A Review of Circulating Tumour Cell Enrichment Technologies
Abstract
:Simple Summary
Abstract
1. Introduction
2. Immunomagnetic Positive Enrichment
3. Immunomagnetic Negative Enrichment
Technology | Trial Identifier | Cancer Type | Status | Study Type | Study Start Date | Estimated/Actual Completion Date | Estimated/Actual Number of Patients Enrolled |
---|---|---|---|---|---|---|---|
RosetteSep | NCT02349867 | Pancreatic | Active | Phase I | January 2015 | September 2024 | 23 |
CTC-Chip | NCT00888134 | Solid neoplasm | Completed * | Phase II | July 2009 | January 2015 | 28 |
NCT01961713 | Prostate | Recruiting | Observational | April 2010 | August 2022 | 200 | |
NCT01734915 | Lung | Completed [73] | Observational | November 2012 | September 2016 | 40 | |
NCT02812680 | Oesophageal | Recruiting | Observational | June 2016 | June 2021 | 200 | |
NCT02630615 | Lung | Active | Observational | September 2017 | August 2021 | 41 | |
GEDI chip | NCT01718353 | Prostate | Completed [74] | Phase II | March 2013 | August 2015 | 63 |
Nanovelcro | NCT01834651 | Prostate | Completed [75] | Phase II | April 2013 | July 2016 | 17 |
FMSA | NCT01722903 | Colorectal | Completed [76] | Observational | April 2012 | June 2015 | 25 |
ScreenCell | NCT03797053 | Melanoma | Unknown | Observational | April 2015 | Junee 2019 | 450 |
NCT02610764 | Oesophageal | Completed * | Interventional | November 2015 | December 2016 | 19 | |
ISET | NCT00818558 | Lung | Unknown | Interventional | October 2008 | October 2012 | 520 |
NCT01776385 | Pleural neoplasms | Completed * | Interventional | February 2012 | March 2018 | 9 | |
NCT02372448 | Lung | Completed [77] | Interventional | January 2015 | November 2019 | 206 | |
NCT02500693 | Lung | Unknown | Interventional | October 2015 | September 2019 | 600 | |
NCT02827344 | Lung | Recruiting | Observational | October 2015 | December 2021 | 200 | |
NCT02554448 | Rectal | Unknown | Interventional | January 2016 | December 2016 | 80 | |
NCT03328559 | Bronchial | Active | Interventional | March 2016 | December 2020 | 6 | |
NCT02979470 | Colorectal | Unknown | Observational | September 2016 | September 2019 | 100 | |
NCT04702633 | Prostate | Not yet recruiting | Observational | February 2021 | February 2024 | 200 | |
Parsortix | NCT02781272 | Ovarian | Active | Observational | June 2016 | June 2022 | 200 |
NCT02785731 | Ovarian | Completed * | Observational | July 2016 | July 2018 | 204 | |
NCT03771404 | Lung | Recruiting | Interventional | January 2018 | December 2021 | 50 | |
NCT03427450 | Breast | Completed * | Observational | March 2018 | December 2019 | 421 | |
NCT04021394 | Prostate | Recruiting | Observational | June 2019 | December 2025 | 40 | |
ClearCell | NCT02370303 | Lung | Completed * | Observational | August 2014 | July 2016 | 23 |
NCT04696744 | Head and neck | Not yet recruiting | Observational | February 2021 | January 2025 | 40 | |
ApoStream | NCT02349867 | Pancreatic | Active | Phase I | January 2015 | September 2024 | 23 |
NCT02466802 | Solid neoplasm | Completed * | Phase I | July 2015 | January 2019 | 32 |
4. Microfluidic Immunocapture
5. Capture Enhancement by Nanomaterials
6. Membrane Filtration
7. Size-Based Microfluidics
8. Density Based
9. Dielectrophoresis
10. In Vivo Enrichment
11. Combined Methods
12. Secondary Isolation Technologies
13. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- 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]
- 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] [PubMed]
- Punnoose, E.A.; Atwal, S.K.; Spoerke, J.M.; Savage, H.; Pandita, A.; Yeh, R.F.; Pirzkall, A.; Fine, B.M.; Amler, L.C.; Chen, D.S.; et al. Molecular biomarker analyses using circulating tumor cells. PLoS ONE 2010, 5, e12517. [Google Scholar] [CrossRef] [PubMed] [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]
- 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]
- Politaki, E.; Agelaki, S.; Apostolaki, S.; Hatzidaki, D.; Strati, A.; Koinis, F.; Perraki, M.; Saloustrou, G.; Stoupis, G.; Kallergi, G.; et al. A Comparison of Three Methods for the Detection of Circulating Tumor Cells in Patients with Early and Metastatic Breast Cancer. Cell. Physiol. Biochem. 2017, 44, 594–606. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mikolajczyk, S.D.; Millar, L.S.; Tsinberg, P.; Coutts, S.M.; Zomorrodi, M.; Pham, T.; Bischoff, F.Z.; Pircher, T.J. Detection of EpCAM-Negative and Cytokeratin-Negative Circulating Tumor Cells in Peripheral Blood. J. Oncol. 2011, 2011, 252361. [Google Scholar] [CrossRef]
- Miltenyi, S.; Müller, W.; Weichel, W.; Radbruch, A. High gradient magnetic cell separation with MACS. Cytometry 1990, 11, 231–238. [Google Scholar] [CrossRef]
- Zhu, D.M.; Wu, L.; Suo, M.; Gao, S.; Xie, W.; Zan, M.H.; Liu, A.; Chen, B.; Wu, W.T.; Ji, L.W.; et al. Engineered red blood cells for capturing circulating tumor cells with high performance. Nanoscale 2018, 10, 6014–6023. [Google Scholar] [CrossRef]
- 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] [Green Version]
- Cann, G.M.; Gulzar, Z.G.; Cooper, S.; Li, R.; Luo, S.; Tat, M.; Stuart, S.; Schroth, G.; Srinivas, S.; Ronaghi, M.; et al. mRNA-Seq of single prostate cancer circulating tumor cells reveals recapitulation of gene expression and pathways found in prostate cancer. PLoS ONE 2012, 7, e49144. [Google Scholar] [CrossRef]
- Schmidt, T.G.; Skerra, A. The Strep-tag system for one-step purification and high-affinity detection or capturing of proteins. Nat. Protoc. 2007, 2, 1528–1535. [Google Scholar] [CrossRef]
- Lu, N.-N.; Xie, M.; Wang, J.; Lv, S.-W.; Yi, J.-S.; Dong, W.-G.; Huang, W.-H. Biotin-Triggered Decomposable Immunomagnetic Beads for Capture and Release of Circulating Tumor Cells. ACS Appl. Mater. Interfaces 2015, 7, 8817–8826. [Google Scholar] [CrossRef] [PubMed]
- Xiong, K.; Wei, W.; Jin, Y.; Wang, S.; Zhao, D.; Wang, S.; Gao, X.; Qiao, C.; Yue, H.; Ma, G.; et al. Biomimetic Immuno-Magnetosomes for High-Performance Enrichment of Circulating Tumor Cells. Adv. Mater. 2016, 28, 7929–7935. [Google Scholar] [CrossRef]
- Drucker, A.; Teh, E.M.; Kostyleva, R.; Rayson, D.; Douglas, S.; Pinto, D.M. Comparative performance of different methods for circulating tumor cell enrichment in metastatic breast cancer patients. PLoS ONE 2020, 15, e0237308. [Google Scholar] [CrossRef] [PubMed]
- 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]
- 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]
