V. Plaks, C. D. Koopman, and . Werb, Z. Cancer. Circulating tumor cells. Science, vol.341, pp.1186-1188, 2013.

M. Tellez-gabriel, B. Ory, F. Lamoureux, M. F. Heymann, D. T. Heymann et al., The Key Advantages of Single-Cell Analysis, Int. J. Mol. Sci, vol.17, p.2142, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-01466086

H. K. Brown, M. Tellez-gabriel, P. F. Cartron, F. Vallette, M. F. Heymann et al., Characterization of circulating tumor cells as a reflection of the tumor heterogeneity: Myth or reality?, Drug Discov. Today, 2018.
URL : https://hal.archives-ouvertes.fr/inserm-01947472

T. M. Gorges, A. Kuske, K. Rock, O. Mauermann, V. Muller et al., Accession of Tumor Heterogeneity by Multiplex Transcriptome Profiling of Single Circulating Tumor Cells, Clin. Chem, vol.62, pp.1504-1515, 2016.

C. Alix-panabieres and K. Pantel, Clinical Applications of Circulating Tumor Cells and Circulating Tumor DNA as Liquid Biopsy, Cancer Discov, vol.6, pp.479-491, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01872958

D. Lorente, D. Olmos, J. Mateo, D. Dolling, D. Bianchini et al., Circulating tumour cell increase as a biomarker of disease progression in metastatic castration-resistant prostate cancer patients with low baseline CTC counts, Ann. Oncol. Off. J. Eur. Soc. Med. Oncol, vol.29, pp.1554-1560, 2018.

E. S. Antonarakis, C. Lu, B. Luber, H. Wang, Y. Chen et al., Clinical Significance of Androgen Receptor Splice Variant-7 mRNA Detection in Circulating Tumor Cells of Men with Metastatic Castration-Resistant Prostate Cancer Treated with First-and Second-Line Abiraterone and Enzalutamide, J. Clin. Oncol, vol.35, pp.2149-2156, 2017.

X. Zhang, H. Li, X. Yu, S. Li, Z. Lei et al., Analysis of Circulating Tumor Cells in Ovarian Cancer and Their Clinical Value as a Biomarker, Cell. Physiol. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol, vol.48, 1983.

K. Pantel and C. Alix-panabieres, Liquid biopsy: Potential and challenges, Mol. Oncol, vol.10, pp.371-373, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01877343

I. Baccelli, A. Schneeweiss, S. Riethdorf, A. Stenzinger, A. Schillert et al., Identification of a population of blood circulating tumor cells from breast cancer patients that initiates metastasis in a xenograft assay, Nat. Biotechnol, vol.31, pp.539-544, 2013.

L. Cayrefourcq, T. Mazard, S. Joosse, J. Solassol, J. Ramos et al., Establishment and characterization of a cell line from human circulating colon cancer cells, Cancer Res, vol.75, pp.892-901, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01990012

M. Yu, S. Stott, M. Toner, S. Maheswaran, and D. A. Haber, Circulating tumor cells: Approaches to isolation and characterization, J. Cell Biol, vol.192, pp.373-382, 2011.

M. M. Ferreira, V. C. Ramani, and S. S. Jeffrey, Circulating tumor cell technologies, Mol. Oncol, vol.10, pp.374-394, 2016.

Z. Shen, A. Wu, and X. Chen, Current detection technologies for circulating tumor cells, Chem. Soc. Rev, vol.46, pp.2038-2056, 2017.

M. T. Gabriel, L. R. Calleja, A. Chalopin, B. Ory, and D. Heymann, Circulating Tumor Cells: A Review of Non-EpCAM-Based Approaches for Cell Enrichment and Isolation, Clin. Chem, vol.62, pp.571-581, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-01466107

H. Cho, J. Kim, H. Song, K. Y. Sohn, M. Jeon et al., Microfluidic technologies for circulating tumor cell isolation, Analyst, vol.143, pp.2936-2970, 2018.

D. Heymann and M. Tellez-gabriel, Circulating Tumor Cells: The Importance of Single Cell Analysis, Adv. Exp. Med. Biol, vol.1068, pp.45-58, 2018.

L. Broncy, B. B. Njima, A. Mejean, C. Beroud, K. B. Romdhane et al., Single-cell genetic analysis validates cytopathological identification of circulating cancer cells in patients with clear cell renal cell carcinoma, Oncotarget, vol.9, 2018.

