The Significance of Circulating Tumor Cells in Patients with Hepatocellular Carcinoma: Real-Time Monitoring and Moving Targets for Cancer Therapy
Abstract
:1. Introduction
2. Overview of CTC Enrichment Strategies
3. The Dynamic Changes of CTCs in Blood Circulation
4. Potential Biomarkers of CTCs in HCC
4.1. Is CTCs Heterogeneity Compatible with EMT Markers?
4.2. Hepatocyte-Specific Markers of CTCs in HCC
4.3. How Do CTCs Respond in the Microenvironment?
4.4. Are CTCs Equivalent to Cancer Stem Cells (CSCs)?
5. Conclusions and Future Perspective
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
Abbreviations
AFP | alpha-fetoprotein |
ANXA3 | Annexin A3 |
ApoA-1 | apolipoprotein A1 |
ASGPR | asialoglycoprotein receptor |
CK | cytokeratin |
CPS1 | carbamoyl phosphate synthetase 1 |
CSCs | cancer stem cells |
CTCs | circulating tumor cells |
ctDNA | circulating tumor DNA |
EMA | epithelial membrane antigen |
EMT | epithelial to mesenchymal transition |
EpCAM | epithelial cell adhesion molecule |
FDA | the US Food and Drug Administration |
GPC | glypican-3; GS: glutamine synthase |
HBV | hepatitis b virus |
HCC | hepatocellular carcinoma |
Hep Par 1 | hepatocyte paraffin-1 |
IGFBP1 | insulin like growth factor binding protein 1 |
OS | overall survival |
pERK | phosphorylated ERK |
PFS | progression-free survival |
ROC | receiver operating characteristic |
RT-PCR | reverse transcriptase polymerase chain reaction |
TACE | transcatheter arterial chemoembolization |
TNM | tumor-node-metastasis |
Treg | regulatory T cells |
References
- Forner, A.; Reig, M.; Bruix, J. Hepatocellular carcinoma. Lancet 2018, 391, 1301–1314. [Google Scholar] [CrossRef]
- Forner, A.; Llovet, J.M.; Bruix, J. Hepatocellular carcinoma. Lancet 2012, 379, 1245–1255. [Google Scholar] [CrossRef]
- Mittal, S.; El-Serag, H.B. Epidemiology of hepatocellular carcinoma: Consider the population. J. Clin. Gastroenterol. 2013, 47, S2–S6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Daher, S.; Massarwa, M.; Benson, A.A.; Khoury, T. Current and Future Treatment of Hepatocellular Carcinoma: An Updated Comprehensive Review. J. Clin. Transl. Hepatol. 2018, 6, 69–78. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bruix, J.; Reig, M.; Sherman, M. Evidence-Based Diagnosis, Staging, and Treatment of Patients with Hepatocellular Carcinoma. Gastroenterology 2016, 150, 835–853. [Google Scholar] [CrossRef] [Green Version]
- Giannini, E.G.; Cucchetti, A.; Erroi, V.; Garuti, F.; Odaldi, F.; Trevisani, F. Surveillance for early diagnosis of hepatocellular carcinoma: How best to do it? World J. Gastroenterol. 2013, 19, 8808–8821. [Google Scholar] [CrossRef]
- Salk, J.J.; Schmitt, M.W.; Loeb, L.A. Enhancing the accuracy of next-generation sequencing for detecting rare and subclonal mutations. Nat. Rev. Genet. 2018, 19, 269–285. [Google Scholar] [CrossRef]
- Zhang, L.; Liang, Y.; Li, S.; Zeng, F.; Meng, Y.; Chen, Z.; Liu, S.; Tao, Y.; Yu, F. The interplay of circulating tumor DNA and chromatin modification, therapeutic resistance, and metastasis. Mol. Cancer 2019, 18, 36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Speicher, M.R.; Pantel, K. Tumor signatures in the blood. Nat. Biotechnol. 2014, 32, 441–443. [Google Scholar] [CrossRef]
- Aceto, N.; Bardia, A.; Miyamoto, D.T.; Donaldson, M.C.; Wittner, B.S.; Spencer, J.A.; Yu, M.; Pely, A.; Engstrom, A.; Zhu, H.; et al. Circulating tumor cell clusters are oligoclonal precursors of breast cancer metastasis. Cell 2014, 158, 1110–1122. [Google Scholar] [CrossRef] [Green Version]
- Kmietowicz, Z. Liquid biopsies will be routine NHS test for cancer “in under five years”. BMJ 2016, 354, i4334. [Google Scholar] [CrossRef] [PubMed]
- Bardelli, A.; Pantel, K. Liquid Biopsies, What We Do Not Know (Yet). Cancer Cell 2017, 31, 172–179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wu, L.-J.; Pan, Y.-D.; Pei, X.-Y.; Chen, H.; Nguyen, S.; Kashyap, A.; Liu, J.; Wu, J. Capturing circulating tumor cells of hepatocellular carcinoma. Cancer Lett. 2012, 326, 17–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, Y.; Li, S.; Li, W.; Yang, R.; Zhang, X.; Ye, Y.; Yu, J.; Ye, L.; Tang, W. Circulating tumor cells undergoing EMT are poorly correlated with clinical stages or predictive of recurrence in hepatocellular carcinoma. Sci. Rep. 2019, 9, 7084. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, C.; Shi, D.; Wang, S.; Wei, C.; Zhang, C.; Xiong, B. Prognostic value of pre- and post-operative circulating tumor cells detection in colorectal cancer patients treated with curative resection: A prospective cohort study based on ISET device. Cancer Manag. Res. 2018, 10, 4135–4144. [Google Scholar] [CrossRef] [Green Version]
- Guo, W.; Yang, X.R.; Sun, Y.F.; Shen, M.N.; Ma, X.L.; Wu, J.; Zhang, C.Y.; Zhou, Y.; Xu, Y.; Hu, B.; 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, 4794–4805. [Google Scholar] [CrossRef] [Green Version]
- Schulze, K.; Gasch, C.; Staufer, K.; Nashan, B.; Lohse, A.W.; Pantel, K.; Riethdorf, S.; Wege, H. 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, 2165–2171. [Google Scholar] [CrossRef]
- Yin, L.C.; Luo, Z.C.; Gao, Y.X.; Li, Y.; Peng, Q.; Gao, Y. Twist Expression in Circulating Hepatocellular Carcinoma Cells Predicts Metastasis and Prognoses. Biomed. Res. Int. 2018, 2018, 3789613. [Google Scholar] [CrossRef]
- Ou, H.; Huang, Y.; Xiang, L.; Chen, Z.; Fang, Y.; Lin, Y.; Cui, Z.; Yu, S.; Li, X.; Yang, D. Circulating Tumor Cell Phenotype Indicates Poor Survival and Recurrence After Surgery for Hepatocellular Carcinoma. Dig. Dis. Sci. 2018, 63, 2373–2380. [Google Scholar] [CrossRef] [PubMed]
- Cheng, Y.; Luo, L.; Zhang, J.; Zhou, M.; Tang, Y.; He, G.; Lu, Y.; Wang, Z.; Pan, M.X. Diagnostic Value of Different Phenotype Circulating Tumor Cells in Hepatocellular Carcinoma. J. Gastrointest. Surg. 2019, 23, 2354–2361. [Google Scholar] [CrossRef] [PubMed]
- Chen, J.; Cao, S.W.; Cai, Z.; Zheng, L.; Wang, Q. Epithelial-mesenchymal transition phenotypes of circulating tumor cells correlate with the clinical stages and cancer metastasis in hepatocellular carcinoma patients. Cancer Biomark. 2017, 20, 487–498. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.M.; Xu, S.C.; Li, J.; Han, K.Q.; Pi, H.F.; Zheng, L.; Zuo, G.H.; Huang, X.B.; Li, H.Y.; Zhao, H.Z.; 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. [Google Scholar] [CrossRef] [PubMed]
