Association Between Liquid Biopsy and Prognosis of Gastric Cancer Patients: A Systematic Review and Meta-Analysis

Yunhe Gao, Hongqing Xi, Bo Wei, Jianxin Cui, Kecheng Zhang, Hua Li, Aizhen Cai, Weishen Shen, Jiyang Li, Rafael Rosell, Joseph Chao, Tianhui Chen, Samuel Klempner, Zhi Qiao, Lin Chen, Yunhe Gao, Hongqing Xi, Bo Wei, Jianxin Cui, Kecheng Zhang, Hua Li, Aizhen Cai, Weishen Shen, Jiyang Li, Rafael Rosell, Joseph Chao, Tianhui Chen, Samuel Klempner, Zhi Qiao, Lin Chen

Abstract

Background: Reports regarding liquid biopsy and gastric cancer (GC) have emerged rapidly in recent decades, yet their prognostic value still remains controversial. This study was aimed to assess the impact of liquid biopsy, including circulating tumor cells (CTCs) and cell-free nucleic acids, on GC patients' prognosis. Methods: PubMed, Medline, EMBASE, and ClinicalTrial.gov databases were searched for studies that report GC patient survival data stratified by CTC/circulating tumor DNA (ctDNA)/circulating miRNAs' status. The hazard ratios (HRs) and their 95% confidence intervals (CIs) for patients' overall survival (OS) and disease-free survival (DFS)/progression-free survival (PFS) were recorded or calculated depending on circulating target status. Results: We initially identified 4,221 studies, from which 43 were eligible for further analysis, comprising 3,814 GC patients. Pooled analyses showed that detection of certain CTCs, ctDNA, and circulating miRNA was associated with poorer OS (CTCs: HR = 1.84, 95%CI 1.50-2.26, p < 0.001; ctDNA: HR = 1.78, 95%CI 1.36-2.34, p < 0.001; circulating miRNA: HR = 1.74, 95%CI 1.13-2.69, p < 0.001) and DFS/PFS (CTCs: HR = 3.39, 95%CI 2.21-5.20, p < 0.001; ctDNA: HR = 2.38, 95%CI 1.31-4.32, p = 0.004; circulating miRNA: HR = 3.30, 95%CI 2.39-4.55, p < 0.001) of GC patients, regardless of disease stage and time point at which sample is taken (at baseline or post-treatment). Conclusions: The presence of CTCs and/or cellular components identifies a group of GC with poorer prognosis. Among circulating markers, CTCs demonstrated a stronger and more stable predictive value for late-stage disease and among Mongolian populations with GC. Less data are available for ctDNA and miRNA; however, their presence may also reflect aggressive biology and warrants further prospective study.

Keywords: circulating mRNA; circulating tumor DNA; circulating tumor cells; gastric cancer; liquid biopsy; prognosis.

Copyright © 2019 Gao, Xi, Wei, Cui, Zhang, Li, Cai, Shen, Li, Rosell, Chao, Chen, Klempner, Qiao and Chen.

Figures

Figure 1
Figure 1
Flowchart of enrolled studies investigating the association of liquid biopsy and gastric cancer patients' prognosis.
Figure 2
Figure 2
Forest plots of HRs for OS and DFS/PFS of GC patients, by CTC status. (A) Overall analysis of HR for OS of GC patients. (B) Subgroup analysis of HR for OS of the GC patients by detection targets. (C) Overall analysis of HR for DFS/PFS of GC patients. (D) Subgroup analysis of HR for DFS/PFS of GC patients by detection targets. HRs, hazard ratios; OS, overall survival; DFS, disease-free survival; PFS, progression-free survival; GC, gastric cancer; CTC, circulating tumor cell.
Figure 3
Figure 3
Forest plots of HRs for (A) OS and (B) DFS/PFS of GC patients, based on detection of circulating tumor DNA status. HRs, hazard ratios; OS, overall survival; DFS, disease-free survival; PFS, progression-free survival; GC, gastric cancer.
