3D Cancer Models: Depicting Cellular Crosstalk within the Tumour Microenvironment
Abstract
:Simple Summary
Abstract
1. Introduction
2. Three-Dimensional Cancer Models
2.1. Spheroids
2.2. Tumour Organoids and Other Scaffold-Based Models
2.3. Microfluidic-Based 3D Models
3. Three-Dimensional Double Co-Cultures Incorporating Non-Malignant Cell Components of the Tumour Microenvironment
3.1. Fibroblasts
3.1.1. Models with Normal Fibroblasts
3.1.2. Models with CAF and Non-Malignant Epithelial Cells
3.1.3. Models with CAF and Tumour Cells
3.1.4. Models with Different CAF Subsets
3.2. Endothelial Cells
3.2.1. Heterotypic Spheroid Approaches
3.2.2. Matrix-Embedding Approaches
3.2.3. Microfluidic Approaches
3.3. Immune Cells
3.3.1. T Lymphocytes
3.3.2. Macrophages
3.3.3. Neutrophils
3.3.4. Dendritic Cells
3.3.5. NK Cells
3.3.6. Other Immune Cells
3.4. Mesenchymal Stromal Cells
3.5. Other Non-Malignant Cell Types of the TME
4. Combination of Multiple Non-Malignant Cell Types
4.1. 3D Tricultures
4.1.1. 3D Tricultures of Tumour and Stromal Cells
4.1.2. Triple Cultures with Immune Cells
4.2. Tetracultures
5. Concluding Remarks and Future Perspectives
5.1. Choice of Scaffold
5.2. Choice of Cell Source
5.3. Physicochemical Parameters
5.4. Current Challenges and Applications
5.5. Towards Increased Comparability and Reproducibility of 3D TME Models
Author Contributions
Funding
Conflicts of Interest
Abbreviations
5-FU | 5-Fluorouracil |
ADCC | Antibody-dependent cell cytotoxicity |
BM | Bone marrow |
BME | Basement membrane extract |
CAF | Cancer-associated fibroblasts |
CAR | Chimeric antigen receptor |
CEA | Carcinoembryonic antigen |
CCL2 | C-C motif chemokine ligand 2 |
CSF1R | Colony stimulating factor 1 receptor |
CTL | Cytotoxic T lymphocytes |
CXCL12 | C-X-C motif chemokine ligand 12 |
CXCR4 | C-X-C chemokine receptor type 4 |
DC | Dendritic cells |
EC | Endothelial cells |
ECM | Extracellular matrix |
EGFR | Epidermal growth factor receptor |
EMT | Epithelial to mesenchymal transition |
FAP | Fibroblast-activation protein |
FGF-2 | Fibroblast growth factor-2 |
G-CSF | Granulocyte colony stimulating factor |
HER2 | Human epidermal growth factor receptor 2 |
HGF | Hepatocyte growth factor |
HUVEC | Human umbilical vein endothelial cells |
ICAM-1 | Intercellular adhesion molecule 1 |
IFN-γ | Interferon- γ |
IL-6 | Interleukin-6 |
LOX | Lysyl oxidase |
LPS | Lipopolysaccharide |
MDSC | Myeloid-derived suppressor cells |
MHC | Major histocompatibility complex |
MMP | Matrix metalloproteinases |
MSC | Mesenchymal stromal cells |
M-CSF | Macrophage-colony stimulating factor |
NK | Natural Killer cells |
PBMC | Peripheral blood mononuclear cells |
PEG | Poly(ethylene) glycol |
PD-1 | Programmed cell death protein 1 |
PD-L1 | Programmed death ligand 1 |
PDGF | Platelet-derived growth factor |
PMA | Phorbol-12-myristate-13-acetate |
ROS | Reactive oxygen species |
SMC | Smooth muscle cells |
TAM | Tumour-associated macrophages |
TGF-β | Transforming growth factor β |
TIMP-1 | Tissue inhibitor of metalloproteinases 1 |
TME | Tumour microenvironment |
TNF-α | Tumour necrosis factor α |
Treg | Regulatory T cells |
VEGF | Vascular endothelial growth factor |
α-SMA | α Smooth muscle actin |
References
- Liu, Z.; Delavan, B.; Roberts, R.; Tong, W. Lessons Learned from Two Decades of Anticancer Drugs. Trends Pharmacol. Sci. 2017, 38, 852–872. [Google Scholar] [CrossRef] [Green Version]
- Moreno, L.; Pearson, A.D.J. How Can Attrition Rates Be Reduced in Cancer Drug Discovery? Expert Opin. Drug Discov. 2013, 8, 363–368. [Google Scholar] [CrossRef] [PubMed]
- Quail, D.F.; Joyce, J.A. Microenvironmental Regulation of Tumor Progression and Metastasis. Nat. Med. 2013, 19, 1423–1437. [Google Scholar] [CrossRef]
- Bissell, M.J.; Radisky, D. Putting Tumours in Context. Nat. Rev. Cancer 2001, 1, 46–54. [Google Scholar] [CrossRef] [Green Version]
- Bizzarri, M.; Cucina, A. Tumor and the Microenvironment: A Chance to Reframe the Paradigm of Carcinogenesis? BioMed Res. Int. 2014, 2014, 934038. [Google Scholar] [CrossRef] [Green Version]
- Vermeulen, L.; De Sousa, E.; Melo, F.; van der Heijden, M.; Cameron, K.; de Jong, J.H.; Borovski, T.; Tuynman, J.B.; Todaro, M.; Merz, C.; et al. Wnt Activity Defines Colon Cancer Stem Cells and Is Regulated by the Microenvironment. Nat. Cell Biol. 2010, 12, 468–476. [Google Scholar] [CrossRef] [PubMed]
- Critelli, R.; Milosa, F.; Faillaci, F.; Condello, R.; Turola, E.; Marzi, L.; Lei, B.; Dituri, F.; Andreani, S.; Sighinolfi, P.; et al. Microenvironment Inflammatory Infiltrate Drives Growth Speed and Outcome of Hepatocellular Carcinoma: A Prospective Clinical Study. Cell Death Dis. 2017, 8, e3017. [Google Scholar] [CrossRef] [PubMed]
- Klemm, F.; Joyce, J.A. Microenvironmental Regulation of Therapeutic Response in Cancer. Trends Cell Biol. 2015, 25, 198–213. [Google Scholar] [CrossRef] [Green Version]
- Junttila, M.R.; de Sauvage, F.J. Influence of Tumour Micro-Environment Heterogeneity on Therapeutic Response. Nature 2013, 501, 346–354. [Google Scholar] [CrossRef] [PubMed]
- Mink, S.R.; Vashistha, S.; Zhang, W.; Hodge, A.; Agus, D.B.; Jain, A. Cancer-Associated Fibroblasts Derived from EGFR-TKI-Resistant Tumors Reverse EGFR Pathway Inhibition by EGFR-TKIs. Mol. Cancer Res. 2010, 8, 809–820. [Google Scholar] [CrossRef] [Green Version]
- Amornsupak, K.; Insawang, T.; Thuwajit, P.; O-Charoenrat, P.; Eccles, S.A.; Thuwajit, C. Cancer-Associated Fibroblasts Induce High Mobility Group Box 1 and Contribute to Resistance to Doxorubicin in Breast Cancer Cells. BMC Cancer 2014, 14, 955. [Google Scholar] [CrossRef] [PubMed]
- Nakasone, E.S.; Askautrud, H.A.; Kees, T.; Park, J.H.; Plaks, V.; Ewald, A.J.; Fein, M.; Rasch, M.G.; Tan, Y.X.; Qiu, J.; et al. Imaging Tumor-Stroma Interactions during Chemotherapy Reveals Contributions of the Microenvironment to Resistance. Cancer Cell 2012, 21, 488–503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jain, R.K.; Duda, D.G.; Clark, J.W.; Loeffler, J.S. Lessons from Phase III Clinical Trials on Anti-VEGF Therapy for Cancer. Nat. Clin. Pract. Oncol. 2006, 3, 24–40. [Google Scholar] [CrossRef]
- Pardoll, D.M. The Blockade of Immune Checkpoints in Cancer Immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef] [Green Version]
- Hill, B.S.; Sarnella, A.; D’Avino, G.; Zannetti, A. Recruitment of Stromal Cells into Tumour Microenvironment Promote the Metastatic Spread of Breast Cancer. Semin. Cancer Biol. 2020, 60, 202–213. [Google Scholar] [CrossRef]
- De Palma, M.; Biziato, D.; Petrova, T.V. Microenvironmental Regulation of Tumour Angiogenesis. Nat. Rev. Cancer 2017, 17, 457–474. [Google Scholar] [CrossRef]
- Winkler, J.; Abisoye-Ogunniyan, A.; Metcalf, K.J.; Werb, Z. Concepts of Extracellular Matrix Remodelling in Tumour Progression and Metastasis. Nat. Commun. 2020, 11, 5120. [Google Scholar] [CrossRef]
- Walker, C.; Mojares, E.; Hernández, A.D.R. Role of Extracellular Matrix in Development and Cancer Progression. Int. J. Mol. Sci. 2018, 19, 3028. [Google Scholar] [CrossRef] [Green Version]
- Liu, T.; Zhou, L.; Li, D.; Andl, T.; Zhang, Y. Cancer-Associated Fibroblasts Build and Secure the Tumor Microenvironment. Front. Cell Dev. Biol. 2019, 7, 60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Friedl, P.; Alexander, S. Cancer Invasion and the Microenvironment: Plasticity and Reciprocity. Cell 2011, 147, 992–1009. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kai, F.B.; Drain, A.P.; Weaver, V.M. The Extracellular Matrix Modulates the Metastatic Journey. Dev. Cell 2019, 49, 332–346. [Google Scholar] [CrossRef] [PubMed]
- Provenzano, P.P.; Inman, D.R.; Eliceiri, K.W.; Knittel, J.G.; Yan, L.; Rueden, C.T.; White, J.G.; Keely, P.J. Collagen Density Promotes Mammary Tumor Initiation and Progression. BMC Med. 2008, 6, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Neve, A.; Cantatore, F.P.; Maruotti, N.; Corrado, A.; Ribatti, D. Extracellular Matrix Modulates Angiogenesis in Physiological and Pathological Conditions. BioMed Res. Int. 2014, 2014, 756078. [Google Scholar] [CrossRef] [Green Version]
- Yamauchi, M.; Gibbons, D.L.; Zong, C.; Fradette, J.J.; Bota-Rabassedas, N.; Kurie, J.M. Fibroblast Heterogeneity and Its Impact on Extracellular Matrix and Immune Landscape Remodeling in Cancer. Matrix Biol. 2020, 91–92, 8–18. [Google Scholar] [CrossRef]
- Provenzano, P.P.; Eliceiri, K.W.; Campbell, J.M.; Inman, D.R.; White, J.G.; Keely, P.J. Collagen Reorganization at the Tumor-Stromal Interface Facilitates Local Invasion. BMC Med. 2006, 4, 38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lu, P.; Weaver, V.M.; Werb, Z. The Extracellular Matrix: A Dynamic Niche in Cancer Progression. J. Cell Biol. 2012, 196, 395–406. [Google Scholar] [CrossRef] [PubMed]
- Kessenbrock, K.; Plaks, V.; Werb, Z. Matrix Metalloproteinases: Regulators of the Tumor Microenvironment. Cell 2010, 141, 52–67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Perentes, J.Y.; Kirkpatrick, N.D.; Nagano, S.; Smith, E.Y.; Shaver, C.M.; Sgroi, D.; Garkavtsev, I.; Munn, L.L.; Jain, R.K.; Boucher, Y. Cancer Cell-Associated MT1-MMP Promotes Blood Vessel Invasion and Distant Metastasis in Triple-Negative Mammary Tumors. Cancer Res. 2011, 71, 4527–4538. [Google Scholar] [CrossRef] [Green Version]
- Mehner, C.; Hockla, A.; Miller, E.; Ran, S.; Radisky, D.C.; Radisky, E.S. Tumor Cell-Produced Matrix Metalloproteinase 9 (MMP-9) Drives Malignant Progression and Metastasis of Basal-like Triple Negative Breast Cancer. Oncotarget 2014, 5, 2736–2749. [Google Scholar] [CrossRef] [Green Version]
- Baker, A.M.; Bird, D.; Lang, G.; Cox, T.R.; Erler, J.T. Lysyl Oxidase Enzymatic Function Increases Stiffness to Drive Colorectal Cancer Progression through FAK. Oncogene 2013, 32, 1863–1868. [Google Scholar] [CrossRef] [Green Version]
- Levental, K.; Yu, H.; Kass, L.; Lakins, J.N.; Egeblad, M.; Erler, J.; Fong, S.F.; Csiszar, K.; Giaccia, A.; Weninger, W.; et al. Matrix Crosslinking Forces Tumor Progression by Enhancing Integrin Signaling. Cell 2009, 139, 891–906. [Google Scholar] [CrossRef] [Green Version]
- Blanco-Fernandez, B.; Gaspar, V.M.; Engel, E.; Mano, J.F. Proteinaceous Hydrogels for Bioengineering Advanced 3D Tumor Models. Adv. Sci. 2021, 8, 2003129. [Google Scholar] [CrossRef]
- Lee, H.J.; Mun, S.; Pham, D.M.; Kim, P. Extracellular Matrix-Based Hydrogels to Tailoring Tumor Organoids. ACS Biomater. Sci. Eng. 2021, 7, 4128–4135. [Google Scholar] [CrossRef]
- Micek, H.M.; Visetsouk, M.R.; Masters, K.S.; Kreeger, P.K. Engineering the Extracellular Matrix to Model the Evolving Tumor Microenvironment. iScience 2020, 23, 101742. [Google Scholar] [CrossRef]
- Nii, T.; Makino, K.; Tabata, Y. Three-Dimensional Culture System of Cancer Cells Combined with Biomaterials for Drug Screening. Cancers 2020, 12, 2754. [Google Scholar] [CrossRef]
- Wilding, J.L.; Bodmer, W. Cancer Cell Lines for Drug Discovery and Development. Cancer Res. 2014, 74, 2377–2384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bhimani, J.; Ball, K.; Stebbing, J. Patient-derived xenograft models—The future of personalised cancer treatment. Br. J. Cancer 2020, 122, 601–602. [Google Scholar] [CrossRef] [PubMed]
- Laplagne, C.; Domagala, M.; Le Naour, A.; Quemerais, C.; Hamel, D.; Fournié, J.-J.; Couderc, B.; Bousquet, C.; Ferrand, A.; Poupot, M. Latest Advances in Targeting the Tumor Microenvironment for Tumor Suppression. Int. J. Mol. Sci. 2019, 20, 4719. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Miki, Y.; Ono, K.; Hata, S.; Suzuki, T.; Kumamoto, H.; Sasano, H. The advantages of co-culture over mono cell culture in simulating in vivo environment. J. Steroid Biochem. Mol. Biol. 2012, 131, 68–75. [Google Scholar] [CrossRef]
- Fong, E.L.; Harrington, D.A.; Farach-Carson, M.C.; Yu, H. Heralding a new paradigm in 3D tumor modeling. Biomaterials 2016, 108, 197–213. [Google Scholar] [CrossRef] [Green Version]
- Choi, S.Y.C.; Lin, D.; Gout, P.W.; Collins, C.C.; Xu, Y.; Wang, Y. Lessons from patient-derived xenografts for better in vitro modeling of human cancer. Adv. Drug Deliv. Rev. 2014, 79–80, 222–237. [Google Scholar] [CrossRef] [Green Version]
- Stock, K.; Estrada, M.; Vidic, S.; Gjerde, K.; Rudisch, A.; Santo, V.E.; Barbier, M.; Blom, S.; Arundkar, S.C.; Selvam, I.; et al. Capturing tumor complexity in vitro: Comparative analysis of 2D and 3D tumor models for drug discovery. Sci. Rep. 2016, 6, 28951. [Google Scholar] [CrossRef] [Green Version]
- Kapałczyńska, M.; Kolenda, T.; Przybyła, W.; Zajączkowska, M.; Teresiak, A.; Filas, V.; Ibbs, M.; Bliźniak, R.; Łuczewski, L.; Lamperska, K. 2D and 3D cell cultures—A comparison of different types of cancer cell cultures. Arch. Med. Sci. 2016, 12, 910–919. [Google Scholar] [CrossRef]
- Breslin, S.; O’Driscoll, L. Three-dimensional cell culture: The missing link in drug discovery. Drug Discov. Today 2013, 18, 240–249. [Google Scholar] [CrossRef] [PubMed]
- Rodrigues, J.; Heinrich, M.A.; Teixeira, L.M.; Prakash, J. 3D In Vitro Model (R)evolution: Unveiling Tumor–Stroma Interactions. Trends Cancer 2021, 7, 249–264. [Google Scholar] [CrossRef]
- Lee, J.M.; Mhawech-Fauceglia, P.; Lee, N.; Parsanian, L.C.; Lin, Y.G.; Gayther, S.A.; Lawrenson, K. A three-dimensional microenvironment alters protein expression and chemosensitivity of epithelial ovarian cancer cells in vitro. Lab. Investig. 2013, 93, 528–542. [Google Scholar] [CrossRef] [Green Version]
- Luca, A.C.; Mersch, S.; Deenen, R.; Schmidt, S.; Messner, I.; Schäfer, K.-L.; Baldus, S.E.; Huckenbeck, W.; Piekorz, R.P.; Knoefel, W.T.; et al. Impact of the 3D Microenvironment on Phenotype, Gene Expression, and EGFR Inhibition of Colorectal Cancer Cell Lines. PLoS ONE 2013, 8, e59689. [Google Scholar] [CrossRef]
- Imamura, Y.; Mukohara, T.; Shimono, Y.; Funakoshi, Y.; Chayahara, N.; Toyoda, M.; Kiyota, N.; Takao, S.; Kono, S.; Nakatsura, T.; et al. Comparison of 2D- and 3D-culture models as drug-testing platforms in breast cancer. Oncol. Rep. 2015, 33, 1837–1843. [Google Scholar] [CrossRef] [Green Version]