- Sequist, L.V.; Nagrath, S.; Toner, M.; Haber, D.A.; Lynch, T.J. The CTC-chip: An exciting new tool to detect circulating tumor cells in lung cancer patients. J. Thorac. Oncol. 2009, 4, 281–283. [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] [PubMed] [Green Version]
- 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]
- 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]
- Adams, A.A.; Okagbare, P.I.; Feng, J.; Hupert, M.L.; Patterson, D.; Göttert, J.; McCarley, R.L.; Nikitopoulos, D.; Murphy, M.C.; Soper, S.A. Highly efficient circulating tumor cell isolation from whole blood and label-free enumeration using polymer-based microfluidics with an integrated conductivity sensor. J. Am. Chem. Soc. 2008, 130, 8633–8641. [Google Scholar] [CrossRef] [Green Version]
- Jan, Y.J.; Chen, J.-F.; Zhu, Y.; Lu, Y.-T.; Chen, S.H.; Chung, H.; Smalley, M.; Huang, Y.-W.; Dong, J.; Chen, L.-C.; et al. NanoVelcro rare-cell assays for detection and characterization of circulating tumor cells. Adv. Drug Deliv. Rev. 2018, 125, 78–93. [Google Scholar] [CrossRef]
- Park, M.-H.; Reátegui, E.; Li, W.; Tessier, S.N.; Wong, K.H.K.; Jensen, A.E.; Thapar, V.; Ting, D.; Toner, M.; Stott, S.L.; et al. Enhanced Isolation and Release of Circulating Tumor Cells Using Nanoparticle Binding and Ligand Exchange in a Microfluidic Chip. J. Am. Chem. Soc. 2017, 139, 2741–2749. [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] [PubMed]
- Yoon, H.J.; Shanker, A.; Wang, Y.; Kozminsky, M.; Jin, Q.; Palanisamy, N.; Burness, M.L.; Azizi, E.; Simeone, D.M.; Wicha, M.S.; et al. Tunable Thermal-Sensitive Polymer–Graphene Oxide Composite for Efficient Capture and Release of Viable Circulating Tumor Cells. Adv. Mater. 2016, 28, 4891–4897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, S.; Wang, H.; Jiao, J.; Chen, K.-J.; Owens, G.E.; Kamei, K.-i.; Sun, J.; Sherman, D.J.; Behrenbruch, C.P.; Wu, H.; et al. Three-Dimensional Nanostructured Substrates toward Efficient Capture of Circulating Tumor Cells. Angew. Chem. Int. Ed. 2009, 48, 8970–8973. [Google Scholar] [CrossRef] [PubMed]
- Loeian, M.S.; Mehdi Aghaei, S.; Farhadi, F.; Rai, V.; Yang, H.W.; Johnson, M.D.; Aqil, F.; Mandadi, M.; Rai, S.N.; Panchapakesan, B. Liquid biopsy using the nanotube-CTC-chip: Capture of invasive CTCs with high purity using preferential adherence in breast cancer patients. Lab Chip 2019, 19, 1899–1915. [Google Scholar] [CrossRef] [Green Version]
- Harouaka, R.A.; Zhou, M.-D.; Yeh, Y.-T.; Khan, W.J.; Das, A.; Liu, X.; Christ, C.C.; Dicker, D.T.; Baney, T.S.; Kaifi, J.T.; et al. Flexible Micro Spring Array Device for High-Throughput Enrichment of Viable Circulating Tumor Cells. Clin. Chem. 2014, 60, 323–333. [Google Scholar] [CrossRef] [Green Version]
- Desitter, I.; Guerrouahen, B.S.; Benali-Furet, N.; Wechsler, J.; Jänne, 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]
- Farace, F.; Massard, C.; Vimond, N.; Drusch, F.; Jacques, N.; Billiot, F.; Laplanche, A.; Chauchereau, A.; Lacroix, L.; Planchard, D.; et al. A direct comparison of CellSearch and ISET for circulating tumour-cell detection in patients with metastatic carcinomas. Br. J. Cancer 2011, 105, 847–853. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kallergi, G.; Politaki, E.; Alkahtani, S.; Stournaras, C.; Georgoulias, V. Evaluation of Isolation Methods for Circulating Tumor Cells (CTCs). Cell. Physiol. Biochem. 2016, 40, 411–419. [Google Scholar] [CrossRef] [PubMed]
- Zhou, M.-D.; Hao, S.; Williams, A.J.; Harouaka, R.A.; Schrand, B.; Rawal, S.; Ao, Z.; Brenneman, R.; Gilboa, E.; Lu, B.; et al. Separable Bilayer Microfiltration Device for Viable Label-free Enrichment of Circulating Tumour Cells. Sci. Rep. 2014, 4, 7392. [Google Scholar] [CrossRef]
- Kim, T.-H.; Lim, M.; Park, J.; Oh, J.M.; Kim, H.; Jeong, H.; Lee, S.J.; Park, H.C.; Jung, S.; Kim, B.C.; et al. FAST: Size-Selective, Clog-Free Isolation of Rare Cancer Cells from Whole Blood at a Liquid–Liquid Interface. Anal. Chem. 2017, 89, 1155–1162. [Google Scholar] [CrossRef]
- Cohen, E.N.; Jayachandran, G.; Hardy, M.R.; Venkata Subramanian, A.M.; Meng, X.; Reuben, J.M. Antigen-agnostic microfluidics-based circulating tumor cell enrichment and downstream molecular characterization. PLoS ONE 2020, 15, e0241123. [Google Scholar] [CrossRef] [PubMed]
- Hosokawa, M.; Hayata, T.; Fukuda, Y.; Arakaki, A.; Yoshino, T.; Tanaka, T.; Matsunaga, T. Size-Selective Microcavity Array for Rapid and Efficient Detection of Circulating Tumor Cells. Anal. Chem. 2010, 82, 6629–6635. [Google Scholar] [CrossRef] [PubMed]
- Lee, Y.; Guan, G.; Bhagat, A.A. ClearCell(R) FX, a label-free microfluidics technology for enrichment of viable circulating tumor cells. Cytom. Part A 2018, 93, 1251–1254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yap, Y.-S.; Leong, M.C.; Chua, Y.W.; Loh, K.W.J.; Lee, G.E.; Lim, E.H.; Dent, R.; Ng, R.C.H.; Lim, J.H.-C.; Singh, G.; et al. Detection and prognostic relevance of circulating tumour cells (CTCs) in Asian breast cancers using a label-free microfluidic platform. PLoS ONE 2019, 14, e0221305. [Google Scholar] [CrossRef]
- Lemaire, C.A.; Liu, S.Z.; Wilkerson, C.L.; Ramani, V.C.; Barzanian, N.A.; Huang, K.-W.; Che, J.; Chiu, M.W.; Vuppalapaty, M.; Dimmick, A.M.; et al. Fast and Label-Free Isolation of Circulating Tumor Cells from Blood: From a Research Microfluidic Platform to an Automated Fluidic Instrument, VTX-1 Liquid Biopsy System. SLAS Technol. Transl. Life Sci. Innov. 2018, 23, 16–29. [Google Scholar] [CrossRef] [Green Version]
- Sollier-Christen, E.; Renier, C.; Kaplan, T.; Kfir, E.; Crouse, S.C. VTX-1 Liquid Biopsy System for Fully-Automated and Label-Free Isolation of Circulating Tumor Cells with Automated Enumeration by BioView Platform. Cytom. Part A 2018, 93, 1240–1245. [Google Scholar] [CrossRef] [Green Version]
- Hyun, K.A.; Kwon, K.; Han, H.; Kim, S.I.; Jung, H.I. Microfluidic flow fractionation device for label-free isolation of circulating tumor cells (CTCs) from breast cancer patients. Biosens. Bioelectron. 2013, 40, 206–212. [Google Scholar] [CrossRef]
- Königsberg, R.; Obermayr, E.; Bises, G.; Pfeiler, G.; Gneist, M.; Wrba, F.; de Santis, M.; Zeillinger, R.; Hudec, M.; Dittrich, C. Detection of EpCAM positive and negative circulating tumor cells in metastatic breast cancer patients. Acta Oncol. 2011, 50, 700–710. [Google Scholar] [CrossRef] [PubMed]
