Relevance of Circulating Tumor Cells as Predictive Markers for Cancer Incidence and Relapse
Abstract
:1. Introduction
2. Enrichment and Isolation of CTCs
3. Clinical Utility of CTCs as a Biomarker in Solid Tumors
3.1. CTC Enumeration as a Predictive or Pharmacodynamics/Response Biomarker for Response to Therapy
3.2. Molecular Analysis of CTCs to Identify Their Role as a Predictive Biomarker
3.2.1. HER2+ CTCs as a Molecular Biomarker for Treatment Prediction
3.2.2. EGFR Expression on CTCs
3.2.3. AR-V7 Expression on CTCs
3.2.4. MET Expression on CTCs
3.2.5. PD-L1 Expression on CTCs
3.3. Genomic Analysis of CTCs for Their Role as a Predictive Biomarker
Trial | Disease Condition | Purpose | Phase | Trial Result | Ref. |
---|---|---|---|---|---|
REMAGUS-02 | Localized HER2+ mBC | Determine if CTCs were present in patients receiving neoadjuvant chemotherapy before initiation of chemotherapy and at the end of chemotherapy before surgery | III | CTCs can be detected in the blood of patients with large operable or locally advanced breast cancers before initiation of neo-adjuvant chemotherapy and can be monitored during treatment | [79] |
DETECT-III NCT01619111 | HER2− mBC | Evaluate the efficacy of HER2-targeted therapy in patients with mBC and HER2 positive CTCs | III | HER2 positive CTCs can be detected in a relevant number of patients with HER2 negative tumors and will be mandatory to correlate the assay-dependent HER2 status of CTCs to clinical response on HER2 targeted therapies | [134] |
SWOG S0500 NCT00382018 | mBC | Study treatment decision-making based on blood levels of tumor cells in women with mBC receiving chemotherapy | III | Confirmed the prognostic significance of CTCs in patients with mBC receiving first-line chemotherapy. Early switching to an alternative cytotoxic therapy was not effective in prolonging OS | [90] |
TREAT CTC NCT01548677 | HER2− mBC | Efficacy study of Herceptin to treat HER2-negative CTC breast cancer | II | Real-time screening of patients with early breast cancer for CTCs is feasible (pilot phase results) | [138] |
STIC-CTC NCT01710605 | HR+ HER2− mBC | Analyze the CTC count (CellSearch®)-driven first-line treatment choice | III | CTC count may be a reliable biomarker method for guiding the treatment choice between chemotherapy and endocrine therapy as the first-line treatment choice | [114] |
Circe T-DM1 NCT01975142 | HER2− mBC | Evaluate the validity of HER2-amplified CTCs to select mBC considered HER2− for trastuzumab-emtansine treatment | II | CTCs with HER2 amplification can be detected in a limited subset of HER2− mBC and treatment with T-DM1 achieved partial response | [137] |
CABAV7 NCT03050866 | mCRPC | Efficacy of cabazitaxel in mCRPC patients with AR-V7+ CTC | II | Awaited (prospective validation is needed to investigate if AR-V7 could fulfil the criteria as a predictive biomarker) | [169] |
PROPHECY NCT02269982 | mCRPC | Evaluate circulating tumor-derived products (CTCs) as a prospective predictor in higher risk mCRPC in the context of AR-directed therapies | NA | Detection of AR-V7 in CTCs is associated with shorter PFS and OS with abiraterone or enzalutamide, and such mCRPC patients should be offered alternative treatments | [170] |
PERSEVERE NCT04849364 | TNBC | Evaluated the association of ctDNA and CTCs after neoadjuvant chemotherapy with disease recurrence | II | Detection of ctDNA and CTCs in patients with early stage TNBC after neoadjuvant chemotherapy was independently associated with disease recurrence | [81] |
CARD NCT02485691 | mCRPC | Evaluated the impact of CTC morphologic subtypes prior to treatment in CARD trial | IV | Presence of chromosome instability at screening is associated with increased CTC morphological diversity, and also had poor rPFS and OS, when treated with cabazitaxel | [220] |
CTCNeoBC NCT03732339 | Locally advanced BC | Evaluate the predictive value of CTC using GILUPI cell collector | NA | Awaited | NA |
PROLIPSY NCT04556916 | PC | Assess the validity of CTC and tumor cell products for early prostate cancer detection | NA | Awaited (Not yet recruiting) | NA |
I-CURE NCT03213041 | HER2− mBC | Evaluate the impact on PFS with carboplatin-pembrolizumab combination in patients with CTC-positive HER2-negative mBC previously treated with anthracyclines and taxanes | II | Awaited (Recruiting) | NA |
IMMUNO-PREDICT NCT02827344 | LC | Demonstrate the feasibility of the analysis of PD-L1 expression on CTCs | NA | Awaited (Recruiting) | NA |
Companion Diagnostic Test | Drugs | Disease Type/ Sample Type | Mutation | Technique for Mutation Detection | Manufacturer |
---|---|---|---|---|---|
cobas EGFR mutation test | erlotinib (Tarceva), osimertinib (Tagrisso), gefitinib (Iressa) | NSCLC/plasma | EGFR mutations (exon 19 deletions, L858R mutation), T790M mutation | Real time polymerase chain reaction (RT-PCR) | Roche |
Guardant 360 CDx | osimertinib (Tagrisso) | NSCLC/lasma | EGFR mutation | Next-generation sequencing | Guardant |
amivantamab (Rybrevant) | EGFR mutation (exon 20 mutation) | ||||
FoundationOne Liquid CDx | rucaparib (Rubarca) | Ovarian cancer/plasma | BRCA1, BRCA2 | Next-generation sequencing | Foundation One |
alectinib (Alecensa) | NSCLC | ALK rearrangement | |||
alpelsib (Piqray) | Breast cancer | PIK3CA | |||
olaparib (Lynparza) | mCRPC/plasma | BRCA1, BRCA2, ATM | |||
rucaparib (Rubarca), AR-directed therapy, Taxane | mCRPC/plasma | BRCA1, BRCA2 | |||
gefitinib (Iressa), osimertinib (Tagrisso), erlotinib (Tarceva) | Lung cancer | EGFR mutations (exon 19 deletions, L858R mutation) | |||
capmatinib (Tabrecta, Novartis) | NSCLC | MET (exon 14 mutations) | |||
therascreen PIK3CA RGQ PCR Kit | alpelsib (Piqray) | Breast cancer/plasma | PIK3CA | Real-time polymerase chain reaction (RT-PCR) | Qiagen GmbH |
ArcherDx’s ArcherMET Assay | tepotinib (Tepmetko, Merck KGaA) | NSCLC/plasma | MET (exon 14 skipping) | Next-generation sequencing | Archer |
4. Limitations in CTC-Based Studies
5. Conclusions and Future Perspectives
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Brown, M.V.; McDunn, J.E.; Gunst, P.R.; Smith, E.M.; Milburn, M.V.; Troyer, D.A.; Lawton, K.A. Cancer detection and biopsy classification using concurrent histopathological and metabolomic analysis of core biopsies. Genome Med. 2012, 4, 33. [Google Scholar] [CrossRef] [Green Version]
- Ilie, M.; Hofman, P. Pros: Can tissue biopsy be replaced by liquid biopsy? Transl. Lung Cancer Res. 2016, 5, 420–423. [Google Scholar] [CrossRef] [Green Version]
- Ashworth, T.R. A case of cancer in which cells similar to those in the tumors were seen in the blood after death. Med. J. Aust. 1869, 14, 146–147. [Google Scholar]
- Ignatiadis, M.; Rack, B.; Rothe, F.; Riethdorf, S.; Decraene, C.; Bonnefoi, H.; Dittrich, C.; Messina, C.; Beauvois, M.; Trapp, E.; et al. Liquid biopsy-based clinical research in early breast cancer: The EORTC 90091-10093 Treat CTC trial. Eur. J. Cancer 2016, 63, 97–104. [Google Scholar] [CrossRef]
- Alix-Panabieres, C.; Pantel, K. Clinical Applications of Circulating Tumor Cells and Circulating Tumor DNA as Liquid Biopsy. Cancer Discov. 2016, 6, 479–491. [Google Scholar] [CrossRef] [Green Version]
- Dive, C.; Brady, G. SnapShot: Circulating Tumor Cells. Cell 2017, 168, 742–742.e1. [Google Scholar] [CrossRef]
- Amin, M.B.; Greene, F.L.; Edge, S.B.; Compton, C.C.; Gershenwald, J.E.; Brookland, R.K.; Meyer, L.; Gress, D.M.; Byrd, D.R.; Winchester, D.P. The Eighth Edition AJCC Cancer Staging Manual: Continuing to build a bridge from a population-based to a more “personalized” approach to cancer staging. CA Cancer J. Clin. 2017, 67, 93–99. [Google Scholar] [CrossRef] [PubMed]
- Lakhani, S.R.; Ellis, I.O.; Schnitt, S.J.; Tan, P.H.; van de Vijver, M.J. (Eds.) WHO Classification of Tumors of Breast, 4th ed.; IARC Press: Lyon, France, 2012. [Google Scholar]
- Banko, P.; Lee, S.Y.; Nagygyorgy, V.; Zrinyi, M.; Chae, C.H.; Cho, D.H.; Telekes, A. Technologies for circulating tumor cell separation from whole blood. J. Hematol. Oncol. 2019, 12, 48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cortes-Hernandez, L.E.; Eslami, S.Z.; Pantel, K.; Alix-Panabieres, C. Molecular and Functional Characterization of Circulating Tumor Cells: From Discovery to Clinical Application. Clin. Chem. 2020, 66, 97–104. [Google Scholar] [CrossRef]
- Cho, P.Y.; Hsieh, C.H.; Wu, M.H. The combination of Immunomagnetic Bead-based Cell isolation and Optically induced Dielectrophoresis (ODEP)-based microfluidic device for the negative-selection based isolation of circulating tumor cells. Front. Bioeng. Biotechnol. 2020, 8, 921. [Google Scholar] [CrossRef]
- 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]
- Riethdorf, S.; Fritsche, H.; Muller, V.; Rau, T.; Schindlbeck, C.; Rack, B.; Janni, W.; Coith, C.; Beck, K.; Janicke, 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]