- Riedl, A.; Schlederer, M.; Pudelko, K.; Stadler, M.; Walter, S.; Unterleuthner, D.; Unger, C.; Kramer, N.; Hengstschläger, M.; Kenner, L.; et al. Comparison of cancer cells in 2D vs 3D culture reveals differences in AKT–mTOR–S6K signaling and drug responses. J. Cell Sci. 2017, 130, 203–218. [Google Scholar] [CrossRef] [Green Version]
- Costa, E.C.; Moreira, A.F.; Diogo, D.M.D.M.; Gaspar, V.; Carvalho, M.P.; Correia, I.J. 3D tumor spheroids: An overview on the tools and techniques used for their analysis. Biotechnol. Adv. 2016, 34, 1427–1441. [Google Scholar] [CrossRef]
- Weiswald, L.-B.; Bellet, D.; Dangles-Marie, V. Spherical Cancer Models in Tumor Biology. Neoplasia 2015, 17, 1–15. [Google Scholar] [CrossRef] [Green Version]
- Fitzgerald, A.A.; Li, E.; Weiner, L.M. 3D Culture Systems for Exploring Cancer Immunology. Cancers 2020, 13, 56. [Google Scholar] [CrossRef]
- Reidy, E.; Leonard, N.A.; Treacy, O.; Ryan, A.E. A 3D View of Colorectal Cancer Models in Predicting Therapeutic Responses and Resistance. Cancers 2021, 13, 227. [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]
- Lazzari, G.; Nicolas, V.; Matsusaki, M.; Akashi, M.; Couvreur, P.; Mura, S. Multicellular spheroid based on a triple co-culture: A novel 3D model to mimic pancreatic tumor complexity. Acta Biomater. 2018, 78, 296–307. [Google Scholar] [CrossRef] [PubMed]
- Santo, V.E.; Estrada, M.; Rebelo, S.P.; Abreu, S.; Silva, I.; Pinto, C.; Veloso, S.C.; Serra, A.T.; Boghaert, E.; Alves, P.; et al. Adaptable stirred-tank culture strategies for large scale production of multicellular spheroid-based tumor cell models. J. Biotechnol. 2016, 221, 118–129. [Google Scholar] [CrossRef]
- Fey, S.J.; Wrzesinski, K. Determination of Drug Toxicity Using 3D Spheroids Constructed From an Immortal Human Hepatocyte Cell Line. Toxicol. Sci. 2012, 127, 403–411. [Google Scholar] [CrossRef]
- Däster, S.; Amatruda, N.; Calabrese, D.; Ivanek, R.; Turrini, E.; Droeser, R.A.; Zajac, P.; Fimognari, C.; Spagnoli, G.C.; Iezzi, G.; et al. Induction of hypoxia and necrosis in multicellular tumor spheroids is associated with resistance to chemotherapy treatment. Oncotarget 2017, 8, 1725–1736. [Google Scholar] [CrossRef] [Green Version]
- Fang, Y.; Eglen, R.M. Three-Dimensional Cell Cultures in Drug Discovery and Development. SLAS Discov. 2017, 22, 456–472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Santo, V.E.; Rebelo, S.P.; Estrada, M.; Alves, P.; Boghaert, E.; Brito, C. Drug screening in 3D in vitro tumor models: Overcoming current pitfalls of efficacy read-outs. Biotechnol. J. 2017, 12, 1600505. [Google Scholar] [CrossRef]
- Schutgens, F.; Clevers, H. Human Organoids: Tools for Understanding Biology and Treating Diseases. Annu. Rev. Pathol. Mech. Dis. 2020, 15, 211–234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Weeber, F.; van de Wetering, M.; Hoogstraat, M.; Dijkstra, K.K.; Krijgsman, O.; Kuilman, T.; Gadellaa-van Hooijdonk, C.G.; van der Velden, D.L.; Peeper, D.S.; Cuppen, E.P.; et al. Preserved genetic diversity in organoids cultured from biopsies of human colorectal cancer metastases. Proc. Natl. Acad. Sci. USA 2015, 112, 13308–13311. [Google Scholar] [CrossRef] [Green Version]
- Rosenbluth, J.M.; Schackmann, R.C.J.; Gray, G.K.; Selfors, L.M.; Li, C.M.-C.; Boedicker, M.; Kuiken, H.J.; Richardson, A.; Brock, J.; Garber, J.; et al. Organoid cultures from normal and cancer-prone human breast tissues preserve complex epithelial lineages. Nat. Commun. 2020, 11, 1711. [Google Scholar] [CrossRef] [Green Version]
- Baker, L.; Tiriac, H.; Clevers, H.; Tuveson, D.A. Modeling Pancreatic Cancer with Organoids. Trends Cancer 2016, 2, 176–190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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]
- Sachs, N.; De Ligt, J.; Kopper, O.; Gogola, E.; Bounova, G.; Weeber, F.; Balgobind, A.V.; Wind, K.; Gracanin, A.; Begthel, H.; et al. A Living Biobank of Breast Cancer Organoids Captures Disease Heterogeneity. Cell 2018, 172, 373–386.e10. [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] [Green Version]
- Tsai, S.; McOlash, L.; Palen, K.; Johnson, B.; Duris, C.; Yang, Q.; Dwinell, M.B.; Hunt, B.; Evans, D.B.; Gershan, J.; et al. Development of primary human pancreatic cancer organoids, matched stromal and immune cells and 3D tumor microenvironment models. BMC Cancer 2018, 18, 335. [Google Scholar] [CrossRef] [PubMed]
- Bar-Ephraim, Y.E.; Kretzschmar, K.; Clevers, H. Organoids in immunological research. Nat. Rev. Immunol. 2020, 20, 279–293. [Google Scholar] [CrossRef]
- Hughes, C.S.; Postovit, L.M.; Lajoie, G.A. Matrigel: A complex protein mixture required for optimal growth of cell culture. Proteomics 2010, 10, 1886–1890. [Google Scholar] [CrossRef]
- Benton, G.; Arnaoutova, I.; George, J.; Kleinman, H.K.; Koblinski, J. Matrigel: From discovery and ECM mimicry to assays and models for cancer research. Adv. Drug Deliv. Rev. 2014, 79–80, 3–18. [Google Scholar] [CrossRef]
- Asghar, W.; El Assal, R.; Shafiee, H.; Pitteri, S.; Paulmurugan, R.; Demirci, U. Engineering cancer microenvironments for in vitro 3-D tumor models. Mater. Today 2015, 18, 539–553. [Google Scholar] [CrossRef]
- de Sampaio, P.C.; Auslaender, D.; Krubasik, D.; Failla, A.V.; Skepper, J.N.; Murphy, G.; English, W.R. A Heterogeneous In Vitro Three Dimensional Model of Tumour-Stroma Interactions Regulating Sprouting Angiogenesis. PLoS ONE 2012, 7, e30753. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Gjorevski, N.; Sachs, N.; Manfrin, A.; Giger, S.; Bragina, M.E.; Ordóñez-Morán, P.; Clevers, N.S.H.; Lutolf, N.G.A.M.S.G.M.E.B.M.P. Designer matrices for intestinal stem cell and organoid culture. Nat. Cell Biol. 2016, 539, 560–564. [Google Scholar] [CrossRef]
- Aisenbrey, E.A.; Murphy, W.L. Synthetic alternatives to Matrigel. Nat. Rev. Mater. 2020, 5, 539–551. [Google Scholar] [CrossRef] [PubMed]
- Chwalek, K.; Tsurkan, M.; Freudenberg, U.; Werner, C. Glycosaminoglycan-based hydrogels to modulate heterocellular communication in in vitro angiogenesis models. Sci. Rep. 2015, 4, 4414. [Google Scholar] [CrossRef] [Green Version]
- Ma, Y.-H.V.; Middleton, K.; You, L.; Sun, Y. A review of microfluidic approaches for investigating cancer extravasation during metastasis. Microsyst. Nanoeng. 2018, 4, 17104. [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]
- Shemesh, J.; Jalilian, I.; Shi, A.; Yeoh, G.H.; Tate, M.L.K.; Warkiani, M.E. Flow-induced stress on adherent cells in microfluidic devices. Lab Chip 2015, 15, 4114–4127. [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] [PubMed] [Green Version]
- 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]
- Sahai, E.; Astsaturov, I.; Cukierman, E.; DeNardo, D.G.; Egeblad, M.; Evans, R.M.; Fearon, D.; Greten, F.R.; Hingorani, S.R.; Hunter, T.; et al. A framework for advancing our understanding of cancer-associated fibroblasts. Nat. Rev. Cancer 2020, 20, 174–186. [Google Scholar] [CrossRef] [Green Version]
- Gascard, P.; Tlsty, T.D. Carcinoma-associated fibroblasts: Orchestrating the composition of malignancy. Genes Dev. 2016, 30, 1002–1019. [Google Scholar] [CrossRef]
- Jeong, S.-Y.; Lee, J.-H.; Shin, Y.; Chung, S.; Kuh, H.-J. Co-Culture of Tumor Spheroids and Fibroblasts in a Collagen Matrix-Incorporated Microfluidic Chip Mimics Reciprocal Activation in Solid Tumor Microenvironment. PLoS ONE 2016, 11, e0159013. [Google Scholar] [CrossRef] [Green Version]
- Öhlund, D.; Handly-Santana, A.; Biffi, G.; Elyada, E.; Almeida, A.S.; Ponz-Sarvise, M.; Corbo, V.; Oni, T.E.; Hearn, S.A.; Lee, E.J.; et al. Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. J. Exp. Med. 2017, 214, 579–596. [Google Scholar] [CrossRef]
- Onion, D.; Argent, R.H.; Reece-Smith, A.M.; Craze, M.L.; Pineda, R.G.; Clarke, P.; Ratan, H.L.; Parsons, S.L.; Lobo, D.; Duffy, J.P.; et al. 3-Dimensional Patient-Derived Lung Cancer Assays Reveal Resistance to Standards-of-Care Promoted by Stromal Cells but Sensitivity to Histone Deacetylase Inhibitors. Mol. Cancer Ther. 2016, 15, 753–763. [Google Scholar] [CrossRef] [Green Version]
- Attieh, Y.; Vignjevic, D.M. The hallmarks of CAFs in cancer invasion. Eur. J. Cell Biol. 2016, 95, 493–502. [Google Scholar] [CrossRef]
- Li, M.; Li, M.; Yin, T.; Shi, H.; Wen, Y.; Zhang, B.; Chen, M.; Xu, G.; Ren, K.; Wei, Y. Targeting of cancer-associated fibroblasts enhances the efficacy of cancer chemotherapy by regulating the tumor microenvironment. Mol. Med. Rep. 2016, 13, 2476–2484. [Google Scholar] [CrossRef] [Green Version]
- Wang, L.-C.S.; Lo, A.; Scholler, J.; Sun, J.; Majumdar, R.S.; Kapoor, V.; Antzis, M.; Cotner, C.; Johnson, L.A.; Durham, A.C.; et al. Targeting Fibroblast Activation Protein in Tumor Stroma with Chimeric Antigen Receptor T Cells Can Inhibit Tumor Growth and Augment Host Immunity without Severe Toxicity. Cancer Immunol. Res. 2014, 2, 154–166. [Google Scholar] [CrossRef] [Green Version]
- Lo, A.; Wang, L.-C.S.; Scholler, J.; Monslow, J.; Avery, D.; Newick, K.; O’Brien, S.; Evans, R.A.; Bajor, D.J.; Clendenin, C.; et al. Tumor-Promoting Desmoplasia Is Disrupted by Depleting FAP-Expressing Stromal Cells. Cancer Res. 2015, 75, 2800–2810. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fabre, M.; Ferrer, C.; Domínguez-Hormaetxe, S.; Bockorny, B.; Murias, L.; Seifert, O.; Eisler, S.A.; Kontermann, R.E.; Pfizenmaier, K.; Lee, S.Y.; et al. OMTX705, a Novel FAP-Targeting ADC Demonstrates Activity in Chemotherapy and Pembrolizumab-Resistant Solid Tumor Models. Clin. Cancer Res. 2020, 26, 3420–3430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Su, S.; Chen, J.; Yao, H.; Liu, J.; Yu, S.; Lao, L.; Wang, M.; Luo, M.; Xing, Y.; Chen, F.; et al. CD10(+)GPR77(+) Cancer-Associated Fibroblasts Promote Cancer Formation and Chemoresistance by Sustaining Cancer Stemness. Cell 2018, 172, 841–856.e16. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.-A.; Lee, E.K.; Kuh, H.-J. Co-culture of 3D tumor spheroids with fibroblasts as a model for epithelial–mesenchymal transition in vitro. Exp. Cell Res. 2015, 335, 187–196. [Google Scholar] [CrossRef]
- Dolznig, H.; Rupp, C.; Puri, C.; Haslinger, C.; Schweifer, N.; Wieser, E.; Kerjaschki, D.; Garin-Chesa, P. Modeling colon adenocarcinomas in vitro a 3D co-culture system induces cancer-relevant pathways upon tumor cell and stromal fibroblast interaction. Am. J. Pathol. 2011, 179, 487–501. [Google Scholar] [CrossRef]
- Attieh, Y.; Clark, A.G.; Grass, C.; Richon, S.; Pocard, M.; Mariani, P.; Elkhatib, N.; Betz, T.; Gurchenkov, B.; Vignjevic, D.M. Cancer-associated fibroblasts lead tumor invasion through integrin-β3–dependent fibronectin assembly. J. Cell Biol. 2017, 216, 3509–3520. [Google Scholar] [CrossRef] [Green Version]
- Horie, M.; Saito, A.; Mikami, Y.; Ohshima, M.; Morishita, Y.; Nakajima, J.; Kohyama, T.; Nagase, T. Characterization of human lung cancer-associated fibroblasts in three-dimensional in vitro co-culture model. Biochem. Biophys. Res. Commun. 2012, 423, 158–163. [Google Scholar] [CrossRef]
- Jacobi, N.; Seeboeck, R.; Hofmann, E.; Schweiger, H.; Smolinska, V.; Mohr, T.; Boyer, A.; Sommergruber, W.; Lechner, P.; Pichler-Huebschmann, C.; et al. Organotypic three-dimensional cancer cell cultures mirror drug responses in vivo: Lessons learned from the inhibition of EGFR signaling. Oncotarget 2017, 8, 107423–107440. [Google Scholar] [CrossRef] [Green Version]
- Wang, W.; Li, Q.; Yamada, T.; Matsumoto, K.; Matsumoto, I.; Oda, M.; Watanabe, G.; Kayano, Y.; Nishioka, Y.; Sone, S.; et al. Crosstalk to Stromal Fibroblasts Induces Resistance of Lung Cancer to Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors. Clin. Cancer Res. 2009, 15, 6630–6638. [Google Scholar] [CrossRef] [Green Version]
- Nii, T.; Makino, K.; Tabata, Y. A Cancer Invasion Model Combined with Cancer-Associated Fibroblasts Aggregates Incorporating Gelatin Hydrogel Microspheres Containing a p53 Inhibitor. Tissue Eng. Part C Methods 2019, 25, 711–720. [Google Scholar] [CrossRef]
- Estrada, M.; Rebelo, S.P.; Davies, E.J.; Pinto, M.; Pereira, H.A.; Santo, V.E.; Smalley, M.; Barry, S.T.; Gualda, E.J.; Alves, P.; et al. Modelling the tumour microenvironment in long-term microencapsulated 3D co-cultures recapitulates phenotypic features of disease progression. Biomaterials 2016, 78, 50–61. [Google Scholar] [CrossRef] [Green Version]
- Dondajewska, E.; Juzwa, W.; Mackiewicz, A.; Dams-Kozlowska, H. Heterotypic breast cancer model based on a silk fibroin scaffold to study the tumor microenvironment. Oncotarget 2017, 9, 4935–4950. [Google Scholar] [CrossRef]
- Kunz-Schughart, L.; Heyder, P.; Schroeder, J.; Knuechel, R. A Heterologous 3-D Coculture Model of Breast Tumor Cells and Fibroblasts to Study Tumor-Associated Fibroblast Differentiation. Exp. Cell Res. 2001, 266, 74–86. [Google Scholar] [CrossRef]
- Seidl, P.; Huettinger, R.; Knuechel, R.; Kunz-Schughart, L. Three-dimensional fibroblast-tumor cell interaction causes downregulation ofRACK1 mRNA expression in breast cancer cellsin vitro. Int. J. Cancer 2002, 102, 129–136. [Google Scholar] [CrossRef]
- Kaur, P.; Ward, B.; Saha, B.; Young, L.; Groshen, S.; Techy, G.; Lu, Y.; Atkinson, R.; Taylor, C.R.; Ingram, M.; et al. Human Breast Cancer Histoid. J. Histochem. Cytochem. 2011, 59, 1087–1100. [Google Scholar] [CrossRef] [Green Version]
- Angelucci, C.; Maulucci, G.; Lama, G.; Proietti, G.; Colabianchi, A.; Papi, M.; Maiorana, A.; De Spirito, M.; Micera, A.; Balzamino, O.B.; et al. Epithelial-Stromal Interactions in Human Breast Cancer: Effects on Adhesion, Plasma Membrane Fluidity and Migration Speed and Directness. PLoS ONE 2012, 7, e50804. [Google Scholar] [CrossRef] [PubMed]
- Anastasov, N.; Höfig, I.; Radulovic, V.; Ströbel, S.; Salomon, M.; Lichtenberg, J.; Rothenaigner, I.; Hadian, K.; Kelm, J.M.; Thirion, C.; et al. A 3D-microtissue-based phenotypic screening of radiation resistant tumor cells with synchronized chemotherapeutic treatment. BMC Cancer 2015, 15, 466. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yakavets, I.; Francois, A.; Benoit, A.; Merlin, J.-L.; Bezdetnaya, L.; Vogin, G. Advanced co-culture 3D breast cancer model for investigation of fibrosis induced by external stimuli: Optimization study. Sci. Rep. 2020, 10, 21273. [Google Scholar] [CrossRef] [PubMed]