- Wang, Z.; Luo, L.; Cheng, Y.; He, G.; Peng, B.; Gao, Y.; Jiang, Z.; Pan, M.X. Correlation between Postoperative Early Recurrence of Hepatocellular Carcinoma and Mesenchymal Circulating Tumor Cells in Peripheral Blood. J. Gastrointest. Surg. 2018, 22, 633–639. [Google Scholar] [CrossRef] [Green Version]
- Liu, Y.K.; Hu, B.S.; Li, Z.L.; He, X.; Li, Y.; Lu, L.G. An improved strategy to detect the epithelial-mesenchymal transition process in circulating tumor cells in hepatocellular carcinoma patients. Hepatol. Int. 2016, 10, 640–646. [Google Scholar] [CrossRef]
- Wu, S.; Liu, S.; Liu, Z.; Huang, J.; Pu, X.; Li, J.; Yang, D.; Deng, H.; Yang, N.; Xu, J. Classification of circulating tumor cells by epithelial-mesenchymal transition markers. PLoS ONE 2015, 10, e0123976. [Google Scholar] [CrossRef]
- Qi, L.N.; Xiang, B.D.; Wu, F.X.; Ye, J.Z.; Zhong, J.H.; Wang, Y.Y.; Chen, Y.Y.; Chen, Z.S.; Ma, L.; Chen, J.; et al. Circulating Tumor Cells Undergoing EMT Provide a Metric for Diagnosis and Prognosis of Patients with Hepatocellular Carcinoma. Cancer Res. 2018, 78, 4731–4744. [Google Scholar] [CrossRef] [Green Version]
- Liu, S.; Wang, M.; Zheng, C.; Zhong, Q.; Shi, Y.; Han, X. Diagnostic value of serum glypican-3 alone and in combination with AFP as an aid in the diagnosis of liver cancer. Clin. Biochem. 2020. [Google Scholar] [CrossRef]
- Hamaoka, M.; Kobayashi, T.; Tanaka, Y.; Mashima, H.; Ohdan, H. Clinical significance of glypican-3-positive circulating tumor cells of hepatocellular carcinoma patients: A prospective study. PLoS ONE 2019, 14, e0217586. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, Y.; Zhang, X.; Zhang, J.; Sun, B.; Zheng, L.; Li, J.; Liu, S.; Sui, G.; Yin, Z. Microfluidic chip for isolation of viable circulating tumor cells of hepatocellular carcinoma for their culture and drug sensitivity assay. Cancer Biol. Ther. 2016, 17, 1177–1187. [Google Scholar] [CrossRef] [Green Version]
- Li, J.; Chen, L.; Zhang, X.; Zhang, Y.; Liu, H.; Sun, B.; Zhao, L.; Ge, N.; Qian, H.; Yang, Y.; et al. Detection of circulating tumor cells in hepatocellular carcinoma using antibodies against asialoglycoprotein receptor, carbamoyl phosphate synthetase 1 and pan-cytokeratin. PLoS ONE 2014, 9, e96185. [Google Scholar] [CrossRef] [Green Version]
- Pang, Y.; Wang, C.; Xiao, R.; Sun, Z. Dual-Selective and Dual-Enhanced SERS Nanoprobes Strategy for Circulating Hepatocellular Carcinoma Cells Detection. Chemistry 2018, 24, 7060–7067. [Google Scholar] [CrossRef]
- Mu, H.; Lin, K.X.; Zhao, H.; Xing, S.; Li, C.; Liu, F.; Lu, H.Z.; Zhang, Z.; Sun, Y.L.; Yan, X.Y.; et al. Identification of biomarkers for hepatocellular carcinoma by semiquantitative immunocytochemistry. World J. Gastroenterol. 2014, 20, 5826–5838. [Google Scholar] [CrossRef]
- Wan, S.; Kim, T.H.; Smith, K.J.; Delanney, R.; Park, G.S.; Guo, H.; Lin, E.; Plegue, T.; Kuo, N.; Steffes, J.; et al. New Labyrinth Microfluidic Device Detects Circulating Tumor Cells Expressing Cancer Stem Cell Marker and Circulating Tumor Microemboli in Hepatocellular Carcinoma. Sci. Rep. 2019, 9, 18575. [Google Scholar] [CrossRef]
- Xu, W.; Cao, L.; Chen, L.; Li, J.; Zhang, X.F.; Qian, H.H.; Kang, X.Y.; Zhang, Y.; Liao, J.; Shi, L.H.; et al. Isolation of circulating tumor cells in patients with hepatocellular carcinoma using a novel cell separation strategy. Clin. Cancer Res. 2011, 17, 3783–3793. [Google Scholar] [CrossRef] [Green Version]
- Liu, H.Y.; Qian, H.H.; Zhang, X.Z.; Li, J.; Yang. X.; Sun, B.; Ma, J.Y.; Chen. L.; Yin, Z.F. Improved method increases sensitivity for circulating hepatocellular carcinoma cells. World J. Gastroenterol. 2015, 21, 2918–2925. [Google Scholar] [CrossRef]
- Nam, S.J.; Yeo, H.Y.; Chang, H.J.; Kim, B.H.; Hong, E.K.; Park, J. A New Cell Block Method for Multiple Immunohistochemical Analysis of Circulating Tumor Cells in Patients with Liver Cancer. Cancer Res. Treat. 2016, 48, 1229–1242. [Google Scholar] [CrossRef] [Green Version]
- Li, J.; Shi, L.; Zhang, X.; Sun, B.; Yang, Y.; Ge, N.; Liu, H.; Yang, X.; Chen, L.; Qian, H.; et al. pERK/pAkt phenotyping in circulating tumor cells as a biomarker for sorafenib efficacy in patients with advanced hepatocellular carcinoma. Oncotarget 2016, 7, 2646–2659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, Y.; Wang, B.; Wu, J.; Zhang, C.; Zhou, Y.; Yang, X.; Zhou, J.; Guo, W.; Fan, J. 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. [Google Scholar] [CrossRef] [Green Version]
- Nel, I.; Baba, H.; Weber, F.; Sitek, B.; Eisenacher, M.; Meyer, H.E.; Schlaak, J.F.; Hoffmann, A.C. IGFBP1 in epithelial circulating tumor cells as a potential response marker to selective internal radiation therapy in hepatocellular carcinoma. Biomark. Med. 2014, 8, 687–698. [Google Scholar] [CrossRef]
- Sun, Y.F.; Xu, Y.; Yang, X.R.; Guo, W.; Zhang, X.; Qiu, S.J.; Shi, R.Y.; Hu, B.; Zhou, J.; Fan, J. Circulating stem cell-like epithelial cell adhesion molecule-positive tumor cells indicate poor prognosis of hepatocellular carcinoma after curative resection. Hepatology 2013, 57, 1458–1468. [Google Scholar] [CrossRef]
- Guo, W.; Sun, Y.F.; Shen, M.N.; Ma, X.L.; Zhang, C.Y.; Zhou, Y.; Xu, Y.; Hu, B.; Zhang, M.; Wang, G.; et al. Circulating Tumor Cells with Stem-Like Phenotypes for Diagnosis, Prognosis, and Therapeutic Response Evaluation in Hepatocellular Carcinoma. Clin. Cancer Res. 2018, 24, 2203–2213. [Google Scholar] [CrossRef] [Green Version]
- Ma, X.L.; Jiang, M.; Zhao, Y.; Wang, B.L.; Shen, M.N.; Zhou, Y.; Zhang, C.Y.; Sun, Y.F.; Chen, J.W.; Hu, B.; et al. Application of Serum Annexin A3 in Diagnosis, Outcome Prediction and Therapeutic Response Evaluation for Patients with Hepatocellular Carcinoma. Ann. Surg. Oncol. 2018, 25, 1686–1694. [Google Scholar] [CrossRef] [PubMed]
- Pantel, K.; Speicher, M.R. The biology of circulating tumor cells. Oncogene 2016, 35, 1216–1224. [Google Scholar] [CrossRef]
- Ilina, O.; Friedl, P. Mechanisms of collective cell migration at a glance. J. Cell Sci. 2009, 122, 3203–3208. [Google Scholar] [CrossRef] [Green Version]
