Patient-Derived In Vitro Models for Drug Discovery in Colorectal Carcinoma
Abstract
:1. Introduction
2. Conventional CRC In Vitro Models
2.1. Patient-Derived Tumor Cell Lines Cultivated in Two Dimensions
2.2. Patient-Derived Cells Cultivated in Three Dimensions
3. CRC Patient-Derived In Vitro Models
3.1. Patient-Derived Tissue Slice Culture
3.2. Liquid Biopsy and Circulating Tumor Cells
3.3. Organ-on-Chip
3.4. Patient-Derived Organoids
3.5. Biomarkers-Based Drug Discovery
3.6. 3D Bioprinting
3.7. Clinical Point of View
4. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Siegel, R.L.; A Torre, L.; Soerjomataram, I.; Hayes, R.B.; Bray, F.; Weber, T.K.; Jemal, A. Global patterns and trends in colorectal cancer incidence in young adults. Gut 2019, 68, 2179–2185. [Google Scholar] [CrossRef] [Green Version]
- Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [Green Version]
- McQuade, R.; Stojanovska, V.; De Leiris, J.; Nurgali, K. Colorectal Cancer Chemotherapy: The Evolution of Treatment and New Approaches. Curr. Med. Chem. 2017, 24, 1537–1557. [Google Scholar] [CrossRef] [PubMed]
- Centenera, M.M.; Raj, G.V.; Knudsen, K.E.; Tilley, W.D.; Butler, L.M. Ex vivo culture of human prostate tissue and drug development. Nat. Rev. Urol. 2013, 10, 483–487. [Google Scholar] [CrossRef] [PubMed]
- Hutchinson, L.; Kirk, R. High drug attrition rates—Where are we going wrong? Nat. Rev. Clin. Oncol. 2011, 8, 189–190. [Google Scholar] [CrossRef] [PubMed]
- Uronis, J.M.; Osada, T.; McCall, S.J.; Yang, X.Y.; Mantyh, C.; Morse, M.A.; Lyerly, H.K.; Clary, B.M.; Hsu, D.S. Histological and Molecular Evaluation of Patient-Derived Colorectal Cancer Explants. PLoS ONE 2012, 7, e38422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Burgenske, D.M.; Monsma, D.J.; MacKeigan, J. Patient-Derived Xenograft Models of Colorectal Cancer: Procedures for Engraftment and Propagation. In Colorectal Cancer; Humana Press: New York, NY, USA, 2018; pp. 307–314. [Google Scholar] [CrossRef]
- Ji, X.; Chen, S.; Guo, Y.; Li, W.; Qi, X.; Yang, H.; Xiao, S.; Fang, G.; Hu, J.; Wen, C.; et al. Establishment and evaluation of four different types of patient-derived xenograft models. Cancer Cell Int. 2017, 17, 122. [Google Scholar] [CrossRef] [Green Version]
- Bertotti, A.; Migliardi, G.; Galimi, F.; Sassi, F.; Torti, D.; Isella, C.; Corà, D.; Di Nicolantonio, F.; Buscarino, M.; Petti, C.; et al. A Molecularly Annotated Platform of Patient-Derived Xenografts (“Xenopatients”) Identifies HER2 as an Effective Therapeutic Target in Cetuximab-Resistant Colorectal Cancer. Cancer Discov. 2011, 1, 508–523. [Google Scholar] [CrossRef] [Green Version]
- Jung, J.; Kim, J.; Lim, H.K.; Kim, K.M.; Lee, Y.S.; Park, J.S.; Yoon, D.-S. Establishing a colorectal cancer liver metastasis patient-derived tumor xenograft model for the evaluation of personalized chemotherapy. Ann. Surg. Treat. Res. 2017, 93, 173–180. [Google Scholar] [CrossRef] [Green Version]
- Maletzki, C.; Bock, S.; Fruh, P.; Macius, K.; Witt, A.; Prall, F.; Linnebacher, M. NSG mice as hosts for oncological precision medicine. Lab. Investig. 2019, 100, 27–37. [Google Scholar] [CrossRef] [PubMed]
- Brown, K.M.; Xue, A.; Mittal, A.; Samra, J.S.; Smith, R.; Hugh, T.J. Patient-derived xenograft models of colorectal cancer in pre-clinical research: A systematic review. Oncotarget 2016, 7, 66212–66225. [Google Scholar] [CrossRef] [Green Version]
- McIntyre, R.E.; Buczacki, S.J.; Arends, M.J.; Adams, D.J. Mouse models of colorectal cancer as preclinical models. BioEssays 2015, 37, 909–920. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gao, H.; Korn, J.M.; Ferretti, S.; Monahan, J.; Wang, Y.; Singh, M.; Zhang, C.; Schnell, C.; Yang, G.; Zhang, Y.; et al. High-throughput screening using patient-derived tumor xenografts to predict clinical trial drug response. Nat. Med. 2015, 21, 1318–1325. [Google Scholar] [CrossRef] [PubMed]
- Okada, S.; Vaeteewoottacharn, K.; Kariya, R. Application of Highly Immunocompromised Mice for the Establishment of Patient-Derived Xenograft (PDX) Models. Cells 2019, 8, 889. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Katsiampoura, A.; Raghav, K.; Jiang, Z.-Q.; Menter, D.G.; Varkaris, A.; Morelli, M.P.; Manuel, S.; Wu, J.; Sorokin, A.V.; Rizi, B.S.; et al. Modeling of Patient-Derived Xenografts in Colorectal Cancer. Mol. Cancer Ther. 2017, 16, 1435–1442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rubio-Viqueira, B.; Jimeno, A.; Cusatis, G.; Zhang, X.; Iacobuzio-Donahue, C.; Karikari, C.; Shi, C.; Danenberg, K.; Danenberg, P.V.; Kuramochi, H.; et al. An In vivo Platform for Translational Drug Development in Pancreatic Cancer. Clin. Cancer Res. 2006, 12, 4652–4661. [Google Scholar] [CrossRef] [Green Version]
- Hidalgo, M.; Bruckheimer, E.; RajeshKumar, N.V.; Garrido-Laguna, I.; De Oliveira, E.; Rubio-Viqueira, B.; Strawn, S.; Wick, M.J.; Martell, J.; Sidransky, D. A pilot clinical study of treatment guided by personalized tumorgrafts in patients with advanced cancer. Mol. Cancer Ther. 2011, 10, 1311–1316. [Google Scholar] [CrossRef] [Green Version]
- Stebbing, J.; Paz, K.; Schwartz, G.K.; Wexler, L.H.; Maki, R.G.; Pollock, R.E.; Morris, R.; Cohen, R.; Shankar, A.; Blackman, G.; et al. Patient-derived xenografts for individualized care in advanced sarcoma. Cancer 2014, 120, 2006–2015. [Google Scholar] [CrossRef] [Green Version]