- Xin, X.; Yang, S.-T. A Dual Fluorescent 3-D Multicellular Coculture of Breast Cancer MCF-7 and Fibroblast NIH-3T3 Cells for High Throughput Cancer Drug Screening. Biochem. Eng. J. 2019, 148, 152–161. [Google Scholar] [CrossRef]
- Saini, H.; Eliato, K.R.; Veldhuizen, J.; Zare, A.; Allam, M.; Silva, C.; Kratz, A.; Truong, D.; Mouneimne, G.; LaBaer, J.; et al. The role of tumor-stroma interactions on desmoplasia and tumorigenicity within a microengineered 3D platform. Biomaterials 2020, 247, 119975. [Google Scholar] [CrossRef] [PubMed]
- Hanley, C.J.; Henriet, E.; Sirka, O.K.; Thomas, G.J.; Ewald, A.J. Tumor-Resident Stromal Cells Promote Breast Cancer Invasion through Regulation of the Basal Phenotype. Mol. Cancer Res. 2020, 18, 1615–1622. [Google Scholar] [CrossRef]
- Liu, C.; Lewin Mejia, D.; Chiang, B.; Luker, K.E.; Luker, G.D. Hybrid collagen alginate hydrogel as a platform for 3D tumor spheroid invasion. Acta Biomater. 2018, 75, 213–225. [Google Scholar] [CrossRef]
- Froeling, F.E.; Mirza, T.A.; Feakins, R.M.; Seedhar, A.; Elia, G.; Hart, I.R.; Kocher, H. Organotypic Culture Model of Pancreatic Cancer Demonstrates that Stromal Cells Modulate E-Cadherin, β-Catenin, and Ezrin Expression in Tumor Cells. Am. J. Pathol. 2009, 175, 636–648. [Google Scholar] [CrossRef] [Green Version]
- Noel, P.; Muñoz, R.; Rogers, G.W.; Neilson, A.; Von Hoff, D.D.; Han, H. Preparation and Metabolic Assay of 3-dimensional Spheroid Co-cultures of Pancreatic Cancer Cells and Fibroblasts. J. Vis. Exp. 2017, e56081. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brancato, V.; Comunanza, V.; Imparato, G.; Corà, D.; Urciuolo, F.; Noghero, A.; Bussolino, F.; Netti, P.A. Bioengineered tumoral microtissues recapitulate desmoplastic reaction of pancreatic cancer. Acta Biomater. 2017, 49, 152–166. [Google Scholar] [CrossRef] [PubMed]
- Ware, M.; Keshishian, V.; Law, J.J.; Ho, J.C.; Favela, C.A.; Rees, P.; Smith, B.; Mohammad, S.; Hwang, R.F.; Rajapakshe, K.; et al. Generation of an in vitro 3D PDAC stroma rich spheroid model. Biomaterials 2016, 108, 129–142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Majety, M.; Pradel, L.P.; Gies, M.; Ries, C.H. Fibroblasts Influence Survival and Therapeutic Response in a 3D Co-Culture Model. PLoS ONE 2015, 10, e0127948. [Google Scholar] [CrossRef] [PubMed]
- Yip, D.; Cho, C.H. A multicellular 3D heterospheroid model of liver tumor and stromal cells in collagen gel for anti-cancer drug testing. Biochem. Biophys. Res. Commun. 2013, 433, 327–332. [Google Scholar] [CrossRef]
- Liu, J.; Li, P.; Wang, L.; Li, M.; Ge, Z.; Noordam, L.; Lieshout, R.; Verstegen, M.M.; Ma, B.; Su, J.; et al. Cancer-Associated Fibroblasts Provide a Stromal Niche for Liver Cancer Organoids That Confers Trophic Effects and Therapy Resistance. Cell. Mol. Gastroenterol. Hepatol. 2021, 11, 407–431. [Google Scholar] [CrossRef] [PubMed]
- Clark, A.K.; Taubenberger, A.; Taylor, R.; Niranjan, B.; Chea, Z.Y.; Zotenko, E.; Sieh, S.; Pedersen, J.S.; Norden, S.; Frydenberg, M.; et al. A bioengineered microenvironment to quantitatively measure the tumorigenic properties of cancer-associated fibroblasts in human prostate cancer. Biomaterials 2013, 34, 4777–4785. [Google Scholar] [CrossRef] [PubMed]
- Liu, T.; Lin, B.; Qin, J. Carcinoma-associated fibroblasts promoted tumor spheroid invasion on a microfluidic 3D co-culture device. Lab Chip 2010, 10, 1671–1677. [Google Scholar] [CrossRef] [PubMed]
- Xu, F.; Celli, J.; Rizvi, I.; Moon, S.; Hasan, T.; Demirci, U. A three-dimensional in vitro ovarian cancer coculture model using a high-throughput cell patterning platform. Biotechnol. J. 2011, 6, 204–212. [Google Scholar] [CrossRef] [PubMed]
- Priwitaningrum, D.L.; Blondé, J.-B.G.; Sridhar, A.; van Baarlen, J.; Hennink, W.E.; Storm, G.; Le Gac, S.; Prakash, J. Tumor stroma-containing 3D spheroid arrays: A tool to study nanoparticle penetration. J. Control. Release 2016, 244, 257–268. [Google Scholar] [CrossRef] [PubMed]
- Wenzel, C.; Otto, S.; Prechtl, S.; Parczyk, K.; Steigemann, P. A novel 3D high-content assay identifies compounds that prevent fibroblast invasion into tissue surrogates. Exp. Cell Res. 2015, 339, 35–43. [Google Scholar] [CrossRef]
- Zhao, L.; Liu, Y.; Liu, Y.; Zhang, M.; Zhang, X. Microfluidic Control of Tumor and Stromal Cell Spheroids Pairing and Merging for Three-Dimensional Metastasis Study. Anal. Chem. 2020, 92, 7638–7645. [Google Scholar] [CrossRef] [PubMed]
- Fang, X.; Sittadjody, S.; Gyabaah, K.; Opara, E.C.; Balaji, K.C. Novel 3D Co-Culture Model for Epithelial-Stromal Cells Interaction in Prostate Cancer. PLoS ONE 2013, 8, e75187. [Google Scholar] [CrossRef] [Green Version]
- Nissen, N.I.; Karsdal, M.; Willumsen, N. Collagens and Cancer associated fibroblasts in the reactive stroma and its relation to Cancer biology. J. Exp. Clin. Cancer Res. 2019, 38, 1–12. [Google Scholar] [CrossRef] [Green Version]
- Rebelo, S.P.; Pinto, C.; Martins, T.R.; Harrer, N.; Estrada, M.; Loza-Alvarez, P.; Cabeçadas, J.; Alves, P.M.; Gualda, E.J.; Sommergruber, W.; et al. 3D-3-culture: A tool to unveil macrophage plasticity in the tumour microenvironment. Biomaterials 2018, 163, 185–197. [Google Scholar] [CrossRef]
- Holliday, D.L.; Brouilette, K.T.; Markert, A.; Gordon, L.A.; Jones, J.L. Novel multicellular organotypic models of normal and malignant breast: Tools for dissecting the role of the microenvironment in breast cancer progression. Breast Cancer Res. 2009, 11, 3–11. [Google Scholar] [CrossRef] [Green Version]
- Cho, J.; Lee, H.-J.; Hwang, S.J.; Min, H.-Y.; Na Kang, H.; Park, A.-Y.; Hyun, S.Y.; Sim, J.Y.; Lee, H.J.; Jang, H.-J.; et al. The interplay between slow-cycling, chemoresistant cancer cells and fibroblasts creates a proinflammatory niche for tumor progression. Cancer Res. 2020, 80, 2257–2272. [Google Scholar] [CrossRef] [Green Version]
- Hwang, R.F.; Moore, T.; Arumugam, T.; Ramachandran, V.; Amos, K.D.; Rivera, A.; Ji, B.; Evans, D.B.; Logsdon, C.D. Cancer-Associated Stromal Fibroblasts Promote Pancreatic Tumor Progression. Cancer Res. 2008, 68, 918–926. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shekhar, M.P.; Werdell, J.; Santner, S.J.; Pauley, R.J.; Tait, L. Breast Stroma Plays a Dominant Regulatory Role in Breast Epi-thelial Growth and Differentiation: Implications for Tumor Development and Progression. Cancer Res. 2001, 61, 1320–1326. [Google Scholar]
- Weigelt, B.; Lo, A.T.; Park, C.C.; Gray, J.W.; Bissell, M.J. HER2 signaling pathway activation and response of breast cancer cells to HER2-targeting agents is dependent strongly on the 3D microenvironment. Breast Cancer Res. Treat. 2009, 122, 35–43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pickl, M.; Ries, C.H. Comparison of 3D and 2D tumor models reveals enhanced HER2 activation in 3D associated with an increased response to trastuzumab. Oncogene 2008, 28, 461–468. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ekert, J.E.; Johnson, K.; Strake, B.; Pardinas, J.; Jarantow, S.; Perkinson, R.; Colter, D.C. Three-Dimensional Lung Tumor Microenvironment Modulates Therapeutic Compound Responsiveness In Vitro—Implication for Drug Development. PLoS ONE 2014, 9, e92248. [Google Scholar] [CrossRef]
- Choe, C.; Shin, Y.-S.; Choi, S.-J.; Lee, J.; Kim, S.Y.; Cho, Y.B.; Kim, J.; Kim, C. Crosstalk with cancer-associated fibroblasts induces resistance of non-small cell lung cancer cells to epidermal growth factor receptor tyrosine kinase inhibition. OncoTargets Ther. 2015, 8, 3665–3678. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Noriega-Guerra, H.; Freitas, V.M. Extracellular Matrix Influencing HGF/c-MET Signaling Pathway: Impact on Cancer Progression. Int. J. Mol. Sci. 2018, 19, 3300. [Google Scholar] [CrossRef] [Green Version]
- Rupp, C.; Dolznig, H.; Puri, C.; Schweifer, N.; Sommergruber, W.; Kraut, N.; Rettig, W.J.; Kerjaschki, D.; Garin-Chesa, P. Laser Capture Microdissection of Epithelial Cancers Guided by Antibodies Against Fibroblast Activation Protein and Endosialin. Diagn. Mol. Pathol. 2006, 15, 35–42. [Google Scholar] [CrossRef]
- Helms, E.; Onate, M.K.; Sherman, M.H. Fibroblast Heterogeneity in the Pancreatic Tumor Microenvironment. Cancer Discov. 2020, 10, 648–656. [Google Scholar] [CrossRef] [Green Version]
- Herrera, M.; Islam, A.B.; Herrera, A.; Martín, P.; García, V.; Silva, J.; Garcia, J.M.; Salas, C.; Casal, I.; De Herreros, A.G.; et al. Functional Heterogeneity of Cancer-Associated Fibroblasts from Human Colon Tumors Shows Specific Prognostic Gene Expression Signature. Clin. Cancer Res. 2013, 19, 5914–5926. [Google Scholar] [CrossRef] [Green Version]
- Sebastian, A.; Hum, N.R.; Martin, K.A.; Gilmore, S.F.; Peran, I.; Byers, S.W.; Wheeler, E.K.; Coleman, M.A.; Loots, G.G. Single-Cell Transcriptomic Analysis of Tumor-Derived Fibroblasts and Normal Tissue-Resident Fibroblasts Reveals Fibroblast Heterogeneity in Breast Cancer. Cancers 2020, 12, 1307. [Google Scholar] [CrossRef]
- Kidd, S.; Spaeth, E.; Watson, K.; Burks, J.; Lu, H.; Klopp, A.; Andreeff, M.; Marini, F.C. Origins of the Tumor Microenvironment: Quantitative Assessment of Adipose-Derived and Bone Marrow–Derived Stroma. PLoS ONE 2012, 7, e30563. [Google Scholar] [CrossRef] [Green Version]
- Räsänen, K.; Vaheri, A. Activation of fibroblasts in cancer stroma. Exp. Cell Res. 2010, 316, 2713–2722. [Google Scholar] [CrossRef]
- Bartoschek, M.; Oskolkov, N.; Bocci, M.; Lövrot, J.; Larsson, C.; Sommarin, M.; Madsen, C.D.; Lindgren, D.; Pekar, G.; Karlsson, G.; et al. Spatially and functionally distinct subclasses of breast cancer-associated fibroblasts revealed by single cell RNA sequencing. Nat. Commun. 2018, 9, 5150. [Google Scholar] [CrossRef] [Green Version]
- Torres, S.; Bartolome, R.A.; Mendes, M.; Barderas, R.; Fernández-Aceñerp, M.J.; Peláez-García, A.; Peña, C.; Lopez-Lucendo, M.; Villar-Vázquez, R.; De Herreros, A.G.; et al. Proteome Profiling of Cancer-Associated Fibroblasts Identifies Novel Proinflammatory Signatures and Prognostic Markers for Colorectal Cancer. Clin. Cancer Res. 2013, 19, 6006–6019. [Google Scholar] [CrossRef] [Green Version]
- González-Silva, L.; Quevedo, L.; Varela, I. Tumor Functional Heterogeneity Unraveled by scRNA-seq Technologies. Trends Cancer 2020, 6, 13–19. [Google Scholar] [CrossRef] [Green Version]
- Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The Next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
- Lu, J.; Ye, X.; Fan, F.; Xia, L.; Bhattacharya, R.; Bellister, S.; Tozzi, F.; Sceusi, E.; Zhou, Y.; Tachibana, I.; et al. Endothelial Cells Promote the Colorectal Cancer Stem Cell Phenotype through a Soluble Form of Jagged-1. Cancer Cell 2013, 23, 171–185. [Google Scholar] [CrossRef] [Green Version]
- Hida, K.; Maishi, N.; Sakurai, Y.; Hida, Y.; Harashima, H. Heterogeneity of Tumor Endothelial Cells and Drug Delivery. Adv. Drug Deliv. Reviews. 2016, 99, 140–147. [Google Scholar] [CrossRef]
- Aird, W.C. Endothelial Cell Heterogeneity. Cold Spring Harb. Perspect. Med. 2011, 2, a006429. [Google Scholar] [CrossRef]
- Gunti, S.; Hoke, A.; Vu, K.; London, N. Organoid and Spheroid Tumor Models: Techniques and Applications. Cancers 2021, 13, 874. [Google Scholar] [CrossRef]
- Upreti, M.; Jamshidi-Parsian, A.; Koonce, N.A.; Webber, J.S.; Sharma, S.K.; Asea, A.; Mader, M.J.; Griffin, R. Tumor-Endothelial Cell Three-dimensional Spheroids: New Aspects to Enhance Radiation and Drug Therapeutics. Transl. Oncol. 2011, 4, 365–376. [Google Scholar] [CrossRef] [Green Version]
- Chiew, G.G.Y.; Wei, N.; Sultania, S.; Lim, S.; Luo, K.Q. Bioengineered three-dimensional co-culture of cancer cells and endothelial cells: A model system for dual analysis of tumor growth and angiogenesis. Biotechnol. Bioeng. 2017, 114, 1865–1877. [Google Scholar] [CrossRef]
- Shoval, H.; Karsch-Bluman, A.; Brill-Karniely, Y.; Stern, T.; Zamir, G.; Hubert, A.; Benny, O. Tumor cells and their crosstalk with endothelial cells in 3D spheroids. Sci. Rep. 2017, 7, 10428. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Ingthorsson, S.; Sigurdsson, V.; Fridriksdottir, A.J.; Jonasson, J.G.; Kjartansson, J.; Magnusson, M.K.; Gudjonsson, T. Endothelial cells stimulate growth of normal and cancerous breast epithelial cells in 3D culture. BMC Res. Notes 2010, 3, 184. [Google Scholar] [CrossRef] [Green Version]
- Roudsari, L.C.; Jeffs, S.E.; Witt, A.S.; Gill, B.J.; West, J.L. A 3D Poly(ethylene glycol)-based Tumor Angiogenesis Model to Study the Influence of Vascular Cells on Lung Tumor Cell Behavior. Sci. Rep. 2016, 6, 32726. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Moya, M.L.; Hsu, Y.-H.; Lee, A.; Hughes, C.C.; George, S.C. In Vitro Perfused Human Capillary Networks. Tissue Eng. Part C Methods 2013, 19, 730–737. [Google Scholar] [CrossRef] [Green Version]
- Aref, A.R.; Huang, Y.; Thiery, J.P.; Kamm, R.D.; Huang, R.Y.J.; Yu, W.; Chua, K.N.; Sun, W.; Tu, T.Y.; Bai, J.; et al. Screening Therapeutic EMT Blocking Agents in a Threedimensional Microenvi-ronment. Integr. Biol. 2013, 5, 381–389. [Google Scholar] [CrossRef] [Green Version]
- 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] [Green Version]
- Roudsari, L.C.; West, J.L. Studying the influence of angiogenesis in in vitro cancer model systems. Adv. Drug Deliv. Rev. 2016, 97, 250–259. [Google Scholar] [CrossRef]
- Song, J.; Cavnar, S.P.; Walker, A.C.; Luker, K.; Gupta, M.; Tung, Y.-C.; Luker, G.D.; Takayama, S. Microfluidic Endothelium for Studying the Intravascular Adhesion of Metastatic Breast Cancer Cells. PLoS ONE 2009, 4, e5756. [Google Scholar] [CrossRef] [Green Version]
- Nagaraju, S.; Truong, D.; Mouneimne, G.; Nikkhah, M. Microfluidic Tumor–Vascular Model to Study Breast Cancer Cell Invasion and Intravasation. Adv. Healthc. Mater. 2018, 7, e1701257. [Google Scholar] [CrossRef]
- Carvalho, M.R.; Barata, D.; Teixeira, L.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]
- Du, X.; Li, W.; Du, G.-S.; Cho, H.; Yu, M.; Fang, Q.; Lee, L.P.; Fang, J. Droplet Array-Based 3D Coculture System for High-Throughput Tumor Angiogenesis Assay. Anal. Chem. 2018, 90, 3253–3261. [Google Scholar] [CrossRef]
- Truong, D.; Fiorelli, R.; Barrientos, E.S.; Melendez, E.L.; Sanai, N.; Mehta, S.; Nikkhah, M. A three-dimensional (3D) organotypic microfluidic model for glioma stem cells—Vascular interactions. Biomaterials 2019, 198, 63–77. [Google Scholar] [CrossRef]
