1
|
Fois MG, Tahmasebi Birgani ZN, López-Iglesias C, Knoops K, van Blitterswijk C, Giselbrecht S, Habibović P, Truckenmüller RK. In vitro vascularization of 3D cell aggregates in microwells with integrated vascular beds. Mater Today Bio 2024; 29:101260. [PMID: 39391792 PMCID: PMC11466645 DOI: 10.1016/j.mtbio.2024.101260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/20/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024] Open
Abstract
Most human tissues possess vascular networks supplying oxygen and nutrients. Engineering of functional tissue and organ models or equivalents often require the integration of artificial vascular networks. Several approaches, such as organs on chips and three-dimensional (3D) bioprinting, have been pursued to obtain vasculature and vascularized tissues in vitro. This technical feasibility study proposes a new approach for the in vitro vascularization of 3D microtissues. For this, we thermoform arrays of round-bottom microwells into thin non-porous and porous polymer films/membranes and culture vascular beds on them from which endothelial sprouting occurs in a Matrigel-based 3D extra cellular matrix. We present two possible culture configurations for the microwell-integrated vascular beds. In the first configuration, human umbilical vein endothelial cells (HUVECs) grow on and sprout from the inner wall of the non-porous microwells. In the second one, HUVECs grow on the outer surface of the porous microwells and sprout through the pores toward the inside. These approaches are extended to lymphatic endothelial cells. As a proof of concept, we demonstrate the in vitro vascularization of spheroids from human mesenchymal stem cells and MG-63 human osteosarcoma cells. Our results show the potential of this approach to provide the spheroids with an abundant outer vascular network and the indication of an inner vasculature.
Collapse
Affiliation(s)
- Maria G. Fois
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Zeinab N. Tahmasebi Birgani
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Carmen López-Iglesias
- Microscopy CORE Lab, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
| | - Kèvin Knoops
- Microscopy CORE Lab, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
| | - Clemens van Blitterswijk
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Stefan Giselbrecht
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Pamela Habibović
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Roman K. Truckenmüller
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| |
Collapse
|
2
|
Abbasi-Malati Z, Khanicheragh P, Narmi MT, Mardi N, Khosrowshahi ND, Hiradfar A, Rezabakhsh A, Sadeghsoltani F, Rashidi S, Chegeni SA, Roozbahani G, Rahbarghazi R. Tumoroids, a valid preclinical screening platform for monitoring cancer angiogenesis. Stem Cell Res Ther 2024; 15:267. [PMID: 39183337 PMCID: PMC11346257 DOI: 10.1186/s13287-024-03880-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 08/06/2024] [Indexed: 08/27/2024] Open
Abstract
In recent years, biologists and clinicians have witnessed prominent advances in in vitro 3D culture techniques related to biomimetic human/animal tissue analogs. Numerous data have confirmed that unicellular and multicellular (tumoroids) tumor spheroids with dense native cells in certain matrices are sensitive and valid analytical tools for drug screening, cancer cell dynamic growth, behavior, etc. in laboratory settings. Angiogenesis/vascularization is a very critical biological phenomenon to support oxygen and nutrients to tumor cells within the deep layer of solid masses. It has been shown that endothelial cell (EC)-incorporated or -free spheroid/tumoroid systems provide a relatively reliable biological platform for monitoring the formation of nascent blood vessels in micron/micrometer scales. Besides, the paracrine angiogenic activity of cells within the spheroid/tumoroid systems can be monitored after being treated with different therapeutic approaches. Here, we aimed to collect recent advances and findings related to the monitoring of cancer angiogenesis using unicellular and multicellular tumor spheroids. Vascularized spheroids/tumoroids can help us in the elucidation of mechanisms related to cancer formation, development, and metastasis by monitoring the main influencing factors.
Collapse
Affiliation(s)
- Zahra Abbasi-Malati
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parisa Khanicheragh
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Narges Mardi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nafiseh Didar Khosrowshahi
- Stem Cell and Tissue Engineering Research Laboratory, Sahand University of Technology, Tabriz, 51335-1996, Iran
| | - Amirataollah Hiradfar
- Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aysa Rezabakhsh
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Somayyeh Rashidi
- Department of Medical Biotechnology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | | | - Golbarg Roozbahani
- Department of Plant, Cell and Molecular Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
3
|
Ascheid D, Baumann M, Pinnecker J, Friedrich M, Szi-Marton D, Medved C, Bundalo M, Ortmann V, Öztürk A, Nandigama R, Hemmen K, Ergün S, Zernecke A, Hirth M, Heinze KG, Henke E. A vascularized breast cancer spheroid platform for the ranked evaluation of tumor microenvironment-targeted drugs by light sheet fluorescence microscopy. Nat Commun 2024; 15:3599. [PMID: 38678014 PMCID: PMC11055956 DOI: 10.1038/s41467-024-48010-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 04/18/2024] [Indexed: 04/29/2024] Open
Abstract
Targeting the supportive tumor microenvironment (TME) is an approach of high interest in cancer drug development. However, assessing TME-targeted drug candidates presents a unique set of challenges. We develop a comprehensive screening platform that allows monitoring, quantifying, and ranking drug-induced effects in self-organizing, vascularized tumor spheroids (VTSs). The confrontation of four human-derived cell populations makes it possible to recreate and study complex changes in TME composition and cell-cell interaction. The platform is modular and adaptable for tumor entity or genetic manipulation. Treatment effects are recorded by light sheet fluorescence microscopy and translated by an advanced image analysis routine in processable multi-parametric datasets. The system proved to be robust, with strong interassay reliability. We demonstrate the platform's utility for evaluating TME-targeted antifibrotic and antiangiogenic drugs side-by-side. The platform's output enabled the differential evaluation of even closely related drug candidates according to projected therapeutic needs.
Collapse
Affiliation(s)
- David Ascheid
- Institute of Anatomy and Cell Biology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Magdalena Baumann
- Institute of Anatomy and Cell Biology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Jürgen Pinnecker
- Chair of Molecular Microscopy, Rudolf-Virchow-Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Mike Friedrich
- Chair of Molecular Microscopy, Rudolf-Virchow-Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Daniel Szi-Marton
- Institute of Anatomy and Cell Biology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Cornelia Medved
- Institute of Anatomy and Cell Biology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Maja Bundalo
- Institute of Experimental Biomedicine, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Vanessa Ortmann
- Institute of Anatomy and Cell Biology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Asli Öztürk
- Institute of Anatomy and Cell Biology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Rajender Nandigama
- Institute of Anatomy and Cell Biology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
- Max Planck Institute of Heart and Lung Research, Bad Nauheim, Germany
| | - Katherina Hemmen
- Chair of Molecular Microscopy, Rudolf-Virchow-Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Süleymann Ergün
- Institute of Anatomy and Cell Biology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Alma Zernecke
- Institute of Experimental Biomedicine, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Matthias Hirth
- Institut für Medientechnik, Technische Universität Illmenau, Illmenau, Germany
| | - Katrin G Heinze
- Chair of Molecular Microscopy, Rudolf-Virchow-Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg, Würzburg, Germany.
| | - Erik Henke
- Institute of Anatomy and Cell Biology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany.
- Graduate School for Life Sciences, Julius-Maximilians-Universität Würzburg, Würzburg, Germany.
| |
Collapse
|
4
|
Griffin KH, Thorpe SW, Sebastian A, Hum NR, Coonan TP, Sagheb IS, Loots GG, Randall RL, Leach JK. Engineered bone marrow as a clinically relevant ex vivo model for primary bone cancer research and drug screening. Proc Natl Acad Sci U S A 2023; 120:e2302101120. [PMID: 37729195 PMCID: PMC10523456 DOI: 10.1073/pnas.2302101120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 08/17/2023] [Indexed: 09/22/2023] Open
Abstract
Osteosarcoma (OS) is the most common primary malignant bone cancer in children and adolescents. While numerous other cancers now have promising therapeutic advances, treatment options for OS have remained unchanged since the advent of standard chemotherapeutics and offer less than a 25% 5-y survival rate for those with metastatic disease. This dearth of clinical progress underscores a lack of understanding of OS progression and necessitates the study of this disease in an innovative system. Here, we adapt a previously described engineered bone marrow (eBM) construct for use as a three-dimensional platform to study how microenvironmental and immune factors affect OS tumor progression. We form eBM by implanting acellular bone-forming materials in mice and explanting the cellularized constructs after 8 wk for study. We interrogate the influence of the anatomical implantation site on eBM tissue quality, test ex vivo stability under normoxic (5% O2) and standard (21% O2) culture conditions, culture OS cells within these constructs, and compare them to human OS samples. We show that eBM stably recapitulates the composition of native bone marrow. OS cells exhibit differential behavior dependent on metastatic potential when cultured in eBM, thus mimicking in vivo conditions. Furthermore, we highlight the clinical applicability of eBM as a drug-screening platform through doxorubicin treatment and show that eBM confers a protective effect on OS cells that parallel clinical responses. Combined, this work presents eBM as a cellular construct that mimics the complex bone marrow environment that is useful for mechanistic bone cancer research and drug screening.