- Fiore, D.; Di Giacomo, F.; Kyriakides, P.; Inghirami, G. Patient-Derived-Tumor-Xenograft: Modeling cancer for basic and translational cancer research. Clin. Diagn. Pathol. 2017, 1, 1. [Google Scholar] [CrossRef] [Green Version]
- Pauli, C.; Hopkins, B.D.; Prandi, D.; Shaw, R.; Fedrizzi, T.; Sboner, A.; Sailer, V.; Augello, M.; Puca, L.; Rosati, R.; et al. Personalized In Vitro and In Vivo Cancer Models to Guide Precision Medicine. Cancer Discov. 2017, 7, 462–477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chao, C.; Widen, S.G.; Wood, T.G.; Zatarain, J.R.; Johnson, P.; Gajjar, A.; Gomez, G.; Qiu, S.; Thompson, J.; Spratt, H.; et al. Patient-Derived Xenografts from Colorectal Carcinoma: A Temporal and Hierarchical Study of Murine Stromal Cell Replacement. Anticancer. Res. 2017, 37, 3405–3412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- O’Rourke, K.P.; Loizou, E.; Livshits, G.; Schatoff, E.M.; Baslan, T.; Manchado, E.; Simon, J.; Romesser, P.B.; Leach, B.; Han, T.; et al. Transplantation of engineered organoids enables rapid generation of metastatic mouse models of colorectal cancer. Nat. Biotechnol. 2017, 35, 577–582. [Google Scholar] [CrossRef] [Green Version]
- Céspedes, M.V.; Espina, C.; García-Cabezas, M.A.; Trias, M.; Boluda, A.; del Pulgar, M.T.G.; Sancho, F.J.; Nistal, M.; Lacal, J.C.; Mangues, R. Orthotopic Microinjection of Human Colon Cancer Cells in Nude Mice Induces Tumor Foci in All Clinically Relevant Metastatic Sites. Am. J. Pathol. 2007, 170, 1077–1085. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Swamy, M.V.; Patlolla, J.M.; Steele, V.E.; Kopelovich, L.; Reddy, B.S.; Rao, C.V. Chemoprevention of Familial Adenomatous Polyposis by Low Doses of Atorvastatin and Celecoxib Given Individually and in Combination to APCMinMice. Cancer Res. 2006, 66, 7370–7377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Washington, M.K.; Powell, A.E.; Sullivan, R.; Sundberg, J.P.; A Wright, N.; Coffey, R.J.; Dove, W. Pathology of rodent models of intestinal cancer: Progress report and recommendations. Gastroenterol. 2013, 144, 705–717. [Google Scholar] [CrossRef] [Green Version]
- Chen, J.; Huang, X.-F. The signal pathways in azoxymethane-induced colon cancer and preventive implications. Cancer Boil. Ther. 2009, 8, 1313–1317. [Google Scholar] [CrossRef] [Green Version]
- Young, M.; Ordonez, L.; Clarke, A. What are the best routes to effectively model human colorectal cancer? Mol. Oncol. 2013, 7, 178–189. [Google Scholar] [CrossRef]
- Golovko, D.; Kedrin, D.; Yilmaz, O.H.; Roper, J. Colorectal cancer models for novel drug discovery. Expert Opin. Drug Discov. 2015, 10, 1217–1229. [Google Scholar] [CrossRef]
- Tannenbaum, J.; Bennett, B.T. Russell and Burch’s 3Rs then and now: The need for clarity in definition and purpose. J. Am. Assoc. Lab. Anim. Sci. JAALAS 2015, 54, 120–132. [Google Scholar]
- Navarro, A.M.; Susanto, E.; Falk, A.; Wilhelm, M. Modeling cancer using patient-derived induced pluripotent stem cells to understand development of childhood malignancies. Cell Death Discov. 2018, 4, 7. [Google Scholar] [CrossRef] [PubMed]
- Curry, E.L.; Moad, M.; Robson, C.N.; Heer, R. Using induced pluripotent stem cells as a tool for modelling carcinogenesis. World J. Stem Cells 2015, 7, 461–469. [Google Scholar] [CrossRef] [PubMed]
- Jesudoss, M.X.D.; Sachinidis, A. Current Challenges of iPSC-Based Disease Modeling and Therapeutic Implications. Cells 2019, 8, 403. [Google Scholar] [CrossRef] [Green Version]
- Rowe, R.G.; Daley, G.Q. Induced pluripotent stem cells in disease modelling and drug discovery. Nat. Rev. Genet. 2019, 20, 377–388. [Google Scholar] [CrossRef]
- Suprynowicz, F.A.; Upadhyay, G.; Krawczyk, E.; Kramer, S.C.; Hebert, J.D.; Liu, X.; Yuan, H.; Cheluvaraju, C.; Clapp, P.; Boucher, R.C.; et al. Conditionally reprogrammed cells represent a stem-like state of adult epithelial cells. Proc. Natl. Acad. Sci. USA 2012, 109, 20035–20040. [Google Scholar] [CrossRef] [Green Version]
- Liu, X.; Krawczyk, E.; A Suprynowicz, F.; Palechor-Ceron, N.; Yuan, H.; Dakic, A.; Simic, V.; Zheng, Y.-L.; Sripadhan, P.; Chen, C.; et al. Conditional reprogramming and long-term expansion of normal and tumor cells from human biospecimens. Nat. Protoc. 2017, 12, 439–451. [Google Scholar] [CrossRef] [PubMed]
- Chapman, S.; Liu, X.; Meyers, C.; Schlegel, R.; McBride, A.A. Human keratinocytes are efficiently immortalized by a Rho kinase inhibitor. J. Clin. Investig. 2010, 120, 2619–2626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Terunuma, A.; Limgala, R.P.; Park, C.J.; Choudhary, I.; Vogel, J.C. Efficient Procurement of Epithelial Stem Cells from Human Tissue Specimens Using a Rho-Associated Protein Kinase Inhibitor Y-27632. Tissue Eng. Part A 2010, 16, 1363–1368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, Z.; Bi, B.; Song, H.; Liu, L.; Zheng, H.; Wang, S.; Shen, Z. Proliferation of human hepatocellular carcinoma cells from surgically resected specimens under conditionally reprogrammed culture. Mol. Med. Rep. 2019, 19, 4623–4630. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Timofeeva, O.A.; Palechor-Ceron, N.; Li, G.; Yuan, H.; Krawczyk, E.; Zhong, X.; Liu, G.; Upadhyay, G.; Dakic, A.; Yu, S.; et al. Conditionally reprogrammed normal and primary tumor prostate epithelial cells: A novel patient-derived cell model for studies of human prostate cancer. Oncotarget 2016, 8, 22741–22758. [Google Scholar] [CrossRef] [Green Version]
