Circulating Tumor Cells from Enumeration to Analysis: Current Challenges and Future Opportunities
Abstract
:Simple Summary
Abstract
1. Introduction
2. CTC Isolation and Detection Techniques
2.1. Isolation Based on Physical Properties
2.1.1. Density-Based Separation
2.1.2. Filtration-Based Separation
2.1.3. Contactless Separation
2.2. Isolation Based on Biological Properties
2.3. Isolation Based on a Combination of Physical and Biological Properties
2.4. In Vivo CTC Detection
3. Downstream Analysis of Circulating Tumor Cells
3.1. Single-Cell Molecular Analysis of CTCs
3.2. Ex Vivo Expansion of CTCs
3.3. Role of CTC-Associated Cells
4. Current Challenges and Future Direction for Clinical Utility
5. Conclusions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Lambert, A.W.; Pattabiraman, D.R.; Weinberg, R.A. Emerging Biological Principles of Metastasis. Cell 2017, 168, 670–691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Micalizzi, D.S.; Maheswaran, S.; Haber, D.A. A conduit to metastasis: Circulating tumor cell biology. Genes Dev. 2017, 31, 1827–1840. [Google Scholar] [CrossRef]
- Azevedo, A.S.; Follain, G.; Patthabhiraman, S.; Harlepp, S.; Goetz, J.G. Metastasis of circulating tumor cells: Favorable soil or suitable biomechanics, or both? Cell Adhes. Migr. 2015, 9, 345–356. [Google Scholar] [CrossRef]
- Cheung, K.J.; Ewald, A.J. A collective route to metastasis: Seeding by tumor cell clusters. Science 2016, 352, 167–169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alix-Panabieres, C.; Pantel, K. Challenges in circulating tumour cell research. Nat. Rev. Cancer 2014, 14, 623–631. [Google Scholar] [CrossRef] [PubMed]
- Ignatiadis, M.; Lee, M.; Jeffrey, S.S. Circulating Tumor Cells and Circulating Tumor DNA: Challenges and Opportunities on the Path to Clinical Utility. Clin. Cancer Res. 2015, 21, 4786–4800. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rawal, S.; Yang, Y.P.; Cote, R.; Agarwal, A. Identification and Quantitation of Circulating Tumor Cells. Annu. Rev. Anal. Chem. 2017, 10, 321–343. [Google Scholar] [CrossRef] [PubMed]
- Krebs, M.G.; Hou, J.M.; Ward, T.H.; Blackhall, F.H.; Dive, C. Circulating tumour cells: Their utility in cancer management and predicting outcomes. Ther. Adv. Med. Oncol. 2010, 2, 351–365. [Google Scholar] [CrossRef] [Green Version]
- Lin, E.; Cao, T.; Nagrath, S.; King, M.R. Circulating Tumor Cells: Diagnostic and Therapeutic Applications. Annu. Rev. Biomed. Eng. 2018, 20, 329–352. [Google Scholar] [CrossRef] [PubMed]
- Vaidyanathan, R.; Soon, R.H.; Zhang, P.; Jiang, K.; Lim, C.T. Cancer diagnosis: From tumor to liquid biopsy and beyond. Lab Chip 2018. [Google Scholar] [CrossRef]
- Heitzer, E.; Auer, M.; Ulz, P.; Geigl, J.B.; Speicher, M.R. Circulating tumor cells and DNA as liquid biopsies. Genome Med. 2013, 5, 73. [Google Scholar] [CrossRef] [PubMed]
- Seal, S.H. Silicone flotation: A simple quantitative method for the isolation of free-floating cancer cells from the blood. Cancer 1959, 12, 590–595. [Google Scholar] [CrossRef]
- Weitz, J.; Kienle, P.; Lacroix, J.; Willeke, F.; Benner, A.; Lehnert, T.; Herfarth, C.; von Knebel Doeberitz, M. Dissemination of tumor cells in patients undergoing surgery for colorectal cancer. Clin. Cancer Res. 1998, 4, 343–348. [Google Scholar]
- Chalfin, H.J.; Glavaris, S.A.; Malihi, P.D.; Sperger, J.M.; Gorin, M.A.; Lu, C.; Goodwin, C.R.; Chen, Y.; Caruso, E.A.; Dumpit, R.; et al. Prostate Cancer Disseminated Tumor Cells are Rarely Detected in the Bone Marrow of Patients with Localized Disease Undergoing Radical Prostatectomy across Multiple Rare Cell Detection Platforms. J. Urol. 2018, 199, 1494–1501. [Google Scholar] [CrossRef] [PubMed]
- Campton, D.E.; Ramirez, A.B.; Nordberg, J.J.; Drovetto, N.; Clein, A.C.; Varshavskaya, P.; Friemel, B.H.; Quarre, S.; Breman, A.; Dorschner, M.; et al. High-recovery visual identification and single-cell retrieval of circulating tumor cells for genomic analysis using a dual-technology platform integrated with automated immunofluorescence staining. BMC Cancer 2015, 15, 360. [Google Scholar] [CrossRef] [Green Version]
- Rosenberg, R.; Gertler, R.; Friederichs, J.; Fuehrer, K.; Dahm, M.; Phelps, R.; Thorban, S.; Nekarda, H.; Siewert, J.R. Comparison of two density gradient centrifugation systems for the enrichment of disseminated tumor cells in blood. Cytometry 2002, 49, 150–158. [Google Scholar] [CrossRef]
- He, W.; Kularatne, S.A.; Kalli, K.R.; Prendergast, F.G.; Amato, R.J.; Klee, G.G.; Hartmann, L.C.; Low, P.S. Quantitation of circulating tumor cells in blood samples from ovarian and prostate cancer patients using tumor-specific fluorescent ligands. Int. J. Cancer 2008, 123, 1968–1973. [Google Scholar] [CrossRef] [Green Version]
- Wu, F.; Zhu, J.; Mao, Y.; Li, X.; Hu, B.; Zhang, D. Associations between the Epithelial-Mesenchymal Transition Phenotypes of Circulating Tumor Cells and the Clinicopathological Features of Patients with Colorectal Cancer. Dis. Markers 2017, 2017, 9474532. [Google Scholar] [CrossRef] [Green Version]
- Cohen, S.J.; Punt, C.J.; Iannotti, N.; Saidman, B.H.; Sabbath, K.D.; Gabrail, N.Y.; Picus, J.; Morse, M.; Mitchell, E.; Miller, M.C.; 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, 3213–3221. [Google Scholar] [CrossRef] [PubMed]
- Vona, G.; Sabile, A.; Louha, M.; Sitruk, V.; Romana, S.; Schutze, K.; Capron, F.; Franco, D.; Pazzagli, M.; Vekemans, M.; et al. Isolation by size of epithelial tumor cells: A new method for the immunomorphological and molecular characterization of circulatingtumor cells. Am. J. Pathol. 2000, 156, 57–63. [Google Scholar] [CrossRef]
- Desitter, I.; Guerrouahen, B.S.; Benali-Furet, N.; Wechsler, J.; Janne, P.A.; Kuang, Y.; Yanagita, M.; Wang, L.; Berkowitz, J.A.; Distel, R.J.; et al. A new device for rapid isolation by size and characterization of rare circulating tumor cells. Anticancer Res. 2011, 31, 427–441. [Google Scholar]
- Lin, H.K.; Zheng, S.; Williams, A.J.; Balic, M.; Groshen, S.; Scher, H.I.; Fleisher, M.; Stadler, W.; Datar, R.H.; Tai, Y.C.; 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]
- Zheng, S.; Lin, H.; Liu, J.Q.; Balic, M.; Datar, R.; Cote, R.J.; Tai, Y.C. Membrane microfilter device for selective capture, electrolysis and genomic analysis of human circulating tumor cells. J. Chromatogr. A 2007, 1162, 154–161. [Google Scholar] [CrossRef] [PubMed]