Collapse
Affiliation(s)
- Katherine H. Griffin
- Department of Orthopaedic Surgery, University of California Davis Health, Sacramento, CA95817
- School of Veterinary Medicine, University of California, Davis, CA95616
| | - Steven W. Thorpe
- Department of Orthopaedic Surgery, University of California Davis Health, Sacramento, CA95817
| | - Aimy Sebastian
- Physical and Life Sciences, Lawrence Livermore National Laboratory, Livermore, CA94550
| | - Nicholas R. Hum
- Physical and Life Sciences, Lawrence Livermore National Laboratory, Livermore, CA94550
| | - Thomas P. Coonan
- Department of Biomedical Engineering, University of California, Davis, CA95616
| | - Isabel S. Sagheb
- Department of Biomedical Engineering, University of California, Davis, CA95616
| | - Gabriela G. Loots
- Department of Orthopaedic Surgery, University of California Davis Health, Sacramento, CA95817
- Physical and Life Sciences, Lawrence Livermore National Laboratory, Livermore, CA94550
| | - R. Lor Randall
- Department of Orthopaedic Surgery, University of California Davis Health, Sacramento, CA95817
| | - J. Kent Leach
- Department of Orthopaedic Surgery, University of California Davis Health, Sacramento, CA95817
- Department of Biomedical Engineering, University of California, Davis, CA95616
| |
Collapse
|
5
|
Mason J, Öhlund D. Key aspects for conception and construction of co-culture models of tumor-stroma interactions. Front Bioeng Biotechnol 2023; 11:1150764. [PMID: 37091337 PMCID: PMC10119418 DOI: 10.3389/fbioe.2023.1150764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/31/2023] [Indexed: 04/25/2023] Open
Abstract
The tumor microenvironment is crucial in the initiation and progression of cancers. The interplay between cancer cells and the surrounding stroma shapes the tumor biology and dictates the response to cancer therapies. Consequently, a better understanding of the interactions between cancer cells and different components of the tumor microenvironment will drive progress in developing novel, effective, treatment strategies. Co-cultures can be used to study various aspects of these interactions in detail. This includes studies of paracrine relationships between cancer cells and stromal cells such as fibroblasts, endothelial cells, and immune cells, as well as the influence of physical and mechanical interactions with the extracellular matrix of the tumor microenvironment. The development of novel co-culture models to study the tumor microenvironment has progressed rapidly over recent years. Many of these models have already been shown to be powerful tools for further understanding of the pathophysiological role of the stroma and provide mechanistic insights into tumor-stromal interactions. Here we give a structured overview of different co-culture models that have been established to study tumor-stromal interactions and what we have learnt from these models. We also introduce a set of guidelines for generating and reporting co-culture experiments to facilitate experimental robustness and reproducibility.
Collapse
Affiliation(s)
- James Mason
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Daniel Öhlund
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
- *Correspondence: Daniel Öhlund,
| |
Collapse
|
6
|
Simpson S, Rizvanov AA, Jeyapalan JN, de Brot S, Rutland CS. Canine osteosarcoma in comparative oncology: Molecular mechanisms through to treatment discovery. Front Vet Sci 2022; 9:965391. [PMID: 36570509 PMCID: PMC9773846 DOI: 10.3389/fvets.2022.965391] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 11/23/2022] [Indexed: 12/13/2022] Open
Abstract
Cancer is a leading cause of non-communicable morbidity and mortality throughout the world, similarly, in dogs, the most frequent cause of mortality is tumors. Some types of cancer, including osteosarcoma (OSA), occur at much higher rates in dogs than people. Dogs therefore not only require treatment themselves but can also act as an effective parallel patient population for the human disease equivalent. It should be noted that although there are many similarities between canine and human OSA, there are also key differences and it is important to research and highlight these features. Despite progress using chorioallantoic membrane models, 2D and 3D in vitro models, and rodent OSA models, many more insights into the molecular and cellular mechanisms, drug development, and treatment are being discovered in a variety of canine OSA patient populations.
Collapse
Affiliation(s)
- Siobhan Simpson
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
| | - Albert A. Rizvanov
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Jennie N. Jeyapalan
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
- Faculty of Medicine and Health Science, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Simone de Brot
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
- Comparative Pathology Platform (COMPATH), Institute of Animal Pathology, University of Bern, Bern, Switzerland
| | - Catrin S. Rutland
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
7
|
Anthon SG, Valente KP. Vascularization Strategies in 3D Cell Culture Models: From Scaffold-Free Models to 3D Bioprinting. Int J Mol Sci 2022; 23:14582. [PMID: 36498908 PMCID: PMC9737506 DOI: 10.3390/ijms232314582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/21/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022] Open
Abstract
The discrepancies between the findings in preclinical studies, and in vivo testing and clinical trials have resulted in the gradual decline in drug approval rates over the past decades. Conventional in vitro drug screening platforms employ two-dimensional (2D) cell culture models, which demonstrate inaccurate drug responses by failing to capture the three-dimensional (3D) tissue microenvironment in vivo. Recent advancements in the field of tissue engineering have made possible the creation of 3D cell culture systems that can accurately recapitulate the cell-cell and cell-extracellular matrix interactions, as well as replicate the intricate microarchitectures observed in native tissues. However, the lack of a perfusion system in 3D cell cultures hinders the establishment of the models as potential drug screening platforms. Over the years, multiple techniques have successfully demonstrated vascularization in 3D cell cultures, simulating in vivo-like drug interactions, proposing the use of 3D systems as drug screening platforms to eliminate the deviations between preclinical and in vivo testing. In this review, the basic principles of 3D cell culture systems are briefly introduced, and current research demonstrating the development of vascularization in 3D cell cultures is discussed, with a particular focus on the potential of these models as the future of drug screening platforms.
Collapse
Affiliation(s)
- Shamapto Guha Anthon
- Department of Biomedical Engineering, University of Victoria, Victoria, BC V8W 2Y2, Canada
| | | |
Collapse
|
8
|
Maia FR, Bastos AR, Oliveira JM, Correlo VM, Reis RL. Recent approaches towards bone tissue engineering. Bone 2022; 154:116256. [PMID: 34781047 DOI: 10.1016/j.bone.2021.116256] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 10/19/2021] [Accepted: 11/09/2021] [Indexed: 12/17/2022]
Abstract
Bone tissue engineering approaches have evolved towards addressing the challenges of tissue mimetic requirements over the years. Different strategies have been combining scaffolds, cells, and biologically active cues using a wide range of fabrication techniques, envisioning the mimicry of bone tissue. On the one hand, biomimetic scaffold-based strategies have been pursuing different biomaterials to produce scaffolds, combining with diverse and innovative fabrication strategies to mimic bone tissue better, surpassing bone grafts. On the other hand, biomimetic scaffold-free approaches mainly foresee replicating endochondral ossification, replacing hyaline cartilage with new bone. Finally, since bone tissue is highly vascularized, new strategies focused on developing pre-vascularized scaffolds or pre-vascularized cellular aggregates have been a motif of study. The recent biomimetic scaffold-based and scaffold-free approaches in bone tissue engineering, focusing on materials and fabrication methods used, are overviewed herein. The biomimetic vascularized approaches are also discussed, namely the development of pre-vascularized scaffolds and pre-vascularized cellular aggregates.