- Deng, G.; Krishnakumar, S.; Powell, A.A.; Zhang, H.; Mindrinos, M.N.; Telli, M.L.; Davis, R.W.; Jeffrey, S.S. Single cell mutational analysis of PIK3CA in circulating tumor cells and metastases in breast cancer reveals heterogeneity, discordance, and mutation persistence in cultured disseminated tumor cells from bone marrow. BMC Cancer 2014, 14, 456. [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] [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] [Green Version]
- Chelakkot, C.; Ryu, J.; Kim, M.Y.; Kim, J.S.; Kim, D.; Hwang, J.; Park, S.H.; Ko, S.B.; Park, J.W.; Jung, M.Y.; et al. An Immune-Magnetophoretic Device for the Selective and Precise Enrichment of Circulating Tumor Cells from Whole Blood. Micromachines 2020, 11, 560. [Google Scholar] [CrossRef]
- 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]
- Earhart, C.M.; Hughes, C.E.; Gaster, R.S.; Ooi, C.C.; Wilson, R.J.; Zhou, L.Y.; Humke, E.W.; Xu, L.; Wong, D.J.; Willingham, S.B.; et al. Isolation and mutational analysis of circulating tumor cells from lung cancer patients with magnetic sifters and biochips. Lab Chip 2014, 14, 78–88. [Google Scholar] [CrossRef] [Green Version]
- Murlidhar, V.; Zeinali, M.; Grabauskiene, S.; Ghannad-Rezaie, M.; Wicha, M.S.; Simeone, D.M.; Ramnath, N.; Reddy, R.M.; Nagrath, S. A radial flow microfluidic device for ultra-high-throughput affinity-based isolation of circulating tumor cells. Small 2014, 10, 4895–4904. [Google Scholar] [CrossRef] [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]
- 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] [PubMed]
- 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]
- 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]
- Dharmasiri, U.; Njoroge, S.K.; Witek, A.M.; Adebiyi, M.G.; Kamande, J.W.; Hupert, M.L.; Barany, F.; Soper, S.A. High-throughput selection, enumeration, electrokinetic manipulation, and molecular profiling of low-abundance circulating tumor cells using a microfluidic system. Anal. Chem. 2011, 15, 2301–2309. [Google Scholar] [CrossRef] [Green Version]
- 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]
- 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] [Green Version]
- Stott, S.L.; Lee, R.J.; Nagrath, S.; Yu, M.; Miyamoto, D.T.; Ulkus, L.; Inserra, E.J.; Ulman, M.; Springer, S.; Nakamura, Z.; et al. Isolation and characterization of circulating tumor cells from patients with localized and metastatic prostate cancer. Sci. Transl. Med. 2010, 2, 25ra23. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Hofman, V.; Ilie, M.I.; Long, E.; Selva, E.; Bonnetaud, C.; Molina, T.; Venissac, N.; Mouroux, J.; Vielh, P.; Hofman, P. Detection of circulating tumor cells as a prognostic factor in patients undergoing radical surgery for non-small-cell lung carcinoma: Comparison of the efficacy of the CellSearch Assay and the isolation by size of epithelial tumor cell method. Int. J. Cancer 2011, 129, 1651–1660. [Google Scholar] [CrossRef]
- Hofman, V.; Long, E.; Ilie, M.; Bonnetaud, C.; Vignaud, J.M.; Flejou, J.F.; Lantuejoul, S.; Piaton, E.; Mourad, N.; Butori, C.; et al. Morphological analysis of circulating tumour cells in patients undergoing surgery for non-small cell lung carcinoma using the isolation by size of epithelial tumour cell (ISET) method. Cytopathology 2012, 23, 30–38. [Google Scholar] [CrossRef] [PubMed]
- 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]
- 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]
- Freidin, M.B.; Tay, A.; Freydina, D.V.; Chudasama, D.; Nicholson, A.G.; Rice, A.; Anikin, V.; Lim, E. An assessment of diagnostic performance of a filter-based antibody-independent peripheral blood circulating tumour cell capture paired with cytomorphologic criteria for the diagnosis of cancer. Lung Cancer 2014, 85, 182–185. [Google Scholar] [CrossRef] [PubMed]
- Khoo, B.L.; Warkiani, M.E.; Tan, D.S.; Bhagat, A.A.; Irwin, D.; Lau, D.P.; Lim, A.S.; Lim, K.H.; Krisna, S.S.; Lim, W.T.; et al. Clinical validation of an ultra high-throughput spiral microfluidics for the detection and enrichment of viable circulating tumor cells. PLoS ONE 2014, 9, e99409. [Google Scholar] [CrossRef] [Green Version]
- Lee, Y.; Guan, G.; Bhagat, A.A. ClearCell FX, a label-free microfluidics technology for enrichment of viable circulating tumor cells. Cytometry 2018, 93A, 1251–1254. [Google Scholar] [CrossRef] [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] [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] [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]
- Gorges, T.M.; Penkalla, N.; Schalk, T.; Joosse, S.A.; Riethdorf, S.; Tucholski, J.; Lucke, K.; Wikman, H.; Jackson, S.; Brychta, N.; et al. Enumeration and Molecular Characterization of Tumor Cells in Lung Cancer Patients Using a Novel In Vivo Device for Capturing Circulating Tumor Cells. Clin. Cancer Res. 2016, 22, 2197–2206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, D.; Ge, C.; Liang, W.; Yang, Q.; Liu, Q.; Ma, W.; Shi, L.; Wu, H.; Zhang, Y.; Wu, Z.; et al. In Vivo Enrichment and Elimination of Circulating Tumor Cells by Using a Black Phosphorus and Antibody Functionalized Intravenous Catheter. Adv. Sci. 2020, 7, 2000940. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Vermesh, O.; Aalipour, A.; Ge, T.J.; Saenz, Y.; Guo, Y.; Alam, I.S.; Park, S.M.; Adelson, C.N.; Mitsutake, Y.; Vilches-Moure, J.; et al. An intravascular magnetic wire for the high-throughput retrieval of circulating tumour cells in vivo. Nat. Biomed. Eng. 2018, 2, 696–705. [Google Scholar] [CrossRef] [PubMed]
- Belotti, Y.; Lim, C.T. Microfluidics for Liquid Biopsies: Recent Advances, Current Challenges, and Future Directions. Anal. Chem. 2021, 93, 4727–4738. [Google Scholar] [CrossRef]
- Wei, X.; Chen, K.; Guo, S.; Liu, W.; Zhao, X.Z. Emerging microfluidic technologies for the detection of circulating tumor cells and fetal nucleated red blood cells. ACS Appl. Bio Mater. 2021, 4, 1140–1155. [Google Scholar] [CrossRef]
- Cristofanilli, M.; Hayes, D.F.; Budd, G.T.; Ellis, M.J.; Stopeck, A.; Reuben, J.M.; Doyle, G.V.; Matera, J.; Allard, W.J.; Miller, M.C.; et al. Circulating tumor cells: A novel prognostic factor for newly diagnosed metastatic breast cancer. J. Clin. Oncol. 2005, 23, 1420–1430. [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]
- Muller, V.; Hayes, D.F.; Pantel, K. Recent translational research: Circulating tumor cells in breast cancer patients. Breast Cancer Res. 2006, 8, 110. [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.A.; Mitchell, E.; Miller, M.C.; et al. Prognostic significance of circulating tumor cells in patients with metastatic colorectal cancer. Ann. Oncol. 2009, 20, 1223–1229. [Google Scholar] [CrossRef]
- de Bono, J.S.; Scher, H.I.; Montgomery, R.B.; Parker, C.; Miller, M.C.; Tissing, H.; Doyle, G.V.; Terstappen, L.W.; Pienta, K.J.; Raghavan, D. Circulating tumor cells predict survival benefit from treatment in metastatic castration-resistant prostate cancer. Clin. Cancer Res. 2008, 14, 6302–6309. [Google Scholar] [CrossRef] [Green Version]
- Battula, V.L.; Evans, K.W.; Hollier, B.G.; Shi, Y.; Marini, F.C.; Ayyanan, A.; Wang, R.Y.; Brisken, C.; Guerra, R.; Andreeff, M.; et al. Epithelial-mesenchymal transition-derived cells exhibit multilineage differentiation potential similar to mesenchymal stem cells. Stem Cells 2010, 28, 1435–1445. [Google Scholar] [CrossRef] [Green Version]
- Chen, J.; Yu, L.; Camacho, J.L.C.; Chai, Y.; Li, D.; Li, Y.; Liu, H.; Ou, L.; Li, W.; Haag, R. Biospecific monolayer coating for multivalent capture of circulating tumor cells with high sensitivity. Adv. Funct. Mater. 2019, 29, 1903960. [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]
- 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]
- Kim, J.; Cho, H.; Han, S.I.; Han, K.H. Single-cell isolation of circulating tumor cells from whole blood by lateral magnetophoretic microseparation and microfluidic dispensing. Anal Chem. 2016, 88, 4857–4863. [Google Scholar] [CrossRef]
- Tang, M.; Wen, C.Y.; Wu, L.L.; Hong, S.L.; Hu, J.; Xu, C.M.; Pang, D.W.; Zhang, Z.L. A Chip assisted immunomagnetic separation system for the efficient capture and in situ identification of circulating tumor cells. Lab Chip 2016, 16, 1214–1223. [Google Scholar] [CrossRef] [PubMed]
- Qin, X.; Park, S.; Duffy, S.P.; Matthews, K.; Ang, R.R.; Todenhofer, T.; Abdi, H.; Azad, A.; Bazov, J.; Chi, K.N.; et al. Size and deformability based separation of circulating tumor cells from castrate resistant prostate cancer patients using resettable cell traps. Lab Chip 2015, 15, 2278–2286. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Sarioglu, A.F.; Aceto, N.; Kojic, N.; Donaldson, M.C.; Zeinali, M.; Hamza, B.; Engstrom, A.; Zhu, H.; Sundaresan, T.K.; Miyamoto, D.T.; et al. A microfluidic device for label-free, physical capture of circulating tumor cell clusters. Nat. Methods 2015, 12, 685–691. [Google Scholar] [CrossRef]
- Ahmed, M.G.; Abate, M.F.; Song, Y.; Zhu, Z.; Yan, F.; Xu, Y.; Wang, X.; Li, Q.; Yang, C. Isolation, Detection, and Antigen-Based Profiling of Circulating Tumor Cells Using a Size-Dictated Immunocapture Chip. Angew. Chem. Int. Ed. Engl. 2017, 56, 10681–10685. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed] [Green Version]