- Hvichia, G.E.; Parveen, Z.; Wagner, C.; Janning, M.; Quidde, J.; Stein, A.; Muller, V.; Loges, S.; Neves, R.P.; Stoecklein, N.H.; et al. A novel microfluidic platform for size and deformability based separation and the subsequent molecular characterization of viable circulating tumor cells. Int. J. Cancer 2016, 138, 2894–2904. [Google Scholar] [CrossRef] [PubMed]
- Xu, L.; Mao, X.; Imrali, A.; Syed, F.; Mutsvangwa, K.; Berney, D.; Cathcart, P.; Hines, J.; Shamash, J.; Lu, Y.J. Optimization and Evaluation of a Novel Size Based Circulating Tumor Cell Isolation System. PLoS ONE 2015, 10, e0138032. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sollier, E.; Go, D.E.; Che, J.; Gossett, D.R.; O’Byrne, S.; Weaver, W.M.; Kummer, N.; Rettig, M.; Goldman, J.; Nickols, N.; et al. Size-selective collection of circulating tumor cells using Vortex technology. Lab Chip 2014, 14, 63–77. [Google Scholar] [CrossRef]
- Hou, H.W.; Warkiani, M.E.; Khoo, B.L.; Li, Z.R.; Soo, R.A.; Tan, D.S.; Lim, W.T.; Han, J.; Bhagat, A.A.; Lim, C.T. Isolation and retrieval of circulating tumor cells using centrifugal forces. Sci. Rep. 2013, 3, 1259. [Google Scholar] [CrossRef] [Green Version]
- Gupta, V.; Jafferji, I.; Garza, M.; Melnikova, V.O.; Hasegawa, D.K.; Pethig, R.; Davis, D.W. ApoStream(TM), a new dielectrophoretic device for antibody independent isolation and recovery of viable cancer cells from blood. Biomicrofluidics 2012, 6, 24133. [Google Scholar] [CrossRef] [Green Version]
- 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] [PubMed] [Green Version]
- Tashireva, L.A.; Savelieva, O.E.; Grigoryeva, E.S.; Nikitin, Y.V.; Denisov, E.V.; Vtorushin, S.V.; Zavyalova, M.V.; Cherdyntseva, N.V.; Perelmuter, V.M. Heterogeneous Manifestations of Epithelial-Mesenchymal Plasticity of Circulating Tumor Cells in Breast Cancer Patients. Int. J. Mol. Sci. 2021, 22, 2504. [Google Scholar] [CrossRef]
- Earl, J.; Garcia-Nieto, S.; Martinez-Avila, J.C.; Montans, J.; Sanjuanbenito, A.; Rodriguez-Garrote, M.; Lisa, E.; Mendia, E.; Lobo, E.; Malats, N.; et al. Circulating tumor cells (Ctc) and kras mutant circulating free Dna (cfdna) detection in peripheral blood as biomarkers in patients diagnosed with exocrine pancreatic cancer. BMC Cancer 2015, 15, 797. [Google Scholar] [CrossRef] [Green Version]
- Hu, C.L.; Zhang, Y.J.; Zhang, X.F.; Fei, X.; Zhang, H.; Li, C.G.; Sun, B. 3D Culture of Circulating Tumor Cells for Evaluating Early Recurrence and Metastasis in Patients with Hepatocellular Carcinoma. Onco. Targets Ther. 2021, 14, 2673–2688. [Google Scholar] [CrossRef]
- Takahashi, K.; Ofuji, K.; Hiramatsu, K.; Nosaka, T.; Naito, T.; Matsuda, H.; Endo, K.; Higuchi, M.; Ohtani, M.; Nemoto, T.; et al. Circulating tumor cells detected with a microcavity array predict clinical outcome in hepatocellular carcinoma. Cancer Med. 2021, 10, 2300–2309. [Google Scholar] [CrossRef] [PubMed]
- Lin, S.Y.; Chang, S.C.; Lam, S.; Irene Ramos, R.; Tran, K.; Ohe, S.; Salomon, M.P.; Bhagat, A.A.S.; Teck Lim, C.; Fischer, T.D.; et al. Prospective Molecular Profiling of Circulating Tumor Cells from Patients with Melanoma Receiving Combinatorial Immunotherapy. Clin. Chem. 2020, 66, 169–177. [Google Scholar] [CrossRef]
- Wang, P.P.; Liu, S.H.; Chen, C.T.; Lv, L.; Li, D.; Liu, Q.Y.; Liu, G.L.; Wu, Y. Circulating tumor cells as a new predictive and prognostic factor in patients with small cell lung cancer. J. Cancer 2020, 11, 2113–2122. [Google Scholar] [CrossRef]
- Klameth, L.; Rath, B.; Hochmaier, M.; Moser, D.; Redl, M.; Mungenast, F.; Gelles, K.; Ulsperger, E.; Zeillinger, R.; Hamilton, G. Small cell lung cancer: Model of circulating tumor cell tumorospheres in chemoresistance. Sci. Rep. 2017, 7, 5337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hsu, C.L.; Tsai, T.H.; Huang, C.K.; Yang, C.Y.; Liao, W.Y.; Ho, C.C.; Ruan, S.Y.; Chen, K.Y.; Shih, J.Y.; Yang, P.C. Monitoring levels of vimentin-positive circulating cancer stem cells and tumor cells in patients with advanced EGFR-mutated non-small cell lung cancer. Lung Cancer 2021, 156, 50–58. [Google Scholar] [CrossRef]
- Lindsay, C.R.; Blackhall, F.H.; Carmel, A.; Fernandez-Gutierrez, F.; Gazzaniga, P.; Groen, H.J.M.; Hiltermann, T.J.N.; Krebs, M.G.; Loges, S.; Lopez-Lopez, R.; et al. EPAC-lung: Pooled analysis of circulating tumour cells in advanced non-small cell lung cancer. Eur. J. Cancer 2019, 117, 60–68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rink, M.; Riethdorf, S.; Yu, H.; Kolker, M.; Vetterlein, M.W.; Dahlem, R.; Fisch, M.; Pantel, K.; Soave, A. The Impact of Circulating Tumor Cells on Venous Thromboembolism and Cardiovascular Events in Bladder Cancer Patients Treated with Radical Cystectomy. J. Clin. Med. 2020, 9, 3478. [Google Scholar] [CrossRef]
- Bergmann, S.; Coym, A.; Ott, L.; Soave, A.; Rink, M.; Janning, M.; Stoupiec, M.; Coith, C.; Peine, S.; von Amsberg, G.; et al. Evaluation of PD-L1 expression on circulating tumor cells (CTCs) in patients with advanced urothelial carcinoma (UC). Oncoimmunology 2020, 9, 1738798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guadagni, S.; Clementi, M.; Masedu, F.; Fiorentini, G.; Sarti, D.; Deraco, M.; Kusamura, S.; Papasotiriou, I.; Apostolou, P.; Aigner, K.R.; et al. A Pilot Study of the Predictive Potential of Chemosensitivity and Gene Expression Assays Using Circulating Tumour Cells from Patients with Recurrent Ovarian Cancer. Int. J. Mol. Sci. 2020, 21, 4813. [Google Scholar] [CrossRef] [PubMed]
- Banys-Paluchowski, M.; Fehm, T.; Neubauer, H.; Paluchowski, P.; Krawczyk, N.; Meier-Stiegen, F.; Wallach, C.; Kaczerowsky, A.; Gebauer, G. Clinical relevance of circulating tumor cells in ovarian, fallopian tube and peritoneal cancer. Arch. Gynecol. Obstet. 2020, 301, 1027–1035. [Google Scholar] [CrossRef] [Green Version]
- Lee, H.J.; Kim, G.H.; Park, S.J.; Kwon, C.H.; Lee, M.W.; Lee, B.E.; Baek, D.H.; Hoseok, I. Clinical Significance of TWIST-Positive Circulating Tumor Cells in Patients with Esophageal Squamous Cell Carcinoma. Gut Liver 2020. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed] [Green Version]
- Muller, C.; Holtschmidt, J.; Auer, M.; Heitzer, E.; Lamszus, K.; Schulte, A.; Matschke, J.; Langer-Freitag, S.; Gasch, C.; Stoupiec, M.; et al. Hematogenous dissemination of glioblastoma multiforme. Sci. Transl. Med. 2014, 6, 247ra101. [Google Scholar] [CrossRef]
