4921. doi: 10.1126/scitranslmed.aat4921.
    1. Lo YM, Zhang J, Leung TN, Lau TK, Chang AM, Hjelm NM. Rapid clearance of fetal DNA from maternal plasma. Am J Hum Genet. 1999;64(1):218–224. doi: 10.1086/302205.
    1. Diehl F, Schmidt K, Choti MA, Romans K, Goodman S, Li M, et al. Circulating mutant DNA to assess tumor dynamics. Nat Med. 2008;14(9):985–990. doi: 10.1038/nm.1789.
    1. Chan KC, Jiang P, Zheng YW, Liao GJ, Sun H, Wong J, 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(1):211–224. doi: 10.1373/clinchem.2012.196014.
    1. Yung TK, Chan KC, Mok TS, Tong J, To KF. Lo YM. 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(6):2076–2084. doi: 10.1158/1078-0432.CCR-08-2622.
    1. Diehl F, Li M, Dressman D, He Y, Shen D, Szabo S, et al. Detection and quantification of mutations in the plasma of patients with colorectal tumors. Proc Natl Acad Sci U S A. 2005;102(45):16368–16373. doi: 10.1073/pnas.0507904102.
    1. Qiu M, Wang J, Xu Y, Ding X, Li M, Jiang F, et al. Circulating tumor DNA is effective for the detection of EGFR mutation in non-small cell lung cancer: a meta-analysis. Cancer Epidemiol Biomark Prev. 2015;24(1):206–212. doi: 10.1158/1055-9965.EPI-14-0895.
    1. Freidin MB, Freydina DV, Leung M, Montero Fernandez A, Nicholson AG, Lim E. Circulating tumor DNA outperforms circulating tumor cells for KRAS mutation detection in thoracic malignancies. Clin Chem. 2015;61(10):1299–1304. doi: 10.1373/clinchem.2015.242453.
    1. Wong IH, Lo YM, Zhang J, Liew CT, Ng MH, Wong N, et al. Detection of aberrant p16 methylation in the plasma and serum of liver cancer patients. Cancer Res. 1999;59(1):71–73.
    1. Chan KC, Lai PB, Mok TS, Chan HL, Ding C, Yeung SW, et al. Quantitative analysis of circulating methylated DNA as a biomarker for hepatocellular carcinoma. Clin Chem. 2008;54(9):1528–1536. doi: 10.1373/clinchem.2008.104653.
    1. Chan KC, Jiang P, Chan CW, Sun K, Wong J, Hui EP, et al. Noninvasive detection of cancer-associated genome-wide hypomethylation and copy number aberrations by plasma DNA bisulfite sequencing. Proc Natl Acad Sci U S A. 2013;110(47):18761–18768. doi: 10.1073/pnas.1313995110.
    1. Balgkouranidou I, Chimonidou M, Milaki G, Tsarouxa EG, Kakolyris S, Welch DR, et al. Breast cancer metastasis suppressor-1 promoter methylation in cell-free DNA provides prognostic information in non-small cell lung cancer. Br J Cancer. 2014;110(8):2054–2062. doi: 10.1038/bjc.2014.104.
    1. Lo YM, Chan LY, Lo KW, Leung SF, Zhang J, Chan AT, et al. Quantitative analysis of cell-free Epstein-Barr virus DNA in plasma of patients with nasopharyngeal carcinoma. Cancer Res. 1999;59(6):1188–1191.
    1. Chan KC, Hung EC, Woo JK, Chan PK, Leung SF, Lai FP, et al. Early detection of nasopharyngeal carcinoma by plasma Epstein-Barr virus DNA analysis in a surveillance program. Cancer. 2013;119(10):1838–1844. doi: 10.1002/cncr.28001.
    1. Campitelli M, Jeannot E, Peter M, Lappartient E, Saada S, de la Rochefordiere A, et al. Human papillomavirus mutational insertion: specific marker of circulating tumor DNA in cervical cancer patients. PLoS One. 2012;7(8):e43393. doi: 10.1371/journal.pone.0043393.