- Ko, J.; Ahn, J.; Kim, S.; Lee, Y.; Lee, J.; Park, D.; Jeon, N.L. Tumor spheroid-on-a-chip: A standardized microfluidic culture platform for investigating tumor angiogenesis. Lab Chip 2019, 19, 2822–2833. [Google Scholar] [CrossRef] [PubMed]
- Nyga, A.; Neves, J.B.; Stamati, K.; Loizidou, M.; Emberton, M.; Cheema, U. The next level of 3D tumour models: Immunocompetence. Drug Discov. Today 2016, 21, 1421–1428. [Google Scholar] [CrossRef]
- Gil Del Alcazar, C.R.; Huh, S.J.; Ekram, M.B.; Trinh, A.; Liu, L.L.; Beca, F.; Zi, X.; Kwak, M.; Bergholtz, H.; Su, Y.; et al. Immune Escape in Breast Cancer During In Situ to Invasive Carcinoma Transition. Cancer Discov. 2017, 7, 1098–1115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Varn, F.S.; Mullins, D.W.; Arias-Pulido, H.; Fiering, S.; Cheng, C. Adaptive immunity programmes in breast cancer. Immunology 2016, 150, 25–34. [Google Scholar] [CrossRef] [Green Version]
- Jiang, X.; Wang, J.; Deng, X.; Xiong, F.; Ge, J.; Xiang, B.; Wu, X.; Ma, J.; Zhou, M.; Li, X.; et al. Role of the tumor microenvironment in PD-L1/PD-1-mediated tumor immune escape. Mol. Cancer 2019, 18, 1–17. [Google Scholar] [CrossRef] [Green Version]
- Haanen, J.B. Converting Cold into Hot Tumors by Combining Immunotherapies. Cell 2017, 170, 1055–1056. [Google Scholar] [CrossRef] [Green Version]
- Ribas, A.; Dummer, R.; Puzanov, I.; VanderWalde, A.; Andtbacka, R.H.I.; Michielin, O.; Olszanski, A.J.; Malvehy, J.; Cebon, J.; Fernandez, E.; et al. Oncolytic Virotherapy Promotes Intratumoral T Cell Infiltration and Improves Anti-PD-1 Immunotherapy. Cell 2017, 170, 1109–1119.e10. [Google Scholar] [CrossRef] [Green Version]
- Maolake, A.; Izumi, K.; Shigehara, K.; Natsagdorj, A.; Iwamoto, H.; Kadomoto, S.; Takezawa, Y.; Machioka, K.; Narimoto, K.; Namiki, M.; et al. Tumor-associated macrophages promote prostate cancer migration through activation of the CCL22-CCR4 axis. Oncotarget 2016, 8, 9739–9751. [Google Scholar] [CrossRef] [Green Version]
- Tevis, K.M.; Cecchi, R.J.; Colson, Y.L.; Grinstaff, M.W. Mimicking the tumor microenvironment to regulate macrophage phenotype and assessing chemotherapeutic efficacy in embedded cancer cell/macrophage spheroid models. Acta Biomater. 2017, 50, 271–279. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dwyer, A.; Ellies, L.G.; Holme, A.L.; Pixley, F.J. A three-dimensional co-culture system to investigate macrophage-dependent tumor cell invasion. J. Biol. Methods 2016, 3, e49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, S.W.L.; Seager, R.J.; Litvak, F.; Spill, F.; Sieow, J.L.; Leong, P.H.; Kumar, D.; Tan, A.S.M.; Wong, S.C.; Adriani, G.; et al. Integrated in silico and 3D in vitro model of macrophage migration in response to physical and chemical factors in the tumor microenvironment. Integr. Biol. 2020, 12, 90–108. [Google Scholar] [CrossRef]
- Ayuso, J.M.; Virumbrales-Muñoz, M.; Lacueva, A.; Lanuza, P.M.; Checa-Chavarria, E.; Botella, P.; Fernandez, E.P.-A.; Doblare, M.; Allison, S.; Phillips, R.M.; et al. Development and characterization of a microfluidic model of the tumour microenvironment. Sci. Rep. 2016, 6, 36086. [Google Scholar] [CrossRef]
- Schnalzger, T.E.; De Groot, M.H.; Zhang, C.; Mosa, M.H.; Michels, B.E.; Röder, J.; Darvishi, T.; Wels, W.S.; Farin, H.F. 3D model for CAR -mediated cytotoxicity using patient-derived colorectal cancer organoids. EMBO J. 2019, 38, e100928. [Google Scholar] [CrossRef]
- Sherman, H.; Gitschier, H.J.; Rossi, A.E. A Novel Three-Dimensional Immune Oncology Model for High-Throughput Testing of Tumoricidal Activity. Front. Immunol. 2018, 9, 857. [Google Scholar] [CrossRef] [PubMed]
- Giannattasio, A.; Weil, S.; Kloess, S.; Ansari, N.; Stelzer, E.H.K.; Cerwenka, A.; Steinle, A.; Koehl, U.; Koch, J. Cytotoxicity and infiltration of human NK cells in in vivo-like tumor spheroids. BMC Cancer 2015, 15, 351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Christakou, A.E.; Ohlin, M.; Önfelt, B.; Wiklund, M. Ultrasonic three-dimensional on-chip cell culture for dynamic studies of tumor immune surveillance by natural killer cells. Lab Chip 2015, 15, 3222–3231. [Google Scholar] [CrossRef] [Green Version]
- Gottfried, E.; Kunz-Schughart, L.; Ebner, S.; Mueller-Klieser, W.; Hoves, S.; Andreesen, R.; Mackensen, A.; Kreutz, M. Tumor-derived lactic acid modulates dendritic cell activation and antigen expression. Blood 2006, 107, 2013–2021. [Google Scholar] [CrossRef] [PubMed]
- Parlato, S.; de Ninno, A.; Molfetta, R.; Toschi, E.; Salerno, D.; Mencattini, A.; Romagnoli, G.; Fragale, A.; Roccazzello, L.; Buoncervello, M.; et al. 3D Microfluidic model for evaluating immunotherapy efficacy by tracking dendritic cell behaviour toward tumor cells. Sci. Rep. 2017, 7, 1093. [Google Scholar] [CrossRef]
- Dangles-Marie, V.; Richon, S.; El Behi, M.; Echchakir, H.; Dorothée, G.; Thiery, J.; Validire, P.; Vergnon, I.; Menez, J.; Ladjimi, M.; et al. A Three-Dimensional Tumor Cell Defect in Activating Autologous CTLs Is As-sociated with Inefficient Antigen Presentation Correlated with Heat Shock Protein-70 down-Regulation. Cancer Res. 2003, 63, 3682–3687. [Google Scholar] [PubMed]
- Ghosh, S.; Rosenthal, R.; Zajac, P.; Weber, W.P.; Oertli, D.; Heberer, M.; Martin, I.; Spagnoli, G.C.; Reschner, A. Culture of Melanoma Cells in 3-Dimensional Architectures Results in Impaired Immunorecognition by Cytotoxic T Lymphocytes Specific for Melan-A/MART-1 Tumor-Associated Antigen. Ann. Surg. 2005, 242, 851–858. [Google Scholar] [CrossRef] [PubMed]
- Alonso-Nocelo, M.; Abuín, C.; López-López, R.; De La Fuente, M.; Freire, M.D.L.F. Development and characterization of a three-dimensional co-culture model of tumor T cell infiltration. Biofabrication 2016, 8, 025002. [Google Scholar] [CrossRef] [PubMed]
- Pavesi, A.; Tan, A.T.; Koh, S.; Chia, A.; Colombo, M.; Antonecchia, E.; Miccolis, C.; Ceccarello, E.; Adriani, G.; Raimondi, M.T.; et al. A 3D microfluidic model for preclinical evaluation of TCR-engineered T cells against solid tumors. JCI Insight 2017, 2, e89762. [Google Scholar] [CrossRef]
- Dijkstra, K.K.; Cattaneo, C.M.; Weeber, F.; Chalabi, M.; Van De Haar, J.; Fanchi, L.F.; Slagter, M.; Van Der Velden, D.L.; Kaing, S.; Kelderman, S.; et al. Generation of Tumor-Reactive T Cells by Co-culture of Peripheral Blood Lymphocytes and Tumor Organoids. Cell 2018, 174, 1586–1598.e12. [Google Scholar] [CrossRef] [Green Version]
- Bar-Ephraim, Y.; Kretzschmar, K.; Asra, P.; de Jongh, E.; Boonekamp, K.; Drost, J.; van Gorp, J.; Pronk, A.; Smakman, N.; Gan, I.; et al. Modelling Cancer Immunomodulation Using Epithelial Organoid Cultures. bioRxiv. 2018. [Google Scholar] [CrossRef]
- Wallstabe, L.; Göttlich, C.; Nelke, L.C.; Kühnemundt, J.; Schwarz, T.; Nerreter, T.; Einsele, H.; Walles, H.; Dandekar, G.; Nietzer, S.L.; et al. ROR1-CAR T cells are effective against lung and breast cancer in advanced microphysiologic 3D tumor models. JCI Insight 2019, 4, e126345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Doumba, P.P.; Nikolopoulou, M.; Gomatos, I.P.; Konstadoulakis, M.M.; Koskinas, J. Co-culture of primary human tumor hepatocytes from patients with hepatocellular carcinoma with autologous peripheral blood mononuclear cells: Study of their in vitro immunological interactions. BMC Gastroenterol. 2013, 13, 17. [Google Scholar] [CrossRef] [Green Version]
- Florczyk, S.; Liu, G.; Kievit, F.; Lewis, A.M.; Wu, J.D.; Zhang, M. 3D Porous Chitosan-Alginate Scaffolds: A New Matrix for Studying Prostate Cancer Cell-Lymphocyte Interactions In Vitro. Adv. Healthc. Mater. 2012, 1, 590–599. [Google Scholar] [CrossRef] [Green Version]
- Hirschhaeuser, F.; Walenta, S.; Mueller-Klieser, W. Efficacy of catumaxomab in tumor spheroid killing is mediated by its trifunctional mode of action. Cancer Immunol. Immunother. 2010, 59, 1675–1684. [Google Scholar] [CrossRef]
- Hoffmann, O.I.; 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] [PubMed] [Green Version]
- Courau, T.; Bonnereau, J.; Chicoteau, J.; Bottois, H.; Remark, R.; Assante Miranda, L.; Toubert, A.; Blery, 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]
- Augustine, T.N.; Dix-Peek, T.; Duarte, R.; Candy, G.P. Establishment of a heterotypic 3D culture system to evaluate the interaction of TREG lymphocytes and NK cells with breast cancer. J. Immunol. Methods 2015, 426, 1–13. [Google Scholar] [CrossRef] [PubMed]
- Feder-Mengus, C.; Ghosh, S.; Weber, W.P.; Wyler, S.; Zajac, P.; Terracciano, L.; Oertli, D.; Heberer, M.; Martin, I.; Spagnoli, G.C.; et al. Multiple mechanisms underlie defective recognition of melanoma cells cultured in three-dimensional architectures by antigen-specific cytotoxic T lymphocytes. Br. J. Cancer 2007, 96, 1072–1082. [Google Scholar] [CrossRef] [Green Version]
- Cohen, I.J.; Blasberg, R. Impact of the Tumor Microenvironment on Tumor-Infiltrating Lymphocytes: Focus on Breast Cancer. Breast Cancer Basic Clin. Res. 2017, 11, 1178223417731565. [Google Scholar] [CrossRef] [Green Version]
- Ohashi, T.; Aoki, M.; Tomita, H.; Akazawa, T.; Sato, K.; Kuze, B.; Mizuta, K.; Hara, A.; Nagaoka, H.; Inoue, N.; et al. M2-like macrophage polarization in high lactic acid-producing head and neck cancer. Cancer Sci. 2017, 108, 1128–1134. [Google Scholar] [CrossRef] [Green Version]
- Gehring, A.; Xue, S.-A.; Ho, Z.Z.; Teoh, D.; Ruedl, C.; Chia, A.; Koh, S.; Lim, S.G.; Maini, M.; Stauss, H.; et al. Engineering virus-specific T cells that target HBV infected hepatocytes and hepatocellular carcinoma cell lines. J. Hepatol. 2011, 55, 103–110. [Google Scholar] [CrossRef]
- Lee, S.W.L.; Adriani, G.; Ceccarello, E.; Pavesi, A.; Tan, A.T.; Bertoletti, A.; Kamm, R.D.; Wong, S.C. Characterizing the Role of Monocytes in T Cell Cancer Immunotherapy Using a 3D Microfluidic Model. Front. Immunol. 2018, 9, 416. [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]
- Nozaki, K.; Mochizuki, W.; Matsumoto, Y.; Matsumoto, T.; Fukuda, M.; Mizutani, T.; Watanabe, M.; Nakamura, T. Co-culture with intestinal epithelial organoids allows efficient expansion and motility analysis of intraepithelial lymphocytes. J. Gastroenterol. 2016, 51, 206–213. [Google Scholar] [CrossRef] [Green Version]
- Rogoz, A.; Reis, B.S.; Karssemeijer, R.A.; Mucida, D. A 3-D enteroid-based model to study T-cell and epithelial cell interaction. J. Immunol. Methods 2015, 421, 89–95. [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, 79-80, 145–154. [Google Scholar] [CrossRef]
- Kang, J.-C.; Chen, J.-S.; Lee, C.-H.; Chang, J.-J.; Shieh, Y.-S. Intratumoral macrophage counts correlate with tumor progression in colorectal cancer. J. Surg. Oncol. 2010, 102, 242–248. [Google Scholar] [CrossRef] [PubMed]
- Erreni, M.; Mantovani, A.; Allavena, P. Tumor-associated Macrophages (TAM) and Inflammation in Colorectal Cancer. Cancer Microenviron. 2011, 4, 141–154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pinto, M.L.; Rios, E.; Durães, C.; Ribeiro, R.; Machado, J.C.; Mantovani, A.; Barbosa, M.A.; Carneiro, F.; Oliveira, M.J. The Two Faces of Tumor-Associated Macrophages and Their Clinical Significance in Colorectal Cancer. Front. Immunol. 2019, 10, 1875. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cortese, N.; Carriero, R.; Laghi, L.; Mantovani, A.; Marchesi, F. Prognostic significance of tumor-associated macrophages: Past, present and future. Semin. Immunol. 2020, 48, 101408. [Google Scholar] [CrossRef] [PubMed]
- Azizi, E.; Carr, A.J.; Plitas, G.; Cornish, A.E.; Konopacki, C.; Prabhakaran, S.; Nainys, J.; Wu, K.; Kiseliovas, V.; Setty, M.; et al. Single-Cell Map of Diverse Immune Phenotypes in the Breast Tumor Microenvironment. Cell 2018, 174, 1293–1308.e36. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Q.; He, Y.; Luo, N.; Patel, S.J.; Han, Y.; Gao, R.; Modak, M.; Carotta, S.; Haslinger, C.; Kind, D.; et al. Landscape and Dynamics of Single Immune Cells in Hepatocellular Carcinoma. Cell 2019, 179, 829–845.e20. [Google Scholar] [CrossRef]
- Johnson, E.E.; Sandgren, A.; Cherayil, B.J.; Murray, M.; Wessling-Resnick, M. Role of Ferroportin in Macrophage-Mediated Immunity. Infect. Immun. 2010, 78, 5099–5106. [Google Scholar] [CrossRef] [Green Version]
- Liguori, M.; Digifico, E.; Vacchini, A.; Avigni, R.; Colombo, F.S.; Borroni, E.M.; Farina, F.M.; Milanesi, S.; Castagna, A.; Mannarino, L.; et al. The soluble glycoprotein NMB (GPNMB) produced by macrophages induces cancer stemness and metastasis via CD44 and IL-33. Cell. Mol. Immunol. 2021, 18, 711–722. [Google Scholar] [CrossRef] [PubMed]
- Tedesco, S.; De Majo, F.; Kim, J.; Trenti, A.; Trevisi, L.; Fadini, G.P.; Bolego, C.; Zandstra, P.W.; Cignarella, A.; Vitiello, L. Convenience versus Biological Significance: Are PMA-Differentiated THP-1 Cells a Reliable Substitute for Blood-Derived Macrophages When Studying in Vitro Polarization? Front. Pharmacol. 2018, 9, 71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bosshart, H.; Heinzelmann, M. THP-1 cells as a model for human monocytes. Ann. Transl. Med. 2016, 4, 438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Taciak, B.; Białasek, M.; Braniewska, A.; Sas, Z.; Sawicka, P.; Kiraga, Ł.; Rygiel, T.; Król, M. Evaluation of phenotypic and functional stability of RAW 264.7 cell line through serial passages. PLoS ONE 2018, 13, e0198943. [Google Scholar] [CrossRef]
- Shiratori, H.; Feinweber, C.; Luckhardt, S.; Linke, B.; Resch, E.; Geisslinger, G.; Weigert, A.; Parnham, M.J. THP-1 and human peripheral blood mononuclear cell-derived macrophages differ in their capacity to polarize in vitro. Mol. Immunol. 2017, 88, 58–68. [Google Scholar] [CrossRef]
- Marshall, L.A.; Marubayashi, S.; Jorapur, A.; Jacobson, S.; Zibinsky, M.; Robles, O.; Hu, D.X.; Jackson, J.J.; Pookot, D.; Sanchez, J.; et al. Tumors establish resistance to immunotherapy by regulating Treg recruitment via CCR4. J. Immunother. Cancer 2020, 8, e000764. [Google Scholar] [CrossRef]
- Noel, G.; Baetz, N.W.; Staab, J.F.; Donowitz, M.; Kovbasnjuk, O.; Pasetti, M.F.; Zachos, N.C. A primary human macrophage-enteroid co-culture model to investigate mucosal gut physiology and host-pathogen interactions. Sci. Rep. 2017, 7, 45270. [Google Scholar] [CrossRef] [Green Version]