- Pluim, D.; Devriese, L.A.; Beijnen, J.H.; Schellens, J.H. Validation of a multiparameter flow cytometry method for the determination of phosphorylated extracellular-signal-regulated kinase and DNA in circulating tumor cells. Cytometry A 2012, 81, 664–671. [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]
- Seal, S.H. A Sieve for the Isolation of Cancer Cells and Other Large Cells from the Blood. Cancer 1964, 17, 637–642. [Google Scholar] [CrossRef]
- Apel, P. Track etching technique in membrane technology. Radiat. Meas. 2001, 34, 559–566. [Google Scholar] [CrossRef]
- Xu, T.; Lu, B.; Tai, Y.C.; Goldkorn, A. A cancer detection platform which measures telomerase activity from live circulating tumor cells captured on a microfilter. Cancer Res. 2010, 70, 6420–6426. [Google Scholar] [CrossRef] [Green Version]
- Ao, Z.; Parasido, E.; Rawal, S.; Williams, A.; Schlegel, R.; Liu, S.; Albanese, C.; Cote, R.J.; Agarwal, A.; Datar, R.H. Thermoresponsive release of viable microfiltrated Circulating Tumor Cells (CTCs) for precision medicine applications. Lab Chip 2015, 15, 4277–4282. [Google Scholar] [CrossRef] [Green Version]
- Miller, M.C.; Robinson, P.S.; Wagner, C.; O’Shannessy, D.J. The Parsortix Cell Separation System-A versatile liquid biopsy platform. Cytometry A 2018. [Google Scholar] [CrossRef]
- Becker, F.F.; Wang, X.B.; Huang, Y.; Pethig, R.; Vykoukal, J.; Gascoyne, P.R. Separation of human breast cancer cells from blood by differential dielectric affinity. Proc. Natl. Acad. Sci. USA 1995, 92, 860–864. [Google Scholar] [CrossRef] [Green Version]
- Manaresi, N.; Romani, A.; Medoro, G.; Altomare, L.; Leonardi, A.; Tartagni, M.; Guerrieri, R. A CMOS chip for individual cell manipulation and detection. IEEE J. Solid State Circuits 2003, 38, 2297–2305. [Google Scholar] [CrossRef]
- Dabighi, A.; Toghraie, D. A new microfluidic device for separating circulating tumor cells based on their physical properties by using electrophoresis and dielectrophoresis forces within an electrical field. Comput. Methods Programs Biomed. 2020, 185, 105147. [Google Scholar] [CrossRef] [PubMed]
- Budd, G.T.; Cristofanilli, M.; Ellis, M.J.; Stopeck, A.; Borden, E.; Miller, M.C.; Matera, J.; Repollet, M.; Doyle, G.V.; Terstappen, L.W.; et al. Circulating tumor cells versus imaging--predicting overall survival in metastatic breast cancer. Clin. Cancer Res. 2006, 12, 6403–6409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shaffer, D.R.; Leversha, M.A.; Danila, D.C.; Lin, O.; Gonzalez-Espinoza, R.; Gu, B.; Anand, A.; Smith, K.; Maslak, P.; Doyle, G.V.; et al. Circulating tumor cell analysis in patients with progressive castration-resistant prostate cancer. Clin. Cancer Res. 2007, 13, 2023–2029. [Google Scholar] [CrossRef] [Green Version]
- Scher, H.I.; Jia, X.; de Bono, J.S.; Fleisher, M.; Pienta, K.J.; Raghavan, D.; Heller, G. Circulating tumour cells as prognostic markers in progressive, castration-resistant prostate cancer: A reanalysis of IMMC38 trial data. Lancet Oncol. 2009, 10, 233–239. [Google Scholar] [CrossRef] [Green Version]
- Scher, H.I.; Armstrong, A.J.; Schonhoft, J.D.; Gill, A.; Zhao, J.L.; Barnett, E.; Carbone, E.; Lu, J.; Antonarakis, E.S.; Luo, J.; et al. Development and validation of circulating tumour cell enumeration (Epic Sciences) as a prognostic biomarker in men with metastatic castration-resistant prostate cancer. Eur. J. Cancer 2021, 150, 83–94. [Google Scholar] [CrossRef]
- Bidard, F.C.; Huguet, F.; Louvet, C.; Mineur, L.; Bouche, O.; Chibaudel, B.; Artru, P.; Desseigne, F.; Bachet, J.B.; Mathiot, C.; et al. Circulating tumor cells in locally advanced pancreatic adenocarcinoma: The ancillary CirCe 07 study to the LAP 07 trial. Ann. Oncol. 2013, 24, 2057–2061. [Google Scholar] [CrossRef]
- Tsai, W.S.; You, J.F.; Hung, H.Y.; Hsieh, P.S.; Hsieh, B.; Lenz, H.J.; Idos, G.; Friedland, S.; Yi-Jiun Pan, J.; Shao, H.J.; et al. Novel Circulating Tumor Cell Assay for Detection of Colorectal Adenomas and Cancer. Clin. Transl. Gastroenterol. 2019, 10, e00088. [Google Scholar] [CrossRef] [PubMed]
- Sullivan, J.P.; Nahed, B.V.; Madden, M.W.; Oliveira, S.M.; Springer, S.; Bhere, D.; Chi, A.S.; Wakimoto, H.; Rothenberg, S.M.; Sequist, L.V.; et al. Brain tumor cells in circulation are enriched for mesenchymal gene expression. Cancer Discov. 2014, 4, 1299–1309. [Google Scholar] [CrossRef] [Green Version]
- Redding, W.H.; Coombes, R.C.; Monaghan, P.; Clink, H.M.; Imrie, S.F.; Dearnaley, D.P.; Ormerod, M.G.; Sloane, J.P.; Gazet, J.C.; Powles, T.J.; et al. Detection of micrometastases in patients with primary breast cancer. Lancet 1983, 2, 1271–1274. [Google Scholar] [CrossRef]
- Andreopoulou, E.; Yang, L.Y.; Rangel, K.M.; Reuben, J.M.; Hsu, L.; Krishnamurthy, S.; Valero, V.; Fritsche, H.A.; Cristofanilli, M. Comparison of assay methods for detection of circulating tumor cells in metastatic breast cancer: AdnaGen AdnaTest BreastCancer Select/Detect versus Veridex CellSearch system. Int. J. Cancer 2012, 130, 1590–1597. [Google Scholar] [CrossRef]
- Lopez-Munoz, E.; Mendez-Montes, M. Markers of circulating breast cancer cells. Adv. Clin. Chem. 2013, 61, 175–224. [Google Scholar]
- Giordano, A.; Gao, H.; Anfossi, S.; Cohen, E.; Mego, M.; Lee, B.N.; Tin, S.; De Laurentiis, M.; Parker, C.A.; Alvarez, R.H.; et al. Epithelial-mesenchymal transition and stem cell markers in patients with HER2-positive metastatic breast cancer. Mol. Cancer Ther. 2012, 11, 2526–2534. [Google Scholar] [CrossRef] [Green Version]
- Talasaz, A.H.; Powell, A.A.; Huber, D.E.; Berbee, J.G.; Roh, K.H.; Yu, W.; Xiao, W.; Davis, M.M.; Pease, R.F.; Mindrinos, M.N.; et al. Isolating highly enriched populations of circulating epithelial cells and other rare cells from blood using a magnetic sweeper device. Proc. Natl. Acad. Sci. USA 2009, 106, 3970–3975. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Karabacak, N.M.; Spuhler, P.S.; Fachin, F.; Lim, E.J.; Pai, V.; Ozkumur, E.; Martel, J.M.; Kojic, N.; Smith, K.; Chen, P.I.; et al. Microfluidic, marker-free isolation of circulating tumor cells from blood samples. Nat. Protoc. 2014, 9, 694–710. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gleghorn, J.P.; Pratt, E.D.; Denning, D.; Liu, H.; Bander, N.H.; Tagawa, S.T.; Nanus, D.M.; Giannakakou, P.A.; Kirby, B.J. Capture of circulating tumor cells from whole blood of prostate cancer patients using geometrically enhanced differential immunocapture (GEDI) and a prostate-specific antibody. Lab Chip 2010, 10, 27–29. [Google Scholar] [CrossRef]