Collapse
Affiliation(s)
- F Raquel Maia
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's PT Government Associate Laboratory, Braga, Guimarães, Portugal.
| | - Ana R Bastos
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Joaquim M Oliveira
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Vitor M Correlo
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's PT Government Associate Laboratory, Braga, Guimarães, Portugal
| |
Collapse
|
9
|
Shoaib Z, Fan TM, Irudayaraj J. Osteosarcoma mechanobiology and therapeutic targets. Br J Pharmacol 2021; 179:201-217. [PMID: 34679192 PMCID: PMC9305477 DOI: 10.1111/bph.15713] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 09/15/2021] [Accepted: 09/22/2021] [Indexed: 11/28/2022] Open
Abstract
Osteosarcoma (OS) is the one of the most common primary tumors of bone with less than a 20% 5-year survival rate after the development of metastases. OS is highly predisposed in Paget's disease (PD) of bone, and both have common characteristic skeletal features due to rapid bone remodeling. OS prognosis is location dependent which further emphasizes the likely contribution of the bone microenvironment in its pathogenesis. Mechanobiology is the phenomenon when mechanical cues from the changing physical microenvironment of bone are transduced to biological pathways through mechanosensitive cellular components. Mechanobiology-driven therapies have been used for curbing tumor progression by direct alteration of the physical microenvironment or inhibition of metastasis-associated mechanosensitive proteins. This review emphasizes the contribution of mechanobiology to OS progression, and sheds light on current mechanobiology-based therapies and potential new targets for improving disease management. Additionally, the variety of 3D models currently used to study OS mechanobiology are summarized.
Collapse
Affiliation(s)
- Zunaira Shoaib
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Timothy M Fan
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Joseph Irudayaraj
- Department of Bioengineering, Nick Holonyak Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, IL, USA.,Biomedical Research Center, Carle Foundation Hospital, Urbana, IL, USA.,Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
10
|
Franchi-Mendes T, Eduardo R, Domenici G, Brito C. 3D Cancer Models: Depicting Cellular Crosstalk within the Tumour Microenvironment. Cancers (Basel) 2021; 13:4610. [PMID: 34572836 PMCID: PMC8468887 DOI: 10.3390/cancers13184610] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 12/11/2022] Open
Abstract
The tumour microenvironment plays a critical role in tumour progression and drug resistance processes. Non-malignant cell players, such as fibroblasts, endothelial cells, immune cells and others, interact with each other and with the tumour cells, shaping the disease. Though the role of each cell type and cell communication mechanisms have been progressively studied, the complexity of this cellular network and its role in disease mechanism and therapeutic response are still being unveiled. Animal models have been mainly used, as they can represent systemic interactions and conditions, though they face recognized limitations in translational potential due to interspecies differences. In vitro 3D cancer models can surpass these limitations, by incorporating human cells, including patient-derived ones, and allowing a range of experimental designs with precise control of each tumour microenvironment element. We summarize the role of each tumour microenvironment component and review studies proposing 3D co-culture strategies of tumour cells and non-malignant cell components. Moreover, we discuss the potential of these modelling approaches to uncover potential therapeutic targets in the tumour microenvironment and assess therapeutic efficacy, current bottlenecks and perspectives.
Collapse
Affiliation(s)
- Teresa Franchi-Mendes
- iBET—Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (T.F.-M.); (R.E.); (G.D.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Rodrigo Eduardo
- iBET—Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (T.F.-M.); (R.E.); (G.D.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Giacomo Domenici
- iBET—Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (T.F.-M.); (R.E.); (G.D.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Catarina Brito
- iBET—Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (T.F.-M.); (R.E.); (G.D.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Av. da República, 2780-157 Oeiras, Portugal
| |
Collapse
|
11
|
Munoz-Garcia J, Jubelin C, Loussouarn A, Goumard M, Griscom L, Renodon-Cornière A, Heymann MF, Heymann D. In vitro three-dimensional cell cultures for bone sarcomas. J Bone Oncol 2021; 30:100379. [PMID: 34307011 PMCID: PMC8287221 DOI: 10.1016/j.jbo.2021.100379] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/24/2021] [Accepted: 06/26/2021] [Indexed: 12/18/2022] Open
Abstract
Bone sarcomas are rare tumour entities that arise from the mesenchyme most of which are highly heterogeneous at the cellular, genetic and epigenetic levels. The three main types are osteosarcoma, Ewing sarcoma, and chondrosarcoma. These oncological entities are characterised by high morbidity and mortality and an absence of significant therapeutic improvement in the last four decades. In the field of oncology, in vitro cultures of cancer cells have been extensively used for drug screening unfortunately with limited success. Indeed, despite the massive knowledge acquired from conventional 2D culture methods, scientific community has been challenged by the loss of efficacy of drugs when moved to clinical trials. The recent explosion of new 3D culture methods is paving the way to more relevant in vitro models mimicking the in vivo tumour environment (e.g. bone structure) with biological responses close to the in vivo context. The present review gives a brief overview of the latest advances of the 3D culture methods used for studying primary bone sarcomas.
Collapse
Affiliation(s)
- Javier Munoz-Garcia
- Université de Nantes, INSERM, Nantes, France.,Institut de Cancérologie de l'Ouest, Tumour Heterogeneity and Precision Medicine Laboratory, Saint-Herblain, France
| | - Camille Jubelin
- Université de Nantes, INSERM, Nantes, France.,Institut de Cancérologie de l'Ouest, Tumour Heterogeneity and Precision Medicine Laboratory, Saint-Herblain, France.,Atlantic Bone Screen, Saint-Herblain, France
| | | | - Matisse Goumard
- Université de Nantes, INSERM, Nantes, France.,Institut de Cancérologie de l'Ouest, Tumour Heterogeneity and Precision Medicine Laboratory, Saint-Herblain, France
| | | | | | - Marie-Françoise Heymann
- Université de Nantes, INSERM, Nantes, France.,Institut de Cancérologie de l'Ouest, Tumour Heterogeneity and Precision Medicine Laboratory, Saint-Herblain, France
| | - Dominique Heymann
- Université de Nantes, INSERM, Nantes, France.,Institut de Cancérologie de l'Ouest, Tumour Heterogeneity and Precision Medicine Laboratory, Saint-Herblain, France.,University of Sheffield, Department of Oncology and Metabolism, Medical School, Sheffield, UK
| |
Collapse
|
12
|
Ma R, Mandell J, Lu F, Heim T, Schoedel K, Duensing A, Watters RJ, Weiss KR. Do Patient-derived Spheroid Culture Models Have Relevance in Chondrosarcoma Research? Clin Orthop Relat Res 2021; 479:477-490. [PMID: 32469486 PMCID: PMC7899730 DOI: 10.1097/corr.0000000000001317] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 04/27/2020] [Indexed: 01/31/2023]
Abstract
BACKGROUND In high-grade chondrosarcoma, 5-year survival is lower than 50%. Therefore, it is important that preclinical models that mimic the disease with the greatest possible fidelity are used to potentially develop new treatments. Accumulating evidence suggests that two-dimensional (2-D) cell culture may not accurately represent the tumor's biology. It has been demonstrated in other cancers that three-dimensional (3-D) cancer cell spheroids may recapitulate tumor biology and response to treatment with greater fidelity than traditional 2-D techniques. To our knowledge, the formation of patient-derived chondrosarcoma spheroids has not been described. QUESTIONS/PURPOSES (1) Can patient-derived chondrosarcoma spheroids be produced? (2) Do spheroids recapitulate human chondrosarcoma better than 2-D cultures, both morphologically and molecularly? (3) Can chondrosarcoma spheroids provide an accurate model to test novel treatments? METHODS Experiments to test the feasibility of spheroid formation of chondrosarcoma cells were performed using HT-1080, an established chondrosarcoma cell line, and two patient-derived populations, TP19-S26 and TP19-S115. Cells were cultured in flasks, trypsinized, and seeded into 96-well ultra-low attachment plates with culture media. After spheroids formed, they were monitored daily by bright-field microscopy. Spheroids were fixed using paraformaldehyde and embedded in agarose. After dehydration with isopropanol, paraffin-embedded spheroids were sectioned, and slides were stained with hematoxylin and eosin. To compare differences and similarities in gene expression between 2-D and 3-D chondrosarcoma cultures and primary tumors, and to determine whether these spheroids recapitulated the biology of chondrosarcoma, RNA was extracted from 2-D cultures, spheroids, and tumors. Quantitative polymerase chain reaction was performed to detect chondrosarcoma markers of interest, including vascular endothelial growth factor alpha, hypoxia-inducible factor 1α, COL2A1, and COL10A1. To determine whether 2-D and 3-D cultures responded differently to novel chondrosarcoma treatments, we compared their sensitivities to disulfiram and copper chloride treatment. To test their sensitivity to disulfiram and copper chloride treatment, 10,000 cells were seeded into 96-well plates for 2-D culturing and 3000 cells in each well for 3-D culturing. After treating the cells with disulfiram and copper for 48 hours, we detected cell viability using quantitative presto-blue staining and measured via plate reader. RESULTS Cell-line and patient-derived