- Hou, J.M.; Krebs, M.; Ward, T.; Sloane, R.; Priest, L.; Hughes, A.; Clack, G.; Ranson, M.; Blackhall, F.; Dive, C. Circulating tumor cells as a window on metastasis biology in lung cancer. Am. J. Pathol. 2011, 178, 989–996. [Google Scholar] [CrossRef]
- Hou, J.M.; Krebs, M.G.; Lancashire, L.; Sloane, R.; Backen, A.; Swain, R.K.; Priest, L.; Greystoke, A.; Zhou, C.; Morrise, K.; et al. Clinical significance and molecular characteristics of circulating tumor cells and circulating tumor microemboli in patients with small-cell lung cancer. J. Clin. Oncol. 2012, 30, 525–532. [Google Scholar] [CrossRef]
- Erpenbeck, L.; Schon, M.P. Deadly allies: The fatal interplay between platelets and metastasizing cancer cells. Blood 2010, 115, 3427–3436. [Google Scholar] [CrossRef]
- Labelle, M.; Begum, S.; Hynes, R.O. Direct signaling between platelets and cancer cells induces an epithelial-mesenchymal-like transition and promotes metastasis. Cancer Cell 2011, 20, 576–590. [Google Scholar] [CrossRef] [Green Version]
- Fawcett, D.W.; Vallee, B.L.; Soule, M.H. A method for concentration and segregation of malignant cells from bloody, pleural, and peritoneal fluids. Science 1950, 111, 34–36. [Google Scholar] [CrossRef]
- Lambros, M.B.; Seed, G.; Sumanasuriya, S.; Gil, V.; Crespo, M.; Fontes, M.; Chandler, R.; Mehra, N.; Fowler, G.; Ebbs, B.; et al. Single-Cell Analyses of Prostate Cancer Liquid Biopsies Acquired by Apheresis. Clin. Cancer Res. 2018, 24, 5635–5644. [Google Scholar] [CrossRef] [Green Version]
- Ogle, L.F.; Orr, J.G.; Willoughby, C.E.; Hutton, C.; Mcpherson, S.; Plummer, R.; Boddy, A.V.; Curtin, N.J.; Jamieson, D.; Reeves, H.L.; et al. Imagestream detection and characterisation of circulating tumour cells—A liquid biopsy for hepatocellular carcinoma? J. Hepatol. 2016, 65, 305–313. [Google Scholar] [CrossRef] [Green Version]
- Boya, M.; Chu, C.; Liu, R.; Ozkaya-Ahmadov, T.; Sarioglu, A.F. Circulating Tumor Cell Enrichment Technologies. Recent Results Cancer Res. 2020, 215, 25–55. [Google Scholar] [PubMed]
- Yu, M.; Stott, S.; Toner, M.; Maheswaran, S.; Haber, D.A. Circulating tumor cells: Approaches to isolation and characterization. J. Cell Biol. 2011, 192, 373–382. [Google Scholar] [CrossRef] [PubMed]
- Song, Y.; Tian, T.; Shi, Y.; Liu, W.; Zou, Y.; Khajvand, T.; Wang, S.; Zhu, Z.; Yang, C. Enrichment and single-cell analysis of circulating tumor cells. Chem. Sci. 2017, 8, 1736–1751. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nagrath, S.; Jack, R.M.; Sahai, V.; Simeone, D.M. Opportunities and Challenges for Pancreatic Circulating Tumor Cells. Gastroenterology 2016, 151, 412–426. [Google Scholar] [CrossRef] [Green Version]
- Franken, A.; Driemel, C.; Behrens, B.; Meier-Stiegen, F.; Endris, V.; Stenzinger, A.; Niederacher, D.; Fischer, J.C.; Stoecklein, N.H.; Ruckhaeberle, E.; et al. Label-Free Enrichment and Molecular Characterization of Viable Circulating Tumor Cells from Diagnostic Leukapheresis Products. Clin. Chem. 2019, 65, 549–558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, H.J.; Cho, H.; Oh, J.H.; Namkoong, K.; Lee, J.G.; Park, J.; Lee, S.S.; Huh, N.; Choi, J. Simultaneous capture and in situ analysis of circulating tumor cells using multiple hybrid nanoparticles. Biosens. Bioelectron. 2013, 47, 508–514. [Google Scholar] [CrossRef]
- Hoshino, K.; Huang, Y.; Lane, N.; Huebschman, M.; Uhr, J.W.; Frenkel, E.P.; Zhang, X. Microchip-based immunomagnetic detection of circulating tumor cells. Lab. Chip 2011, 11, 3449–3457. [Google Scholar] [CrossRef]
- Went, P.T.; Lugli, A.; Meier, S.; Bundi, M.; Mirlacher, M.; Sauter, G.; Dirnhofer, S. Frequent EpCam protein expression in human carcinomas. Hum. Pathol. 2004, 35, 122–128. [Google Scholar] [CrossRef]
- Lin, H.K.; Zheng, S.; Williams, A.J.; Balic, M.; Groshen, S.; Scher, H.I.; Fleisher, M.; Stadler, W.; Datar, R.H.; Tai, Y.; et al. Portable filter-based microdevice for detection and characterization of circulating tumor cells. Clin. Cancer Res. 2010, 16, 5011–5018. [Google Scholar] [CrossRef] [Green Version]
- Joosse, S.A.; Gorges, T.M.; Pantel, K. Biology, detection, and clinical implications of circulating tumor cells. EMBO Mol. Med. 2015, 7, 1–11. [Google Scholar] [CrossRef]
- Gires, O.; Stoecklein, N.H. Dynamic EpCAM expression on circulating and disseminating tumor cells: Causes and consequences. Cell Mol. Life Sci. 2014, 71, 4393–4402. [Google Scholar] [CrossRef]
- Barriere, G.; Tartary, M.; Rigaud, M. Epithelial mesenchymal transition: A new insight into the detection of circulating tumor cells. ISRN Oncol. 2012, 2012, 382010. [Google Scholar] [CrossRef] [Green Version]
- Ferreira, M.M.; Ramani, V.C.; Jeffrey, S.S. Circulating tumor cell technologies. Mol. Oncol. 2016, 10, 374–394. [Google Scholar] [CrossRef] [Green Version]
- Ahn, J.C.; Teng, P.; Chen, P.; Posadas, E.; Tseng, H.; Lu, S.C.; Yang, J.D. Detection of circulating tumor cells and their implications as a novel biomarker for diagnosis, prognostication, and therapeutic monitoring in hepatocellular carcinoma. Hepatology 2020. [Google Scholar] [CrossRef]
- Wang, W.C.; Zhang, X.F.; Peng, J.; Li, X.F.; Wang, A.L.; Bie, Y.Q.; Shi, L.H.; Lin, M.B.; Zhang, X.F. Survival Mechanisms and Influence Factors of Circulating Tumor Cells. Biomed. Res. Int. 2018, 2018, 6304701. [Google Scholar] [CrossRef] [PubMed]
- Ross, A.A.; Cooper, B.W.; Lazarus, H.M.; Mackay, W.; Moss, T.J.; Ciobanu, N.; Tallman, M.S.; Kennedy, M.J.; Davidson, N.E.; Sweet, D.; et al. Detection and viability of tumor cells in peripheral blood stem cell collections from breast cancer patients using immunocytochemical and clonogenic assay techniques. Blood 1993, 82, 2605–2610. [Google Scholar] [CrossRef]
- Gazzaniga, P.; Gandini, O.; Giuliani, L.; Magnanti, M.; Gradilone, A.; Silvestri, I.; Gianni, W.; Gallucci, M.; Frati, L.; Agliano, A.M. Detection of epidermal growth factor receptor mRNA in peripheral blood: A new marker of circulating neoplastic cells in bladder cancer patients. Clin. Cancer Res. 2001, 7, 577–583. [Google Scholar]