- Gregory, A.D.; Houghton, A.M. Tumor-Associated Neutrophils: New Targets for Cancer Therapy. Cancer Res. 2011, 71, 2411–2416. [Google Scholar] [CrossRef] [Green Version]
- Giese, M.A.; Hind, L.E.; Huttenlocher, A. Neutrophil plasticity in the tumor microenvironment. Blood 2019, 133, 2159–2167. [Google Scholar] [CrossRef]
- Shaul, M.E.; Fridlender, Z.G. Tumour-associated neutrophils in patients with cancer. Nat. Rev. Clin. Oncol. 2019, 16, 601–620. [Google Scholar] [CrossRef]
- Jensen, H.K.; Donskov, F.; Marcussen, N.; Nordsmark, M.; Lundbeck, F.; von der Maase, H. Presence of Intratumoral Neutrophils Is an Independent Prognostic Factor in Localized Renal Cell Carcinoma. J. Clin. Oncol. 2009, 27, 4709–4717. [Google Scholar] [CrossRef] [PubMed]
- Eruslanov, E.B.; Bhojnagarwala, P.S.; Quatromoni, J.G.; Stephen, T.L.; Ranganathan, A.; Deshpande, C.; Akimova, T.; Vachani, A.; Litzky, L.; Hancock, W.W.; et al. Tumor-associated neutrophils stimulate T cell responses in early-stage human lung cancer. J. Clin. Investig. 2014, 124, 5466–5480. [Google Scholar] [CrossRef] [Green Version]
- Huh, S.J.; Liang, S.; Sharma, A.; Dong, C.; Robertson, G.P. Transiently Entrapped Circulating Tumor Cells Interact with Neutrophils to Facilitate Lung Metastasis Development. Cancer Res. 2010, 70, 6071–6082. [Google Scholar] [CrossRef] [Green Version]
- Granot, Z.; Henke, E.; Comen, E.A.; King, T.A.; Norton, L.; Benezra, R. Tumor entrained neutrophils inhibit seeding in the premetastatic lung. Cancer Cell 2011, 20, 300–314. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mouchemore, K.; Anderson, R.; Hamilton, J.A. Neutrophils, G-CSF and their contribution to breast cancer metastasis. FEBS J. 2018, 285, 665–679. [Google Scholar] [CrossRef] [Green Version]
- Li, P.; Lu, M.; Shi, J.; Hua, L.; Gong, Z.; Li, Q.; Shultz, L.D.; Ren, G. Dual roles of neutrophils in metastatic colonization are governed by the host NK cell status. Nat. Commun. 2020, 11, 4387. [Google Scholar] [CrossRef] [PubMed]
- Sun, B.; Qin, W.; Song, M.; Liu, L.; Yu, Y.; Qi, X.; Sun, H. Neutrophil Suppresses Tumor Cell Proliferation via Fas /Fas Ligand Pathway Mediated Cell Cycle Arrested. Int. J. Biol. Sci. 2018, 14, 2103–2113. [Google Scholar] [CrossRef] [PubMed]
- Gonzalez, M.G.; Cichon, I.; Scislowska-Czarnecka, A.; Kolaczkowska, E. Challenges in 3D culturing of neutrophils: Assessment of cell viability. J. Immunol. Methods 2018, 457, 73–77. [Google Scholar] [CrossRef]
- McMINN, P.H.; Hind, L.E.; Huttenlocher, A.; Beebe, D.J. Neutrophil trafficking on-a-chip: An in vitro, organotypic model for investigating neutrophil priming, extravasation, and migration with spatiotemporal control. Lab Chip 2019, 19, 3697–3705. [Google Scholar] [CrossRef] [PubMed]
- Heydarian, M.; Schweinlin, M.; Schwarz, T.; Rawal, R.; Walles, H.; Metzger, M.; Rudel, T.; Kozjak-Pavlovic, V. Triple co-culture and perfusion bioreactor for studying the interaction between Neisseria gonorrhoeae and neutrophils: A novel 3D tissue model for bacterial infection and immunity. J. Tissue Eng. 2021, 12, 2041731420988802. [Google Scholar] [CrossRef] [PubMed]
- Gardner, A.; Ruffell, B. Dendritic Cells and Cancer Immunity. Trends Immunol. 2016, 37, 855–865. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fong, L.; Engleman, E.G. Dendritic Cells in Cancer Immunotherapy. Annu. Rev. Immunol. 2000, 18, 245–273. [Google Scholar] [CrossRef]
- Curiel, T.J.; Cheng, P.; Mottram, P.; Alvarez, X.; Moons, L.; Evdemon-Hogan, M.; Wei, S.; Zou, L.; Kryczek, I.; Hoyle, G.; et al. Dendritic Cell Subsets Differentially Regulate Angiogenesis in Human Ovarian Cancer. Cancer Res. 2004, 64, 5535–5538. [Google Scholar] [CrossRef] [Green Version]
- Cheever, M.A.; Higano, C.S. PROVENGE (Sipuleucel-T) in Prostate Cancer: The First FDA-Approved Therapeutic Cancer Vaccine. Clin. Cancer Res. 2011, 17, 3520–3526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bol, K.F.; Schreibelt, G.; Gerritsen, W.R.; de Vries, J.; Figdor, C.G. Dendritic Cell–Based Immunotherapy: State of the Art and Beyond. Clin. Cancer Res. 2016, 22, 1897–1906. [Google Scholar] [CrossRef] [Green Version]
- Constantino, J.; Gomes, C.; Falcão, A.; Cruz, M.T.; Neves, B.M. Antitumor dendritic cell–based vaccines: Lessons from 20 years of clinical trials and future perspectives. Transl. Res. 2016, 168, 74–95. [Google Scholar] [CrossRef]
- Sabado, R.L.; Balan, S.; Bhardwaj, N. Dendritic cell-based immunotherapy. Cell Res. 2017, 27, 74–95. [Google Scholar] [CrossRef] [Green Version]
- Steponavicius-Cruz, K.; Freitas, V.M.; Barbuto, J.A.M. Dendritic Cells and T Lymphocytes Interactions in a Novel 3D System. Procedia Eng. 2013, 59, 166–173. [Google Scholar] [CrossRef] [Green Version]
- Shimasaki, N.; Jain, A.; Campana, D. NK cells for cancer immunotherapy. Nat. Rev. Drug Discov. 2020, 19, 200–218. [Google Scholar] [CrossRef]
- Guillerey, C.; Huntington, N.D.; Smyth, M. Targeting natural killer cells in cancer immunotherapy. Nat. Immunol. 2016, 17, 1025–1036. [Google Scholar] [CrossRef]
- Ljunggren, H.-G.; Malmberg, K.-J. Prospects for the use of NK cells in immunotherapy of human cancer. Nat. Rev. Immunol. 2007, 7, 329–339. [Google Scholar] [CrossRef]
- Waldhauer, I.; Steinle, A. NK cells and cancer immunosurveillance. Oncogene 2008, 27, 5932–5943. [Google Scholar] [CrossRef] [Green Version]
- Zhang, S.; Liu, W.; Hu, B.; Wang, P.; Lv, X.; Chen, S.; Shao, Z. Prognostic Significance of Tumor-Infiltrating Natural Killer Cells in Solid Tumors: A Systematic Review and Meta-Analysis. Front. Immunol. 2020, 11, 1242. [Google Scholar] [CrossRef]
- Takanami, I.; Takeuchi, K.; Giga, M. The prognostic value of natural killer cell infiltration in resected pulmonary adenocarcinoma. J. Thorac. Cardiovasc. Surg. 2001, 121, 1058–1063. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, Y.; Feng, Y.; Mao, Q.; Ma, P.; Liu, J.; Lu, W.; Liu, Y.; Chen, X.; Hu, Y.; Xue, W. Diagnostic and prognostic value of the peripheral natural killer cell levels in gastric cancer. Exp. Ther. Med. 2020, 20, 3816–3822. [Google Scholar] [CrossRef]
- Tang, Y.-P.; Xie, M.-Z.; Li, K.-Z.; Li, J.-L.; Cai, Z.-M.; Hu, B.-L. Prognostic value of peripheral blood natural killer cells in colorectal cancer. BMC Gastroenterol. 2020, 20, 31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Osswald, A.; Hedrich, V.; Sommergruber, W. 3D-3 Tumor Models in Drug Discovery for Analysis of Immune Cell Infiltration. In Methods in Molecular Biology; Springer Science and Business Media LLC: Berlin, Germany, 2019. [Google Scholar]
- Silva-Santos, B.; Serre, K.; Norell, H. γδ T cells in cancer. Nat. Rev. Immunol. 2015, 15, 683–691. [Google Scholar] [CrossRef] [PubMed]
- Yazdanifar, M.; Barbarito, G.; Bertaina, A.; Airoldi, I. γδ T Cells: The Ideal Tool for Cancer Immunotherapy. Cells 2020, 9, 1305. [Google Scholar] [CrossRef] [PubMed]
- Rossi, C.; Gravelle, P.; Decaup, E.; Bordenave, J.; Poupot, M.; Tosolini, M.; Franchini, D.M.; Laurent, C.; Morin, R.; Lagarde, J.-M.; et al. Boosting γδ T cell-mediated antibody-dependent cellular cytotoxicity by PD-1 blockade in follicular lymphoma. OncoImmunology 2019, 8, 1554175. [Google Scholar] [CrossRef] [PubMed]
- Bronte, V.; Brandau, S.; Chen, S.-H.; Colombo, M.P.; Frey, A.B.; Greten, T.F.; Mandruzzato, S.; Murray, P.J.; Ochoa, A.; Ostrand-Rosenberg, S.; et al. Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat. Commun. 2016, 7, 12150. [Google Scholar] [CrossRef] [Green Version]
- Husain, Z.; Huang, Y.; Seth, P.; Sukhatme, V.P. Tumor-Derived Lactate Modifies Antitumor Immune Response: Effect on Myeloid-Derived Suppressor Cells and NK Cells. J. Immunol. 2013, 191, 1486–1495. [Google Scholar] [CrossRef] [PubMed]
- Solito, S.; Falisi, E.; Diaz-Montero, C.M.; Doni, A.; Pinton, L.; Rosato, A.; Francescato, S.; Basso, G.; Zanovello, P.; Onicescu, G.; et al. A human promyelocytic-like population is responsible for the immune suppression mediated by myeloid-derived suppressor cells. Blood 2011, 118, 2254–2265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Safarzadeh, E.; Hashemzadeh, S.; Duijf, P.H.; Mansoori, B.; Khaze, V.; Mohammadi, A.; Kazemi, T.; Yousefi, M.; Asadi, M.; Mohammadi, H.; et al. Circulating myeloid-derived suppressor cells: An independent prognostic factor in patients with breast cancer. J. Cell. Physiol. 2019, 234, 3515–3525. [Google Scholar] [CrossRef]
- Jung, Y.; Kim, J.K.; Shiozawa, Y.; Wang, J.; Mishra, A.; Joseph, J.; Berry, J.E.; Mcgee, S.; Lee, E.; Sun, H.; et al. Recruitment of Mesenchymal Stem Cells Into Prostate Tu-mors Promotes Metastasis. Nat. Commun. 2013, 4, 1795. [Google Scholar] [CrossRef] [Green Version]
- Poggi, A.; Varesano, S.; Zocchi, M.R. How to Hit Mesenchymal Stromal Cells and Make the Tumor Microenvironment Immunostimulant Rather Than Immunosuppressive. Front. Immunol. 2018, 9, 262. [Google Scholar] [CrossRef] [Green Version]
- Zhu, W.; Holmes, B.; Glazer, R.I.; Zhang, L.G. 3D printed nanocomposite matrix for the study of breast cancer bone metastasis. Nanomed. Nanotechnol. Biol. Med. 2016, 12, 69–79. [Google Scholar] [CrossRef]
- Zhou, X.; Zhu, W.; Nowicki, M.; Miao, S.; Cui, H.; Holmes, B.; Glazer, R.I.; Zhang, L.G. 3D Bioprinting a Cell-Laden Bone Matrix for Breast Cancer Metastasis Study. ACS Appl. Mater. Interfaces 2016, 8, 30017–30026. [Google Scholar] [CrossRef]
- Bersini, S.; Jeon, J.; Dubini, G.; Arrigoni, C.; Chung, S.; Charest, J.L.; Moretti, M.; Kamm, R.D. A microfluidic 3D in vitro model for specificity of breast cancer metastasis to bone. Biomaterials 2014, 35, 2454–2461. [Google Scholar] [CrossRef]
- Mosaad, E.; Chambers, K.; Futrega, K.; Clements, J.; Doran, M.R. Using high throughput microtissue culture to study the difference in prostate cancer cell behavior and drug response in 2D and 3D co-cultures. BMC Cancer 2018, 18, 592. [Google Scholar] [CrossRef]
- Liu, C.; Liu, Y.; Xu, X.-X.; Guo, X.; Sun, G.-W.; Ma, X.-J. Mesenchymal stem cells enhance the metastasis of 3D-cultured hepatocellular carcinoma cells. BMC Cancer 2016, 16, 566. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chao, K.-C.; Yang, H.-T.; Chen, M.-W. Human umbilical cord mesenchymal stem cells suppress breast cancer tumourigenesis through direct cell-cell contact and internalization. J. Cell. Mol. Med. 2012, 16, 1803–1815. [Google Scholar] [CrossRef] [PubMed]
- Rossignoli, F.; Spano, C.; Grisendi, G.; Foppiani, E.M.; Golinelli, G.; Mastrolia, I.; Bestagno, M.; Candini, O.; Petrachi, T.; Recchia, A.; et al. MSC-Delivered Soluble TRAIL and Paclitaxel as Novel Combinatory Treatment for Pancreatic Adenocarcinoma. Theranostics 2019, 9, 436–448. [Google Scholar] [CrossRef]
- Silva, M.; Monteiro, G.A.; Fialho, A.; Bernardes, N.; Da Silva, C.L. Conditioned Medium From Azurin-Expressing Human Mesenchymal Stromal Cells Demonstrates Antitumor Activity Against Breast and Lung Cancer Cell Lines. Front. Cell Dev. Biol. 2020, 8, 471. [Google Scholar] [CrossRef] [PubMed]
- Ham, J.; Lever, L.; Fox, M.; Reagan, M.R. In Vitro 3D Cultures to Reproduce the Bone Marrow Niche. JBMR Plus 2019, 3, e10228. [Google Scholar] [CrossRef] [Green Version]
- Görgün, G.; Calabrese, E.; Soydan, E.; Hideshima, T.; Perrone, G.; Bandi, M.; Cirstea, D.; Santo, L.; Hu, Y.; Tai, Y.-T.; et al. Immunomodulatory effects of lenalidomide and pomalidomide on interaction of tumor and bone marrow accessory cells in multiple myeloma. Blood 2010, 116, 3227–3237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Purroy, N.; Abrisqueta, P.; Carabia, J.; Carpio, C.; Palacio, C.; Bosch, F.; Crespo, M. Co-culture of primary CLL cells with bone marrow mesenchymal cells, CD40 ligand and CpG ODN promotes proliferation of chemoresistant CLL cells phenotypically comparable to those proliferating in vivo. Oncotarget 2014, 6, 7632–7643. [Google Scholar] [CrossRef] [Green Version]
- Raic, A.; Naolou, T.; Mohra, A.; Chatterjee, C.; Lee-Thedieck, C. 3D models of the bone marrow in health and disease: Yesterday, today, and tomorrow. MRS Commun. 2019, 9, 37–52. [Google Scholar] [CrossRef]
- de la Puente, P.; Muz, B.; Gilson, R.; Azab, F.; Luderer, M.; King, J.; Achilefu, S.; Vij, R.; Azab, A.K. 3D tissue-engineered bone marrow as a novel model to study pathophysiology and drug resistance in multiple myeloma. Biomaterials 2015, 73, 70–84. [Google Scholar] [CrossRef] [Green Version]
- Aljitawi, O.S.; Li, D.; Xiao, Y.; Zhang, D.; Ramachandran, K.; Stehno-Bittel, L.; Van Veldhuizen, P.; Lin, T.; Kambhampati, S.; Garimella, R. A novel three-dimensional stromal-based model forin vitrochemotherapy sensitivity testing of leukemia cells. Leuk. Lymphoma 2014, 55, 378–391. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shen, Z.-H.; Zeng, D.-F.; Wang, X.-Y.; Ma, Y.-Y.; Zhang, X.; Kong, P.-Y. Targeting of the leukemia microenvironment by c(RGDfV) overcomes the resistance to chemotherapy in acute myeloid leukemia in biomimetic polystyrene scaffolds. Oncol. Lett. 2016, 12, 3278–3284. [Google Scholar] [CrossRef]
- Corrêa, L.H.; Corrêa, R.; Farinasso, C.M.; Dourado, L.P.D.S.; Magalhães, K.G. Adipocytes and Macrophages Interplay in the Orchestration of Tumor Microenvironment: New Implications in Cancer Progression. Front. Immunol. 2017, 8, 1129. [Google Scholar] [CrossRef] [PubMed]
- Spencer, M.; Yao-Borengasser, A.; Unal, R.; Rasouli, N.; Gurley, C.M.; Zhu, B.; Peterson, C.A.; Kern, P.A. Adipose tissue macrophages in insulin-resistant subjects are associated with collagen VI and fibrosis and demonstrate alternative activation. Am. J. Physiol. Metab. 2010, 299, E1016–E1027. [Google Scholar] [CrossRef]
- Yue, X.; Nguyen, T.D.; Zellmer, V.; Zhang, S.; Zorlutuna, P. Stromal cell-laden 3D hydrogel microwell arrays as tumor microenvironment model for studying stiffness dependent stromal cell-cancer interactions. Biomaterials 2018, 170, 37–48. [Google Scholar] [CrossRef] [PubMed]