- 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]
- 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] [PubMed] [Green Version]
- Rugo, H.S.; Cortes, J.; Awada, A.; O’Shaughnessy, J.; Twelves, C.; Im, S.A.; Hannah, A.; Lu, L.; Sy, S.; Caygill, K.; et al. Change in Topoisomerase 1-Positive Circulating Tumor Cells Affects Overall Survival in Patients with Advanced Breast Cancer after Treatment with Etirinotecan Pegol. Clin. Cancer Res. 2018, 24, 3348–3357. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gupta, V.; Jafferji, I.; Garza, M.; Melnikova, V.O.; Hasegawa, D.K.; Pethig, R.; Davis, D.W. ApoStream(™), a new dielectrophoretic device for antibody independent isolation and recovery of viable cancer cells from blood. Biomicrofluidics 2012, 6, 24133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fehm, T.N.; Meier-Stiegen, F.; Driemel, C.; Jäger, B.; Reinhardt, F.; Naskou, J.; Franken, A.; Neubauer, H.; Neves, R.P.L.; van Dalum, G.; et al. Diagnostic leukapheresis for CTC analysis in breast cancer patients: CTC frequency, clinical experiences and recommendations for standardized reporting. Cytom. Part A 2018, 93, 1213–1219. [Google Scholar] [CrossRef] [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]
- Ozkumur, E.; Shah, A.M.; Ciciliano, J.C.; Emmink, B.L.; Miyamoto, D.T.; Brachtel, E.; Yu, M.; Chen, P.-I.; Morgan, B.; Trautwein, J.; et al. Inertial Focusing for Tumor Antigen–Dependent and –Independent Sorting of Rare Circulating Tumor Cells. Sci. Transl. Med. 2013, 5, 179ra147. [Google Scholar] [CrossRef] [Green Version]
- Mishra, A.; Dubash, T.D.; Edd, J.F.; Jewett, M.K.; Garre, S.G.; Karabacak, N.M.; Rabe, D.C.; Mutlu, B.R.; Walsh, J.R.; Kapur, R.; et al. Ultrahigh-throughput magnetic sorting of large blood volumes for epitope-agnostic isolation of circulating tumor cells. Proc. Natl. Acad. Sci. USA 2020, 117, 16839–16847. [Google Scholar] [CrossRef]
- Lee, A.C.; Svedlund, J.; Darai, E.; Lee, Y.; Lee, D.; Lee, H.-B.; Kim, S.-M.; Kim, O.; Bae, H.J.; Choi, A.; et al. OPENchip: An on-chip in situ molecular profiling platform for gene expression analysis and oncogenic mutation detection in single circulating tumour cells. Lab Chip 2020, 20, 912–922. [Google Scholar] [CrossRef]
- Hayes, D.F.; Cristofanilli, M.; Budd, G.T.; Ellis, M.J.; Stopeck, A.; Miller, M.C.; Matera, J.; Allard, W.J.; Doyle, G.V.; Terstappen, L.W. Circulating tumor cells at each follow-up time point during therapy of metastatic breast cancer patients predict progression-free and overall survival. Clin. Cancer Res. 2006, 12, 4218–4224. [Google Scholar] [CrossRef] [Green Version]
- Ligthart, S.T.; Coumans, F.A.; Attard, G.; Cassidy, A.M.; de Bono, J.S.; Terstappen, L.W. Unbiased and automated identification of a circulating tumour cell definition that associates with overall survival. PLoS ONE 2011, 6, e27419. [Google Scholar] [CrossRef]
- Riethdorf, S.; Müller, V.; Zhang, L.; Rau, T.; Loibl, S.; Komor, M.; Roller, M.; Huober, J.; Fehm, T.; Schrader, I.; et al. Detection and HER2 expression of circulating tumor cells: Prospective monitoring in breast cancer patients treated in the neoadjuvant GeparQuattro trial. Clin. Cancer Res. 2010, 16, 2634–2645. [Google Scholar] [CrossRef] [PubMed] [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] [PubMed] [Green Version]
- Wang, L.; Balasubramanian, P.; Chen, A.P.; Kummar, S.; Evrard, Y.A.; Kinders, R.J. Promise and limits of the CellSearch platform for evaluating pharmacodynamics in circulating tumor cells. Semin. Oncol. 2016, 43, 464–475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Riethdorf, S.; Fritsche, H.; Müller, V.; Rau, T.; Schindlbeck, C.; Rack, B.; Janni, W.; Coith, C.; Beck, K.; Jänicke, F.; et al. Detection of Circulating Tumor Cells in Peripheral Blood of Patients with Metastatic Breast Cancer: A Validation Study of the CellSearch System. Clin. Cancer Res. 2007, 13, 920–928. [Google Scholar] [CrossRef] [Green Version]
- Miller, M.C.; Doyle, G.V.; Terstappen, L.W.M.M. Significance of Circulating Tumor Cells Detected by the CellSearch System in Patients with Metastatic Breast Colorectal and Prostate Cancer. J. Oncol. 2010, 2010, 617421. [Google Scholar] [CrossRef]
- ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/results?recrs=&cond=cancer&term=cellsearch&cntry=&state=&city=&dist= (accessed on 20 February 2021).
- Swennenhuis, J.F.; van Dalum, G.; Zeune, L.L.; Terstappen, L.W. Improving the CellSearch® system. Expert Rev. Mol. Diagn. 2016, 16, 1291–1305. [Google Scholar] [CrossRef] [Green Version]
- Kölbl, A.C.; Jeschke, U.; Andergassen, U. The Significance of Epithelial-to-Mesenchymal Transition for Circulating Tumor Cells. Int. J. Mol. Sci. 2016, 17, 1308. [Google Scholar] [CrossRef] [Green Version]
- Iwatsuki, M.; Mimori, K.; Yokobori, T.; Ishi, H.; Beppu, T.; Nakamori, S.; Baba, H.; Mori, M. Epithelial–mesenchymal transition in cancer development and its clinical significance. Cancer Sci. 2010, 101, 293–299. [Google Scholar] [CrossRef] [PubMed]
- Pan, L.; Yan, G.; Chen, W.; Sun, L.; Wang, J.; Yang, J. Distribution of circulating tumor cell phenotype in early cervical cancer. Cancer Manag. Res. 2019, 11, 5531–5536. [Google Scholar] [CrossRef] [Green Version]
- Han, D.; Chen, K.; Che, J.; Hang, J.; Li, H. Detection of Epithelial-Mesenchymal Transition Status of Circulating Tumor Cells in Patients with Esophageal Squamous Carcinoma. Biomed. Res. Int. 2018, 2018, 7610154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aktas, B.; Tewes, M.; Fehm, T.; Hauch, S.; Kimmig, R.; Kasimir-Bauer, S. Stem cell and epithelial-mesenchymal transition markers are frequently overexpressed in circulating tumor cells of metastatic breast cancer patients. Breast Cancer Res. 2009, 11, R46. [Google Scholar] [CrossRef] [Green Version]
- Powell, A.A.; Talasaz, A.H.; Zhang, H.; Coram, M.A.; Reddy, A.; Deng, G.; Telli, M.L.; Advani, R.H.; Carlson, R.W.; Mollick, J.A.; et al. Single cell profiling of circulating tumor cells: Transcriptional heterogeneity and diversity from breast cancer cell lines. PLoS ONE 2012, 7, e33788. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Abal, M.; Andreu, J.M.; Barasoain, I. Taxanes: Microtubule and centrosome targets, and cell cycle dependent mechanisms of action. Curr. Cancer Drug Targets 2003, 3, 193–203. [Google Scholar] [CrossRef]