spheroids were cultured and monitored over 12 days. Qualitatively, we observed that HT-1080 demonstrated unlimited growth, while TP19-S26 and TP19-S115 contracted during culturing relative to their initial size. Hematoxylin and eosin staining of HT-1080 spheroids revealed that cell-cell attachments were more pronounced at the periphery of the spheroid structure than at the core, while the core was less dense. Spheroids derived from the intermediate-grade chondrosarcoma TP19-S26 were abundant in extracellular matrix, and spheroids derived from the dedifferentiated chondrosarcoma TP19-S115 had a higher cellularity and heterogeneity with spindle cells at the periphery. In the HT-1080 cells, differences in gene expression were appreciated with spheroids demonstrating greater expressions of VEGF-α (1.01 ± 0.16 versus 6.48 ± 0.55; p = 0.003), COL2A1 (1.00 ± 0.10 versus 7.46 ± 2.52; p < 0.001), and COL10A1 (1.01 ± 0.19 versus 22.53 ± 4.91; p < 0.001). Differences in gene expressions were also noted between primary tumors, spheroids, and 2-D cultures in the patient-derived samples TP19-S26 and TP19-S115. TP19-S26 is an intermediate-grade chondrosarcoma. With the numbers we had, we could not detect a difference in VEGF-α and HIF1α gene expression compared with the primary tumor. COL2A1 (1.00 ± 0.14 versus 1.76 ± 0.10 versus 335.66 ± 31.13) and COL10A1 (1.06 ± 0.378 versus 5.98 ± 0.45 versus 138.82 ± 23.4) expressions were both greater in the tumor (p (COL2A1) < 0.001; p (COL10A1) < 0.0001) and 3-D cultures (p (COL2A1) = 0.004; p (COL10A1) < 0.0001) compared with 2-D cultures. We could not demonstrate a difference in VEGF-α and HIF1α expressions in TP19-S115, a dedifferentiated chondrosarcoma, in the tumor compared with 2-D and 3-D cultures. COL2A1 (1.00 ± 0.02 versus 1.86 ± 0.18 versus 2.95 ± 0.56) and COL10A1 (1.00 ± 0.03 versus 5.52 ± 0.66 versus 3.79 ± 0.36) expressions were both greater in spheroids (p (COL2A1) = 0.003; p (COL10A1) < 0.0001) and tumors (p (COL2A1) < 0.001; p (COL10A1) < 0.0001) compared with 2-D cultures. Disulfiram-copper chloride treatment demonstrated high cytotoxicity in HT-1080 and SW-1353 chondrosarcoma cells grown in the 2-D monolayer, but 3-D spheroids were highly resistant to this treatment. CONCLUSION We provide preliminary findings that it is possible to generate 3-D spheroids from chondrosarcoma cell lines and two human chondrosarcomas (one dedifferentiated chondrosarcoma and one intermediate-grade chondrosarcoma). Chondrosarcoma spheroids derived from human tumors demonstrated morphology more reminiscent of primary tumors than cells grown in 2-D culture. Spheroids displayed similar expressions of cartilage markers as the primary tumor, and we observed a higher expression of collagen markers in the spheroids compared with cells grown in monolayer. Spheroids also demonstrated greater chemotherapy resistance than monolayer cells, but more patient-derived spheroids are needed to further conclude that 3-D cultures may mimic the chemoresistance that chondrosarcomas demonstrate clinically. Additional studies on patient-derived chondrosarcoma spheroids are warranted. CLINICAL RELEVANCE Chondrosarcomas demonstrate resistance to chemotherapy and radiation, and we believe that if they can be replicated, models such as 3-D spheroids may provide a method to test novel treatments for human chondrosarcoma. Additional comprehensive genomic studies are required to compare 2-D and 3-D models with the primary tumor to determine the most effective way to study this disease in vitro.
Collapse
Affiliation(s)
- Ruichen Ma
- R. Ma, J. Mandell, F. Lu, T. Heim, R. Watters, K. R. Weiss, Musculoskeletal Oncology Laboratory, University of Pittsburgh School of Medicine Department of Orthopaedic Surgery, Pittsburgh, PA, USA
- R. Ma, F. Lu, School of Medicine, Tsinghua University, Beijing, China
- J. Mandell, Department of Infectious Diseases and Microbiology, University of Pittsburgh, PA, USA
- K. Schoedel, A. Duensing, K. R. Weiss, Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
- A. Duensing, R. Watters, K. R. Weiss, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Jonathan Mandell
- R. Ma, J. Mandell, F. Lu, T. Heim, R. Watters, K. R. Weiss, Musculoskeletal Oncology Laboratory, University of Pittsburgh School of Medicine Department of Orthopaedic Surgery, Pittsburgh, PA, USA
- R. Ma, F. Lu, School of Medicine, Tsinghua University, Beijing, China
- J. Mandell, Department of Infectious Diseases and Microbiology, University of Pittsburgh, PA, USA
- K. Schoedel, A. Duensing, K. R. Weiss, Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
- A. Duensing, R. Watters, K. R. Weiss, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Feiqi Lu
- R. Ma, J. Mandell, F. Lu, T. Heim, R. Watters, K. R. Weiss, Musculoskeletal Oncology Laboratory, University of Pittsburgh School of Medicine Department of Orthopaedic Surgery, Pittsburgh, PA, USA
- R. Ma, F. Lu, School of Medicine, Tsinghua University, Beijing, China
- J. Mandell, Department of Infectious Diseases and Microbiology, University of Pittsburgh, PA, USA
- K. Schoedel, A. Duensing, K. R. Weiss, Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
- A. Duensing, R. Watters, K. R. Weiss, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Tanya Heim
- R. Ma, J. Mandell, F. Lu, T. Heim, R. Watters, K. R. Weiss, Musculoskeletal Oncology Laboratory, University of Pittsburgh School of Medicine Department of Orthopaedic Surgery, Pittsburgh, PA, USA
- R. Ma, F. Lu, School of Medicine, Tsinghua University, Beijing, China
- J. Mandell, Department of Infectious Diseases and Microbiology, University of Pittsburgh, PA, USA
- K. Schoedel, A. Duensing, K. R. Weiss, Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
- A. Duensing, R. Watters, K. R. Weiss, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Karen Schoedel
- R. Ma, J. Mandell, F. Lu, T. Heim, R. Watters, K. R. Weiss, Musculoskeletal Oncology Laboratory, University of Pittsburgh School of Medicine Department of Orthopaedic Surgery, Pittsburgh, PA, USA
- R. Ma, F. Lu, School of Medicine, Tsinghua University, Beijing, China
- J. Mandell, Department of Infectious Diseases and Microbiology, University of Pittsburgh, PA, USA
- K. Schoedel, A. Duensing, K. R. Weiss, Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
- A. Duensing, R. Watters, K. R. Weiss, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Anette Duensing
- R. Ma, J. Mandell, F. Lu, T. Heim, R. Watters, K. R. Weiss, Musculoskeletal Oncology Laboratory, University of Pittsburgh School of Medicine Department of Orthopaedic Surgery, Pittsburgh, PA, USA
- R. Ma, F. Lu, School of Medicine, Tsinghua University, Beijing, China
- J. Mandell, Department of Infectious Diseases and Microbiology, University of Pittsburgh, PA, USA
- K. Schoedel, A. Duensing, K. R. Weiss, Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
- A. Duensing, R. Watters, K. R. Weiss, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Rebecca J Watters
- R. Ma, J. Mandell, F. Lu, T. Heim, R. Watters, K. R. Weiss, Musculoskeletal Oncology Laboratory, University of Pittsburgh School of Medicine Department of Orthopaedic Surgery, Pittsburgh, PA, USA
- R. Ma, F. Lu, School of Medicine, Tsinghua University, Beijing, China
- J. Mandell, Department of Infectious Diseases and Microbiology, University of Pittsburgh, PA, USA
- K. Schoedel, A. Duensing, K. R. Weiss, Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
- A. Duensing, R. Watters, K. R. Weiss, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Kurt R Weiss
- R. Ma, J. Mandell, F. Lu, T. Heim, R. Watters, K. R. Weiss, Musculoskeletal Oncology Laboratory, University of Pittsburgh School of Medicine Department of Orthopaedic Surgery, Pittsburgh, PA, USA
- R. Ma, F. Lu, School of Medicine, Tsinghua University, Beijing, China
- J. Mandell, Department of Infectious Diseases and Microbiology, University of Pittsburgh, PA, USA
- K. Schoedel, A. Duensing, K. R. Weiss, Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
- A. Duensing, R. Watters, K. R. Weiss, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| |
Collapse
|
13
|
Chow T, Wutami I, Lucarelli E, Choong PF, Duchi S, Di Bella C. Creating In Vitro Three-Dimensional Tumor Models: A Guide for the Biofabrication of a Primary Osteosarcoma Model. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:514-529. [PMID: 33138724 DOI: 10.1089/ten.teb.2020.0254] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Osteosarcoma (OS) is a highly aggressive primary bone tumor. The mainstay for its treatment is multiagent chemotherapy and surgical resection, with a 50-70% 5-year survival rate. Despite the huge effort made by clinicians and researchers in the past 30 years, limited progress has been made to improve patient outcomes. As novel therapeutic approaches for OS become available, such as monoclonal antibodies, small molecules, and immunotherapies, the need for OS preclinical model development becomes equally pressing. Three-dimensional (3D) OS models represent an alternative system to study this tumor: In contrast to two-dimensional monolayers, 3D matrices can recapitulate key elements of the tumor microenvironment (TME), such as the cellular interaction with the bone mineralized matrix. The advancement of tissue engineering and biofabrication techniques enables the incorporation of specific TME aspects into 3D models, to investigate the contribution of individual components to tumor progression and enhance understanding of basic OS biology. The use of biomaterials that mimic the extracellular matrix could also facilitate the testing of drugs targeting the TME itself, allowing a larger range of therapeutics to be tested, while averting the ethical implications and high cost associated with in vivo preclinical models. This review aims at serving as a practical guide by delineating the OS TME ("what it is like") and, in turn, propose various biofabrication strategies to create a 3D model ("how to recreate it"), to improve the in vitro representation of the OS tumor and ultimately generate more accurate drug response profiles.
