1
|
Brunmaier LAE, Ozdemir T, Walker TW. Angiogenesis: Biological Mechanisms and In Vitro Models. Ann Biomed Eng 2025:10.1007/s10439-025-03721-2. [PMID: 40210793 DOI: 10.1007/s10439-025-03721-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/25/2025] [Indexed: 04/12/2025]
Abstract
The translation of biomedical devices and drug research is an expensive and long process with a low probability of receiving FDA approval. Developing physiologically relevant in vitro models with human cells offers a solution to not only improving the odds of FDA approval but also to expand our ability to study complex in vivo systems in a simpler fashion. Animal models remain the standard for pre-clinical testing; however, the data from animal models is an unreliable extrapolation when anticipating a human response in clinical trials, thus contributing to the low rates of translation. In this review, we focus on in vitro vascular or angiogenic models because of the incremental role that the vascular system plays in the translation of biomedical research. The first section of this review discusses the most common angiogenic cytokines that are used in vitro to initiate angiogenesis, followed by angiogenic inhibitors where both initiators and inhibitors work to maintain vascular homeostasis. Next, we evaluate previously published in vitro models, where we evaluate capturing the physical environment for biomimetic in vitro modeling. These topics provide a foundation of parameters that must be considered to improve and achieve vascular biomimicry. Finally, we summarize these topics to suggest a path forward with the goal of engineering human in vitro models that emulate the in vivo environment and provide a platform for biomedical device and drug screening that produces data to support clinical translation.
Collapse
Affiliation(s)
- Laura A E Brunmaier
- Nanoscience and Biomedical Engineering Department, South Dakota School of Mines & Technology, 501 E St. Joseph St., Rapid City, SD, 57701, USA
| | - Tugba Ozdemir
- Nanoscience and Biomedical Engineering Department, South Dakota School of Mines & Technology, 501 E St. Joseph St., Rapid City, SD, 57701, USA
| | - Travis W Walker
- Karen M. Swindler Department of Chemical and Biological Engineering, South Dakota School of Mines & Technology, 501 E St. Joseph St., Rapid City, SD, 57701, USA.
| |
Collapse
|
2
|
Zhou R, Brislinger D, Fuchs J, Lyons A, Langthaler S, Hauser CAE, Baumgartner C. Vascularised organoids: Recent advances and applications in cancer research. Clin Transl Med 2025; 15:e70258. [PMID: 40045486 PMCID: PMC11882480 DOI: 10.1002/ctm2.70258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 03/09/2025] Open
Abstract
Organoids are three-dimensional (3D) cellular models designed to replicate human tissues and organs while preserving their physiological complexity and functionality. Among these, vascularised organoids represent a groundbreaking advancement in 3D tissue engineering, incorporating vascular networks into engineered tissues to more accurately mimic the in vivo tumour microenvironment. These models offer significantly improved physiological relevance compared to conventional two-dimensional cultures or animal models, positioning them as invaluable tools in cancer research. Despite their potential, the rapid proliferation of techniques and materials for developing vascularised organoids presents challenges for researchers navigating this dynamic field. This systematic review provides a comprehensive examination of methodologies for fabricating vascularised organoids, with a focus on strategies that enhance vascularisation and support organoid growth. It critically evaluates the materials used, emphasising those that effectively mimic the extracellular matrix and facilitate vascular network formation. Key advancements in engineered organoids models are highlighted, emphasising their potential for studying interactions between vasculature and cancer cells, conducting drug screening, and understanding cytokine regulation. In summary, this review provides an in-depth overview of the current landscape of vascularised organoid fabrication and functionality, addressing challenges and opportunities within the field. A detailed understanding of the scope and future trajectories is essential for advancing organoid development and expanding their applications in both basic cancer research and clinical practice. KEY POINTS: Comparative analysis: Evaluation of organoids, animal models, and 2D models, highlighting their respective strengths and limitations in replicating physiological conditions and studying disease processes. Vascularisation techniques: Comparative evaluation of vascularised organoid fabrication methods, emphasising their efficiency, scalability and ability to replicate physiological vascular networks. Material selection: Thorough evaluation of materials for vascularised organoid culture system, focusing on those that effectively mimic the extracellular matrix and support vascular network formation. Applications: Overview of organoid applications in basic cancer research and clinical settings, with an emphasis on their potential in drug discovery, disease modelling and exploring complex biological processes.
Collapse
Affiliation(s)
- Rui Zhou
- Institute of Health Care Engineering with European Testing Center of Medical DevicesGraz University of TechnologyGrazAustria
| | - Dagmar Brislinger
- Department of Cell BiologyHistology and EmbryologyGottfried Schatz Research CenterMedical University of GrazGrazAustria
| | - Julia Fuchs
- Institute of Health Care Engineering with European Testing Center of Medical DevicesGraz University of TechnologyGrazAustria
- Department of Cell BiologyHistology and EmbryologyGottfried Schatz Research CenterMedical University of GrazGrazAustria
| | - Alicia Lyons
- Institute of Health Care Engineering with European Testing Center of Medical DevicesGraz University of TechnologyGrazAustria
| | - Sonja Langthaler
- Institute of Health Care Engineering with European Testing Center of Medical DevicesGraz University of TechnologyGrazAustria
| | - Charlotte A. E. Hauser
- Institute of Health Care Engineering with European Testing Center of Medical DevicesGraz University of TechnologyGrazAustria
| | - Christian Baumgartner
- Institute of Health Care Engineering with European Testing Center of Medical DevicesGraz University of TechnologyGrazAustria
| |
Collapse
|
3
|
Cantoni F, Barbe L, Roy A, Wicher G, Simonsson S, Forsberg-Nilsson K, Tenje M. On-chip fabrication of tailored 3D hydrogel scaffolds to model cancer cell invasion and interaction with endothelial cells. APL Bioeng 2024; 8:046113. [PMID: 39634677 PMCID: PMC11617029 DOI: 10.1063/5.0227135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 12/09/2024] [Accepted: 11/15/2024] [Indexed: 12/07/2024] Open
Abstract
The high mortality associated with certain cancers can be attributed to the invasive nature of the tumor cells. Yet, the complexity of studying invasion hinders our understanding of how the tumor spreads. This work presents a microengineered three-dimensional (3D) in vitro model for studying cancer cell invasion and interaction with endothelial cells. The model was generated by printing a biomimetic hydrogel scaffold directly on a chip using 2-photon polymerization that simulates the brain's extracellular matrix. The scaffold's geometry was specifically designed to facilitate the growth of a continuous layer of endothelial cells on one side, while also allowing for the introduction of tumor cells on the other side. This arrangement confines the cells spatially and enables in situ microscopy of the cancer cells as they invade the hydrogel scaffold and interact with the endothelial layer. We examined the impact of 3D printing parameters on the hydrogel's physical properties and used patient derived glioblastoma cells to study their effect on cell invasion. Notably, the tumor cells tended to infiltrate faster when an endothelial cell barrier was present. The potential for adjusting the hydrogel scaffold's properties, coupled with the capability for real-time observation of tumor-endothelial cell interactions, offers a platform for studying tumor invasion and tumor-endothelial cell interactions.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Maria Tenje
- Author to whom correspondence should be addressed:
| |
Collapse
|
4
|
Suurmond CE, Leeuwenburgh SCG, van den Beucken JJJP. Modelling bone metastasis in spheroids to study cancer progression and screen cisplatin efficacy. Cell Prolif 2024; 57:e13693. [PMID: 38899562 PMCID: PMC11503253 DOI: 10.1111/cpr.13693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/27/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024] Open
Abstract
Most bone metastases are caused by primary breast or prostate cancer cells settling in the bone microenvironment, affecting normal bone physiology and function and reducing 5-year survival rates to 10% and 6%, respectively. To expedite clinical availability of novel and effective bone metastases treatments, reliable and predictive in vitro models are urgently required to screen for novel therapies as current in vitro 2D planar mono-culture models do not accurately predict the clinical efficacy. We herein engineered a novel human in vitro 3D co-culture model based on spheroids to study dynamic cellular quantities of (breast or prostate) cancer cells and human bone marrow stromal cells and screen chemotherapeutic efficacy and specificity of the common anticancer drug cisplatin. Bone metastatic spheroids (BMSs) were formed rapidly within 24 h, while the morphology of breast versus prostate cancer BMS differed in terms of size and circularity upon prolonged culture periods. Prestaining cell types prior to BMS formation enabled confocal imaging and quantitative image analysis of in-spheroid cellular dynamics for up to 7 days of BMS culture. We found that cancer cells in BMS proliferated faster and were less susceptible to cisplatin treatment compared to 2D control cultures. Based on these findings and the versatility of our methodology, BMS represent a feasible 3D in vitro model for screening of new bone cancer metastases therapies.
Collapse
|
5
|
Zhang Z, Chen X, Gao S, Fang X, Ren S. 3D bioprinted tumor model: a prompt and convenient platform for overcoming immunotherapy resistance by recapitulating the tumor microenvironment. Cell Oncol (Dordr) 2024; 47:1113-1126. [PMID: 38520648 PMCID: PMC11322267 DOI: 10.1007/s13402-024-00935-9] [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] [Accepted: 03/05/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND Cancer immunotherapy is receiving worldwide attention for its induction of an anti-tumor response. However, it has had limited efficacy in some patients who acquired resistance. The dynamic and sophisticated complexity of the tumor microenvironment (TME) is the leading contributor to this clinical dilemma. Through recapitulating the physiological features of the TME, 3D bioprinting is a promising research tool for cancer immunotherapy, which preserves in vivo malignant aggressiveness, heterogeneity, and the cell-cell/matrix interactions. It has been reported that application of 3D bioprinting holds potential to address the challenges of immunotherapy resistance and facilitate personalized medication. CONCLUSIONS AND PERSPECTIVES In this review, we briefly summarize the contributions of cellular and noncellular components of the TME in the development of immunotherapy resistance, and introduce recent advances in 3D bioprinted tumor models that served as platforms to study the interactions between tumor cells and the TME. By constructing multicellular 3D bioprinted tumor models, cellular and noncellular crosstalk is reproduced between tumor cells, immune cells, fibroblasts, adipocytes, and the extracellular matrix (ECM) within the TME. In the future, by quickly preparing 3D bioprinted tumor models with patient-derived components, information on tumor immunotherapy resistance can be obtained timely for clinical reference. The combined application with tumoroid or other 3D culture technologies will also help to better simulate the complexity and dynamics of tumor microenvironment in vitro. We aim to provide new perspectives for overcoming cancer immunotherapy resistance and inspire multidisciplinary research to improve the clinical application of 3D bioprinting technology.