G. Kallergi, D. Aggouraki, N. Zacharopoulou, C. Stournaras, V. Georgoulias et al., Evaluation of alpha-tubulin, detyrosinated alpha-tubulin, and vimentin in CTCs: Identification of the interaction between CTCs and blood cells through cytoskeletal elements, Breast Cancer Res, p.67, 1920.

M. Yanagita, J. J. Luke, F. S. Hodi, P. A. Janne, and C. P. Paweletz, Isolation and characterization of circulating melanoma cells by size filtration and fluorescent in-situ hybridization, Melanoma Res, vol.28, pp.89-95, 2018.

S. Kruspe, D. D. Dickey, K. T. Urak, G. N. Blanco, M. J. Miller et al., Rapid and Sensitive Detection of Breast Cancer Cells in Patient Blood with Nuclease-Activated Probe Technology, Mol. Ther. Nucleic Acids, vol.8, pp.542-557, 2017.

D. L. Adams, S. Stefansson, C. Haudenschild, S. S. Martin, M. Charpentier et al., Cytometric characterization of circulating tumor cells captured by microfiltration and their correlation to the CellSearch((R)) CTC test, Cytom. Part A J. Int. Soc. Anal. Cytol, vol.87, pp.137-144, 2015.

D. L. Adams, S. S. Martin, R. K. Alpaugh, M. Charpentier, S. Tsai et al., Circulating giant macrophages as a potential biomarker of solid tumors, Proc. Natl. Acad. Sci, vol.111, pp.3514-3519, 2014.

R. A. Harouaka, M. D. Zhou, Y. T. Yeh, W. J. Khan, A. Das et al., Flexible micro spring array device for high-throughput enrichment of viable circulating tumor cells, Clin. Chem, vol.60, pp.323-333, 2014.

I. Krol, F. Castro-giner, M. Maurer, S. Gkountela, B. M. Szczerba et al., Detection of circulating tumour cell clusters in human glioblastoma, Br. J. Cancer, vol.119, pp.487-491, 2018.

A. El-heliebi, C. Hille, N. Laxman, J. Svedlund, C. Haudum et al., Situ Detection and Quantification of AR-V7, AR-FL, PSA, and KRAS Point Mutations in Circulating Tumor Cells, vol.64, pp.536-546, 2018.

X. Qin, S. Park, S. P. Duffy, K. Matthews, R. R. Ang et al., Size and deformability based separation of circulating tumor cells from castrate resistant prostate cancer patients using resettable cell traps, Lab Chip, vol.15, pp.2278-2286, 2015.

A. F. Sarioglu, N. Aceto, N. Kojic, M. C. Donaldson, M. Zeinali et al., A microfluidic device for label-free, physical capture of circulating tumor cell clusters, Nat. Methods, vol.12, pp.685-691, 2015.

A. Yusa, M. Toneri, T. Masuda, S. Ito, S. Yamamoto et al., Development of a new rapid isolation device for circulating tumor cells (CTCs) using 3D palladium filter and its application for genetic analysis, PLoS ONE, vol.9, p.88821, 2014.

B. Markl, N. Wilhelms, M. Anthuber, G. Schenkirsch, G. Schlimok et al., Circulating cytokeratin-positive cells and tumor budding in colorectal cancer, World J. Clin. Oncol, vol.7, pp.433-440, 2016.

B. A. Jaeger, J. Jueckstock, U. Andergassen, J. Salmen, F. Schochter et al., Evaluation of two different analytical methods for circulating tumor cell detection in peripheral blood of patients with primary breast cancer, Biomed. Res. Int, p.491459, 2014.

A. Soler, L. Cayrefourcq, M. Mazel, and C. Alix-panabieres, EpCAM-Independent Enrichment and Detection of Viable Circulating Tumor Cells Using the EPISPOT Assay, Methods Mol. Biol, vol.1634, pp.263-276, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01755569

A. Kuske, T. M. Gorges, P. Tennstedt, A. K. Tiebel, R. Pompe et al., Improved detection of circulating tumor cells in non-metastatic high-risk prostate cancer patients, Sci. Rep, vol.6, p.39736, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01814421

H. D. Axelrod, K. J. Pienta, and K. C. Valkenburg, Optimization of Immunofluorescent Detection of Bone Marrow Disseminated Tumor Cells, Biol. Proced. Online, vol.20, p.13, 2018.