Collapse
Affiliation(s)
- Thomas Chow
- Melbourne Medical School, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Australia.,BioFab3D-ACMD, St Vincent's Hospital Melbourne, Fitzroy, Australia
| | - Ilycia Wutami
- Melbourne Medical School, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Australia.,BioFab3D-ACMD, St Vincent's Hospital Melbourne, Fitzroy, Australia
| | - Enrico Lucarelli
- Unit of Orthopaedic Pathology and Osteoarticular Tissue Regeneration, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Peter F Choong
- BioFab3D-ACMD, St Vincent's Hospital Melbourne, Fitzroy, Australia.,Department of Surgery, The University of Melbourne, St Vincent's Hospital Melbourne, Fitzroy, Australia.,Department of Orthopaedics, St Vincent's Hospital Melbourne, Fitzroy, Australia
| | - Serena Duchi
- BioFab3D-ACMD, St Vincent's Hospital Melbourne, Fitzroy, Australia.,Department of Surgery, The University of Melbourne, St Vincent's Hospital Melbourne, Fitzroy, Australia
| | - Claudia Di Bella
- BioFab3D-ACMD, St Vincent's Hospital Melbourne, Fitzroy, Australia.,Department of Surgery, The University of Melbourne, St Vincent's Hospital Melbourne, Fitzroy, Australia.,Department of Orthopaedics, St Vincent's Hospital Melbourne, Fitzroy, Australia
| |
Collapse
|
14
|
Sarkar S, Peng CC, Tung YC. Comparison of VEGF-A secretion from tumor cells under cellular stresses in conventional monolayer culture and microfluidic three-dimensional spheroid models. PLoS One 2020; 15:e0240833. [PMID: 33175874 PMCID: PMC7657494 DOI: 10.1371/journal.pone.0240833] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 10/02/2020] [Indexed: 01/05/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) is a major cytokine in tumor biology affecting tumor survival, aggressiveness and pro-angiogenetic activities. In addition, cellular stresses often result in aggressive pro-angiogenetic behavior in tumors. For in vitro study, conventional monolayer cell culture has been broadly exploited; however, it often provides limited information due to its different microenvironment from that in vivo. Recently, three-dimensional (3D) cell spheroid culture provides in vivo-like microenvironments to study tumor biology and their survival mechanisms with better predictive power. In this work, vascular endothelial growth factor of type A (VEGF-A) secretion from osteosarcoma (MG-63) cells cultured using monolayer and 3D spheroid models under two stress conditions: nutrient deficiency (reduced serum culture) and hypoxia-inducible factor (HIF) inhibition (HIF inhibitor, YC-1) are characterized and systematically compared. In order to obtain ample sample size for consistent characterization of cellular responses from cancer spheroids under the stresses and compare the responses to those from the conventional monolayer model, a microfluidic spheroid formation and culture device is utilized in the experiments. In the analysis, cell viability is estimated from captured images, and quantification of VEGF-A secreted from the cells is achieved using enzyme-linked immunosorbent assay (ELISA). The experimental results show that the viabilities decrease when the cells face higher stress levels in both monolayer and 3D spheroid culture models; however, the VEGF-A secretion profiles between the cell culture models are different. The VEGF-A secretion decreases when the cells face higher stress conditions in the monolayer cell culture. In contrast, for the 3D spheroid culture, the VEGF-A concentration decreases for low stress levels but increases while the stress level is high. The VEGF-A regulation in the 3D models mimics in vivo cases of tumor survival and can provide insightful information to investigate tumor angiogenesis in vitro. The approach developed in this paper provides an efficient method to quantitatively and statistically study tumor growth kinetics and stress responses from highly uniform samples and it can also be applied to compare the underlying biomolecular mechanisms in monolayer and 3D spheroid culture models to elucidate the effects of microenvironments on cellular response in cancer research.
Collapse
Affiliation(s)
- Sreerupa Sarkar
- Department of Engineering and System Science, National Tsing Hua University, Hsinchu, Taiwan
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan
- Taiwan International Graduate Program (TIGP), Nano Science and Technology Program, Academia Sinica, Taipei, Taiwan
| | - Chien-Chung Peng
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan
| | - Yi-Chung Tung
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan
- Taiwan International Graduate Program (TIGP), Nano Science and Technology Program, Academia Sinica, Taipei, Taiwan
- College of Engineering, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
15
|
Cui J, Dean D, Hornicek FJ, Chen Z, Duan Z. The role of extracelluar matrix in osteosarcoma progression and metastasis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:178. [PMID: 32887645 PMCID: PMC7650219 DOI: 10.1186/s13046-020-01685-w] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 08/20/2020] [Indexed: 02/07/2023]
Abstract
Osteosarcoma (OS) is the most common primary bone malignancy and responsible for considerable morbidity and mortality due to its high rates of pulmonary metastasis. Although neoadjuvant chemotherapy has improved 5-year survival rates for patients with localized OS from 20% to over 65%, outcomes for those with metastasis remain dismal. In addition, therapeutic regimens have not significantly improved patient outcomes over the past four decades, and metastases remains a primary cause of death and obstacle in curative therapy. These limitations in care have given rise to numerous works focused on mechanisms and novel targets of OS pathogenesis, including tumor niche factors. OS is notable for its hallmark production of rich extracellular matrix (ECM) of osteoid that goes beyond simple physiological growth support. The aberrant signaling and structural components of the ECM are rich promoters of OS development, and very recent works have shown the specific pathogenic phenotypes induced by these macromolecules. Here we summarize the current developments outlining how the ECM contributes to OS progression and metastasis with supporting mechanisms. We also illustrate the potential of tumorigenic ECM elements as prognostic biomarkers and therapeutic targets in the evolving clinical management of OS.
Collapse
Affiliation(s)
- Juncheng Cui
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, 69 Chuanshan Road, Hengyang, 421001, Hunan, China.,Department of Orthopedic Surgery, Sarcoma Biology Laboratory, David Geffen School of Medicine at UCLA, 615 Charles E. Young Dr. South, Los Angeles, CA, 90095, USA
| | - Dylan Dean
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, David Geffen School of Medicine at UCLA, 615 Charles E. Young Dr. South, Los Angeles, CA, 90095, USA
| | - Francis J Hornicek
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, David Geffen School of Medicine at UCLA, 615 Charles E. Young Dr. South, Los Angeles, CA, 90095, USA
| | - Zhiwei Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, 69 Chuanshan Road, Hengyang, 421001, Hunan, China.
| | - Zhenfeng Duan
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, David Geffen School of Medicine at UCLA, 615 Charles E. Young Dr. South, Los Angeles, CA, 90095, USA.