- Junttila, M.R.; Saarinen, S.; Schmidt, T.; Kast, J.; Westermarck, J. Single-step Strep-tag purification for the isolation and identification of protein complexes from mammalian cells. Proteomics 2005, 5, 1199–1203. [Google Scholar] [CrossRef]
- Xu, Y.; Liu, B.; Ding, F.; Zhou, X.; Tu, P.; Yu, B.; He, Y.; Huang, P. Circulating tumor cell detection: A direct comparison between negative and unbiased enrichment in lung cancer. Oncol. Lett. 2017, 13, 4882–4886. [Google Scholar] [CrossRef] [Green Version]
- Boyer, M.; Cayrefourcq, L.; Garima, F.; Foulongne, V.; Dereure, O.; Alix-Panabières, C. Circulating Tumor Cell Detection and Polyomavirus Status in Merkel Cell Carcinoma. Sci. Rep. 2020, 10, 1612. [Google Scholar] [CrossRef] [Green Version]
- Szczerba, B.M.; Castro-Giner, F.; Vetter, M.; Krol, I.; Gkountela, S.; Landin, J.; Scheidmann, M.C.; Donato, C.; Scherrer, R.; Singer, J.; et al. Neutrophils escort circulating tumour cells to enable cell cycle progression. Nature 2019, 566, 553–557. [Google Scholar] [CrossRef]
- Amantini, C.; Morelli, M.B.; Nabissi, M.; Piva, F.; Marinelli, O.; Maggi, F.; Bianchi, F.; Bittoni, A.; Berardi, R.; Giampieri, R.; et al. Expression Profiling of Circulating Tumor Cells in Pancreatic Ductal Adenocarcinoma Patients: Biomarkers Predicting Overall Survival. Front. Oncol. 2019, 9. [Google Scholar] [CrossRef] [PubMed]
- Sundaresan, T.K.; Sequist, L.V.; Heymach, J.V.; Riely, G.J.; Jänne, P.A.; Koch, W.H.; Sullivan, J.P.; Fox, D.B.; Maher, R.; Muzikansky, A.; et al. Detection of T790M, the Acquired Resistance EGFR Mutation, by Tumor Biopsy versus Noninvasive Blood-Based Analyses. Clin. Cancer Res. 2016, 22, 1103–1110. [Google Scholar] [CrossRef] [Green Version]
- Antonarakis, E.S.; Tagawa, S.T.; Galletti, G.; Worroll, D.; Ballman, K.; Vanhuyse, M.; Sonpavde, G.; North, S.; Albany, C.; Tsao, C.K.; et al. Randomized, Noncomparative, Phase II Trial of Early Switch From Docetaxel to Cabazitaxel or Vice Versa, With Integrated Biomarker Analysis, in Men With Chemotherapy-Naïve, Metastatic, Castration-Resistant Prostate Cancer. J. Clin. Oncol. 2017, 35, 3181–3188. [Google Scholar] [CrossRef] [Green Version]
- Huynh, R.; Chen, J.-F.; Tighiouart, M.; Sievert, M.; Oppenheim, A.; Moldawer, N.P.; Knudsen, B.; Freeman, M.; Bhowmick, N.; Rogatko, A.; et al. A phase II study of cabozantinib in metastatic castration-resistant prostate cancer (mCRPC) with visceral metastases (VM) with very small nuclear circulating tumor cell (vsnCTC) association studies. J. Clin. Oncol. 2016, 34, 208. [Google Scholar] [CrossRef]
- Kaifi, J.T.; Kunkel, M.; Das, A.; Harouaka, R.A.; Dicker, D.T.; Li, G.; Zhu, J.; Clawson, G.A.; Yang, Z.; Reed, M.F.; et al. Circulating tumor cell isolation during resection of colorectal cancer lung and liver metastases: A prospective trial with different detection techniques. Cancer Biol. Ther. 2015, 16, 699–708. [Google Scholar] [CrossRef] [Green Version]
- Ilie, M.; MaziÈRes, J.; Chamorey, E.; Heeke, S.; Benzaquen, J.; Thamphya, B.; Boutros, J.; Tiotiu, A.; Fayada, J.; Cadranel, J.; et al. Prospective multi-center validation of the detection of ALK rearrangements of circulating tumor cells for non-invasive longitudinal management of patients with advanced non-small cell lung cancer. J. Thorac. Oncol. 2021. [Google Scholar] [CrossRef] [PubMed]
- Zou, D.; Cui, D. Advances in isolation and detection of circulating tumor cells based on microfluidics. Cancer Biol. Med. 2018, 15, 335–353. [Google Scholar] [CrossRef] [Green Version]
- Maheswaran, S.; Sequist, L.V.; Nagrath, S.; Ulkus, L.; Brannigan, B.; Collura, C.V.; Inserra, E.; Diederichs, S.; Iafrate, A.J.; Bell, D.W.; et al. Detection of Mutations in EGFR in Circulating Lung-Cancer Cells. N. Engl. J. Med. 2008, 359, 366–377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kaburagi, T.; Kiyoshima, M.; Nawa, T.; Ichimura, H.; Saito, T.; Hayashihara, K.; Yamada, H.; Satoh, H.; Endo, T.; Inage, Y.; et al. Acquired EGFR T790M Mutation After Relapse Following EGFR-TKI Therapy: A Population-based Multi-institutional Study. Anticancer Res. 2018, 38, 3145–3150. [Google Scholar] [CrossRef]
- Xue, P.; Wu, Y.; Guo, J.; Kang, Y. Highly efficient capture and harvest of circulating tumor cells on a microfluidic chip integrated with herringbone and micropost arrays. Biomed. Microdevices 2015, 17, 39. [Google Scholar] [CrossRef] [PubMed]
- Kirby, B.J.; Jodari, M.; Loftus, M.S.; Gakhar, G.; Pratt, E.D.; Chanel-Vos, C.; Gleghorn, J.P.; Santana, S.M.; Liu, H.; Smith, J.P.; et al. Functional characterization of circulating tumor cells with a prostate-cancer-specific microfluidic device. PLoS ONE 2012, 7, e35976. [Google Scholar] [CrossRef]
- Galletti, G.; Sung, M.S.; Vahdat, L.T.; Shah, M.A.; Santana, S.M.; Altavilla, G.; Kirby, B.J.; Giannakakou, P. Isolation of breast cancer and gastric cancer circulating tumor cells by use of an anti HER2-based microfluidic device. Lab Chip 2014, 14, 147–156. [Google Scholar] [CrossRef]
- 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] [PubMed] [Green Version]
- Ankeny, J.S.; Court, C.M.; Hou, S.; Li, Q.; Song, M.; Wu, D.; Chen, J.F.; Lee, T.; Lin, M.; Sho, S.; et al. Circulating tumour cells as a biomarker for diagnosis and staging in pancreatic cancer. Br. J. Cancer 2016, 114, 1367–1375. [Google Scholar] [CrossRef] [Green Version]
- Liu, S.; Tian, Z.; Zhang, L.; Hou, S.; Hu, S.; Wu, J.; Jing, Y.; Sun, H.; Yu, F.; Zhao, L.; et al. Combined cell surface carbonic anhydrase 9 and CD147 antigens enable high-efficiency capture of circulating tumor cells in clear cell renal cell carcinoma patients. Oncotarget 2016, 7, 59877–59891. [Google Scholar] [CrossRef] [PubMed]
- Chen, J.F.; Ho, H.; Lichterman, J.; Lu, Y.T.; Zhang, Y.; Garcia, M.A.; Chen, S.F.; Liang, A.J.; Hodara, E.; Zhau, H.E.; et al. Subclassification of prostate cancer circulating tumor cells by nuclear size reveals very small nuclear circulating tumor cells in patients with visceral metastases. Cancer 2015, 121, 3240–3251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fend, F.; Raffeld, M. Laser capture microdissection in pathology. J. Clin. Pathol. 2000, 53, 666–672. [Google Scholar] [CrossRef] [Green Version]
- Hou, S.; Zhao, L.; Shen, Q.; Yu, J.; Ng, C.; Kong, X.; Wu, D.; Song, M.; Shi, X.; Xu, X.; et al. Polymer Nanofiber-Embedded Microchips for Detection, Isolation, and Molecular Analysis of Single Circulating Melanoma Cells. Angew. Chem. Int. Ed. 2013, 52, 3379–3383. [Google Scholar] [CrossRef] [Green Version]