Figure 4
Figure 4
Forest plots of HRs for (A) OS and (B) DFS/PFS of GC patients, based on detection of circulating miRNA status. HRs, hazard ratios; OS, overall survival; DFS, disease-free survival; PFS, progression-free survival; GC, gastric cancer.

References

    1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. (2018) 68:394–424. 10.3322/caac.21492
    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA Cancer J Clin. (2017) 67:7–30. 10.3322/caac.21387
    1. Thrumurthy SG, Chaudry MA, Hochhauser D, Mughal M. The diagnosis and management of gastric cancer. BMJ. (2013) 347:f6367. 10.1136/bmj.f6367
    1. Zhao Z, Yin Z, Wang S, Wang J, Bai B, Qiu Z, et al. . Meta-analysis: the diagnostic efficacy of chromoendoscopy for early gastric cancer and premalignant gastric lesions. J Gastroenterol Hepatol. (2016) 31:1539–45. 10.1111/jgh.13313
    1. Pasechnikov V, Chukov S, Fedorov E, Kikuste I, Leja M. Gastric cancer: prevention, screening and early diagnosis. World J Gastroenterol. (2014) 20:13842–62. 10.3748/wjg.v20.i38.13842
    1. Ruibal Morell A. CEA serum levels in non-neoplastic disease. Int J Biol Markers. (1992) 7:160–6. 10.1177/172460089200700307
    1. Emoto S, Ishigami H, Yamashita H, Yamaguchi H, Kaisaki S, Kitayama J. Clinical significance of CA125 and CA72-4 in gastric cancer with peritoneal dissemination. Gastric Cancer. (2012) 15:154–61. 10.1007/s10120-011-0091-8
    1. Yang AP, Liu J, Lei HY, Zhang QW, Zhao L, Yang GH. CA72-4 combined with CEA, CA125 and CAl9-9 improves the sensitivity for the early diagnosis of gastric cancer. Clin Chim Acta. (2014) 437:183–6. 10.1016/j.cca.2014.07.034
    1. Shimada H, Noie T, Ohashi M, Oba K, Takahashi Y. Clinical significance of serum tumor markers for gastric cancer: a systematic review of literature by the Task Force of the Japanese Gastric Cancer Association. Gastric Cancer. (2014) 17:26–33. 10.1007/s10120-013-0259-5
    1. Alix-Panabières C, Pantel K. Circulating tumor cells: liquid biopsy of cancer. Clin Chem. (2013) 59:110–8. 10.1373/clinchem.2012.194258
    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) 71:5955–60. 10.1158/0008-5472.CAN-11-1254
    1. Crowley E, Di Nicolantonio F, Loupakis F, Bardelli A. Liquid biopsy: monitoring cancer-genetics in the blood. Nat Rev Clin Oncol. (2013) 10:472–84. 10.1038/nrclinonc.2013.110
    1. Schwarzenbach H, Hoon DS, Pantel K. Cell-free nucleic acids as biomarkers in cancer patients. Nat Rev Cancer. (2011) 11:426–37. 10.1038/nrc3066
    1. Hayes DF, Cristofanilli M, Budd GT, Ellis MJ, Stopeck A, Miller MC, et al. . Circulating tumor cells at each follow-up time point during therapy of metastatic breast cancer patients predict progression-free and overall survival. Clin Cancer Res. (2006) 12:4218–24. 10.1158/1078-0432.CCR-05-2821
    1. de Bono JS, Scher HI, Montgomery RB, Parker C, Miller MC, Tissing H, et al. . Circulating tumor cells predict survival benefit from treatment in metastatic castration-resistant prostate cancer. Clin Cancer Res. (2008) 14:6302–9. 10.1158/1078-0432.CCR-08-0872
    1. Cohen SJ, Punt CJ, Iannotti N, Saidman BH, Sabbath KD, Gabrail NY, et al. . Relationship of circulating tumor cells to tumor response, progression-free survival, and overall survival in patients with metastatic colorectal cancer. J Clin Oncol. (2008) 26:3213–21. 10.1007/978-1-59745-183-3
    1. Kim ST, Banks KC, Pectasides E, Kim SY, Kim K, Lanman RB, et al. Impact of genomic alterations on lapatinib treatment outcome and cell-free genomic landscape during HER2 therapy in HER2-positive gastric cancer patients. Ann Oncol. (2018) 29:1037–48. 10.1093/annonc/mdy034