Collapse
Affiliation(s)
- Zhanyi Zhang
- Bethune Third Clinical Medical College, Jilin University, Changchun, 130021, China
| | - Xuebo Chen
- Department of Gastrointestinal, Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, NO. 126, Xiantai Street, Changchun, 130033, China
| | - Sujie Gao
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Xuedong Fang
- Department of Gastrointestinal, Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, NO. 126, Xiantai Street, Changchun, 130033, China.
| | - Shengnan Ren
- Department of Breast Surgery, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, NO. 519, Kunzhou Street, Kunming, 650118, China.
| |
Collapse
|
6
|
Nuckhir M, Withey D, Cabral S, Harrison H, Clarke RB. State of the Art Modelling of the Breast Cancer Metastatic Microenvironment: Where Are We? J Mammary Gland Biol Neoplasia 2024; 29:14. [PMID: 39012440 PMCID: PMC11252219 DOI: 10.1007/s10911-024-09567-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 05/09/2024] [Indexed: 07/17/2024] Open
Abstract
Metastatic spread of tumour cells to tissues and organs around the body is the most frequent cause of death from breast cancer. This has been modelled mainly using mouse models such as syngeneic mammary cancer or human in mouse xenograft models. These have limitations for modelling human disease progression and cannot easily be used for investigation of drug resistance and novel therapy screening. To complement these approaches, advances are being made in ex vivo and 3D in vitro models, which are becoming progressively better at reliably replicating the tumour microenvironment and will in the future facilitate drug development and screening. These approaches include microfluidics, organ-on-a-chip and use of advanced biomaterials. The relevant tissues to be modelled include those that are frequent and clinically important sites of metastasis such as bone, lung, brain, liver for invasive ductal carcinomas and a distinct set of common metastatic sites for lobular breast cancer. These sites all have challenges to model due to their unique cellular compositions, structure and complexity. The models, particularly in vivo, provide key information on the intricate interactions between cancer cells and the native tissue, and will guide us in producing specific therapies that are helpful in different context of metastasis.
Collapse
Affiliation(s)
- Mia Nuckhir
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Oglesby Cancer Research Building, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M20 4GJ, UK
| | - David Withey
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Oglesby Cancer Research Building, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M20 4GJ, UK
| | - Sara Cabral
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Oglesby Cancer Research Building, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M20 4GJ, UK
| | - Hannah Harrison
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Oglesby Cancer Research Building, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M20 4GJ, UK.
| | - Robert B Clarke
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Oglesby Cancer Research Building, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M20 4GJ, UK.
| |
Collapse
|
7
|
Vinnacombe-Willson GA, García-Astrain C, Troncoso-Afonso L, Wagner M, Langer J, González-Callejo P, Silvio DD, Liz-Marzán LM. Growing Gold Nanostars on 3D Hydrogel Surfaces. CHEMISTRY OF MATERIALS : A PUBLICATION OF THE AMERICAN CHEMICAL SOCIETY 2024; 36:5192-5203. [PMID: 38828187 PMCID: PMC11137816 DOI: 10.1021/acs.chemmater.4c00564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 06/05/2024]
Abstract
Nanocomposites comprising hydrogels and plasmonic nanoparticles are attractive materials for tissue engineering, bioimaging, and biosensing. These materials are usually fabricated by adding colloidal nanoparticles to the uncured polymer mixture and thus require time-consuming presynthesis, purification, and ligand-exchange steps. Herein, we introduce approaches for rapid synthesis of gold nanostars (AuNSt) in situ on hydrogel substrates, including those with complex three-dimensional (3D) features. These methods enable selective AuNSt growth at the surface of the substrate, and the growth conditions can be tuned to tailor the nanoparticle size and density (coverage). We additionally demonstrate proof-of-concept applications of these nanocomposites for SERS sensing and imaging. High surface coverage with AuNSt enabled 1-2 orders of magnitude higher SERS signals compared to plasmonic hydrogels loaded with premade colloids. Importantly, AuNSt can be prepared without the addition of any potentially cytotoxic surfactants, thereby ensuring a high biocompatibility. Overall, in situ growth becomes a versatile and straightforward approach for the fabrication of plasmonic biomaterials.
Collapse
Affiliation(s)
| | - Clara García-Astrain
- CIC
biomaGUNE, Basque Research and Technology
Alliance (BRTA), Donostia-San
Sebastián 20014, Spain
- Centro
de Investigación Biomédica en Red de Bioingeniería
Biomateriales, y Nanomedicina (CIBER-BBN), Donostia-San Sebastián 20014, Spain
| | - Lara Troncoso-Afonso
- CIC
biomaGUNE, Basque Research and Technology
Alliance (BRTA), Donostia-San
Sebastián 20014, Spain
- Department
of Applied Chemistry, University of the
Basque Country (UPV-EHU), Donostia-San
Sebastián 20018, Spain
| | - Marita Wagner
- CIC
biomaGUNE, Basque Research and Technology
Alliance (BRTA), Donostia-San
Sebastián 20014, Spain
- Department
of Applied Chemistry, University of the
Basque Country (UPV-EHU), Donostia-San
Sebastián 20018, Spain
- CIC
nanoGUNE, Basque Research and Technology
Alliance (BRTA), Donostia-San Sebastián 20018, Spain
| | - Judith Langer
- CIC
biomaGUNE, Basque Research and Technology
Alliance (BRTA), Donostia-San
Sebastián 20014, Spain
| | | | - Desirè Di Silvio
- CIC
biomaGUNE, Basque Research and Technology
Alliance (BRTA), Donostia-San
Sebastián 20014, Spain
| | - Luis M. Liz-Marzán
- CIC
biomaGUNE, Basque Research and Technology
Alliance (BRTA), Donostia-San
Sebastián 20014, Spain
- Centro
de Investigación Biomédica en Red de Bioingeniería
Biomateriales, y Nanomedicina (CIBER-BBN), Donostia-San Sebastián 20014, Spain
- Ikerbasque
Basque Foundation for Science, Bilbao 48009, Spain
- Cinbio, Universidade de Vigo, Vigo 36310, Spain
| |
Collapse
|
8
|
García-Astrain C, Henriksen-Lacey M, Lenzi E, Renero-Lecuna C, Langer J, Piñeiro P, Molina-Martínez B, Plou J, Jimenez de Aberasturi D, Liz-Marzán LM. A Scaffold-Assisted 3D Cancer Cell Model for Surface-Enhanced Raman Scattering-Based Real-Time Sensing and Imaging. ACS NANO 2024; 18:11257-11269. [PMID: 38632933 PMCID: PMC11064228 DOI: 10.1021/acsnano.4c00543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/02/2024] [Accepted: 04/09/2024] [Indexed: 04/19/2024]
Abstract
Despite recent advances in the development of scaffold-based three-dimensional (3D) cell models, challenges persist in imaging and monitoring cell behavior within these complex structures due to their heterogeneous cell distribution and geometries. Incorporating sensors into 3D scaffolds provides a potential solution for real-time, in situ sensing and imaging of biological processes such as cell growth and disease development. We introduce a 3D printed hydrogel-based scaffold capable of supporting both surface-enhanced Raman scattering (SERS) biosensing and imaging of 3D breast cancer cell models. The scaffold incorporates plasmonic nanoparticles and SERS tags, for sensing and imaging, respectively. We demonstrate the scaffold's adaptability and modularity in supporting breast cancer spheroids, thereby enabling spatial and temporal monitoring of tumor evolution.
Collapse
Affiliation(s)
- Clara García-Astrain
- CIC
biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
- Centro
de Investigación Biomédica en Red, Bioingeniería,
Biomateriales y Nanomedicina (CIBER-BBN), 20014 Donostia-San Sebastián, Spain
| | - Malou Henriksen-Lacey
- CIC
biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
- Centro
de Investigación Biomédica en Red, Bioingeniería,
Biomateriales y Nanomedicina (CIBER-BBN), 20014 Donostia-San Sebastián, Spain
| | - Elisa Lenzi
- CIC
biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
| | - Carlos Renero-Lecuna
- CIC
biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
- Cinbio,
University of Vigo, 36310 Vigo, Spain
| | - Judith Langer
- CIC
biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
| | - Paula Piñeiro
- CIC
biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
- Department
of Applied Chemistry, University of the
Basque Country (UPV-EHU), 20018 Donostia-San Sebastián, Spain
| | - Beatriz Molina-Martínez
- CIC
biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
| | - Javier Plou
- CIC
biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
| | - Dorleta Jimenez de Aberasturi
- CIC
biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
- Centro
de Investigación Biomédica en Red, Bioingeniería,
Biomateriales y Nanomedicina (CIBER-BBN), 20014 Donostia-San Sebastián, Spain
- Ikerbasque,
Basque Foundation for Science, 48009 Bilbao, Spain
| | - Luis M. Liz-Marzán
- CIC
biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
- Centro
de Investigación Biomédica en Red, Bioingeniería,
Biomateriales y Nanomedicina (CIBER-BBN), 20014 Donostia-San Sebastián, Spain
- Cinbio,
University of Vigo, 36310 Vigo, Spain
- Ikerbasque,
Basque Foundation for Science, 48009 Bilbao, Spain
| |
Collapse
|
9
|
Laowpanitchakorn P, Zeng J, Piantino M, Uchida K, Katsuyama M, Matsusaki M. Biofabrication of engineered blood vessels for biomedical applications. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2024; 25:2330339. [PMID: 38633881 PMCID: PMC11022926 DOI: 10.1080/14686996.2024.2330339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/10/2024] [Indexed: 04/19/2024]
Abstract
To successfully engineer large-sized tissues, establishing vascular structures is essential for providing oxygen, nutrients, growth factors and cells to prevent necrosis at the core of the tissue. The diameter scale of the biofabricated vasculatures should range from 100 to 1,000 µm to support the mm-size tissue while being controllably aligned and spaced within the diffusion limit of oxygen. In this review, insights regarding biofabrication considerations and techniques for engineered blood vessels will be presented. Initially, polymers of natural and synthetic origins can be selected, modified, and combined with each other to support maturation of vascular tissue while also being biocompatible. After they are shaped into scaffold structures by different fabrication techniques, surface properties such as physical topography, stiffness, and surface chemistry play a major role in the endothelialization process after transplantation. Furthermore, biological cues such as growth factors (GFs) and endothelial cells (ECs) can be incorporated into the fabricated structures. As variously reported, fabrication techniques, especially 3D printing by extrusion and 3D printing by photopolymerization, allow the construction of vessels at a high resolution with diameters in the desired range. Strategies to fabricate of stable tubular structures with defined channels will also be discussed. This paper provides an overview of the many advances in blood vessel engineering and combinations of different fabrication techniques up to the present time.