- Lin, E.; Rivera-Baez, L.; Fouladdel, S.; Yoon, H.J.; Guthrie, S.; Wieger, J.; Deol, Y.; Keller, E.; Sahai, V.; Simeone, D.M.; et al. High-Throughput Microfluidic Labyrinth for the Label-free Isolation of Circulating Tumor Cells. Cell Syst. 2017, 5, 295–304.e4. [Google Scholar] [CrossRef] [Green Version]
- 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. 2018, 23, 16–29. [Google Scholar] [CrossRef] [Green Version]
- Zheng, S.; Lin, H.K.; Lu, B.; Williams, A.; Datar, R.; Cote, R.J.; Tai, Y.C. 3D microfilter device for viable circulating tumor cell (CTC) enrichment from blood. Biomed. Microdevices 2011, 13, 203–213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hosokawa, M.; Yoshikawa, T.; Negishi, R.; Yoshino, T.; Koh, Y.; Kenmotsu, H.; Naito, T.; Takahashi, T.; Yamamoto, N.; Kikuhara, Y.; et al. Microcavity array system for size-based enrichment of circulating tumor cells from the blood of patients with small-cell lung cancer. Anal. Chem. 2013, 85, 5692–5698. [Google Scholar] [CrossRef]
- Di Carlo, D. Inertial microfluidics. Lab Chip 2009, 9, 3038–3046. [Google Scholar] [CrossRef]
- Di Carlo, D.; Irimia, D.; Tompkins, R.G.; Toner, M. Continuous inertial focusing, ordering, and separation of particles in microchannels. Proc. Natl. Acad. Sci. USA 2007, 104, 18892–18897. [Google Scholar] [CrossRef] [Green Version]
- Abdulla, A.; Liu, W.; Gholamipour-Shirazi, A.; Sun, J.; Ding, X. High-Throughput Isolation of Circulating Tumor Cells Using Cascaded Inertial Focusing Microfluidic Channel. Anal. Chem. 2018, 90, 4397–4405. [Google Scholar] [CrossRef]
- Kuntaegowdanahalli, S.S.; Bhagat, A.A.; Kumar, G.; Papautsky, I. Inertial microfluidics for continuous particle separation in spiral microchannels. Lab Chip 2009, 9, 2973–2980. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nivedita, N.; Garg, N.; Lee, A.P.; Papautsky, I. A high throughput microfluidic platform for size-selective enrichment of cell populations in tissue and blood samples. Analyst 2017, 142, 2558–2569. [Google Scholar] [CrossRef]
- Huang, L.R.; Cox, E.C.; Austin, R.H.; Sturm, J.C. Continuous particle separation through deterministic lateral displacement. Science 2004, 304, 987–990. [Google Scholar] [CrossRef]
- McGrath, J.; Jimenez, M.; Bridle, H. Deterministic lateral displacement for particle separation: A review. Lab Chip 2014, 14, 4139–4158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gascoyne, P.R.; Shim, S. Isolation of circulating tumor cells by dielectrophoresis. Cancers 2014, 6, 545–579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cappelletti, V.; Verzoni, E.; Ratta, R.; Vismara, M.; Silvestri, M.; Montone, R.; Miodini, P.; Reduzzi, C.; Claps, M.; Sepe, P.; et al. Analysis of Single Circulating Tumor Cells in Renal Cell Carcinoma Reveals Phenotypic Heterogeneity and Genomic Alterations Related to Progression. Int. J. Mol. Sci. 2020, 21, 1475. [Google Scholar] [CrossRef] [Green Version]
- Bidard, F.C.; Mathiot, C.; Delaloge, S.; Brain, E.; Giachetti, S.; de Cremoux, P.; Marty, M.; Pierga, J.Y. Single circulating tumor cell detection and overall survival in nonmetastatic breast cancer. Ann. Oncol. 2010, 21, 729–733. [Google Scholar] [CrossRef]
- Bidard, F.C.; Belin, L.; Delaloge, S.; Lerebours, F.; Ngo, C.; Reyal, F.; Alran, S.; Giacchetti, S.; Marty, M.; Lebofsky, R.; et al. Time-Dependent Prognostic Impact of Circulating Tumor Cells Detection in Non-Metastatic Breast Cancer: 70-Month Analysis of the REMAGUS02 Study. Int. J. Breast Cancer 2013, 2013, 130470. [Google Scholar] [CrossRef]
- Giacchetti, S.; Hamy, A.S.; Delaloge, S.; Brain, E.; Berger, F.; Sigal-Zafrani, B.; Mathieu, M.C.; Bertheau, P.; Guinebretiere, J.M.; Saghatchian, M.; et al. Long-term outcome of the REMAGUS 02 trial, a multicenter randomised phase II trial in locally advanced breast cancer patients treated with neoadjuvant chemotherapy with or without celecoxib or trastuzumab according to HER2 status. Eur. J. Cancer 2017, 75, 323–332. [Google Scholar] [CrossRef]
- Sparano, J.; O’Neill, A.; Alpaugh, K.; Wolff, A.C.; Northfelt, D.W.; Dang, C.T.; Sledge, G.W.; Miller, K.D. Association of Circulating Tumor Cells with Late Recurrence of Estrogen Receptor-Positive Breast Cancer: A Secondary Analysis of a Randomized Clinical Trial. JAMA Oncol. 2018, 4, 1700–1706. [Google Scholar] [CrossRef] [PubMed]
- Radovich, M.; Jiang, G.; Hancock, B.A.; Chitambar, C.; Nanda, R.; Falkson, C.; Lynce, F.C.; Gallagher, C.; Isaacs, C.; Blaya, M.; et al. Association of Circulating Tumor DNA and Circulating Tumor Cells After Neoadjuvant Chemotherapy with Disease Recurrence in Patients with Triple-Negative Breast Cancer: Preplanned Secondary Analysis of the BRE12-158 Randomized Clinical Trial. JAMA Oncol. 2020, 6, 1410–1415. [Google Scholar] [CrossRef]
- Heidrich, I.; Ackar, L.; Mossahebi Mohammadi, P.; Pantel, K. Liquid biopsies: Potential and challenges. Int. J. Cancer 2020, 148, 528–545. [Google Scholar] [CrossRef]
- Kelley, S.O.; Pantel, K. A New Era in Liquid Biopsy: From Genotype to Phenotype. Clin. Chem. 2020, 66, 89–96. [Google Scholar] [CrossRef]
- 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]
- 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]
- Larsson, A.M.; Jansson, S.; Bendahl, P.O.; Levin Tykjaer Jorgensen, C.; Loman, N.; Graffman, C.; Lundgren, L.; Aaltonen, K.; Ryden, L. Longitudinal enumeration and cluster evaluation of circulating tumor cells improve prognostication for patients with newly diagnosed metastatic breast cancer in a prospective observational trial. Breast Cancer Res. 2018, 20, 48. [Google Scholar] [CrossRef]
- 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]
- Basso, U.; Facchinetti, A.; Rossi, E.; Maruzzo, M.; Conteduca, V.; Aieta, M.; Massari, F.; Fraccon, A.P.; Mucciarini, C.; Sava, T.; et al. Prognostic Role of Circulating Tumor Cells in Metastatic Renal Cell Carcinoma: A Large, Multicenter, Prospective Trial. Oncologist 2021, 26, 740–750. [Google Scholar] [CrossRef] [PubMed]
- Cristofanilli, M.; Pierga, J.Y.; Reuben, J.; Rademaker, A.; Davis, A.A.; Peeters, D.J.; Fehm, T.; Nole, F.; Gisbert-Criado, R.; Mavroudis, D.; et al. The clinical use of circulating tumor cells (CTCs) enumeration for staging of metastatic breast cancer (MBC): International expert consensus paper. Crit. Rev. Oncol. Hematol. 2019, 134, 39–45. [Google Scholar] [CrossRef] [PubMed]
- Smerage, J.B.; Barlow, W.E.; Hortobagyi, G.N.; Winer, E.P.; Leyland-Jones, B.; Srkalovic, G.; Tejwani, S.; Schott, A.F.; O’Rourke, M.A.; Lew, D.L.; et al. Circulating tumor cells and response to chemotherapy in metastatic breast cancer: SWOG S0500. J. Clin. Oncol. 2014, 32, 3483–3489. [Google Scholar] [CrossRef]
- Pierga, J.Y.; Bidard, F.C.; Cropet, C.; Tresca, P.; Dalenc, F.; Romieu, G.; Campone, M.; Mahier Ait-Oukhatar, C.; Le Rhun, E.; Goncalves, A.; et al. Circulating tumor cells and brain metastasis outcome in patients with HER2-positive breast cancer: The LANDSCAPE trial. Ann. Oncol. 2013, 24, 2999–3004. [Google Scholar] [CrossRef] [PubMed]
- Giuliano, M.; Shaikh, A.; Lo, H.C.; Arpino, G.; De Placido, S.; Zhang, X.H.; Cristofanilli, M.; Schiff, R.; Trivedi, M.V. Perspective on Circulating Tumor Cell Clusters: Why It Takes a Village to Metastasize. Cancer Res. 2018, 78, 845–852. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Reduzzi, C.; Di Cosimo, S.; Gerratana, L.; Motta, R.; Martinetti, A.; Vingiani, A.; D’Amico, P.; Zhang, Y.; Vismara, M.; Depretto, C.; et al. Circulating Tumor Cell Clusters Are Frequently Detected in Women with Early-Stage Breast Cancer. Cancers 2021, 13, 2356. [Google Scholar] [CrossRef] [PubMed]
- Murlidhar, V.; Reddy, R.M.; Fouladdel, S.; Zhao, L.; Ishikawa, M.K.; Grabauskiene, S.; Zhang, Z.; Lin, J.; Chang, A.C.; Carrott, P.; et al. Poor Prognosis Indicated by Venous Circulating Tumor Cell Clusters in Early-Stage Lung Cancers. Cancer Res. 2017, 77, 5194–5206. [Google Scholar] [CrossRef] [Green Version]
- Jansson, S.; Bendahl, P.O.; Larsson, A.M.; Aaltonen, K.E.; Ryden, L. Prognostic impact of circulating tumor cell apoptosis and clusters in serial blood samples from patients with metastatic breast cancer in a prospective observational cohort. BMC Cancer 2016, 16, 433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hou, J.M.; Krebs, M.G.; Lancashire, L.; Sloane, R.; Backen, A.; Swain, R.K.; Priest, L.J.; Greystoke, A.; Zhou, C.; Morris, K.; et al. Clinical significance and molecular characteristics of circulating tumor cells and circulating tumor microemboli in patients with small-cell lung cancer. J. Clin. Oncol. 2012, 30, 525–532. [Google Scholar] [CrossRef]
- Wang, C.; Mu, Z.; Chervoneva, I.; Austin, L.; Ye, Z.; Rossi, G.; Palazzo, J.P.; Sun, C.; Abu-Khalaf, M.; Myers, R.E.; et al. Longitudinally collected CTCs and CTC-clusters and clinical outcomes of metastatic breast cancer. Breast Cancer Res. Treat. 2017, 161, 83–94. [Google Scholar] [CrossRef]
