Trends in Bone Metastasis Modeling
Abstract
1. Introduction
2. Metastasis Process and Bone Metastatic Microenvironment
2.1. 3D Models
2.2. 3D Models and Applications to Bone Cancer Metastasis Studies
2.2.1. 3D Scaffold-Free Systems-Cells Spheroids
2.2.2. Organoids
2.2.3. 3D Scaffold or Hydrogels Based Tumor Models
2.3. Cultivation and Biofabrication Systems
2.3.1. Bioreactors
2.3.2. Microfluidic
2.3.3. Organ-on-a Chip
2.3.4. Bioprinting
2.4. Ex Vivo Models
2.4.1. Main Application of Ex Vivo Explants: Viability for Long Term Culture, Analysis of Tissue Architecture, and Response to Therapies
2.4.2. Ex vivo Bone Metastasis Models and Applications
2.4.3. A Comparison between In Vitro and Ex Vivo Models
3. Discussion
4. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- McGranahan, N.; Swanton, C. Clonal Heterogeneity and Tumor Evolution: Past, Present, and the Future. Cell 2017, 168, 613–628. [Google Scholar] [CrossRef] [PubMed]
- Davila, D.; Antoniou, A.; Chaudhry, M.A. Evaluation of osseous metastasis in bone scintigraphy. Semin. Nucl. Med. 2015, 45, 3–15. [Google Scholar] [CrossRef] [PubMed]
- Mark, R.; Wick, M.D. Metastases to Bones. Seminars in Diagnostic Pathology. 2014. Available online: https://acsjournals.onlinelibrary.wiley.com/doi/full/10.1002/cncr.23151 (accessed on 27 November 2007).
- Yan, W.; Tu, B.; Liu, Y.Y.; Wang, T.Y.; Qiao, H.; Zhai, Z.J.; Li, H.W.; Tang, T.T. Suppressive Effects of Plumbagin on Invasion and Migration of Breast Cancer Cells via the Inhibition of STAT3 Signaling and Down-regulation of Inflammatory Cytokine Expressions. Bone Res. 2013, 1, 362–370. [Google Scholar] [CrossRef] [PubMed]
- Salamanna, F.; Contartese, D.; Maglio, M.; Fini, M. A systematic review on in vitro 3d bone metastases models. A new horizon to recapitulate the native clinical scenario? Oncotarget 2016, 7, 44803–44820. [Google Scholar] [CrossRef]
- Fatehullah, A.; Tan, S.H.; Barker, N. Organoids as an in vitro model of human development and disease. Nat. Cell Biol. 2016, 18, 246–254. [Google Scholar] [CrossRef]
- Brancato, V.; Oliveira, J.M.; Correlo, V.M.; Reis, R.L.; Kundu, S.C. Could 3D models of cancer enhance drug screening? Biomaterials 2020, 232, 119744. [Google Scholar] [CrossRef]
- Van Tienderen, G.S.; Koerkamp, B.G.; Ijzermans, J.N.M.; van der Laan, L.J.W.; Verstegen, M.M.A. Recreating tumour complexity in a dish: Organoid models to study liver cancer cells and their extracellular environment. Cancers 2019, 11, 1706. [Google Scholar] [CrossRef]
- Paget, S. The distribution of secondary growths in cancer of the breast. Lancet 1889, 133, 571–573. [Google Scholar] [CrossRef]
- Ewing, J. Neoplastic Diseases: A Treatise on Tumours. By James Ewing, A.M., M.D., Sc.D., Professor of Pathology at Cornell University Medical College, N.Y.; Pathologist to the Memorial Hospital. Third edition. Royal 8vo. Pp. 1127, with 546 illustrations. Phil. Br. J. Surg. 1928, 16, 174–175. Available online: https://bjssjournals.onlinelibrary.wiley.com/doi/abs/10.1002/bjs.1800166126 (accessed on 6 December 2005).
- Fidler, I.J.; Poste, G. The biologic diversity of cancer metastases. Hosp. Pract. 1982, 17, 57–64. [Google Scholar] [CrossRef]
- Psaila, B.; Lyden, D. The metastatic niche: Adapting the foreign soil. Nat. Rev. Cancer 2009, 9, 285–293. [Google Scholar] [CrossRef] [PubMed]
- Kaplan, R.N.; Riba, R.D.; Zacharoulis, S.; Bramley, A.H.; Vincent, L.; Costa, C.; MacDonald, D.D.; Jin, D.K.; Shido, K.; Kerns, S.A.; et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature 2005, 438, 820–827. [Google Scholar] [CrossRef] [PubMed]
- Sleeman, J.P. The lymph node pre-metastatic niche. J. Mol. Med. 2015, 93, 1173–1184. [Google Scholar] [CrossRef] [PubMed]
- Peinado, H.; Zhang, H.; Matei, I.R.; Costa-Silva, B.; Hoshino, A.; Rodrigues, G.; Psaila, B.; Kaplan, R.N.; Bromberg, J.F.; Kang, Y.; et al. Pre-metastatic niches: Organ-specific homes for metastases. Nat. Rev. Cancer 2017, 17, 302–317. [Google Scholar] [CrossRef]
- Sleeman, J.P.; Christofori, G.; Fodde, R.; Collard, J.G.; Berx, G.; Decraene, C.; Rüegg, C. Concepts of metastasis in flux: The stromal progression model. Semin. Cancer Biol. 2012, 22, 174–186. [Google Scholar] [CrossRef]
- Ren, G.; Esposito, M.; Kang, Y. Bone metastasis and the metastatic niche. J. Mol. Med. 2015, 93, 1203–1212. [Google Scholar] [CrossRef]
- Shen, Y.; Nilsson, S.K. Bone, microenvironment and hematopoiesis. Curr. Opin. Hematol. 2012, 19, 250–255. [Google Scholar] [CrossRef]
- Frenette, P.S.; Pinho, S.; Lucas, D.; Scheiermann, C. Mesenchymal Stem Cell: Keystone of the Hematopoietic Stem Cell Niche and a Stepping-Stone for Regenerative Medicine. Annu. Rev. Immunol. 2013, 31, 285–316. [Google Scholar] [CrossRef]
- Morrison, S.J.; Scadden, D.T. The bone marrow niche for haematopoietic stem cells. Nature 2014, 505, 327–334. [Google Scholar] [CrossRef]
- Méndez-Ferrer, S.; Michurina, T.V.; Ferraro, F.; Mazloom, A.R.; MacArthur, B.D.; Lira, S.A.; Scadden, D.T.; Ma’ayan, A.; Enikolopov, G.N.; Frenette, P.S. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature 2010, 466, 829–834. [Google Scholar] [CrossRef]
- Ng, A.P.; Alexander, W.S. Haematopoietic stem cells: Past, present and future. Cell Death Discov. 2017, 3, 2–5. [Google Scholar] [CrossRef]
- Coughlin, T.R.; Romero-Moreno, R.; Mason, D.E.; Nystrom, L.; Boerckel, J.D.; Niebur, G.; Littlepage, L.E. Bone: A Fertile Soil for Cancer Metastasis. Curr. Drug Targets 2017, 18, 1281–1295. [Google Scholar] [CrossRef]