S. Roy, H. D. Axelrod, K. C. Valkenburg, S. Amend, and K. J. Pienta, Optimization of prostate cancer cell detection using multiplex tyramide signal amplification, J. Cell. Biochem, 2018.

A. Cubero, M. J. Lorente, J. A. Robles-fernandez, I. Rodriguez-martinez, A. Puche et al., Circulating tumor cells: Markers and methodologies for enrichment and detection, Methods Mol. Biol, vol.1634, pp.283-303, 2017.

D. J. O'shannessy, D. W. Davis, K. Anderes, and E. B. Somers, Isolation of Circulating Tumor Cells from Multiple Epithelial Cancers with ApoStream((R)) for Detecting (or Monitoring) the Expression of Folate Receptor Alpha, Biomark. Insights, vol.11, pp.7-18, 2016.

P. Balasubramanian, R. J. Kinders, S. Kummar, V. Gupta, D. Hasegawa et al., Antibody-independent capture of circulating tumor cells of non-epithelial origin with the ApoStream(R) system, PLoS ONE, vol.12, p.175414, 2017.

A. Chalopin, M. Tellez-gabriel, H. K. Brown, F. Vallette, M. F. Heymann et al., Isolation of circulating tumor cells in a preclinical model of osteosarcoma: Effect of chemotherapy, J. Bone Oncol, vol.12, pp.83-90, 2018.
URL : https://hal.archives-ouvertes.fr/inserm-01851614

B. Mesquita, D. G. Rothwell, D. J. Burt, F. Chemi, F. Fernandez-gutierrez et al., Molecular analysis of single circulating tumour cells following long-term storage of clinical samples, Mol. Oncol, vol.11, pp.1687-1697, 2017.

J. Y. Chan, A. B. Ahmad-kayani, M. A. Ali, C. K. Kok, B. Majlis et al., Dielectrophoresis-based microfluidic platforms for cancer diagnostics, Biomicrofluidics, vol.12, p.11503, 2018.

A. Goldkorn, B. Ely, D. I. Quinn, C. M. Tangen, L. M. Fink et al., Circulating tumor cell counts are prognostic of overall survival in SWOG S0421: A phase III trial of docetaxel with or without atrasentan for metastatic castration-resistant prostate cancer, J. Clin. Oncol, vol.32, pp.1136-1142, 2014.

C. Aggarwal, N. J. Meropol, C. J. Punt, N. Iannotti, B. H. Saidman et al., Relationship among circulating tumor cells, CEA and overall survival in patients with metastatic colorectal cancer, Ann. Oncol. Off. J. Eur. Soc. Med. Oncol, vol.24, pp.420-428, 2013.

H. Y. Wang, S. Ahn, S. Kim, S. Park, D. Jung et al., Detection of circulating tumor cell-specific markers in breast cancer patients using the quantitative RT-PCR assay, Int. J. Clin. Oncol, vol.20, pp.878-890, 2015.

G. Deng, S. Krishnakumar, A. A. Powell, H. Zhang, M. N. Mindrinos et al., 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, vol.14, p.456, 2014.

G. M. Cann, Z. G. Gulzar, S. Cooper, R. Li, S. Luo et al., mRNA-Seq of single prostate cancer circulating tumor cells reveals recapitulation of gene expression and pathways found in prostate cancer, PLoS ONE, vol.7, p.49144, 2012.

M. H. Park, E. Reategui, W. Li, S. N. Tessier, K. H. Wong et al., Enhanced Isolation and Release of Circulating Tumor Cells Using Nanoparticle Binding and Ligand Exchange in a Microfluidic Chip, J. Am. Chem. Soc, vol.139, pp.2741-2749, 2017.

G. Galletti, M. S. Sung, L. T. Vahdat, M. A. Shah, S. M. Santana et al., Isolation of breast cancer and gastric cancer circulating tumor cells by use of an anti HER2-based microfluidic device, Lab Chip, vol.14, pp.147-156, 2014.

H. J. Yoon, A. Shanker, Y. Wang, M. Kozminsky, Q. Jin et al., Tunable Thermal-Sensitive Polymer-Graphene Oxide Composite for Efficient Capture and Release of Viable Circulating Tumor Cells, Adv. Mater, vol.28, pp.4891-4897, 2016.

E. A. Punnoose, S. K. Atwal, J. M. Spoerke, H. Savage, A. Pandita et al., Molecular biomarker analyses using circulating tumor cells, PLoS ONE, vol.5, p.12517, 2010.