- Au, S.H.; Edd, J.; Stoddard, A.E.; Wong, K.H.K.; Fachin, F.; Maheswaran, S.; Haber, D.A.; Stott, S.L.; Kapur, R.; Toner, M. Microfluidic Isolation of Circulating Tumor Cell Clusters by Size and Asymmetry. Sci. Rep. 2017, 7, 2433. [Google Scholar] [CrossRef]
- Cheng, S.B.; Xie, M.; Chen, Y.; Xiong, J.; Liu, Y.; Chen, Z.; Guo, S.; Shu, Y.; Wang, M.; Yuan, B.F.; et al. Three-Dimensional Scaffold Chip with Thermosensitive Coating for Capture and Reversible Release of Individual and Cluster of Circulating Tumor Cells. Anal. Chem. 2017, 89, 7924–7932. [Google Scholar] [CrossRef]
- Kulasinghe, A.; Zhou, J.; Kenny, L.; Papautsky, I.; Punyadeera, C. Capture of Circulating Tumour Cell Clusters Using Straight Microfluidic Chips. Cancers 2019, 11, 89. [Google Scholar] [CrossRef] [Green Version]
- Wang, C.; Zhang, Z.; Chong, W.; Luo, R.; Myers, R.E.; Gu, J.; Lin, J.; Wei, Q.; Li, B.; Rebbeck, T.R.; et al. Improved Prognostic Stratification Using Circulating Tumor Cell Clusters in Patients with Metastatic Castration-Resistant Prostate Cancer. Cancers 2021, 13, 268. [Google Scholar] [CrossRef] [PubMed]
- Krol, I.; Schwab, F.D.; Carbone, R.; Ritter, M.; Picocci, S.; De Marni, M.L.; Stepien, G.; Franchi, G.M.; Zanardi, A.; Rissoglio, M.D.; et al. Detection of clustered circulating tumour cells in early breast cancer. Br. J. Cancer 2021, 125, 23–27. [Google Scholar] [CrossRef] [PubMed]
- Costa, C.; Muinelo-Romay, L.; Cebey-Lopez, V.; Pereira-Veiga, T.; Martinez-Pena, I.; Abreu, M.; Abalo, A.; Lago-Leston, R.M.; Abuin, C.; Palacios, P.; et al. Analysis of a Real-World Cohort of Metastatic Breast Cancer Patients Shows Circulating Tumor Cell Clusters (CTC-clusters) as Predictors of Patient Outcomes. Cancers 2020, 12, 1111. [Google Scholar] [CrossRef] [PubMed]
- Paoletti, C.; Miao, J.; Dolce, E.M.; Darga, E.P.; Repollet, M.I.; Doyle, G.V.; Gralow, J.R.; Hortobagyi, G.N.; Smerage, J.B.; Barlow, W.E.; et al. Circulating Tumor Cell Clusters in Patients with Metastatic Breast Cancer: A SWOG S0500 Translational Medicine Study. Clin. Cancer Res. 2019, 25, 6089–6097. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ried, K.; Eng, P.; Sali, A. Screening of Circulating Tumor Cells Allow Early Detection of Cancer and Monitoring of Treatment Effectiveness: An Observational Study. Asian Pac. J. Cancer Prev. 2017, 18, 2275–2285. [Google Scholar] [PubMed]
- Nimgaonkar, A.; Segurado, O.; Tsai, W.S.; Pang, S.T.; Hou, M.F.; Chang, Y.; Watson, D.; Chang, Y.H.; Lin, P.H.; Wu, J.C.; et al. A novel circulating utmor cell blood test for early detection of colorectal, prostate and breast cancers: Result from 709 samples. J. Clin. Oncol. 2018, 36, e13549. [Google Scholar] [CrossRef]
- Ludwig, J.A.; Weinstein, J.N. Biomarkers in Cancer Staging, Prognosis and Treatment Selection. Nat. Rev. Cancer 2005, 5, 845–856. [Google Scholar] [CrossRef]
- Sauter, E.R. Reliable Biomarkers to Identify New and Recurrent Cancer. Eur. J. Breat Health 2017, 13, 162–167. [Google Scholar] [CrossRef]
- Herrmann, K.; Walch, A.; Balluff, B.; Tanzer, M.; Hofler, H.; Krause, B.J.; Schwaiger, M.; Friess, H.; Schmid, R.M.; Ebert, M.P.A. Proteomic and metabolic prediction of response to therapy in gastrointestinal cancers. Nat. Rev. Gastroenterol. Hepatol. 2009, 6, 170–183. [Google Scholar] [CrossRef]
- Pantel, K.; Alix-Panabieres, C. Liquid biopsy and minimal residual disease-latest advances and implications for cure. Nat. Rev. Clin. Oncol 2019, 16, 409–424. [Google Scholar] [CrossRef]
- Goodman, C.R.; Seagle, B.L.; Friedl, T.W.P.; Rack, B.; Lato, K.; Fink, V.; Cristofanilli, M.; Donnelly, E.D.; Janni, W.; Shahabi, S.; et al. Association of Circulating Tumor Cell Status With Benefit of Radiotherapy and Survival in Early-Stage Breast Cancer. JAMA Oncol. 2018, 4, e180163. [Google Scholar] [CrossRef] [Green Version]
- Bidard, F.C.; Mathiot, C.; Degeorges, A.; Etienne-Grimaldi, M.C.; Delva, R.; Pivot, X.; Veyret, C.; Bergougnoux, L.; de Cremoux, P.; Milano, G.; et al. Clinical value of circulating endothelial cells and circulating tumor cells in metastatic breast cancer patients treated first line with bevacizumab and chemotherapy. Ann. Oncol. 2010, 21, 1765–1771. [Google Scholar] [CrossRef]
- Giuliano, M.; Giordano, A.; Jackson, S.; Hess, K.R.; De Giorgi, U.; Mego, M.; Handy, B.C.; Ueno, N.T.; Alvarez, R.H.; De Laurentiis, M.; et al. Circulating tumor cells as prognostic and predictive markers in metastatic breast cancer patients receiving first-line systemic treatment. Breast Cancer Res. 2011, 13, R67. [Google Scholar] [CrossRef] [Green Version]
- Bidard, F.C.; Jacot, W.; Kiavue, N.; Dureau, S.; Kadi, A.; Brain, E.; Bachelot, T.; Bourgeois, H.; Goncalves, A.; Ladoire, S.; et al. Efficacy of Circulating Tumor Cell Count-Driven vs Clinician-Driven First-line Therapy Choice in Hormone Receptor-Positive, ERBB2-Negative Metastatic Breast Cancer: The STIC CTC Randomized Clinical Trial. JAMA Oncol. 2020, 7, 34–41. [Google Scholar] [CrossRef] [PubMed]
- Punnoose, E.A.; Atwal, S.; Liu, W.; Raja, R.; Fine, B.M.; Hughes, B.G.; Hicks, R.J.; Hampton, G.M.; Amler, L.C.; Pirzkall, A.; et al. Evaluation of circulating tumor cells and circulating tumor DNA in non-small cell lung cancer: Association with clinical endpoints in a phase II clinical trial of pertuzumab and erlotinib. Clin. Cancer Res. 2012, 18, 2391–2401. [Google Scholar] [CrossRef] [Green Version]
- Hiltermann, T.J.N.; Pore, M.M.; van den Berg, A.; Timens, W.; Boezen, H.M.; Liesker, J.J.W.; Schouwink, J.H.; Wijnands, W.J.A.; Kerner, G.; Kruyt, F.A.E.; et al. Circulating tumor cells in small-cell lung cancer: A predictive and prognostic factor. Ann. Oncol. 2012, 23, 2937–2942. [Google Scholar] [CrossRef] [PubMed]
- Fu, L.; Liu, F.; Fu, H.; Liu, L.; Yuan, S.; Gao, Y.; Fu, Z.; Yu, J. Circulating tumor cells correlate with recurrence in stage III small-cell lung cancer after systemic chemoradiotherapy and prophylactic cranial irradiation. Jpn. J. Clin. Oncol. 2014, 44, 948–955. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fu, L.; Zhu, Y.; Jing, W.; Guo, D.; Kong, L.; Yu, J. Incorporation of circulating tumor cells and whole-body metabolic tumor volume of (18)F-FDG PET/CT improves prediction of outcome in IIIB stage small-cell lung cancer. Chin. J. Cancer Res. 2018, 30, 596–604. [Google Scholar] [CrossRef]
- Heller, G.; McCormack, R.; Kheoh, T.; Molina, A.; Smith, M.R.; Dreicer, R.; Saad, F.; de Wit, R.; Aftab, D.T.; Hirmand, M.; et al. Circulating Tumor Cell Number as a Response Measure of Prolonged Survival for Metastatic Castration-Resistant Prostate Cancer: A Comparison With Prostate-Specific Antigen Across Five Randomized Phase III Clinical Trials. J. Clin. Oncol. 2018, 36, 572–580. [Google Scholar] [CrossRef]
- Goldkorn, A.; Tangen, C.; Plets, M.; Morrison, G.J.; Cunha, A.; Xu, T.; Pinski, J.K.; Ingles, S.A.; Triche, T.; Harzstark, A.L.; et al. Baseline Circulating Tumor Cell Count as a Prognostic Marker of PSA Response and Disease Progression in Metastatic Castrate-Sensitive Prostate Cancer (SWOG S1216). Clin. Cancer Res. 2021, 27, 1967–1973. [Google Scholar] [CrossRef]
- Yu, M.; Bardia, A.; Aceto, N.; Bersani, F.; Madden, M.W.; Donaldson, M.C.; Desai, R.; Zhu, H.; Comaills, V.; Zheng, Z.; et al. Ex Vivo culture of circulating breast tumor cells for individualized testing of drug susceptibility. Science 2014, 345, 6193. [Google Scholar] [CrossRef] [Green Version]
- Lallo, A.; Schenk, M.W.; Frese, K.K.; Blackhall, F.; Dive, C. Circulating tumor cells and CDx models as a tool for preclinical drug development. Transl. Lung Cancer Res. 2017, 6, 397–408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Conteduca, V.; Ku, S.Y.; Fernandez, L.; Rodriquez, A.D.; Lee, J.; Jendrisak, A.; Slade, M.; Gilbertson, C.; Manohar, J.; Sigouros, M.; et al. Circulating tumor cell heterogeneity in neuroendocrine prostate cancer by single cell copy number analysis. NPJ Precis. Oncol. 2021, 5, 1–8. [Google Scholar]
- Kong, S.L.; Liu, X.; Tan, S.J.; Tai, J.A.; Phua, L.Y.; Poh, H.M.; Yeo, T.; Chua, Y.W.; Haw, Y.X.; Ling, W.H.; et al. Complementary Sequential Circulating Tumor Cell (CTC) and Cell-Free Tumor DNA (ctDNA) Profiling Reveals Metastatic Heterogeneity and Genomic Changes in Lung Cancer and Breast Cancer. Front. Oncol. 2021, 11, 698551. [Google Scholar] [CrossRef]
- Genna, A.; Vanwynsberghe, A.M.; Villard, A.V.; Pottier, C.; Ancel, J.; Polette, M.; Gilles, C. EMT-Associated Heterogeneity in Circulating Tumor Cells: Sticky Friends on the Road to Metastasis. Cancers 2020, 12, 1632. [Google Scholar] [CrossRef]
- 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]
- Koch, C.; Kuske, A.; Joosse, S.A.; Yigit, G.; Sflomos, G.; Thanler, S.; Smit, D.J.; Werner, S.; Borgmann, K.; Gartner, S.; et al. Characterization of circulating breast cancer cells with tumorigenic and metastatic capacity. EMBO Mol. Med. 2020, 12, e11908. [Google Scholar] [CrossRef]
- Castro-Giner, F.; Aceto, N. Tracking cancer progression: From circulating tumor cells to metastasis. Genome Med. 2020, 12, 31. [Google Scholar] [CrossRef] [Green Version]