    1. Schrock AB, Pavlick D, Klempner SJ, Chung JH, Forcier B, Welsh A, et al. Hybrid capture-based genomic profiling of circulating tumor DNA from patients with advanced cancers of the gastrointestinal tract or anus. Clin Cancer Res. (2018) 24:1881–90. 10.1158/1078-0432.CCR-17-3103
    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 Biomarkers Prev. (2015) 24:206–12. 10.1158/1055-9965.EPI-14-0895
    1. Zhang L, Riethdorf S, Wu G, Wang T, Yang K, Peng G, et al. Meta-analysis of the prognostic value of circulating tumor cells in breast cancer. Clin Cancer Res. (2012) 18:5701–10. 10.1158/1078-0432.CCR-12-1587
    1. Rahbari NN, Aigner M, Thorlund K, Mollberg N, Motschall E, Jensen K, et al. . Meta-analysis shows that detection of circulating tumor cells indicates poor prognosis in patients with colorectal cancer. Gastroenterology. (2010) 138:1714–26. 10.1053/j.gastro.2010.01.008
    1. Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y, Agrawal N, et al. . Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med. (2014) 6:224ra24. 10.1126/scitranslmed.3007094
    1. Huang YK, Yu JC. Circulating microRNAs and long non-coding RNAs in gastric cancer diagnosis: an update and review. World J Gastroenterol. (2015) 21:9863–86. 10.3748/wjg.v21.i34.9863
    1. Alix-Panabières C, Pantel K. Challenges in circulating tumour cell research. Nat Rev Cancer. (2014) 14:623–31. 10.1038/nrc3820
    1. Stroup DF, Berlin JA, Morton SC, Olkin I, Williamson GD, Rennie D, et al. . Meta-analysis of observational studies in epidemiology: a proposal for reporting. Meta-analysis Of Observational Studies in Epidemiology (MOOSE) group. JAMA. (2000) 283:2008–12. 10.1001/jama.283.15.2008
    1. Moher D, Liberati A, Tetzlaff J, Altman DG. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. BMJ. (2009) 339:b2535. 10.1136/bmj.b2535
    1. Higgins JP GS. Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 [updated March 2011]. Cochrane Collaboration (2011).
    1. Tierney JF, Stewart LA, Ghersi D, Burdett S, Sydes MR. Practical methods for incorporating summary time-to-event data into meta-analysis. Trials. (2007) 8:16. 10.1186/1745-6215-8-16
    1. DerSimonian R, Laird N. Meta-analysis in clinical trials. Control Clin Trials. (1986) 7:177–88. 10.1016/0197-2456(86)90046-2
    1. Hiraiwa K, Takeuchi H, Hasegawa H, Saikawa Y, Suda K, Ando T, et al. . Clinical significance of circulating tumor cells in blood from patients with gastrointestinal cancers. Ann Surg Oncol. (2008) 15:3092–100. 10.1245/s10434-008-0122-9
    1. Matsusaka S, Chìn K, Ogura M, Suenaga M, Shinozaki E, Mishima Y, et al. . Circulating tumor cells as a surrogate marker for determining response to chemotherapy in patients with advanced gastric cancer. Cancer Sci. (2010) 101:1067–71. 10.1111/j.1349-7006.2010.01492.x
    1. Uenosono Y, Arigami T, Kozono T, Yanagita S, Hagihara T, Haraguchi N, et al. . Clinical significance of circulating tumor cells in peripheral blood from patients with gastric cancer. Cancer. (2013) 119:3984–91. 10.1002/cncr.28309
    1. Li Y, Gong J, Zhang Q, Lu Z, Gao J, Li Y, et al. . Dynamic monitoring of circulating tumour cells to evaluate therapeutic efficacy in advanced gastric cancer. Br J Cancer. (2016) 114:138–45. 10.1038/bjc.2015.417
    1. Okabe H, Tsunoda S, Hosogi H, Hisamori S, Tanaka E, Tanaka S, et al. . Circulating tumor cells as an independent predictor of survival in advanced gastric cancer. Ann Surg Oncol. (2015) 22:3954–61. 10.1245/s10434-015-4483-6