- Jiang, R.; Lu, Y.T.; Ho, H.; Li, B.; Chen, J.F.; Lin, M.; Li, F.; Wu, K.; Wu, H.; Lichterman, J.; et al. A comparison of isolated circulating tumor cells and tissue biopsies using whole-genome sequencing in prostate cancer. Oncotarget 2015, 6, 44781–44793. [Google Scholar] [CrossRef] [Green Version]
- Hou, S.; Zhao, H.; Zhao, L.; Shen, Q.; Wei, K.S.; Suh, D.Y.; Nakao, A.; Garcia, M.A.; Song, M.; Lee, T.; et al. Capture and Stimulated Release of Circulating Tumor Cells on Polymer-Grafted Silicon Nanostructures. Adv. Mater. 2013, 25, 1547–1551. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ke, Z.; Lin, M.; Chen, J.-F.; Choi, J.-s.; Zhang, Y.; Fong, A.; Liang, A.-J.; Chen, S.-F.; Li, Q.; Fang, W.; et al. Programming Thermoresponsiveness of NanoVelcro Substrates Enables Effective Purification of Circulating Tumor Cells in Lung Cancer Patients. ACS Nano. 2015, 9, 62–70. [Google Scholar] [CrossRef]
- Shen, M.-Y.; Chen, J.-F.; Luo, C.-H.; Lee, S.; Li, C.-H.; Yang, Y.-L.; Tsai, Y.-H.; Ho, B.-C.; Bao, L.-R.; Lee, T.-J.; et al. Glycan Stimulation Enables Purification of Prostate Cancer Circulating Tumor Cells on PEDOT NanoVelcro Chips for RNA Biomarker Detection. Adv. Healthc. Mater. 2018, 7, 1700701. [Google Scholar] [CrossRef]
- Springsteen, G.; Wang, B. A detailed examination of boronic acid–diol complexation. Tetrahedron 2002, 58, 5291–5300. [Google Scholar] [CrossRef]
- Agerbæk, M.; 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] [PubMed] [Green Version]
- Tseng, H.-C.; Lee, A.-W.; Wei, P.-L.; Chang, Y.-J.; Chen, J.-K. Clinical diagnosis of colorectal cancer using electrospun triple-blend fibrous mat-based capture assay of circulating tumor cells. J. Mater. Chem. B 2016, 4, 6565–6580. [Google Scholar] [CrossRef]
- Sánchez-Lorencio, M.I.; Ramirez, P.; Saenz, L.; Martínez Sánchez, M.V.; De La Orden, V.; Mediero-Valeros, B.; Veganzones-De-Castro, S.; Baroja-Mazo, A.; Revilla Nuin, B.; Gonzalez, M.R.; et al. Comparison of Two Types of Liquid Biopsies in Patients With Hepatocellular Carcinoma Awaiting Orthotopic Liver Transplantation. Transplant. Proc. 2015, 47, 2639–2642. [Google Scholar] [CrossRef] [PubMed]
- Alva, A.; Friedlander, T.; Clark, M.; Huebner, T.; Daignault, S.; Hussain, M.; Lee, C.; Hafez, K.; Hollenbeck, B.; Weizer, A.; et al. Circulating Tumor Cells as Potential Biomarkers in Bladder Cancer. J. Urol. 2015, 194, 790–798. [Google Scholar] [CrossRef] [PubMed]
- 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, 1849454418782617. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nimir, M.; Ma, Y.; Jeffreys, S.A.; Opperman, T.; Young, F.; Khan, T.; Ding, P.; Chua, W.; Balakrishnar, B.; Cooper, A.; et al. Detection of AR-V7 in Liquid Biopsies of Castrate Resistant Prostate Cancer Patients: A Comparison of AR-V7 Analysis in Circulating Tumor Cells, Circulating Tumor RNA and Exosomes. Cells 2019, 8, 688. [Google Scholar] [CrossRef] [Green Version]
- Gonzalez-Rivera, M.; Picornell, A.C.; Alvarez, E.L.; Martin, M. A Cross-Sectional Comparison of Druggable Mutations in Primary Tumors, Metastatic Tissue, Circulating Tumor Cells, and Cell-Free Circulating DNA in Patients with Metastatic Breast Cancer: The MIRROR Study Protocol. JMIR Res. Protoc. 2016, 5, e167. [Google Scholar] [CrossRef] [PubMed]
- Ionescu-Zanetti, C.; Brobey, R.; Tseng, H.; Rosenblatt, K.P.; Kamps-Hughes, N.; Tran, T.; Amato, R.J. High sensitivity and specificity NGS analysis of prostate and kidney cancer samples using a novel liquid biopsy workflow. J. Clin. Oncol. 2016, 34, e16607. [Google Scholar] [CrossRef]
- Ma, Y.; Luk, A.; Young, F.P.; Lynch, D.; Chua, W.; Balakrishnar, B.; De Souza, P.; Becker, T.M. Droplet Digital PCR Based Androgen Receptor Variant 7 (AR-V7) Detection from Prostate Cancer Patient Blood Biopsies. Int. J. Mol. Sci. 2016, 17, 1264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chan, S.C.; Li, Y.; Dehm, S.M. Androgen receptor splice variants activate androgen receptor target genes and support aberrant prostate cancer cell growth independent of canonical androgen receptor nuclear localization signal. J. Biol. Chem. 2012, 287, 19736–19749. [Google Scholar] [CrossRef] [Green Version]
- Antonarakis, E.S.; Lu, C.; Wang, H.; Luber, B.; Nakazawa, M.; Roeser, J.C.; Chen, Y.; Mohammad, T.A.; Chen, Y.; Fedor, H.L.; et al. AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer. N. Engl. J. Med. 2014, 371, 1028–1038. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Antonarakis, E.S.; Lu, C.; Luber, B.; Wang, H.; Chen, Y.; Nakazawa, M.; Nadal, R.; Paller, C.J.; Denmeade, S.R.; Carducci, M.A.; et al. Androgen Receptor Splice Variant 7 and Efficacy of Taxane Chemotherapy in Patients With Metastatic Castration-Resistant Prostate Cancer. JAMA Oncol. 2015, 1, 582–591. [Google Scholar] [CrossRef] [Green Version]
- Steinestel, J.; Luedeke, M.; Arndt, A.; Schnoeller, T.J.; Lennerz, J.K.; Wurm, C.; Maier, C.; Cronauer, M.V.; Steinestel, K.; Schrader, A.J. Detecting predictive androgen receptor modifications in circulating prostate cancer cells. Oncotarget 2019, 10, 4213–4223. [Google Scholar] [CrossRef] [Green Version]
- Chudasama, D.; Burnside, N.; Beeson, J.; Karteris, E.; Rice, A.; Anikin, V. Perioperative detection of circulating tumour cells in patients with lung cancer. Oncol. Lett. 2017, 14, 1281–1286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nicolazzo, C.; Colangelo, L.; Corsi, A.; Carpino, G.; Gradilone, A.; Sonato, C.; Raimondi, C.; Gaudio, E.; Gazzaniga, P.; Gianni, W. Liquid Biopsy in Rare Cancers: Lessons from Hemangiopericytoma. Anal. Cell. Pathol. 2018, 2018, 9718585. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vona, G.; Sabile, A.; Louha, M.; Sitruk, V.; Romana, S.; Schütze, 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 Circulating Tumor Cells. Am. J. Pathol. 2000, 156, 57–63. [Google Scholar] [CrossRef]
- Bai, M.; Zou, B.; Wang, Z.; Li, P.; Wang, H.; Ou, Y.; Cui, K.; Bian, J.; Li, S.; Xu, X. Comparison of two detection systems for circulating tumor cells among patients with renal cell carcinoma. Int. Urol. Nephrol. 2018, 50, 1801–1809. [Google Scholar] [CrossRef]
- Kang, H.M.; Kim, G.H.; Jeon, H.K.; Kim, D.H.; Jeon, T.Y.; Park, D.Y.; Jeong, H.; Chun, W.J.; Kim, M.H.; Park, J.; et al. Circulating tumor cells detected by lab-on-a-disc: Role in early diagnosis of gastric cancer. PLoS ONE 2017, 12, e0180251. [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. Cytom. Part A 2018, 93, 1234–1239. [Google Scholar] [CrossRef] [PubMed]