- Nora Dickson, M.; Tsinberg, P.; Tang, Z.; Bischoff, F.Z.; Wilson, T.; Leonard, E.F. Efficient capture of circulating tumor cells with a novel immunocytochemical microfluidic device. Biomicrofluidics 2011, 5, 034119. [Google Scholar] [CrossRef] [Green Version]
- Harb, W.; Fan, A.; Tran, T.; Danila, D.C.; Keys, D.; Schwartz, M.; Ionescu-Zanetti, C. Mutational Analysis of Circulating Tumor Cells Using a Novel Microfluidic Collection Device and qPCR Assay. Transl. Oncol. 2013, 6, 528–538. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Winer-Jones, J.P.; Vahidi, B.; Arquilevich, N.; Fang, C.; Ferguson, S.; Harkins, D.; Hill, C.; Klem, E.; Pagano, P.C.; Peasley, C.; et al. Circulating tumor cells: Clinically relevant molecular access based on a novel CTC flow cell. PLoS ONE 2014, 9, e86717. [Google Scholar] [CrossRef] [PubMed]
- Stott, S.L.; Hsu, C.H.; Tsukrov, D.I.; Yu, M.; Miyamoto, D.T.; Waltman, B.A.; Rothenberg, S.M.; Shah, A.M.; Smas, M.E.; Korir, G.K.; et al. Isolation of circulating tumor cells using a microvortex-generating herringbone-chip. Proc. Natl. Acad. Sci. USA 2010, 107, 18392–18397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sheng, W.; Ogunwobi, O.O.; Chen, T.; Zhang, J.; George, T.J.; Liu, C.; Fan, Z.H. Capture, release and culture of circulating tumor cells from pancreatic cancer patients using an enhanced mixing chip. Lab Chip 2014, 14, 89–98. [Google Scholar] [CrossRef] [Green Version]
- Yoon, H.J.; Kim, T.H.; Zhang, Z.; Azizi, E.; Pham, T.M.; Paoletti, C.; Lin, J.; Ramnath, N.; Wicha, M.S.; Hayes, D.F.; et al. Sensitive capture of circulating tumour cells by functionalized graphene oxide nanosheets. Nat. Nanotechnol. 2013, 8, 735–741. [Google Scholar] [CrossRef]
- Kamande, J.W.; Hupert, M.L.; Witek, M.A.; Wang, H.; Torphy, R.J.; Dharmasiri, U.; Njoroge, S.K.; Jackson, J.M.; Aufforth, R.D.; Snavely, A.; et al. Modular microsystem for the isolation, enumeration, and phenotyping of circulating tumor cells in patients with pancreatic cancer. Anal. Chem. 2013, 85, 9092–9100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lu, Y.T.; Zhao, L.; Shen, Q.; Garcia, M.A.; Wu, D.; Hou, S.; Song, M.; Xu, X.; Ouyang, W.H.; Ouyang, W.W.; et al. NanoVelcro Chip for CTC enumeration in prostate cancer patients. Methods 2013, 64, 144–152. [Google Scholar] [CrossRef] [Green Version]
- Wang, S.; Liu, K.; Liu, J.; Yu, Z.T.; Xu, X.; Zhao, L.; Lee, T.; Lee, E.K.; Reiss, J.; Lee, Y.K.; et al. Highly efficient capture of circulating tumor cells by using nanostructured silicon substrates with integrated chaotic micromixers. Angew. Chem. Int. Ed. 2011, 50, 3084–3088. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lustberg, M.B.; Balasubramanian, P.; Miller, B.; Garcia-Villa, A.; Deighan, C.; Wu, Y.; Carothers, S.; Berger, M.; Ramaswamy, B.; Macrae, E.R.; et al. Heterogeneous atypical cell populations are present in blood of metastatic breast cancer patients. Breast Cancer Res. 2014, 16, R23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, Z.; Fusi, A.; Klopocki, E.; Schmittel, A.; Tinhofer, I.; Nonnenmacher, A.; Keilholz, U. Negative enrichment by immunomagnetic nanobeads for unbiased characterization of circulating tumor cells from peripheral blood of cancer patients. J. Transl. Med. 2011, 9, 70. [Google Scholar] [CrossRef] [Green Version]
- Lara, O.; Tong, X.; Zborowski, M.; Chalmers, J.J. Enrichment of rare cancer cells through depletion of normal cells using density and flow-through, immunomagnetic cell separation. Exp. Hematol. 2004, 32, 891–904. [Google Scholar] [CrossRef] [PubMed]
- Poudineh, M.; Aldridge, P.M.; Ahmed, S.; Green, B.J.; Kermanshah, L.; Nguyen, V.; Tu, C.; Mohamadi, R.M.; Nam, R.K.; Hansen, A.; et al. Tracking the dynamics of circulating tumour cell phenotypes using nanoparticle-mediated magnetic ranking. Nat. Nanotechnol. 2017, 12, 274–281. [Google Scholar] [CrossRef]
- Sinkala, E.; Sollier-Christen, E.; Renier, C.; Rosas-Canyelles, E.; Che, J.; Heirich, K.; Duncombe, T.A.; Vlassakis, J.; Yamauchi, K.A.; Huang, H.; et al. Profiling protein expression in circulating tumour cells using microfluidic western blotting. Nat. Commun. 2017, 8, 14622. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huang, Q.; Wang, Y.; Chen, X.; Wang, Y.; Li, Z.; Du, S.; Wang, L.; Chen, S. Nanotechnology-Based Strategies for Early Cancer Diagnosis Using Circulating Tumor Cells as a Liquid Biopsy. Nanotheranostics 2018, 2, 21–41. [Google Scholar] [CrossRef] [Green Version]
- Li, W.; Wang, H.; Zhao, Z.; Gao, H.; Liu, C.; Zhu, L.; Wang, C.; Yang, Y. Emerging Nanotechnologies for Liquid Biopsy: The Detection of Circulating Tumor Cells and Extracellular Vesicles. Adv. Mater. 2019, 31, e1805344. [Google Scholar] [CrossRef]
- Sheng, W.; Chen, T.; Tan, W.; Fan, Z.H. Multivalent DNA nanospheres for enhanced capture of cancer cells in microfluidic devices. ACS Nano 2013, 7, 7067–7076. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhao, L.; Pang, X.; Adhikary, R.; Petrich, J.W.; Jeffries-El, M.; Lin, Z. Organic-inorganic nanocomposites by placing conjugated polymers in intimate contact with quantum rods. Adv. Mater. 2011, 23, 2844–2849. [Google Scholar] [CrossRef] [PubMed]
- Abdolahad, M.; Taghinejad, M.; Taghinejad, H.; Janmaleki, M.; Mohajerzadeh, S. A vertically aligned carbon nanotube-based impedance sensing biosensor for rapid and high sensitive detection of cancer cells. Lab. Chip 2012, 12, 1183–1190. [Google Scholar] [CrossRef] [PubMed]