- Guise, T. Examining the metastatic niche: Targeting the microenvironment. Semin. Oncol. 2010, 37 (Suppl 2), S2–S14. [Google Scholar] [CrossRef]
- Roodman, G.D. Mechanisms of Bone Metastasis. N. Engl. J. Med. 2004, 350, 1655–1664. [Google Scholar] [CrossRef] [PubMed]
- Fazilaty, H.; Mehdipour, P. Genetics of breast cancer bone metastasis: A sequential multistep pattern. Clin. Exp. Metastasis 2014, 31, 595–612. [Google Scholar] [CrossRef] [PubMed]
- Qiao, H.; Tang, T. Engineering 3D approaches to model the dynamic microenvironments of cancer bone metastasis. Bone Res. 2018, 6, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Auguste, P.; Lemiere, S.; Larrieu-Lahargue, F.; Bikfalvi, A. Molecular mechanisms of tumor vascularization. Crit. Rev. Oncol. Hematol. 2005, 54, 53–61. [Google Scholar] [CrossRef]
- Thiery, J.P.; Sleeman, J.P. Complex networks orchestrate epithelial-mesenchymal transitions. Nat. Rev. Mol. Cell Biol. 2006, 7, 131–142. [Google Scholar] [CrossRef]
- Douma, S.; Van Laar, T.; Zevenhoven, J.; Meuwissen, R.; Van Garderen, E.; Peeper, D.S. Suppression of anoikis and induction of metastasis by the neurotrophic receptor TrkB. Nature 2004, 430, 1034–1040. [Google Scholar] [CrossRef]
- Giancotti, F.G. HHS Public Access. Cell 2013, 155, 750–764. [Google Scholar] [CrossRef]
- Hensel, J.; Thalmann, G.N. Biology of Bone Metastases in Prostate Cancer. Urology 2016, 92, 6–13. [Google Scholar] [CrossRef] [PubMed]
- Fitzgerald, K.A.; Malhotra, M.; Curtin, C.M.; O’Brien, F.J.; O’Driscoll, C.M. Life in 3D is never flat: 3D models to optimise drug delivery. J. Control. Release 2015, 215, 39–54. [Google Scholar] [CrossRef] [PubMed]
- Kinney, M.A.; Hookway, T.A.; Wang, Y.; McDevitt, T.C. Eingineering three-diemnsional stem cell morphogenesis for the development of tissue models and scalable regenerative therapeutics. Ann Biomed Eng. 2014, 42, 352–367. [Google Scholar] [CrossRef] [PubMed]
- Grinnell, F. Fibroblast biology in three-dimensional collagen matrices. Trends Cell Biol. 2003, 13, 264–269. [Google Scholar] [CrossRef]
- Peela, N.; Sam, F.S.; Christenson, W.; Truong, D.; Watson, A.W.; Mouneimne, G.; Ros, R.; Nikkhah, M. A three dimensional micropatterned tumor model for breast cancer cell migration studies. Biomaterials 2016, 81, 72–83. [Google Scholar] [CrossRef]
- Chen, S.H.; Lei, M.; Xie, X.H.; Zheng, L.Z.; Yao, D.; Wang, X.L.; Li, W.; Zhao, Z.; Kong, A.; Xiao, D.M.; et al. PLGA/TCP composite scaffold incorporating bioactive phytomolecule icaritin for enhancement of bone defect repair in rabbits. Acta Biomater. 2013, 9, 6711–6722. [Google Scholar] [CrossRef]
- Candini, O.; Grisendi, G.; Foppiani, E.M.; Brogli, M.; Aramini, B.; Masciale, V.; Spano, C.; Petrachi, T.; Veronesi, E.; Conte, P.; et al. A Novel 3D In vitro Platform for Pre-Clinical Investigations in Drug Testing, Gene Therapy, and Immuno-oncology. Sci. Rep. 2019, 9, 1–12. [Google Scholar] [CrossRef]
- Antoni, D.; Burckel, H.; Josset, E.; Noel, G. Three-dimensional cell culture: A breakthrough in vivo. Int. J. Mol. Sci. 2015, 16, 5517–5527. [Google Scholar] [CrossRef]
- Fischbach, C.; Chen, R.; Matsumoto, T.; Schmelzle, T.; Brugge, J.S.; Polverini, P.J.; Mooney, D.J. Engineering tumors with 3D scaffolds. Nat. Methods 2007, 4, 855–860. [Google Scholar] [CrossRef]
- Tamaki, M.; McDonald, W.; Amberger, V.R.; Moore, E.; Del Maestro, R.F. Implantation of C6 astrocytoma spheroid into collagen type I gels: Invasive, proliferative, and enzymatic characterizations. J. Neurosurg. 1997, 87, 602–609. [Google Scholar] [CrossRef]
- Chaddad, H.; Kuchler-Bopp, S.; Fuhrmann, G.; Gegout, H.; Ubeaud-Sequier, G.; Schwinté, P.; Bornert, F.; Benkirane-Jessel, N.; Idoux-Gillet, Y. Combining 2D angiogenesis and 3D osteosarcoma microtissues to improve vascularization. Exp. Cell Res. 2017, 360, 138–145. [Google Scholar] [CrossRef] [PubMed]
- Cheung, K.J.; Gabrielson, E.; Werb, Z.; Ewald, A.J. Collective Invasion in Breast Cancer Requires a Conserved Basal Epithelial Program. Cell 2013, 155, 1639–1651. [Google Scholar] [CrossRef] [PubMed]
- Vellinga, T.T.; Den Uil, S.; Rinkes, I.; Marvin, D.; Ponsioen, B.; Alvarez-Varela, A.; Fatrai, S.; Scheele, C.; Zwijnenburg, D.A.; Snippert, H.; et al. Collagen-rich stroma in aggressive colon tumors induces mesenchymal gene expression and tumor cell invasion. Oncogene 2016, 35, 5263–5271. [Google Scholar] [CrossRef] [PubMed]
- O’Rourke, K.P.; Loizou, E.; Livshits, G.; Schatoff, E.M.; Baslan, T.; Manchado, E.; Simon, J.; Romesser, P.; Leach, B.; Han, T.; et al. Transplantation of engineered organoids enables rapid generation of metastatic mouse models of colorectal cancer HHS Public Access Author manuscript. Nat Biotechnol 2017, 35, 577–582. [Google Scholar] [CrossRef]
- Pathi, S.P.; Kowalczewski, C.; Tadipatri, R.; Fischbach, C. A novel 3-D mineralized tumor model to study breast cancer bone metastasis. PLoS ONE 2010, 5. [Google Scholar] [CrossRef]
- Talukdar, S.; Mandal, M.; Hutmacher, D.W.; Russell, P.J.; Soekmadji, C.; Kundu, S.C. Engineered silk fibroin protein 3D matrices for in vitro tumor model. Ipswich Rd 2011. [Google Scholar] [CrossRef]
- Cox, R.F.; Hernandez-Santana, A.; Ramdass, S.; McMahon, G.; Harmey, J.H.; Morgan, M.P. Microcalcifications in breast cancer: Novel insights into the molecular mechanism and functional consequence of mammary mineralisation. Br. J. Cancer 2012, 106, 525–537. [Google Scholar] [CrossRef]