- Cohen, S.J.; Punt, C.J.; Iannotti, N.; Saidman, B.H.; Sabbath, K.D.; Gabrail, N.Y.; Picus, J.; Morse, M.A.; Mitchell, E.; Miller, M.C.; et al. Prognostic significance of circulating tumor cells in patients with metastatic colorectal cancer. Ann. Oncol. 2009, 20, 1223–1229. [Google Scholar] [CrossRef] [PubMed]
- Cristofanilli, M.; Budd, G.T.; Ellis, M.J.; Stopeck, A.; Matera, J.; Miller, M.C.; Reuben, J.M.; Doyle, G.V.; Allard, W.J.; Terstappen, L.W.; et al. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N. Engl. J. Med. 2004, 351, 781–791. [Google Scholar] [CrossRef] [Green Version]
- De Bono, J.S.; Scher, H.I.; Montgomery, R.B.; Parker, C.; Miller, M.C.; Tissing, H.; Doyle, G.V.; Terstappen, L.W.; Pienta, K.J.; Raghavan, D. Circulating tumor cells predict survival benefit from treatment in metastatic castration-resistant prostate cancer. Clin. Cancer Res. 2008, 14, 6302–6309. [Google Scholar] [CrossRef] [Green Version]
- Krebs, M.G.; Sloane, R.; Priest, L.; Lancashire, L.; Hou, J.M.; Greystoke, A.; Ward, T.H.; Ferraldeschi, R.; Hughes, A.; Clack, G.; et al. Evaluation and prognostic significance of circulating tumor cells in patients with non-small-cell lung cancer. J. Clin. Oncol. 2011, 29, 1556–1563. [Google Scholar] [CrossRef] [PubMed]
- Meng, S.; Tripathy, D.; Frenkel, E.P.; Shete, S.; Naftalis, E.Z.; Huth, J.F.; Beitsch, P.D.; Leitch, M.; Hoover, S.; Euhus, D.; et al. Circulating tumor cells in patients with breast cancer dormancy. Clin. Cancer Res. 2004, 10, 8152–8162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hesketh, R.L.; Zhu, A.X.; Oklu, R. Hepatocellular carcinoma: Can circulating tumor cells and radiogenomics deliver personalized care? Am. J. Clin. Oncol. 2015, 38, 431–436. [Google Scholar] [CrossRef] [Green Version]
- Chang, Y.S.; Tomaso, E.D.; McDonald, D.M.; Jones, R.; Jain, R.K.; Munn, L.L. Mosaic blood vessels in tumors: Frequency of cancer cells in contact with flowing blood. Proc. Natl. Acad. Sci. USA 2000, 97, 14608–14613. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhao, R.; Cai, Z.; Li, S.; Cheng, Y.; Gao, H.; Liu, F.; Wu, S.; Liu, S.; Dong, Y.; Zheng, L.; et al. Expression and clinical relevance of epithelial and mesenchymal markers in circulating tumor cells from colorectal cancer. Oncotarget 2017, 8, 9293–9302. [Google Scholar] [CrossRef] [Green Version]
- Hay, E.D. The mesenchymal cell, its role in the embryo, and the remarkable signaling mechanisms that create it. Dev. Dyn. 2005, 233, 706–720. [Google Scholar] [CrossRef]
- Stott, S.L.; Lee, R.J.; Nagrath, S.; Yu, M.; Miyamoto, D.T.; Ulkus, L.; Inserra, E.J.; Ulman, M.; Springer, S.; Nakamura, Z.; et al. Isolation and characterization of circulating tumor cells from patients with localized and metastatic prostate cancer. Sci. Transl. Med. 2010, 2, 25ra23. [Google Scholar] [CrossRef] [Green Version]
- Nagrath, S.; Sequist, L.V.; Maheswaran, S.; Bell, D.W.; Irimia, D.; Ulkus, L.; Smith, M.R.; Kwak, E.L.; Digumarthy, S.; Muzikansky, A.; et al. Isolation of rare circulating tumour cells in cancer patients by microchip technology. Nature 2007, 450, 1235–1239. [Google Scholar] [CrossRef] [Green Version]
- Allard, W.J.; Matera, J.; Miller, M.C.; Repollet, M.; Connelly, M.C.; Rao, C.; Tibbe, A.G.; Uhr, J.W.; Terstappen, L.W. Tumor cells circulate in the peripheral blood of all major carcinomas but not in healthy subjects or patients with nonmalignant diseases. Clin. Cancer Res. 2004, 10, 6897–6904. [Google Scholar] [CrossRef] [Green Version]
- Baccelli, I.; Schneeweiss, A.; Riethdorf, S.; Stenzinger, A.; Schillert, A.; Vogel, V.; Klein, C.; Saini, M.; Bäuerle, T.; Wallwiener, M.; et al. Identification of a population of blood circulating tumor cells from breast cancer patients that initiates metastasis in a xenograft assay. Nat. Biotechnol. 2013, 31, 539–544. [Google Scholar] [CrossRef] [PubMed]
- Sun, Y.F.; Guo, W.; Xu, Y.; Shi, Y.H.; Gong, Z.J.; Ji, Y.; Du, M.; Zhang, X.; Hu, B.; Huang, A.; et al. Circulating Tumor Cells from Different Vascular Sites Exhibit Spatial Heterogeneity in Epithelial and Mesenchymal Composition and Distinct Clinical Significance in Hepatocellular Carcinoma. Clin. Cancer Res. 2018, 24, 547–559. [Google Scholar] [CrossRef] [Green Version]
- Wan, L.; Pantel, K.; Kang, Y. Tumor metastasis: Moving new biological insights into the clinic. Nat. Med. 2013, 19, 1450–1464. [Google Scholar] [CrossRef] [PubMed]
- Hodgkinson, C.L.; Morrow, C.J.; Li, Y.; Metcalf, R.L.; Rothwell, D.G.; Trapani, F.; Polanski, R.; Burt, D.J.; Simpson, K.L.; Morris, K.; et al. Tumorigenicity and genetic profiling of circulating tumor cells in small-cell lung cancer. Nat. Med. 2014, 20, 897–903. [Google Scholar] [CrossRef]
- Yang, J.; Weinberg, R.A. Epithelial-mesenchymal transition: At the crossroads of development and tumor metastasis. Dev. Cell 2008, 14, 818–829. [Google Scholar] [CrossRef] [Green Version]
- Skrypek, N.; Goossens, S.; De Smedt, E.; Vandamme, N.; Berx, G. Epithelial-to-Mesenchymal Transition: Epigenetic Reprogramming Driving Cellular Plasticity. Trends Genet. 2017, 33, 943–959. [Google Scholar] [CrossRef]
- Polyak, K.; Weinberg, R.A. Transitions between epithelial and mesenchymal states: Acquisition of malignant and stem cell traits. Nat. Rev. Cancer 2009, 9, 265–273. [Google Scholar] [CrossRef]
- Nieto, M.A.; Huang, R.Y.; Jackson, R.A.; Thiery, J.P. Emt: 2016. Cell 2016, 166, 21–45. [Google Scholar] [CrossRef] [Green Version]
- Thiery, J.P.; Acloque, H.; Huang, R.Y.; Nieto, M.A. Epithelial-mesenchymal transitions in development and disease. Cell 2009, 139, 871–890. [Google Scholar] [CrossRef]