- Palechor-Ceron, N.; Krawczyk, E.; Dakic, A.; Simic, V.; Yuan, H.; Blancato, J.; Wang, W.; Hubbard, F.; Zheng, Y.L.; Dan, H.; et al. Conditional Reprogramming for Patient-Derived Cancer Models and Next-Generation Living Biobanks. Cells 2019, 8, 1327. [Google Scholar] [CrossRef] [Green Version]
- Correa, B.R.S.; Hu, J.; Penalva, L.O.F.; Schlegel, R.; Rimm, D.L.; Galante, P.A.F.; Agarwal, S. Patient-derived conditionally reprogrammed cells maintain intra-tumor genetic heterogeneity. Sci. Rep. 2018, 8, 4097. [Google Scholar] [CrossRef] [Green Version]
- Kodack, D.P.; Farago, A.F.; Dastur, A.; Held, M.A.; Dardaei, L.; Friboulet, L.; Von Flotow, F.; Damon, L.J.; Lee, D.-J.; Parks, M.; et al. Primary Patient-Derived Cancer Cells and Their Potential for Personalized Cancer Patient Care. Cell Rep. 2017, 21, 3298–3309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dame, M.K.; Bhagavathula, N.; Mankey, C.; DaSilva, M.; Paruchuri, T.; Aslam, M.N.; Varani, J. Human colon tissue in organ culture: Preservation of normal and neoplastic characteristics. Vitr. Cell. Dev. Boil. Anim. 2010, 46, 114–122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zirvi, K.A. Development of serum-free media for the growth of human gastrointestinal adenocarcinoma xenografts as primary tissue cultures. J. Cancer Res. Clin. Oncol. 1991, 117, 515–518. [Google Scholar] [CrossRef] [PubMed]
- Sato, T.; Stange, D.E.; Ferrante, M.; Vries, R.G.; Van Es, J.H.; Brink, S.V.D.; Van Houdt, W.J.; Pronk, A.; Van Gorp, J.M.; Siersema, P.D.; et al. Long-term Expansion of Epithelial Organoids from Human Colon, Adenoma, Adenocarcinoma, and Barrett’s Epithelium. Gastroenterology 2011, 141, 1762–1772. [Google Scholar] [CrossRef] [PubMed]
- Miyoshi, H.; Maekawa, H.; Kakizaki, F.; Yamaura, T.; Kawada, K.; Sakai, Y.; Taketo, M.M. An improved method for culturing patient-derived colorectal cancer spheroids. Oncotarget 2018, 9, 21950–21964. [Google Scholar] [CrossRef] [Green Version]
- Edmondson, R.; Broglie, J.J.; Adcock, A.F.; Yang, L. Three-Dimensional Cell Culture Systems and Their Applications in Drug Discovery and Cell-Based Biosensors. ASSAY Drug Dev. Technol. 2014, 12, 207–218. [Google Scholar] [CrossRef] [Green Version]
- Zanoni, M.; Piccinini, F.; Arienti, C.; Zamagni, A.; Santi, S.; Polico, R.; Bevilacqua, A.; Tesei, A. 3D tumor spheroid models for in vitro therapeutic screening: A systematic approach to enhance the biological relevance of data obtained. Sci. Rep. 2016, 6, 19103. [Google Scholar] [CrossRef]
- Cattin, S.; Ramont, L.; Ruegg, C. Characterization and In Vivo Validation of a Three-Dimensional Multi-Cellular Culture Model to Study Heterotypic Interactions in Colorectal Cancer Cell Growth, Invasion and Metastasis. Front. Bioeng. Biotechnol. 2018, 6, 97. [Google Scholar] [CrossRef]
- Zoetemelk, M.; Rausch, M.; Colin, D.J.; Dormond, O.; Nowak-Sliwinska, P. Short-term 3D culture systems of various complexity for treatment optimization of colorectal carcinoma. Sci. Rep. 2019, 9, 7103. [Google Scholar] [CrossRef] [PubMed]
- Jeppesen, M.; Hagel, G.; Glenthoj, A.; Vainer, B.; Ibsen, P.; Harling, H.; Thastrup, O.; Jørgensen, L.N.; Thastrup, J. Short-term spheroid culture of primary colorectal cancer cells as an in vitro model for personalizing cancer medicine. PLoS ONE 2017, 12, e0183074. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Weiswald, L.-B.; Richon, S.; Validire, P.; Briffod, M.; Lai-Kuen, R.; Cordelières, F.P.; Bertrand, F.; Dargere, D.; Massonnet, G.; Marangoni, E.; et al. Newly characterised ex vivo colospheres as a three-dimensional colon cancer cell model of tumour aggressiveness. Br. J. Cancer 2009, 101, 473–482. [Google Scholar] [CrossRef] [PubMed]
- Ashley, N.; Jones, M.; Ouaret, D.; Wilding, J.; Bodmer, W.F. Rapidly derived colorectal cancer cultures recapitulate parental cancer characteristics and enable personalized therapeutic assays. J. Pathol. 2014, 234, 34–45. [Google Scholar] [CrossRef]
- Kondo, J.; Endo, H.; Okuyama, H.; Ishikawa, O.; Iishi, H.; Tsujii, M.; Ohue, M.; Inoue, M. Retaining Cell-Cell Contact Enables Preparation and Culture of Spheroids Composed of Pure Primary Cancer Cells from Colorectal Cancer. Gastroenterology 2011, 140, 6235–6240. [Google Scholar] [CrossRef]
- Qureshi-Baig, K.; Ullmann, P.; Rodriguez, F.; Frasquilho, S.; Nazarov, P.V.; Haan, S.; Letellier, E. What Do We Learn from Spheroid Culture Systems? Insights from Tumorspheres Derived from Primary Colon Cancer Tissue. PLoS ONE 2016, 11, e0146052. [Google Scholar] [CrossRef]
- Hoffmann, O.; Ilmberger, C.; Magosch, S.; Joka, M.; Jauch, K.-W.; Mayer, B. Impact of the spheroid model complexity on drug response. J. Biotechnol. 2015, 205, 14–23. [Google Scholar] [CrossRef] [Green Version]
- Hirt, C.; Papadimitropoulos, A.; Mele, V.; Muraro, M.G.; Mengus, C.; Iezzi, G.; Terracciano, L.; Martin, I.; Spagnoli, G.C. “In vitro” 3D models of tumor-immune system interaction. Adv. Drug Deliv. Rev. 2014, 79, 145–154. [Google Scholar] [CrossRef]