- Marlow, R.; Honeth, G.; Lombardi, S.; Cariati, M.; Hessey, S.; Pipili, A.; Mariotti, V.; Buchupalli, B.; Foster, K.; Bonnet, D.; et al. A novel model of dormancy for bone metastatic breast cancer cells. Cancer Res. 2013, 73, 6886–6899. [Google Scholar] [CrossRef]
- Shahi Thakuri, P.; Liu, C.; Luker, G.D.; Pitcher Place, Z.; Arbor, A.; Hossein Tavana, U. Biomaterials-Based Approaches to Tumor Spheroid and Organoid Modeling The table of contents entry HHS Public Access. Adv. Healthc. Mater. 2018, 7, 1700980. [Google Scholar] [CrossRef]
- Sieh, S.; Lubik, A.A.; Clements, J.A.; Nelson, C.C.; Hutmacher, D.W. Interactions between human osteoblasts and prostate cancer cells in a novel 3D in vitro model. Organogenesis 2010, 6, 181–188. [Google Scholar] [CrossRef]
- Fong, E.L.S.; Wan, X.; Yang, J.; Morgado, M.; Mikos, A.G.; Harrington, D.A.; Navone, N.M.; Farach-Carson, M.C. A 3D in vitro model of patient-derived prostate cancer xenograft for controlled interrogation of in vivo tumor-stromal interactions. Biomaterials 2016, 77, 164–172. [Google Scholar] [CrossRef] [PubMed]
- Godugu, C.; Patel, A.R.; Desai, U.; Andey, T.; Sams, A.; Singh, M. AlgiMatrixTM Based 3D Cell Culture System as an In-Vitro Tumor Model for Anticancer Studies. PLoS ONE 2013, 8, e53708. [Google Scholar] [CrossRef] [PubMed]
- Dai, X.; Ma, C.; Lan, Q.; Xu, T. 3D Bioprinted Glioma Stem Cells for Brain Tumor Model and Applications of Drug Susceptibility—IOPscience. Available online: https://iopscience.iop.org/article/10.1088/1758-5090/8/4/045005 (accessed on 11 May 2020).
- Phan, N.; Hong, J.J.; Tofig, B.; Mapua, M.; Elashoff, D.; Moatamed, N.A.; Huang, J.; Memarzadeh, S.; Damoiseaux, R.; Soragni, A. A simple high-throughput approach identifies actionable drug sensitivities in patient-derived tumor organoids. Commun. Biol. 2019, 2, 1–11. [Google Scholar] [CrossRef] [PubMed]
- Mollica, P.A.; Booth-Creech, E.N.; Reid, J.A.; Zamponi, M.; Sullivan, S.M.; Palmer, X.L.; Sachs, P.C.; Bruno, R.D. 3D bioprinted mammary organoids and tumoroids in human mammary derived ECM hydrogels. Acta Biomater. 2019, 95, 201–213. [Google Scholar] [CrossRef] [PubMed]
- Sardi, M.; Lubitz, A.; Giese, C. Modeling Human Immunity In vitro: Improving Artificial Lymph Node Physiology by Stromal Cells. Appl. Vitr. Toxicol. 2016, 2, 143–150. [Google Scholar] [CrossRef]
- Sood, D.; Tang-Schomer, M.; Pouli, D.; Mizzoni, C.; Raia, N.; Tai, A.; Arkun, K.; Wu, J.; Black, L.D.; Scheffler, B.; et al. 3D extracellular matrix microenvironment in bioengineered tissue models of primary pediatric and adult brain tumors. Nat. Commun. 2019, 10, 1–14. [Google Scholar] [CrossRef]
- Grikscheit, K.; Frank, T.; Wang, Y.; Grosse, R. Junctional actin assembly is mediated by Formin-like 2 downstream of Rac1. J. Cell Biol. 2015, 209, 367–376. [Google Scholar] [CrossRef]
- Lal-Nag, M.; McGee, L.; Guha, R.; Lengyel, E.; Kenny, H.A.; Ferrer, M. A High-Throughput Screening Model of the Tumor Microenvironment for Ovarian Cancer Cell Growth. SLAS Discov. Adv. Sci. Drug Discov. 2017, 22, 494–506. [Google Scholar] [CrossRef]
- Yeung, P.; Sin, H.S.; Chan, S.; Chan, G.C.F.; Chan, B.P. Microencapsulation of Neuroblastoma Cells and Mesenchymal Stromal Cells in Collagen Microspheres: A 3D Model for Cancer Cell Niche Study. PLoS ONE 2015, 10, e0144139. [Google Scholar] [CrossRef]
- Li, C.; Zhao, S.; Zhao, Y.; Qian, Y.; Li, J.; Yin, Y. Chemically crosslinked alginate porous microcarriers modified with bioactive molecule for expansion of human hepatocellular carcinoma cells. J. Biomed. Mater. Res. Part B Appl. Biomater. 2014, 102, 1648–1658. [Google Scholar] [CrossRef]
- Krishnan, V.; Shuman, L.A.; Sosnoski, D.M.; Dhurjati, R.; Vogler, E.A.; Mastro, A.M. Dynamic interaction between breast cancer cells and osteoblastic tissue: Comparison of Two- and Three-dimensional cultures. J. Cell. Physiol. 2011, 226, 2150–2158. [Google Scholar] [CrossRef] [PubMed]
- Dhurjati, R.; Krishnan, V.; Shuman, L.A.; Mastro, A.M.; Vogler, E.A. Metastatic breast cancer cells colonize and degrade three-dimensional osteoblastic tissue in vitro. Clin. Exp. Metastasis 2008, 25, 741–752. [Google Scholar] [CrossRef] [PubMed]
- Sung, S.Y.; Hsieh, C.L.; Law, A.; Zhau, H.E.; Pathak, S.; Multani, A.S.; Lim, S.; Coleman, I.M.; Wu, L.C.; Figg, W.D.; et al. Coevolution of prostate cancer and bone stroma in three-dimensional coculture: Implications for cancer growth and metastasis. Cancer Res. 2008, 68, 9996–10003. [Google Scholar] [CrossRef] [PubMed]
- Brancato, V.; Garziano, A.; Gioiella, F.; Urciuolo, F.; Imparato, G.; Panzetta, V.; Fusco, S.; Netti, P.A. 3D is not enough: Building up a cell instructive microenvironment for tumoral stroma microtissues. Acta Biomater. 2017, 47, 1–13. [Google Scholar] [CrossRef] [PubMed]
- Brancato, V.; Gioiella, F.; Profeta, M.; Imparato, G.; Guarnieri, D.; Urciuolo, F.; Melone, P.; Netti, P.A. 3D tumor microtissues as an in vitro testing platform for microenvironmentally-triggered drug delivery systems. Acta Biomater. 2017, 57, 47–58. [Google Scholar] [CrossRef] [PubMed]
- Gioiella, F.; Urciuolo, F.; Imparato, G.; Brancato, V.; Netti, P.A. An Engineered Breast Cancer Model on a Chip to Replicate ECM-Activation In vitro during Tumor Progression. Adv. Healthc. Mater. 2016, 5, 3074–3084. [Google Scholar] [CrossRef] [PubMed]