Collapse
Affiliation(s)
| | - Jinfeng Zeng
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
| | - Marie Piantino
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
- The Consortium for Future Innovation by Cultured Meat, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
| | - Kentaro Uchida
- Materials Solution Department, Product Analysis Center, Panasonic Holdings Corporation, Kadoma, Osaka, Japan
| | - Misa Katsuyama
- Materials Solution Department, Product Analysis Center, Panasonic Holdings Corporation, Kadoma, Osaka, Japan
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
- The Consortium for Future Innovation by Cultured Meat, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
10
|
Kolahi Azar H, Gharibshahian M, Rostami M, Mansouri V, Sabouri L, Beheshtizadeh N, Rezaei N. The progressive trend of modeling and drug screening systems of breast cancer bone metastasis. J Biol Eng 2024; 18:14. [PMID: 38317174 PMCID: PMC10845631 DOI: 10.1186/s13036-024-00408-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/22/2024] [Indexed: 02/07/2024] Open
Abstract
Bone metastasis is considered as a considerable challenge for breast cancer patients. Various in vitro and in vivo models have been developed to examine this occurrence. In vitro models are employed to simulate the intricate tumor microenvironment, investigate the interplay between cells and their adjacent microenvironment, and evaluate the effectiveness of therapeutic interventions for tumors. The endeavor to replicate the latency period of bone metastasis in animal models has presented a challenge, primarily due to the necessity of primary tumor removal and the presence of multiple potential metastatic sites.The utilization of novel bone metastasis models, including three-dimensional (3D) models, has been proposed as a promising approach to overcome the constraints associated with conventional 2D and animal models. However, existing 3D models are limited by various factors, such as irregular cellular proliferation, autofluorescence, and changes in genetic and epigenetic expression. The imperative for the advancement of future applications of 3D models lies in their standardization and automation. The utilization of artificial intelligence exhibits the capability to predict cellular behavior through the examination of substrate materials' chemical composition, geometry, and mechanical performance. The implementation of these algorithms possesses the capability to predict the progression and proliferation of cancer. This paper reviewed the mechanisms of bone metastasis following primary breast cancer. Current models of breast cancer bone metastasis, along with their challenges, as well as the future perspectives of using these models for translational drug development, were discussed.
Collapse
Affiliation(s)
- Hanieh Kolahi Azar
- Department of Pathology, Tabriz University of Medical Sciences, Tabriz, Iran
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Maliheh Gharibshahian
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mohammadreza Rostami
- Division of Food Safety and Hygiene, Department of Environmental Health Engineering, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Food Science and Nutrition Group (FSAN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Vahid Mansouri
- Gene Therapy Research Center, Digestive Diseases Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Leila Sabouri
- Department of Tissue Engineering and Applied Cell Sciences, School of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Beheshtizadeh
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Nima Rezaei
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
11
|
Gugulothu S, Asthana S, Homer-Vanniasinkam S, Chatterjee K. Trends in Photopolymerizable Bioinks for 3D Bioprinting of Tumor Models. JACS AU 2023; 3:2086-2106. [PMID: 37654587 PMCID: PMC10466332 DOI: 10.1021/jacsau.3c00281] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 09/02/2023]
Abstract
Three-dimensional (3D) bioprinting technologies involving photopolymerizable bioinks (PBs) have attracted enormous attention in recent times owing to their ability to recreate complex structures with high resolution, mechanical stability, and favorable printing conditions that are suited for encapsulating cells. 3D bioprinted tissue constructs involving PBs can offer better insights into the tumor microenvironment and offer platforms for drug screening to advance cancer research. These bioinks enable the incorporation of physiologically relevant cell densities, tissue-mimetic stiffness, and vascularized channels and biochemical gradients in the 3D tumor models, unlike conventional two-dimensional (2D) cultures or other 3D scaffold fabrication technologies. In this perspective, we present the emerging techniques of 3D bioprinting using PBs in the context of cancer research, with a specific focus on the efforts to recapitulate the complexity of the tumor microenvironment. We describe printing approaches and various PB formulations compatible with these techniques along with recent attempts to bioprint 3D tumor models for studying migration and metastasis, cell-cell interactions, cell-extracellular matrix interactions, and drug screening relevant to cancer. We discuss the limitations and identify unexplored opportunities in this field for clinical and commercial translation of these emerging technologies.
Collapse
Affiliation(s)
- Sriram
Bharath Gugulothu
- Department
of Materials Engineering Indian Institute of Science, Bangalore, Karnataka 560012, India
| | - Sonal Asthana
- Department
of Materials Engineering Indian Institute of Science, Bangalore, Karnataka 560012, India
- Department
of Hepatobiliary and Multi-Organ Transplantation Surgery, Aster CMI Hospital, Bangalore 560024, India
| | - Shervanthi Homer-Vanniasinkam
- Department
of Materials Engineering Indian Institute of Science, Bangalore, Karnataka 560012, India
- Department
of Mechanical Engineering and Division of Surgery, University College, London WC1E 7JE, U.K.
| | - Kaushik Chatterjee
- Department
of Materials Engineering Indian Institute of Science, Bangalore, Karnataka 560012, India
| |
Collapse
|
12
|
Tronolone JJ, Mathur T, Chaftari CP, Jain A. Evaluation of the Morphological and Biological Functions of Vascularized Microphysiological Systems with Supervised Machine Learning. Ann Biomed Eng 2023; 51:1723-1737. [PMID: 36913087 DOI: 10.1007/s10439-023-03177-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 02/22/2023] [Indexed: 03/14/2023]
Abstract
Vascularized microphysiological systems and organoids are contemporary preclinical experimental platforms representing human tissue or organ function in health and disease. While vascularization is emerging as a necessary physiological organ-level feature required in most such systems, there is no standard tool or morphological metric to measure the performance or biological function of vascularized networks within these models. Further, the commonly reported morphological metrics may not correlate to the network's biological function-oxygen transport. Here, a large library of vascular network images was analyzed by the measure of each sample's morphology and oxygen transport potential. The oxygen transport quantification is computationally expensive and user-dependent, so machine learning techniques were examined to generate regression models relating morphology to function. Principal component and factor analyses were applied to reduce dimensionality of the multivariate dataset, followed by multiple linear regression and tree-based regression analyses. These examinations reveal that while several morphological data relate poorly to the biological function, some machine learning models possess a relatively improved, but still moderate predictive potential. Overall, random forest regression model correlates to the biological function of vascular networks with relatively higher accuracy than other regression models.
Collapse
Affiliation(s)
- James J Tronolone
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, 101 Bizzell Street, College Station, TX, 77843, USA
| | - Tanmay Mathur
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, 101 Bizzell Street, College Station, TX, 77843, USA
| | - Christopher P Chaftari
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, 101 Bizzell Street, College Station, TX, 77843, USA
| | - Abhishek Jain
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, 101 Bizzell Street, College Station, TX, 77843, USA.
- Department of Medical Physiology, School of Medicine, Texas A&M University, Bryan, TX, USA.
- Department of Cardiovascular Sciences, Houston Methodist Academic Institute, Houston, USA.
| |
Collapse
|
13
|
Souto-Lopes M, Fernandes MH, Monteiro FJ, Salgado CL. Bioengineering Composite Aerogel-Based Scaffolds That Influence Porous Microstructure, Mechanical Properties and In Vivo Regeneration for Bone Tissue Application. MATERIALS (BASEL, SWITZERLAND) 2023; 16:4483. [PMID: 37374666 PMCID: PMC10305395 DOI: 10.3390/ma16124483] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/13/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023]
Abstract
Tissue regeneration of large bone defects is still a clinical challenge. Bone tissue engineering employs biomimetic strategies to produce graft composite scaffolds that resemble the bone extracellular matrix to guide and promote osteogenic differentiation of the host precursor cells. Aerogel-based bone scaffold preparation methods have been increasingly improved to overcome the difficulties in balancing the need for an open highly porous and hierarchically organized microstructure with compression resistance to withstand bone physiological loads, especially in wet conditions. Moreover, these improved aerogel scaffolds have been implanted in vivo in critical bone defects, in order to test their bone regeneration potential. This review addresses recently published studies on aerogel composite (organic/inorganic)-based scaffolds, having in mind the various cutting-edge technologies and raw biomaterials used, as well as the improvements that are still a challenge in terms of their relevant properties. Finally, the lack of 3D in vitro models of bone tissue for regeneration studies is emphasized, as well as the need for further developments to overcome and minimize the requirement for studies using in vivo animal models.
Collapse
Affiliation(s)
- Mariana Souto-Lopes
- i3S—Instituto de Investigação e Inovação em Saúde da Universidade do Porto, 4200-135 Porto, Portugal; (M.S.-L.); (F.J.M.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- Departamento de Engenharia Metalúrgica e de Materiais, Faculdade de Engenharia da Universidade do Porto, 4200-465 Porto, Portugal
| | - Maria Helena Fernandes
- Bonelab–Laboratory for Bone Metabolism and Regeneration, Faculdade de Medicina Dentária da Universidade do Porto, 4200-393 Porto, Portugal
- LAQV/REQUIMTE—Laboratório Associado para a Química Verde/Rede de Química e Tecnologia, 4169-007 Porto, Portugal
| | - Fernando Jorge Monteiro
- i3S—Instituto de Investigação e Inovação em Saúde da Universidade do Porto, 4200-135 Porto, Portugal; (M.S.-L.); (F.J.M.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- Departamento de Engenharia Metalúrgica e de Materiais, Faculdade de Engenharia da Universidade do Porto, 4200-465 Porto, Portugal
- Porto Comprehensive Cancer Center (P.CCC), R. Dr. António Bernardino de Almeida, 4200–072 Porto, Portugal
| | - Christiane Laranjo Salgado
- i3S—Instituto de Investigação e Inovação em Saúde da Universidade do Porto, 4200-135 Porto, Portugal; (M.S.-L.); (F.J.M.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| |
Collapse
|
14
|
Parodi I, Di Lisa D, Pastorino L, Scaglione S, Fato MM. 3D Bioprinting as a Powerful Technique for Recreating the Tumor Microenvironment. Gels 2023; 9:482. [PMID: 37367152 DOI: 10.3390/gels9060482] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/06/2023] [Accepted: 06/10/2023] [Indexed: 06/28/2023] Open
Abstract
In vitro three-dimensional models aim to reduce and replace animal testing and establish new tools for oncology research and the development and testing of new anticancer therapies. Among the various techniques to produce more complex and realistic cancer models is bioprinting, which allows the realization of spatially controlled hydrogel-based scaffolds, easily incorporating different types of cells in order to recreate the crosstalk between cancer and stromal components. Bioprinting exhibits other advantages, such as the production of large constructs, the repeatability and high resolution of the process, as well as the possibility of vascularization of the models through different approaches. Moreover, bioprinting allows the incorporation of multiple biomaterials and the creation of gradient structures to mimic the heterogeneity of the tumor microenvironment. The aim of this review is to report the main strategies and biomaterials used in cancer bioprinting. Moreover, the review discusses several bioprinted models of the most diffused and/or malignant tumors, highlighting the importance of this technique in establishing reliable biomimetic tissues aimed at improving disease biology understanding and high-throughput drug screening.