    1. Yu JJ, Xiao W, Dong SL, Liang HF, Zhang ZW, Zhang BX, et al. Effect of surgical liver resection on circulating tumor cells in patients with hepatocellular carcinoma. BMC Cancer. 2018;18(1):835. doi: 10.1186/s12885-018-4744-4.
    1. Qi Lu-Nan, Xiang Bang-De, Wu Fei-Xiang, Ye Jia-Zhou, Zhong Jian-Hong, Wang Yan-Yan, Chen Yuan-Yuan, Chen Zu-Shun, Ma Liang, Chen Jie, Gong Wen-Feng, Han Ze-Guang, Lu Yan, Shang Jin-Jie, Li Le-Qun. Circulating Tumor Cells Undergoing EMT Provide a Metric for Diagnosis and Prognosis of Patients with Hepatocellular Carcinoma. Cancer Research. 2018;78(16):4731–4744. doi: 10.1158/0008-5472.CAN-17-2459.
    1. von Felden J, Schulze K, Krech T, Ewald F, Nashan B, Pantel K, et al. Circulating tumor cells as liquid biomarker for high HCC recurrence risk after curative liver resection. Oncotarget. 2017;8(52):89978–89987.
    1. Ogle LF, Orr JG, Willoughby CE, Hutton C, McPherson S, Plummer R, et al. Imagestream detection and characterisation of circulating tumour cells - a liquid biopsy for hepatocellular carcinoma? J Hepatol. 2016;65(2):305–313. doi: 10.1016/j.jhep.2016.04.014.
    1. Zhang Y, Zhang X, Zhang J, Sun B, Zheng L, Li J, et al. Microfluidic chip for isolation of viable circulating tumor cells of hepatocellular carcinoma for their culture and drug sensitivity assay. Cancer Biol Ther. 2016;17(11):1177–1187. doi: 10.1080/15384047.2016.1235665.
    1. Wang S, Zhang C, Wang G, Cheng B, Wang Y, Chen F, et al. Aptamer-mediated transparent-biocompatible nanostructured surfaces for Hepotocellular circulating tumor cells enrichment. Theranostics. 2016;6(11):1877–1886. doi: 10.7150/thno.15284.
    1. Kelley RK, Magbanua MJ, Butler TM, Collisson EA, Hwang J, Sidiropoulos N, et al. Circulating tumor cells in hepatocellular carcinoma: a pilot study of detection, enumeration, and next-generation sequencing in cases and controls. BMC Cancer. 2015;15:206. doi: 10.1186/s12885-015-1195-z.
    1. Choi GH, Kim GI, Yoo JE, Na DC, Han DH, Roh YH, et al. Increased expression of circulating Cancer stem cell markers during the perioperative period predicts early recurrence after curative resection of hepatocellular carcinoma. Ann Surg Oncol. 2015;22(Suppl 3):S1444–S1452. doi: 10.1245/s10434-015-4480-9.
    1. Bahnassy AA, Zekri AR, El-Bastawisy A, Fawzy A, Shetta M, Hussein N, et al. Circulating tumor and cancer stem cells in hepatitis C virus-associated liver disease. World J Gastroenterol. 2014;20(48):18240–18248. doi: 10.3748/wjg.v20.i48.18240.
    1. Guo W, Yang XR, Sun YF, Shen MN, Ma XL, Wu J, et al. Clinical significance of EpCAM mRNA-positive circulating tumor cells in hepatocellular carcinoma by an optimized negative enrichment and qRT-PCR-based platform. Clin Cancer Res. 2014;20(18):4794–4805. doi: 10.1158/1078-0432.CCR-14-0251.
    1. Morris KL, Tugwood JD, Khoja L, Lancashire M, Sloane R, Burt D, et al. Circulating biomarkers in hepatocellular carcinoma. Cancer Chemother Pharmacol. 2014;74(2):323–332. doi: 10.1007/s00280-014-2508-7.
    1. Fang ZT, Zhang W, Wang GZ, Zhou B, Yang GW, Qu XD, et al. Circulating tumor cells in the central and peripheral venous compartment - assessing hematogenous dissemination after transarterial chemoembolization of hepatocellular carcinoma. Onco Targets Ther. 2014;7:1311–1318. doi: 10.2147/OTT.S62605.