    1. Yie SM, Lou B, Ye SR, Cao M, He X, Li P, et al. . Detection of survivin-expressing circulating cancer cells (CCCs) in peripheral blood of patients with gastric and colorectal cancer reveals high risks of relapse. Ann Surg Oncol. (2008) 15:3073–82. 10.1245/s10434-008-0069-x
    1. Bertazza L, Mocellin S, Marchet A, Pilati P, Gabrieli J, Scalerta R, et al. . Survivin gene levels in the peripheral blood of patients with gastric cancer independently predict survival. J Transl Med. (2009) 7:111. 10.1186/1479-5876-7-111
    1. Cao W, Yang W, Li H, Lou G, Jiang J, Geng M, et al. . Using detection of survivin-expressing circulating tumor cells in peripheral blood to predict tumor recurrence following curative resection of gastric cancer. J Surg Oncol. (2011) 103:110–5. 10.1002/jso.21777
    1. Majima T, Ichikura T, Takayama E, Chochi K, Mochizuki H. Detecting circulating cancer cells using reverse transcriptase-polymerase chain reaction for cytokeratin mRNA in peripheral blood from patients with gastric cancer. Jpn J Clin Oncol. (2000) 30:499–503. 10.1093/jjco/hyd130
    1. Pituch-Noworolska A, Kolodziejczyk P, Kulig J, Drabik G, Szczepanik A, Czupryna A, et al. . Circulating tumour cells and survival of patients with gastric cancer. Anticancer Res. (2007) 27:635–40.
    1. Illert B, Fein M, Otto C, Cording F, Stehle D, Thiede A, et al. . Disseminated tumor cells in the blood of patients with gastric cancer are an independent predictive marker of poor prognosis. Scand J Gastroenterol. (2005) 40:843–9. 10.1080/00365520510015557
    1. Saad AA, Awed NM, Abd Elkerim NN, El-Shennawy D, Alfons MA, Elserafy ME, et al. . Prognostic significance of E-cadherin expression and peripheral blood micrometastasis in gastric carcinoma patients. Ann Surg Oncol. (2010) 17:3059–67. 10.1245/s10434-010-1151-8
    1. Liu Y, Ling Y, Qi Q, Lan F, Zhu M, Zhang Y, et al. . Prognostic value of circulating tumor cells in advanced gastric cancer patients receiving chemotherapy. Mol Clin Oncol. (2017) 6:235–42. 10.3892/mco.2017.1125
    1. Uen YH, Lin SR, Wu CH, Hsieh JS, Lu CY, Yu FJ, et al. . Clinical significance of MUC1 and c-Met RT-PCR detection of circulating tumor cells in patients with gastric carcinoma. Clin Chim Acta. (2006) 367:55–61. 10.1016/j.cca.2005.11.013
    1. Wu CH, Lin SR, Yu FJ, Wu DC, Pan YS, Hsieh JS, et al. . Development of a high-throughput membrane-array method for molecular diagnosis of circulating tumor cells in patients with gastric cancers. Int J Cancer. (2006) 119:373–9. 10.1002/ijc.21856
    1. Ikeguchi M, Kaibara N. Detection of circulating cancer cells after a gastrectomy for gastric cancer. Surg Today. (2005) 35:436–41. 10.1007/s00595-004-2978-z
    1. Ishigami S, Sakamoto A, Uenosono Y, Nakajo A, Okumura H, Matsumoto M, et al. . Carcinoembryonic antigen messenger RNA expression in blood can predict relapse in gastric cancer. J Surg Res. (2008) 148:205–9. 10.1016/j.jss.2007.08.013
    1. Qiu MZ, Li ZH, Zhou ZW, Li YH, Wang ZQ, Wang FH, et al. . Detection of carcinoembryonic antigen messenger RNA in blood using quantitative real-time reverse transcriptase-polymerase chain reaction to predict recurrence of gastric adenocarcinoma. J Transl Med. (2010) 8:107. 10.1186/1479-5876-8-107
    1. Arigami T, Uenosono Y, Hirata M, Yanagita S, Ishigami S, Natsugoe S. B7-H3 expression in gastric cancer: a novel molecular blood marker for detecting circulating tumor cells. Cancer Sci. (2011) 102:1019–24. 10.1111/j.1349-7006.2011.01877.x