Collapse
Affiliation(s)
- Ilaria Parodi
- Department of Computer Science, Bioengineering, Robotics and Systems Engineering, University of Genoa, 16126 Genoa, Italy
- National Research Council of Italy, Institute of Electronic, Computer and Telecommunications Engineering (IEIIT), 16149 Genoa, Italy
| | - Donatella Di Lisa
- Department of Computer Science, Bioengineering, Robotics and Systems Engineering, University of Genoa, 16126 Genoa, Italy
| | - Laura Pastorino
- Department of Computer Science, Bioengineering, Robotics and Systems Engineering, University of Genoa, 16126 Genoa, Italy
| | - Silvia Scaglione
- National Research Council of Italy, Institute of Electronic, Computer and Telecommunications Engineering (IEIIT), 16149 Genoa, Italy
- React4life S.p.A., 16152 Genova, Italy
| | - Marco Massimo Fato
- Department of Computer Science, Bioengineering, Robotics and Systems Engineering, University of Genoa, 16126 Genoa, Italy
- National Research Council of Italy, Institute of Electronic, Computer and Telecommunications Engineering (IEIIT), 16149 Genoa, Italy
| |
Collapse
|
15
|
Pasquier E, Rosendahl J, Solberg A, Ståhlberg A, Håkansson J, Chinga-Carrasco G. Polysaccharides and Structural Proteins as Components in Three-Dimensional Scaffolds for Breast Cancer Tissue Models: A Review. Bioengineering (Basel) 2023; 10:682. [PMID: 37370613 PMCID: PMC10295496 DOI: 10.3390/bioengineering10060682] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 05/26/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Breast cancer is the most common cancer among women, and even though treatments are available, efficiency varies with the patients. In vitro 2D models are commonly used to develop new treatments. However, 2D models overestimate drug efficiency, which increases the failure rate in later phase III clinical trials. New model systems that allow extensive and efficient drug screening are thus required. Three-dimensional printed hydrogels containing active components for cancer cell growth are interesting candidates for the preparation of next generation cancer cell models. Macromolecules, obtained from marine- and land-based resources, can form biopolymers (polysaccharides such as alginate, chitosan, hyaluronic acid, and cellulose) and bioactive components (structural proteins such as collagen, gelatin, and silk fibroin) in hydrogels with adequate physical properties in terms of porosity, rheology, and mechanical strength. Hence, in this study attention is given to biofabrication methods and to the modification with biological macromolecules to become bioactive and, thus, optimize 3D printed structures that better mimic the cancer cell microenvironment. Ink formulations combining polysaccharides for tuning the mechanical properties and bioactive polymers for controlling cell adhesion is key to optimizing the growth of the cancer cells.
Collapse
Affiliation(s)
- Eva Pasquier
- RISE PFI AS, Høgskoleringen 6b, NO-7491 Trondheim, Norway; (E.P.); (A.S.)
| | - Jennifer Rosendahl
- RISE Unit of Biological Function, Division Materials and Production, RISE Research Institutes of Sweden, Box 857, 50115 Borås, Sweden; (J.R.); (J.H.)
| | - Amalie Solberg
- RISE PFI AS, Høgskoleringen 6b, NO-7491 Trondheim, Norway; (E.P.); (A.S.)
| | - Anders Ståhlberg
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 41390 Gothenburg, Sweden;
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, 41390 Gothenburg, Sweden
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden
| | - Joakim Håkansson
- RISE Unit of Biological Function, Division Materials and Production, RISE Research Institutes of Sweden, Box 857, 50115 Borås, Sweden; (J.R.); (J.H.)
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, 40530 Gothenburg, Sweden
- Department of Chemistry and Molecular Biology, University of Gothenburg, 40530 Gothenburg, Sweden
| | | |
Collapse
|
16
|
Yang X, Li S, Sun X, Ren Y, Qiang L, Liu Y, Wang J, Dai K. Swelling compensation of engineered vasculature fabricated by additive manufacturing and sacrifice-based technique using thermoresponsive hydrogel. Int J Bioprint 2023; 9:749. [PMID: 37457939 PMCID: PMC10339422 DOI: 10.18063/ijb.749] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/02/2023] [Indexed: 07/18/2023] Open
Abstract
Engineered vasculature is widely employed to maintain the cell viability within in vitro tissues. A variety of fabrication techniques for engineered vasculature have been explored, with combination of additive manufacturing with a sacrifice-based technique being the most common approach. However, the size deformation of vasculature caused by the swelling of sacrificial materials remains unaddressed. In this study, Pluronic F-127 (PF-127), the most widely used sacrificial material, was employed to study the deformation of the vasculature. Then, a thermoresponsive hydrogel comprising poly(N-isopropylacrylamide) (PNIPAM) and gelatin methacrylate (GelMA) was used to induce volume shrinkage at 37°C to compensate for the deformation of vasculature caused by the swelling of a three-dimensional (3D)-printed sacrificial template, and to generate vasculature of a smaller size than that after deformation. Our results showed that the vasculature diameter increased after the sacrificial template was removed, whereas it decreased to the designed diameter after the volume shrinkage. Human umbilical vein endothelial cells (HUVECs) formed an endothelial monolayer in the engineered vasculature. Osteosarcoma cells (OCs) were loaded into a hierarchical vasculature within the thermoresponsive hydrogel to investigate the interaction between HUVECs and OCs. New blood vessel infiltration was observed within the lumen of the engineered vasculature after in vivo subcutaneous implantation for 4 weeks. In addition, engineered vasculature was implanted in a rat ischemia model to further study the function of engineered vasculature for blood vessel infiltration. This study presents a small method aiming to accurately create engineered vasculature by additive manufacturing and a sacrificebased technique.
Collapse
Affiliation(s)
- Xue Yang
- College of Medicine, Southwest Jiaotong University, No. 144 Jiaoda Road, Chengdu 610031, People’s Republic of China
| | - Shuai Li
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Rd, Hangzhou 310003, People’s Republic of China
| | - Xin Sun
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Rd, Shanghai 200011, People’s Republic of China
| | - Ya Ren
- College of Medicine, Southwest Jiaotong University, No. 144 Jiaoda Road, Chengdu 610031, People’s Republic of China
| | - Lei Qiang
- School of Materials Science and Engineering, Southwest Jiaotong University, No. 111 2nd Ring Rd, Chengdu 611756, People’s Republic of China
| | - Yihao Liu
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Rd, Shanghai 200011, People’s Republic of China
| | - Jinwu Wang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Rd, Shanghai 200011, People’s Republic of China
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, No. 1956 Huashan Rd, Shanghai 200030, People’s Republic of China
| | - Kerong Dai
- College of Medicine, Southwest Jiaotong University, No. 144 Jiaoda Road, Chengdu 610031, People’s Republic of China
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Rd, Shanghai 200011, People’s Republic of China
| |
Collapse
|
17
|
Ren B, Jiang Z, Murfee WL, Katz AJ, Siemann D, Huang Y. Realizations of vascularized tissues: From in vitro platforms to in vivo grafts. BIOPHYSICS REVIEWS 2023; 4:011308. [PMID: 36938117 PMCID: PMC10015415 DOI: 10.1063/5.0131972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 02/07/2023] [Indexed: 03/18/2023]
Abstract
Vascularization is essential for realizing thick and functional tissue constructs that can be utilized for in vitro study platforms and in vivo grafts. The vasculature enables the transport of nutrients, oxygen, and wastes and is also indispensable to organ functional units such as the nephron filtration unit, the blood-air barrier, and the blood-brain barrier. This review aims to discuss the latest progress of organ-like vascularized constructs with specific functionalities and realizations even though they are not yet ready to be used as organ substitutes. First, the human vascular system is briefly introduced and related design considerations for engineering vascularized tissues are discussed. Second, up-to-date creation technologies for vascularized tissues are summarized and classified into the engineering and cellular self-assembly approaches. Third, recent applications ranging from in vitro tissue models, including generic vessel models, tumor models, and different human organ models such as heart, kidneys, liver, lungs, and brain, to prevascularized in vivo grafts for implantation and anastomosis are discussed in detail. The specific design considerations for the aforementioned applications are summarized and future perspectives regarding future clinical applications and commercialization are provided.
Collapse
Affiliation(s)
- Bing Ren
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, Florida 32611, USA
| | - Zhihua Jiang
- Department of Surgery, University of Florida, Gainesville, Florida 32610, USA
| | - Walter Lee Murfee
- Department of Biomedical Engineering, University of Florida, Gainesville, Florida 32611, USA
| | - Adam J. Katz
- Department of Plastic and Reconstructive Surgery, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, USA
| | - Dietmar Siemann
- Department of Radiation Oncology, University of Florida, Gainesville, Florida 32610, USA
| | - Yong Huang
- Author to whom correspondence should be addressed:
| |
Collapse
|
18
|
Tronolone JJ, Mathur T, Chaftari CP, Jain A. Evaluation of the morphological and biological functions of vascularized microphysiological systems with supervised machine learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.12.523755. [PMID: 36711458 PMCID: PMC9882172 DOI: 10.1101/2023.01.12.523755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Vascularized microphysiological systems and organoids are contemporary preclinical experimental platforms representing human tissue or organ function in health and disease. While vascularization is emerging as a necessary physiological organ-level feature required in most such systems, there is no standard tool or morphological metric to measure the performance or biological function of vascularized networks within these models. Further, the commonly reported morphological metrics may not correlate to the network's biological function - oxygen transport. Here, a large library of vascular network images was analyzed by the measure of each sample's morphology and oxygen transport potential. The oxygen transport quantification is computationally expensive and user-dependent, so machine learning techniques were examined to generate regression models relating morphology to function. Principal component and factor analyses were applied to reduce dimensionality of the multivariate dataset, followed by multiple linear regression and tree-based regression analyses. These examinations reveal that while several morphological data relate poorly to the biological function, some machine learning models possess a relatively improved, but still moderate predictive potential. Overall, random forest regression model correlates to the biological function of vascular networks with relatively higher accuracy than other regression models.
Collapse
|
19
|
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: 8] [Impact Index Per Article: 4.0] [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
|
20
|
Azimian Zavareh V, Rafiee L, Sheikholeslam M, Shariati L, Vaseghi G, Savoji H, Haghjooy Javanmard S. Three-Dimensional in Vitro Models: A Promising Tool To Scale-Up Breast Cancer Research. ACS Biomater Sci Eng 2022; 8:4648-4672. [PMID: 36260561 DOI: 10.1021/acsbiomaterials.2c00277] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Common models used in breast cancer studies, including two-dimensional (2D) cultures and animal models, do not precisely model all aspects of breast tumors. These models do not well simulate the cell-cell and cell-stromal interactions required for normal tumor growth in the body and lake tumor like microenvironment. Three-dimensional (3D) cell culture models are novel approaches to studying breast cancer. They do not have the restrictions of these conventional models and are able to recapitulate the structural architecture, complexity, and specific function of breast tumors and provide similar in vivo responses to therapeutic regimens. These models can be a link between former traditional 2D culture and in vivo models and are necessary for further studies in cancer. This review attempts to summarize the most common 3D in vitro models used in breast cancer studies, including scaffold-free (spheroid and organoid), scaffold-based, and chip-based models, particularly focused on the basic and translational application of these 3D models in drug screening and the tumor microenvironment in breast cancer.