- Cao, Y. Angiogenesis and Vascular Functions in Modulation of Obesity, Adipose Metabolism, and Insulin Sensitivity. Cell Metab. 2013, 18, 478–489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mason, B.N.; Starchenko, A.; Williams, R.; Bonassar, L.J.; Reinhart-King, C.A. Tuning three-dimensional collagen matrix stiffness independently of collagen concentration modulates endothelial cell behavior. Acta Biomater. 2013, 9, 4635–4644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Agarwal, P.; Wang, H.; Sun, M.; Xu, J.; Zhao, S.; Liu, Z.; Gooch, K.J.; Zhao, Y.; Lu, X.; He, X. Microfluidics Enabled Bottom-Up Engineering of 3D Vascularized Tumor for Drug Discovery. ACS Nano 2017, 11, 6691–6702. [Google Scholar] [CrossRef] [PubMed]
- Herroon, M.K.; Diedrich, J.D.; Podgorski, I. New 3D-Culture Approaches to Study Interactions of Bone Marrow Adipocytes with Metastatic Prostate Cancer Cells. Front. Endocrinol. 2016, 7, 84. [Google Scholar] [CrossRef] [Green Version]
- Devarasetty, M.; Skardal, A.; Cowdrick, K.; Marini, F.; Soker, S. Bioengineered Submucosal Organoids for In Vitro Modeling of Colorectal Cancer. Tissue Eng. Part A 2017, 23, 1026–1041. [Google Scholar] [CrossRef]
- Drifka, C.R.; Loeffler, A.G.; Mathewson, K.; Keikhosravi, A.; Eickhoff, J.C.; Liu, Y.; Weber, S.M.; Kao, W.J.; Eliceiri, K.W. Highly aligned stromal collagen is a negative prognostic factor following pancreatic ductal adenocarcinoma resection. Oncotarget 2016, 7, 76197–76213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pietras, K.; Östman, A. Hallmarks of cancer: Interactions with the tumor stroma. Exp. Cell Res. 2010, 316, 1324–1331. [Google Scholar] [CrossRef] [PubMed]
- McMillin, D.W.; Negri, J.M.; Mitsiades, C.S. The role of tumour–stromal interactions in modifying drug response: Challenges and opportunities. Nat. Rev. Drug Discov. 2013, 12, 217–228. [Google Scholar] [CrossRef]
- Mao, Y.; Keller, E.T.; Garfield, D.H.; Shen, K.; Wang, J. Stromal cells in tumor microenvironment and breast cancer. Cancer Metastasis Rev. 2013, 32, 303–315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Barnas, J.L.; Simpson-Abelson, M.R.; Yokota, S.J.; Kelleher, R.J.; Bankert, R.B. T Cells and Stromal Fibroblasts in Human Tumor Microenvironments Represent Potential Therapeutic Targets. Cancer Microenviron. 2010, 3, 29–47. [Google Scholar] [CrossRef] [Green Version]
- Tang, D.; Yuan, Z.; Xue, X.; Lu, Z.; Zhang, Y.; Wang, H.; Chen, M.; An, Y.; Wei, J.; Zhu, Y.; et al. High expression of Galectin-1 in pancreatic stellate cells plays a role in the development and maintenance of an immunosuppressive microenvironment in pancreatic cancer. Int. J. Cancer 2011, 130, 2337–2348. [Google Scholar] [CrossRef]
- Barnas, J.L.; Simpson-Abelson, M.R.; Brooks, S.P.; Kelleher, R.J.; Bankert, R.B.; Kelleher, R.J., Jr. Reciprocal Functional Modulation of the Activation of T Lymphocytes and Fibroblasts Derived from Human Solid Tumors. J. Immunol. 2010, 185, 2681–2692. [Google Scholar] [CrossRef] [PubMed]
- Buckanovich, R.J.; Facciabene, A.; Kim, S.; Benencia, F.; Sasaroli, D.; Balint, K.; Katsaros, D.; O’Brien-Jenkins, A.; Gimotty, P.A.; Coukos, G. Endothelin B receptor mediates the endothelial barrier to T cell homing to tumors and disables immune therapy. Nat. Med. 2008, 14, 28–36. [Google Scholar] [CrossRef]
- Griffioen, A.W.; Damen, C.A.; Martinotti, S.; Blijham, G.H.; Groenewegen, G. Endothelial intercellular adhesion molecule-1 expression is suppressed in human malignancies: The role of angiogenic factors. Cancer Res. 1996, 56, 1111–1117. [Google Scholar]
- Tang, N.; Gao, J.; Wang, S.; Ye, N.; Chong, Y.; Huang, Y.; Wang, J.; Li, B.; Yin, W.; Wang, D. Cancer-associated fibroblasts promote angiogenesis in gastric cancer through galectin-1 expression. Tumor Biol. 2016, 37, 1889–1899. [Google Scholar] [CrossRef]
- De Francesco, E.M.; Lappano, R.; Santolla, M.F.; Marsico, S.; Caruso, A.; Maggiolini, M. HIF-1α/GPER signaling mediates the expression of VEGF induced by hypoxia in breast cancer associated fibroblasts (CAFs). Breast Cancer Res. 2013, 15, R64. [Google Scholar] [CrossRef] [Green Version]
- Leung, C.S.; Yeung, T.-L.; Yip, K.-P.; Wong, K.-K.; Ho, S.Y.; Mangala, L.S.; Sood, A.K.; Lopez-Berestein, G.; Sheng, J.; Wong, S.T.; et al. Cancer-associated fibroblasts regulate endothelial adhesion protein LPP to promote ovarian cancer chemoresistance. J. Clin. Investig. 2017, 128, 589–606. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Esposito, I.; Menicagli, M.; Funel, N.; Bergmann, F.; Boggi, U.; Mosca, F.; Bevilacqua, G.; Campani, D. Inflammatory cells contribute to the generation of an angiogenic phenotype in pancreatic ductal adenocarcinoma. J. Clin. Pathol. 2004, 57, 630–636. [Google Scholar] [CrossRef] [Green Version]
- Shrimali, R.K.; Yu, Z.; Theoret, M.R.; Chinnasamy, D.; Restifo, N.P.; Rosenberg, S.A. Antiangiogenic Agents Can Increase Lymphocyte Infiltration into Tumor and Enhance the Effectiveness of Adoptive Immunotherapy of Cancer. Cancer Res. 2010, 70, 6171–6180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huang, Y.; Kim, B.Y.S.; Chan, C.K.; Hahn, S.M.; Weissman, I.L.; Jiang, W. Improving immune–vascular crosstalk for cancer immunotherapy. Nat. Rev. Immunol. 2018, 18, 195–203. [Google Scholar] [CrossRef]
- Gabrusiewicz, K.; Liu, D.; Cortes-Santiago, N.; Hossain, M.B.; Conrad, C.A.; Aldape, K.D.; Fuller, G.; Marini, F.C.; Alonso, M.; Idoate-Gastearena, M.-A.; et al. Anti-vascular endothelial growth factor therapy-induced glioma invasion is associated with accumulation of Tie2-expressing monocytes. Oncotarget 2014, 5, 2208–2220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peranzoni, E.; Lemoine, J.; Vimeux, L.; Feuillet, V.; Barrin, S.; Kantari-Mimoun, C.; Bercovici, N.; Guérin, M.; Biton, J.; Ouakrim, H.; et al. Macrophages impede CD8 T cells from reaching tumor cells and limit the efficacy of anti–PD-1 treatment. Proc. Natl. Acad. Sci. USA 2018, 115, E4041–E4050. [Google Scholar] [CrossRef] [Green Version]
- Butowski, N.; Colman, H.; De Groot, J.F.; Omuro, A.M.; Nayak, L.; Wen, P.Y.; Cloughesy, T.F.; Marimuthu, A.; Haidar, S.; Perry, A.; et al. Orally administered colony stimulating factor 1 receptor inhibitor PLX3397 in recurrent glioblastoma: An Ivy Foundation Early Phase Clinical Trials Consortium phase II study. Neuro-Oncology 2015, 18, 557–564. [Google Scholar] [CrossRef] [Green Version]
- Kumar, V.; Donthireddy, L.; Marvel, D.; Condamine, T.; Wang, F.; Lavilla-Alonso, S.; Hashimoto, A.; Vonteddu, P.; Behera, R.; Goins, M.A.; et al. Cancer-Associated Fibroblasts Neutralize the Anti-tumor Effect of CSF1 Receptor Blockade by Inducing PMN-MDSC Infiltration of Tumors. Cancer Cell 2017, 32, 654–668.e5. [Google Scholar] [CrossRef] [Green Version]
- Cannarile, M.A.; Weisser, M.; Jacob, W.; Jegg, A.-M.; Ries, C.H.; Rüttinger, D. Colony-stimulating factor 1 receptor (CSF1R) inhibitors in cancer therapy. J. Immunother. Cancer 2017, 5, 53. [Google Scholar] [CrossRef]
- Ehsan, S.M.; Welch-Reardon, K.M.; Waterman, M.L.; Hughes, C.C.W.; George, S.C. A three-dimensional in vitro model of tumor cell intravasation. Integr. Biol. 2014, 6, 603–610. [Google Scholar] [CrossRef]
- Pape, J.; Magdeldin, T.; Stamati, K.; Nyga, A.; Loizidou, M.; Emberton, M.; Cheema, U. Cancer-associated fibroblasts mediate cancer progression and remodel the tumouroid stroma. Br. J. Cancer 2020, 123, 1178–1190. [Google Scholar] [CrossRef] [PubMed]
- Lee, S.W.; Kwak, H.S.; Kang, M.-H.; Park, Y.-Y.; Jeong, G.S. Fibroblast-associated tumour microenvironment induces vascular structure-networked tumouroid. Sci. Rep. 2018, 8, 2365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Franchi-Mendes, T.; Lopes, N.; Brito, C. Heterotypic Tumor Spheroids in Agitation-Based Cultures: A Scaffold-Free Cell Model That Sustains Long-Term Survival of Endothelial Cells. Front. Bioeng. Biotechnol. 2021, 9, 447. [Google Scholar] [CrossRef] [PubMed]
- Liu, X.-Q.; Kiefl, R.; Roskopf, C.; Tian, F.; Huber, R.M. Interactions among Lung Cancer Cells, Fibroblasts, and Macrophages in 3D Co-Cultures and the Impact on MMP-1 and VEGF Expression. PLoS ONE 2016, 11, e0156268. [Google Scholar] [CrossRef]
- Linde, N.; Gutschalk, C.M.; Hoffmann, C.; Yilmaz, D.; Mueller, M.M. Integrating Macrophages into Organotypic Co-Cultures: A 3D In Vitro Model to Study Tumor-Associated Macrophages. PLoS ONE 2012, 7, e40058. [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]
- Teixeira, F.C.; Chaves, S.; Torres, A.L.; Barrias, C.C.; Bidarra, S.J. Engineering a Vascularized 3D Hybrid System to Model Tumor-Stroma Interactions in Breast Cancer. Front. Bioeng. Biotechnol. 2021, 9, 166. [Google Scholar] [CrossRef]
- Kwak, T.J.; Lee, E. In vitro modeling of solid tumor interactions with perfused blood vessels. Sci. Rep. 2020, 10, 20142. [Google Scholar] [CrossRef]
- Rausch, M.; Blanc, L.; Silva, O.D.S.; Dormond, O.; Griffioen, A.; Nowak-Sliwinska, P. Characterization of Renal Cell Carcinoma Heterotypic 3D Co-Cultures with Immune Cell Subsets. Cancers 2021, 13, 2551. [Google Scholar] [CrossRef]
- Qiao, S.; Zhao, Y.; Tian, H.; Manike, I.; Ma, L.; Yan, H.; Tian, W. 3D Co-cultured Endothelial Cells and Monocytes Promoted Cancer Stem Cells’ Stemness and Malignancy. ACS Appl. Bio Mater. 2021, 4, 441–450. [Google Scholar] [CrossRef]
- Campisi, M.; Sundararaman, S.K.; Shelton, S.E.; Knelson, E.H.; Mahadevan, N.R.; Yoshida, R.; Tani, T.; Ivanova, E.; Cañadas, I.; Osaki, T.; et al. Tumor-Derived cGAMP Regulates Activation of the Vasculature. Front. Immunol. 2020, 11, 2090. [Google Scholar] [CrossRef]
- Nashimoto, Y.; Okada, R.; Hanada, S.; Arima, Y.; Nishiyama, K.; Miura, T.; Yokokawa, R. Vascularized cancer on a chip: The effect of perfusion on growth and drug delivery of tumor spheroid. Biomaterials 2020, 229, 119547. [Google Scholar] [CrossRef]
- Kim, H.; Chung, H.; Kim, J.; Choi, D.; Shin, Y.; Kang, Y.G.; Kim, B.; Seo, S.; Chung, S.; Seok, S.H. Macrophages-Triggered Sequential Remodeling of Endothelium-Interstitial Matrix to Form Pre-Metastatic Niche in Microfluidic Tumor Microenvironment. Adv. Sci. 2019, 6, 1900195. [Google Scholar] [CrossRef] [PubMed]
- Cui, X.; Morales, R.-T.T.; Qian, W.; Wang, H.; Gagner, J.-P.; Dolgalev, I.; Placantonakis, D.; Zagzag, D.; Cimmino, L.; Snuderl, M.; et al. Hacking macrophage-associated immunosuppression for regulating glioblastoma angiogenesis. Biomaterials 2018, 161, 164–178. [Google Scholar] [CrossRef] [PubMed]
- Yamazoe, H.; Hagihara, Y.; Kobayashi, H. Multicomponent Coculture System of Cancer Cells and Two Types of Stromal Cells for In Vitro Evaluation of Anticancer Drugs. Tissue Eng. Part C Methods 2016, 22, 20–29. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Sobrino, A.; Phan, D.T.T.; Datta, R.; Wang, X.; Hachey, S.J.; Romero-López, M.; Gratton, E.; Lee, A.P.; George, S.C.; Hughes, C.C.W. 3D microtumors in vitro supported by perfused vascular networks. Sci. Rep. 2016, 6, 31589. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Magdeldin, T.; López-Dávila, V.; Pape, J.; Cameron, G.W.W.; Emberton, M.; Loizidou, M.; Cheema, U. Engineering a vascularised 3D in vitro model of cancer progression. Sci. Rep. 2017, 7, 44045. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aung, A.; Kumar, V.; Theprungsirikul, J.; Davey, S.K.; Varghese, S. An Engineered Tumor-on-a-Chip Device with Breast Cancer–Immune Cell Interactions for Assessing T-cell Recruitment. Cancer Res. 2020, 80, 263–275. [Google Scholar] [CrossRef] [PubMed]
- Nii, T.; Kuwahara, T.; Makino, K.; Tabata, P.Y. A Co-Culture System of Three-Dimensional Tumor-Associated Macrophages and Three-Dimensional Cancer-Associated Fibroblasts Combined with Biomolecule Release for Cancer Cell Migration. Tissue Eng. Part A 2020, 26, 1272–1282. [Google Scholar] [CrossRef]
- Rama-Esendagli, D.; Esendagli, G.; Yilmaz, G.; Guc, D. Spheroid formation and invasion capacity are differentially influenced by co-cultures of fibroblast and macrophage cells in breast cancer. Mol. Biol. Rep. 2014, 41, 2885–2892. [Google Scholar] [CrossRef] [PubMed]
- Kuen, J.; Darowski, D.; Kluge, T.; Majety, M. Pancreatic cancer cell/fibroblast co-culture induces M2 like macrophages that influence therapeutic response in a 3D model. PLoS ONE 2017, 12, e0182039. [Google Scholar] [CrossRef] [PubMed]
- Nguyen, M.; De Ninno, A.; Mencattini, A.; Mermet-Meillon, F.; Fornabaio, G.; Evans, S.S.; Cossutta, M.; Khira, Y.; Han, W.; Sirven, P.; et al. Dissecting Effects of Anti-cancer Drugs and Cancer-Associated Fibroblasts by On-Chip Reconstitution of Immunocompetent Tumor Microenvironments. Cell Rep. 2018, 25, 3884–3893.e3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Donovan, D.; Brown, N.; Bishop, E.; Lewis, C. Comparison of three in vitro human ‘angiogenesis’ assays with capillaries formed in vivo. Angiogenesis 2001, 4, 113–121. [Google Scholar] [CrossRef]
- Amann, A.; Zwierzina, M.; Koeck, S.; Gamerith, G.; Pechriggl, E.; Huber, J.M.; Lorenz, E.; Kelm, J.M.; Hilbe, W.; Zwierzina, H.; et al. Development of a 3D angiogenesis model to study tumour—Endothelial cell interactions and the effects of anti-angiogenic drugs. Sci. Rep. 2017, 7, 2963. [Google Scholar] [CrossRef] [Green Version]
- Lamichhane, S.P.; Arya, N.; Kohler, E.; Xiang, S.; Christensen, J.; Shastri, V.P. Recapitulating epithelial tumor microenvironment in vitro using three dimensional tri-culture of human epithelial, endothelial, and mesenchymal cells. BMC Cancer 2016, 16, 581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Weinberg, F.; Ramnath, N.; Nagrath, D. Reactive Oxygen Species in the Tumor Microenvironment: An Overview. Cancers 2019, 11, 1191. [Google Scholar] [CrossRef] [Green Version]
- Bray, L.J.; Binner, M.; Körner, Y.; Von Bonin, M.; Bornhäuser, M.; Werner, C. A three-dimensional ex vivo tri-culture model mimics cell-cell interactions between acute myeloid leukemia and the vascular niche. Haematologica 2017, 102, 1215–1226. [Google Scholar] [CrossRef] [Green Version]
- Bray, L.J.; Binner, M.; Holzheu, A.; Friedrichs, J.; Freudenberg, U.; Hutmacher, D.W.; Werner, C. Multi-parametric hydrogels support 3D in vitro bioengineered microenvironment models of tumour angiogenesis. Biomaterials 2015, 53, 609–620. [Google Scholar] [CrossRef] [Green Version]