    1. Ito H, Sato J, Tsujino Y, Yamaguchi N, Kimura S, Gohda K, et al. . Long-term prognostic impact of circulating tumour cells in gastric cancer patients. World J Gastroenterol. (2016) 22:10232–41. 10.3748/wjg.v22.i46.10232
    1. Leung WK, To KF, Chu ES, Chan MW, Bai AH, Ng EK, et al. . Potential diagnostic and prognostic values of detecting promoter hypermethylation in the serum of patients with gastric cancer. Br J Cancer. (2005) 92:2190–4. 10.1038/sj.bjc.6602636
    1. Balgkouranidou I, Karayiannakis A, Matthaios D, Bolanaki H, Tripsianis G, Tentes AA, et al. . Assessment of SOX17 DNA methylation in cell free DNA from patients with operable gastric cancer. Association with prognostic variables and survival. Clin Chem Lab Med. (2013) 51:1505–10. 10.1515/cclm-2012-0320
    1. Yang Q, Gao J, Xu L, Zeng Z, Sung JJ, Yu J. Promoter hypermethylation of BCL6B gene is a potential plasma DNA biomarker for gastric cancer. Biomarkers. (2013) 18:721–5. 10.3109/1354750X.2013.853839
    1. Ling ZQ, Lv P, Lu XX, Yu JL, Han J, Ying LS, et al. . Circulating methylated XAF1 DNA indicates poor prognosis for gastric cancer. PLoS ONE. (2013) 8:e67195. 10.1371/journal.pone.0067195
    1. Han J, Lv P, Yu JL, Wu YC, Zhu X, Hong LL, et al. . Circulating methylated MINT2 promoter DNA is a potential poor prognostic factor in gastric cancer. Dig Dis Sci. (2014) 59:1160–8. 10.1007/s10620-013-3007-0
    1. Wu YC, Lv P, Han J, Yu JL, Zhu X, Hong LL, et al. . Enhanced serum methylated p16 DNAs is associated with the progression of gastric cancer. Int J Clin Exp Pathol. (2014) 7:1553–62.
    1. Yu JL, Lv P, Han J, Zhu X, Hong LL, Zhu WY, et al. . Methylated TIMP-3 DNA in body fluids is an independent prognostic factor for gastric cancer. Arch Pathol Lab Med. (2014) 138:1466–73. 10.5858/arpa.2013-0285-OA
    1. Balgkouranidou I, Matthaios D, Karayiannakis A, Bolanaki H, Michailidis P, Xenidis N, et al. . Prognostic role of APC and RASSF1A promoter methylation status in cell free circulating DNA of operable gastric cancer patients. Mutat Res. (2015) 778:46–51. 10.1016/j.mrfmmm.2015.05.002
    1. Pimson C, Ekalaksananan T, Pientong C, Promthet S, Putthanachote N, Suwanrungruang K, et al. . Aberrant methylation of PCDH10 and RASSF1A genes in blood samples for non-invasive diagnosis and prognostic assessment of gastric cancer. PeerJ. (2016) 4:e2112. 10.7717/peerj.2112
    1. Fang WL, Lan YT, Huang KH, Liu CA, Hung YP, Lin CH, et al. . Clinical significance of circulating plasma DNA in gastric cancer. Int J Cancer. (2016) 138:2974–83. 10.1002/ijc.30018
    1. Valladares-Ayerbes M, Reboredo M, Medina-Villaamil V, Iglesias-Díaz P, Lorenzo-Patiño MJ, Haz M, et al. . Circulating miR-200c as a diagnostic and prognostic biomarker for gastric cancer. J Transl Med. (2012) 10:186. 10.1186/1479-5876-10-186
    1. Zhang HP, Sun FB, Li SJ. Serum miR-200c expression level as a prognostic biomarker for gastric cancer. Genet Mol Res. (2015) 14:15913–20. 10.4238/2015.December.7.2
    1. Yang R, Fu Y, Zeng Y, Xiang M, Yin Y, Li L, et al. . Serum miR-20a is a promising biomarker for gastric cancer. Biomed Rep. (2017) 6:429–34. 10.3892/br.2017.862
    1. Wang M, Gu H, Wang S, Qian H, Zhu W, Zhang L, et al. . Circulating miR-17-5p and miR-20a: molecular markers for gastric cancer. Mol Med Rep. (2012) 5:1514–20. 10.3892/mmr.2012.828
    1. Komatsu S, Ichikawa D, Tsujiura M, Konishi H, Takeshita H, Nagata H, et al. . Prognostic impact of circulating miR-21 in the plasma of patients with gastric carcinoma. Anticancer Res. (2013) 33:271−6.