- Tarin, D.; Thompson, E.W.; Newgreen, D.F. The fallacy of epithelial mesenchymal transition in neoplasia. Cancer Res. 2005, 65, 5996–6000, discussion 6000-1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ledford, H. Cancer theory faces doubts. Nature 2011, 472, 273. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.; Ridgway, L.D.; Wetzel, M.D.; Ngo, J.; Yin, W.; Kumar, D.; Goodman, J.C.; Groves, M.D.; Marchetti, D. The identification and characterization of breast cancer CTCs competent for brain metastasis. Sci. Transl. Med. 2013, 5, 180ra48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yu, M.; Bardia, A.; Wittner, B.S.; Stott, S.L.; Smas, M.E.; Ting, D.T.; Isakoff, S.J.; Ciciliano, J.C.; Wells, M.N.; Shah, A.M.; et al. Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition. Science 2013, 339, 580–584. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lecharpentier, A.; Vielh, P.; Perez-Moreno, P.; Planchard, D.; Soria, J.C.; Farace, F. Detection of circulating tumour cells with a hybrid (epithelial/mesenchymal) phenotype in patients with metastatic non-small cell lung cancer. Br. J. Cancer 2011, 105, 1338–1341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Armstrong, A.J.; Marengo, M.S.; Oltean, S.; Kemeny, G.; Bitting, R.L.; Turnbull, J.D.; Herold, C.I.; Marcom, P.K.; George, D.J.; Garcia-Blanco, M.A. Circulating tumor cells from patients with advanced prostate and breast cancer display both epithelial and mesenchymal markers. Mol. Cancer Res. 2011, 9, 997–1007. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Min, A.L.; Marengo, M.S.; Oltean, S.; Kemeny, G.; Bitting, R.L.; Turnbull, J.D.; Herold, C.I.; Marcom, P.K.; George, D.J.; Garcia-Blanco, M.A. High expression of Snail mRNA in blood from hepatocellular carcinoma patients with extra-hepatic metastasis. Clin. Exp. Metastasis 2009, 26, 759–767. [Google Scholar] [CrossRef]
- Wang, Q.; Duan, L.X.; Xu, Z.S.; Wang, J.G.; Xi, S.M. The protective effect of the earthworm active ingredients on hepatocellular injury induced by endoplasmic reticulum stress. Biomed. Pharmacother. 2016, 82, 304–311. [Google Scholar] [CrossRef]
- Zeisberg, M.; Neilson, E.G. Biomarkers for epithelial-mesenchymal transitions. J. Clin. Investig. 2009, 119, 1429–1437. [Google Scholar] [CrossRef] [Green Version]
- Sleeman, J.P.; Thiery, J.P. SnapShot: The epithelial-mesenchymal transition. Cell 2011, 145, 162. [Google Scholar] [CrossRef] [Green Version]
- Gorges, T.M.; Tinhofer, I.; Drosch, M.; Röse, L.; Zollner, T.M.; Krahn, T.; von Ahsen, O. Circulating tumour cells escape from EpCAM-based detection due to epithelial-to-mesenchymal transition. BMC Cancer 2012, 12, 178. [Google Scholar] [CrossRef] [Green Version]
- Škovierová, H.; Okajčeková, T.; Strnádel, J.; Vidomanová, E.; Halašová, E. Molecular regulation of epithelial-to-mesenchymal transition in tumorigenesis (Review). Int. J. Mol. Med. 2018, 41, 1187–1200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lou, J.; Zhang, L.; Lv, S.; Zhang, C.; Jiang, S. Biomarkers for Hepatocellular Carcinoma. Biomark. Cancer 2017, 9, 1–9. [Google Scholar] [CrossRef] [PubMed]
- Yang, J.D.; Kim, W.R.; Park, K.W.; Chaiteerakij, R.; Kim, B.; Sanderson, S.O.; Larson, J.J.; Pedersen, R.A.; Therneau, T.M.; Gores, G.J.; et al. Model to estimate survival in ambulatory patients with hepatocellular carcinoma. Hepatology 2012, 56, 614–621. [Google Scholar] [CrossRef] [PubMed]
- Marrero, J.A.; Kulik, L.M.; Sirlin, C.B.; Zhu, A.X.; Finn, R.S.; Abecassis, M.M.; Roberts, L.R.; Heimbach, J.K. Diagnosis, Staging, and Management of Hepatocellular Carcinoma: 2018 Practice Guidance by the American Association for the Study of Liver Diseases. Hepatology 2018, 68, 723–750. [Google Scholar] [CrossRef] [Green Version]
- European Association for the Study of the Liver. Electronic address, e.e.e. L. European Association for the Study of the EASL Clinical Practice Guidelines: Management of hepatocellular carcinoma. J. Hepatol. 2018, 69, 182–236. [Google Scholar] [CrossRef] [Green Version]
- Omata, M.; Cheng, A.L.; Kokudo, N.; Kudo, M.; Lee, J.M.; Jia, J.; Tateishi, R.; Han, K.H.; Chawla, Y.K.; Shiina, S.; et al. Asia-Pacific clinical practice guidelines on the management of hepatocellular carcinoma: A 2017 update. Hepatol. Int. 2017, 11, 317–370. [Google Scholar] [CrossRef] [Green Version]
- Tzartzeva, K.; Obi, J.; Rich, N.E.; Parikh, N.D.; Marrero, J.A.; Yopp, A.; Waljee, A.K.; Singal, A.G. Surveillance Imaging and Alpha Fetoprotein for Early Detection of Hepatocellular Carcinoma in Patients with Cirrhosis: A Meta-analysis. Gastroenterology 2018, 154, 1706–1718.e1. [Google Scholar] [CrossRef] [Green Version]
- Ma, X.L.; Gao, X.H.; Gong, Z.J.; Wu, J.; Tian, L.; Zhang, C.Y.; Zhou, Y.; Sun, Y.F.; Hu, B.; Qiu, S.J.; et al. Apolipoprotein A1: A novel serum biomarker for predicting the prognosis of hepatocellular carcinoma after curative resection. Oncotarget 2016, 7, 70654–70668. [Google Scholar] [CrossRef]
- Hsu, H.C.; Cheng, W.; Lai, P.L. Cloning and expression of a developmentally regulated transcript MXR7 in hepatocellular carcinoma: Biological significance and temporospatial distribution. Cancer Res. 1997, 57, 5179–5184. [Google Scholar]
- Wang, H.L.; Anatelli, F.; Zhai, Q.J.; Adley, B.; Chuang, S.T.; Yang, X.J. Glypican-3 as a useful diagnostic marker that distinguishes hepatocellular carcinoma from benign hepatocellular mass lesions. Arch. Pathol. Lab. Med. 2008, 132, 1723–1728. [Google Scholar]
- Libbrecht, L.; Severi, T.; Cassiman, D.; Vander Borght, S.; Pirenne, J.; Nevens, F.; Verslype, C.; van Pelt, J.; Roskams, T. Glypican-3 expression distinguishes small hepatocellular carcinomas from cirrhosis, dysplastic nodules, and focal nodular hyperplasia-like nodules. Am. J. Surg. Pathol. 2006, 30, 1405–1411. [Google Scholar] [CrossRef] [PubMed]
- Sun, B.; Huang, Z.; Wang, B.; Yu, Y.; Lin, S.; Luo, L.; Wang, Y.; Huang, Z. Significance of Glypican-3 (GPC3) Expression in Hepatocellular Cancer Diagnosis. Med. Sci. Monit. 2017, 23, 850–855. [Google Scholar] [CrossRef] [Green Version]
- Capurro, M.; Wanless, I.R.; Sherman, M.; Deboer, G.; Shi, W.; Miyoshi, E.; Filmus, J. Glypican-3: A novel serum and histochemical marker for hepatocellular carcinoma. Gastroenterology 2003, 125, 89–97. [Google Scholar] [CrossRef]
- Park, J.H.; Cho, E.W.; Shin, S.Y.; Lee, Y.J.; Kim, K.L. Detection of the asialoglycoprotein receptor on cell lines of extrahepatic origin. Biochem. Biophys. Res. Commun. 1998, 244, 304–311. [Google Scholar] [CrossRef] [PubMed]
- Wennerberg, A.E.; Nalesnik, M.A.; Coleman, W.B. Hepatocyte paraffin 1: A monoclonal antibody that reacts with hepatocytes and can be used for differential diagnosis of hepatic tumors. Am. J. Pathol. 1993, 143, 1050–1054. [Google Scholar] [PubMed]
- Lamps, L.W.; Folpe, A.L. The diagnostic value of hepatocyte paraffin antibody 1 in differentiating hepatocellular neoplasms from nonhepatic tumors: A review. Adv. Anat. Pathol. 2003, 10, 39–43. [Google Scholar] [CrossRef]