- Rogers, M.; Sobolik, T.; Schaffer, D.K.; Samson, P.C.; Johnson, A.C.; Owens, P.; Codreanu, S.G.; Sherrod, S.D.; McLean, J.A.; Wikswo, J.P.; et al. Engineered microfluidic bioreactor for examining the three-dimensional breast tumor microenvironment. Biomicrofluidics 2018, 12, 034102. [Google Scholar] [CrossRef] [PubMed]
- Herrera-Perez, R.M.; Voytik-Harbin, S.L.; Sarkaria, J.N.; Pollok, K.E.; Fishel, M.; Rickus, J.L. Presence of stromal cells in a bioengineered tumor microenvironment alters glioblastoma migration and response to STAT3 inhibition. PLoS ONE 2018, 13, e0194183. [Google Scholar] [CrossRef]
- Kessenbrock, K.; Wang, C.-Y.; Werb, Z. Matrix metalloproteinases in stem cell regulation and cancer. Matrix Biol. 2015, 44-46, 184–190. [Google Scholar] [CrossRef] [PubMed]
- Carmeliet, P. VEGF as a Key Mediator of Angiogenesis in Cancer. Oncology 2005, 69, 4–10. [Google Scholar] [CrossRef] [PubMed]
- De Biasi, A.R.; Villena-Vargas, J.; Adusumilli, P.S. Cisplatin-Induced Antitumor Immunomodulation: A Review of Preclinical and Clinical Evidence. Clin. Cancer Res. 2014, 20, 5384–5391. [Google Scholar] [CrossRef] [Green Version]
- Laoui, D.; Van Overmeire, E.; De Baetselier, P.; Van Ginderachter, J.; Raes, G. Functional Relationship between Tumor-Associated Macrophages and Macrophage Colony-Stimulating Factor as Contributors to Cancer Progression. Front. Immunol. 2014, 5, 489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Baleia, P. 8th International conference on Tumor Microenvironment, Lisbon, Portugal June 10-14, 2018. Cancer Microenviron. 2018, 11 (Suppl. S1), 1–91. [Google Scholar] [CrossRef] [PubMed]
- Saraiva, D.P.; Matias, A.T.; Braga, S.; Jacinto, A.; Cabral, M.G. Establishment of a 3D Co-culture With MDA-MB-231 Breast Cancer Cell Line and Patient-Derived Immune Cells for Application in the Development of Immunotherapies. Front. Oncol. 2020, 10, 1543. [Google Scholar] [CrossRef]
- Koeck, S.; Kern, J.; Zwierzina, M.; Gamerith, G.; Lorenz, E.; Sopper, S.; Zwierzina, H.; Amann, A. The influence of stromal cells and tumor-microenvironment-derived cytokines and chemokines on CD3+CD8+ tumor infiltrating lymphocyte subpopulations. OncoImmunology 2017, 6, e1323617. [Google Scholar] [CrossRef] [Green Version]
- Herter, S.; Morra, L.; Schlenker, R.; Sulcova, J.; Fahrni, L.; Waldhauer, I.; Lehmann, S.; Reisländer, T.; Agarkova, I.; Kelm, J.M.; et al. A novel three-dimensional heterotypic spheroid model for the assessment of the activity of cancer immunotherapy agents. Cancer Immunol. Immunother. 2017, 66, 129–140. [Google Scholar] [CrossRef] [PubMed]
- Kuang, D.-M.; Zhao, Q.; Peng, C.; Xu, J.; Zhang, J.-P.; Wu, C.; Zheng, L. Activated monocytes in peritumoral stroma of hepatocellular carcinoma foster immune privilege and disease progression through PD-L1. J. Exp. Med. 2009, 206, 1327–1337. [Google Scholar] [CrossRef]
- Mpekris, F.; Voutouri, C.; Baish, J.W.; Duda, D.G.; Munn, L.L.; Stylianopoulos, T.; Jain, R.K. Combining microenvironment normalization strategies to improve cancer immunotherapy. Proc. Natl. Acad. Sci. USA 2020, 117, 3728–3737. [Google Scholar] [CrossRef] [Green Version]
- Fukumura, D.; Kloepper, J.; Amoozgar, Z.; Duda, D.G.; Jain, R.K. Enhancing cancer immunotherapy using antiangiogenics: Opportunities and challenges. Nat. Rev. Clin. Oncol. 2018, 15, 325–340. [Google Scholar] [CrossRef]
- Tang, M.; Xie, Q.; Gimple, R.C.; Zhong, Z.; Tam, T.; Tian, J.; Kidwell, R.L.; Wu, Q.; Prager, B.C.; Qiu, Z.; et al. Three-dimensional bioprinted glioblastoma microenvironments model cellular dependencies and immune interactions. Cell Res. 2020, 30, 833–853. [Google Scholar] [CrossRef]
- Neufeld, L.; Yeini, E.; Reisman, N.; Shtilerman, Y.; Ben-Shushan, D.; Pozzi, S.; Madi, A.; Tiram, G.; Eldar-Boock, A.; Ferber, S.; et al. Microengineered perfusable 3D-bioprinted glioblastoma model for in vivo mimicry of tumor microenvironment. Sci. Adv. 2021, 7, eabi9119. [Google Scholar] [CrossRef] [PubMed]
- Qiao, H.; Tang, T. Engineering 3D approaches to model the dynamic microenvironments of cancer bone metastasis. Bone Res. 2018, 6, 3. [Google Scholar] [CrossRef] [Green Version]
- 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] [Green Version]
- Carvalho, M.; Lima, D.; Reis, R.L.; Correlo, V.M.; Oliveira, J.M. Evaluating Biomaterial- and Microfluidic-Based 3D Tumor Models. Trends Biotechnol. 2015, 33, 667–678. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gu, L.; Mooney, D.J. Biomaterials and emerging anticancer therapeutics: Engineering the microenvironment. Nat. Rev. Cancer 2016, 16, 56–66. [Google Scholar] [CrossRef] [Green Version]
- Qiao, S.-P.; Zhao, Y.-F.; Li, C.-F.; Yin, Y.-B.; Meng, Q.-Y.; Lin, F.-H.; Liu, Y.; Hou, X.-L.; Guo, K.; Chen, X.-b.; et al. An alginate-based platform for cancer stem cell research. Acta Biomater. 2016, 37, 83–92. [Google Scholar] [CrossRef]
- Amorim, S.; Da Costa, D.S.; Pashkuleva, I.; Reis, C.A.; Reis, R.L.; Pires, R.A. 3D hydrogel mimics of the tumor microenvironment: The interplay among hyaluronic acid, stem cells and cancer cells. Biomater. Sci. 2021, 9, 252–260. [Google Scholar] [CrossRef]
- Egeblad, M.; Rasch, M.G.; Weaver, V.M. Dynamic interplay between the collagen scaffold and tumor evolution. Curr. Opin. Cell Biol. 2010, 22, 697–706. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Benton, G.; Kleinman, H.K.; George, J.; Arnaoutova, I. Multiple uses of basement membrane-like matrix (BME/Matrigel) in vitro and in vivo with cancer cells. Int. J. Cancer 2011, 128, 1751–1757. [Google Scholar] [CrossRef] [PubMed]
- Tsao, C.-T.; Kievit, F.M.; Wang, K.; Erickson, A.E.; Ellenbogen, R.G.; Zhang, M. Chitosan-Based Thermoreversible Hydrogel as an in Vitro Tumor Microenvironment for Testing Breast Cancer Therapies. Mol. Pharm. 2014, 11, 2134–2142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cavo, M.; Fato, M.M.; Peñuela, L.; Beltrame, F.; Raiteri, R.; Scaglione, S. Microenvironment complexity and matrix stiffness regulate breast cancer cell activity in a 3D in vitro model. Sci. Rep. 2016, 6, 35367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Paszek, M.J.; Zahir, N.; Johnson, K.R.; Lakins, J.N.; Rozenberg, G.I.; Gefen, A.; Reinhart-King, C.; Margulies, S.S.; Dembo, M.; Boettiger, D.; et al. Tensional homeostasis and the malignant phenotype. Cancer Cell 2005, 8, 241–254. [Google Scholar] [CrossRef] [Green Version]
- Rice, A.J.; Cortes, E.; Lachowski, D.; Cheung, B.C.H.; Karim, S.A.; Morton, J.; Hernández, A.D.R. Matrix stiffness induces epithelial–mesenchymal transition and promotes chemoresistance in pancreatic cancer cells. Oncogenesis 2017, 6, e352. [Google Scholar] [CrossRef] [Green Version]
- Hofer, M.; Lutolf, M.P. Engineering organoids. Nat. Rev. Mater. 2021, 6, 402–420. [Google Scholar] [CrossRef]
- Luecken, M.D.; Theis, F.J. Current best practices in single-cell RNA-seq analysis: A tutorial. Mol. Syst. Biol. 2019, 15, e8746. [Google Scholar] [CrossRef] [PubMed]
- Wu, H.; Zhu, J.; Huang, Y.; Wu, D.; Sun, J. Microfluidic-Based Single-Cell Study: Current Status and Future Perspective. Molecules 2018, 23, 2347. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vargo-Gogola, T.; Rosen, J.M. Modelling breast cancer: One size does not fit all. Nat. Rev. Cancer 2007, 7, 659–672. [Google Scholar] [CrossRef]
- Holliday, D.L.; Speirs, V. Choosing the right cell line for breast cancer research. Breast Cancer Res. 2011, 13, 215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Prasad, V.V.; Gopalan, R.O. Continued use of MDA-MB-435, a melanoma cell line, as a model for human breast cancer, even in year, 2014. NPJ Breast Cancer 2015, 1, 15002. [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] [Green Version]
- Weeber, F.; Ooft, S.; Dijkstra, K.; Voest, E.E. Tumor Organoids as a Pre-clinical Cancer Model for Drug Discovery. Cell Chem. Biol. 2017, 24, 1092–1100. [Google Scholar] [CrossRef]
- Neal, J.T.; Kuo, C.J. Organoids as Models for Neoplastic Transformation. Annu. Rev. Pathol. Mech. Dis. 2016, 11, 199–220. [Google Scholar] [CrossRef]
- Wörsdörfer, P.; Dalda, N.; Kern, A.; Krüger, S.; Wagner, N.; Kwok, C.K.; Henke, E.; Ergün, S. Generation of complex human organoid models including vascular networks by incorporation of mesodermal progenitor cells. Sci. Rep. 2019, 9, 15663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fong, E.L.; 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] [Green Version]
- 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, 6169. [Google Scholar] [CrossRef] [Green Version]
- 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, 6, e1331798. [Google Scholar] [CrossRef] [Green Version]
- Jeppesen, M.; Hagel, G.; Glenthoj, A.; Vainer, B.; Ibsen, P.; Harling, H.; Thastrup, O.; Jorgensen, 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]
- Liao, D.; Johnson, R.S. Hypoxia: A key regulator of angiogenesis in cancer. Cancer Metastasis Rev. 2007, 26, 281–290. [Google Scholar] [CrossRef] [PubMed]
- Park, J.E.; Tan, H.S.; Datta, A.; Lai, R.C.; Zhang, H.; Meng, W.; Lim, S.K.; Sze, S.K. Hypoxic Tumor Cell Modulates Its Microenvironment to Enhance Angiogenic and Metastatic Potential by Secretion of Proteins and Exosomes. Mol. Cell. Proteom. 2010, 9, 1085–1099. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Laoui, D.; Van Overmeire, E.; Di Conza, G.; Aldeni, C.; Keirsse, J.; Morias, Y.; Movahedi, K.; Houbracken, I.; Schouppe, E.; Elkrim, Y.; et al. Tumor Hypoxia Does Not Drive Differentiation of Tumor-Associated Macrophages but Rather Fine-Tunes the M2-like Macrophage Population. Cancer Res. 2014, 74, 24–30. [Google Scholar] [CrossRef] [Green Version]
- Onozuka, H.; Tsuchihara, K.; Esumi, H. Hypoglycemic/hypoxic condition in vitro mimicking the tumor microenvironment markedly reduced the efficacy of anticancer drugs. Cancer Sci. 2011, 102, 975–982. [Google Scholar] [CrossRef]
- Fischer, K.; Hoffmann, P.; Voelkl, S.; Meidenbauer, N.; Ammer, J.; Edinger, M.; Gottfried, E.; Schwarz, S.; Rothe, G.; Hoves, S.; et al. Inhibitory effect of tumor cell–derived lactic acid on human T cells. Blood 2007, 109, 3812–3819. [Google Scholar] [CrossRef]
- Goetze, K.; Walenta, S.; Ksiazkiewicz, M.; Kunz-Schughart, L.; Mueller-Klieser, W. Lactate enhances motility of tumor cells and inhibits monocyte migration and cytokine release. Int. J. Oncol. 2011, 39, 453–463. [Google Scholar] [CrossRef]
- Radhakrishnan, J.; Varadaraj, S.; Dash, S.K.; Sharma, A.; Verma, R.S. Organotypic cancer tissue models for drug screening: 3D constructs, bioprinting and microfluidic chips. Drug Discov. Today 2020, 25, 879–890. [Google Scholar] [CrossRef]
- Miller, C.P.; Shin, W.; Ahn, E.H.; Kim, H.J.; Kim, D.-H. Engineering Microphysiological Immune System Responses on Chips. Trends Biotechnol. 2020, 38, 857–872. [Google Scholar] [CrossRef] [PubMed]
- Perego, P.; On behalf of the EORTC PAMM Group; Hempel, G.; Linder, S.; Bradshaw, T.D.; Larsen, A.K.; Peters, G.J.; Phillips, R.M. Cellular pharmacology studies of anticancer agents: Recommendations from the EORTC-PAMM group. Cancer Chemother. Pharmacol. 2018, 81, 427–441. [Google Scholar] [CrossRef] [PubMed]
- Wennerberg, K. Cancer Cell Drug Response Transcriptomes in 3D. Cell Chem. Biol. 2016, 23, 1323–1324. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Langhans, S.A. Three-Dimensional in Vitro Cell Culture Models in Drug Discovery and Drug Repositioning. Front. Pharmacol. 2018, 9, 6. [Google Scholar] [CrossRef] [PubMed]
- Sampson, J.H.; Gunn, M.D.; Fecci, P.E.; Ashley, D.M. Brain immunology and immunotherapy in brain tumours. Nat. Rev. Cancer 2020, 20, 12–25. [Google Scholar] [CrossRef]
- Ma, S.; Li, X.; Wang, X.; Cheng, L.; Li, Z.; Zhang, C.; Ye, Z.; Qian, Q. Current Progress in CAR-T Cell Therapy for Solid Tumors. Int. J. Biol. Sci. 2019, 15, 2548–2560. [Google Scholar] [CrossRef] [Green Version]
- Salmon, H.; Franciszkiewicz, K.; Damotte, D.; Dieu-Nosjean, M.-C.; Validire, P.; Trautmann, A.; Mami-Chouaib, F.; Donnadieu, E. Matrix architecture defines the preferential localization and migration of T cells into the stroma of human lung tumors. J. Clin. Investig. 2012, 122, 899–910. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kuczek, D.E.; Larsen, A.M.H.; Thorseth, M.-L.; Carretta, M.; Kalvisa, A.; Siersbæk, M.S.; Simões, A.M.C.; Roslind, A.; Engelholm, L.H.; Noessner, E.; et al. Collagen density regulates the activity of tumor-infiltrating T cells. J. Immunother. Cancer 2019, 7, 68. [Google Scholar] [CrossRef] [Green Version]
- Carragher, N.; Piccinini, F.; Tesei, A.; Trask Jr, O.J.; Bickle, M.; Horvath, P. Concerns, challenges and promises of high-content analysis of 3D cellular models. Nat. Rev. Drug Discov. 2018, 17, 606. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Tumour Cells | Fibroblast Source | Platform and Matrix | Main Outcomes | Ref. | |
---|---|---|---|---|---|
Cancer Type | Source | ||||
Colorectal | Human cell line (HT-29) | Human cell line (CCD-18Co) | Collagen I Microfluidics | Increased spheroid size but decreased cell proliferation in co-culture; lower sensitivity to paclitaxel in co-culture | [85] |
Human cell line (HT-29) | Human cell line (CCD-18Co) | Transwell | Fibroblast activation and increased tumour cell migration and proliferation in co-culture; no resistance to 5-FU in co-culture | [94] | |
Multiple human cell lines (HCT116, Caco-2) | Human primary (CAF and NF) | Collagen with nylon mesh | Increased signs of tumour cell invasion and enrichment in pathways involved in hypoxia, ECM, EMT and angiogenesis; no differences between CAF and NF | [95] | |
Murine cell line (CT26) | Human primary (CAF and NF) | Collagen | Increased signs of tumour cell invasion in co-culture with CAF in comparison with co-culture with NF or tumour cell monoculture | [96] | |
Lung | Human cell line (A549) | Human primary (CAF and NF) | Collagen | Increased signs of tumour cell invasion and no differences in tumour cell proliferation in co-cultures with CAF | [97] |
Human cell lines (HCC827, NCI-H1975 and NCI-H1437) | Human primary (CAF) | Spheroids in collagen I and Matrigel | Increased signs of tumour cell invasion and decreased drug resistance to EGFR inhibitor in co-cultures | [98] | |
Human cell lines (PC-9 and HCC827) | Murine cell line (MRC5) | Transwell | Increased drug resistance to EGFR inhibitor in co-cultures | [99] | |