- Courau, T.; Bonnereau, J.; Chicoteau, J.; Bottois, H.; Remark, R.; Miranda, L.A.; Toubert, A.; Bléry, M.; Aparicio, T.; Allez, M.; et al. Cocultures of human colorectal tumor spheroids with immune cells reveal the therapeutic potential of MICA/B and NKG2A targeting for cancer treatment. J. Immunother. Cancer 2019, 7, 74. [Google Scholar] [CrossRef] [Green Version]
- Failli, A.; Consolini, R.; Legitimo, A.; Spisni, R.; Castagna, M.; Romanini, A.; Crimaldi, G.; Miccoli, P. The challenge of culturing human colorectal tumor cells: Establishment of a cell culture model by the comparison of different methodological approaches. Tumori J. 2009, 95, 343–347. [Google Scholar] [CrossRef]
- Fan, F.; Bellister, S.; Lü, J.; Ye, X.; Boulbès, D.R.; Tozzi, F.; Sceusi, E.; Kopetz, S.; Tian, F.; Xia, L.; et al. The requirement for freshly isolated human colorectal cancer (CRC) cells in isolating CRC stem cells. Br. J. Cancer 2014, 112, 539–546. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pereira, J.F.D.S.; Awatade, N.T.; Loureiro, C.; Matos, P.; Amaral, M.D.; Jordan, P. The third dimension: New developments in cell culture models for colorectal research. Cell. Mol. Life Sci. 2016, 73, 3971–3989. [Google Scholar] [CrossRef] [PubMed]
- Meijer, T.G.; Naipal, K.A.; Jager, A.; Van Gent, D.C. Ex vivotumor culture systems for functional drug testing and therapy response prediction. Future Sci. OA 2017, 3, FSO190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Davies, E.J.; Dong, M.; Gutekunst, M.; Närhi, K.; Van Zoggel, H.J.A.A.; Blom, S.; Nagaraj, A.; Metsalu, T.; Oswald, E.; Erkens-Schulze, S.; et al. Capturing complex tumour biology in vitro: Histological and molecular characterisation of precision cut slices. Sci. Rep. 2015, 5, 17187. [Google Scholar] [CrossRef] [PubMed]
- Brand, D.V.D.; Massuger, L.F.; Brock, R.; Verdurmen, W.P. Mimicking Tumors: Toward More Predictive In Vitro Models for Peptide- and Protein-Conjugated Drugs. Bioconjug. Chem. 2017, 28, 846–856. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bläuer, M.; Tammela, T.L.; Ylikomi, T. A novel tissue-slice culture model for non-malignant human prostate. Cell Tissue Res. 2008, 332, 489–498. [Google Scholar] [CrossRef]
- Van De Merbel, M.; Van Der Horst, G.; Van Der Mark, M.H.; Van Uhm, J.I.M.; Van Gennep, E.J.; Kloen, P.; Beimers, L.; Pelger, R.C.M.; Van Der Pluijm, G. An ex vivo Tissue Culture Model for the Assessment of Individualized Drug Responses in Prostate and Bladder Cancer. Front. Oncol. 2018, 8. [Google Scholar] [CrossRef]
- Carranza-Torres, I.E.; Guzmán-Delgado, N.E.; Coronado-Martínez, C.; Bañuelos-García, J.I.; Valdez, E.V.; Morán-Martínez, J.; Carranza-Rosales, P. Organotypic Culture of Breast Tumor Explants as a Multicellular System for the Screening of Natural Compounds with Antineoplastic Potential. BioMed Res. Int. 2015, 2015, 618021. [Google Scholar] [CrossRef]
- Sönnichsen, R.; Hennig, L.; Blaschke, V.; Winter, K.; Körfer, J.; Hähnel, S.; Monecke, A.; Wittekind, C.; Jansen-Winkeln, B.; Thieme, R.; et al. Individual Susceptibility Analysis Using Patient-derived Slice Cultures of Colorectal Carcinoma. Clin. Color. Cancer 2018, 17, e189–e199. [Google Scholar] [CrossRef]
- Martin, S.Z.; Wagner, D.C.; Hörner, N.; Horst, D.; Lang, H.; Tagscherer, K.E.; Roth, W. Ex vivo tissue slice culture system to measure drug-response rates of hepatic metastatic colorectal cancer. BMC Cancer 2019, 19, 1030. [Google Scholar] [CrossRef]
- Zhang, Y.; Huang, W.; Yang, Q.; Zhang, H.; Zhu, X.; Zeng, M.; Zhou, X.; Wang, Z.; Li, W.; Jing, H.; et al. Cryopreserved biopsy tissues of rectal cancer liver metastasis for assessment of anticancer drug response in vitro and in vivo. Oncol. Rep. 2019, 43, 405–414. [Google Scholar] [CrossRef] [Green Version]
- Siravegna, G.; Marsoni, S.; Siena, S.; Bardelli, A. Integrating liquid biopsies into the management of cancer. Nat. Rev. Clin. Oncol. 2017, 14, 531–548. [Google Scholar] [CrossRef] [PubMed]
- Normanno, N.; Cervantes, A.; Ciardiello, F.; De Luca, A.; Pinto, C. The liquid biopsy in the management of colorectal cancer patients: Current applications and future scenarios. Cancer Treat. Rev. 2018, 70, 1–8. [Google Scholar] [CrossRef] [PubMed]
- Gkountela, S.; Castro-Giner, F.; Szczerba, B.M.; Vetter, M.; Landin, J.; Scherrer, R.; Krol, I.; Scheidmann, M.C.; Beisel, C.; Stirnimann, C.U.; et al. Circulating Tumor Cell Clustering Shapes DNA Methylation to Enable Metastasis Seeding. Cell 2019, 176, 98–112.e14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cabel, L.; Proudhon, C.; Gortais, H.; Loirat, D.; Coussy, F.; Pierga, J.-Y.; Bidard, F.-C. Circulating tumor cells: Clinical validity and utility. Int. J. Clin. Oncol. 2017, 22, 421–430. [Google Scholar] [CrossRef] [PubMed]
- Bork, U.; Rahbari, N.N.; Schölch, S.; Reissfelder, C.; Kahlert, C.; Büchler, M.W.; Weitz, J.; Koch, M. Circulating tumour cells and outcome in non-metastatic colorectal cancer: A prospective study. Br. J. Cancer 2015, 112, 1306–1313. [Google Scholar] [CrossRef]
- Aceto, N.; Bardia, A.; Miyamoto, D.T.; Donaldson, M.C.; Wittner, B.S.; Spencer, J.A.; Yu, M.; Pely, A.; Engstrom, A.; Zhu, H.; et al. Circulating tumor cell clusters are oligoclonal precursors of breast cancer metastasis. Cell 2014, 158, 1110–1122. [Google Scholar] [CrossRef] [Green Version]