- Pradhan, S.; Clary, J.M.; Seliktar, D.; Lipke, E.A. A three-dimensional spheroidal cancer model based on PEG-fibrinogen hydrogel microspheres. Biomaterials 2017, 115, 141–154. [Google Scholar] [CrossRef]
- Zhuo, S.; Chen, J.; Xie, S.; Hong, Z.; Jiang, X. Extracting diagnostic stromal organization features based on intrinsic two-photon excited fluorescence and second-harmonic generation signals. J. Biomed. Opt. 2009, 14, 020503. [Google Scholar] [CrossRef] [PubMed][Green Version]
- Zervantonakis, I.K.; Hughes-Alford, S.K.; Charest, J.L.; Condeelis, J.S.; Gertler, F.B.; Kamm, R.D. Three-dimensional microfluidic model for tumor cell intravasation and endothelial barrier function. Proc. Natl. Acad. Sci. USA 2012, 109, 13515–13520. [Google Scholar] [CrossRef] [PubMed]
- Bersini, S.; Jeon, J.S.; Dubini, G.; Arrigoni, C.; Chung, S.; Charest, J.L.; Moretti, M.; Kamm, R.D. A microfluidic 3D invitro model for specificity of breast cancer metastasis to bone. Biomaterials 2014, 35, 2454–2461. [Google Scholar] [CrossRef] [PubMed]
- Jeon, J.S.; Bersini, S.; Gilardi, M.; Dubini, G.; Charest, J.L.; Moretti, M.; Kamm, R.D. Human 3D vascularized organotypic microfluidic assays to study breast cancer cell extravasation. Proc. Natl. Acad. Sci. USA 2015, 112, 214–219. [Google Scholar] [CrossRef] [PubMed]
- Hsiao, A.Y.; Torisawa, Y.S.; Tung, Y.C.; Sud, S.; Taichman, R.S.; Pienta, K.J.; Takayama, S. Microfluidic system for formation of PC-3 prostate cancer co-culture spheroids. Biomaterials 2009, 30, 3020–3027. [Google Scholar] [CrossRef] [PubMed]
- Sitarski, A.M.; Fairfield, H.; Falank, C.; Reagan, M.R. 3D Tissue Engineered in vitro Models of Cancer in Bone. ACS Biomater. Sci. Eng. 2018, 4, 324–336. [Google Scholar] [CrossRef] [PubMed]
- Kumar, V.; Varghese, S. Ex vivo Tumor-on-a-Chip Platforms to Study Intercellular Interactions within the Tumor Microenvironment. Adv. Healthc. Mater. 2019, 8, 1–12. [Google Scholar] [CrossRef]
- Whitesides, G.M. The origins and the future of microfluidics. Nature 2006, 442, 368–373. [Google Scholar] [CrossRef]
- Huh, D.; Matthews, B.D.; Mammoto, A.; Montoya-Zavala, M.; Yuan Hsin, H.; Ingber, D.E. Reconstituting organ-level lung functions on a chip. Science 2010, 328, 1662–1668. [Google Scholar] [CrossRef]
- Xu, Z.; Li, E.; Guo, Z.; Yu, R.; Hao, H.; Xu, Y.; Sun, Z.; Li, X.; Lyu, J.; Wang, Q. Design and Construction of a Multi-Organ Microfluidic Chip Mimicking the in vivo Microenvironment of Lung Cancer Metastasis. ACS Appl. Mater. Interfaces 2016, 8, 25840–25847. [Google Scholar] [CrossRef]
- Lanz, H.L.; Saleh, A.; Kramer, B.; Cairns, J.; Ng, C.P.; Yu, J.; Trietsch, S.J.; Hankemeier, T.; Joore, J.; Vulto, P.; et al. Therapy response testing of breast cancer in a 3D high-throughput perfused microfluidic platform. BMC Cancer 2017, 17, 709. [Google Scholar] [CrossRef]
- Edington, C.D.; Chen, W.L.K.; Geishecker, E.; Kassis, T.; Soenksen, L.R.; Bhushan, B.M.; Freake, D.; Kirschner, J.; Maass, C.; Tsamandouras, N.; et al. Interconnected Microphysiological Systems for Quantitative Biology and Pharmacology Studies. Sci. Rep. 2018, 8, 1–18. [Google Scholar] [CrossRef]
- Vanderburgh, J.; Sterling, J.A.; Guelcher, S.A. 3D Printing of Tissue Engineered Constructs for In vitro Modeling of Disease Progression and Drug Screening. Ann. Biomed. Eng. 2017, 45, 164–179. [Google Scholar] [CrossRef]
- Mou, H.; Wang, J.; Hu, H.; Xu, W.; Chen, Q. Non-small cell lung cancer 95D cells co-cultured with 3D-bioprinted scaffold to construct a lung cancer model in vitro. Zhonghua Zhong Liu Za Zhi 2015, 37, 736–740. [Google Scholar] [PubMed]
- Ling, K.; Huang, G.; Liu, J.; Zhang, X.; Ma, Y.; Lu, T.; Xu, F. Bioprinting-Based High-Throughput Fabrication of Three-Dimensional MCF-7 Human Breast Cancer Cellular Spheroids. Engineering 2015, 1, 269–274. [Google Scholar] [CrossRef]
- Huang, T.Q.; Qu, X.; Liu, J.; Chen, S. 3D printing of biomimetic microstructures for cancer cell migration. Biomed. Microdevices 2014, 16, 127–132. [Google Scholar] [CrossRef]
- Zhu, C.; He, L.; Zhou, X.; Nie, X.; Gu, Y. Sulfatase 2 promotes breast cancer progression through regulating some tumor-related factors. Oncol. Rep. 2016, 35, 1318–1328. [Google Scholar] [CrossRef]
- Liu, T.; Delavaux, C.; Zhang, Y.S. 3D bioprinting for oncology applications. J. 3D Print. Med. 2019, 3, 55–58. [Google Scholar] [CrossRef] [PubMed]
- Meng, F.; Meyer, C.M.; Joung, D.; Vallera, D.A.; McAlpine, M.C.; Panoskaltsis-Mortari, A. 3D Bioprinted In vitro Metastatic Models via Reconstruction of Tumor Microenvironments. Adv. Mater. 2019, 31, 1–10. [Google Scholar] [CrossRef]
- Stanton, M.M.; Samitier, J.; Sánchez, S. Bioprinting of 3D hydrogels. Lab Chip 2015, 15, 3111–3115. [Google Scholar] [CrossRef]
- Sutherland, R.M.; Inch, W.R.; McCredie, J.A.; Kruuv, J. A multi-component radiation survival curve using an in vitro tumour model. Int. J. Radiat. Biol. 1970, 18, 491–495. [Google Scholar]
- Grimes, D.R.; Fletcher, A.G.; Partridge, M. Oxygen consumption dynamics in steady-state tumour models Author for correspondence. J. R. Soc. Interface 2014, 1. [Google Scholar] [CrossRef]
- Kelm, J.M.; Timmins, N.E.; Brown, C.J.; Fussenegger, M.; Nielsen, L.K. Method for generation of homogeneous multicellular tumor spheroids applicable to a wide variety of cell types. Biotechnol. Bioeng. 2003, 83, 173–180. [Google Scholar] [CrossRef]
- Page, H.; Flood, P.; Reynaud, E.G. Three-dimensional tissue cultures: Current trends and beyond. Cell Tissue Res. 2013, 352, 123–131. [Google Scholar] [CrossRef] [PubMed]