    1. Li YM, Xu SC, Li J, Han KQ, Pi HF, Zheng L, et al. Epithelial-mesenchymal transition markers expressed in circulating tumor cells in hepatocellular carcinoma patients with different stages of disease. Cell Death Dis. 2013;4:e831. doi: 10.1038/cddis.2013.347.
    1. Schulze K, Gasch C, Staufer K, Nashan B, Lohse AW, Pantel K, et al. Presence of EpCAM-positive circulating tumor cells as biomarker for systemic disease strongly correlates to survival in patients with hepatocellular carcinoma. Int J Cancer. 2013;133(9):2165–2171. doi: 10.1002/ijc.28230.
    1. Sun YF, Xu Y, Yang XR, Guo W, Zhang X, Qiu SJ, et al. Circulating stem cell-like epithelial cell adhesion molecule-positive tumor cells indicate poor prognosis of hepatocellular carcinoma after curative resection. Hepatology. 2013;57(4):1458–1468. doi: 10.1002/hep.26151.
    1. Liu S, Li N, Yu X, Xiao X, Cheng K, Hu J, et al. Expression of intercellular adhesion molecule 1 by hepatocellular carcinoma stem cells and circulating tumor cells. Gastroenterology. 2013;144(5):1031–1041. doi: 10.1053/j.gastro.2013.01.046.
    1. Xu W, Cao L, Chen L, Li J, Zhang XF, Qian HH, et al. Isolation of circulating tumor cells in patients with hepatocellular carcinoma using a novel cell separation strategy. Clin Cancer Res. 2011;17(11):3783–3793. doi: 10.1158/1078-0432.CCR-10-0498.
    1. Fan ST, Yang ZF, Ho DW, Ng MN, Yu WC, Wong J. Prediction of posthepatectomy recurrence of hepatocellular carcinoma by circulating cancer stem cells: a prospective study. Ann Surg. 2011;254(4):569–576. doi: 10.1097/SLA.0b013e3182300a1d.
    1. Vona G, Estepa L, Beroud C, Damotte D, Capron F, Nalpas B, et al. Impact of cytomorphological detection of circulating tumor cells in patients with liver cancer. Hepatology. 2004;39(3):792–797. doi: 10.1002/hep.20091.
    1. Mou DC, Cai SL, Peng JR, Wang Y, Chen HS, Pang XW, et al. Evaluation of MAGE-1 and MAGE-3 as tumour-specific markers to detect blood dissemination of hepatocellular carcinoma cells. Br J Cancer. 2002;86(1):110–116. doi: 10.1038/sj.bjc.6600016.
    1. Ikeda S, Tsigelny IF, Skjevik AA, Kono Y, Mendler M, Kuo A, et al. Next-generation sequencing of circulating tumor DNA reveals frequent alterations in advanced hepatocellular carcinoma. Oncologist. 2018;23(5):586–593. doi: 10.1634/theoncologist.2017-0479.
    1. Wu HC, Yang HI, Wang Q, Chen CJ, Santella RM. Plasma DNA methylation marker and hepatocellular carcinoma risk prediction model for the general population. Carcinogenesis. 2017;38(10):1021–1028. doi: 10.1093/carcin/bgx078.
    1. Lu CY, Chen SY, Peng HL, Kan PY, Chang WC, Yen CJ. Cell-free methylation markers with diagnostic and prognostic potential in hepatocellular carcinoma. Oncotarget. 2017;8(4):6406–6418.
    1. Liao W, Yang H, Xu H, Wang Y, Ge P, Ren J, et al. Noninvasive detection of tumor-associated mutations from circulating cell-free DNA in hepatocellular carcinoma patients by targeted deep sequencing. Oncotarget. 2016;7(26):40481–40490. doi: 10.18632/oncotarget.9629.
    1. Huang A, Zhang X, Zhou SL, Cao Y, Huang XW, Fan J, et al. Detecting circulating tumor DNA in hepatocellular carcinoma patients using droplet digital PCR is feasible and reflects Intratumoral heterogeneity. J Cancer. 2016;7(13):1907–1914. doi: 10.7150/jca.15823.