- Andree, K.C.; Van Dalum, G.; Terstappen, L.W.M.M. Challenges in circulating tumor cell detection by the CellSearch system. Mol. Oncol. 2015, 10, 395–407. [Google Scholar] [CrossRef] [Green Version]
- Wang, L.; Balasubramanian, P.; Chen, A.P.; Kummar, S.; Evrard, Y.A.; Kinders, R.J. Promise and limits of the CellSearch platform for evaluating pharmacodynamics in circulating tumor cells. Semin. Oncol. 2016, 43, 464–475. [Google Scholar] [CrossRef] [Green Version]
- Williams, S.C.P. Circulating tumor cells. Proc. Natl. Acad. Sci. USA 2013, 110, 4861. [Google Scholar] [CrossRef] [Green Version]
- Gao, D.; Vela, I.; Sboner, A.; Iaquinta, P.J.; Karthaus, W.R.; Gopalan, A.; Dowling, C.; Wanjala, J.N.; Undvall, E.A.; Arora, V.K.; et al. Organoid cultures derived from patients with advanced prostate cancer. Cell 2014, 159, 176–187. [Google Scholar] [CrossRef]
- Zhang, L.; Ridgway, L.D.; Wetzel, M.D.; Ngo, J.; Yin, W.; Kumar, D.; Goodman, J.C.; Groves, M.D.; Marchetti, D. The Identification and Characterization of Breast Cancer CTCs Competent for Brain Metastasis. Sci. Transl. Med. 2013, 5, 180ra48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cayrefourcq, L.; Mazard, T.; Joosse, S.; Solassol, J.; Ramos, J.; Assenat, E.; Schumacher, U.; Costes-Martineau, V.; Maudelonde, T.; Pantel, K.; et al. Establishment and Characterization of a Cell Line from Human Circulating Colon Cancer Cells. Cancer Res. 2015, 75, 892–901. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Souglakos, J.; Androulakis, N.; Syrigos, K.; Polyzos, A.; Ziras, N.; Athanasiadis, A.; Kakolyris, S.; Tsousis, S.; Kouroussis, C.; Vamvakas, L.; et al. FOLFOXIRI (folinic acid, 5-fluorouracil, oxaliplatin and irinotecan) vs FOLFIRI (folinic acid, 5-fluorouracil and irinotecan) as first-line treatment in metastatic colorectal cancer (MCC): A multicentre randomised phase III trial from the Hellenic Oncology Research Group (HORG). Br. J. Cancer 2006, 94, 798–805. [Google Scholar] [CrossRef] [PubMed]
- Grillet, F.; Bayet, E.; Villeronce, O.; Zappia, L.; Lagerqvist, E.L.; Lunke, S.; Charafe-Jauffret, E.; Pham, K.; Molck, C.; Rolland, N.; et al. Circulating tumour cells from patients with colorectal cancer have cancer stem cell hallmarks inex vivoculture. Gut 2016, 66, 1802–1810. [Google Scholar] [CrossRef] [Green Version]
- 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] [Green Version]
- Zhou, J.; Kulasinghe, A.; Bogseth, A.; O’Byrne, K.; Punyadeera, C.; Papautsky, I. Isolation of circulating tumor cells in non-small-cell-lung-cancer patients using a multi-flow microfluidic channel. Microsyst. Nanoeng. 2019, 5, 8. [Google Scholar] [CrossRef] [Green Version]
- Esch, E.W.; Bahinski, A.; Huh, D. Organs-on-chips at the frontiers of drug discovery. Nat. Rev. Drug Discov. 2015, 14, 248–260. [Google Scholar] [CrossRef] [Green Version]
- Sontheimer-Phelps, A.; Hassell, B.A.; Ingber, D.E. Modelling cancer in microfluidic human organs-on-chips. Nat. Rev. Cancer 2019, 19, 65–81. [Google Scholar] [CrossRef]
- Zhang, Y.S.; Zhang, Y.-N.; Zhang, W. Cancer-on-a-chip systems at the frontier of nanomedicine. Drug Discov. Today 2017, 22, 1392–1399. [Google Scholar] [CrossRef]
- Akay, M.; Hite, J.; Avci, N.G.; Fan, Y.; Akay, Y.; Lu, G.; Zhu, J.-J. Drug Screening of Human GBM Spheroids in Brain Cancer Chip. Sci. Rep. 2018, 8, 15423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, P.-F.; Cao, Y.-W.; Zhang, S.-D.; Zhao, Y.; Liu, X.-G.; Shi, H.-Q.; Hu, K.-Y.; Zhu, G.-Q.; Ma, B.; Niu, H.-T. A bladder cancer microenvironment simulation system based on a microfluidic co-culture model. Oncotarget 2015, 6, 37695–37705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pradhan, S.; Smith, A.M.; Garson, C.J.; Hassani, I.; Seeto, W.J.; Pant, K.; Arnold, R.; Prabhakarpandian, B.; Lipke, E. A Microvascularized Tumor-mimetic Platform for Assessing Anti-cancer Drug Efficacy. Sci. Rep. 2018, 8, 3171. [Google Scholar] [CrossRef] [PubMed]
- Yu, T.; Guo, Z.; Fan, H.; Song, J.; Liu, Y.; Gao, Z.; Wang, Q. Cancer-associated fibroblasts promote non-small cell lung cancer cell invasion by upregulation of glucose-regulated protein 78 (GRP78) expression in an integrated bionic microfluidic device. Oncotarget 2016, 7, 25593–25603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zheng, Y.; Sun, Y.; Yu, X.; Shao, Y.; Zhang, P.; Dai, G.; Fu, J. Angiogenesis in Liquid Tumors: An In Vitro Assay for Leukemic-Cell-Induced Bone Marrow Angiogenesis. Adv. Heal. Mater. 2016, 5, 1014–1024. [Google Scholar] [CrossRef] [Green Version]
- Bein, A.; Shin, W.; Jalili-Firoozinezhad, S.; Park, M.H.; Sontheimer-Phelps, A.; Tovaglieri, A.; Chalkiadaki, A.; Kim, H.J.; Ingber, D.E. Microfluidic Organ-on-a-Chip Models of Human Intestine. Cell. Mol. Gastroenterol. Hepatol. 2018, 5, 659–668. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, H.J.; Li, H.; Collins, J.J.; Ingber, D.E. Contributions of microbiome and mechanical deformation to intestinal bacterial overgrowth and inflammation in a human gut-on-a-chip. Proc. Natl. Acad. Sci. USA 2015, 113, E7–E15. [Google Scholar] [CrossRef] [Green Version]
- Ahmad, A.A.; Wang, Y.; Gracz, A.D.; Sims, C.E.; Magness, S.T.; Allbritton, N.L. Optimization of 3-D organotypic primary colonic cultures for organ-on-chip applications. J. Boil. Eng. 2014, 8, 9. [Google Scholar] [CrossRef] [Green Version]