- Hosokawa, M.; Kenmotsu, H.; Koh, Y.; Yoshino, T.; Yoshikawa, T.; Naito, T.; Takahashi, T.; Murakami, H.; Nakamura, Y.; Tsuya, A.; et al. Size-Based Isolation of Circulating Tumor Cells in Lung Cancer Patients Using a Microcavity Array System. PLoS ONE 2013, 8, e67466. [Google Scholar] [CrossRef] [Green Version]
- Hou, H.W.; Warkiani, M.E.; Khoo, B.L.; Li, Z.R.; Soo, R.A.; Tan, D.S.-W.; Lim, W.-T.; Han, J.; Bhagat, A.A.S.; Lim, C.T. Isolation and retrieval of circulating tumor cells using centrifugal forces. Sci. Rep. 2013, 3, 1259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yin, J.; Wang, Z.; Li, G.; Lin, F.; Shao, K.; Cao, B.; Hou, Y. Characterization of circulating tumor cells in breast cancer patients by spiral microfluidics. Cell Biol. Toxicol. 2019, 35, 59–66. [Google Scholar] [CrossRef] [PubMed]
- Aya-Bonilla, C.A.; Morici, M.; Hong, X.; McEvoy, A.C.; Sullivan, R.J.; Freeman, J.; Calapre, L.; Khattak, M.A.; Meniawy, T.; Millward, M.; et al. Detection and prognostic role of heterogeneous populations of melanoma circulating tumour cells. Br. J. Cancer 2020, 122, 1059–1067. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Kidess-Sigal, E.; Liu, H.E.; Triboulet, M.M.; Che, J.; Ramani, V.C.; Visser, B.C.; Poultsides, G.A.; Longacre, T.A.; Marziali, A.; Vysotskaia, V.; et al. Enumeration and targeted analysis of KRAS, BRAF and PIK3CA mutations in CTCs captured by a label-free platform: Comparison to ctDNA and tissue in metastatic colorectal cancer. Oncotarget 2016, 7, 85349–85364. [Google Scholar] [CrossRef] [Green Version]
- Roock, W.D.; Vriendt, V.D.; Normanno, N.; Ciardiello, F.; Tejpar, S. KRAS, BRAF, PIK3CA, and PTEN mutations: Implications for targeted therapies in metastatic colorectal cancer. Lancet Oncol. 2011, 12, 594–603. [Google Scholar] [CrossRef]
- Müller, V.; Stahmann, N.; Riethdorf, S.; Rau, T.; Zabel, T.; Goetz, A.; Jänicke, F.; Pantel, K. Circulating Tumor Cells in Breast Cancer: Correlation to Bone Marrow Micrometastases, Heterogeneous Response to Systemic Therapy and Low Proliferative Activity. Clin. Cancer Res. 2005, 11, 3678–3685. [Google Scholar] [CrossRef] [Green Version]
- Königsberg, R.; Gneist, M.; Jahn-Kuch, D.; Pfeiler, G.; Hager, G.; Hudec, M.; Dittrich, C.; Zeillinger, R. Circulating tumor cells in metastatic colorectal cancer: Efficacy and feasibility of different enrichment methods. Cancer Lett. 2010, 293, 117–123. [Google Scholar] [CrossRef]
- Balic, M.; Dandachi, N.; Hofmann, G.; Samonigg, H.; Loibner, H.; Obwaller, A.; van der Kooi, A.; Tibbe, A.G.J.; Doyle, G.V.; Terstappen, L.W.M.M.; et al. Comparison of two methods for enumerating circulating tumor cells in carcinoma patients. Cytom. Part B Clin. Cytom. 2005, 68B, 25–30. [Google Scholar] [CrossRef]
- Ramirez, A.B.; U’Ren, L.; Campton, D.E.; Stewart, D.; Nordberg, J.J.; Stilwell, J.L.; Kaldjian, E.P. RareCyte® CTC Analysis Step 1: AccuCyte® Sample Preparation for the Comprehensive Recovery of Nucleated Cells from Whole Blood. Methods Mol. Biol. 2017, 1634, 163–172. [Google Scholar] [CrossRef]
- Gabriel, M.T.; Calleja, L.R.; Chalopin, A.; Ory, B.; Heymann, D. Circulating Tumor Cells: A Review of Non-EpCAM-Based Approaches for Cell Enrichment and Isolation. Clin. Chem. 2016, 62, 571–581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Le Du, F.; Fujii, T.; Kida, K.; Davis, D.W.; Park, M.; Liu, D.D.; Wu, W.; Chavez-MacGregor, M.; Barcenas, C.H.; Valero, V.; et al. EpCAM-independent isolation of circulating tumor cells with epithelial-to-mesenchymal transition and cancer stem cell phenotypes using ApoStream(R) in patients with breast cancer treated with primary systemic therapy. PLoS ONE 2020, 15, e0229903. [Google Scholar] [CrossRef]
- Balasubramanian, P.; Kinders, R.J.; Kummar, S.; Gupta, V.; Hasegawa, D.; Menachery, A.; Lawrence, S.M.; Wang, L.; Ferry-Galow, K.; Davis, D.; et al. Antibody-independent capture of circulating tumor cells of non-epithelial origin with the ApoStream® system. PLoS ONE 2017, 12, e0175414. [Google Scholar] [CrossRef] [Green Version]
- Fischer, J.C.; Niederacher, D.; Topp, S.A.; Honisch, E.; Schumacher, S.; Schmitz, N.; Zacarias Föhrding, L.; Vay, C.; Hoffmann, I.; Kasprowicz, N.S.; et al. Diagnostic leukapheresis enables reliable detection of circulating tumor cells of nonmetastatic cancer patients. Proc. Natl. Acad. Sci. USA 2013, 110, 16580–16585. [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]
- Reinhardt, F.; Franken, A.; Meier-Stiegen, F.; Driemel, C.; Stoecklein, N.H.; Fischer, J.C.; Niederacher, D.; Ruckhaeberle, E.; Fehm, T.; Neubauer, H. Diagnostic Leukapheresis Enables Reliable Transcriptomic Profiling of Single Circulating Tumor Cells to Characterize Inter-Cellular Heterogeneity in Terms of Endocrine Resistance. Cancers 2019, 11, 903. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tamminga, M.; Andree, K.C.; Hiltermann, T.J.N.; Jayat, M.; Schuuring, E.; van den Bos, H.; Spierings, D.C.J.; Lansdorp, P.M.; Timens, W.; Terstappen, L.; et al. Detection of Circulating Tumor Cells in the Diagnostic Leukapheresis Product of Non-Small-Cell Lung Cancer Patients Comparing CellSearch(®) and ISET. Cancers 2020, 12, 896. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Scherag, F.D.; Niestroj-Pahl, R.; Krusekopf, S.; Lücke, K.; Brandstetter, T.; Rühe, J. Highly Selective Capture Surfaces on Medical Wires for Fishing Tumor Cells in Whole Blood. Anal. Chem. 2017, 89, 1846–1854. [Google Scholar] [CrossRef]
- Dizdar, L.; Fluegen, G.; van Dalum, G.; Honisch, E.; Neves, R.P.; Niederacher, D.; Neubauer, H.; Fehm, T.; Rehders, A.; Krieg, A.; et al. Detection of circulating tumor cells in colorectal cancer patients using the GILUPI CellCollector: Results from a prospective, single-center study. Mol. Oncol. 2019, 13, 1548–1558. [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] [Green Version]
- Fachin, F.; Spuhler, P.; Martel-Foley, J.M.; Edd, J.F.; Barber, T.A.; Walsh, J.; Karabacak, M.; Pai, V.; Yu, M.; Smith, K.; et al. Monolithic Chip for High-throughput Blood Cell Depletion to Sort Rare Circulating Tumor Cells. Sci. Rep. 2017, 7, 10936. [Google Scholar] [CrossRef]