    1. Kuo CC, Lin CY, Shih YL, Hsieh CB, Lin PY, Guan SB, et al. Frequent methylation of HOXA9 gene in tumor tissues and plasma samples from human hepatocellular carcinomas. Clin Chem Lab Med. 2014;52(8):1235–1245. doi: 10.1515/cclm-2013-0780.
    1. Ji XF, Fan YC, Gao S, Yang Y, Zhang JJ, Wang K. MT1M and MT1G promoter methylation as biomarkers for hepatocellular carcinoma. World J Gastroenterol. 2014;20(16):4723–4729. doi: 10.3748/wjg.v20.i16.4723.
    1. Huang G, Krocker JD, Kirk JL, Merwat SN, Ju H, Soloway RD, et al. Evaluation of INK4A promoter methylation using pyrosequencing and circulating cell-free DNA from patients with hepatocellular carcinoma. Clin Chem Lab Med. 2014;52(6):899–909. doi: 10.1515/cclm-2013-0885.
    1. Han LY, Fan YC, Mu NN, Gao S, Li F, Ji XF, et al. Aberrant DNA methylation of G-protein-coupled bile acid receptor Gpbar1 (TGR5) is a potential biomarker for hepatitis B virus associated hepatocellular carcinoma. Int J Med Sci. 2014;11(2):164–171. doi: 10.7150/ijms.6745.
    1. Zhang P, Wen X, Gu F, Deng X, Li J, Dong J, et al. Methylation profiling of serum DNA from hepatocellular carcinoma patients using an Infinium human methylation 450 BeadChip. Hepatol Int. 2013;7(3):893–900. doi: 10.1007/s12072-013-9437-0.
    1. Sun FK, Fan YC, Zhao J, Zhang F, Gao S, Zhao ZH, et al. Detection of TFPI2 methylation in the serum of hepatocellular carcinoma patients. Dig Dis Sci. 2013;58(4):1010–1015. doi: 10.1007/s10620-012-2462-3.
    1. Piciocchi M, Cardin R, Vitale A, Vanin V, Giacomin A, Pozzan C, et al. Circulating free DNA in the progression of liver damage to hepatocellular carcinoma. Hepatol Int. 2013;7(4):1050–1057. doi: 10.1007/s12072-013-9481-9.
    1. Mohamed NA, Swify EM, Amin NF, Soliman MM, Tag-Eldin LM, Elsherbiny NM. Is serum level of methylated RASSF1A valuable in diagnosing hepatocellular carcinoma in patients with chronic viral hepatitis C? Arab J Gastroenterol. 2012;13(3):111–115. doi: 10.1016/j.ajg.2012.06.009.
    1. Iizuka N, Oka M, Sakaida I, Moribe T, Miura T, Kimura N, et al. Efficient detection of hepatocellular carcinoma by a hybrid blood test of epigenetic and classical protein markers. Clin Chim Acta. 2011;412(1–2):152–158. doi: 10.1016/j.cca.2010.09.028.
    1. Huang ZH, Hu Y, Hua D, Wu YY, Song MX, Cheng ZH. Quantitative analysis of multiple methylated genes in plasma for the diagnosis and prognosis of hepatocellular carcinoma. Exp Mol Pathol. 2011;91(3):702–707. doi: 10.1016/j.yexmp.2011.08.004.
    1. Yang YJ, Chen H, Huang P, Li CH, Dong ZH, Hou YL. Quantification of plasma hTERT DNA in hepatocellular carcinoma patients by quantitative fluorescent polymerase chain reaction. Clin Invest Med. 2011;34(4):E238. doi: 10.25011/cim.v34i4.15366.
    1. Iyer P, Zekri AR, Hung CW, Schiefelbein E, Ismail K, Hablas A, et al. Concordance of DNA methylation pattern in plasma and tumor DNA of Egyptian hepatocellular carcinoma patients. Exp Mol Pathol. 2010;88(1):107–111. doi: 10.1016/j.yexmp.2009.09.012.
    1. Szymanska K, Chen JG, Cui Y, Gong YY, Turner PC, Villar S, et al. TP53 R249S mutations, exposure to aflatoxin, and occurrence of hepatocellular carcinoma in a cohort of chronic hepatitis B virus carriers from Qidong, China. Cancer Epidemiol Biomark Prev. 2009;18(5):1638–1643. doi: 10.1158/1055-9965.EPI-08-1102.
