101
|
Sayed IM, El-Hafeez AAA, Maity PP, Das S, Ghosh P. Modeling colorectal cancers using multidimensional organoids. Adv Cancer Res 2021; 151:345-383. [PMID: 34148617 PMCID: PMC8221168 DOI: 10.1016/bs.acr.2021.02.005] [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] [Indexed: 10/21/2022]
Abstract
Organoids have revolutionized cancer research as highly adaptable models that enable an array of experimental techniques to interrogate tissue morphology and function. Because they preserve the genetic, phenotypic, and behavioral traits of their source tissue, organoids have gained traction as the most relevant models for drug discovery, tracking therapeutic response and for personalized medicine. As organoids are indisputably becoming a mainstay of cancer research, this review specifically addresses how colon-derived organoids can be perfected as multidimensional, scalable, reproducible models of healthy, pre-neoplastic and neoplastic conditions of the colon and for use in high-throughput "Phase-0" human clinical trials-in-a-dish.
Collapse
Affiliation(s)
- Ibrahim M Sayed
- Department of Pathology, University of California, San Diego, CA, United States
| | - Amer Ali Abd El-Hafeez
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA, United States
| | - Priti P Maity
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA, United States
| | - Soumita Das
- Department of Pathology, University of California, San Diego, CA, United States; Rebecca and John Moore Comprehensive Cancer Center, University of California, San Diego, CA, United States; HUMANOID Center of Research Excellence (CoRE), University of California, San Diego, CA, United States.
| | - Pradipta Ghosh
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA, United States; Rebecca and John Moore Comprehensive Cancer Center, University of California, San Diego, CA, United States; Department of Medicine, University of California, San Diego, CA, United States; Veterans Affairs Medical Center, San Diego, CA, United States; HUMANOID Center of Research Excellence (CoRE), University of California, San Diego, CA, United States.
| |
Collapse
|
102
|
Sung JH. Multi-organ-on-a-chip for pharmacokinetics and toxicokinetic study of drugs. Expert Opin Drug Metab Toxicol 2021; 17:969-986. [PMID: 33764248 DOI: 10.1080/17425255.2021.1908996] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Accurate prediction of pharmacokinetic (PK) and toxicokinetics (TK) of drugs is imperative for successful development of new pharmaceutics. Although conventional in vitro methods for predicting the PK and TK of drugs are well established, limitations still exist and more advanced chip-based in vitro platforms combined with mathematical models can help researchers overcome the limitations. Areas covered: We will review recent progress in the development of multi-organ-on-a-chip platforms for predicting PK and TK of drugs, as well as mathematical approaches that can be combined with these platforms for experiment design, data analysis and in vitro-in vivo extrapolation (IVIVE) for application to humans. Expert opinion: Although there remain some challenges to be addressed, the remarkable progress in the area of multi-organ-on-a-chip in recent years indicate that we will see tangible outcomes that can be utilized in the pharmaceutical industry in near future.
Collapse
Affiliation(s)
- Jong Hwan Sung
- Department of Chemical Engineering, Hongik University, Seoul, sejong, Republic of Korea
| |
Collapse
|
103
|
Duzagac F, Saorin G, Memeo L, Canzonieri V, Rizzolio F. Microfluidic Organoids-on-a-Chip: Quantum Leap in Cancer Research. Cancers (Basel) 2021; 13:737. [PMID: 33578886 PMCID: PMC7916612 DOI: 10.3390/cancers13040737] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 01/24/2021] [Accepted: 01/29/2021] [Indexed: 02/06/2023] Open
Abstract
Organ-like cell clusters, so-called organoids, which exhibit self-organized and similar organ functionality as the tissue of origin, have provided a whole new level of bioinspiration for ex vivo systems. Microfluidic organoid or organs-on-a-chip platforms are a new group of micro-engineered promising models that recapitulate 3D tissue structure and physiology and combines several advantages of current in vivo and in vitro models. Microfluidics technology is used in numerous applications since it allows us to control and manipulate fluid flows with a high degree of accuracy. This system is an emerging tool for understanding disease development and progression, especially for personalized therapeutic strategies for cancer treatment, which provide well-grounded, cost-effective, powerful, fast, and reproducible results. In this review, we highlight how the organoid-on-a-chip models have improved the potential of efficiency and reproducibility of organoid cultures. More widely, we discuss current challenges and development on organoid culture systems together with microfluidic approaches and their limitations. Finally, we describe the recent progress and potential utilization in the organs-on-a-chip practice.