- Christa, L.; Simon, M.T.; Flinois, J.P.; Gebhardt, R.; Brechot, C.; Lasserre, C. Overexpression of glutamine synthetase in human primary liver cancer. Gastroenterology 1994, 106, 1312–1320. [Google Scholar] [CrossRef]
- Osada, T.; Sakamoto, M.; Nagawa, H.; Yamamoto, J.; Matsuno, Y.; Iwamatsu, A.; Muto, T.; Hirohashi, S. Acquisition of glutamine synthetase expression in human hepatocarcinogenesis: Relation to disease recurrence and possible regulation by ubiquitin-dependent proteolysis. Cancer 1999, 85, 819–831. [Google Scholar] [CrossRef]
- Butler, S.L.; Dong, H.; Cardona, D.; Jia, M.; Zheng, R.; Zhu, H.; Crawford, J.M.; Liu, C. The antigen for Hep Par 1 antibody is the urea cycle enzyme carbamoyl phosphate synthetase 1. Lab. Investig. 2008, 88, 78–88. [Google Scholar] [CrossRef] [Green Version]
- Heeke, S.; Mograbi, B.; Alix-Panabières, C.; Hofman, P. Never Travel Alone: The Crosstalk of Circulating Tumor Cells and the Blood Microenvironment. Cells 2019, 8, 714. [Google Scholar] [CrossRef] [Green Version]
- Jie, X.X.; Zhang, X.Y.; Xu, C.J. Epithelial-to-mesenchymal transition, circulating tumor cells and cancer metastasis: Mechanisms and clinical applications. Oncotarget 2017, 8, 81558–81571. [Google Scholar] [CrossRef] [Green Version]
- Steinert, G.; Schölch, S.; Niemietz, T.; Iwatam, N.; García, S.A.; Behrens, B.; Voigt, A.; Kloor, M.; Benner, A.; Bork, U.; et al. Immune escape and survival mechanisms in circulating tumor cells of colorectal cancer. Cancer Res. 2014, 74, 1694–1704. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lang, L. FDA approves sorafenib for patients with inoperable liver cancer. Gastroenterology 2008, 134, 379. [Google Scholar] [CrossRef] [PubMed]
- Cheng, A.L.; Kang, Y.K.; Chen, Z.; Tsao, C.J.; Qin, S.; Kim, J.S.; Luo, R.; Feng, J.; Ye, S.; Yang, T.S.; et al. Efficacy and safety of sorafenib in patients in the Asia-Pacific region with advanced hepatocellular carcinoma: A phase III randomised, double-blind, placebo-controlled trial. Lancet Oncol. 2009, 10, 25–34. [Google Scholar] [CrossRef]
- Llovet, J.M.; Ricci, S.; Mazzaferro, V.; Hilgard, P.; Gane, E.; Blanc, J.F.; de Oliveira, A.C.; Santoro, A.; Raoul, J.L.; Forner, A.; et al. Sorafenib in advanced hepatocellular carcinoma. N. Engl. J. Med. 2008, 359, 378–390. [Google Scholar] [CrossRef]
- Liu, Q.; Liao, Q.; Zhao, Y. Myeloid-derived suppressor cells (MDSC) facilitate distant metastasis of malignancies by shielding circulating tumor cells (CTC) from immune surveillance. Med. Hypotheses 2016, 87, 34–39. [Google Scholar] [CrossRef] [PubMed]
- Hamilton, G.; Rath, B.; Klameth, L.; Hochmair, M.J. Small cell lung cancer: Recruitment of macrophages by circulating tumor cells. Oncoimmunology 2016, 5, e1093277. [Google Scholar] [CrossRef] [Green Version]
- Hamilton, G.; Rath, B. Circulating tumor cell interactions with macrophages: Implications for biology and treatment. Transl. Lung Cancer Res. 2017, 6, 418–430. [Google Scholar] [CrossRef] [Green Version]
- Mishra, L.; Banker, T.; Murray, J.; Byers, S.; Thenappan, A.; He, A.R.; Shetty, K.; Johnson, L.; Reddy, E.P. Liver stem cells and hepatocellular carcinoma. Hepatology 2009, 49, 318–329. [Google Scholar] [CrossRef] [Green Version]
- Singh, S.K.; Clarke, I.D.; Terasaki, M.; Bonn, V.E.; Hawkins, C.; Squire, J.; Dirks, P.B. Identification of a cancer stem cell in human brain tumors. Cancer Res. 2003, 63, 5821–5828. [Google Scholar]
- Li, C.; Heidt, D.G.; Dalerba, P.; Burant, C.F.; Zhang, L.; Adsay, V.; Wicha, M.; Clarke, M.F.; Simeone, D.M. Identification of pancreatic cancer stem cells. Cancer Res. 2007, 67, 1030–1037. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ho, M.M.; Ng, A.V.; Lam, S.; Hung, J.Y. Side population in human lung cancer cell lines and tumors is enriched with stem-like cancer cells. Cancer Res. 2007, 67, 4827–4833. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bertolini, G.; D’Amico, L.; Moro, M.; Landoni, E.; Perego, P.; Miceli, R.; Gatti, L.; Andriani, F.; Wong, D.; Caserini, R.; et al. Microenvironment-Modulated Metastatic CD133+/CXCR4+/EpCAM- Lung Cancer-Initiating Cells Sustain Tumor Dissemination and Correlate with Poor Prognosis. Cancer Res. 2015, 75, 3636–3649. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sarvi, S.; Mackinnon, A.C.; Avlonitis, N.; Bradley, M.; Rintoul, R.C.; Rassl, D.M.; Wang, W.; Forbes, S.J.; Gregory, C.D.; Sethi, T. CD133+ cancer stem-like cells in small cell lung cancer are highly tumorigenic and chemoresistant but sensitive to a novel neuropeptide antagonist. Cancer Res. 2014, 74, 1554–1565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Haraguchi, N.; Ishii, H.; Mimori, K.; Tanaka, F.; Ohkuma, M.; Kim, H.M.; Akita, H.; Takiuchi, D.; Hatano, H.; Nagano, H.; et al. CD13 is a therapeutic target in human liver cancer stem cells. J. Clin. Investig. 2010, 120, 3326–3339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Suetsugu, A.; Nagaki, M.; Aoki, H.; Motohashi, T.; Kunisada, T.; Moriwaki, H. Characterization of CD133+ hepatocellular carcinoma cells as cancer stem/progenitor cells. Biochem. Biophys. Res. Commun. 2006, 351, 820–824. [Google Scholar] [CrossRef]
- Ma, S.; Chan, K.W.; Hu, L.; Lee, T.K.; Wo, J.Y.; Ng, I.O.; Zheng, B.J.; Guan, X.Y. Identification and characterization of tumorigenic liver cancer stem/progenitor cells. Gastroenterology 2007, 132, 2542–2556. [Google Scholar] [CrossRef]
- Yamashita, T.; Ji, J.; Budhu, A.; Forgues, M.; Yang, W.; Wang, H.Y.; Jia, H.; Ye, Q.; Qin, L.X.; Wauthier, E.; et al. EpCAM-positive hepatocellular carcinoma cells are tumor-initiating cells with stem/progenitor cell features. Gastroenterology 2009, 136, 1012–1024. [Google Scholar] [CrossRef] [Green Version]
- Yang, Z.F.; Ho, D.W.; Ng, M.N.; Lau, C.K.; Yu, W.C.; Ngai, P.; Chu, P.W.; Lam, C.T.; Poon, R.T.; Fan, S.T. Significance of CD90+ cancer stem cells in human liver cancer. Cancer Cell 2008, 13, 153–166. [Google Scholar] [CrossRef] [Green Version]
- Fan, S.T.; Yang, Z.F.; Ho, D.W.; Ng, M.N.; Yu, W.C.; Wong, J. Prediction of posthepatectomy recurrence of hepatocellular carcinoma by circulating cancer stem cells: A prospective study. Ann. Surg. 2011, 254, 569–576. [Google Scholar] [CrossRef]