    1. Song J, Bai Z, Zhang J, Meng H, Cai J, Deng W, et al. . Serum microRNA-21 levels are related to tumor size in gastric cancer patients but cannot predict prognosis. Oncol Lett. (2013) 6:1733–7. 10.3892/ol.2013.1626
    1. Hou CG, Luo XY, Li G. Diagnostic and prognostic value of serum MicroRNA-206 in patients with gastric cancer. Cell Physiol Biochem. (2016) 39:1512–20. 10.1159/000447854
    1. Imaoka H, Toiyama Y, Okigami M, Yasuda H, Saigusa S, Ohi M, et al. . Circulating microRNA-203 predicts metastases, early recurrence, and poor prognosis in human gastric cancer. Gastric Cancer. (2016) 19:744–53. 10.1007/s10120-015-0521-0
    1. Fu Z, Qian F, Yang X, Jiang H, Chen Y, Liu S. Circulating miR-222 in plasma and its potential diagnostic and prognostic value in gastric cancer. Med Oncol. (2014) 31:164. 10.1007/s12032-014-0164-8
    1. Huang D, Wang H, Liu R, Li H, Ge S, Bai M, et al. miRNA27a is a biomarker for predicting chemosensitivity and prognosis in metastatic or recurrent gastric cancer. J Cell Biochem. (2014) 115:549–56. 10.1002/jcb.24689
    1. Zhuang K, Han K, Tang H, Yin X, Zhang J, Zhang X, et al. . Up-Regulation of plasma miR-23b is associated with poor prognosis of gastric cancer. Med Sci Monit. (2016) 22:356–61. 10.12659/MSM.895428
    1. Tsai MM, Wang CS, Tsai CY, Huang CG, Lee KF, Huang HW, et al. . Circulating microRNA-196a/b are novel biomarkers associated with metastatic gastric cancer. Eur J Cancer. (2016) 64:137–48. 10.1016/j.ejca.2016.05.007
    1. Chen Q, Ge X, Zhang Y, Xia H, Yuan D, Tang Q, et al. . Plasma miR-122 and miR-192 as potential novel biomarkers for the early detection of distant metastasis of gastric cancer. Oncol Rep. (2014) 31:1863–70. 10.3892/or.2014.3004
    1. Huang X, Gao P, Sun J, Chen X, Song Y, Zhao J, et al. . Clinicopathological and prognostic significance of circulating tumor cells in patients with gastric cancer: a meta-analysis. Int J Cancer. (2015) 136:21–33. 10.1002/ijc.28954
    1. Zhang ZY, Dai ZL, Yin XW, Li SH, Li SP, Ge HY. Meta-analysis shows that circulating tumor cells including circulating microRNAs are useful to predict the survival of patients with gastric cancer. BMC Cancer. (2014) 14:773. 10.1186/1471-2407-14-773
    1. Gao Y, Zhang K, Xi H, Cai A, Wu X, Cui J, et al. . Diagnostic and prognostic value of circulating tumor DNA in gastric cancer: a meta-analysis. Oncotarget. (2017) 8:6330–40. 10.18632/oncotarget.14064
    1. Oyama K, Fushida S, Kinoshita J, Okamoto K, Makino I, Nakamura K, et al. . Serum cytokeratin 18 as a biomarker for gastric cancer. Clin Exp Med. (2013) 13:289–95. 10.1007/s10238-012-0202-9
    1. Zhao S, Yang H, Zhang M, Zhang D, Liu Y, Liu Y, et al. . Circulating tumor cells (CTCs) detected by triple-marker EpCAM, CK19, and hMAM RT-PCR and their relation to clinical outcome in metastatic breast cancer patients. Cell Biochem Biophys. (2013) 65:263–73. 10.1007/s12013-012-9426-2