Y. Deng, Y. Zhang, S. Sun, Z. Wang, M. Wang et al., An integrated microfluidic chip system for single-cell secretion profiling of rare circulating tumor cells, Sci. Rep, vol.4, p.7499, 2014.

S. Wang, A. Thomas, E. Lee, S. Yang, X. Cheng et al., Highly efficient and selective isolation of rare tumor cells using a microfluidic chip with wavy-herringbone micro-patterned surfaces, Analyst, vol.141, pp.2228-2237, 2016.

J. Autebert, B. Coudert, J. Champ, L. Saias, E. T. Guneri et al., High purity microfluidic sorting and analysis of circulating tumor cells: Towards routine mutation detection, Lab Chip, vol.15, pp.2090-2101, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01852039

D. J. Klionsky, K. Abdelmohsen, A. Abe, M. J. Abedin, H. Abeliovich et al., Guidelines for the use and interpretation of assays for monitoring autophagy, Autophagy, vol.12, pp.1-222, 2016.
URL : https://hal.archives-ouvertes.fr/hal-00214269

S. M. Park, D. J. Wong, C. C. Ooi, D. M. Kurtz, O. Vermesh et al., Molecular profiling of single circulating tumor cells from lung cancer patients, Proc. Natl. Acad. Sci, vol.113, pp.8379-8386, 2016.

M. Gonzalez-rivera, A. C. Picornell, E. L. Alvarez, and M. Martin, 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, vol.5, p.167, 2016.

W. Harb, A. Fan, T. Tran, D. C. Danila, D. Keys et al., Mutational Analysis of Circulating Tumor Cells Using a Novel Microfluidic Collection Device and qPCR Assay, Transl. Oncol, vol.6, pp.528-538, 2013.

N. Aceto, A. Bardia, B. S. Wittner, M. C. Donaldson, R. O'keefe et al., AR Expression in Breast Cancer CTCs Associates with Bone Metastases, Mol. Cancer Res, vol.16, pp.720-727, 2018.

F. Fachin, P. Spuhler, J. M. Martel-foley, J. F. Edd, T. A. Barber et al., Monolithic Chip for High-throughput Blood Cell Depletion to Sort Rare Circulating Tumor Cells, Sci. Rep, vol.7, p.10936, 2017.

G. Theil, K. Fischer, E. Weber, R. Medek, R. Hoda et al., The Use of a New CellCollector to Isolate Circulating Tumor Cells from the Blood of Patients with Different Stages of Prostate Cancer and Clinical Outcomes-A Proof-of-Concept Study, PLoS ONE, vol.11, p.158354, 2016.

H. Zhang, S. Gong, Y. Liu, L. Liang, S. He et al., Enumeration and molecular characterization of circulating tumor cell using an in vivo capture system in squamous cell carcinoma of head and neck, Chin. J. Cancer Res, vol.29, pp.196-203, 2017.

M. Lapin, K. Tjensvoll, S. Oltedal, T. Buhl, B. Gilje et al., MINDEC-An Enhanced Negative Depletion Strategy for Circulating Tumour Cell Enrichment, Sci. Rep, 2016.

J. Yin, Y. Wang, H. Yin, W. Chen, G. Jin et al., Circulating Tumor Cells Enriched by the Depletion of Leukocytes with Bi-Antibodies in Non-Small Cell Lung Cancer: Potential Clinical Application, PLoS ONE, vol.10, p.137076, 2015.

Y. Wu, C. J. Deighan, B. L. Miller, P. Balasubramanian, M. B. Lustberg et al., Isolation and analysis of rare cells in the blood of cancer patients using a negative depletion methodology, vol.64, pp.169-182, 2013.

L. Zhang, L. D. Ridgway, M. D. Wetzel, J. Ngo, W. Yin et al., The identification and characterization of breast cancer CTCs competent for brain metastasis, Sci. Transl. Med, issue.5, pp.180-148, 2013.

V. Bobek, R. Matkowski, R. Gurlich, K. Grabowski, J. Szelachowska et al., Cultivation of circulating tumor cells in esophageal cancer, Folia Histochem. Cytobiol, vol.52, pp.171-177, 2014.

G. Hamilton, O. Burghuber, and R. Zeillinger, Circulating tumor cells in small cell lung cancer: Ex vivo expansion, Lung, vol.193, pp.451-452, 2015.