- Franses, J.W.; Philipp, J.; Missios, P.; Bhan, I.; Liu, A.; Yashaswini, C.; Tai, E.; Zhu, H.; Ligorio, M.; Nicholson, B.; et al. Pancreatic circulating tumor cell profiling identifies LIN28B as a metastasis driver and drug target. Nat. Commun. 2020, 11, 3303. [Google Scholar]
- Pailler, E.; Faugeroux, V.; Oulhen, M.; Mezquita, L.; Laporte, M.; Honore, A.; Lecluse, Y.; queffelec, P.; NgoCamus, M.; Nicorta, C.; et al. Acquired Resistance Mutations to ALK inhibitors Identified by Single Circulating Tumor Cell Sequencing in ALK-Rearranged Non-Small-Cell Lung Cancer. Clin. Can. Res. 2019, 25, 6671–6682. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Medford, A.J.; Dubash, T.D.; Juric, D.; Spring, L.; Niemierko, A.; Vidula, N.; Peppercorn, J.; Isakoff, S.; Reeves, B.A.; LiCausi, J.A.; et al. Blood-based monitoring identifies acquired and targetable driver mutations in endocrine-resistant metastatic breasr cancer. Npj Precis. Oncol. 2019, 3, 18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, J.; Xu, B. Targeted therapeutic options and future perspectives for HER2-positive breast cancer. Signal Transduct. Target. Ther. 2019, 4, 34. [Google Scholar] [CrossRef] [Green Version]
- Beije, N.; Onstenk, W.; Kraan, J.; Sieuwerts, A.M.; Hamberg, P.; Dirix, L.Y.; Brouwer, A.; de Jongh, F.E.; Jager, A.; Seynaeve, C.M.; et al. Prognostic Impact of HER2 and ER Status of Circulating Tumor Cells in Metastatic Breast Cancer Patients with a HER2-Negative Primary Tumor. Neoplasia 2016, 18, 647–653. [Google Scholar] [CrossRef] [Green Version]
- Fehm, T.; Muller, V.; Aktas, B.; Janni, W.; Schneeweiss, A.; Stickeler, E.; Lattrich, C.; Lohberg, C.R.; Solomayer, E.; Rack, B.; et al. HER2 status of circulating tumor cells in patients with metastatic breast cancer: A prospective, multicenter trial. Breast Cancer Res. Treat. 2010, 124, 403–412. [Google Scholar] [CrossRef] [Green Version]
- Liu, Y.; Liu, Q.; Wang, T.; Bian, L.; Zhang, S.; Hu, H.; Li, S.; Hu, Z.; Wu, S.; Liu, B.; et al. Circulating tumor cells in HER2-positive metastatic breast cancer patients: A valuable prognostic and predictive biomarker. BMC Cancer 2013, 13, 202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, S.; Li, L.; Wang, T.; Bian, L.; Hu, H.; Xu, C.; Liu, B.; Liu, Y.; Cristofanilli, M.; Jiang, Z. Real-time HER2 status detected on circulating tumor cells predicts different outcomes of anti-HER2 therapy in histologically HER2-positive metastatic breast cancer patients. BMC Cancer 2016, 16, 526. [Google Scholar] [CrossRef] [Green Version]
- Jacot, W.; Cottu, P.; Berger, F.; Dubot, C.; Venat-Bouvet, L.; Lortholary, A.; Bourgeois, H.; Bollet, M.; Servent, V.; Luporsi, E.; et al. Actionability of HER2-amplified circulating tumor cells in HER2-negative metastatic breast cancer: The CirCe T-DM1 trial. Breast Cancer Res. 2019, 21, 121. [Google Scholar] [CrossRef]
- Ignatiadis, M.; Litiere, S.; Rothe, F.; Riethdorf, S.; Proudhon, C.; Fehm, T.; Aalders, K.; Forstbauer, H.; Fasching, P.A.; Brain, E.; et al. Trastuzumab versus observation for HER2 nonamplified early breast cancer with circulating tumor cells (EORTC 90091-10093, BIG 1-12, Treat CTC): A randomized phase II trial. Ann. Oncol. 2018, 29, 1777–1783. [Google Scholar] [CrossRef] [PubMed]
- Riethdorf, S.; Muller, 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]
- Pobiruchin, M.; Bochum, S.; Martens, U.M.; Kieser, M.; Schramm, W. Transition probabilities of HER2-positive and HER2-negative breast cancer patients treated with Trastuzumab obtained from a clinical cancer registry dataset. Data Brief. 2016, 7, 654–657. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Deutsch, T.M.; Riethdorf, S.; Fremd, C.; Feisst, M.; Nees, J.; Fischer, C.; Hartkopf, A.D.; Pantel, K.; Trumpp, A.; Schutz, F.; et al. HER2-targeted therapy influences CTC status in metastatic breast cancer. Breast Cancer Res. Treat. 2020, 182, 127–136. [Google Scholar] [CrossRef] [PubMed]
- Serrano, M.J.; Alvarez-Cubero, M.J.; De Miguel Perez, D.; Rodriguez-Martinez, A.; Gonzalez-Herrera, L.; Robles-Fernandez, I.; Hernandez, J.E.; Puche, J.L.G.; Lorente, J.A. Significance of EGFR Expression in Circulating Tumor Cells. Adv. Exp. Med. Biol. 2017, 994, 285–296. [Google Scholar]
- 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]
- Godin-Heymann, N.; Ulkus, L.; Brannigan, B.W.; McDermott, U.; Lamb, J.; Maheswaran, S.; Settleman, J.; Haber, D.A. The T790M “gatekeeper” mutation in EGFR mediates resistance to low concentrations of an irreversible EGFR inhibitor. Mol. Cancer Ther. 2008, 7, 874–879. [Google Scholar] [CrossRef] [Green Version]
- Wang, D.D.; Ma, L.; Wong, M.P.; Lee, V.H.; Yan, H. Contribution of EGFR and ErbB-3 Heterodimerization to the EGFR Mutation-Induced Gefitinib- and Erlotinib-Resistance in Non-Small-Cell Lung Carcinoma Treatments. PLoS ONE 2015, 10, e0128360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kuboki, Y.; Matsusaka, S.; Minowa, S.; Shibata, H.; Suenaga, M.; Shinozaki, E.; Mizunuma, N.; Ueno, M.; Yamaguchi, T.; Hatake, K. Circulating tumor cell (CTC) count and epithelial growth factor receptor expression on CTCs as biomarkers for cetuximab efficacy in advanced colorectal cancer. Anticancer Res. 2013, 33, 3905–3910. [Google Scholar]
- Okegawa, T.; Itaya, N.; Hara, H.; Tambo, M.; Nutahara, K. Epidermal Growth Factor Receptor Status in Circulating Tumor Cells as a Predictive Biomarker of Sensitivity in Castration-Resistant Prostate Cancer Patients Treated with Docetaxel Chemotherapy. Int. J. Mol. Sci. 2016, 17, 2008. [Google Scholar] [CrossRef]
- Serrano, M.J.; Ortega, F.G.; Alvarez-Cubero, M.J.; Nadal, R.; Sanchez-Rovira, P.; Salido, M.; Rodriguez, M.; Garcia-Puche, J.L.; Delgado-Rodriguez, M.; Sole, F.; et al. EMT and EGFR in CTCs cytokeratin negative non-metastatic breast cancer. Oncotarget 2014, 5, 7486–7497. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Braun, A.C.; de Mello, C.A.L.; Corassa, M.; Abdallah, E.A.; Urvanegia, A.C.; Alves, V.S.; Flores, B.; Diaz, M.; Nicolau, U.R.; Silva, V.S.E.; et al. EGFR expression in circulating tumor cells from high-grade metastatic soft tissue sarcomas. Cancer Biol. Ther. 2018, 19, 454–460. [Google Scholar] [CrossRef] [Green Version]
- Kallergi, G.; Agelaki, S.; Kalykaki, A.; Stournaras, C.; Mavroudis, D.; Georgoulias, V. Phosphorylated EGFR and PI3K/Akt signaling kinases are expressed in circulating tumor cells of breast cancer patinets. Breast Cancer Res. 2008, 10, 1–11. [Google Scholar] [CrossRef] [Green Version]
- Meador, C.B.; Jin, H.; de Stanchina, E.; Nebhan, C.A.; Pirazzoli, V.; Wang, L.; Lu, P.; Vuong, H.; Hutchinson, K.E.; Jia, P.; et al. Optimizing the sequence of anti-EGFR-targeted therapy in EGFR-mutant lung cancer. Mol. Cancer Ther. 2015, 14, 542–552. [Google Scholar] [CrossRef] [Green Version]
- Nguyen, K.S.; Kobayashi, S.; Costa, D.B. Acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors in non-small-cell lung cancers dependent on the epidermal growth factor receptor pathway. Clin. Lung Cancer 2009, 10, 281–289. [Google Scholar] [CrossRef] [Green Version]
- Matsuo, N.; Azuma, K.; Sakai, K.; Hattori, S.; Kawahara, A.; Ishii, H.; Tokito, T.; Kinoshita, T.; Yamada, K.; Nishio, K.; et al. Association of EGFR Exon 19 Deletion and EGFR-TKI Treatment Duration with Frequency of T790M Mutation in EGFR-Mutant Lung Cancer Patients. Sci. Rep. 2016, 6, 36458. [Google Scholar] [CrossRef] [PubMed]
- Gao, W.; He, J.; Jin, S.D.; Xu, J.; Yu, T.F.; Wang, W.; Zhu, Q.; Dai, H.; Wu, H.; Liu, Y.Q.; et al. Association of Initial Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors Treatment and EGFR Exon 19 Deletion with Frequency of The T790M Mutation in Non-Small Cell Lung Cancer Patients After Resistance To First-Line Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors. Onco. Targets Ther. 2019, 12, 9495–9504. [Google Scholar]
- Wang, Z.F.; Ren, S.X.; Li, W.; Gao, G.H. Frequency of the acquired resistant mutation T790 M in non-small cell lung cancer patients with active exon 19Del and exon 21 L858R: A systematic review and meta-analysis. BMC Cancer 2018, 18, 148. [Google Scholar] [CrossRef] [Green Version]
- Sundaresan, T.K.; Sequist, L.V.; Heymach, J.V.; Riely, G.J.; Janne, 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]
- Huang, C.W.; Chen, Y.T.; Tsai, H.L.; Yeh, Y.S.; Su, W.C.; Ma, C.J.; Tsai, T.N.; Wang, J.Y. EGFR expression in patients with stage III colorectal cancer after adjuvant chemotherapy and on cancer cell function. Oncotarget 2017, 8, 114663–114676. [Google Scholar] [CrossRef] [Green Version]
- Liu, H.E.; Vuppalapaty, M.; Wilkerson, C.; Renier, C.; Chiu, M.; Lemaire, C.; Che, J.; Matsumoto, M.; Carroll, J.; Crouse, S.; et al. Detection of EGFR Mutations in cfDNA and CTCs, and Comparison to Tumor Tissue in Non-Small-Cell-Lung-Cancer (NSCLC) Patients. Front. Oncol. 2020, 10, 572895. [Google Scholar] [CrossRef]
- Schweizer, M.T.; Antonarakis, E.S. Liquid biopsy: Clues on prostate cancer drug resistance. Sci. Transl. Med. 2015, 7, 312fs45. [Google Scholar] [CrossRef] [PubMed]