    1. Govaere O, Komuta M, Berkers J, Spee B, Janssen C, de Luca F, et al. . Keratin 19: a key role player in the invasion of human hepatocellular carcinomas. Gut. (2014) 63:674–85. 10.1136/gutjnl-2012-304351
    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) 313:139–42. 10.1016/S0009-8981(01)00665-9
    1. Tsujiura M, Ichikawa D, Konishi H, Komatsu S, Shiozaki A, Otsuji E. Liquid biopsy of gastric cancer patients: circulating tumor cells and cell-free nucleic acids. World J Gastroenterol. (2014) 20:3265–86. 10.3748/wjg.v20.i12.3265
    1. Ye T, Chen Y, Fang J. DNA methylation biomarkers in serum for gastric cancer screening. Mini Rev Med Chem. (2010) 10:1034–8. 10.2174/1389557511009011034
    1. Dias-Santagata D, Akhavanfard S, David SS, Vernovsky K, Kuhlmann G, Boisvert SL, et al. . Rapid targeted mutational analysis of human tumours: a clinical platform to guide personalized cancer medicine. EMBO Mol Med. (2010) 2:146–58. 10.1002/emmm.201000070
    1. Merker JD, Oxnard GR, Compton C, Diehn M, Hurley P, Lazar AJ, et al. . Circulating tumor DNA analysis in patients with cancer: American Society of Clinical Oncology and College of American Pathologists Joint Review. J Clin Oncol. (2018) 36:1631–41. 10.1200/JCO.2017.76.8671
    1. Zhang Y, Yang P, Sun T, Li D, Xu X, Rui Y, et al. . miR-126 and miR-126* repress recruitment of mesenchymal stem cells and inflammatory monocytes to inhibit breast cancer metastasis. Nat Cell Biol. (2013) 15:284–94. 10.1038/ncb2690
    1. Zhang Z, Li Z, Li Y, Zang A. MicroRNA and signaling pathways in gastric cancer. Cancer Gene Ther. (2014) 21:305–16. 10.1038/cgt.2014.37
    1. Wicha MS, Hayes DF. Circulating tumor cells: not all detected cells are bad and not all bad cells are detected. J Clin Oncol. (2011) 29:1508–11. 10.1200/JCO.2010.34.0026
    1. Liang Z, Cheng Y, Chen Y, Hu Y, Liu WP, Lu Y, et al. . EGFR T790M ctDNA testing platforms and their role as companion diagnostics: correlation with clinical outcomes to EGFR-TKIs. Cancer Lett. (2017) 403:186–94. 10.1016/j.canlet.2017.06.008
    1. Diaz LA, Bardelli A. Liquid biopsies: genotyping circulating tumor DNA. J Clin Oncol. (2014) 32:579–86. 10.1200/JCO.2012.45.2011
    1. Visvanathan K, Fackler MS, Zhang Z, Lopez-Bujanda ZA, Jeter SC, Sokoll LJ, et al. . Monitoring of serum DNA methylation as an early independent marker of response and survival in metastatic breast cancer: TBCRC 005 Prospective Biomarker Study. J Clin Oncol. (2017) 35:751–8. 10.1200/JCO.2015.66.2080
    1. Cohen JD, Li L, Wang Y, Thoburn C, Afsari B, Danilova L, et al. . Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science. (2018) 359:926–30. 10.1126/science.aar3247
    1. Qin Z, Ljubimov VA, Zhou C, Tong Y, Liang J. Cell-free circulating tumor DNA in cancer. Chin J Cancer. (2016) 35:36. 10.1186/s40880-016-0092-4

Source: PubMed

3
Abonner