- Cortini, M.; Baldini, N.; Avnet, S. New advances in the study of bone tumors: A lesson from the 3D environment. Front. Physiol. 2019, 10, 814. [Google Scholar] [CrossRef] [PubMed]
- Tevis, K.M.; Colson, Y.L.; Grinstaff, M.W.; Colson, Y.; Author, A.B. Embedded Spheroids as Models of the Cancer Microenvironment Corresponding Authors HHS Public Access Author manuscript. Adv Biosyst 2017, 1. [Google Scholar] [CrossRef] [PubMed]
- 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. The Netherlands 3 Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus. Cell 2015, 161, 933–945. [Google Scholar] [CrossRef]
- 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]
- Fang, Y.; Eglen, R.M. Three-Dimensional Cell Cultures in Drug Discovery and Development. SLAS Discov. Adv. Life Sci. R&D 2017, 247255521769679. [Google Scholar] [CrossRef]
- Lancaster, M.A.; Knoblich, J.A. Organogenesis in a dish: Modeling development and disease using organoid technologies. Science 2014, 345, 1247125. [Google Scholar] [CrossRef]
- Kwon, H.; Sun, L.; Cairns, D.M.; Rainbow, R.S.; Preda, R.C.; Kaplan, D.L.; Zeng, L. The influence of scaffold material on chondrocytes under inflammatory conditions. Acta Biomater. 2013, 9, 6563–6575. [Google Scholar] [CrossRef]
- Hiraga, T.; Myoui, A.; Hashimoto, N.; Sasaki, A.; Hata, K.; Morita, Y.; Yoshikawa, H.; Rosen, C.J.; Mundy, G.R.; Yoneda, T. Bone-Derived IGF Mediates Crosstalk between Bone and Breast Cancer Cells in Bony Metastases. Cancer Res. 2012, 72, 4238–4249. [Google Scholar] [CrossRef]
- Malda, J.; Klein, T.J.; Upton, Z. The roles of hypoxia in the in vitro engineering of tissues. Tissue Eng. 2007, 13, 2153–2162. [Google Scholar] [CrossRef]
- Keith, B.; Johnson, R.S.; Simon, M.C. HIF1 α and HIF2 α: Sibling rivalry in hypoxic tumour growth and progression. Nat. Rev. Cancer 2012, 12, 9–22. [Google Scholar] [CrossRef]
- Johnson, R.W.; Sowder, M.E.; Giaccia, A.J. Hypoxia and Bone Metastatic Disease. Curr. Osteoporos. Rep. 2017, 15, 231–238. [Google Scholar] [CrossRef] [PubMed]
- Spencer, J.A.; Ferraro, F.; Roussakis, E.; Klein, A.; Wu, J.; Runnels, J.M.; Zaher, W.; Mortensen, L.J.; Alt, C.; Turcotte, R.; et al. Direct measurement of local oxygen concentration in the bone marrow of live animals. Nature 2014, 508, 269–273. [Google Scholar] [CrossRef] [PubMed]
- Mohyeldin, A.; Garzón-Muvdi, T.; Quiñones-Hinojosa, A. Oxygen in stem cell biology: A critical component of the stem cell niche. Cell Stem Cell 2010, 7, 150–161. [Google Scholar] [CrossRef] [PubMed]
- Minardi, S.; Sandri, M.; Martinez, J.O.; Yazdi, I.K.; Liu, X.; Ferrari, M.; Weiner, B.K.; Tampieri, A.; Tasciotti, E. Multiscale Patterning of a Biomimetic Scaffold Integrated with Composite Microspheres. Small 2014, 10, 3943–3953. [Google Scholar] [CrossRef]
- Liverani, C.; De Vita, A.; Minardi, S.; Kang, Y.; Mercatali, L.; Amadori, D.; Bongiovanni, A.; La Manna, F.; Ibrahim, T.; Tasciotti, E. A biomimetic 3D model of hypoxia-driven cancer progression. Sci. Rep. 2019, 9, 1–13. [Google Scholar] [CrossRef]
- Prat, A.; Perou, C.M. Deconstructing the molecular portraits of breast cancer. Mol. Oncol. 2011, 5, 5–23. [Google Scholar] [CrossRef]
- Sourla, A.; Doillon, C.; Koutsilieris, M. Three-dimensional type I collagen gel system containing MG-63 osteoblasts-like cells as a model for studying local bone reaction caused by metastatic cancer cells. Anticancer Res. 1996, 16, 2773–2780. [Google Scholar]
- Jin, M.Z.; Han, R.R.; Qiu, G.Z.; Ju, X.C.; Lou, G.; Jin, W.L. Organoids: An intermediate modeling platform in precision oncology. Cancer Lett. 2018, 414, 174–180. [Google Scholar] [CrossRef]
- Liu, M.; Zeng, X.; Ma, C.; Yi, H.; Ali, Z.; Mou, X.; Li, S.; Deng, Y.; He, N. Injectable hydrogels for cartilage and bone tissue engineering. Bone Res. 2017, 5. [Google Scholar] [CrossRef]
- Xu, X.; Farach-Carson, M.C.; Jia, X. Three-dimensional in vitro tumor models for cancer research and drug evaluation. Biotechnol. Adv. 2014, 32, 1256–1268. [Google Scholar] [CrossRef] [PubMed]
- Fong, E.L.S.; Martinez, M.; Yang, J.; Mikos, A.G.; Navone, N.M.; Harrington, D.A.; Farach-Carson, M.C. Hydrogel-based 3D model of patient-derived prostate xenograft tumors suitable for drug screening. Mol. Pharm. 2014, 11, 2040–2050. [Google Scholar] [CrossRef] [PubMed]
- Huang, Y.J.; Hsu, S. hui Acquisition of epithelial-mesenchymal transition and cancer stem-like phenotypes within chitosan-hyaluronan membrane-derived 3D tumor spheroids. Biomaterials 2014, 35, 10070–10079. [Google Scholar] [CrossRef] [PubMed]
- Sung, K.E.; Beebe, D.J. Microfluidic 3D models of cancer. Adv. Drug Deliv. Rev. 2014, 79, 68–78. [Google Scholar] [CrossRef] [PubMed]
- Guller, A.E.; Grebenyuk, P.N.; Shekhter, A.B.; Zvyagin, A.V.; Deyev, S.M. Bioreactor-Based Tumor Tissue Engineering. Acta Naturae 2016, 8, 44. [Google Scholar] [CrossRef] [PubMed]
- Hickman, J.A.; Graeser, R.; de Hoogt, R.; Vidic, S.; Brito, C.; Gutekunst, M.; van der Kuip, H. Imi Predect consortium Three-dimensional models of cancer for pharmacology and cancer cell biology: Capturing tumor complexity in vitro/ex vivo. Biotechnol. J. 2014, 9, 1115–1128. [Google Scholar] [CrossRef]
- Paolillo, M.; Colombo, R.; Serra, M.; Belvisi, L.; Papetti, A.; Ciusani, E.; Comincini, S.; Schinelli, S. Stem-like Cancer Cells in a Dynamic 3D Culture System: A Model to Study Metastatic Cell Adhesion and Anti-cancer Drugs. Cells 2019, 8, 1434. [Google Scholar] [CrossRef]