M. Cegan, K. Kolostova, R. Matkowski, M. Broul, J. Schraml et al., In vitro culturing of viable circulating tumor cells of urinary bladder cancer, Int. J. Clin. Exp. Pathol, vol.7, pp.7164-7171, 2014.

V. Bobek, R. Gurlich, P. Eliasova, and K. Kolostova, Circulating tumor cells in pancreatic cancer patients: Enrichment and cultivation, World J. Gastroenterol, vol.20, pp.17163-17170, 2014.

K. Kolostova, R. Matkowski, R. Gurlich, K. Grabowski, K. Soter et al., Detection and cultivation of circulating tumor cells in gastric cancer, Cytotechnology, vol.68, pp.1095-1102, 2016.

V. Bobek, G. Kacprzak, A. Rzechonek, and K. Kolostova, Detection and cultivation of circulating tumor cells in malignant pleural mesothelioma, Anticancer Res, vol.34, pp.2565-2569, 2014.

L. Klameth, B. Rath, M. Hochmaier, D. Moser, M. Redl et al., Small cell lung cancer: Model of circulating tumor cell tumorospheres in chemoresistance, vol.7, p.5337, 2017.

K. Duval, H. Grover, L. H. Han, Y. Mou, A. F. Pegoraro et al., Modeling Physiological Events in 2D vs. 3D Cell Culture, Physiology, vol.32, pp.266-277, 2017.

M. Delarue, F. Montel, D. Vignjevic, J. Prost, J. F. Joanny et al., Compressive stress inhibits proliferation in tumor spheroids through a volume limitation, Biophys. J, vol.107, pp.1821-1828, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01123922

J. Hoarau-vechot, A. Rafii, C. Touboul, and J. Pasquier, Halfway between 2D and Animal Models: Are 3D Cultures the Ideal Tool to Study Cancer-Microenvironment Interactions?, Int. J. Mol. Sci, vol.19, p.181, 2018.

M. Yu, A. Bardia, N. Aceto, F. Bersani, M. W. Madden et al., Cancer therapy. Ex vivo culture of circulating breast tumor cells for individualized testing of drug susceptibility, Science, vol.345, pp.216-220, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01948490

Z. Zhang, H. Shiratsuchi, J. Lin, G. Chen, R. M. Reddy et al., Expansion of CTCs from early stage lung cancer patients using a microfluidic co-culture model, Oncotarget, vol.5, pp.12383-12397, 2014.

S. Nath and G. R. Devi, Three-dimensional culture systems in cancer research: Focus on tumor spheroid model, Pharmacol. Ther, vol.163, pp.94-108, 2016.

Y. Zhang, X. Zhang, J. Zhang, B. Sun, L. Zheng et al., Microfluidic chip for isolation of viable circulating tumor cells of hepatocellular carcinoma for their culture and drug sensitivity assay, Cancer Biol. Ther, vol.17, pp.1177-1187, 2016.

M. Vishnoi, S. Peddibhotla, W. ;. Yin, T. S. George, G. C. Hong et al., The isolation and characterization of CTC subsets related to breast cancer dormancy, Sci. Rep, vol.5, p.17533, 2015.

Y. Takahashi, Y. Hori, T. Yamamoto, T. Urashima, Y. Ohara et al., 3D spheroid cultures improve the metabolic gene expression profiles of HepaRG cells, Biosci. Rep, vol.35, p.208, 2015.

K. Bloch, H. Smith, . Van-hamel, V. Parsons, D. Gavaghan et al., Metabolic alterations during the growth of tumour spheroids, Cell Biochem. Biophys, vol.68, pp.615-628, 2014.

J. A. Hickman, R. Graeser, R. De-hoogt, S. Vidic, C. Brito et al., Three-dimensional models of cancer for pharmacology and cancer cell biology: Capturing tumor complexity in vitro/ex vivo, Biotechnol. J, vol.9, pp.1115-1128, 2014.

Y. R. Lou and A. W. Leung, Next generation organoids for biomedical research and applications, Biotechnol. Adv, vol.36, pp.132-149, 2018.

D. Gao, I. Vela, A. Sboner, P. J. Iaquinta, W. R. Karthaus et al., Organoid cultures derived from patients with advanced prostate cancer, Cell, vol.159, pp.176-187, 2014.

F. Weeber, M. Van-de-wetering, M. Hoogstraat, K. K. Dijkstra, O. Krijgsman et al., Preserved genetic diversity in organoids cultured from biopsies of human colorectal cancer metastases, Proc. Natl. Acad. Sci, vol.112, pp.13308-13311, 2015.