- Carvalho, M.; Barata, D.; Teixeira, L.S.M.; Giselbrecht, S.; Reis, R.L.; Oliveira, J.M.; Truckenmüller, R.; Habibovic, P. Colorectal tumor-on-a-chip system: A 3D tool for precision onco-nanomedicine. Sci. Adv. 2019, 5, eaaw1317. [Google Scholar] [CrossRef] [Green Version]
- Edwards, E.E.; Birmingham, K.G.; O’Melia, M.J.; Oh, J.; Thomas, S.N. Fluorometric Quantification of Single-Cell Velocities to Investigate Cancer Metastasis. Cell Syst. 2018, 7, 496–509.e6. [Google Scholar] [CrossRef] [Green Version]
- Zhao, Y.; Kankala, R.K.; Wang, S.-B.; Chen, A.-Z. Multi-Organs-on-Chips: Towards Long-Term Biomedical Investigations. Molecules 2019, 24, 675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Skardal, A.; Shupe, T.; Atala, A. Organoid-on-a-chip and body-on-a-chip systems for drug screening and disease modeling. Drug Discov. Today 2016, 21, 1399–1411. [Google Scholar] [CrossRef]
- Oleaga, C.; Bernabini, C.; Smith, A.S.; Srinivasan, B.; Jackson, M.; McLamb, W.; Platt, V.; Bridges, R.; Cai, Y.; Santhanam, N.; et al. Multi-Organ toxicity demonstration in a functional human in vitro system composed of four organs. Sci. Rep. 2016, 6, 20030. [Google Scholar] [CrossRef] [PubMed]
- Esch, M.B.; Ueno, H.; Applegate, D.R.; Shuler, M.L. Modular, pumpless body-on-a-chip platform for the co-culture of GI tract epithelium and 3D primary liver tissue. Lab Chip 2016, 16, 2719–2729. [Google Scholar] [CrossRef] [PubMed]
- Kasendra, M.; Tovaglieri, A.; Sontheimer-Phelps, A.; Jalili-Firoozinezhad, S.; Bein, A.; Chalkiadaki, A.; Scholl, W.; Zhang, C.; Rickner, H.; Richmond, C.A.; et al. Development of a primary human Small Intestine-on-a-Chip using biopsy-derived organoids. Sci. Rep. 2018, 8, 2871. [Google Scholar] [CrossRef]
- Lou, Y.-R.; Leung, A. Next generation organoids for biomedical research and applications. Biotechnol. Adv. 2018, 36, 132–149. [Google Scholar] [CrossRef]
- Dutta, D.; Heo, I.; Clevers, H. Disease Modeling in Stem Cell-Derived 3D Organoid Systems. Trends Mol. Med. 2017, 23, 393–410. [Google Scholar] [CrossRef]
- Drost, J.; Clevers, H. Organoids in cancer research. Nat. Rev. Cancer 2018, 18, 407–418. [Google Scholar] [CrossRef]
- Van De Wetering, M.; Francies, H.E.; Francis, J.M.; Bounova, G.; Iorio, F.; Pronk, A.; Van Houdt, W.; Van Gorp, J.; Taylor-Weiner, A.; Kester, L.; et al. Prospective derivation of a living organoid biobank of colorectal cancer patients. Cell 2015, 161, 933–945. [Google Scholar] [CrossRef] [Green Version]
- Nardella, C.; Lunardi, A.; Patnaik, A.; Cantley, L.C.; Pandolfi, P.P. The APL Paradigm and the “Co-Clinical Trial” Project. Cancer Discov. 2011, 2011, 108–116. [Google Scholar] [CrossRef] [Green Version]
- Vlachogiannis, G.; Hedayat, S.; Vatsiou, A.; Jamin, Y.; Fernández-Mateos, J.; Khan, K.; Lampis, A.; Eason, K.; Huntingford, I.; Burke, R.; et al. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science 2018, 359, 920–926. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, X.; Ootani, A.; Kuo, C. An Air–Liquid Interface Culture System for 3D Organoid Culture of Diverse Primary Gastrointestinal Tissues. In Gastrointestinal Physiology and Diseases: Methods and Protocols; Ivanov, A.I., Ed.; Springer: New York, NY, USA, 2016; pp. 33–40. [Google Scholar] [CrossRef]
- Katano, T.; Ootani, A.; Mizoshita, T.; Tanida, S.; Tsukamoto, H.; Ozeki, K.; Ebi, M.; Mori, Y.; Kataoka, H.; Kamiya, T.; et al. Establishment of a long-term three-dimensional primary culture of mouse glandular stomach epithelial cells within the stem cell niche. Biochem. Biophys. Res. Commun. 2013, 432, 558–563. [Google Scholar] [CrossRef] [PubMed]
- Li, X.; Nadauld, L.; Ootani, A.; Corney, D.C.; Pai, R.K.; Gevaert, O.; Cantrell, M.A.; Rack, P.D.; Neal, J.; Chan, C.W.-M.; et al. Oncogenic transformation of diverse gastrointestinal tissues in primary organoid culture. Nat. Med. 2014, 20, 769–777. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Elbadawy, M.; Usui, T.; Yamawaki, H.; Sasaki, K. Development of an Experimental Model for Analyzing Drug Resistance in Colorectal Cancer. Cancers 2018, 10, 164. [Google Scholar] [CrossRef] [Green Version]
- Lancaster, M.A.; Corsini, N.S.; Wolfinger, S.; Gustafson, E.H.; Phillips, A.; Burkard, T.R.; Otani, T.; Livesey, F.J.; Knoblich, J.A. Guided self-organization and cortical plate formation in human brain organoids. Nat. Biotechnol. 2017, 35, 659–666. [Google Scholar] [CrossRef] [Green Version]
- Urbischek, M.; Rannikmae, H.; Foets, T.; Ravn, K.; Hyvönen, M.; De La Roche, M. Organoid culture media formulated with growth factors of defined cellular activity. Sci. Rep. 2019, 9, 6193. [Google Scholar] [CrossRef] [Green Version]
- Lancaster, M.A.; Huch, M. Disease modelling in human organoids. Dis. Model. Mech. 2019, 12, dmm039347. [Google Scholar] [CrossRef] [Green Version]
- Sasaki, N.; Clevers, H. Studying cellular heterogeneity and drug sensitivity in colorectal cancer using organoid technology. Curr. Opin. Genet. Dev. 2018, 52, 117–122. [Google Scholar] [CrossRef]
- Finnberg, N.K.; Gokare, P.; Lev, A.; Grivennikov, S.I.; Macfarlane, A.W.; Campbell, K.S.; Winters, R.M.; Kaputa, K.; Farma, J.M.; Abbas, A.E.-S.; et al. Application of 3D tumoroid systems to define immune and cytotoxic therapeutic responses based on tumoroid and tissue slice culture molecular signatures. Oncotarget 2017, 8, 66747–66757. [Google Scholar] [CrossRef] [Green Version]