- Zhao, L.; Lu, Y.T.; Li, F.; Wu, K.; Hou, S.; Yu, J.; Shen, Q.; Wu, D.; Song, M.; OuYang, W.H.; et al. High-purity prostate circulating tumor cell isolation by a polymer nanofiber-embedded microchip for whole exome sequencing. Adv. Mater. 2013, 25, 2897–2902. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Saucedo-Zeni, N.; Mewes, S.; Niestroj, R.; Gasiorowski, L.; Murawa, D.; Nowaczyk, P.; Tomasi, T.; Weber, E.; Dworacki, G.; Morgenthaler, N.G.; et al. A novel method for the in vivo isolation of circulating tumor cells from peripheral blood of cancer patients using a functionalized and structured medical wire. Int. J. Oncol. 2012, 41, 1241–1250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, T.H.; Wang, Y.; Oliver, C.R.; Thamm, D.H.; Cooling, L.; Paoletti, C.; Smith, K.J.; Nagrath, S.; Hayes, D.F. A temporary indwelling intravascular aphaeretic system for in vivo enrichment of circulating tumor cells. Nat. Commun. 2019, 10, 1478. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nolan, J.; Nedosekin, D.A.; Galanzha, E.I.; Zharov, V.P. Detection of Apoptotic Circulating Tumor Cells Using in vivo Fluorescence Flow Cytometry. Cytometry A 2019, 95, 664–671. [Google Scholar] [CrossRef] [PubMed]
- Galanzha, E.I.; Menyaev, Y.A.; Yadem, A.C.; Sarimollaoglu, M.; Juratli, M.A.; Nedosekin, D.A.; Foster, S.R.; Jamshidi-Parsian, A.; Siegel, E.R.; Makhoul, I.; et al. In vivo liquid biopsy using Cytophone platform for photoacoustic detection of circulating tumor cells in patients with melanoma. Sci. Transl. Med. 2019, 11. [Google Scholar] [CrossRef] [PubMed]
- Ilie, M.; Hofman, V.; Long-Mira, E.; Selva, E.; Vignaud, J.M.; Padovani, B.; Mouroux, J.; Marquette, C.H.; Hofman, P. “Sentinel” circulating tumor cells allow early diagnosis of lung cancer in patients with chronic obstructive pulmonary disease. PLoS ONE 2014, 9, e111597. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pierga, J.Y.; Bidard, F.C.; Autret, A.; Petit, T.; Andre, F.; Dalenc, F.; Levy, C.; Ferrero, J.M.; Romieu, G.; Bonneterre, J.; et al. Circulating tumour cells and pathological complete response: Independent prognostic factors in inflammatory breast cancer in a pooled analysis of two multicentre phase II trials (BEVERLY-1 and -2) of neoadjuvant chemotherapy combined with bevacizumab. Ann. Oncol. 2017, 28, 103–109. [Google Scholar] [CrossRef]
- Miyamoto, D.T.; Ting, D.T.; Toner, M.; Maheswaran, S.; Haber, D.A. Single-Cell Analysis of Circulating Tumor Cells as a Window into Tumor Heterogeneity. Cold Spring Harb. Symp. Quant. Biol. 2016, 81, 269–274. [Google Scholar] [CrossRef] [Green Version]
- Czyz, Z.T.; Hoffmann, M.; Schlimok, G.; Polzer, B.; Klein, C.A. Reliable single cell array CGH for clinical samples. PLoS ONE 2014, 9, e85907. [Google Scholar] [CrossRef]
- Babayan, A.; Alawi, M.; Gormley, M.; Muller, V.; Wikman, H.; McMullin, R.P.; Smirnov, D.A.; Li, W.; Geffken, M.; Pantel, K.; et al. Comparative study of whole genome amplification and next generation sequencing performance of single cancer cells. Oncotarget 2017, 8, 56066–56080. [Google Scholar] [CrossRef] [Green Version]
- Carter, L.; Rothwell, D.G.; Mesquita, B.; Smowton, C.; Leong, H.S.; Fernandez-Gutierrez, F.; Li, Y.; Burt, D.J.; Antonello, J.; Morrow, C.J.; et al. Molecular analysis of circulating tumor cells identifies distinct copy-number profiles in patients with chemosensitive and chemorefractory small-cell lung cancer. Nat. Med. 2017, 23, 114–119. [Google Scholar] [CrossRef]
- Gao, Y.; Ni, X.; Guo, H.; Su, Z.; Ba, Y.; Tong, Z.; Guo, Z.; Yao, X.; Chen, X.; Yin, J.; et al. Single-cell sequencing deciphers a convergent evolution of copy number alterations from primary to circulating tumor cells. Genome Res. 2017, 27, 1312–1322. [Google Scholar] [CrossRef]
- Zhu, S.; Qing, T.; Zheng, Y.; Jin, L.; Shi, L. Advances in single-cell RNA sequencing and its applications in cancer research. Oncotarget 2017, 8, 53763–53779. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Miyamoto, D.T.; Lee, R.J.; Stott, S.L.; Ting, D.T.; Wittner, B.S.; Ulman, M.; Smas, M.E.; Lord, J.B.; Brannigan, B.W.; Trautwein, J.; et al. Androgen receptor signaling in circulating tumor cells as a marker of hormonally responsive prostate cancer. Cancer Discov. 2012, 2, 995–1003. [Google Scholar] [CrossRef] [Green Version]
- Miyamoto, D.T.; Zheng, Y.; Wittner, B.S.; Lee, R.J.; Zhu, H.; Broderick, K.T.; Desai, R.; Fox, D.B.; Brannigan, B.W.; Trautwein, J.; et al. RNA-Seq of single prostate CTCs implicates noncanonical Wnt signaling in antiandrogen resistance. Science 2015, 349, 1351–1356. [Google Scholar] [CrossRef] [Green Version]
- Ting, D.T.; Wittner, B.S.; Ligorio, M.; Vincent Jordan, N.; Shah, A.M.; Miyamoto, D.T.; Aceto, N.; Bersani, F.; Brannigan, B.W.; Xega, K.; et al. Single-cell RNA sequencing identifies extracellular matrix gene expression by pancreatic circulating tumor cells. Cell Rep. 2014, 8, 1905–1918. [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] [Green Version]
- Patel, A.P.; Tirosh, I.; Trombetta, J.J.; Shalek, A.K.; Gillespie, S.M.; Wakimoto, H.; Cahill, D.P.; Nahed, B.V.; Curry, W.T.; Martuza, R.L.; et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science 2014, 344, 1396–1401. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tirosh, I.; Izar, B.; Prakadan, S.M.; Wadsworth, M.H., 2nd; Treacy, D.; Trombetta, J.J.; Rotem, A.; Rodman, C.; Lian, C.; Murphy, G.; et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 2016, 352, 189–196. [Google Scholar] [CrossRef] [Green Version]
- Reece, A.; Xia, B.; Jiang, Z.; Noren, B.; McBride, R.; Oakey, J. Microfluidic techniques for high throughput single cell analysis. Curr. Opin. Biotechnol. 2016, 40, 90–96. [Google Scholar] [CrossRef] [Green Version]
- Gao, D.; Jin, F.; Zhou, M.; Jiang, Y. Recent advances in single cell manipulation and biochemical analysis on microfluidics. Analyst 2019, 144, 766–781. [Google Scholar] [CrossRef] [PubMed]
- Rich-Griffin, C.; Stechemesser, A.; Finch, J.; Lucas, E.; Ott, S.; Schafer, P. Single-Cell Transcriptomics: A High-Resolution Avenue for Plant Functional Genomics. Trends Plant. Sci. 2020, 25, 186–197. [Google Scholar] [CrossRef] [PubMed]
- Luo, T.; Fan, L.; Zhu, R.; Sun, D. Microfluidic Single-Cell Manipulation and Analysis: Methods and Applications. Micromachines 2019, 10, 104. [Google Scholar] [CrossRef] [Green Version]
- Caen, O.; Lu, H.; Nizard, P.; Taly, V. Microfluidics as a Strategic Player to Decipher Single-Cell Omics? Trends Biotechnol. 2017, 35, 713–727. [Google Scholar] [CrossRef]
- Iyer, A.; Gupta, K.; Sharma, S.; Hari, K.; Lee, Y.F.; Ramalingam, N.; Yap, Y.S.; West, J.; Bhagat, A.A.; Subramani, B.V.; et al. Integrative Analysis and Machine Learning based Characterization of Single Circulating Tumor Cells. J. Clin. Med. 2020, 9, 1206. [Google Scholar] [CrossRef] [Green Version]