Human cell line | Murine cell line (WA-mFib) | Transwell Gelatine microspheres | CAF activation measured by αSMA Enhanced tumour cell invasion | [100] | |
Breast | Human cell line (MCF-7) | Human primary (dermal NF) | Spheroids in alginate microcapsules Spinner flasks | Loss of tumour epithelial phenotype, deposition of ECM proteins and increased proangiogenic potential in co-cultures | [101] |
Murine cell line (EMT6) | Murine cell line (NIH3T3) | Silk fibroin | Acquisition of CAF phenotype in co-culture, with decreased proliferation and signs of EMT; Enhanced resistance to doxorubicin in 3D vs. 2D monocultures than in co-cultures vs. monocultures | [102] | |
Human cell lines (BT474, T47D, MCF-7 and SKBR3) | Human primary (CAF and NF) | Spheroids | Fibroblast infiltration dependent on tumour cell line | [103,104] | |
Human cell lines (UACC-893, BT20, MDA-MB-453) | Human primary (foreskin NF) | Rotary suspension | Cancer cell invasion into fibroblast core; Deposition of ECM proteins | [105] | |
Human cell lines (MDA-MB-231 and MCF-7) | Human primary (breast CAF and skin NF) | Spheroids | Increased tumour cell proliferation and migration in co-culture with CAF but not NF; Increased α-SMA in CAF co-cultured with MDA-MB-231; NF not activated by MDA-MB-231 or MCF-7 | [106] | |
Human cell lines (T47D, MDA-MB-361 and MDA-MB-231) | Human primary (dermal NF) | Spheroids | Similar tumour growth in mono and co-cultures; No differences between mono and co-cultures in sensitivity to combination of chemotherapy and radiotherapy | [107] | |
Human cell line (MCF-7) | Murine cell line (MRC-5) | Spheroids | Increased tumour cell growth in co-cultures, with formation of necrotic spheroid cores. | [108] | |
Human cell line (MCF-7) | Murine cell line (3T3) | PET scaffold Microbioreactor with agitation | Increased resistance to tamoxifen, oxaliplatin and cisplatin in co-cultures | [109] | |
Human cell lines (MDA-MB-231 and MCF-7) | Human cell line (HTB-125) | Collagen I Microfluidics | Increased signs of tumour cell invasion, collagen deposition and stiffness in co-culture | [110] | |
Murine primary mammary tumour cells | Murine primary (CAF) | Organoids Matrigel | Co-culture increased signs of invasion through release of TGF-β | [111] | |
Human cell line (MDA-MB-231) | Human mammary fibroblasts (HMF) | Spheroids Alginate and Collagen I | Increased tumour and fibroblast invasion through alginate and collagen mixed gel than collagen only matrix Invasion potentiated by CXCL12-secreting fibroblasts | [112] | |
Pancreatic | Human cell line (Capan-1 and Paca-3) | Murine cell line (MRC-5); human immortalized | Collagen I and Matrigel | No alterations in tumour cell proliferation; Modulation of adhesion molecules | [113] |
Human cell line (Patu8902) | Human immortalized | Spheroids Bioprinting | Generation of heterotypic spheroids | [114] | |
Human cell line (PT45) | Human primary (CAF or normal) | Microcarriers Spinner flask | ECM deposition in co-cultures with NF and CAF; NF acquired activated phenotype | [115] | |
Human cell lines (PANC-1, AsPc-1, BxPC-3, Capan-1 and MIA PaCa-2) | Human primary (CAF) | Spheroids | Spheroids more compact in co-culture, with collagen deposition; Higher gemcitabine resistance in co-culture than tumour monospheroids | [116] | |
Lung Colorectal Esophageal Pancreatic | Patient-derived xenografts | Human primary (CAF) | BME | Drug resistance to different chemotherapeutics in co-culture | [87] |
Lung, breast, pancreatic | Human cell lines (e.g., A549, MCF-7, Panc1) | Murine cell line (MRC5); Human primary (CAF) and cell lines | Spheroid | Increased proliferation in co-cultures; Differential secretion of cytokines depending on the tumour cell line; Decreased drug sensitivity to targeted therapy in co-culture for lung tumour cell lines, but not for breast cancer cell lines | [117] |
Liver | Human cell line (HepG2) | Murine cell line (3T3-J2) | Spheroids Collagen | Higher drug resistance to doxorubicin in co-cultures | [118] |
Murine and human primary mammary and breast tumour cells | Murine and human primary (CAF) | Transwell Organoids Matrigel | Co-culture increased organoid growth but not organoid number In response to sorafenib, regorafenib or 5-FU, less organoid growth inhibition in co-cultures | [119] | |
Prostate | Human cell line (BPH-1) | Human primary (CAF and NF) | Fibroblast produced matrix | Increased signs of migration and invasion in co-culture | [120] |
Salivary gland adenoid cystic carcinoma | Human cell line (ACC-M) | Human Primary and cell line (HFL1) | BME Microfluidics | Increased signs of invasive phenotype in co-cultures with CAF, but not with NF MMP inhibitor blocked CAF-induced invasion | [121] |
Ovarian | Human cell line (OVCAR5) | Murine cell line (MRC-5) | Matrigel Bioprinting | Generation of co-cultures with different sizes and cell densities | [122] |
Breast Pancreatic | Cell lines (murine 4T1 and human MDA-MB-231 and Panc-1) | Murine and human cell lines (3T3, BJ-hTERT) and human primary (CAF, NF) | Spheroids | Increased α-SMA and collagen in co-culture; Decreased penetration of nanoparticles in co-culture | [123] |
Breast Lung | Human cell lines (MCF-7, SKBR3, A549) | Human primary (CAF from chemo-sensitive or chemo-resistant tumours) | Transwell | Increased drug resistance in co-cultures with CAF isolated from chemo-resistant tumours | [93] |
Breast Lung | Human cell lines (T47D, MCF-7; H1299) | Murine cell line (MRC5) and human NF | Spheroids | Fibroblast localized preferentially in the inner part of the spheroids; identification of specific compounds that inhibited fibroblast migration | [124] |
Breast Colorectal | Human cell lines (MCF-7 HCT-116) | Primary (human dermal NF) | Spheroids Microfluidics | Imaging and quantification of tumour cell spheroid invasion into fibroblast spheroid | [125] |
Tumour Cells | EC Source | Platform and Matrix | Main Outcomes | Ref. | |
---|---|---|---|---|---|
Cancer Type | Source | ||||
Liver Breast | Human cell lines (HepG2, MCF-7 and MDA-MB-231) | HUVEC | Spheroids | EC formed tube-like structures in co-cultures with HepG2 but not with breast cancer cell lines; structures declined after 3 days of culture | [153] |
Breast | Human cell lines (MCF7, T47-D and MDA-MB-231) | Primary (breast tissue) | BME | Increased breast cancer spheroid growth (size and proliferation) in co-culture with EC | [156] |
Murine cell line (4T1) | Murine tumour-like EC line (2H1) | Spheroids | EC infiltrated tumour spheroids; increased sensitivity to chemotherapy in co-culture but not to radiation | [152] | |
Human cell line (MDA-MB-231) | Human dermal microvascular cells | Microfluidics | CXCR4 or CXCR7 on breast cancer cells promoted adhesion to EC | [162] | |
Human cell line (MDA-MB-231) | HUVEC | Collagen and fibrin Microfluidics | Increased MDA-MB-231 invasion in collagen, in co-culture | [163] | |
Human cell line (MDA-MB-231) | Immortalized microvascular EC | Collagen Microfluidics | EC formed a confluent layer aligned with flow direction; upregulation of proangiogenic genes in flow vs. static conditions | [160] | |
Lung | Human cell line (A549) | HUVEC | Spheroids Microfluidics | Evidence of EMT in co-cultures, reverted by EGFR inhibitor | [159] |
Melanoma, Breast, Pancreatic | Multiple cell lines or patient-derived | HUVEC | Spheroids | Formation of capillary-like structures | [154] |
Liver | Human cell line (HepG2) | HUVEC | Glycosaminoglycan-based hydrogel | EC infiltration and tumour cell migration in co-culture | [77] |
Colorectal | Human cell line (HCT-116) | Human Colonic microvascular EC | Matrigel Microfluidics | EC formed tube-like structures; gemcitabine nanoparticles decreased tumour cell proliferation | [164] |
Colorectal Glioma | Cell lines (rat glioma C6 and human colorectal LoVo and HT29) | HUVEC | Microfluidics Matrigel | Spheroid secreted higher levels of VEGF than monolayers; EC formed more tube-like structures in 3D than in 2D co-cultures. | [165] |
Glioma/Glioblastoma | Patient-derived glioma cell line (GB3) | HUVEC | Matrigel and fibrin Microfluidics | Enhanced tumour cell migration in co-culture and no effect on proliferation; CXCR4 inhibitor decreased tumour cell migration. | [166] |
Human glioblastoma cell line U87MG | HUVEC | Spheroid Microfluidics Fibrin | EC tube-like formation towards tumour spheroid Antiangiogenics (bevacizumab and sunitinib) reduced EC migration | [167] |
Immune Cells | Tumour Cells | Platform and Matrix | Main Outcomes | Ref. | ||
---|---|---|---|---|---|---|
Cell Type | Source | Cancer Type | Source | |||
Monocytes/Macrophages | Human monocytic cell line (THP-1) | Prostate | Human cell line (BHP-1) | Transwell | Polarization to M2-like macrophages; increased tumour cell migration | [174] |
Murine leukaemia cell line (RAW 264.7) | Breast | Human cell line (MDA-MB-231) | Spheroids embedded in collagen | Polarization to M2-like macrophages; increased resistance of tumour cells to paclitaxel in co-cultures | [175] | |
Murine BM-derived macrophages | Murine cell line (Py8119) | Spheroids embedded in Matrigel | Macrophage infiltration in 3D; Increased tumour cell invasion in co-culture | [176] | ||
Peripheral blood- derived | Pancreatic | Human cell line (Panc-1) | Microfluidics | Increased macrophage migration in co-culture or induced by flow Partial blocking of macrophage migration by anti-IL-8 and anti-CCL2 | [177] | |
Natural killer cells (NK) | Peripheral blood- derived | Colorectal | Human cell line (HCT-116) | Collagen; Microfluidics | Increased migration of NK towards tumour cells | [178] |
Engineered NK cells (CAR-NK) | Colorectal | Human primary | Organoids in Matrigel | CAR-NK recognizing different antigens; increased cytotoxicity of CAR-NK towards tumour organoids than normal counterparts. | [179] | |
Human cell line (NK-92) | Lung | Human cell line (A549) | Spheroids Transwell | Enhanced migration and cytotoxicity of NK in the presence of CXCL12 | [180] | |
Peripheral blood- derived | Cervical | Human cell lines (CaSki, SiHa) | Spheroids | NK infiltration in spheroids and cytotoxicity towards tumour cells | [181] | |
Peripheral blood- derived | Liver | Human cell line (HepG2) | Spheroids | NK-mediated tumour lysis mainly at the periphery of the spheroids | [182] | |
Dendritic cells (DC) | Monocytes from peripheral blood | Urothelial Melanoma | Several human cell lines | Spheroids | Distinct DC phenotypes dependent on tumour cell line | [183] |
Monocytes from peripheral blood | Colorectal | Human cell line (SW620) | Microfluidics | DC migration towards tumour cells; histone deacetylase inhibitor and IFN-α led to increased DC migration, through activation of CXCR4/CCL12 axis | [184] | |
T lymphocytes | Human CTL clone | Lung | Human cell line (IGR-Heu) | Spheroids | Less CTL activation in 3D co-cultures than in 2D | [185] |
Human CTL clone | Metastatic melanoma | Human cell line (HBL) | Spheroids | Tumour associated antigen recognition by CTL decreased in 3D | [186] | |
Human cell line (Jurkat E6.1) | Lung | Human cell line (A549) | Transwell | Co-culture secretome enriched in proangiogenic and proinflammatory EMT-inducing factors. | [187] | |
Engineered T cells | Liver | Human cell line (HepG2) | Microfluidics | T cell infiltration and induction of tumour cell death | [188] | |
Peripheral blood lymphocytes | Colorectal and lung | Human organoids | 96-well U-bottom | Autologous tumour T cells recognized tumour organoids but not healthy counterparts | [189] | |
αβ T cells carrying a transgenic TCR peptide-specific | Colorectal | Human organoids | BME | Engineered T cells induced death of antigen-specific tumour cells | [190] | |
CAR-T | Lung Breast | Human cell line (A549; MDA-MB-231) | Microfluidics Porcine decellularized matrix | CAR-T decreased tumour cell volume and increased tumour apoptosis relative to untreated or co-culture with non-engineered T lymphocytes | [191] | |
PBMC | Human | Liver | Human primary hepatocytes (tumour and healthy) | 2D | T CD8+ showed increased activation but less viability in co-cultures | [192] |
Human | Prostate | Cell lines (human LNCaP, C4-2, C4-2B and murine TRAMP-C2) | Spheroids in chitosan– alginate or Matrigel | Decreased proliferation of tumour cells in comparison to Matrigel; immune cells infiltrated the tumour spheroids | [193] | |
Human | HNSCC | Human cell line (EpCAM-positive FaDu) | Spheroids Spinner flask | Immune cell infiltration into spheroids; bispecific antibody (anti-EpCAM and anti-CD3) alone or combined with cisplatin decreased spheroid viability | [194] | |
Human | HNSCC | Human cell lines (UD-SCC 4, 5, 6) | Spheroids | Anti-EGFR antibody induced leukocyte infiltration into tumour spheroids and the effect was abrogated by anti-CCL2 antibody | [195] | |
T and NK from healthy donors or patients | Colorectal | Human cell line (HT-29) or primary | Spheroids | T and NK infiltration into spheroids and increased tumour apoptosis in co-cultures; both processes enhanced by IL-15 supplementation | [196] | |
Treg and NK | Breast | Human cell lines (MCF-7 and MDA-MB-231) | Matrigel | Model establishment and implementation of analytical methods (RNA extraction, immunohistochemistry) | [197] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Franchi-Mendes, T.; Eduardo, R.; Domenici, G.; Brito, C. 3D Cancer Models: Depicting Cellular Crosstalk within the Tumour Microenvironment. Cancers 2021, 13, 4610. https://doi.org/10.3390/cancers13184610
Franchi-Mendes T, Eduardo R, Domenici G, Brito C. 3D Cancer Models: Depicting Cellular Crosstalk within the Tumour Microenvironment. Cancers. 2021; 13(18):4610. https://doi.org/10.3390/cancers13184610
Chicago/Turabian StyleFranchi-Mendes, Teresa, Rodrigo Eduardo, Giacomo Domenici, and Catarina Brito. 2021. "3D Cancer Models: Depicting Cellular Crosstalk within the Tumour Microenvironment" Cancers 13, no. 18: 4610. https://doi.org/10.3390/cancers13184610
APA StyleFranchi-Mendes, T., Eduardo, R., Domenici, G., & Brito, C. (2021). 3D Cancer Models: Depicting Cellular Crosstalk within the Tumour Microenvironment. Cancers, 13(18), 4610. https://doi.org/10.3390/cancers13184610