- Luo, Y.T.; Cheng, J.; Feng, X.; He, S.J.; Wang, Y.W.; Huang, Q. The viable circulating tumor cells with cancer stem cells feature, where is the way out? J. Exp. Clin. Cancer Res. 2018, 37, 38. [Google Scholar] [CrossRef]
- Terris, B.; Cavard, C.; Perret, C. EpCAM, a new marker for cancer stem cells in hepatocellular carcinoma. J. Hepatol. 2010, 52, 280–281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Munz, M.; Baeuerle, P.A.; Gires, O. The emerging role of EpCAM in cancer and stem cell signaling. Cancer Res. 2009, 69, 5627–5629. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, L.; Li, Y.; Xu, J.; Zhang, A.; Wang, X.; Tang, R.; Zhang, X.; Yin, H.; Liu, M.; Wang, D.D.; et al. Quantified postsurgical small cell size CTCs and EpCAM(+) circulating tumor stem cells with cytogenetic abnormalities in hepatocellular carcinoma patients determine cancer relapse. Cancer Lett. 2018, 412, 99–107. [Google Scholar] [CrossRef] [PubMed]
- Wan, S.; Zhao, E.; Kryczek, I.; Vatan, L.; Sadovskaya, A.; Ludema, G.; Simeone, D.M.; Zou, W.; Welling, T.H. Tumor-associated macrophages produce interleukin 6 and signal via STAT3 to promote expansion of human hepatocellular carcinoma stem cells. Gastroenterology 2014, 147, 1393–1404. [Google Scholar] [CrossRef] [Green Version]
- Chaffer, C.L.; Weinberg, R.A. A perspective on cancer cell metastasis. Science 2011, 331, 1559–1564. [Google Scholar] [CrossRef]
- Wu, N.; Liu, S.; Guo, C.; Hou, Z.; Sun, M.Z. The role of annexin A3 playing in cancers. Clin. Transl. Oncol. 2013, 15, 106–110. [Google Scholar] [CrossRef]
- Pan, Q.Z.; Pan, K.; Weng, D.S.; Zhao, J.J.; Zhang, X.F.; Wang, D.D.; Lv, L.; Jiang, S.S.; Zheng, H.X.; Xia, J.C. Annexin A3 promotes tumorigenesis and resistance to chemotherapy in hepatocellular carcinoma. Mol. Carcinog. 2015, 54, 598–607. [Google Scholar] [CrossRef]
- Pan, Q.Z.; Pan, K.; Wang, Q.J.; Weng, D.S.; Zhao, J.J.; Zheng, H.X.; Zhang, X.F.; Jiang, S.S.; Lv, L.; Tang, Y.; et al. Annexin A3 as a potential target for immunotherapy of liver caner stem-like cells. Stem. Cells 2015, 33, 354–366. [Google Scholar] [CrossRef]
- Tong, M.; Fung, T.M.; Luk, S.T.; Ng, K.Y.; Lee, T.K.; Lin, C.H.; Yam, J.W.; Chan, K.W.; Ng, F.; Zheng, B.J.; et al. ANXA3/JNK Signaling Promotes Self-Renewal and Tumor Growth, and Its Blockade Provides a Therapeutic Target for Hepatocellular Carcinoma. Stem. Cell Rep. 2015, 5, 45–59. [Google Scholar] [CrossRef] [Green Version]
- Farinati, F.; Marino, D.; De Giorgio, M.; Baldan, A.; Cantarini, M.; Cursaro, C.; Rapaccini, G.; Del Poggio, P.; Di Nolfo, M.A.; Benvegnù, L.; et al. Diagnostic and prognostic role of alpha-fetoprotein in hepatocellular carcinoma: Both or neither? Am. J. Gastroenterol. 2006, 101, 524–532. [Google Scholar] [CrossRef] [PubMed]
- Taketa, K. Alpha-fetoprotein: Reevaluation in hepatology. Hepatology 1990, 12, 1420–1432. [Google Scholar] [CrossRef] [PubMed]
- Johnson, P.J. The role of serum alpha-fetoprotein estimation in the diagnosis and management of hepatocellular carcinoma. Clin. Liver Dis. 2001, 5, 145–159. [Google Scholar] [CrossRef]
Phenotypic Markers | Enrichment Method | Specimen | Key Findings | Ref |
---|---|---|---|---|
Is CTCs heterogeneity compatible with EMT (epithelial to mesenchymal transition) markers? | ||||
EpCAM | qRT-PCR-based platform | 299 HCC patients and 120 control subjects | Compared with pre-operation, the population of EpCAM+ CTCs decreased significantly after operation, and all the patients with CTC reduction showed tumor remission. | [16] |
EpCAM | CellSearch system | 59 HCC patients and 19 control patients | CTCs in the presence of EpCAM were strongly correlated with tumor aggressiveness, and this allowed adequate stratification of HCC patients for curative or systemic therapy. | [17] |
Twist, GPC-3 | CanPatrol system | 80 HCC patients and 10 healthy volunteers | The ratio of twist+ CTCs was closely correlated with the rate of metastasis or recurrence and the mortality rate; the prognostic evaluation of twist+ CTCs was better than CTCs alone. | [18] |
EpCAM, CK8/18/19, and vimentin, twist | CanPatrol system | 165 HCC patients | The presence of mesenchymal CTCs tended to occur in patients with advanced stage, and was associated with decreased relapse-free survival. | [19] |
EpCAM, CK8/18/19, and vimentin, twist | CanPatrol system | 113 HCC patients | The use of total CTCs was more effective than AFP for the diagnosis of HCC, and the combination of total CTCs and AFP could enhance diagnostic effectiveness. | [20] |
EpCAM, CK8/18/19, E-cadherin, vimentin, twist, AKT2, and snail | CanPatrol system | 195 HCC patients | Mesenchymal and hybrid CTCs had higher invasive and metastatic abilities than E type CTCs. | [21] |
E-cadherin, vimentin, and twist | Flow cytometric analysis, and immunofluorescence staining | 46 HCC patients | Co-expression of twist and vimentin in CTCs was significantly correlated with portal vein tumor thrombus, TNM classification, and tumor size. | [22] |
EpCAM, CK8/18/19, and vimentin, twist | CanPatrol system | 62 HCC patients | HCC patients with positive peripheral mesenchymal CTCs had a higher risk of early recurrence. | [23] |
EpCAM, CK8/18/19, and vimentin, twist | CanPatrol system | 33 HCC patients and 10 healthy volunteers | Epithelial-mesenchymal-mixed CTCs played an important role in EMT transition of HCC. The mixed CTCs might be a vital factor for intrahepatic metastasis, and mesenchymal CTCs could have potential to be a predictor of extrahepatic metastasis. | [24] |
EpCAM, CK8/18/19, and vimentin, twist | CanPatrol system | 40 HCC patients | The average ratio of mesenchymal CTCs in each sample was increased in the later stages of cancer compared with the earlier stages of cancer. | [25] |
EpCAM, E-cadherin, CK8/18/19, vimentin, and twist, BCAT1 | CanPatrol system | 112 HCC patients | The percentage of BCAT1 was positively correlated with EMT process, suggesting a potential marker for CTCs in evaluating tumor metastasis or recurrence. | [26] |
Hepatocyte-specific markers of CTCs in HCC | ||||