H. Clevers, Modeling Development and Disease with Organoids, Cell, vol.165, pp.1586-1597, 2016.

J. Drost, R. H. Van-jaarsveld, B. Ponsioen, C. Zimberlin, R. Van-boxtel et al., Sequential cancer mutations in cultured human intestinal stem cells, Nature, vol.521, pp.43-47, 2015.

P. P. Praharaj, S. K. Bhutia, S. Nagrath, R. L. Bitting, and G. Deep, Circulating tumor cell-derived organoids: Current challenges and promises in medical research and precision medicine, Biochim. Biophys. Acta, vol.1869, pp.117-127, 2018.

Z. Zhang, H. Shiratsuchi, N. Palanisamy, S. Nagrath, and N. Ramnath, Expanded Circulating Tumor Cells from a Patient with ALK-Positive Lung Cancer Present with EML4-ALK Rearrangement Along with Resistance Mutation and Enable Drug Sensitivity Testing: A Case Study, J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer, vol.12, pp.397-402, 2017.

C. L. Hodgkinson, C. J. Morrow, Y. Li, R. L. Metcalf, D. G. Rothwell et al., Tumorigenicity and genetic profiling of circulating tumor cells in small-cell lung cancer, Nat. Med, vol.20, pp.897-903, 2014.

N. H. Tran, L. L. Cavalcante, S. J. Lubner, D. L. Mulkerin, N. K. Loconte et al., Precision medicine in colorectal cancer: The molecular profile alters treatment strategies, Ther. Adv. Med. Oncol, vol.7, pp.252-262, 2015.

K. Jin, L. Teng, Y. Shen, K. He, Z. Xu et al., Patient-derived human tumour tissue xenografts in immunodeficient mice: A systematic review, Clin. Transl. Oncol, vol.12, pp.473-480, 2010.

J. J. Tentler, A. C. Tan, C. D. Weekes, A. Jimeno, S. Leong et al., Patient-derived tumour xenografts as models for oncology drug development, Nat. Rev. Clin. Oncol, vol.9, pp.338-350, 2012.

E. Heitzer, M. Auer, C. Gasch, M. Pichler, P. Ulz et al., Complex tumor genomes inferred from single circulating tumor cells by array-CGH and next-generation sequencing, Cancer Res, vol.73, pp.2965-2975, 2013.

R. Bernards, A missing link in genotype-directed cancer therapy, Cell, vol.151, pp.465-468, 2012.

S. Scholch, S. A. Garcia, N. Iwata, T. Niemietz, A. M. Betzler et al., Circulating tumor cells exhibit stem cell characteristics in an orthotopic mouse model of colorectal cancer, Oncotarget, vol.7, pp.27232-27242, 2016.

D. Park, D. Wang, G. Chen, and X. Deng, Establishment of Patient-derived Xenografts in Mice, Bio-Protocol, vol.6, 2016.

A. Lallo, K. K. Frese, C. J. Morrow, R. Sloane, S. Gulati et al., The Combination of the PARP Inhibitor Olaparib and the WEE1 Inhibitor AZD1775 as a New Therapeutic Option for Small Cell Lung Cancer, Clin. Cancer Res, vol.24, pp.5153-5164, 2018.

B. J. Drapkin, J. George, C. L. Christensen, M. Mino-kenudson, R. Dries et al., Genomic and Functional Fidelity of Small Cell Lung Cancer Patient-Derived Xenografts, Cancer Discov, vol.8, pp.600-615, 2018.

S. Kimbung and N. Loman, Hedenfalk, I. Clinical and molecular complexity of breast cancer metastases, Semin. Cancer Biol, vol.35, pp.85-95, 2015.

C. J. Morrow, F. Trapani, R. L. Metcalf, G. Bertolini, C. L. Hodgkinson et al., Tumourigenic non-small-cell lung cancer mesenchymal circulating tumour cells: A clinical case study, Ann. Oncol. Off. J. Eur. Soc. Med. Oncol, vol.27, pp.1155-1160, 2016.

S. Khetani, M. Mohammadi, and A. S. Nezhad, Filter-based isolation, enrichment, and characterization of circulating tumor cells, Biotechnol. Bioeng, vol.115, pp.2504-2529, 2018.