- Becker, J.L.; Souza, G.R. Using space-based investigations to inform cancer research on Earth. Nat. Rev. Cancer 2013, 13, 315–327. [Google Scholar] [CrossRef]
- 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]
- Chung, M.; Ahn, J.; Son, K.; Kim, S.; Jeon, N.L. Biomimetic Model of Tumor Microenvironment on Microfluidic Platform. Adv. Healthc. Mater. 2017, 6. [Google Scholar] [CrossRef]
- Mitchell, M.J.; King, M.R. Computational and experimental models of cancer cell response to fluid shear stress. Front. Oncol. 2013, 3, 44. [Google Scholar] [CrossRef] [PubMed]
- Phillips, K.G.; Kuhn, P.; McCarty, O.J.T. Physical biology in cancer. 2. The physical biology of circulating tumor cells. Am. J. Physiol. Cell Physiol. 2014, 306. [Google Scholar] [CrossRef] [PubMed]
- Boussommier-Calleja, A.; Li, R.; Chen, M.B.; Wong, S.C.; Kamm, R.D. Microfluidics: A New Tool for Modeling Cancer–Immune Interactions. Trends Cancer 2016, 2, 6–19. [Google Scholar] [CrossRef] [PubMed]
- Buchanan, C.F.; Voigt, E.E.; Szot, C.S.; Freeman, J.W.; Vlachos, P.P.; Rylander, M.N. Three-dimensional microfluidic collagen hydrogels for investigating flow-mediated tumor-endothelial signaling and vascular organization. Tissue Eng. Part C Methods 2014, 20, 64–75. [Google Scholar] [CrossRef]
- Park, S.E.; Georgescu, A.; Huh, D. Organoids on a chip. Science 2019, 965, 837–842. [Google Scholar] [CrossRef]
- Apoorva, F.; Loiben, A.M.; Shah, S.B.; Purwada, A.; Fontan, L.; Goldstein, R.; Kirby, B.J.; Melnick, A.M.; Cosgrove, B.D.; Singh, A. How Biophysical Forces Regulate Human B Cell Lymphomas. Cell Rep. 2018, 23, 499–511. [Google Scholar] [CrossRef]
- Ando, Y.; Ta, H.P.; Yen, D.P.; Lee, S.S.; Raola, S.; Shen, K. A microdevice platform recapitulating hypoxic tumor microenvironments. Sci. Rep. 2017, 7. [Google Scholar] [CrossRef]
- Takebe, T.; Zhang, B.; Radisic, M. Synergistic Engineering: Organoids Meet Organs-on-a-Chip. Cell Stem Cell 2017, 21, 297–300. [Google Scholar] [CrossRef]
- Fan, H.; Demirci, U.; Chen, P. Emerging organoid models: Leaping forward in cancer research. J. Hematol. Oncol. 2019, 12, 1–10. [Google Scholar] [CrossRef]
- Charbe, N.; Mccarron, P.A.; Tambuwala, M.M. Three-dimensional bio-printing: A new frontier in oncology research. World J. Clin. Oncol. 2017, 8, 21–36. [Google Scholar] [CrossRef]
- Zhang, Y.S.; Arneri, A.; Bersini, S.; Shin, S.-R.; Zhu, K.; Goli-Malekabadi, Z.; Aleman, J.; Colosi, C.; Busignani, F.; Dell’Erba, V.; et al. Bioprinting 3D microfibrous scaffolds for engineering endothelialized myocardium and heart-on-a-chip. Biomaterials 2016, 110, 45–59. [Google Scholar] [CrossRef] [PubMed]
- Soman, P.; Kelber, J.A.; Lee, J.W.; Wright, T.N.; Vecchio, K.S.; Klemke, R.L.; Chen, S. Cancer cell migration within 3D layer-by-layer microfabricated photocrosslinked PEG scaffolds with tunable stiffness. Biomaterials 2012, 33, 7064–7070. [Google Scholar] [CrossRef] [PubMed]
- Zhu, W.; Castro, N.J.; Cui, H.; Zhou, X.; Boualam, B.; McGrane, R.; Glazer, R.I.; Zhang, L.G. A 3D Printed Nano Bone Matrix for Characterization of Breast Cancer Cell and Osteoblast Interactions. Nanotechnology 2016, 27, 315103. [Google Scholar] [CrossRef] [PubMed]
- Katt, M.E.; Placone, A.L.; Wong, A.D.; Xu, Z.S.; Searson, P.C. In vitro tumor models: Advantages, disadvantages, variables, and selecting the right platform. Front. Bioeng. Biotechnol. 2016, 4, 12. [Google Scholar] [CrossRef]
- Bellido, T.; Delgado-Calle, J. Ex vivo Organ Cultures as Models to Study Bone Biology. JBMR Plus 2020, 4, 1–10. [Google Scholar] [CrossRef]
- Staines, K.A.; Brown, G.; Farquharson, C. The Ex Vivo Organ Culture of Bone. In Methods in Molecular Biology; Humana Press Inc.: Totowa, NJ, USA, 2019; Volume 1914, pp. 199–215. [Google Scholar]
- Lovitt, C.J.; Shelper, T.B.; Avery, V.M. Cancer drug discovery: Recent innovative approaches to tumor modeling. Expert Opin. Drug Discov. 2016, 11, 885–894. [Google Scholar] [CrossRef]
- van der Kuip, H.; Mürdter, T.E.; Sonnenberg, M.; McClellan, M.; Gutzeit, S.; Gerteis, A.; Simon, W.; Fritz, P.; Aulitzky, W.E. Short term culture of breast cancer tissues to study the activity of the anticancer drug taxol in an intact tumor environment. BMC Cancer 2006, 6, 86. [Google Scholar] [CrossRef]
- Place, T.L.; Domann, F.E.; Case, A.J. Limitations of oxygen delivery to cells in culture: An underappreciated problem in basic and translational research. Free Radic. Biol. Med. 2017, 113, 311–322. [Google Scholar] [CrossRef]
- Nath, S.; Devi, G.R. Three-dimensional culture systems in cancer research: Focus on tumor spheroid model. Pharmacol. Ther. 2016, 163, 94–108. [Google Scholar] [CrossRef]
- Davies, E.J.; Dong, M.; Gutekunst, M.; Närhi, K.; Wedge, S.R.; Hällström, T.M.; Schueler, J.; Weerden, W.M. Van Capturing complex tumour biology in vitro: Histological and molecular characterisation of precision cut slices. Nat. Publ. Gr. 2015, 1–17. [Google Scholar] [CrossRef]
- Davies, E.; Jones, A.; Cumberbatch, M.; Eberlein, C.; Pritchard, A.; Gray, N.; Wedge, S.; Smalley, M.; Barry, S.; Park, A. Tissue Slice Culture: An ex-vivo tumour culture platform for pharmacodynamic analysis and dose response readouts. 2013, p. 115188. Available online: http://www.predect.eu/midcom-serveattachmentguid-1e2e57d7518d94ce57d11e285b7859e50d023802380/201211_bacr-davies.pdf (accessed on 10 May 2020).