    1. Chang H, Yi B, Li L, Zhang HY, Sun F, Dong SQ, et al. Methylation of tumor associated genes in tissue and plasma samples from liver disease patients. Exp Mol Pathol. 2008;85(2):96–100. doi: 10.1016/j.yexmp.2008.07.001.
    1. Zhang YJ, Wu HC, Shen J, Ahsan H, Tsai WY, Yang HI, et al. Predicting hepatocellular carcinoma by detection of aberrant promoter methylation in serum DNA. Clin Cancer Res. 2007;13(8):2378–2384. doi: 10.1158/1078-0432.CCR-06-1900.
    1. Tan SH, Ida H, Lau QC, Goh BC, Chieng WS, Loh M, et al. Detection of promoter hypermethylation in serum samples of cancer patients by methylation-specific polymerase chain reaction for tumour suppressor genes including RUNX3. Oncol Rep. 2007;18(5):1225–1230.
    1. Ren N, Qin LX, Tu H, Liu YK, Zhang BH, Tang ZY. The prognostic value of circulating plasma DNA level and its allelic imbalance on chromosome 8p in patients with hepatocellular carcinoma. J Cancer Res Clin Oncol. 2006;132(6):399–407. doi: 10.1007/s00432-005-0049-5.
    1. Iizuka N, Sakaida I, Moribe T, Fujita N, Miura T, Stark M, et al. Elevated levels of circulating cell-free DNA in the blood of patients with hepatitis C virus-associated hepatocellular carcinoma. Anticancer Res. 2006;26(6C):4713–4719.
    1. Yeo W, Wong N, Wong WL, Lai PB, Zhong S, Johnson PJ. High frequency of promoter hypermethylation of RASSF1A in tumor and plasma of patients with hepatocellular carcinoma. Liver Int. 2005;25(2):266–272. doi: 10.1111/j.1478-3231.2005.01084.x.
    1. Chu HJ, Heo J, Seo SB, Kim GH, Kang DH, Song GA, et al. Detection of aberrant p16INK4A methylation in sera of patients with liver cirrhosis and hepatocellular carcinoma. J Korean Med Sci. 2004;19(1):83–86. doi: 10.3346/jkms.2004.19.1.83.
    1. Wong IH, Zhang J, Lai PB, Lau WY, Lo YM. Quantitative analysis of tumor-derived methylated p16INK4a sequences in plasma, serum, and blood cells of hepatocellular carcinoma patients. Clin Cancer Res. 2003;9(3):1047–1052.
    1. Huang XH, Sun LH, Lu DD, Sun Y, Ma LJ, Zhang XR, et al. Codon 249 mutation in exon 7 of p53 gene in plasma DNA: maybe a new early diagnostic marker of hepatocellular carcinoma in Qidong risk area, China. World J Gastroenterol. 2003;9(4):692–695. doi: 10.3748/wjg.v9.i4.692.
    1. Wong IH, Lo YM, Yeo W, Lau WY, Johnson PJ. Frequent p15 promoter methylation in tumor and peripheral blood from hepatocellular carcinoma patients. Clin Cancer Res. 2000;6(9):3516–3521.
    1. Patil MA, Gutgemann I, Zhang J, Ho C, Cheung ST, Ginzinger D, et al. Array-based comparative genomic hybridization reveals recurrent chromosomal aberrations and Jab1 as a potential target for 8q gain in hepatocellular carcinoma. Carcinogenesis. 2005;26(12):2050–2057. doi: 10.1093/carcin/bgi178.
    1. Krebs MG, Metcalf RL, Carter L, Brady G, Blackhall FH, Dive C. Molecular analysis of circulating tumour cells-biology and biomarkers. Nat Rev Clin Oncol. 2014;11(3):129–144. doi: 10.1038/nrclinonc.2013.253.