J. Mong and M. H. Tan, Size-based enrichment technologies for non-cancerous tumor-derived cells in blood, Trends Biotechnol, vol.36, pp.511-522, 2018.

S. J. Hao, Y. Wan, Y. Q. Xia, X. Zou, and S. Y. Zheng, Size-based separation methods of circulating tumor cells, Adv. Drug Deliv. Rev, vol.125, pp.3-20, 2018.

Q. Kang, N. L. Henry, C. Paoletti, H. Jiang, P. Vats et al., Comparative analysis of circulating tumor DNA stability In K3EDTA, Streck, and CellSave blood collection tubes, Clin. Biochem, vol.49, pp.1354-1360, 2016.

H. K. Lin, S. Zheng, A. J. Williams, M. Balic, S. Groshen et al., Portable filter-based microdevice for detection and characterization of circulating tumor cells, Clin. Cancer Res, vol.16, pp.5011-5018, 2010.

K. Kolostova, M. Broul, J. Schraml, M. Cegan, R. Matkowski et al., Circulating tumor cells in localized prostate cancer: Isolation, cultivation in vitro and relationship to T-stage and Gleason score, Anticancer Res, vol.34, pp.3641-3646, 2014.

K. Kolostova, Y. Zhang, R. M. Hoffman, and V. Bobek, In vitro culture and characterization of human lung cancer circulating tumor cells isolated by size exclusion from an orthotopic nude-mouse model expressing fluorescent protein, J. Fluoresc, vol.24, pp.1531-1536, 2014.

K. Kolostova, J. Spicka, R. Matkowski, and V. Bobek, Isolation, primary culture, morphological and molecular characterization of circulating tumor cells in gynecological cancers, Am. J. Transl. Res, vol.7, pp.1203-1213, 2015.

B. L. Khoo, G. Grenci, T. Jing, Y. B. Lim, S. C. Lee et al., Liquid biopsy and therapeutic response: Circulating tumor cell cultures for evaluation of anticancer treatment, Sci. Adv, 2016.

B. L. Khoo, G. Grenci, Y. B. Lim, S. C. Lee, J. Han et al., Expansion of patient-derived circulating tumor cells from liquid biopsies using a CTC microfluidic culture device, Nat. Protoc, vol.13, pp.34-58, 2018.

R. Wang, G. C. Chu, S. Mrdenovic, A. A. Annamalai, A. E. Hendifar et al., Cultured circulating tumor cells and their derived xenografts for personalized oncology, Asian J. Urol, vol.3, pp.240-253, 2016.

Y. F. Sun, W. Guo, Y. Xu, Y. H. Shi, Z. J. Gong et al., Circulating Tumor Cells from Different Vascular Sites Exhibit Spatial Heterogeneity in Epithelial and Mesenchymal Composition and Distinct Clinical Significance in Hepatocellular Carcinoma, Clin. Cancer Res, vol.24, pp.547-559, 2018.

S. Daster, N. Amatruda, D. Calabrese, R. Ivanek, E. Turrini et al., Induction of hypoxia and necrosis in multicellular tumor spheroids is associated with resistance to chemotherapy treatment, Oncotarget, vol.8, pp.1725-1736, 2017.

O. I. Hoffmann, C. Ilmberger, S. Magosch, M. Joka, K. W. Jauch et al., Impact of the spheroid model complexity on drug response, J. Biotechnol, vol.205, pp.14-23, 2015.

T. B. Shelper, C. J. Lovitt, and V. M. Avery, Assessing Drug Efficacy in a Miniaturized Pancreatic Cancer In Vitro 3D Cell Culture Model, Assay Drug Dev. Technol, vol.14, pp.367-380, 2016.

J. L. Albritton and J. S. Miller, 3D bioprinting: Improving in vitro models of metastasis with heterogeneous tumor microenvironments, Dis. Model Mech, vol.10, pp.3-14, 2017.

P. Ter-brugge, P. Kristel, E. Van-der-burg, U. Boon, M. De-maaker et al., Mechanisms of Therapy Resistance in Patient-Derived Xenograft Models of BRCA1-Deficient Breast Cancer, J. Natl. Cancer Inst, vol.108, 2016.

R. Ito, T. Takahashi, and M. Ito, Humanized mouse models: Application to human diseases, J. Cell. Physiol, vol.233, pp.3723-3728, 2018.

, © 2018 by the authors. Submitted for possible open access publication under the terms and conditions of the Creative Commons Attribution (CC BY) license