- Pinto, C.; Estrada, M.F.; Brito, C. In Vitro and Ex Vivo Models—The Tumor Microenvironment in a Flask. In Advances in Experimental Medicine and Biology; Springer: Berlin/Heidelberg, Germany, 2020; Volume 1219, pp. 431–443. [Google Scholar]
- Tanos, T.; Sflomos, G.; Echeverria, P.C.; Ayyanan, A.; Gutierrez, M.; Delaloye, J.F.; Raffoul, W.; Fiche, M.; Dougall, W.; Schneider, P.; et al. Progesterone/RANKL is a major regulatory axis in the human breast. Sci. Transl. Med. 2013, 5, 182ra55. [Google Scholar] [CrossRef] [PubMed]
- Muraro, M.G.; Muenst, S.; Mele, V.; Quagliata, L.; Iezzi, G.; Tzankov, A.; Weber, W.P.; Spagnoli, G.C.; Soysal, S.D. Ex-vivo assessment of drug response on breast cancer primary tissue with preserved microenvironments. Oncoimmunology 2017, e1331798. [Google Scholar] [CrossRef] [PubMed]
- Majumder, B.; Baraneedharan, U.; Thiyagarajan, S.; Radhakrishnan, P.; Narasimhan, H.; Dhandapani, M.; Brijwani, N.; Pinto, D.D.; Prasath, A.; Shanthappa, B.U.; et al. Predicting clinical response to anticancer drugs using an ex vivo platform that captures tumour heterogeneity. Nat. Commun. 2015, 6, 1–14. [Google Scholar] [CrossRef] [PubMed]
- Garrett, I.R.; Mundy, G.R. Relationship between interleukin-1 and prostaglandins in resorbing neonatal calvaria. J. Bone Miner. Res. 1989, 4, 789–794. [Google Scholar] [CrossRef]
- Abubakar, A.A.; Noordin, M.M.; Azmi, T.I.; Kaka, U.; Loqman, M.Y. The use of rats and mice as animal models in ex vivo bone growth and development studies. Bone Joint Res. 2016, 5, 610–618. [Google Scholar] [CrossRef]
- Fell, H.B.; Robison, R. The growth, development and phosphatase activity of embryonic avian femora and limb-buds cultivated in vitro. Biochem. J. 1929, 23, 767–784.5. [Google Scholar] [CrossRef]
- Pfeilschifter, J.; Mundy, G.R. Modulation of type beta transforming growth factor activity in bone cultures by osteotropic hormones. Proc. Natl. Acad. Sci. USA 1987, 84, 2024–2028. [Google Scholar] [CrossRef]
- Rao, S.R.; Edwards, C.M.; Edwards, J.R. Modeling the Human Bone–Tumor Niche: Reducing and Replacing the Need for Animal Data. JBMR Plus 2020, 4. [Google Scholar] [CrossRef]
- Weir, E.C.; Lowik, C.W.G.M.; Paliwal, I.; Insogna, K.L. Colony stimulating factor-1 plays a role in osteoclast formation and function in bone resorption induced by parathyroid hormone and parathyroid hormone-related protein. J. Bone Miner. Res. 2009, 11, 1474–1481. [Google Scholar] [CrossRef]
- Mundy, G.; Garrett, R.; Harris, S.; Chan, J.; Chen, D.; Rossini, G.; Boyce, B.; Zhao, M.; Gutierrez, G. Stimulation of bone formation in vitro and in rodents by statins. Science 1999, 286, 1946–1949. [Google Scholar] [CrossRef] [PubMed]
- Song, W.; Fhu, C.W.; Ang, K.H.; Liu, C.H.; Johari, N.A.B.; Lio, D.; Abraham, S.; Hong, W.; Moss, S.E.; Greenwood, J.; et al. The fetal mouse metatarsal bone explant as a model of angiogenesis. Nat. Protoc. 2015, 10, 1459–1473. [Google Scholar] [CrossRef] [PubMed]
- Nordstrand, A.; Nilsson, J.; Tieva, Å.; Wikström, P.; Lerner, U.H.; Widmark, A. Establishment and validation of an in vitro co-culture model to study the interactions between bone and prostate cancer cells. Clin. Exp. Metastasis 2009, 26, 945–953. [Google Scholar] [CrossRef] [PubMed]
- Curtin, P.; Youm, H.; Salih, E. Three-dimensional cancer-bone metastasis model using ex-vivo co-cultures of live calvarial bones and cancer cells. Biomaterials 2012, 33, 1065–1078. [Google Scholar] [CrossRef] [PubMed]
- Tulotta, C.; Lefley, D.V.; Freeman, K.; Gregory, W.M.; Hanby, A.M.; Heath, P.R.; Nutter, F.; Mark Wilkinson, J.; Spicer-Hadlington, A.R.; Liu, X.; et al. Endogenous production of IL1B by breast cancer cells drives metastasis and colonization of the bone microenvironment. Clin. Cancer Res. 2019, 25, 2769–2782. [Google Scholar] [CrossRef]
- Lefley, D.; Howard, F.; Arshad, F.; Bradbury, S.; Brown, H.; Tulotta, C.; Eyre, R.; Alférez, D.; Wilkinson, J.M.; Holen, I.; et al. Development of clinically relevant in vivo metastasis models using human bone discs and breast cancer patient-derived xenografts. Breast Cancer Res. 2019, 21, 1–21. [Google Scholar] [CrossRef]
- Suva, L.J.; Washam, C.; Nicholas, R.W.; Griffin, R.J. Bone metastasis: Mechanisms and therapeutic opportunities. Nat. Rev. Endocrinol. 2011, 7, 208–218. [Google Scholar] [CrossRef]
- Berish, R.B.; Ali, A.N.; Telmer, P.G.; Ronald, J.A.; Leong, H.S. Translational models of prostate cancer bone metastasis. Nat. Rev. Urol. 2018, 15, 403–421. [Google Scholar] [CrossRef]
- Jinnah, A.H.; Zacks, B.C.; Gwam, C.U.; Kerr, B.A. Emerging and established models of bone metastasis. Cancers 2018, 10, 176. [Google Scholar] [CrossRef]
- Krishnan, V.; Vogler, E.A.; Mastro, A.M. Three-Dimensional in vitro Model to Study Osteobiology and Osteopathology. J. Cell. Biochem. 2015, 116, 2715–2723. [Google Scholar] [CrossRef]
- Holen, I.; Nutter, F.; Wilkinson, J.M.; Evans, C.A.; Avgoustou, P.; Ottewell, P.D. Human breast cancer bone metastasis in vitro and in vivo: A novel 3D model system for studies of tumour cell-bone cell interactions. Clin. Exp. Metastasis 2015, 32, 689–702. [Google Scholar] [CrossRef] [PubMed]