Collapse
Affiliation(s)
- Vajihe Azimian Zavareh
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 81746 73461, Iran.,Core Research Facilities (CRF), Isfahan University of Medical Sciences, Isfahan 81746 73461, Iran
| | - Laleh Rafiee
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 81746 73461, Iran
| | - Mohammadali Sheikholeslam
- Department of Biomaterials, Nanotechnology and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan 81746 73461, Iran.,Biosensor Research Center, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan 81746 73461, Iran
| | - Laleh Shariati
- Department of Biomaterials, Nanotechnology and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan 81746 73461, Iran.,Cancer Prevention Research Center, Omid Hospital, Isfahan University of Medical Sciences, Isfahan 81746 73461, Iran
| | - Golnaz Vaseghi
- Isfahan Cardiovascular Research Center, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 81746 73461, Iran
| | - Houman Savoji
- Institute of Biomedical Engineering, Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada.,Research Center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC H3T 1C5, Canada.,Montreal TransMedTech Institute, Montreal, QC H3T 1J4, Canada
| | - Shaghayegh Haghjooy Javanmard
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 81746 73461, Iran
| |
Collapse
|
21
|
Inflammation- and Metastasis-Related Proteins Expression Changes in Early Stages in Tumor and Non-Tumor Adjacent Tissues of Colorectal Cancer Samples. Cancers (Basel) 2022; 14:cancers14184487. [PMID: 36139645 PMCID: PMC9497293 DOI: 10.3390/cancers14184487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/05/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Non-tumor adjacent tissue plays a key role in colorectal cancer development, as well as chronic inflammation, but their role has not yet been dilucidated. In addition, inflammation is a process which is related to epithelial-mesenchymal transition and metastasis, but their changes across the different colorectal cancer stages are not fully studied. Understanding how these processes participate in all colorectal cancer phases can be key to a better understanding of the disease. Abstract Chronic inflammation can induce malignant cell transformation, having an important role in all colorectal cancer (CRC) phases. Non-tumor adjacent tissue plays an important role in tumor progression, but its implication in CRC has not yet been fully elucidated. The aim was to analyze the expression of inflammatory, epithelial-mesenchymal transition (EMT), and metastasis-related proteins in both tumor and non-tumor adjacent tissues from CRC patients by western blot. Tumor tissue presented an increase in metastasis and EMT-related proteins compared to non-tumor adjacent tissue, especially in stage II. Tumor tissue stage II also presented an increase in inflammatory-related proteins compared to other stages, which was also seen in non-tumor adjacent tissue stage II. Additionally, the relapse-free survival study of Vimentin and VEGF-B expression levels in stage II patients showed that the higher the expression levels of each protein, the lower 10-year relapse-free survival. These could suggest that some metastasis-related signalling pathways may be activated in stage II in tumor tissue, accompanied by an increase in inflammation. Furthermore, non-tumor adjacent tissue presented an increase of the inflammatory status that could be the basis for future tumor progression. In conclusion, these proteins could be useful as biomarkers of diagnosis for CRC at early stages.
Collapse
|
22
|
Zeng T, Sun C, Liang Y, Yang F, Yan X, Bao S, Zhang Y, Huang X, Fu Z, Li W, Yin Y. A Real-World Multicentre Retrospective Study of Low-Dose Apatinib for Human Epidermal Growth Factor Receptor 2-Negative Metastatic Breast Cancer. Cancers (Basel) 2022; 14:cancers14174084. [PMID: 36077621 PMCID: PMC9454649 DOI: 10.3390/cancers14174084] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/13/2022] [Accepted: 08/20/2022] [Indexed: 11/16/2022] Open
Abstract
Treatment options for human epidermal growth factor receptor (HER2)-negative breast cancer patients are limited in comparison to the HER2-positive patients, particularly for metastatic breast cancer patients. Apatinib is a small-molecule tyrosine kinase inhibitor that targets the vascular endothelial growth factor receptor 2 (VEGFR-2). Here, we reported the apatinib-based therapy data in HER2-negative metastatic breast cancer. Apatinib was taken at a dose of 250 mg orally once per day and combined with standard chemotherapy regimens. The PFS and OS of 128 patients were 4.7 months and 15.3 months, respectively. The objective response rate (ORR) and the disease control rate (DCR) were 22.7% and 80.5%, respectively. Patients with breast cancer susceptibility gene (BRCA) mutations were found to have a longer PFS and OS. Moreover, combination immunotherapy or paclitaxel-platinum regimens shared an improved response to other regimens. Most of the adverse effects (hypertension, anaemia, and hand-foot syndrome) were grade 1 to 2. Metastatic breast cancer patients could benefit from apatinib therapy at a low dosage, and the adverse effects are mild in real-world clinical practice. Furthermore, BRCA may be a putative biomarker for apatinib in HER2-negative breast cancer. Immunotherapy or paclitaxel-platinum regimens may be recommended to combine with apatinib therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Wei Li
- Correspondence: (W.L.); (Y.Y.); Tel.: +86-025-68307102 (W.L. & Y.Y.)
| | - Yongmei Yin
- Correspondence: (W.L.); (Y.Y.); Tel.: +86-025-68307102 (W.L. & Y.Y.)
| |
Collapse
|
23
|
Badr-Eldin SM, Aldawsari HM, Kotta S, Deb PK, Venugopala KN. Three-Dimensional In Vitro Cell Culture Models for Efficient Drug Discovery: Progress So Far and Future Prospects. Pharmaceuticals (Basel) 2022; 15:926. [PMID: 36015074 PMCID: PMC9412659 DOI: 10.3390/ph15080926] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/16/2022] [Accepted: 07/18/2022] [Indexed: 12/13/2022] Open
Abstract
Despite tremendous advancements in technologies and resources, drug discovery still remains a tedious and expensive process. Though most cells are cultured using 2D monolayer cultures, due to lack of specificity, biochemical incompatibility, and cell-to-cell/matrix communications, they often lag behind in the race of modern drug discovery. There exists compelling evidence that 3D cell culture models are quite promising and advantageous in mimicking in vivo conditions. It is anticipated that these 3D cell culture methods will bridge the translation of data from 2D cell culture to animal models. Although 3D technologies have been adopted widely these days, they still have certain challenges associated with them, such as the maintenance of a micro-tissue environment similar to in vivo models and a lack of reproducibility. However, newer 3D cell culture models are able to bypass these issues to a maximum extent. This review summarizes the basic principles of 3D cell culture approaches and emphasizes different 3D techniques such as hydrogels, spheroids, microfluidic devices, organoids, and 3D bioprinting methods. Besides the progress made so far in 3D cell culture systems, the article emphasizes the various challenges associated with these models and their potential role in drug repositioning, including perspectives from the COVID-19 pandemic.
Collapse
Affiliation(s)
- Shaimaa M. Badr-Eldin
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (H.M.A.); (S.K.)
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Hibah M. Aldawsari
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (H.M.A.); (S.K.)
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Sabna Kotta
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (H.M.A.); (S.K.)
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Pran Kishore Deb
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Philadelphia University, P.O. Box 1, Amman 19392, Jordan
| | - Katharigatta N. Venugopala
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia;
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, Durban 4001, South Africa
| |
Collapse
|
24
|
Staros R, Michalak A, Rusinek K, Mucha K, Pojda Z, Zagożdżon R. Perspectives for 3D-Bioprinting in Modeling of Tumor Immune Evasion. Cancers (Basel) 2022; 14:cancers14133126. [PMID: 35804898 PMCID: PMC9265021 DOI: 10.3390/cancers14133126] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 05/31/2022] [Accepted: 06/23/2022] [Indexed: 02/07/2023] Open
Abstract
In a living organism, cancer cells function in a specific microenvironment, where they exchange numerous physical and biochemical cues with other cells and the surrounding extracellular matrix (ECM). Immune evasion is a clinically relevant phenomenon, in which cancer cells are able to direct this interchange of signals against the immune effector cells and to generate an immunosuppressive environment favoring their own survival. A proper understanding of this phenomenon is substantial for generating more successful anticancer therapies. However, classical cell culture systems are unable to sufficiently recapture the dynamic nature and complexity of the tumor microenvironment (TME) to be of satisfactory use for comprehensive studies on mechanisms of tumor immune evasion. In turn, 3D-bioprinting is a rapidly evolving manufacture technique, in which it is possible to generate finely detailed structures comprised of multiple cell types and biomaterials serving as ECM-analogues. In this review, we focus on currently used 3D-bioprinting techniques, their applications in the TME research, and potential uses of 3D-bioprinting in modeling of tumor immune evasion and response to immunotherapies.
Collapse
Affiliation(s)
- Rafał Staros
- Department of Immunology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-006 Warsaw, Poland; (R.S.); (K.M.)
| | - Agata Michalak
- Department of Regenerative Medicine, Maria Sklodowska-Curie National Institute of Oncology, 02-781 Warsaw, Poland; (A.M.); (K.R.); (Z.P.)
| | - Kinga Rusinek
- Department of Regenerative Medicine, Maria Sklodowska-Curie National Institute of Oncology, 02-781 Warsaw, Poland; (A.M.); (K.R.); (Z.P.)
| | - Krzysztof Mucha
- Department of Immunology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-006 Warsaw, Poland; (R.S.); (K.M.)
| | - Zygmunt Pojda
- Department of Regenerative Medicine, Maria Sklodowska-Curie National Institute of Oncology, 02-781 Warsaw, Poland; (A.M.); (K.R.); (Z.P.)
| | - Radosław Zagożdżon
- Department of Immunology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-006 Warsaw, Poland; (R.S.); (K.M.)
- Department of Regenerative Medicine, Maria Sklodowska-Curie National Institute of Oncology, 02-781 Warsaw, Poland; (A.M.); (K.R.); (Z.P.)
- Department of Clinical Immunology, Medical University of Warsaw, 02-006 Warsaw, Poland
- Correspondence: ; Tel.: +48-22-502-14-72; Fax: +48-22-502-21-59
| |
Collapse
|
25
|
Tian H, Shi H, Yu J, Ge S, Ruan J. Biophysics Role and Biomimetic Culture Systems of ECM Stiffness in Cancer EMT. GLOBAL CHALLENGES (HOBOKEN, NJ) 2022; 6:2100094. [PMID: 35712024 PMCID: PMC9189138 DOI: 10.1002/gch2.202100094] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 02/14/2022] [Indexed: 06/15/2023]
Abstract
Oncological diseases have become the second leading cause of death from noncommunicable diseases worldwide and a major threat to human health. With the continuous progress in cancer research, the mechanical cues from the tumor microenvironment environment (TME) have been found to play an irreplaceable role in the progression of many cancers. As the main extracellular mechanical signal carrier, extracellular matrix (ECM) stiffness may influence cancer progression through biomechanical transduction to modify downstream gene expression, promote epithelial-mesenchymal transition (EMT), and regulate the stemness of cancer cells. EMT is an important mechanism that induces cancer cell metastasis and is closely influenced by ECM stiffness, either independently or in conjunction with other molecules. In this review, the unique role of ECM stiffness in EMT in different kinds of cancers is first summarized. By continually examining the significance of ECM stiffness in cancer progression, a biomimetic culture system based on 3D manufacturing and novel material technologies is developed to mimic ECM stiffness. The authors then look back on the novel development of the ECM stiffness biomimetic culture systems and finally provide new insights into ECM stiffness in cancer progression which can broaden the fields' horizons with a view toward developing new cancer diagnosis methods and therapies.