- Antonarakis, E.S. Predicting treatment response in castration-resistant prostate cancer: Could androgen receptor variant-7 hold the key? Expert Rev. Anticancer Ther. 2015, 15, 143–145. [Google Scholar] [CrossRef]
- 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]
- Guo, Z.; Yang, X.; Sun, F.; Jiang, R.; Linn, D.E.; Chen, H.; Chen, H.; Kong, X.; Melamed, J.; Tepper, C.G.; et al. A novel androgen receptor splice variant is up-regulated during prostate cancer progression and promotes androgen depletion-resistant growth. Cancer Res. 2009, 69, 2305–2313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, Y.; Chan, S.C.; Brand, L.J.; Hwang, T.H.; Silverstein, K.A.; Dehm, S.M. Androgen receptor splice variants mediate enzalutamide resistance in castration-resistant prostate cancer cell lines. Cancer Res. 2013, 73, 483–489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Scher, H.I.; Lu, D.; Schreiber, N.A.; Louw, J.; Graf, R.P.; Vargas, H.A.; Johnson, A.; Jendrisak, A.; Bambury, R.; Danila, D.; et al. Association of AR-V7 on Circulating Tumor Cells as a Treatment-Specific Biomarker with Outcomes and Survival in Castration-Resistant Prostate Cancer. JAMA Oncol. 2016, 2, 1441–1449. [Google Scholar] [CrossRef]
- Scher, H.I.; Graf, R.P.; Schreiber, N.A.; Jayaram, A.; Winquist, E.; McLaughlin, B.; Lu, D.; Fleisher, M.; Orr, S.; Lowes, L.; et al. Assessment of the Validity of Nuclear-Localized Androgen Receptor Splice Variant 7 in Circulating Tumor Cells as a Predictive Biomarker for Castration-Resistant Prostate Cancer. JAMA Oncol. 2018, 4, 1179–1186. [Google Scholar] [CrossRef]
- Hille, C.; Gorges, T.M.; Riethdorf, S.; Mazel, M.; Steuber, T.; Amsberg, G.V.; Konig, F.; Peine, S.; Alix-Panabieres, C.; Pantel, K. Detection of Androgen Receptor Variant 7 (ARV7) mRNA Levels in EpCAM-Enriched CTC Fractions for Monitoring Response to Androgen Targeting Therapies in Prostate Cancer. Cells 2019, 8, 1067. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, T.; Karsh, L.I.; Nissenblatt, M.J.; Canfield, S.E. Androgen Receptor Splice Variant, AR-V7, as a Biomarker of Resistance to Androgen Axis-Targeted Therapies in Advanced Prostate Cancer. Clin. Genitourin Cancer 2020, 18, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Belderbos, B.P.S.; Sieuwerts, A.M.; Hoop, E.O.; Mostert, B.; Kraan, J.; Hamberg, P.; Van, M.N.; Beaufort, C.M.; Onstenk, W.; van Soest, R.J.; et al. Associations between AR-V7 status in circulating tumour cells, circulating tumour cell count and survival in men with metastatic castration-resistant prostate cancer. Eur. J. Cancer 2019, 121, 48–54. [Google Scholar] [CrossRef] [PubMed]
- Armstrong, A.J.; Halabi, S.; Luo, J.; Nanus, D.M.; Giannakakou, P.; Szmulewitz, R.Z.; Danila, D.C.; Healy, P.; Anand, M.; Rothwell, C.J.; et al. Prospective Multicenter Validation of Androgen Receptor Splice Variant 7 and Hormone Therapy Resistance in High-Risk Castration-Resistant Prostate Cancer: The PROPHECY Study. J. Clin. Oncol. 2019, 37, 1120–1129. [Google Scholar] [CrossRef] [PubMed]
- Hench, I.B.; Cathomas, R.; Costa, L.; Fischer, N.; Gillessen, S.; Hench, J.; Hermanns, T.; Kremer, E.; Mingrone, W.; Mestre, R.P.; et al. Analysis of AR/ARV7 Expression in Isolated Circulating Tumor Cells of Patients with Metastatic Castration-Resistant Prostate Cancer (SAKK 08/14 IMPROVE Trial). Cancers 2019, 11, 1099. [Google Scholar] [CrossRef] [Green Version]
- Sakamoto, S. Current status of circulating tumor cell androgen receptor splice variant-7 in metastatic castration-resistant prostate cancer. Ann. Transl. Med. 2019, 7 (Suppl. 8), S375. [Google Scholar] [CrossRef]
- Sepe, P.; Verzoni, E.; Miodini, P.; Claps, M.; Ratta, R.; Martinetti, A.; Mennitto, R.; Sottotetti, E.; Procopio, G.; Cappelletti, V.; et al. Could Circulating Tumor Cells and ARV7 Detection Improve Clinical Decisions in Metastatic Castration-Resistant Prostate Cancer? The Istituto Nazionale dei Tumori (INT) Experience. Cancers 2019, 11, 980. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Engelman, J.A.; Zejnullahu, K.; Mitsudomi, T.; Song, Y.; Hyland, C.; Park, J.O.; Lindeman, N.; Gale, C.M.; Zhao, X.; Christensen, J.; et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 2007, 316, 1039–1043. [Google Scholar] [CrossRef]
- Turke, A.B.; Zejnullahu, K.; Wu, Y.L.; Song, Y.; Dias-Santagata, D.; Lifshits, E.; Toschi, L.; Rogers, A.; Mok, T.; Sequist, L.; et al. Preexistence and clonal selection of MET amplification in EGFR mutant NSCLC. Cancer Cell 2010, 17, 77–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, A.P.; Zhang, J.; Liu, H.; Zhao, S.P.; Dai, S.Z.; Sun, X.L. Association of EGFR expression with angiogenesis and chemoresistance in ovarian carcinoma. Zhonghua Zhong Liu Za Zhi 2009, 31, 48–52. [Google Scholar]
- Bardelli, A.; Corso, S.; Bertotti, A.; Hobor, S.; Valtorta, E.; Siravegna, G.; Sartore-Bianchi, A.; Scala, E.; Cassingena, A.; Zecchin, D.; et al. Amplification of the MET receptor drives resistance to anti-EGFR therapies in colorectal cancer. Cancer Discov. 2013, 3, 658–673. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liska, D.; Chen, C.T.; Bachleitner-Hofmann, T.; Christensen, J.G.; Weiser, M.R. HGF rescues colorectal cancer cells from EGFR inhibition via MET activation. Clin. Cancer Res. 2011, 17, 472–482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tada, H.; Takahashi, H.; Kawabata-Iwakawa, R.; Nagata, Y.; Uchida, M.; Shino, M.; Ida, S.; Mito, I.; Matsuyama, T.; Chikamatsu, K. Molecular phenotypes of circulating tumor cells and efficacy of nivolumab treatment in patients with head and neck squamous cell carcinoma. Sci. Rep. 2020, 10, 21573. [Google Scholar] [CrossRef]
- Tada, H.; Takahashi, H.; Kuwabara-Yokobori, Y.; Shino, M.; Chikamatsu, K. Molecular profiling of circulating tumor cells predicts clinical outcome in head and neck squamous cell carcinoma. Oral Oncol. 2020, 102, 104558. [Google Scholar] [CrossRef]
- Mondelo-Macia, P.; Rodriguez-Lopez, C.; Valina, L.; Aguin, S.; Leon-Mateos, L.; Garcia-Gonzalez, J.; Abalo, A.; Rapado-Gonzalez, O.; Suarez-Cunqueiro, M.; Diaz-Lagares, A.; et al. Detection of MET Alterations Using Cell Free DNA and Circulating Tumor Cells from Cancer Patients. Cells 2020, 9, 522. [Google Scholar] [CrossRef] [Green Version]
- Zhang, T.; Boominathan, R.; Foulk, B.; Rao, C.; Kemeny, G.; Strickler, J.H.; Abbruzzese, J.L.; Harrison, M.R.; Hsu, D.S.; Healy, P.; et al. Development of a Novel c-MET-Based CTC Detection Platform. Mol. Cancer Res. 2016, 14, 539–547. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wu, S.; Li, G.; Zhao, X.; Xiang, J.; Lizaso, A.; Ye, J.; Shi, C.; Chen, L. High-level gain of mesenchymal-epithelial transition factor (MET) copy number using next-generation sequencing as a predictive biomarker for MET inhibitor efficacy. Ann. Transl. Med. 2020, 8, 685. [Google Scholar] [CrossRef] [PubMed]
- Wolf, J.; Seto, T.; Han, J.Y.; Reguart, N.; Garon, E.B.; Groen, H.J.M.; Tan, D.S.W.; Hida, T.; de Jonge, M.; Orlov, S.V.; et al. Capmatinib in MET Exon 14-Mutated or MET-Amplified Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2020, 383, 944–957. [Google Scholar] [CrossRef]
- Kim, J.; Park, K.E.; Jeong, Y.S.; Kim, Y.; Park, H.; Nam, J.H.; Jung, K.; Son, W.S.; Jung, H.S.; Lee, J.H.; et al. Therapeutic Efficacy of ABN401, a Highly Potent and Selective MET Inhibitor, Based on Diagnostic Biomarker Test in MET-Addicted Cancer. Cancers 2020, 12, 1575. [Google Scholar] [CrossRef]
- Janning, M.; Kobus, F.; Babayan, A.; Wikman, H.; Velthaus, J.L.; Bergmann, S.; Schatz, S.; Falk, M.; Berger, L.A.; Bottcher, L.M.; et al. Determination of PD-L1 Expression in Circulating Tumor Cells of NSCLC Patients and Correlation with Response to PD-1/PD-L1 Inhibitors. Cancers 2019, 11, 835. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Manjunath, Y.; Upparahalli, S.V.; Avella, D.M.; Deroche, C.B.; Kimchi, E.T.; Staveley-O’Carroll, K.F.; Smith, C.J.; Li, G.; Kaifi, J.T. PD-L1 Expression with Epithelial Mesenchymal Transition of Circulating Tumor Cells Is Associated with Poor Survival in Curatively Resected Non-Small Cell Lung Cancer. Cancers 2019, 11, 806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nicolazzo, C.; Raimondi, C.; Mancini, M.; Caponnetto, S.; Gradilone, A.; Gandini, O.; Mastromartino, M.; Del Bene, G.; Prete, A.; Longo, F.; et al. Monitoring PD-L1 positive circulating tumor cells in non-small cell lung cancer patients treated with the PD-1 inhibitor Nivolumab. Sci. Rep. 2016, 6, 31726. [Google Scholar] [CrossRef]
- Adams, D.L.; Adams, D.K.; He, J.; Kalhor, N.; Zhang, M.; Xu, T.; Gao, H.; Reuben, J.M.; Qiao, Y.; Komaki, R.; et al. Sequential Tracking of PD-L1 Expression and RAD50 Induction in Circulating Tumor and Stromal Cells of Lung Cancer Patients Undergoing Radiotherapy. Clin. Cancer Res. 2017, 23, 5948–5958. [Google Scholar] [CrossRef] [Green Version]