| |
Collapse
|
16
|
Laranga R, Duchi S, Ibrahim T, Guerrieri AN, Donati DM, Lucarelli E. Trends in Bone Metastasis Modeling. Cancers (Basel) 2020; 12:E2315. [PMID: 32824479 PMCID: PMC7464021 DOI: 10.3390/cancers12082315] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/11/2020] [Accepted: 08/14/2020] [Indexed: 12/12/2022] Open
Abstract
Bone is one of the most common sites for cancer metastasis. Bone tissue is composed by different kinds of cells that coexist in a coordinated balance. Due to the complexity of bone, it is impossible to capture the intricate interactions between cells under either physiological or pathological conditions. Hence, a variety of in vivo and in vitro approaches have been developed. Various models of tumor-bone diseases are routinely used to provide valuable information on the relationship between metastatic cancer cells and the bone tissue. Ideally, when modeling the metastasis of human cancers to bone, models would replicate the intra-tumor heterogeneity, as well as the genetic and phenotypic changes that occur with human cancers; such models would be scalable and reproducible to allow high-throughput investigation. Despite the continuous progress, there is still a lack of solid, amenable, and affordable models that are able to fully recapitulate the biological processes happening in vivo, permitting a correct interpretation of results. In the last decades, researchers have demonstrated that three-dimensional (3D) methods could be an innovative approach that lies between bi-dimensional (2D) models and animal models. Scientific evidence supports that the tumor microenvironment can be better reproduced in a 3D system than a 2D cell culture, and the 3D systems can be scaled up for drug screening in the same way as the 2D systems thanks to the current technologies developed. However, 3D models cannot completely recapitulate the inter- and intra-tumor heterogeneity found in patients. In contrast, ex vivo cultures of fragments of bone preserve key cell-cell and cell-matrix interactions and allow the study of bone cells in their natural 3D environment. Moreover, ex vivo bone organ cultures could be a better model to resemble the human pathogenic metastasis condition and useful tools to predict in vivo response to therapies. The aim of our review is to provide an overview of the current trends in bone metastasis modeling. By showing the existing in vitro and ex vivo systems, we aspire to contribute to broaden the knowledge on bone metastasis models and make these tools more appealing for further translational studies.
Collapse
Affiliation(s)
- Roberta Laranga
- Unit of Orthopaedic Pathology and Osteoarticular Tissue Regeneration, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (R.L.); (D.M.D.); (E.L.)
| | - Serena Duchi
- BioFab3D@ACMD, St Vincent’s Hospital, Melbourne, VIC 3065, Australia;
- Department of Surgery, St Vincent’s Hospital, University of Melbourne, Melbourne, VIC 3065, Australia
| | - Toni Ibrahim
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy;
| | - Ania Naila Guerrieri
- Unit of Orthopaedic Pathology and Osteoarticular Tissue Regeneration, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (R.L.); (D.M.D.); (E.L.)
| | - Davide Maria Donati
- Unit of Orthopaedic Pathology and Osteoarticular Tissue Regeneration, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (R.L.); (D.M.D.); (E.L.)
- Rizzoli Laboratory Unit, Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum University of Bologna, Via di Barbiano 1/10, 40136 Bologna, Italy
- 3rd Orthopaedic and Traumatologic Clinic Prevalently Oncologic, IRCCS Istituto Ortopedico Rizzoli, Via Pupilli 1, 40136 Bologna, Italy
| | - Enrico Lucarelli
- Unit of Orthopaedic Pathology and Osteoarticular Tissue Regeneration, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (R.L.); (D.M.D.); (E.L.)
| |
Collapse
|
17
|
The Role of Pre-Clinical 3-Dimensional Models of Osteosarcoma. Int J Mol Sci 2020; 21:ijms21155499. [PMID: 32752092 PMCID: PMC7432883 DOI: 10.3390/ijms21155499] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 07/29/2020] [Accepted: 07/30/2020] [Indexed: 11/17/2022] Open
Abstract
Treatment for osteosarcoma (OS) has been largely unchanged for several decades, with typical therapies being a mixture of chemotherapy and surgery. Although therapeutic targets and products against cancer are being continually developed, only a limited number have proved therapeutically active in OS. Thus, the understanding of the OS microenvironment and its interactions are becoming more important in developing new therapies. Three-dimensional (3D) models are important tools in increasing our understanding of complex mechanisms and interactions, such as in OS. In this review, in vivo animal models, in vitro 3D models and in ovo chorioallantoic membrane (CAM) models, are evaluated and discussed as to their contribution in understanding the progressive nature of OS, and cancer research. We aim to provide insight and prospective future directions into the potential translation of 3D models in OS.
Collapse
|
18
|
Molina ER, Chim LK, Barrios S, Ludwig JA, Mikos AG. Modeling the Tumor Microenvironment and Pathogenic Signaling in Bone Sarcoma. TISSUE ENGINEERING. PART B, REVIEWS 2020; 26:249-271. [PMID: 32057288 PMCID: PMC7310212 DOI: 10.1089/ten.teb.2019.0302] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 01/07/2020] [Indexed: 12/17/2022]
Abstract
Investigations of cancer biology and screening of potential therapeutics for efficacy and safety begin in the preclinical laboratory setting. A staple of most basic research in cancer involves the use of tissue culture plates, on which immortalized cell lines are grown in monolayers. However, this practice has been in use for over six decades and does not account for vital elements of the tumor microenvironment that are thought to aid in initiation, propagation, and ultimately, metastasis of cancer. Furthermore, information gleaned from these techniques does not always translate to animal models or, more crucially, clinical trials in cancer patients. Osteosarcoma (OS) and Ewing sarcoma (ES) are the most common primary tumors of bone, but outcomes for patients with metastatic or recurrent disease have stagnated in recent decades. The unique elements of the bone tumor microenvironment have been shown to play critical roles in the pathogenesis of these tumors and thus should be incorporated in the preclinical models of these diseases. In recent years, the field of tissue engineering has leveraged techniques used in designing scaffolds for regenerative medicine to engineer preclinical tumor models that incorporate spatiotemporal control of physical and biological elements. We herein review the clinical aspects of OS and ES, critical elements present in the sarcoma microenvironment, and engineering approaches to model the bone tumor microenvironment. Impact statement The current paradigm of cancer biology investigation and therapeutic testing relies heavily on monolayer, monoculture methods developed over half a century ago. However, these methods often lack essential hallmarks of the cancer microenvironment that contribute to tumor pathogenesis. Tissue engineers incorporate scaffolds, mechanical forces, cells, and bioactive signals into biological environments to drive cell phenotype. Investigators of bone sarcomas, aggressive tumors that often rob patients of decades of life, have begun to use tissue engineering techniques to devise in vitro models for these diseases. Their efforts highlight how critical elements of the cancer microenvironment directly affect tumor signaling and pathogenesis.
Collapse
Affiliation(s)
- Eric R. Molina
- Department of Bioengineering, Rice University, Houston, Texas
| | - Letitia K. Chim
- Department of Bioengineering, Rice University, Houston, Texas
| | - Sergio Barrios
- Department of Bioengineering, Rice University, Houston, Texas
| | - Joseph A. Ludwig
- Division of Cancer Medicine, Department of Sarcoma Medical Oncology, MD Anderson Cancer Center, The University of Texas, Houston, Texas
| | | |
Collapse
|
19
|
Komez A, Buyuksungur A, Antmen E, Swieszkowski W, Hasirci N, Hasirci V. A two-compartment bone tumor model to investigate interactions between healthy and tumor cells. ACTA ACUST UNITED AC 2020; 15:035007. [PMID: 31935707 DOI: 10.1088/1748-605x/ab6b31] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We produced a novel three-dimensional (3D) bone tumor model (BTM) to study the interactions between healthy and tumor cells in a tumor microenvironment, the migration tendency of the tumor cells, and the efficacy of an anticancer drug, Doxorubicin, on the cancer cells. The model consisted of two compartments: (a) a healthy bone tissue mimic, made of poly(lactic acid-co-glycolic acid) (PLGA)/beta-tricalcium phosphate (β-TCP) sponge seeded with human fetal osteoblastic cells (hFOB) and human umbilical vein endothelial cells (HUVECs), and (b) a tumor mimic, made of lyophilized collagen sponge seeded with human osteosarcoma cells (Saos-2). The tumor mimic component was placed into a central cavity created in the healthy bone mimic and together they constituted the complete 3D bone tumor model (3D-BTM). The porosities of both sponges were higher than 85% and the diameters of the pores were 199 ± 52 μm for the PLGA/TCP and 50-150 μm for the collagen scaffolds. The compression Young's modulus of the PLGA/TCP and the collagen sponges were determined to be 4.76 MPa and 140 kPa, respectively. Cell proliferation, morphology, calcium phosphate forming capacity and alkaline phosphatase production were studied separately on both the healthy and tumor mimics. All cells demonstrated cellular extensions and spread well in porous scaffolds indicating good cell-material interactions. Confocal microscopy analysis showed direct contact between the cells present in different parts of the 3D-BTM. Migration of HUVECs from the healthy bone mimic to the tumor compartment was confirmed by the increase in the levels of angiogenic factors vascular endothelial growth factor, basic fibroblast growth factor, and interleukin 8 in the tumor component. Doxorubicin (2.7 μg.ml-1) administered to the 3D-BTM caused a seven-fold decrease in the cell number after 24 h of interaction with the anticancer drug. Caspase-3 enzyme activity assay results demonstrated apoptosis of the osteosarcoma cells. This novel 3D-BTM has a high potential for use in studying the metastatic capabilities of cancer cells, and in determining the effective drug types and combinations for personalized treatments.