- D’Avola, D.; Villacorta-Martin, C.; Martins-Filho, S.N.; Craig, A.; Labgaa, I.; von Felden, J.; Kimaada, A.; Bonaccorso, A.; Tabrizian, P.; Hartmann, B.M.; et al. High-density single cell mRNA sequencing to characterize circulating tumor cells in hepatocellular carcinoma. Sci. Rep. 2018, 8, 11570. [Google Scholar] [CrossRef] [PubMed]
- Leong, S.M.; Tan, K.M.; Chua, H.W.; Huang, M.C.; Cheong, W.C.; Li, M.H.; Tucker, S.; Koay, E.S. Paper-Based MicroRNA Expression Profiling from Plasma and Circulating Tumor Cells. Clin. Chem. 2017, 63, 731–741. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pixberg, C.F.; Raba, K.; Muller, F.; Behrens, B.; Honisch, E.; Niederacher, D.; Neubauer, H.; Fehm, T.; Goering, W.; Schulz, W.A.; et al. Analysis of DNA methylation in single circulating tumor cells. Oncogene 2017, 36, 3223–3231. [Google Scholar] [CrossRef]
- Gerdtsson, E.; Pore, M.; Thiele, J.-A.; Gerdtsson, A.S.; Malihi, P.D.; Nevarez, R.; Kolatkar, A.; Velasco, C.R.; Wix, S.; Singh, M.; et al. Multiplex protein detection on circulating tumor cells from liquid biopsies using imaging mass cytometry. Converg. Sci. Phys. Oncol. 2018, 4, 015002. [Google Scholar] [CrossRef] [PubMed]
- Masters, J.R. Human cancer cell lines: Fact and fantasy. Nat. Rev. Mol. Cell Biol. 2000, 1, 233–236. [Google Scholar] [CrossRef]
- Alix-Panabieres, C. EPISPOT assay: Detection of viable DTCs/CTCs in solid tumor patients. Recent Results Cancer Res. 2012, 195, 69–76. [Google Scholar] [CrossRef]
- 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, 180ra148. [Google Scholar] [CrossRef] [Green Version]
- Yu, M.; Bardia, A.; Aceto, N.; Bersani, F.; Madden, M.W.; Donaldson, M.C.; Desai, R.; Zhu, H.; Comaills, V.; Zheng, Z.; et al. Cancer therapy. Ex vivo culture of circulating breast tumor cells for individualized testing of drug susceptibility. Science 2014, 345, 216–220. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Z.; Shiratsuchi, H.; Lin, J.; Chen, G.; Reddy, R.M.; Azizi, E.; Fouladdel, S.; Chang, A.C.; Lin, L.; Jiang, H.; et al. Expansion of CTCs from early stage lung cancer patients using a microfluidic co-culture model. Oncotarget 2014, 5, 12383–12397. [Google Scholar] [CrossRef] [Green Version]
- Clevers, H. Modeling Development and Disease with Organoids. Cell 2016, 165, 1586–1597. [Google Scholar] [CrossRef] [Green Version]
- Lancaster, M.A.; Knoblich, J.A. Organogenesis in a dish: Modeling development and disease using organoid technologies. Science 2014, 345, 1247125. [Google Scholar] [CrossRef] [PubMed]
- Dart, A. Organoid diversity. Nat. Rev. Cancer 2018, 18, 404–405. [Google Scholar] [CrossRef]
- Xu, H.; Lyu, X.; Yi, M.; Zhao, W.; Song, Y.; Wu, K. Organoid technology and applications in cancer research. J. Hematol. Oncol. 2018, 11, 116. [Google Scholar] [CrossRef]
- Weeber, F.; Ooft, S.N.; Dijkstra, K.K.; Voest, E.E. Tumor Organoids as a Pre-clinical Cancer Model for Drug Discovery. Cell Chem. Biol. 2017, 24, 1092–1100. [Google Scholar] [CrossRef]
- Gao, D.; Vela, I.; Sboner, A.; Iaquinta, P.J.; Karthaus, W.R.; Gopalan, A.; Dowling, C.; Wanjala, J.N.; Undvall, E.A.; Arora, V.K.; et al. Organoid cultures derived from patients with advanced prostate cancer. Cell 2014, 159, 176–187. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Chang, M.C.; Chang, Y.T.; Chen, J.Y.; Jeng, Y.M.; Yang, C.Y.; Tien, Y.W.; Yang, S.H.; Chen, H.L.; Liang, T.Y.; Wang, C.F.; et al. Clinical Significance of Circulating Tumor Microemboli as a Prognostic Marker in Patients with Pancreatic Ductal Adenocarcinoma. Clin. Chem 2016, 62, 505–513. [Google Scholar] [CrossRef] [Green Version]
- Au, S.H.; Storey, B.D.; Moore, J.C.; Tang, Q.; Chen, Y.L.; Javaid, S.; Sarioglu, A.F.; Sullivan, R.; Madden, M.W.; O’Keefe, R.; et al. Clusters of circulating tumor cells traverse capillary-sized vessels. Proc. Natl. Acad. Sci. USA 2016, 113, 4947–4952. [Google Scholar] [CrossRef] [Green Version]
- Voutsadakis, I.A. Thrombocytosis as a prognostic marker in gastrointestinal cancers. World J. Gastrointest. Oncol. 2014, 6, 34–40. [Google Scholar] [CrossRef]
- Gay, L.J.; Felding-Habermann, B. Contribution of platelets to tumour metastasis. Nat. Rev. Cancer 2011, 11, 123–134. [Google Scholar] [CrossRef]
- Li, J.; King, M.R. Adhesion receptors as therapeutic targets for circulating tumor cells. Front. Oncol. 2012, 2, 79. [Google Scholar] [CrossRef] [Green Version]
- Stegner, D.; Dutting, S.; Nieswandt, B. Mechanistic explanation for platelet contribution to cancer metastasis. Thromb. Res. 2014, 133 (Suppl. S2), S149–S157. [Google Scholar] [CrossRef]
- Jiang, X.; Wong, K.H.K.; Khankhel, A.H.; Zeinali, M.; Reategui, E.; Phillips, M.J.; Luo, X.; Aceto, N.; Fachin, F.; Hoang, A.N.; et al. Microfluidic isolation of platelet-covered circulating tumor cells. Lab. Chip 2017, 17, 3498–3503. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Ai, Y.; Wang, L.; Bu, P.; Sharkey, C.C.; Wu, Q.; Wun, B.; Roy, S.; Shen, X.; King, M.R. Targeted drug delivery to circulating tumor cells via platelet membrane-functionalized particles. Biomaterials 2016, 76, 52–65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, J.; Sharkey, C.C.; Wun, B.; Liesveld, J.L.; King, M.R. Genetic engineering of platelets to neutralize circulating tumor cells. J. Control. Release 2016, 228, 38–47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Noy, R.; Pollard, J.W. Tumor-associated macrophages: From mechanisms to therapy. Immunity 2014, 41, 49–61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Adams, D.L.; Martin, S.S.; Alpaugh, R.K.; Charpentier, M.; Tsai, S.; Bergan, R.C.; Ogden, I.M.; Catalona, W.; Chumsri, S.; Tang, C.M.; et al. Circulating giant macrophages as a potential biomarker of solid tumors. Proc. Natl. Acad. Sci. USA 2014, 111, 3514–3519. [Google Scholar] [CrossRef] [Green Version]
- Adams, D.L.; Adams, D.K.; Alpaugh, R.K.; Cristofanilli, M.; Martin, S.S.; Chumsri, S.; Tang, C.M.; Marks, J.R. Circulating Cancer-Associated Macrophage-Like Cells Differentiate Malignant Breast Cancer and Benign Breast Conditions. Cancer Epidemiol. Biomark. Prev. 2016, 25, 1037–1042. [Google Scholar] [CrossRef] [Green Version]