- Kallergi, G.; Vetsika, E.K.; Aggouraki, D.; Lagoudaki, E.; Koutsopoulos, A.; Koinis, F.; Katsarlinos, P.; Trypaki, M.; Messaritakis, I.; Stournaras, C.; et al. Evaluation of PD-L1/PD-1 on circulating tumor cells in patients with advanced non-small cell lung cancer. Ther. Adv. Med. Oncol. 2018, 10, 1758834017750121. [Google Scholar] [CrossRef]
- Wang, Y.; Kim, T.H.; Fouladdel, S.; Zhang, Z.; Soni, P.; Qin, A.; Zhao, L.; Azizi, E.; Lawrence, T.S.; Ramnath, N.; et al. PD-L1 Expression in Circulating Tumor Cells Increases during Radio(chemo)therapy and Indicates Poor Prognosis in Non-small Cell Lung Cancer. Sci. Rep. 2019, 9, 566. [Google Scholar] [CrossRef] [Green Version]
- Grizzi, F.; Castello, A.; Qehajaj, D.; Toschi, L.; Rossi, S.; Pistillo, D.; Paleari, V.; Veronesi, G.; Novellis, P.; Monterisi, S.; et al. Independent expression of circulating and tissue levels of PD-L1: Correlation of clusters with tumor metabolism and outcome in patients with non-small cell lung cancer. Cancer Immunol. Immunother. 2019, 68, 1537–1545. [Google Scholar] [CrossRef]
- Cheng, Y.; Wang, T.; Lv, X.; Li, R.; Yuan, L.; Shen, J.; Li, Y.; Yan, T.; Liu, B.; Wang, L. Detection of PD-L1 Expression and Its Clinical Significance in Circulating Tumor Cells from Patients with Non-Small-Cell Lung Cancer. Cancer Manag. Res. 2020, 12, 2069–2078. [Google Scholar] [CrossRef] [Green Version]
- Mazel, M.; Jacot, W.; Pantel, K.; Bartkowiak, K.; Topart, D.; Cayrefourcq, L.; Rossille, D.; Maudelonde, T.; Fest, T.; Alix-Panabieres, C. Frequent expression of PD-L1 on circulating breast cancer cells. Mol. Oncol. 2015, 9, 1773–1782. [Google Scholar] [CrossRef] [Green Version]
- 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] [Green Version]
- Liu, M.; Wang, R.; Sun, X.; Liu, Y.; Wang, Z.; Yan, J.; Kong, X.; Liang, S.; Liu, Q.; Zhao, T.; et al. Prognostic significance of PD-L1 expression on cell-surface vimentin-positive circulating tumor cells in gastric cancer patients. Mol. Oncol. 2020, 14, 865–881. [Google Scholar] [CrossRef] [PubMed]
- Khattak, M.A.; Reid, A.; Freeman, J.; Pereira, M.; McEvoy, A.; Lo, J.; Frank, M.H.; Meniawy, T.; Didan, A.; Spencer, I.; et al. PD-L1 Expression on Circulating Tumor Cells May Be Predictive of Response to Pembrolizumab in Advanced Melanoma: Results from a Pilot Study. Oncologist 2020, 25, e520–e527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ilie, M.; Szafer-Glusman, E.; Hofman, V.; Chamorey, E.; Lalvee, S.; Selva, E.; Leroy, S.; Marquette, C.H.; Kowanetz, M.; Hedge, P.; et al. Detection of PD-L1 in circulating tumor cells and white blood cells from patients with advanced non-small-cell lung cancer. Ann. Oncol. 2018, 29, 193–199. [Google Scholar] [CrossRef] [PubMed]
- Wendel, M.; Bazhenova, L.; Boshuizen, R.; Kolatkar, A.; Honnatti, M.; Cho, E.H.; Marrinucci, D.; Sandhu, A.; Perricone, A.; Thistlethwaite, P.; et al. Fluid biopsy for circulating tumor cell identification in patients with early-and late-stage non-small cell lung cancer: A glimpse into lung cancer biology. Phys. Biol 2012, 9, 016005. [Google Scholar] [CrossRef] [Green Version]
- Liu, H.E.; Triboulet, M.; Zia, A.; Vuppalapaty, M.; Kidess-Sigal, E.; Coller, J.; Natu, V.S.; Shokoohi, V.; Che, J.; Renier, C.; et al. Workflow optimization of genome amplification and targeted panel sequencing for CTC mutation detection. NPJ Genome Med. 2017, 2, 34. [Google Scholar] [CrossRef]
- Picelli, S.; Faridani, O.R.; Bjorklund, A.K.; Winberg, G.; Sagasser, S.; Sandberg, R. Full-length RNA-seq from single cells using Smart-seq2. Nat. Protoc. 2014, 9, 171–181. [Google Scholar] [CrossRef]
- Sasagawa, Y.; Nikaido, I.; Hayashi, T.; Danno, H.; Uno, K.D.; Imai, T.; Ueda, H.R. Quartz-Seq: A highly reproducible and sensitive single-cell RNA sequencing method, reveals non-genetic gene expression heterogeneity. Genome Biol. 2013, 14, 3097. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hochegerner, H.; Lonnerberg, P.; Hodge, R.; Mikes, J.; Heskol, A.; Hubschle, H.; Lin, P.; Picelli, S.; Manno, G.L.; Ratz, M.; et al. STRT-seq-2i: Dual-index 5’ single cell and nucleus RNA-seq on an addressable microwell array. Sci. Rep. 2017, 7, 16327. [Google Scholar] [CrossRef] [Green Version]
- Wang, Z.; Gerstein, M.; Snyder, M. RNA-Seq: A revolutionary tool for transcriptomics. Nat. Rev. Genet. 2009, 10, 57–63. [Google Scholar] [CrossRef]
- Sho, S.; Court, C.M.; Winograd, P.; Lee, S.; Hou, S.; Graeber, T.G.; Tseng, H.R.; Tomlinson, J.S. Precision oncology using a limited number of cells: Optimization of whole genome amplification products for sequencing applications. BMC Cancer 2017, 17, 457. [Google Scholar] [CrossRef] [Green Version]
- Wang, Q.; Zhao, L.; Han, L.; Tuo, X.; Ma, S.; Wang, Y.; Feng, X.; Liang, D.; Sun, C.; Wang, Q.; et al. The Discordance of Gene Mutations between Circulating Tumor Cells and Primary/Metastatic Tumor. Mol. Ther Oncolytics 2019, 15, 21–29. [Google Scholar] [CrossRef] [Green Version]
- Gulbahce, N.; Magbanua, M.J.M.; Chin, R.; Agarwal, M.R.; Luo, X.; Liu, J.; Hayden, D.M.; Mao, Q.; Ciotlos, S.; Li, Z.; et al. Quantitative Whole Genome Sequencing of Circulating Tumor Cells Enables Personalized Combination Therapy of Metastatic Cancer. Cancer Res. 2017, 77, 4530–4541. [Google Scholar] [CrossRef] [Green Version]
- De Luca, F.; Rotunno, G.; Salvianti, F.; Galardi, F.; Pestrin, M.; Gabellini, S.; Simi, L.; Mancini, I.; Vannucchi, A.M.; Pazzagli, M.; et al. Mutational analysis of single circulating tumor cells by next generation sequencing in metastatic breast cancer. Oncotarget 2016, 7, 26107–26119. [Google Scholar] [CrossRef] [PubMed]
- Ni, X.; Zhuo, M.; Su, Z.; Duan, J.; Gao, Y.; Wang, Z.; Zong, C.; Bai, H.; Chapman, A.R.; Zhao, J.; et al. Reproducible copy number variation patterns among single circulating tumor cells of lung cancer patients. Proc. Natl. Acad. Sci. USA 2013, 110, 21083–21088. [Google Scholar] [CrossRef] [Green Version]
- Scher, H.I.; Graf, R.P.; Schreiber, N.A.; McLaughlin, B.; Jendrisak, A.; Wang, Y.; Lee, J.; Greene, S.; Krupa, R.; Lu, D.; et al. Phenotypic Heterogeneity of Circulating Tumor Cells Informs Clinical Decisions between AR Signaling Inhibitors and Taxanes in Metastatic Prostate Cancer. Cancer Res. 2017, 77, 5687–5698. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- D’Oronzo, S.; Lovero, D.; Palmirotta, R.; Stucci, L.S.; Tucci, M.; Felici, C.; Cascardi, E.; Giardina, C.; Cafforio, P.; Silvestris, F. Dissection of major cancer gene variants in subsets of circulating tumor cells in advanced breast cancer. Sci. Rep. 2019, 9, 17276. [Google Scholar] [CrossRef] [Green Version]
- Oulhen, M.; Pawlikowska, P.; Tayoun, T.; Garonzi, M.; Buson, G.; Forcato, C.; Manaresi, N.; Aberlenc, A.; Mezquita, L.; Lecluse, Y.; et al. Circulating tumor cell copy number heterogeneity in ALK-rearranged non-small-cell lung cancer resistant to ALK inhibitors. NPJ Precis. Oncol. 2021, 5, 67. [Google Scholar] [CrossRef]
- Gracia, D.F.; Nteliopoulos, G.; Hastings, R.; Rushton, A.; Page, K.; Martinson, L.J.; Gray, M.; Guttery, D.S.; Ferrarini, A.; Manaresi, N.; et al. Genomic copy number profiling of single CTCs reveals clonal evolution in metastatic breast cancer and identifies actionable targets for informing treatment decisions. AACR Cancer Res. 2021, 81 (Suppl. 13), 587. [Google Scholar]
- Chang, Y.; Wang, Y.; Li, B.; Lu, X.; Wang, R.; Li, H.; Yan, B.; Gu, A.; Wang, W.; Huang, A.; et al. Whole-Exome Sequencing on Circulating Tumor Cells Explores Platinum-Drug Resistance Mutations in Advanced Non-small Cell Lung Cancer. Front. Genet. 2021, 12, 722078. [Google Scholar] [CrossRef] [PubMed]
- Rossi, E.; Zamarchi, R. Single-Cell analysis of circulating tumor cells: How far have we come in the -omics era? Front. Genet. 2019, 10, 958. [Google Scholar] [CrossRef] [Green Version]
- Hofman, P. Next Generation sequencing with liquid biopsies from treatment naïve non small cell lung carcinoma patients. Cancers 2021, 13, 2049. [Google Scholar] [CrossRef] [PubMed]
- Gawad, C.; Koh, W.; Quake, S.R. Single-cell genome sequencing: Current state of the science. Nat. Rev. Genet. 2016, 17, 175–188. [Google Scholar] [CrossRef]
- Lu, S.; Chang, C.J.; Guan, Y.; Szafer-Glusman, E.; Punnoose, E.; Do, A.; Suttmann, B.; Gagnon, R.; Rodriguez, A.; Landers, M.; et al. Genomic Analysis of Circulating Tumor Cells at the Single-Cell Level. J. Mol. Diagn. 2020, 22, 770–781. [Google Scholar] [CrossRef]
- Wu, Y.; Niu, Y.; Gao, W.; Shen, X. Morphology classification of circulating tumor cells could be a predictor of recurrent disease in patients with non-small cell lung cancer after surgery. J. Clin. Oncol. 2021, 38, e15530. [Google Scholar] [CrossRef]
- de Bono, J.S.; Pantel, K.; Efstathiou, E.; Sternberg, C.N.; Castellano Gauna, D.; Fizazi, K.; Tomabl, B.; Wulfing, C.; Schonhoft, J.D.; Tubbs, A.; et al. 614P Circulating tumor cell morphologic sub-types present prior to treatment in the CARD trial identify therapy resistance. Ann. Oncol. 2021, 32, S653–S654. [Google Scholar] [CrossRef]
- 20 Years of Precision Medicine in Oncology; Lancet: London, UK, 2021; Volume 397, p. 1781, Editorial.