Collapse
Affiliation(s)
- Aylin Komez
- Graduate Department of Biotechnology, Middle East Technical University (METU), Ankara, 06800, Turkey. BIOMATEN, METU Center of Excellence in Biomaterials and Tissue Engineering, Ankara, 06800, Turkey
| | | | | | | | | | | |
Collapse
|
20
|
Fan TM, Roberts RD, Lizardo MM. Understanding and Modeling Metastasis Biology to Improve Therapeutic Strategies for Combating Osteosarcoma Progression. Front Oncol 2020; 10:13. [PMID: 32082995 PMCID: PMC7006476 DOI: 10.3389/fonc.2020.00013] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/07/2020] [Indexed: 12/12/2022] Open
Abstract
Osteosarcoma is a malignant primary tumor of bone, arising from transformed progenitor cells with osteoblastic differentiation and osteoid production. While categorized as a rare tumor, most patients diagnosed with osteosarcoma are adolescents in their second decade of life and underscores the potential for life changing consequences in this vulnerable population. In the setting of localized disease, conventional treatment for osteosarcoma affords a cure rate approaching 70%; however, survival for patients suffering from metastatic disease remain disappointing with only 20% of individuals being alive past 5 years post-diagnosis. In patients with incurable disease, pulmonary metastases remain the leading cause for osteosarcoma-associated mortality; yet identifying new strategies for combating metastatic progression remains at a scientific and clinical impasse, with no significant advancements for the past four decades. While there is resonating clinical urgency for newer and more effective treatment options for managing osteosarcoma metastases, the discovery of druggable targets and development of innovative therapies for inhibiting metastatic progression will require a deeper and more detailed understanding of osteosarcoma metastasis biology. Toward the goal of illuminating the processes involved in cancer metastasis, a convergent science approach inclusive of diverse disciplines spanning the biology and physical science domains can offer novel and synergistic perspectives, inventive, and sophisticated model systems, and disruptive experimental approaches that can accelerate the discovery and characterization of key processes operative during metastatic progression. Through the lens of trans-disciplinary research, the field of comparative oncology is uniquely positioned to advance new discoveries in metastasis biology toward impactful clinical translation through the inclusion of pet dogs diagnosed with metastatic osteosarcoma. Given the spontaneous course of osteosarcoma development in the context of real-time tumor microenvironmental cues and immune mechanisms, pet dogs are distinctively valuable in translational modeling given their faithful recapitulation of metastatic disease progression as occurs in humans. Pet dogs can be leveraged for the exploration of novel therapies that exploit tumor cell vulnerabilities, perturb local microenvironmental cues, and amplify immunologic recognition. In this capacity, pet dogs can serve as valuable corroborative models for realizing the science and best clinical practices necessary for understanding and combating osteosarcoma metastases.
Collapse
Affiliation(s)
- Timothy M Fan
- Comparative Oncology Research Laboratory, Department of Veterinary Clinical Medicine, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Ryan D Roberts
- Center for Childhood Cancer and Blood Disorders, Abigail Wexner Research Institute at Nationwide Children's Hospital, The James Comprehensive Cancer Center at The Ohio State University, Columbus, OH, United States
| | - Michael M Lizardo
- Poul Sorensen Laboratory, Department of Molecular Oncology, BC Cancer, Part of the Provincial Health Services Authority in British Columbia, Vancouver, BC, Canada
| |
Collapse
|
21
|
Cortini M, Baldini N, Avnet S. New Advances in the Study of Bone Tumors: A Lesson From the 3D Environment. Front Physiol 2019; 10:814. [PMID: 31316395 PMCID: PMC6611422 DOI: 10.3389/fphys.2019.00814] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 06/11/2019] [Indexed: 01/10/2023] Open
Abstract
Bone primary tumors, such as osteosarcoma, are highly aggressive pediatric tumors that in 30% of the cases develop lung metastasis and are characterized by poor prognosis. Bone is also the third most common metastatic site in patients with advanced cancer and once tumor cells become homed to the skeleton, the disease is usually considered incurable, and treatment is only palliative. Bone sarcoma and bone metastasis share the same tissue microenvironment and niches. 3D cultures represent a new promising approach for the study of interactions between tumor cells and other cellular or acellular components of the tumor microenvironment (i.e., fibroblasts, mesenchymal stem cells, bone ECM). Indeed, 3D models can mimic physiological interactions that are crucial to modulate response to soluble paracrine factors, tumor drug resistance and aggressiveness and, in all, these innovative models might be able of bypassing the use of animal-based preclinical cancer models. To date, both static and dynamic 3D cell culture models have been shown to be particularly suited for screening of anticancer agents and might provide accurate information, translating in vitro cell cultures into precision medicine. In this mini-review, we will summarize the current state-of-the-art in the field of bone tumors, both primary and metastatic, illustrating the different methods and techniques employed to realize 3D cell culture systems and new results achieved in a field that paves the way toward personalized medicine.
Collapse
Affiliation(s)
- Margherita Cortini
- Orthopaedic Pathophysiology and Regenerative Medicine Unit, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Nicola Baldini
- Orthopaedic Pathophysiology and Regenerative Medicine Unit, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Sofia Avnet
- Orthopaedic Pathophysiology and Regenerative Medicine Unit, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| |
Collapse
|
22
|
Chramiec A, Vunjak-Novakovic G. Tissue engineered models of healthy and malignant human bone marrow. Adv Drug Deliv Rev 2019; 140:78-92. [PMID: 31002835 PMCID: PMC6663611 DOI: 10.1016/j.addr.2019.04.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 02/14/2019] [Accepted: 04/14/2019] [Indexed: 12/20/2022]
Abstract
Tissue engineering is becoming increasingly successful in providing in vitro models of human tissues that can be used for ex vivo recapitulation of functional tissues as well as predictive testing of drug efficacy and safety. From simple tissue models to microphysiological platforms comprising multiple tissue types connected by vascular perfusion, these "tissues on a chip" are emerging as a fast track application for tissue engineering, with great potential for modeling diseases and supporting the development of new drugs and therapeutic targets. We focus here on tissue engineering of the hematopoietic stem and progenitor cell compartment and the malignancies that can develop in the human bone marrow. Our overall goal is to demonstrate the utility and interconnectedness of improvements in bioengineering methods developed in one area of bone marrow studies for the remaining, seemingly disparate, bone marrow fields.
Collapse
|
23
|
Monteiro CF, Custódio CA, Mano JF. Three-Dimensional Osteosarcoma Models for Advancing Drug Discovery and Development. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800108] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Cátia F. Monteiro
- Department of Chemistry, CICECO; University of Aveiro, Campus Universitário de Santiago; 3810-193 Aveiro Portugal
| | - Catarina A. Custódio
- Department of Chemistry, CICECO; University of Aveiro, Campus Universitário de Santiago; 3810-193 Aveiro Portugal
| | - João F. Mano
- Department of Chemistry, CICECO; University of Aveiro, Campus Universitário de Santiago; 3810-193 Aveiro Portugal
| |
Collapse
|
24
|
Bugueno IM, Batool F, Keller L, Kuchler-Bopp S, Benkirane-Jessel N, Huck O. Porphyromonas gingivalis bypasses epithelial barrier and modulates fibroblastic inflammatory response in an in vitro 3D spheroid model. Sci Rep 2018; 8:14914. [PMID: 30297793 PMCID: PMC6175856 DOI: 10.1038/s41598-018-33267-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 09/25/2018] [Indexed: 02/07/2023] Open
Abstract
Porphyromonas gingivalis-induced inflammatory effects are mostly investigated in monolayer cultured cells. The aim of this study was to develop a 3D spheroid model of gingiva to take into account epithelio-fibroblastic interactions. Human gingival epithelial cells (ECs) and human oral fibroblasts (FBs) were cultured by hanging drop method to generate 3D microtissue (MT) whose structure was analyzed on histological sections and the cell-to-cell interactions were observed by scanning and transmission electron microscopy (SEM and TEM). MTs were infected by P. gingivalis and the impact on cell death (Apaf-1, caspase-3), inflammatory markers (TNF-α, IL-6, IL-8) and extracellular matrix components (Col-IV, E-cadherin, integrin β1) was evaluated by immunohistochemistry and RT-qPCR. Results were compared to those observed in situ in experimental periodontitis and in human gingival biopsies. MTs exhibited a well-defined spatial organization where ECs were organized in an external cellular multilayer, while, FBs constituted the core. The infection of MT demonstrated the ability of P. gingivalis to bypass the epithelial barrier in order to reach the fibroblastic core and induce disorganization of the spheroid structure. An increased cell death was observed in fibroblastic core. The development of such 3D model may be useful to define the role of EC–FB interactions on periodontal host-immune response and to assess the efficacy of new therapeutics.