- Eifler, R.L.; Lind, J.; Falkenhagen, D.; Weber, V.; Fischer, M.B.; Zeillinger, R. Enrichment of circulating tumor cells from a large blood volume using leukapheresis and elutriation: Proof of concept. Cytom. Part B Clin. Cytom. 2011, 80B, 100–111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mutlu, B.R.; Smith, K.C.; Edd, J.F.; Nadar, P.; Dlamini, M.; Kapur, R.; Toner, M. Non-equilibrium Inertial Separation Array for High-throughput, Large-volume Blood Fractionation. Sci. Rep. 2017, 7, 9915. [Google Scholar] [CrossRef] [Green Version]
- Thery, L.; Meddis, A.; Cabel, L.; Proudhon, C.; Latouche, A.; Pierga, J.-Y.; Bidard, F.-C. Circulating Tumor Cells in Early Breast Cancer. JNCI Cancer Spectr. 2019, 3. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed] [Green Version]
- Edd, J.F.; Mishra, A.; Dubash, T.D.; Herrera, S.; Mohammad, R.; Williams, E.K.; Hong, X.; Mutlu, B.R.; Walsh, J.R.; Machado de Carvalho, F.; et al. Microfluidic concentration and separation of circulating tumor cell clusters from large blood volumes. Lab Chip 2020, 20, 558–567. [Google Scholar] [CrossRef] [PubMed]
- Ortiz, V.; Yu, M. Analyzing Circulating Tumor Cells One at a Time. Trends Cell Biol. 2018, 28, 764–775. [Google Scholar] [CrossRef]
- Hu, P.; Zhang, W.; Xin, H.; Deng, G. Single Cell Isolation and Analysis. Front. Cell Dev. Biol. 2016, 4, 116. [Google Scholar] [CrossRef] [Green Version]
- Donato, C.; Szczerba, B.M.; Scheidmann, M.C.; Castro-Giner, F.; Aceto, N. Micromanipulation of Circulating Tumor Cells for Downstream Molecular Analysis and Metastatic Potential Assessment. JoVE 2019, 147, e59677. [Google Scholar] [CrossRef] [PubMed]
- Nelep, C.; Eberhardt, J. Automated rare single cell picking with the ALS cellcelector™. Cytom. Part A 2018, 93, 1267–1270. [Google Scholar] [CrossRef] [Green Version]
- Neumann, M.H.; Schneck, H.; Decker, Y.; Schömer, S.; Franken, A.; Endris, V.; Pfarr, N.; Weichert, W.; Niederacher, D.; Fehm, T.; et al. Isolation and characterization of circulating tumor cells using a novel workflow combining the CellSearch(®) system and the CellCelector(™). Biotechnol. Prog. 2017, 33, 125–132. [Google Scholar] [CrossRef]
- Lohr, J.G.; Adalsteinsson, V.A.; Cibulskis, K.; Choudhury, A.D.; Rosenberg, M.; Cruz-Gordillo, P.; Francis, J.M.; Zhang, C.Z.; Shalek, A.K.; Satija, R.; et al. Whole-exome sequencing of circulating tumor cells provides a window into metastatic prostate cancer. Nat. Biotechnol. 2014, 32, 479–484. [Google Scholar] [CrossRef] [Green Version]
- Yao, X.; Williamson, C.; Adalsteinsson, V.A.; D’Agostino, R.S.; Fitton, T.; Smaroff, G.G.; William, R.T.; Wittrup, K.D.; Love, J.C. Tumor cells are dislodged into the pulmonary vein during lobectomy. J. Thorac. Cardiovasc. Surg. 2014, 148, 3224–3231. [Google Scholar] [CrossRef] [Green Version]
- Lampignano, R.; Yang, L.; Neumann, M.H.D.; Franken, A.; Fehm, T.; Niederacher, D.; Neubauer, H. A Novel Workflow to Enrich and Isolate Patient-Matched EpCAM(high) and EpCAM(low/negative) CTCs Enables the Comparative Characterization of the PIK3CA Status in Metastatic Breast Cancer. Int. J. Mol. Sci. 2017, 18, 1885. [Google Scholar] [CrossRef] [Green Version]
- Fontana, F.; Rapone, C.; Bregola, G.; Aversa, R.; de Meo, A.; Signorini, G.; Sergio, M.; Ferrarini, A.; Lanzellotto, R.; Medoro, G.; et al. Isolation and genetic analysis of pure cells from forensic biological mixtures: The precision of a digital approach. Forensic Sci. Int. Genet. 2017, 29, 225–241. [Google Scholar] [CrossRef] [Green Version]
- Di Trapani, M.; Manaresi, N.; Medoro, G. DEPArray™ system: An automatic image-based sorter for isolation of pure circulating tumor cells. Cytom. Part A 2018, 93, 1260–1266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gambari, R.; Borgatti, M.; Altomare, L.; Manaresi, N.; Medoro, G.; Romani, A.; Tartagni, M.; Guerrieri, R. Applications to cancer research of “lab-on-a-chip” devices based on dielectrophoresis (DEP). Technol. Cancer Res. Treat. 2003, 2, 31–40. [Google Scholar] [CrossRef] [PubMed]
- Polzer, B.; Medoro, G.; Pasch, S.; Fontana, F.; Zorzino, L.; Pestka, A.; Andergassen, U.; Meier-Stiegen, F.; Czyz, Z.T.; Alberter, B.; et al. Molecular profiling of single circulating tumor cells with diagnostic intention. EMBO Mol. Med. 2014, 6, 1371–1386. [Google Scholar] [CrossRef]
- 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] [PubMed]
- Hyun, K.A.; Koo, G.B.; Han, H.; Sohn, J.; Choi, W.; Kim, S.I.; Jung, H.I.; Kim, Y.S. Epithelial-to-mesenchymal transition leads to loss of EpCAM and different physical properties in circulating tumor cells from metastatic breast cancer. Oncotarget 2016, 7, 24677–24687. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bednarz-Knoll, N.; Alix-Panabières, C.; Pantel, K. Plasticity of disseminating cancer cells in patients with epithelial malignancies. Cancer Metastasis Rev. 2012, 31, 673–687. [Google Scholar] [CrossRef] [PubMed]
Subcategory | Name | Capture Efficiency (%) | Recovery Rate (%) | Advantages | Disadvantages |
---|---|---|---|---|---|
Immunomagnetic enrichment | |||||
Immunomagnetic positive enrichment | CellSearch [3,4,5,6,7] | 42–90 | Semi automated Can process up to 8 samples at a time In device staining CTC enumeration via CellTracks Analyser FDA approved | Recovery of EpCAM+ CTCs only Only able to detect CTCs expressing high levels of EpCAM | |
MACS [8,9] | 25–90 | Cocktail of antibodies available to increase CTC capture Able to process up to 15 mL blood Easy elution of CTCs Pro Separator can process up to 6 samples at once | Recovery of EpCAM+ CTCs only Suggested the MACS system is better suited for tissue samples | ||
MagSweeper [10,11] | 60–70 | Nonadherent plastic sleeves allow for multiple rounds of capture to increase capture efficiency | Recovery of EpCAM+ CTCs only | ||
Strep-tag [12,13] | 79–86 | 70 | Easy release of CTCs by simple addition of d-biotin Possibility to use a cocktail of antibodies to increase capture | Recovery of EpCAM+ CTCs only | |
IMS [14] | 92 | Leukocytes repelled so high purity recoveries | Recovery of EpCAM+ CTCs only Not yet tested on patient samples | ||
Immunomagnetic negative enrichment | EasySep [15,16] | 19–65 | Recovery of heterogeneous population of CTCs | Exclusion of CTC-WBC clusters Variable recovery rates May inadvertantly remove CTCs | |
RosetteSep [17] | 62.5 | Recovery of heterogeneous population of CTCs Cocktail of antibodies used to maximise depletion | Exclusion of CTC-WBC clusters, May inadvertantly remove CTCs | ||