- Ignatiadis, M.; Sledge, G.W.; Jeffrey, S.S. Liquid biopsy enters the clinic-implementaion issues and future challenges. Nat. Rev. Clin. Oncol. 2021, 18, 297–312. [Google Scholar] [CrossRef] [PubMed]
- Hamza, B.; Miller, A.B.; Meier, L.; Stockslager, M.; Ng, S.R.; King, E.M.; Lin, L.; DeGouveia, K.L.; Mulugeta, N.; Calistri, N.L.; et al. Measuring kinetics and metastatic propensity of CTCs by blood exchange between mice. Nat. Commun. 2021, 12, 5680. [Google Scholar] [CrossRef] [PubMed]
- Scherag, F.D.; Niestroj-Pahl, R.; Krusekopf, S.; Lucke, K.; Brandstetter, T.; Ruhe, J. Highly Selective Capture Surfaces on Medical Wires for Fishing Tumor Cells in Whole Blood. Anal. Chem. 2017, 89, 1846–1854. [Google Scholar] [CrossRef] [PubMed]
- Jaeger, B.A.S.; Neugebauer, J.; Andergassen, U.; Melcher, C.; Schochter, F.; Mouarrawy, D.; Ziemendorff, G.; Clemens, M.; Abel, E.V.; Heinrich, G.; et al. The HER2 phenotype of circulating tumor cells in HER2-positive early breast cancer: A translational research project of a prospective randomized phase III trial. PLoS ONE 2017, 12, e0173593. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ignatiadis, M.; Rothe, F.; Chaboteaux, C.; Durbecq, V.; Rouas, G.; Criscitiello, C.; Metallo, J.; Kheddoumi, N.; Singhal, S.K.; Michiels, S.; et al. HER2-positive circulating tumor cells in breast cancer. PLoS ONE 2011, 6, e15624. [Google Scholar] [CrossRef]
- Cao, S.; Li, Y.; Li, J.; Li, C.F.; Zhang, W.; Yang, Z.Q.; Meng, S.D. Quantitative determination of HER2 expression by confocal microscopy assay in CTCs of breast cancer. Oncol. Rep. 2010, 23, 423–428. [Google Scholar]
- Romero, D. Breast cancer: CTC heterogeneity is dynamic. Nat. Rev. Clin. Oncol. 2016, 13, 654. [Google Scholar] [CrossRef]
- Keller, L.; Pantel, K. Unravelling tumour heterogeneity by single-cell profiling of circulating tumour cells. Nat. Rev. Cancer 2019, 19, 553–567. [Google Scholar] [CrossRef] [Green Version]
- de Wit, S.; Manicone, M.; Rossi, E.; Lampignano, R.; Yang, L.; Zill, B.; Rengel-Puertas, A.; Ouhlen, M.; Crespo, M.; Berghuis, A.M.S.; et al. EpCAM(high) and EpCAM(low) circulating tumor cells in metastatic prostate and breast cancer patients. Oncotarget 2018, 9, 35705–35716. [Google Scholar] [CrossRef] [Green Version]
- de Wit, S.; van Dalum, G.; Terstappen, L.W. Detection of circulating tumor cells. Scientifica 2014, 2014, 819362. [Google Scholar] [CrossRef] [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]
- Raimondi, C.; Nicolazzo, C.; Gradilone, A. Circulating tumor cells isolation: The “post-EpCAM era”. Chin. J. Cancer Res. 2015, 27, 461–470. [Google Scholar]
- Konigsberg, 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]
- Wu, S.; Liu, S.; Liu, Z.; Huang, J.; Pu, X.; Li, J.; Yang, D.; Deng, H.; Yang, N.; Xu, J. Classification of circulating tumor cells by epithelial-mesenchymal transition markers. PLoS ONE 2015, 10, e0123976. [Google Scholar] [CrossRef] [PubMed]
- Papadaki, M.A.; Kallergi, G.; Zafeiriou, Z.; Manouras, L.; Theodoropoulos, P.A.; Mavroudis, D.; Georgoulias, V.; Agelaki, S. Co-expression of putative stemness and epithelial-to-mesenchymal transition markers on single circulating tumour cells from patients with early and metastatic breast cancer. BMC Cancer 2014, 14, 651. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Qi, L.N.; Xiang, B.D.; Wu, F.X.; Ye, J.Z.; Zhong, J.H.; Wang, Y.Y.; Chen, Y.Y.; Chen, Z.S.; Ma, L.; Chen, J.; et al. Circulating Tumor Cells Undergoing EMT Provide a Metric for Diagnosis and Prognosis of Patients with Hepatocellular Carcinoma. Cancer Res. 2018, 78, 4731–4744. [Google Scholar] [CrossRef] [Green Version]
- Chen, Y.; Li, S.; Li, W.; Yang, R.; Zhang, X.; Ye, Y.; Yu, J.; Ye, L.; Tang, W. Circulating tumor cells undergoing EMT are poorly correlated with clinical stages or predictive of recurrence in hepatocellular carcinoma. Sci. Rep. 2019, 9, 7084. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Murlidhar, V.; Rivera-Baez, L.; Nagrath, S. Affinity versus Label-Free isolation of Circulating tumor cells: Who Wins? Small 2016, 12, 450–463. [Google Scholar] [CrossRef]
- Wang, Y.; Guo, L.; Feng, L.; Zhang, W.; Xiao, T.; Di, X.; Chen, G.; Zhang, K. Single nucleotide variant profiles of viable single circulating tumour cells reveal CTC behaviours in breast cancer. Oncol. Rep. 2018, 39, 2147–2159. [Google Scholar] [CrossRef] [Green Version]
- Pestrin, M.; Salvianti, F.; Galardi, F.; De Luca, F.; Turner, N.; Malorni, L.; Pazzagli, M.; Di Leo, A.; Pinzani, P. Heterogeneity of PIK3CA mutational status at the single cell level in circulating tumor cells from metastatic breast cancer patients. Mol. Oncol. 2015, 9, 749–757. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Heitzer, E.; Auer, M.; Gasch, C.; Pichler, M.; Ulz, P.; Hoffmann, E.M.; Lax, S.; Waldispuehl-Geigl, J.; Mauermann, O.; Lackner, C.; et al. Complex tumor genomes inferred from single circulating tumor cells by array-CGH and next-generation sequencing. Cancer Res. 2013, 73, 2965–2975. [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]
- Kwan, T.T.; Bardia, A.; Spring, L.M.; Giobbie-Hurder, A.; Kalinich, M.; Dubash, T.; Sundaresan, T.; Hong, X.; LiCausi, J.A.; Ho, U.; et al. A Digital RNA Signature of Circulating Tumor Cells Predicting Early Therapeutic Response in Localized and Metastatic Breast Cancer. Cancer Discov. 2018, 8, 1286–1299. [Google Scholar] [CrossRef] [Green Version]
- Miyamoto, D.T.; Lee, R.J.; Kalinich, M.; LiCausi, J.A.; Zheng, Y.; Chen, T.; Milner, J.D.; Emmons, E.; Ho, U.; Broderick, K.; et al. An RNA-Based Digital Circulating Tumor Cell Signature Is Predictive of Drug Response and Early Dissemination in Prostate Cancer. Cancer Discov. 2018, 8, 288–303. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Di Trapani, M.; Manaresi, N.; Medoro, G. DEPArray system: An automatic image-based sorter for isolation of pure circulating tumor cells. Cytometry A 2018, 93, 1260–1266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nelep, C.; Eberhardt, J. Automated rare single cell picking with the ALS cellcelector. Cytom. A 2018, 93, 1267–1270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Swennenhuis, J.F.; Tibbe, A.G.; Stevens, M.; Katika, M.R.; van Dalum, J.; Tong, H.D.; van Rijn, C.J.; Terstappen, L.W. Self-seeding microwell chip for the isolation and characterization of single cells. Lab Chip 2015, 15, 3039–3046. [Google Scholar] [CrossRef]
- Khoo, B.L.; Lee, S.C.; Kumar, P.; Tan, T.Z.; Warkiani, M.E.; Ow, S.G.; Nandi, S.; Lim, C.T.; Thiery, J.P. Short-term expansion of breast circulating cancer cells predicts response to anti-cancer therapy. Oncotarget 2015, 6, 15578–15593. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Balakrishnan, A.; Koppaka, D.; Anand, A.; Deb, B.; Grenci, G.; Viasnoff, V.; Thompson, E.W.; Gowda, H.; Bhat, R.; Rangarajan, A.; et al. Circulating Tumor Cell cluster phenotype allows monitoring response to treatment and predicts survival. Sci. Rep. 2019, 9, 7933. [Google Scholar] [CrossRef]
- Chen, X.; Gole, J.; Gore, A.; He, Q.; Lu, M.; Min, J.; Yuan, Z.; Yang, X.; Jiang, Y.; Zhang, T.; et al. Noninvasive early detection of cancer four years before conventional diagnosis using a blood-test. Nat. Commun. 2020, 100, 3475. [Google Scholar] [CrossRef] [PubMed]
- Nabet, B.Y.; Esfashin, M.S.; Moding, E.J.; Hamilton, E.G.; Chabon, J.J.; Rizvi, H.; Steen, C.B.; Chaudhuri, A.A.; Liu, C.L.; Hui, A.B.; et al. Noninvasive early identification of therapeutic benefit from immune checkpoin inhibition. Cell 2020, 183, 363–376. [Google Scholar] [CrossRef]
Technique | CTC Isolation Platform | Marker | Capture Efficiency | Recovery | Purity | Viability | Clinical Detection Rate (No. of Patients) | Ref. |
---|---|---|---|---|---|---|---|---|
Immuno magnetic | CellSearch | EpCAM | - | 85% | - | Non-viable | 71% | [12,13] |
Magsweeper | EpCAM | 62–70% | - | 50% | Viable | 100% | [14] | |
EasySep | CD45 (Negative Selection) | 79% | - | 42% | - | - | [15] | |
Immuno magnetic and microfluidic | Isoflux | EpCAM | 73–81% | - | - | - | 96% | [16] |
GenoCTC | EpCAM, MET, Vimentin | - | 77% | 90% | Viable | 94% (n = 16) | [17] | |
CTC-iCHIP | EpCAM, CD45+ Size-based sorting | 77–98% | - | 99% | - | 90% (n = 41) | [18] | |
Magnetic Sifter | EpCAM | 74% | - | - | Viable | 100% (n = 4) | [19] | |
OncoBean Chip | EpCAM | 90% | - | - | Viable | 100% (n = 4) | [20] | |
LiquidBiopsy | Trop2, Muc1, Her3, MelCAM, EpCAM | - | 70–80% | 70–77% | - | - | [21] | |
Microfluidic immunocapture | CEE | Trop1, Trop2, MET, FBP, N-Cadherin, CD318, HER2, MUC1, EGFR, MSCA-1 | 76% | 89% | - | - | 63% (n = 24) | [22] |
CTC-CHIP | EpCAM | 65% | >65% | 52–67% | Viable | 99% | [23] | |
Biofluidica HT_CTC chip | EpCAM, Seprase | 95% | 90% | >86% | Viable | 100% (n = 7) | [24,25] | |
CytoQuest | EpCAM, Cell Surface Vimentin | - | - | - | Non-viable | - | [26,27] | |
Herringbone Chip | EpCAM | 80% | - | - | Viable | 93% (n = 15) | [28,29] | |
Micro filtration | Parsortix | Size based sorting | 45–70% | - | 54–69% | Viable | 39% (n = 26) | [30] |
ISET | Size and deformability-based filtration | - | - | - | Non-viable | 80% (n = 40) | [31,32,33] | |
ScreenCell | Size and deformability-based filtration | - | - | - | Viable | 77% (n = 76) | [34,35] | |
Inertial focusing and microfluidics | ClearCell | Size-based | - | - | - | Viable | 100% (n = 56; n = 10) | [36,37] |
VTX-1 | Size-based | ~80% | 54–72% | 57–94% | Viable | 73–80% (n = 15–41) | [38] | |
Density | OncoQuick | Density fractionation | 74–91% | 87% | - | Viable | 23% (n = 61) | [39] |
Accucyte | Density fractionation | - | 90% | - | Viable | 81% (n = 27) | [40] | |
In vivo immune capture | GILUPI CellCollector | EpCAM | - | - | - | Non-viable | 58% (n = 108; n = 185) | [41,42] |
BPNS- Catheter | EpCAM antibody functionalized catheter | 2.1% (in 5 min) | ~80% | - | NA | NA | [43] | |
Intravascular aphaeretic system | Blood allowed to pass through herringbone graphene oxide chip-coated with anti-EpCAM antibody | 80–90% | - | - | Viable | NA | [44] | |
MagWire | EpCAM-coated magnetic particle | 39–70% | - | - | Viable | NA | [45] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 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
Chelakkot, C.; Yang, H.; Shin, Y.K. Relevance of Circulating Tumor Cells as Predictive Markers for Cancer Incidence and Relapse. Pharmaceuticals 2022, 15, 75. https://doi.org/10.3390/ph15010075
Chelakkot C, Yang H, Shin YK. Relevance of Circulating Tumor Cells as Predictive Markers for Cancer Incidence and Relapse. Pharmaceuticals. 2022; 15(1):75. https://doi.org/10.3390/ph15010075
Chicago/Turabian StyleChelakkot, Chaithanya, Hobin Yang, and Young Kee Shin. 2022. "Relevance of Circulating Tumor Cells as Predictive Markers for Cancer Incidence and Relapse" Pharmaceuticals 15, no. 1: 75. https://doi.org/10.3390/ph15010075
APA StyleChelakkot, C., Yang, H., & Shin, Y. K. (2022). Relevance of Circulating Tumor Cells as Predictive Markers for Cancer Incidence and Relapse. Pharmaceuticals, 15(1), 75. https://doi.org/10.3390/ph15010075