Collapse
Affiliation(s)
- Hao Tian
- Department of OphthalmologyShanghai Key Laboratory of Orbital Diseases and Ocular OncologyNinth People's HospitalShanghai JiaoTong University School of MedicineShanghaiP. R. China
| | - Hanhan Shi
- Department of OphthalmologyShanghai Key Laboratory of Orbital Diseases and Ocular OncologyNinth People's HospitalShanghai JiaoTong University School of MedicineShanghaiP. R. China
| | - Jie Yu
- Department of OphthalmologyShanghai Key Laboratory of Orbital Diseases and Ocular OncologyNinth People's HospitalShanghai JiaoTong University School of MedicineShanghaiP. R. China
| | - Shengfang Ge
- Department of OphthalmologyShanghai Key Laboratory of Orbital Diseases and Ocular OncologyNinth People's HospitalShanghai JiaoTong University School of MedicineShanghaiP. R. China
| | - Jing Ruan
- Department of OphthalmologyShanghai Key Laboratory of Orbital Diseases and Ocular OncologyNinth People's HospitalShanghai JiaoTong University School of MedicineShanghaiP. R. China
| |
Collapse
|
26
|
Rodrigues J, Sarmento B, Pereira CL. Osteosarcoma tumor microenvironment: the key for the successful development of biologically relevant 3D in vitro models. IN VITRO MODELS 2022; 1:5-27. [PMID: 39872973 PMCID: PMC11756501 DOI: 10.1007/s44164-022-00008-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 01/09/2022] [Accepted: 01/09/2022] [Indexed: 01/30/2025]
Abstract
Osteosarcoma (OS) is the most common primary bone cancer in children and young adults. This type of cancer is characterized by a high mortality rate, especially for patients with resistant lung metastases. Given its low incidence, high genetic heterogeneity, the lack of effective targets, and poor availability of relevant in vitro and in vivo models to study the tumor progression and the metastatic cascade, the pathophysiology of OS is still poorly understood and the translation of novel drugs into the market has become stagnant. Due to the importance of the tumor microenvironment (TME) in the development of metastases and the growing interest in targeting TME-specific pathways for novel therapeutics in cancer, models that closely represent these interactions are crucial for a better understanding of cancer-related events. In OS research, most studies rely on oversimplified two-dimensional (2D) assays and complex animal models that do not faithfully recapitulate OS development and progression. In turn, three-dimensional (3D) models are able to mimic not only the physical 3D environment in which cancer cells grow but also involve interactions with the TME, including its extracellular matrix, and thus are promising tools for drug screening studies. In this review, the existing and innovative OS in vitro 3D models are highlighted, focusing on how the TME is crucial to develop effective platforms for OS tumor and metastasis modeling in a physiologically relevant context. Graphical abstract
Collapse
Affiliation(s)
- João Rodrigues
- Instituto de Investigação E Inovação Em Saúde (i3S), University of Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Instituto de Engenharia Biomédica (INEB), University of Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Instituto Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Rua Jorge Viterbo 228, 4150-180 Porto, Portugal
- Faculdade de Engenharia da Universidade Do Porto (FEUP), University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Bruno Sarmento
- Instituto de Investigação E Inovação Em Saúde (i3S), University of Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Instituto de Engenharia Biomédica (INEB), University of Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- CESPU, Instituto de Investigação E Formação Avançada Em Ciências E Tecnologias da Saúde, Rua Central da Gandra 1317, 4585-116 Gandra, Portugal
| | - Catarina Leite Pereira
- Instituto de Investigação E Inovação Em Saúde (i3S), University of Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Instituto de Engenharia Biomédica (INEB), University of Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| |
Collapse
|
27
|
Abstract
AbstractThe multidisciplinary research field of bioprinting combines additive manufacturing, biology and material sciences to create bioconstructs with three-dimensional architectures mimicking natural living tissues. The high interest in the possibility of reproducing biological tissues and organs is further boosted by the ever-increasing need for personalized medicine, thus allowing bioprinting to establish itself in the field of biomedical research, and attracting extensive research efforts from companies, universities, and research institutes alike. In this context, this paper proposes a scientometric analysis and critical review of the current literature and the industrial landscape of bioprinting to provide a clear overview of its fast-changing and complex position. The scientific literature and patenting results for 2000–2020 are reviewed and critically analyzed by retrieving 9314 scientific papers and 309 international patents in order to draw a picture of the scientific and industrial landscape in terms of top research countries, institutions, journals, authors and topics, and identifying the technology hubs worldwide. This review paper thus offers a guide to researchers interested in this field or to those who simply want to understand the emerging trends in additive manufacturing and 3D bioprinting.
Graphic abstract
Collapse
|
28
|
Ban J, Fock V, Aryee DNT, Kovar H. Mechanisms, Diagnosis and Treatment of Bone Metastases. Cells 2021; 10:2944. [PMID: 34831167 PMCID: PMC8616226 DOI: 10.3390/cells10112944] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/22/2021] [Accepted: 10/27/2021] [Indexed: 12/24/2022] Open
Abstract
Bone and bone marrow are among the most frequent metastatic sites of cancer. The occurrence of bone metastasis is frequently associated with a dismal disease outcome. The prevention and therapy of bone metastases is a priority in the treatment of cancer patients. However, current therapeutic options for patients with bone metastatic disease are limited in efficacy and associated with increased morbidity. Therefore, most current therapies are mainly palliative in nature. A better understanding of the underlying molecular pathways of the bone metastatic process is warranted to develop novel, well-tolerated and more successful treatments for a significant improvement of patients' quality of life and disease outcome. In this review, we provide comparative mechanistic insights into the bone metastatic process of various solid tumors, including pediatric cancers. We also highlight current and innovative approaches to biologically targeted therapy and immunotherapy. In particular, we discuss the role of the bone marrow microenvironment in the attraction, homing, dormancy and outgrowth of metastatic tumor cells and the ensuing therapeutic implications. Multiple signaling pathways have been described to contribute to metastatic spread to the bone of specific cancer entities, with most knowledge derived from the study of breast and prostate cancer. However, it is likely that similar mechanisms are involved in different types of cancer, including multiple myeloma, primary bone sarcomas and neuroblastoma. The metastatic rate-limiting interaction of tumor cells with the various cellular and noncellular components of the bone-marrow niche provides attractive therapeutic targets, which are already partially exploited by novel promising immunotherapies.
Collapse
Affiliation(s)
- Jozef Ban
- St. Anna Children’s Cancer Research Institute, 1090 Vienna, Austria; (J.B.); (V.F.); (D.N.T.A.)
| | - Valerie Fock
- St. Anna Children’s Cancer Research Institute, 1090 Vienna, Austria; (J.B.); (V.F.); (D.N.T.A.)
| | - Dave N. T. Aryee
- St. Anna Children’s Cancer Research Institute, 1090 Vienna, Austria; (J.B.); (V.F.); (D.N.T.A.)
- Department of Pediatrics, Medical University Vienna, 1090 Vienna, Austria
| | - Heinrich Kovar
- St. Anna Children’s Cancer Research Institute, 1090 Vienna, Austria; (J.B.); (V.F.); (D.N.T.A.)
- Department of Pediatrics, Medical University Vienna, 1090 Vienna, Austria
| |
Collapse
|
29
|
Dellaquila A, Le Bao C, Letourneur D, Simon‐Yarza T. In Vitro Strategies to Vascularize 3D Physiologically Relevant Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100798. [PMID: 34351702 PMCID: PMC8498873 DOI: 10.1002/advs.202100798] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/23/2021] [Indexed: 05/04/2023]
Abstract
Vascularization of 3D models represents a major challenge of tissue engineering and a key prerequisite for their clinical and industrial application. The use of prevascularized models built from dedicated materials could solve some of the actual limitations, such as suboptimal integration of the bioconstructs within the host tissue, and would provide more in vivo-like perfusable tissue and organ-specific platforms. In the last decade, the fabrication of vascularized physiologically relevant 3D constructs has been attempted by numerous tissue engineering strategies, which are classified here in microfluidic technology, 3D coculture models, namely, spheroids and organoids, and biofabrication. In this review, the recent advancements in prevascularization techniques and the increasing use of natural and synthetic materials to build physiological organ-specific models are discussed. Current drawbacks of each technology, future perspectives, and translation of vascularized tissue constructs toward clinics, pharmaceutical field, and industry are also presented. By combining complementary strategies, these models are envisioned to be successfully used for regenerative medicine and drug development in a near future.
Collapse
Affiliation(s)
- Alessandra Dellaquila
- Université de ParisINSERM U1148X Bichat HospitalParisF‐75018France
- Elvesys Microfluidics Innovation CenterParis75011France
- Biomolecular PhotonicsDepartment of PhysicsUniversity of BielefeldBielefeld33615Germany
| | - Chau Le Bao
- Université de ParisINSERM U1148X Bichat HospitalParisF‐75018France
- Université Sorbonne Paris NordGalilée InstituteVilletaneuseF‐93430France
| | | | | |
Collapse
|
30
|
Parisi C, Qin K, Fernandes FM. Colonization versus encapsulation in cell-laden materials design: porosity and process biocompatibility determine cellularization pathways. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2021; 379:20200344. [PMID: 34334019 DOI: 10.1098/rsta.2020.0344] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/28/2021] [Indexed: 06/13/2023]
Abstract
Seeding materials with living cells has been-and still is-one of the most promising approaches to reproduce the complexity and the functionality of living matter. The strategies to associate living cells with materials are limited to cell encapsulation and colonization, however, the requirements for these two approaches have been seldom discussed systematically. Here we propose a simple two-dimensional map based on materials' pore size and the cytocompatibility of their fabrication process to draw, for the first time, a guide to building cellularized materials. We believe this approach may serve as a straightforward guideline to design new, more relevant materials, able to seize the complexity and the function of biological materials. This article is part of the theme issue 'Bio-derived and bioinspired sustainable advanced materials for emerging technologies (part 1)'.