Collapse
Affiliation(s)
- Isaac Maximiliano Bugueno
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), Fédération de Médecine Translationnelle de Strasbourg (FMTS), 11 rue Humann, Strasbourg, 67000, France.,Université de Strasbourg (UDS), Faculté de Chirurgie-dentaire, 8 rue Sainte-Elisabeth, Strasbourg, 67000, France
| | - Fareeha Batool
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), Fédération de Médecine Translationnelle de Strasbourg (FMTS), 11 rue Humann, Strasbourg, 67000, France.,Université de Strasbourg (UDS), Faculté de Chirurgie-dentaire, 8 rue Sainte-Elisabeth, Strasbourg, 67000, France
| | - Laetitia Keller
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), Fédération de Médecine Translationnelle de Strasbourg (FMTS), 11 rue Humann, Strasbourg, 67000, France.,Université de Strasbourg (UDS), Faculté de Chirurgie-dentaire, 8 rue Sainte-Elisabeth, Strasbourg, 67000, France
| | - Sabine Kuchler-Bopp
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), Fédération de Médecine Translationnelle de Strasbourg (FMTS), 11 rue Humann, Strasbourg, 67000, France
| | - Nadia Benkirane-Jessel
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), Fédération de Médecine Translationnelle de Strasbourg (FMTS), 11 rue Humann, Strasbourg, 67000, France.,Université de Strasbourg (UDS), Faculté de Chirurgie-dentaire, 8 rue Sainte-Elisabeth, Strasbourg, 67000, France
| | - Olivier Huck
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), Fédération de Médecine Translationnelle de Strasbourg (FMTS), 11 rue Humann, Strasbourg, 67000, France. .,Université de Strasbourg (UDS), Faculté de Chirurgie-dentaire, 8 rue Sainte-Elisabeth, Strasbourg, 67000, France. .,Hôpitaux Universitaires de Strasbourg (HUS), Department of Periodontology, 1 place de l'Hôpital, Strasbourg, 67000, France.
| |
Collapse
|
25
|
Sarcoma Spheroids and Organoids-Promising Tools in the Era of Personalized Medicine. Int J Mol Sci 2018; 19:ijms19020615. [PMID: 29466296 PMCID: PMC5855837 DOI: 10.3390/ijms19020615] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 02/13/2018] [Accepted: 02/16/2018] [Indexed: 02/06/2023] Open
Abstract
Cancer treatment is rapidly evolving toward personalized medicine, which takes into account the individual molecular and genetic variability of tumors. Sophisticated new in vitro disease models, such as three-dimensional cell cultures, may provide a tool for genetic, epigenetic, biomedical, and pharmacological research, and help determine the most promising individual treatment. Sarcomas, malignant neoplasms originating from mesenchymal cells, may have a multitude of genomic aberrations that give rise to more than 70 different histopathological subtypes. Their low incidence and high level of histopathological heterogeneity have greatly limited progress in their treatment, and trials of clinical sarcoma are less frequent than trials of other carcinomas. The main advantage of 3D cultures from tumor cells or biopsy is that they provide patient-specific models of solid tumors, and they overcome some limitations of traditional 2D monolayer cultures by reflecting cell heterogeneity, native histologic architectures, and cell-extracellular matrix interactions. Recent advances promise that these models can help bridge the gap between preclinical and clinical research by providing a relevant in vitro model of human cancer useful for drug testing and studying metastatic and dormancy mechanisms. However, additional improvements of 3D models are expected in the future, specifically the inclusion of tumor vasculature and the immune system, to enhance their full ability to capture the biological features of native tumors in high-throughput screening. Here, we summarize recent advances and future perspectives of spheroid and organoid in vitro models of rare sarcomas that can be used to investigate individual molecular biology and predict clinical responses. We also highlight how spheroid and organoid culture models could facilitate the personalization of sarcoma treatment, provide specific clinical scenarios, and discuss the relative strengths and limitations of these models.
Collapse
|
26
|
De Luca A, Raimondi L, Salamanna F, Carina V, Costa V, Bellavia D, Alessandro R, Fini M, Giavaresi G. Relevance of 3d culture systems to study osteosarcoma environment. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:2. [PMID: 29304852 PMCID: PMC5756329 DOI: 10.1186/s13046-017-0663-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 12/11/2017] [Indexed: 12/21/2022]
Abstract
Osteosarcoma (OS) is the most common primary malignant tumor of bone, which preferentially develops lung metastasis. Although standard chemotherapy has significantly improved long-term survival over the past few decades, the outcome for patients with metastatic or recurrent OS remains dramatically poor. Novel therapies are therefore required to slow progression and eradicate the disease. Furthermore, to better understand the cellular and molecular mechanisms responsible for OS onset and progression, the development of novel predictive culture systems resembling the native three-dimensional (3D) tumor microenvironment are mandatory. ‘Tumor engineering’ approaches radically changed the previous scenario, through the development of advanced and alternative 3D cell culture in vitro models able to tightly mimic the in vivo tumor microenvironment. In this review, we will summarize the state of the art in this novel area, illustrating the different methods and techniques employed to realize 3D OS cell culture models and we report the achieved results, which highlight the efficacy of these models in reproducing the tumor milieu. Although data need to be further validated, the scientific studies reviewed here are certainly promising and give new insights into the clinical practice.
Collapse
Affiliation(s)
- Angela De Luca
- IRCCS Rizzoli Orthopedic Institute, Bologna, Italy. .,IRCCS Rizzoli Orthopedic Institute, Innovative Technologic Platform for Tissue Engineering, Theranostics and Oncology, Via Divisi, 83, 90133, Palermo, Italy.
| | - Lavinia Raimondi
- IRCCS Rizzoli Orthopedic Institute, Bologna, Italy.,IRCCS Rizzoli Orthopedic Institute, Innovative Technologic Platform for Tissue Engineering, Theranostics and Oncology, Via Divisi, 83, 90133, Palermo, Italy
| | | | - Valeria Carina
- IRCCS Rizzoli Orthopedic Institute, Bologna, Italy.,IRCCS Rizzoli Orthopedic Institute, Innovative Technologic Platform for Tissue Engineering, Theranostics and Oncology, Via Divisi, 83, 90133, Palermo, Italy
| | - Viviana Costa
- IRCCS Rizzoli Orthopedic Institute, Bologna, Italy.,IRCCS Rizzoli Orthopedic Institute, Innovative Technologic Platform for Tissue Engineering, Theranostics and Oncology, Via Divisi, 83, 90133, Palermo, Italy
| | - Daniele Bellavia
- IRCCS Rizzoli Orthopedic Institute, Bologna, Italy.,IRCCS Rizzoli Orthopedic Institute, Innovative Technologic Platform for Tissue Engineering, Theranostics and Oncology, Via Divisi, 83, 90133, Palermo, Italy
| | - Riccardo Alessandro
- Biology and Genetics Unit, Department of Biopathology and Medical Biotechnology, University of Palermo, Palermo, Italy.,Institute of Biomedicine and Molecular Immunology (IBIM), National Research Council, Palermo, Italy
| | - Milena Fini
- IRCCS Rizzoli Orthopedic Institute, Laboratory BITTA, Bologna, Italy.,IRCCS Rizzoli Orthopedic Institute, Laboratory of Preclinical and Surgical Studies, Bologna, Italy
| | - Gianluca Giavaresi
- IRCCS Rizzoli Orthopedic Institute, Innovative Technologic Platform for Tissue Engineering, Theranostics and Oncology, Via Divisi, 83, 90133, Palermo, Italy.,IRCCS Rizzoli Orthopedic Institute, Laboratory of Preclinical and Surgical Studies, Bologna, Italy
| |
Collapse
|