    1. Thompson EW, Haviv I. The social aspects of EMT-MET plasticity. Nat Med. 2011;17(9):1048–1049. doi: 10.1038/nm.2437.
    1. Yin Chang-Qing, Yuan Chun-Hui, Qu Zhen, Guan Qing, Chen Hao, Wang Fu-Bing. Liquid Biopsy of Hepatocellular Carcinoma: Circulating Tumor-Derived Biomarkers. Disease Markers. 2016;2016:1–11.
    1. de Boer CJ, van Krieken JH, Janssen-van Rhijn CM, Litvinov SV. Expression of ep-CAM in normal, regenerating, metaplastic, and neoplastic liver. J Pathol. 1999;188(2):201–206. doi: 10.1002/(SICI)1096-9896(199906)188:2<201::AID-PATH339>;2-8.
    1. Went PT, Lugli A, Meier S, Bundi M, Mirlacher M, Sauter G, et al. Frequent EpCam protein expression in human carcinomas. Hum Pathol. 2004;35(1):122–128. doi: 10.1016/j.humpath.2003.08.026.
    1. Nagrath S, Sequist LV, Maheswaran S, Bell DW, Irimia D, Ulkus L, et al. Isolation of rare circulating tumour cells in cancer patients by microchip technology. Nature. 2007;450(7173):1235–1239. doi: 10.1038/nature06385.
    1. Yu X, Wang B, Zhang N, Yin C, Chen H, Zhang L, et al. Capture and release of Cancer cells by combining on-Chip purification and off-Chip enzymatic treatment. ACS Appl Mater Interfaces. 2015;7(43):24001–24007. doi: 10.1021/acsami.5b06791.
    1. Taly V, Pekin D, Benhaim L, Kotsopoulos SK, Le Corre D, Li X, et al. Multiplex picodroplet digital PCR to detect KRAS mutations in circulating DNA from the plasma of colorectal cancer patients. Clin Chem. 2013;59(12):1722–1731. doi: 10.1373/clinchem.2013.206359.
    1. Kinde I, Wu J, Papadopoulos N, Kinzler KW, Vogelstein B. Detection and quantification of rare mutations with massively parallel sequencing. Proc Natl Acad Sci U S A. 2011;108(23):9530–9535. doi: 10.1073/pnas.1105422108.
    1. Newman AM, Bratman SV, To J, Wynne JF, Eclov NC, Modlin LA, et al. An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage. Nat Med. 2014;20(5):548–554. doi: 10.1038/nm.3519.
    1. Forshew T, Murtaza M, Parkinson C, Gale D, Tsui DW, Kaper F, et al. Noninvasive identification and monitoring of cancer mutations by targeted deep sequencing of plasma DNA. Sci Transl Med. 2012;4(136):136ra68. doi: 10.1126/scitranslmed.3003726.
    1. Murtaza M, Dawson SJ, Tsui DW, Gale D, Forshew T, Piskorz AM, et al. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature. 2013;497(7447):108–112. doi: 10.1038/nature12065.
    1. Rothe F, Laes JF, Lambrechts D, Smeets D, Vincent D, Maetens M, et al. Plasma circulating tumor DNA as an alternative to metastatic biopsies for mutational analysis in breast cancer. Ann Oncol. 2014;25(10):1959–1965. doi: 10.1093/annonc/mdu288.
    1. Sun C, Liao W, Deng Z, Li E, Feng Q, Lei J, et al. The diagnostic value of assays for circulating tumor cells in hepatocellular carcinoma: a meta-analysis. Medicine. 2017;96(29):e7513. doi: 10.1097/MD.0000000000007513.
    1. Zhang Y, Li J, Cao L, Xu W, Yin Z. Circulating tumor cells in hepatocellular carcinoma: detection techniques, clinical implications, and future perspectives. Semin Oncol. 2012;39(4):449–460. doi: 10.1053/j.seminoncol.2012.05.012.
    1. Zhou Y, Wang B, Wu J, Zhang C, Zhou Y, Yang X, et al. Association of preoperative EpCAM circulating tumor cells and peripheral Treg cell levels with early recurrence of hepatocellular carcinoma following radical hepatic resection. BMC Cancer. 2016;16:506. doi: 10.1186/s12885-016-2526-4.