Collapse
Affiliation(s)
- Cleo Parisi
- Laboratoire de Chimie de la Matière Condensée de Paris, Sorbonne Université, UMR7574, 4 Place Jussieu, 75005 Paris, France
| | - Kankan Qin
- Laboratoire de Chimie de la Matière Condensée de Paris, Sorbonne Université, UMR7574, 4 Place Jussieu, 75005 Paris, France
| | - Francisco M Fernandes
- Laboratoire de Chimie de la Matière Condensée de Paris, Sorbonne Université, UMR7574, 4 Place Jussieu, 75005 Paris, France
| |
Collapse
|
31
|
Kim J, Jang J, Cho DW. Recapitulating the Cancer Microenvironment Using Bioprinting Technology for Precision Medicine. MICROMACHINES 2021; 12:1122. [PMID: 34577765 PMCID: PMC8472267 DOI: 10.3390/mi12091122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/09/2021] [Accepted: 09/15/2021] [Indexed: 12/20/2022]
Abstract
The complex and heterogenous nature of cancer contributes to the development of cancer cell drug resistance. The construction of the cancer microenvironment, including the cell-cell interactions and extracellular matrix (ECM), plays a significant role in the development of drug resistance. Traditional animal models used in drug discovery studies have been associated with feasibility issues that limit the recapitulation of human functions; thus, in vitro models have been developed to reconstruct the human cancer system. However, conventional two-dimensional and three-dimensional (3D) in vitro cancer models are limited in their ability to emulate complex cancer microenvironments. Advances in technologies, including bioprinting and cancer microenvironment reconstruction, have demonstrated the potential to overcome some of the limitations of conventional models. This study reviews some representative bioprinted in vitro models used in cancer research, particularly fabrication strategies for modeling and consideration of essential factors needed for the reconstruction of the cancer microenvironment. In addition, we highlight recent studies that applied such models, including application in precision medicine using advanced bioprinting technologies to fabricate biomimetic cancer models. Furthermore, we discuss current challenges in 3D bioprinting and suggest possible strategies to construct in vitro models that better mimic the pathophysiology of the cancer microenvironment for application in clinical settings.
Collapse
Affiliation(s)
- Jisoo Kim
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
| | - Jinah Jang
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
- Department of Creative IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul 03722, Korea
| | - Dong-Woo Cho
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
32
|
Ravanbakhsh H, Bao G, Luo Z, Mongeau LG, Zhang YS. Composite Inks for Extrusion Printing of Biological and Biomedical Constructs. ACS Biomater Sci Eng 2021; 7:4009-4026. [PMID: 34510905 DOI: 10.1021/acsbiomaterials.0c01158] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Extrusion-based three-dimensional (3D) printing is an emerging technology for the fabrication of complex structures with various biological and biomedical applications. The method is based on the layer-by-layer construction of the product using a printable ink. The material used as the ink should possess proper rheological properties and desirable performances. Composite materials, which are extensively used in 3D printing applications, can improve the printability and offer superior performances for the printed constructs. Herein, we review composite inks with a focus on composite hydrogels. The properties of different additives including fibers and nanoparticles are discussed. The performances of various composite inks in biological and biomedical systems are delineated through analyzing the synergistic effects between the composite ink components. Different applications, including tissue engineering, tissue model engineering, soft robotics, and four-dimensional printing, are selected to demonstrate how 3D-printable composite inks are exploited to achieve various desired functionality. This review finally presents an outlook of future perspectives on the design of composite inks.
Collapse
Affiliation(s)
- Hossein Ravanbakhsh
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, Massachusetts 02139, United States.,Department of Mechanical Engineering, McGill University, Montreal, QC H3A0C3, Canada
| | - Guangyu Bao
- Department of Mechanical Engineering, McGill University, Montreal, QC H3A0C3, Canada
| | - Zeyu Luo
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, Massachusetts 02139, United States.,Department of Orthopedics, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Luc G Mongeau
- Department of Mechanical Engineering, McGill University, Montreal, QC H3A0C3, Canada
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
33
|
Shin DS, Anseth KS. Recent advances in 3D models of tumor invasion. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2021; 19:100310. [PMID: 34308009 PMCID: PMC8294077 DOI: 10.1016/j.cobme.2021.100310] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
This review presents recent advances in the design of in vitro cancer models to study tumor cell migration, metastasis, and invasion in three-dimensions (3D). These cancer models are divided into two categories based on the biophysiological processes and structures simulated, namely (i) spheroid invasion models or (ii) vascularization models. Some recent advances to spheroid invasion models include new methods to make them amenable to high-throughput settings. In vascularization models, cancer cell extravasation, intravasation, and angiogenesis have been emulated. Finally, 3D bioprinting and microfluidic technologies are allowing researchers to recapitulate some of the complex architectural and microenvironmental changes that can drive cancer cells migration from the extracellular matrix and basement membrane to blood vessels.
Collapse
Affiliation(s)
- Della S. Shin
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA 80303
| | - Kristi S. Anseth
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA 80303
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA 80303
| |
Collapse
|
34
|
3D Printing and Bioprinting to Model Bone Cancer: The Role of Materials and Nanoscale Cues in Directing Cell Behavior. Cancers (Basel) 2021; 13:cancers13164065. [PMID: 34439218 PMCID: PMC8391202 DOI: 10.3390/cancers13164065] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/04/2021] [Accepted: 08/06/2021] [Indexed: 12/12/2022] Open
Abstract
Bone cancer, both primary and metastatic, is characterized by a low survival rate. Currently, available models lack in mimicking the complexity of bone, of cancer, and of their microenvironment, leading to poor predictivity. Three-dimensional technologies can help address this need, by developing predictive models that can recapitulate the conditions for cancer development and progression. Among the existing tools to obtain suitable 3D models of bone cancer, 3D printing and bioprinting appear very promising, as they enable combining cells, biomolecules, and biomaterials into organized and complex structures that can reproduce the main characteristic of bone. The challenge is to recapitulate a bone-like microenvironment for analysis of stromal-cancer cell interactions and biological mechanics leading to tumor progression. In this review, existing approaches to obtain in vitro 3D-printed and -bioprinted bone models are discussed, with a focus on the role of biomaterials selection in determining the behavior of the models and its degree of customization. To obtain a reliable 3D bone model, the evaluation of different polymeric matrices and the inclusion of ceramic fillers is of paramount importance, as they help reproduce the behavior of both normal and cancer cells in the bone microenvironment. Open challenges and future perspectives are discussed to solve existing shortcomings and to pave the way for potential development strategies.
Collapse
|
35
|
Balachander GM, Kotcherlakota R, Nayak B, Kedaria D, Rangarajan A, Chatterjee K. 3D Tumor Models for Breast Cancer: Whither We Are and What We Need. ACS Biomater Sci Eng 2021; 7:3470-3486. [PMID: 34286955 DOI: 10.1021/acsbiomaterials.1c00230] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Three-dimensional (3D) models have led to a paradigm shift in disease modeling in vitro, particularly for cancer. The past decade has seen a phenomenal increase in the development of 3D models for various types of cancers with a focus on studying stemness, invasive behavior, angiogenesis, and chemoresistance of cancer cells, as well as contributions of its stroma, which has expanded our understanding of these processes. Cancer biology is moving into exploring the emerging hallmarks of cancer, such as inflammation, immune evasion, and reprogramming of energy metabolism. Studies into these emerging concepts have provided novel targets and treatment options such as antitumor immunotherapy. However, 3D models that can investigate the emerging hallmarks are few and underexplored. As commonly used immunocompromised mice and syngenic mice cannot accurately mimic human immunology, stromal interactions, and metabolism and require the use of prohibitively expensive humanized mice, there is tremendous scope to develop authentic 3D tumor models in these areas. Taking the specific case of breast cancer, we discuss the currently available 3D models, their applications to mimic signaling in cancer, tumor-stroma interactions, drug responses, and assessment of drug delivery systems and therapies. We discuss the lacunae in the development of 3D tumor models for the emerging hallmarks of cancer, for lesser-explored forms of breast cancer, and provide insights to develop such models. We discuss how the next generation of 3D models can provide a better mimic of human cancer modeling compared to xenograft models and the scope toward preclinical models and precision medicine.
Collapse
Affiliation(s)
- Gowri Manohari Balachander
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore-560012, India.,Department of Physiology, Yong Loo Lin School of Medicine, National University Health System, MD9-04-11, 2 Medical Drive, Singapore 117593, Singapore
| | - Rajesh Kotcherlakota
- Department of Materials Engineering, Indian Institute of Science, Bangalore-560012, India
| | - Biswadeep Nayak
- Department of Materials Engineering, Indian Institute of Science, Bangalore-560012, India.,Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843, United States.,Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore-560012, India
| | - Dhaval Kedaria
- Department of Materials Engineering, Indian Institute of Science, Bangalore-560012, India
| | - Annapoorni Rangarajan
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore-560012, India.,Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore-560012, India
| | - Kaushik Chatterjee
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore-560012, India.,Department of Materials Engineering, Indian Institute of Science, Bangalore-560012, India
| |
Collapse
|
36
|
Boix-Montesinos P, Soriano-Teruel PM, Armiñán A, Orzáez M, Vicent MJ. The past, present, and future of breast cancer models for nanomedicine development. Adv Drug Deliv Rev 2021; 173:306-330. [PMID: 33798642 PMCID: PMC8191594 DOI: 10.1016/j.addr.2021.03.018] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/19/2021] [Accepted: 03/26/2021] [Indexed: 02/07/2023]
Abstract
Even given recent advances in nanomedicine development of breast cancer treatment in recent years and promising results in pre-clinical models, cancer nanomedicines often fail at the clinical trial stage. Limitations of conventional in vitro models include the lack of representation of the stromal population, the absence of a three-dimensional (3D) structure, and a poor representation of inter-tumor and intra-tumor heterogeneity. Herein, we review those cell culture strategies that aim to overcome these limitations, including cell co-cultures, advanced 3D cell cultures, patient-derived cells, bioprinting, and microfluidics systems. The in vivo evaluation of nanomedicines must consider critical parameters that include the enhanced permeability and retention effect, the host's immune status, and the site of tumor implantation. Here, we critically discuss the advantages and limitations of current in vivo models and report how the improved selection and application of breast cancer models can improve the clinical translation of nanomedicines.
Collapse
Affiliation(s)
- Paz Boix-Montesinos
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Laboratory, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - Paula M Soriano-Teruel
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Laboratory, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain; Centro de Investigación Príncipe Felipe, Targeted Therapies on Cancer and Inflammation Laboratory, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - Ana Armiñán
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Laboratory, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - Mar Orzáez
- Centro de Investigación Príncipe Felipe, Targeted Therapies on Cancer and Inflammation Laboratory, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - María J Vicent
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Laboratory, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| |
Collapse
|
37
|
Tronolone JJ, Jain A. Engineering new microvascular networks on-chip: ingredients, assembly, and best practices. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2007199. [PMID: 33994903 PMCID: PMC8114943 DOI: 10.1002/adfm.202007199] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Indexed: 05/23/2023]
Abstract
Tissue engineered grafts show great potential as regenerative implants for diseased or injured tissues within the human body. However, these grafts suffer from poor nutrient perfusion and waste transport, thus decreasing their viability post-transplantation. Graft vascularization is therefore a major area of focus within tissue engineering because biologically relevant conduits for nutrient and oxygen perfusion can improve viability post-implantation. Many researchers utilize microphysiological systems as testing platforms for potential grafts due to an ability to integrate vascular networks as well as biological characteristics such as fluid perfusion, 3D architecture, compartmentalization of tissue-specific materials, and biophysical and biochemical cues. While many methods of vascularizing these systems exist, microvascular self-assembly has great potential for bench-to-clinic translation as it relies on naturally occurring physiological events. In this review, we highlight the past decade of literature and critically discuss the most important and tunable components yielding a self-assembled vascular network on chip: endothelial cell source, tissue-specific supporting cells, biomaterial scaffolds, biochemical cues, and biophysical forces. This article discusses the bioengineered systems of angiogenesis, vasculogenesis, and lymphangiogenesis, and includes a brief overview of multicellular systems. We conclude with future avenues of research to guide the next generation of vascularized microfluidic models and future tissue engineered grafts.