- Schettini, F.; Buono, G.; Cardalesi, C.; Desideri, I.; De Placido, S.; Del Mastro, L. Hormone Receptor/Human Epidermal Growth Factor Receptor 2-positive breast cancer: Where we are now and where we are going? Cancer Treat. Rev. 2016, 46, 20–26. [Google Scholar] [CrossRef] [Green Version]
- Salles, G.; Barrett, M.; Foà, R.; Maurer, J.; O’Brien, S.; Valente, N.; Wenger, M.; Maloney, D.G. Rituximab in B-Cell Hematologic Malignancies: A Review of 20 Years of Clinical Experience. Adv. Ther. 2017, 34, 2232–2273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ciombor, K.K.; Goldberg, R. Hypermutated Tumors and Immune Checkpoint Inhibition. Drugs 2018, 78, 155–162. [Google Scholar] [CrossRef] [PubMed]
- Gibney, G.T.; Weiner, L.M.; Atkins, M.B. Predictive biomarkers for checkpoint inhibitor-based immunotherapy. Lancet Oncol. 2016, 17, e542–e551. [Google Scholar] [CrossRef] [Green Version]
- Singh, M.; Jadhav, H.R. Targeting non-small cell lung cancer with small-molecule EGFR tyrosine kinase inhibitors. Drug Discov. Today 2018, 23, 745–753. [Google Scholar] [CrossRef]
- Luke, J.J.; Flaherty, K.T.; Ribas, A.; Long, G.V. Targeted agents and immunotherapies: Optimizing outcomes in melanoma. Nat. Rev. Clin. Oncol. 2017, 14, 463–482. [Google Scholar] [CrossRef] [Green Version]
- Giesen, C.; A O Wang, H.; Schapiro, D.; Zivanovic, N.; Jacobs, A.; Hattendorf, B.; Schüffler, P.; Grolimund, D.; Buhmann, J.M.; Brandt, S.; et al. Highly multiplexed imaging of tumor tissues with subcellular resolution by mass cytometry. Nat. Methods 2014, 11, 417–422. [Google Scholar] [CrossRef]
- O’Connor, M.J. Targeting the DNA Damage Response in Cancer. Mol. Cell 2015, 60, 547–560. [Google Scholar] [CrossRef] [Green Version]
- Coppé, J.-P.; Mori, M.; Pan, B.; Yau, C.; Wolf, D.M.; Ruiz-Saenz, A.; Brunen, D.; Prahallad, A.; Cornelissen-Steijger, P.; Kemper, K.; et al. Mapping phospho-catalytic dependencies of therapy-resistant tumours reveals actionable vulnerabilities. Nature 2019, 21, 778–790. [Google Scholar] [CrossRef]
- Kopetz, S.; Desai, J.; Chan, E.; Hecht, J.R.; O’Dwyer, P.J.; Maru, D.; Morris, V.; Janku, F.; Dasari, A.; Chung, W.; et al. Phase II Pilot Study of Vemurafenib in Patients with Metastatic BRAF-Mutated Colorectal Cancer. J. Clin. Oncol. 2015, 33, 4032–4038. [Google Scholar] [CrossRef]
- Xu, T.; Jin, J.; Gregory, C.; Hickman, J.J.; Boland, T. Inkjet printing of viable mammalian cells. Biomaterials 2005, 26, 93–99. [Google Scholar] [CrossRef]
- Murphy, S.V.; Atala, A. 3D bioprinting of tissues and organs. Nat. Biotechnol. 2014, 32, 773–785. [Google Scholar] [CrossRef] [PubMed]
- Moroni, L.; Burdick, J.A.; Highley, C.; Lee, S.J.; Morimoto, Y.; Takeuchi, S.; Yoo, J.J. Biofabrication strategies for 3D in vitro models and regenerative medicine. Nat. Rev. Mater. 2018, 3, 21–37. [Google Scholar] [CrossRef] [PubMed]
- Hospodiuk, M.; Dey, M.; Sosnoski, D.; Ozbolat, I.T. The bioink: A comprehensive review on bioprintable materials. Biotechnol. Adv. 2017, 35, 217–239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Satpathy, A.; Datta, P.; Wu, Y.; Ayan, B.; Bayram, E.; Ozbolat, I.T. Developments with 3D bioprinting for novel drug discovery. Expert Opin. Drug Discov. 2018, 13, 1115–1129. [Google Scholar] [CrossRef] [PubMed]
- Madden, L.R.; Nguyen, T.V.; Garcia-Mojica, S.; Shah, V.; Le, A.V.; Peier, A.; Visconti, R.; Parker, E.M.; Presnell, S.C.; Nguyen, D.G.; et al. Bioprinted 3D Primary Human Intestinal Tissues Model Aspects of Native Physiology and ADME/Tox Functions. iScience 2018, 2, 156–167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sambuy, Y.; De Angelis, I.; Ranaldi, G.; Scarino, M.L.; Stammati, A.; Zucco, F. The Caco-2 cell line as a model of the intestinal barrier: Influence of cell and culture-related factors on Caco-2 cell functional characteristics. Cell Boil. Toxicol. 2005, 21, 1–26. [Google Scholar] [CrossRef]
- McDonnell, A.M.; Dang, C. Basic Review of the Cytochrome P450 System. J. Adv. Pract. Oncol. 2013, 4, 263–268. [Google Scholar] [CrossRef]
- Langer, E.M.; Allen-Petersen, B.; King, S.M.; Kendsersky, N.D.; Turnidge, M.A.; Kuziel, G.M.; Riggers, R.; Samatham, R.; Amery, T.S.; Jacques, S.L.; et al. Modeling Tumor Phenotypes In Vitro with Three-Dimensional Bioprinting. Cell Rep. 2019, 26, 608–623.e6. [Google Scholar] [CrossRef] [Green Version]
- Colosi, C.; Shin, S.R.; Manoharan, V.; Massa, S.; Costantini, M.; Barbetta, A.; Dokmeci, M.R.; Dentini, M.; Khademhosseini, A. Microfluidic Bioprinting of Heterogeneous 3D Tissue Constructs Using Low-Viscosity Bioink. Adv. Mater. 2015, 28, 677–684. [Google Scholar] [CrossRef]
- Grigoryan, B.; Paulsen, S.J.; Corbett, D.C.; Sazer, D.W.; Fortin, C.L.; Zaita, A.J.; Greenfield, P.T.; Calafat, N.J.; Gounley, J.; Ta, A.H.; et al. Multivascular networks and functional intravascular topologies within biocompatible hydrogels. Science 2019, 364, 458–464. [Google Scholar] [CrossRef]
- Wilhelm, S.M.; Dumas, J.; Adnane, L.; Lynch, M.; Carter, C.A.; Schütz, G.; Thierauch, K.-H.; Zopf, D. Regorafenib (BAY 73-4506): A new oral multikinase inhibitor of angiogenic, stromal and oncogenic receptor tyrosine kinases with potent preclinical antitumor activity. Int. J. Cancer 2011, 129, 245–255. [Google Scholar] [CrossRef] [PubMed]