    1. Wang Zhong, Luo Lei, Cheng Yuan, He Guolin, Peng Bangjian, Gao Yi, Jiang Ze-sheng, Pan MingXin. Correlation Between Postoperative Early Recurrence of Hepatocellular Carcinoma and Mesenchymal Circulating Tumor Cells in Peripheral Blood. Journal of Gastrointestinal Surgery. 2017;22(4):633–639. doi: 10.1007/s11605-017-3619-3.
    1. Bergman Y, Cedar H. DNA methylation dynamics in health and disease. Nat Struct Mol Biol. 2013;20(3):274–281. doi: 10.1038/nsmb.2518.
    1. Xu RH, Wei W, Krawczyk M, Wang W, Luo H, Flagg K, et al. Circulating tumour DNA methylation markers for diagnosis and prognosis of hepatocellular carcinoma. Nat Mater. 2017;16(11):1155–1161. doi: 10.1038/nmat4997.
    1. Huang Z, Hua D, Hu Y, Cheng Z, Zhou X, Xie Q, et al. Quantitation of plasma circulating DNA using quantitative PCR for the detection of hepatocellular carcinoma. Pathol Oncol Res. 2012;18(2):271–276. doi: 10.1007/s12253-011-9438-z.
    1. Chen K, Zhang H, Zhang LN, Ju SQ, Qi J, Huang DF, et al. Value of circulating cell-free DNA in diagnosis of hepatocelluar carcinoma. World J Gastroenterol. 2013;19(20):3143–3149. doi: 10.3748/wjg.v19.i20.3143.
    1. Hardy T, Zeybel M, Day CP, Dipper C, Masson S, McPherson S, et al. Plasma DNA methylation: a potential biomarker for stratification of liver fibrosis in non-alcoholic fatty liver disease. Gut. 2017;66(7):1321–1328. doi: 10.1136/gutjnl-2016-311526.
    1. Ren N, Ye QH, Qin LX, Zhang BH, Liu YK, Tang ZY. Circulating DNA level is negatively associated with the long-term survival of hepatocellular carcinoma patients. World J Gastroenterol. 2006;12(24):3911–3914. doi: 10.3748/wjg.v12.i24.3911.
    1. Tokuhisa Y, Iizuka N, Sakaida I, Moribe T, Fujita N, Miura T, et al. Circulating cell-free DNA as a predictive marker for distant metastasis of hepatitis C virus-related hepatocellular carcinoma. Br J Cancer. 2007;97(10):1399–1403. doi: 10.1038/sj.bjc.6604034.
    1. Ono A, Fujimoto A, Yamamoto Y, Akamatsu S, Hiraga N, Imamura M, et al. Circulating tumor DNA analysis for liver cancers and its usefulness as a liquid biopsy. Cell Mol Gastroenterol Hepatol. 2015;1(5):516–534. doi: 10.1016/j.jcmgh.2015.06.009.
    1. Fan ZC, Yan J, Liu GD, Tan XY, Weng XF, Wu WZ, et al. Real-time monitoring of rare circulating hepatocellular carcinoma cells in an orthotopic model by in vivo flow cytometry assesses resection on metastasis. Cancer Res. 2012;72(10):2683–2691. doi: 10.1158/0008-5472.CAN-11-3733.
    1. Nel I, Baba HA, Ertle J, Weber F, Sitek B, Eisenacher M, et al. Individual profiling of circulating tumor cell composition and therapeutic outcome in patients with hepatocellular carcinoma. Transl Oncol. 2013;6(4):420–428. doi: 10.1593/tlo.13271.
    1. Li J, Shi L, Zhang X, Sun B, Yang Y, Ge N, et al. pERK/pAkt phenotyping in circulating tumor cells as a biomarker for sorafenib efficacy in patients with advanced hepatocellular carcinoma. Oncotarget. 2016;7(3):2646–2659.
    1. Cai ZX, Chen G, Zeng YY, Dong XQ, Lin MJ, Huang XH, et al. Circulating tumor DNA profiling reveals clonal evolution and real-time disease progression in advanced hepatocellular carcinoma. Int J Cancer. 2017;141(5):977–985. doi: 10.1002/ijc.30798.