3D Models | Cancer Feature Studied | Pros | Cons | Other References |
---|---|---|---|---|
Scaffold free | Cancer cell invasion into the ECM [41]; Formation of tubule-like structure that mimics vessel sprouting and angiogenesis [42]; Collective invasion and intravasation [43]; Collective invasion [44]. | High Reproducibility; Low cost; Co-culture; HTS approach. | No support or porosity; Not optically transparent; Poor control over spheroids/organoids size; No cell/ECM interactions. | [45,46,47,48,49,50,51,52,53] |
Scaffold based | Cell adhesion, proliferation, activation, and differentiation to hold up metastatic cancer cell growth and to resemble TME complexity [45]; Role of HA and IL8 in switching mammary tumor cells toward a more invasive phenotype [46]; Cancer cells and BM interplay is influenced by spatial organization, osteogenic factors, and stromal cell type [47]; Osteomimicry, the BM [48]; BM model of the BC metastatic niche [49]; PC cells in contact with osteoblast-like cells embedded in 3D collagen gel system produced morphological evidence of blastic reaction and of local invasion [50]; Evaluation of metastasis development from endometrial cancer, PC, and BC co-culture and expression of biomarkers associated with PC cells BM [51]. | Co-cultures; Large variety of materials; Customizable; Affordable cost; High similarity to the in vivo conditions; Promotion of cellular attachment, proliferation, and differentiation; HTS approach sustainable. | Possible scaffold-to-scaffold variation; Not always optically transparent; Difficult cells removal; HTS options limited; Gelling mechanisms; Batch to batch variations; Undefined constituents in natural gels; Poor mechanical properties. | [52,53,54,55,56,57,58,59,60,61,62] |
Cultivation and Biofabrication Systems | Cancer Feature Studied | Pros | Cons | Other References |
Bioreactors | Reconstruction of a bone surrogate to study the early stages of BC invasion to bone [63]; Co-culture of OT with metastatic BC cells [64]; Reproduction of the interaction between bone stroma, PC cells, and human osteosarcoma cell line [65]. | High similarity to the in vivo conditions; High volume of cells production; Customizable and controlled culture parameters. | Space required for dynamic cell culture; High costs for dynamic cultures; HTS options laborious. | [62,66,67,68,69,70] |
Microfluidic | Angiogenesis, intravasation [71]; Study of molecular pathways implicated in BC cells extravasation, mediated by cell surface receptor CXCR2 and bone-secreted chemokine CXCL5 [72]; Microvascularized bone-mimicking microenvironment, defined by active differentiated bone cells, which generated spontaneously molecular gradients affecting both microvasculature and cancer cells [73]; 3D multicellular spheroid composed by PC-3 metastatic PC cells, osteoblasts, and endothelial cells [74]. | Co-cultures (cell–cell, cell–tissue); Control of cell shape and function; Tune dynamic; Fluid flow and spatiotemporal gradient; Customizable; Commercial availability. | Required expertise; High cost of microfabrication; HTS options limited; Microenvironment parameters not measurable; Cell growing media for co-culture not well established. | [75,76,77,78] |
Organ-on-a-chip | Tumor multiorgan metastasis and cancer microenvironment interaction [73]; Development of a four organ-on-a-chip system [79]. | In vitro organ specific systems; High gas permeability; Optically transparent; Commercial availability. | Required expertise; High cost for the microfabrication; HTS options limited | [80,81] |
3D bioprinting | 3D bioprinting fibers embedded in hydrogels to recreate microvessels and study cancer-related angiogenesis [82]; Proliferation and invasion ability [83]; Modeling tumor microenvironment [84]; Migration of osteotropic cells during bone metastasis [85]; In vitro bone matrices to mimic the native BM for the investigation of BC bone metastasis [86]. | Automated robotic processes; Spatially assembling multiple types of cells; Large variety of biomaterials and printing technologies; Bimolecular gradient production; Printable, crosslinkable, biocompatible and bioactive bioinks. | High cost; Required expertise. | [56,75,87,88,89] |
© 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
Laranga, R.; Duchi, S.; Ibrahim, T.; Guerrieri, A.N.; Donati, D.M.; Lucarelli, E. Trends in Bone Metastasis Modeling. Cancers 2020, 12, 2315. https://doi.org/10.3390/cancers12082315
Laranga R, Duchi S, Ibrahim T, Guerrieri AN, Donati DM, Lucarelli E. Trends in Bone Metastasis Modeling. Cancers. 2020; 12(8):2315. https://doi.org/10.3390/cancers12082315
Chicago/Turabian StyleLaranga, Roberta, Serena Duchi, Toni Ibrahim, Ania Naila Guerrieri, Davide Maria Donati, and Enrico Lucarelli. 2020. "Trends in Bone Metastasis Modeling" Cancers 12, no. 8: 2315. https://doi.org/10.3390/cancers12082315
APA StyleLaranga, R., Duchi, S., Ibrahim, T., Guerrieri, A. N., Donati, D. M., & Lucarelli, E. (2020). Trends in Bone Metastasis Modeling. Cancers, 12(8), 2315. https://doi.org/10.3390/cancers12082315