- Manjunath, Y.; Porciani, D.; Mitchem, J.B.; Suvilesh, K.N.; Avella, D.M.; Kimchi, E.T.; Staveley-O’Carroll, K.F.; Burke, D.H.; Li, G.; Kaifi, J.T. Tumor-Cell-Macrophage Fusion Cells as Liquid Biomarkers and Tumor Enhancers in Cancer. Int. J. Mol. Sci. 2020, 21, 1872. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Ohki, S.; Shibata, M.; Gonda, K.; Machida, T.; Shimura, T.; Nakamura, I.; Ohtake, T.; Koyama, Y.; Suzuki, S.; Ohto, H.; et al. Circulating myeloid-derived suppressor cells are increased and correlate to immune suppression, inflammation and hypoproteinemia in patients with cancer. Oncol. Rep. 2012, 28, 453–458. [Google Scholar] [CrossRef] [Green Version]
- Angell, T.E.; Lechner, M.G.; Smith, A.M.; Martin, S.E.; Groshen, S.G.; Maceri, D.R.; Singer, P.A.; Epstein, A.L. Circulating Myeloid-Derived Suppressor Cells Predict Differentiated Thyroid Cancer Diagnosis and Extent. Thyroid 2016, 26, 381–389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yamauchi, Y.; Safi, S.; Blattner, C.; Rathinasamy, A.; Umansky, L.; Juenger, S.; Warth, A.; Eichhorn, M.; Muley, T.; Herth, F.J.F.; et al. Circulating and Tumor Myeloid-derived Suppressor Cells in Resectable Non-Small Cell Lung Cancer. Am. J. Respir. Crit. Care Med. 2018, 198, 777–787. [Google Scholar] [CrossRef]
- Bussard, K.M.; Mutkus, L.; Stumpf, K.; Gomez-Manzano, C.; Marini, F.C. Tumor-associated stromal cells as key contributors to the tumor microenvironment. Breast Cancer Res. 2016, 18, 84. [Google Scholar] [CrossRef] [Green Version]
- Ao, Z.; Shah, S.H.; Machlin, L.M.; Parajuli, R.; Miller, P.C.; Rawal, S.; Williams, A.J.; Cote, R.J.; Lippman, M.E.; Datar, R.H.; et al. Identification of Cancer-Associated Fibroblasts in Circulating Blood from Patients with Metastatic Breast Cancer. Cancer Res. 2015, 75, 4681–4687. [Google Scholar] [CrossRef] [Green Version]
- Yadav, A.; Kumar, B.; Yu, J.G.; Old, M.; Teknos, T.N.; Kumar, P. Tumor-Associated Endothelial Cells Promote Tumor Metastasis by Chaperoning Circulating Tumor Cells and Protecting Them from Anoikis. PLoS ONE 2015, 10, e0141602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lalmahomed, Z.S.; Kraan, J.; Gratama, J.W.; Mostert, B.; Sleijfer, S.; Verhoef, C. Circulating tumor cells and sample size: The more, the better. J. Clin. Oncol. 2010, 28, e288–e289. [Google Scholar] [CrossRef] [PubMed]
- Terai, M.; Mu, Z.; Eschelman, D.J.; Gonsalves, C.F.; Kageyama, K.; Chervoneva, I.; Orloff, M.; Weight, R.; Mastrangelo, M.J.; Cristofanilli, M.; et al. Arterial Blood, Rather Than Venous Blood, is a Better Source for Circulating Melanoma Cells. EBioMedicine 2015, 2, 1821–1826. [Google Scholar] [CrossRef] [Green Version]
- Wong, K.H.K.; Tessier, S.N.; Miyamoto, D.T.; Miller, K.L.; Bookstaver, L.D.; Carey, T.R.; Stannard, C.J.; Thapar, V.; Tai, E.C.; Vo, K.D.; et al. Whole blood stabilization for the microfluidic isolation and molecular characterization of circulating tumor cells. Nat. Commun. 2017, 8, 1733. [Google Scholar] [CrossRef]
- Satelli, A.; Brownlee, Z.; Mitra, A.; Meng, Q.H.; Li, S. Circulating tumor cell enumeration with a combination of epithelial cell adhesion molecule- and cell-surface vimentin-based methods for monitoring breast cancer therapeutic response. Clin. Chem. 2015, 61, 259–266. [Google Scholar] [CrossRef] [Green Version]
- Satelli, A.; Mitra, A.; Brownlee, Z.; Xia, X.; Bellister, S.; Overman, M.J.; Kopetz, S.; Ellis, L.M.; Meng, Q.H.; Li, S. Epithelial-mesenchymal transitioned circulating tumor cells capture for detecting tumor progression. Clin. Cancer Res. 2015, 21, 899–906. [Google Scholar] [CrossRef] [Green Version]
- Agerbaek, M.O.; Bang-Christensen, S.R.; Yang, M.H.; Clausen, T.M.; Pereira, M.A.; Sharma, S.; Ditlev, S.B.; Nielsen, M.A.; Choudhary, S.; Gustavsson, T.; et al. The VAR2CSA malaria protein efficiently retrieves circulating tumor cells in an EpCAM-independent manner. Nat. Commun. 2018, 9, 3279. [Google Scholar] [CrossRef] [Green Version]
- Po, J.W.; Roohullah, A.; Lynch, D.; DeFazio, A.; Harrison, M.; Harnett, P.R.; Kennedy, C.; de Souza, P.; Becker, T.M. Improved ovarian cancer EMT-CTC isolation by immunomagnetic targeting of epithelial EpCAM and mesenchymal N-cadherin. J. Circ. Biomark 2018, 7. [Google Scholar] [CrossRef] [Green Version]
- Wan, Y.; Zhou, Y.G.; Poudineh, M.; Safaei, T.S.; Mohamadi, R.M.; Sargent, E.H.; Kelley, S.O. Highly specific electrochemical analysis of cancer cells using multi-nanoparticle labeling. Angew. Chem. Int. Ed. 2014, 53, 13145–13149. [Google Scholar] [CrossRef]
- Ghazani, A.A.; Castro, C.M.; Gorbatov, R.; Lee, H.; Weissleder, R. Sensitive and direct detection of circulating tumor cells by multimarker micro-nuclear magnetic resonance. Neoplasia 2012, 14, 388–395. [Google Scholar] [CrossRef] [Green Version]
- Nima, Z.A.; Mahmood, M.; Xu, Y.; Mustafa, T.; Watanabe, F.; Nedosekin, D.A.; Juratli, M.A.; Fahmi, T.; Galanzha, E.I.; Nolan, J.P.; et al. Circulating tumor cell identification by functionalized silver-gold nanorods with multicolor, super-enhanced SERS and photothermal resonances. Sci. Rep. 2014, 4, 4752. [Google Scholar] [CrossRef]
- Gast, C.E.; Silk, A.D.; Zarour, L.; Riegler, L.; Burkhart, J.G.; Gustafson, K.T.; Parappilly, M.S.; Roh-Johnson, M.; Goodman, J.R.; Olson, B.; et al. Cell fusion potentiates tumor heterogeneity and reveals circulating hybrid cells that correlate with stage and survival. Sci. Adv. 2018, 4, eaat7828. [Google Scholar] [CrossRef] [Green Version]
Technology | Detection Method | Description | References |
---|---|---|---|
The Ficoll-Paque® | Centrifugation | Cell separation based on relative density | [12,13] |
AccuCyte CTC | Special device for collecting buffy coat that further spreads on a slide for staining CTCs | [14,15] | |
OncoQuick® | Centrifugation/filtration | Enriched CTCs in the interphase between the porous barrier and the separation medium | [16] |
The RosetteSep™ CTC Enrichment Cocktail | Immunoaffinity centrifugation | Standard density gradient centrifugation combined with tetrameric antibody complexes for removing blood cells (negative selection) | [17,18] |
Quadrupole Magnetic Separator (QMS) | Immunoaffinity/filtration | The flow channels and the permanent magnet assembly for negative selection of CTC enrichment | [19] |
The ISET® | Filtration | Isolation of CTCs and CTM based on size | [20] |
ScreenCell® System | Size exclusion technology for isolating circulating rare cells (CRCs) | [21] | |