- Fang, Y.; Eglen, R.M. Three-Dimensional Cell Cultures in Drug Discovery and Development. SLAS Discov. Adv. Life Sci. R&D 2017, 22, 456–472. [Google Scholar] [CrossRef] [Green Version]
- Horvath, P.; Aulner, N.; Bickle, M.; Davies, A.M.; Del Nery, E.; Ebner, D.; Montoya, M.; Östling, P.; Pietiäinen, V.; Price, L.; et al. Screening out irrelevant cell-based models of disease. Nat. Rev. Drug Discov. 2016, 15, 751–769. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Prahallad, A.; Sun, C.; Huang, S.; Di Nicolantonio, F.; Salazar, R.; Zecchin, D.; Beijersbergen, R.L.; Bardelli, A.; Bernards, R. Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature 2012, 483, 100–103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Van Cutsem, E.; Huijberts, S.; Grothey, A.; Yaeger, R.; Cuyle, P.-J.; Elez, E.; Fakih, M.; Montagut, C.; Peeters, M.; Yoshino, T.; et al. Binimetinib, Encorafenib, and Cetuximab Triplet Therapy for Patients With BRAF V600E–Mutant Metastatic Colorectal Cancer: Safety Lead-In Results From the Phase III BEACON Colorectal Cancer Study. J. Clin. Oncol. 2019, 37, 1460–1469. [Google Scholar] [CrossRef] [PubMed]
- Rolfo, C.; Mack, P.C.; Scagliotti, G.V.; Baas, P.; Barlesi, F.; Bivona, T.G.; Herbst, R.S.; Mok, T.S.; Peled, N.; Pirker, R.; et al. Liquid Biopsy for Advanced Non-Small Cell Lung Cancer (NSCLC): A Statement Paper from the IASLC. J. Thorac. Oncol. 2018, 13, 1248–1268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bychkov, D.; Linder, N.; Turkki, R.; Nordling, S.; Kovanen, P.E.; Verrill, C.; Walliander, M.; Lundin, M.; Haglund, C.; Lundin, J. Deep learning based tissue analysis predicts outcome in colorectal cancer. Sci. Rep. 2018, 8, 3395. [Google Scholar] [CrossRef]
- FOxTROT Collaborative Group. Feasibility of preoperative chemotherapy for locally advanced, operable colon cancer: The pilot phase of a randomised controlled trial. Lancet Oncol. 2012, 13, 1152–1160. [Google Scholar] [CrossRef] [Green Version]
- Kamps, R.; Brandão, R.; Bosch, B.J.V.D.; Paulussen, A.D.C.; Xanthoulea, S.; Blok, M.J.; Romano, A. Next-Generation Sequencing in Oncology: Genetic Diagnosis, Risk Prediction and Cancer Classification. Int. J. Mol. Sci. 2017, 18, 308. [Google Scholar] [CrossRef]
- Berger, M.F.; Mardis, E.R. The emerging clinical relevance of genomics in cancer medicine. Nat. Rev. Clin. Oncol. 2018, 15, 353–365. [Google Scholar] [CrossRef]
- Figueiras, R.G.; Baleato-González, S.; Padhani, A.R.; Luna-Alcalá, A.; Marhuenda, A.; Vilanova, J.C.; Osorio-Vázquez, I.; Martinez-De-Alegria, A.; Gomez-Caamaño, A. Advanced Imaging Techniques in Evaluation of Colorectal Cancer. Radiographics 2018, 38, 740–765. [Google Scholar] [CrossRef] [Green Version]
- Nowak-Sliwinska, P.; Weiss, A.; Ding, X.; Dyson, P.J.; Bergh, H.V.D.; Griffioen, A.W.; Ho, C.-M. Optimization of drug combinations using Feedback System Control. Nat. Protoc. 2016, 11, 302–315. [Google Scholar] [CrossRef] [PubMed]
- Ding, X.; Liu, W.; Weiss, A.; Li, Y.; Wong, I.; Griffioen, A.W.; Bergh, H.V.D.; Xu, H.; Nowak-Sliwinska, P.; Ho, C.-M. Discovery of a low order drug-cell response surface for applications in personalized medicine. Phys. Boil. 2014, 11, 065003. [Google Scholar] [CrossRef] [PubMed]
- Weiss, A.; Nowak-Sliwinska, P. Current Trends in Multidrug Optimization: An Alley of Future Successful Treatment of Complex Disorders. SLAS Technol. Transl. Life Sci. Innov. 2016, 22, 254–275. [Google Scholar] [CrossRef] [PubMed]
- Weiss, A.; Le Roux-Bourdieu, M.; Zoetemelk, M.; Ramzy, G.; Rausch, M.; Harry, D.; Miljkovic-Licina, M.; Falamaki, K.; Wehrle-Haller, B.; Meraldi, P.; et al. Identification of a Synergistic Multi-Drug Combination Active in Cancer Cells via the Prevention of Spindle Pole Clustering. Cancers 2019, 11, 1612. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Prasetyanti, P.R.; Van Hooff, S.R.; Van Herwaarden, T.; De Vries, N.; Kalloe, K.; Rodermond, H.; Van Leersum, R.; De Jong, J.H.; Franitza, M.; Nürnberg, P.; et al. Capturing colorectal cancer inter-tumor heterogeneity in patient-derived xenograft (PDX) models. Int. J. Cancer 2018, 144, 366–371. [Google Scholar] [CrossRef]
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Ramzy, G.M.; Koessler, T.; Ducrey, E.; McKee, T.; Ris, F.; Buchs, N.; Rubbia-Brandt, L.; Dietrich, P.-Y.; Nowak-Sliwinska, P. Patient-Derived In Vitro Models for Drug Discovery in Colorectal Carcinoma. Cancers 2020, 12, 1423. https://doi.org/10.3390/cancers12061423
Ramzy GM, Koessler T, Ducrey E, McKee T, Ris F, Buchs N, Rubbia-Brandt L, Dietrich P-Y, Nowak-Sliwinska P. Patient-Derived In Vitro Models for Drug Discovery in Colorectal Carcinoma. Cancers. 2020; 12(6):1423. https://doi.org/10.3390/cancers12061423
Chicago/Turabian StyleRamzy, George M., Thibaud Koessler, Eloise Ducrey, Thomas McKee, Frédéric Ris, Nicolas Buchs, Laura Rubbia-Brandt, Pierre-Yves Dietrich, and Patrycja Nowak-Sliwinska. 2020. "Patient-Derived In Vitro Models for Drug Discovery in Colorectal Carcinoma" Cancers 12, no. 6: 1423. https://doi.org/10.3390/cancers12061423
APA StyleRamzy, G. M., Koessler, T., Ducrey, E., McKee, T., Ris, F., Buchs, N., Rubbia-Brandt, L., Dietrich, P. -Y., & Nowak-Sliwinska, P. (2020). Patient-Derived In Vitro Models for Drug Discovery in Colorectal Carcinoma. Cancers, 12(6), 1423. https://doi.org/10.3390/cancers12061423