Microfluidic immunocapture positive enrichment | CTC-Chip [18,19] | >60 | Large surface area for CTC capture High viability of recovered cells | Recovery of EpCAM+ CTCs only Slow processing rate Complex geometry of chip difficult to scale up Geometry prevents passage of CTC clusters | |
HB-chip [20] | 74.5–97 | HB grooves increase CTC-antibody contact for increased cell capture | Recovery of EpCAM+ CTCs only | ||
GEDI chip [21] | 80–90 | Large surface area for CTC capture Possibility to functionalise with alternative antibodies | May miss heterogeneity of CTCs | ||
HTMSU [22] | >97 | Quick processing On-chip single-cell conductometric counting for enumeration | Recovery of EpCAM+ CTCs only | ||
Nanovelcro [23] | 70–95 | 4 generations developed for different clinical utilities 3rd and 4th generation chips adapted for easy CTC release | Recovery of EpCAM+ CTCs only | ||
Isoflux [4] | 74–90 | 64–75 | Utilises microfluidic approach to increase EpCAM sensitivity Up to 4 samples can be processed in parallel Multiple kits including cocktails of antibodies to capture heterogeneity IsoFlux Cytation Imager for sample scanning | ||
Capture enhancement by nanomaterials | NP-HBCTC-Chip [24] | 79–97 | Simple release of CTCs by addition of glutathione (GSH) Chip surface can be functionalised with a cocktail of antibodies for enhanced capture efficiency | Recovery of EpCAM+ CTCs only Very low throughput | |
GO chip [25,26] | 67–100 | 91–95 | Simple chip design Large surface area for increased CTC capture | Recovery of EpCAM+ CTCs only | |
SiNP [27] | 84–91 | Large surface area for CTC capture | Recovery of EpCAM+ CTCs only | ||
Capture enhancement by nanomaterials | Nanotube-CTC-chip [28] | 89–100 | Preferential adherence negates need for EpCAM antibodies Planar enrichment surface makes chip visualisation and imaging easy | Time taken for optimal CTC adherence to substrate is too long | |
Size based enrichment | |||||
Membrane filtration | FMSA [29] | 90 | Recovery of heterogeneous population of CTCs Cheap and easy to produce Quick processing time | Filter clogging highly likely | |
ScreenCell [30] | 74–91 | Recovery of heterogeneous population of CTCs Cheap and easy to produce Three different devices offered depending on downstream requirements Quick processing time | Unevenly distributed or fused pores can reduce capture efficiency | ||
ISET [31,32] | 83–100 | Recovery of heterogeneous population of CTCs Cheap and easy to produce Ability to process 12 samples in parallel | Slow processing time Blood must be diluted 1:10 to prevent membrane clogging | ||
SB microfilter [33] | 78–83 | Recovery of heterogeneous population of CTCs Cheap and easy to produce Quick processing time | Only 1 mL blood can be processed at a time due to device clogging | ||
FAST [34] | 94–98 | Recovery of heterogeneous population of CTCs Cheap and easy to produce Quick processing time | |||
Microfluidics | Parsortix [35] | 42–70 | 54–69 | Recovery of heterogeneous population of CTCs Ability to capture CTC clusters Option for on-chip staining | Slow processing time On-chip imaging difficult |
MCA [36] | >90 | 68–100 | Recovery of heterogeneous population of CTCs Option for on-chip staining Ability to process up to 4 samples in parallel | ||
ClearCell FX1 [37,38] | 52–79 | Recovery of heterogeneous population of CTCs Quick processing time No channel clogging observed | |||
Vortex VTX-1 [39,40] | 53.8–71.6 | Recovery of heterogeneous population of CTCs Filters at channel inlet prevent channel clogging Fully automated process Quick processing time Associated BioView for enumeration Option to run in “high recovery” or “high purity” mode | |||
p-MOFF [41] | 91.6–93.75 | Recovery of heterogeneous population of CTCs Quick processing time No channel clogging observed | RBC lysis and Ficoll density centrifugation required | ||
Density based | OncoQuick [42,43] | 25–87 | Recovery of heterogeneous population of CTCs Up to 25 mL blood can be processed per tube | Low detection and recoveryrates | |
AccuCyte [44] | 81–90.5 | Recovery of heterogeneous population of CTCs Allows for processing of multiple samples in parallel Associated CyteFinder and CytePicker systems for imaging and mechanical selection of CTCs | |||
Other | |||||
Dielectrophoresis | ApoStream [45,46] | 55–78.5 | Recovery of heterogeneous population of CTCs Quick processing time iCys laser scanning cytometer for enumeration High viability of recovered cells | ||
In vivo | Diagnostic leukapheresis (DLA) [47] | Recovery of heterogeneous population of CTCs Recovery of much greater numbers of CTCs | Only a pre-enrichment step so must be used in combination with another enrichment technology Huge leukocyte background | ||
GILUPI CellCollector [48] | Potential for much greater numbers recovered | More invasive for the patient than a simple blood draw Recovery of EpCAM+ CTCs only | |||
Combined | CTC-iChip [49] | 70–100 | Option for positive or negative enrichment approach Inertial focusing provides high sensitivity selection Quick processing time | Positive enrichment only allows for recovery of EpCAM+ CTCs Negative enrichment will exclude CTC-WBC clusters | |
LPCTC-iChip [50] | 85.5–100 | Potential for much greater numbers recovered Magnetic field directs WBCs to centre of channel to prevent channel clogging Extremely high throughput | Disregards CTC-WBC clusters Initial debulking step may result in CTC loss | ||
OPENchip [51] | 50 | Chip allows for CTC enrichment and on-chip downstream molecular analysis | Low throughput, low recovery rates |
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 (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Rushton, A.J.; Nteliopoulos, G.; Shaw, J.A.; Coombes, R.C. A Review of Circulating Tumour Cell Enrichment Technologies. Cancers 2021, 13, 970. https://doi.org/10.3390/cancers13050970
Rushton AJ, Nteliopoulos G, Shaw JA, Coombes RC. A Review of Circulating Tumour Cell Enrichment Technologies. Cancers. 2021; 13(5):970. https://doi.org/10.3390/cancers13050970
Chicago/Turabian StyleRushton, Amelia J., Georgios Nteliopoulos, Jacqueline A. Shaw, and R. Charles Coombes. 2021. "A Review of Circulating Tumour Cell Enrichment Technologies" Cancers 13, no. 5: 970. https://doi.org/10.3390/cancers13050970
APA StyleRushton, A. J., Nteliopoulos, G., Shaw, J. A., & Coombes, R. C. (2021). A Review of Circulating Tumour Cell Enrichment Technologies. Cancers, 13(5), 970. https://doi.org/10.3390/cancers13050970