    1. Jahr S, Hentze H, Englisch S, Hardt D, Fackelmayer FO, Hesch RD, et al. DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res. 2001;61(4):1659–1665.
    1. Thierry AR, El Messaoudi S, Gahan PB, Anker P, Stroun M. Origins, structures, and functions of circulating DNA in oncology. Cancer Metastasis Rev. 2016;35(3):347–376. doi: 10.1007/s10555-016-9629-x.
    1. Cortese R, Almendros I, Wang Y, Gozal D. Tumor circulating DNA profiling in xenografted mice exposed to intermittent hypoxia. Oncotarget. 2015;6(1):556–569. doi: 10.18632/oncotarget.2785.
    1. Gai Wanxia, Sun Kun. Epigenetic Biomarkers in Cell-Free DNA and Applications in Liquid Biopsy. Genes. 2019;10(1):32. doi: 10.3390/genes10010032.
    1. Sun K, Jiang P, Chan KC, Wong J, Cheng YK, Liang RH, et al. Plasma DNA tissue mapping by genome-wide methylation sequencing for noninvasive prenatal, cancer, and transplantation assessments. Proc Natl Acad Sci U S A. 2015;112(40):E5503–E5512. doi: 10.1073/pnas.1508736112.
    1. Lehmann-Werman R, Neiman D, Zemmour H, Moss J, Magenheim J, Vaknin-Dembinsky A, et al. Identification of tissue-specific cell death using methylation patterns of circulating DNA. Proc Natl Acad Sci U S A. 2016;113(13):E1826–E1834. doi: 10.1073/pnas.1519286113.
    1. Moss J, Magenheim J, Neiman D, Zemmour H, Loyfer N, Korach A, et al. Comprehensive human cell-type methylation atlas reveals origins of circulating cell-free DNA in health and disease. Nat Commun. 2018;9(1):5068. doi: 10.1038/s41467-018-07466-6.
    1. Guo S, Diep D, Plongthongkum N, Fung HL, Zhang K, Zhang K. Identification of methylation haplotype blocks aids in deconvolution of heterogeneous tissue samples and tumor tissue-of-origin mapping from plasma DNA. Nat Genet. 2017;49(4):635–642. doi: 10.1038/ng.3805.
    1. Snyder MW, Kircher M, Hill AJ, Daza RM, Shendure J. Cell-free DNA comprises an in vivo nucleosome footprint that informs its tissues-of-origin. Cell. 2016;164(1–2):57–68. doi: 10.1016/j.cell.2015.11.050.
    1. Jiang Peiyong, Sun Kun, Tong Yu K., Cheng Suk Hang, Cheng Timothy H. T., Heung Macy M. S., Wong John, Wong Vincent W. S., Chan Henry L. Y., Chan K. C. Allen, Lo Y. M. Dennis, Chiu Rossa W. K. Preferred end coordinates and somatic variants as signatures of circulating tumor DNA associated with hepatocellular carcinoma. Proceedings of the National Academy of Sciences. 2018;115(46):E10925–E10933. doi: 10.1073/pnas.1814616115.
    1. Scherer F, Kurtz DM, Newman AM, Stehr H, Craig AF, Esfahani MS, et al. Distinct biological subtypes and patterns of genome evolution in lymphoma revealed by circulating tumor DNA. Sci Transl Med. 2016;8(364):364ra155. doi: 10.1126/scitranslmed.aai8545.
    1. Martínez-Ricarte Francisco, Mayor Regina, Martínez-Sáez Elena, Rubio-Pérez Carlota, Pineda Estela, Cordero Esteban, Cicuéndez Marta, Poca Maria A., López-Bigas Nuria, Ramon y Cajal Santiago, Vieito María, Carles Joan, Tabernero Josep, Vivancos Ana, Gallego Soledad, Graus Francesc, Sahuquillo Juan, Seoane Joan. Molecular Diagnosis of Diffuse Gliomas through Sequencing of Cell-Free Circulating Tumor DNA from Cerebrospinal Fluid. Clinical Cancer Research. 2018;24(12):2812–2819. doi: 10.1158/1078-0432.CCR-17-3800.

Source: PubMed

3
Předplatit