faCTChecker—Circulogix | Automatic filtration-based CTC enrichment system | [22,23] | |
Parsortix™ | Microfiltration | Microfluidic technology that uses a cassette and captures CTCs based on their less deformable nature and larger size | [24,25] |
The VTX-1 Liquid Biopsy System | Laminar microvortices to isolate and concentrate CTCs | [26] | |
ClearCell® FX1 System and CTChip® FR | Under the influence of inherent centrifugal forces, the CTCs are separated based on the size in a spiral microchannel | [27] | |
ApoStream® | Polarizability | A dielectrophoretic (DEP) force arises when an electric field is applied to the cells with different dielectric characteristics producing the separation of the cells | [28] |
The CellSearch® system technology (Menarini-Silicon Biosystems) | Immunomagnetic cell selection system | Antibody-labeled magnetic nanoparticles that target epithelial cell adhesion molecules of CTCs | [29] |
AdnaTest | Tumor cell enrichment based on magnetic beads that are coupled to a mixture of antibodies | [30] | |
MagSweeper | The capture of CTCs using magnetic rods covered with removable plastic sleeves | [31] | |
CTC-iChip | Microfluidic system | CTC isolation by lateral displacement, inertial focusing, two-stage magnetophoresis, and depletion antibodies against leukocytes | [32] |
GEDI chip | The chip is composed of a row of posts coated with antibodies. The larger cancer cells collide with the row of posts and stick on as the other cells flow through | [33] | |
The OncoCEE® (Biocept, Inc.) | The platform utilizes an antibody capture cocktail and CTCs are captured in a microchannel | [34] | |
The IsoFlux System™ | The CTCs and other rare cells flow through the microfluidic channel and reach an isolation zone where the cells get pulled as they pass through an external magnetic field | [35] | |
LiquidBiopsy® | Cells captured on a flow-chip technology that processes the reagents for the immunomagnetic isolation of target rare cells | [36] | |
Herringbone-chip or HB-chip | A microvortex mixing device that ensures contacts of cells with antibody-coated surfaces for the capture of CTCs | [37] | |
GEM chip | The staggered herringbone grooves disrupt streamlines inducing microvortex and capture of CTCs inside the channels | [38] | |
Graphene oxide (GO) chip | Graphene oxide nanosheets stuck to gold nanoparticles promote the growth of molecular chains that grab onto CTCs | [39] | |
BioFluidica | Sinusoidally-shaped channels are coated with antibodies, which isolate specific CTCs from whole blood | [40] | |
NanoVelcro CTC chip | Capture agents and embedded nanosubstrates increases the affinity between CTCs and surface capture | [41,42] | |
The magnetic ranking cytometry (MagRC) | A microfluidic device sorts the cells into different zones based on magnetic labeling levels | [43] | |
Gold nanoparticles (AuNPs) | Gold nanoparticles are an efficient platform for assembling multivalent DNA aptamers for high efficiency cell capture | [44] | |
Microfluidic Western blot | Single-cell resolution Western blot (scWB) to measure a panel of proteins in single CTCs | [45] | |
Gilupi Nanodetector® | Therapeutic apheresis | The nanodetector or thin wire is inserted into the vein of the patient. Cells are docked onto “nano polymer threads” containing antibodies | [46] |
In vivo flow cytometry (IVFC) | Quantitative analysis of CTC without the need for the blood collection | [47] |
Cancer Type | Surface Markers | References |
---|---|---|
Breast cancer | Epithelial cell adhesion molecule, EpCAM | [29,30,56] |
Cytokeratins 7, 8, 18, 19 | ||
Epidermal growth factor receptor 2, HER2 | ||
Mucin1, MUC-1 | ||
Zinc finger protein SNAI1, Snail | ||
Neural cadherin, N-cadherin | ||
Vimentin | ||
Castration-resistant prostate cancer | Epithelial cell adhesion molecule, EpCAM | [57,58,59] |
Cytokeratin | ||
Pancreatic cancer | Epithelial cell adhesion molecule, EpCAM | [31,60] |
Cytokeratins 8, 18, 19 | ||
Epidermal growth factor receptor, EGFR | ||
Hepatocellular carcinoma | Epithelial cell adhesion molecule, EpCAM | [32,33] |
Cytokeratin | ||
β-catenin | ||
Melanoma | Melanoma-associated antigen, MLANA | [34] |
Small-cell lung cancer | Epithelial cell adhesion molecule, EpCAM | [35,36] |
Chromosome 8 centromere probe, CEP8 | ||
Synaptophysin, SYP | ||
Enolase-2, ENOS | ||
Chromogranin A, CHGA | ||
Neural cell adhesion molecule, CD56/NCAM | ||
Non-small cell lung cancer | Epithelial cell adhesion molecule, EpCAM | [37,38] |
Cytokeratins 8, 18, 19 | ||
Cell surface vimentin, CSV | ||
Urinary bladder cancer | Epithelial cell adhesion molecule, EpCAM | [39,40] |
Cytokeratins 8, 18, 19 | ||
Ovarian cancer | Epithelial cell adhesion molecule, EpCAM | [41,42] |
Cytokeratin | ||
Colon and rectum cancer | Epithelial cell adhesion molecule, EpCAM | [18,19,61] |
Cytokeratin | ||
Cell surface vimentin, CSV | ||
Esophageal squamous cell carcinoma | Twist2Cla | [43] |
Pan-cytokeratin (AE1/AE3) | ||
Epithelial cell adhesion molecule, EpCAM | ||
Brain cancer and glioblastoma | Glial fibrillary acidic protein, GFAP | [45,62] |
SRY-related HMG box, Sox2 | ||
Tubulin beta-3, TUBB3 | ||
Epidermal growth factor receptor, EGFR | ||
A2B5 | ||
Hepatocyte growth factor receptor, MET | ||
OLIG2 |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 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 (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Yang, Y.-P.; Giret, T.M.; Cote, R.J. Circulating Tumor Cells from Enumeration to Analysis: Current Challenges and Future Opportunities. Cancers 2021, 13, 2723. https://doi.org/10.3390/cancers13112723
Yang Y-P, Giret TM, Cote RJ. Circulating Tumor Cells from Enumeration to Analysis: Current Challenges and Future Opportunities. Cancers. 2021; 13(11):2723. https://doi.org/10.3390/cancers13112723
Chicago/Turabian StyleYang, Yu-Ping, Teresa M. Giret, and Richard J. Cote. 2021. "Circulating Tumor Cells from Enumeration to Analysis: Current Challenges and Future Opportunities" Cancers 13, no. 11: 2723. https://doi.org/10.3390/cancers13112723
APA StyleYang, Y. -P., Giret, T. M., & Cote, R. J. (2021). Circulating Tumor Cells from Enumeration to Analysis: Current Challenges and Future Opportunities. Cancers, 13(11), 2723. https://doi.org/10.3390/cancers13112723