GPC3, AFP | Enzyme-linked immunoassay | 68 HCC patients | The combination of GPC3 and AFP improved the overall sensitivity for HCC; the positive rate of GPC3 was significantly higher than that of AFP in HCC patients. | [27] |
GPC3 | Density gradient centrifugation and immunomagnetic positive enrichment | 85 HCC patients | Pre-operative GPC3-positive CTCs was a risk factor of microscopic portal vein invasion and poor prognosis, and therefore it might be a useful biomarker for HCC patient outcomes. | [28] |
ASGPR | Microfluidic chip | 36 HCC patients | CTCs were detected in all the examined patients with HCC. | [29] |
ASGPR, CPS1, P-CK | Density gradient Ficoll-Paque PLUS, and magnetic labeling and separation | 27 HCC patients | All the 16 HCC tissues had ASGPR staining on the membranes of the HCC cells, and CTCs in the presence of CPS1 and P-CK were detected in the majority of patients with HCC. | [30] |
ASGPR, GPC3 | Magnetically assisted surface-enhanced Raman scattering biosensor | Eight HCC patients, three breast cancer patients, and three healthy controls | Dual labelling of ASGPR and GPC3 was effective in detecting HCC CTCs with a small volume of blood samples in clinical settings. | [31] |
ASGPR, GPC3, CK | Semiquantitative immunocytochemistry | 62 HCC patients, seven HBV-infected patients, and 15 healthy individuals | The cells obtained from the blood of HCC patients had significantly higher levels of ASGPR, GPC3, and CK than cells derived from chronic HBV-infected patients or healthy controls; ASGPR, GPC3, and CK might be valuable as HCC biomarkers for CTC detection; the expression of ASGPR and GPC3 might be helpful for understanding OS of the patients. | [32] |
Hep Par 1, GPC3, GS | Label-free Labyrinth technology, and immunoaffinity-based CTC-Chip (Microfluidic chip) | 42 HCC patients, four non-HCC patients | The HCC CTC detection rate was improved by using three HCC markers compared to EpCAM-based identification method. | [33] |
ASGPR, Hep Par 1 | Magnetic separation and immunoidentification | 85 HCC patients, 37 patients with benign liver diseases, 20 healthy volunteers, and 14 patients with other advanced cancers | No healthy, benign liver disease, or non-HCC cancer subjects were detected with CTCs. CTCs were identified in 69 of 85 HCC patients. | [34] |
ASGPR, CPS1 | Density gradient Ficoll-Paque PLUS, magnetic labeling, and separation | 32 HCC patients, 17 patients with other types of cancer, 40 patients with other liver diseases, and 20 healthy volunteers | CTCs that tested positive for ASGPR and CPS1 were detected in 91% of patients with HCC, and there were no CTCs detected in healthy volunteers and in patients with any other kinds of cancers, including breast, lung, esophageal, gastric, and colorectal cancer. | [35] |
CK, EpCAM, EMA, CK18, AFP, GPC-3, and Hep Par 1 | BenchMark XT Slide Preparation system | 23 HCC patients, six patients with non-HCC | 57.1% of patients tested positive for EpCAM, 42.9% for EMA, and 21.4% for AFP. | [36] |
How do CTCs respond in tumor microenvironment? | ||||
phosphorylated ERK (pERK) and pAkt CTC | Density gradient centrifugation, magnetic separation | 109 HCC patients | Phosphorylated ERK (pERK) and pAkt expressions in CTCs were correlated to sorafenib efficacy in HCC patients; pERK+/pAkt− CTCs were mostly responsive to sorafenib; the population of pERK+/pAkt− CTCs could be a potential predictive factor for HCC patients treated with sorafenib. | [37] |
CD4+CD25+Foxp3+ Treg cells | PCR and fluorescence-activated cell sorting | 49 HCC patients | The early recurrence rate in the group with combined higher EpCAM+ CTCs and Treg/CD4+ population was significantly higher than in the combined lower CTCs and Treg group; the combined detection of EpCAM+ CTCs and Treg/CD4+ might provide a novel prognostic predictor for HCC patients. | [38] |
IGFBP1 | Density gradient centrifugation, and immunomagnetic beads | 25 HCC patients | IGFBP1 was correlated with the responsiveness to selective internal radiation therapy. | [39] |
Are CTCs equivalent to CSCs (Cancer Stem Cells)? | ||||
EpCAM, CD133 | CellSearch system | 123 HCC patients | CSC biomarkers CD133 and ABCG2 were observed in the blood samples of HCC patients with positive EpCAM+ CTCs. | [40] |
GPC3, GS, Hep Par 1, and CD44 | Label-free Labyrinth technology, and immunoaffinity-based CTC-Chip | 37 HCC patients | CTCs with the expression of CD44 were observed in all the stages of HCC; CTCs with these three markers, GPC3, GS, and Hep Par 1 had a cancer stemness phenotype. | [33] |
EpCAM, CD133, CD90, CK19, ABCG2, CD44, ICAM1, CD24, and Nestin | qRT-PCR | 956 HCC patients and 50 healthy donors | Compared with EpCAM, the prognostic significance of CTC panel (EpCAM, CD90, CD133, and CK19) was still retained in the EpCAM− subgroup. | [41] |
CD133, ANXA3 | Enzyme-linked immunosorbent assay | 368 HCC patients | Serum ANXA3 could stimulate and maintain the stem cell-like traits of CD133 CTCs to promote tumor recurrence and metastasis; combining ANXA3 with AFP significantly improved the outcome prediction. | [42] |
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Chen, F.; Zhong, Z.; Tan, H.-Y.; Wang, N.; Feng, Y. The Significance of Circulating Tumor Cells in Patients with Hepatocellular Carcinoma: Real-Time Monitoring and Moving Targets for Cancer Therapy. Cancers 2020, 12, 1734. https://doi.org/10.3390/cancers12071734
Chen F, Zhong Z, Tan H-Y, Wang N, Feng Y. The Significance of Circulating Tumor Cells in Patients with Hepatocellular Carcinoma: Real-Time Monitoring and Moving Targets for Cancer Therapy. Cancers. 2020; 12(7):1734. https://doi.org/10.3390/cancers12071734
Chicago/Turabian StyleChen, Feiyu, Zhangfeng Zhong, Hor-Yue Tan, Ning Wang, and Yibin Feng. 2020. "The Significance of Circulating Tumor Cells in Patients with Hepatocellular Carcinoma: Real-Time Monitoring and Moving Targets for Cancer Therapy" Cancers 12, no. 7: 1734. https://doi.org/10.3390/cancers12071734
APA StyleChen, F., Zhong, Z., Tan, H. -Y., Wang, N., & Feng, Y. (2020). The Significance of Circulating Tumor Cells in Patients with Hepatocellular Carcinoma: Real-Time Monitoring and Moving Targets for Cancer Therapy. Cancers, 12(7), 1734. https://doi.org/10.3390/cancers12071734