Collapse
Affiliation(s)
- James J Tronolone
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Abhishek Jain
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77808, USA
| |
Collapse
|
38
|
Guyon J, Chapouly C, Andrique L, Bikfalvi A, Daubon T. The Normal and Brain Tumor Vasculature: Morphological and Functional Characteristics and Therapeutic Targeting. Front Physiol 2021; 12:622615. [PMID: 33746770 PMCID: PMC7973205 DOI: 10.3389/fphys.2021.622615] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 01/25/2021] [Indexed: 12/20/2022] Open
Abstract
Glioblastoma is among the most common tumor of the central nervous system in adults. Overall survival has not significantly improved over the last decade, even with optimizing standard therapeutic care including extent of resection and radio- and chemotherapy. In this article, we review features of the brain vasculature found in healthy cerebral tissue and in glioblastoma. Brain vessels are of various sizes and composed of several vascular cell types. Non-vascular cells such as astrocytes or microglia also interact with the vasculature and play important roles. We also discuss in vitro engineered artificial blood vessels which may represent useful models for better understanding the tumor-vessel interaction. Finally, we summarize results from clinical trials with anti-angiogenic therapy alone or in combination, and discuss the value of these approaches for targeting glioblastoma.
Collapse
Affiliation(s)
- Joris Guyon
- INSERM, LAMC, U1029, University Bordeaux, Pessac, France
| | - Candice Chapouly
- INSERM, Biology of Cardiovascular Diseases, U1034, University Bordeaux, Pessac, France
| | - Laetitia Andrique
- INSERM, LAMC, U1029, University Bordeaux, Pessac, France.,VoxCell 3D Plateform, UMS TBMcore 3427, Bordeaux, France
| | | | - Thomas Daubon
- University Bordeaux, CNRS, IBGC, UMR 5095, Bordeaux, France
| |
Collapse
|
39
|
Torre M, Giannitelli SM, Mauri E, Trombetta M, Rainer A. Additive manufacturing of biomaterials. Soft Robot 2021. [DOI: 10.1016/bs.ache.2021.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
40
|
Craig M, Jenner AL, Namgung B, Lee LP, Goldman A. Engineering in Medicine To Address the Challenge of Cancer Drug Resistance: From Micro- and Nanotechnologies to Computational and Mathematical Modeling. Chem Rev 2020; 121:3352-3389. [PMID: 33152247 DOI: 10.1021/acs.chemrev.0c00356] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Drug resistance has profoundly limited the success of cancer treatment, driving relapse, metastasis, and mortality. Nearly all anticancer drugs and even novel immunotherapies, which recalibrate the immune system for tumor recognition and destruction, have succumbed to resistance development. Engineers have emerged across mechanical, physical, chemical, mathematical, and biological disciplines to address the challenge of drug resistance using a combination of interdisciplinary tools and skill sets. This review explores the developing, complex, and under-recognized role of engineering in medicine to address the multitude of challenges in cancer drug resistance. Looking through the "lens" of intrinsic, extrinsic, and drug-induced resistance (also referred to as "tolerance"), we will discuss three specific areas where active innovation is driving novel treatment paradigms: (1) nanotechnology, which has revolutionized drug delivery in desmoplastic tissues, harnessing physiochemical characteristics to destroy tumors through photothermal therapy and rationally designed nanostructures to circumvent cancer immunotherapy failures, (2) bioengineered tumor models, which have benefitted from microfluidics and mechanical engineering, creating a paradigm shift in physiologically relevant environments to predict clinical refractoriness and enabling platforms for screening drug combinations to thwart resistance at the individual patient level, and (3) computational and mathematical modeling, which blends in silico simulations with molecular and evolutionary principles to map mutational patterns and model interactions between cells that promote resistance. On the basis that engineering in medicine has resulted in discoveries in resistance biology and successfully translated to clinical strategies that improve outcomes, we suggest the proliferation of multidisciplinary science that embraces engineering.
Collapse
Affiliation(s)
- Morgan Craig
- Department of Mathematics and Statistics, University of Montreal, Montreal, Quebec H3C 3J7, Canada.,Sainte-Justine University Hospital Research Centre, Montreal, Quebec H3S 2G4, Canada
| | - Adrianne L Jenner
- Department of Mathematics and Statistics, University of Montreal, Montreal, Quebec H3C 3J7, Canada.,Sainte-Justine University Hospital Research Centre, Montreal, Quebec H3S 2G4, Canada
| | - Bumseok Namgung
- Division of Engineering in Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, United States.,Department of Medicine, Harvard Medical School, Boston, Massachusetts 02139, United States
| | - Luke P Lee
- Division of Engineering in Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, United States.,Department of Medicine, Harvard Medical School, Boston, Massachusetts 02139, United States
| | - Aaron Goldman
- Division of Engineering in Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, United States.,Department of Medicine, Harvard Medical School, Boston, Massachusetts 02139, United States
| |
Collapse
|
41
|
Kim J, Kong JS, Han W, Kim BS, Cho DW. 3D Cell Printing of Tissue/Organ-Mimicking Constructs for Therapeutic and Drug Testing Applications. Int J Mol Sci 2020; 21:E7757. [PMID: 33092184 PMCID: PMC7589604 DOI: 10.3390/ijms21207757] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/15/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023] Open
Abstract
The development of artificial tissue/organs with the functional maturity of their native equivalents is one of the long-awaited panaceas for the medical and pharmaceutical industries. Advanced 3D cell-printing technology and various functional bioinks are promising technologies in the field of tissue engineering that have enabled the fabrication of complex 3D living tissue/organs. Various requirements for these tissues, including a complex and large-volume structure, tissue-specific microenvironments, and functional vasculatures, have been addressed to develop engineered tissue/organs with native relevance. Functional tissue/organ constructs have been developed that satisfy such criteria and may facilitate both in vivo replenishment of damaged tissue and the development of reliable in vitro testing platforms for drug development. This review describes key developments in technologies and materials for engineering 3D cell-printed constructs for therapeutic and drug testing applications.
Collapse
Affiliation(s)
- Jongmin Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
| | - Jeong Sik Kong
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
| | - Wonil Han
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
| | - Byoung Soo Kim
- Future IT Innovation Laboratory, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
- Institute of Convergence Science, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
42
|
Yu C, Schimelman J, Wang P, Miller KL, Ma X, You S, Guan J, Sun B, Zhu W, Chen S. Photopolymerizable Biomaterials and Light-Based 3D Printing Strategies for Biomedical Applications. Chem Rev 2020; 120:10695-10743. [PMID: 32323975 PMCID: PMC7572843 DOI: 10.1021/acs.chemrev.9b00810] [Citation(s) in RCA: 241] [Impact Index Per Article: 48.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Since the advent of additive manufacturing, known commonly as 3D printing, this technology has revolutionized the biofabrication landscape and driven numerous pivotal advancements in tissue engineering and regenerative medicine. Many 3D printing methods were developed in short course after Charles Hull first introduced the power of stereolithography to the world. However, materials development was not met with the same enthusiasm and remained the bottleneck in the field for some time. Only in the past decade has there been deliberate development to expand the materials toolbox for 3D printing applications to meet the true potential of 3D printing technologies. Herein, we review the development of biomaterials suited for light-based 3D printing modalities with an emphasis on bioprinting applications. We discuss the chemical mechanisms that govern photopolymerization and highlight the application of natural, synthetic, and composite biomaterials as 3D printed hydrogels. Because the quality of a 3D printed construct is highly dependent on both the material properties and processing technique, we included a final section on the theoretical and practical aspects behind light-based 3D printing as well as ways to employ that knowledge to troubleshoot and standardize the optimization of printing parameters.
Collapse
Affiliation(s)
- Claire Yu
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Jacob Schimelman
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Pengrui Wang
- Materials Science and Engineering Program, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Kathleen L Miller
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Xuanyi Ma
- Department of Bioengineering, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Shangting You
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Jiaao Guan
- Department of Electrical and Computer Engineering, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Bingjie Sun
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Wei Zhu
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Shaochen Chen
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Materials Science and Engineering Program, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Department of Bioengineering, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Chemical Engineering Program, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| |
Collapse
|
43
|
Lee M, Rizzo R, Surman F, Zenobi-Wong M. Guiding Lights: Tissue Bioprinting Using Photoactivated Materials. Chem Rev 2020; 120:10950-11027. [DOI: 10.1021/acs.chemrev.0c00077] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Mihyun Lee
- Tissue Engineering + Biofabrication HPL J22, ETH Zürich, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| | - Riccardo Rizzo
- Tissue Engineering + Biofabrication HPL J22, ETH Zürich, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| | - František Surman
- Tissue Engineering + Biofabrication HPL J22, ETH Zürich, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| | - Marcy Zenobi-Wong
- Tissue Engineering + Biofabrication HPL J22, ETH Zürich, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| |
Collapse
|
44
|
Mahmoodi M, Ferdowsi S, Ebrahimi-Barough S, Kamian S, Ai J. Tissue engineering applications in breast cancer. J Med Eng Technol 2020; 44:162-168. [PMID: 32401543 DOI: 10.1080/03091902.2020.1757771] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
In Iran, breast cancer (BC) is the most prevalent cancer among women. The standard treatment for this cancer is partial or total removal of breast tissue, followed by chemotherapy and radiation. Tissue engineering (TE) has made new treatments for tissue loss in these patients by creating functional substitutes in the laboratory. In addition, cancer biology combined with TE provides a new strategy for evaluation of anti-BC therapy. Several innovations in TE have led to the design of scaffold or matrix based culture systems that more closely mimic the native extracellular matrix (ECM). Currently, engineered three-dimensional (3D) cultures are being developed for modelling of the tumour microenvironment. These 3D cultures fulfil the need for in vitro approaches that allow an accurate study of the molecular mechanisms and a better analysis of the drugs effect. In the present study, we review recent developments in utilising of TE in BC. Moreover, this review describes achievements of Iranian researchers in the field of breast TE.
Collapse
Affiliation(s)
- Mozaffar Mahmoodi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technology in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Radiology, Faculty of Paramedical Sciences, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Shirin Ferdowsi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technology in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shaghayegh Kamian
- Department of Radiotherapy Oncology, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technology in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|