Collapse
Affiliation(s)
- Fahriye Duzagac
- Department of Molecular Sciences and Nanosystems, Ca’ Foscari University of Venice, 30123 Venezia, Italy; (F.D.); (G.S.)
| | - Gloria Saorin
- Department of Molecular Sciences and Nanosystems, Ca’ Foscari University of Venice, 30123 Venezia, Italy; (F.D.); (G.S.)
| | - Lorenzo Memeo
- Department of Experimental Oncology, Mediterranean Institute of Oncology (IOM), 95029 Catania, Italy;
| | - Vincenzo Canzonieri
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy;
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy
| | - Flavio Rizzolio
- Department of Molecular Sciences and Nanosystems, Ca’ Foscari University of Venice, 30123 Venezia, Italy; (F.D.); (G.S.)
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy;
| |
Collapse
|
104
|
Picollet-D'hahan N, Zuchowska A, Lemeunier I, Le Gac S. Multiorgan-on-a-Chip: A Systemic Approach To Model and Decipher Inter-Organ Communication. Trends Biotechnol 2021; 39:788-810. [PMID: 33541718 DOI: 10.1016/j.tibtech.2020.11.014] [Citation(s) in RCA: 185] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 12/14/2022]
Abstract
Multiorgan-on-a-chip (multi-OoC) platforms have great potential to redefine the way in which human health research is conducted. After briefly reviewing the need for comprehensive multiorgan models with a systemic dimension, we highlight scenarios in which multiorgan models are advantageous. We next overview existing multi-OoC platforms, including integrated body-on-a-chip devices and modular approaches involving interconnected organ-specific modules. We highlight how multi-OoC models can provide unique information that is not accessible using single-OoC models. Finally, we discuss remaining challenges for the realization of multi-OoC platforms and their worldwide adoption. We anticipate that multi-OoC technology will metamorphose research in biology and medicine by providing holistic and personalized models for understanding and treating multisystem diseases.
Collapse
Affiliation(s)
- Nathalie Picollet-D'hahan
- Université Grenoble Alpes, Institut National de la Santé et de la Recherche Médicale (INSERM), Commissariat à l'Energie Atomique (CEA) Interdisciplinary Research Institute of Grenoble (IRIG) Biomicrotechnology and Functional Genomics (BIOMICS), Grenoble, France.
| | - Agnieszka Zuchowska
- Applied Microfluidics for Bioengineering Research (AMBER), MESA+ Institute for Nanotechnology, TechMed Center, University of Twente, 7500AE Enschede, The Netherlands
| | - Iris Lemeunier
- Université Grenoble Alpes, Institut National de la Santé et de la Recherche Médicale (INSERM), Commissariat à l'Energie Atomique (CEA) Interdisciplinary Research Institute of Grenoble (IRIG) Biomicrotechnology and Functional Genomics (BIOMICS), Grenoble, France
| | - Séverine Le Gac
- Applied Microfluidics for Bioengineering Research (AMBER), MESA+ Institute for Nanotechnology, TechMed Center, University of Twente, 7500AE Enschede, The Netherlands.
| |
Collapse
|
105
|
Ziółkowska-Suchanek I. Mimicking Tumor Hypoxia in Non-Small Cell Lung Cancer Employing Three-Dimensional In Vitro Models. Cells 2021; 10:cells10010141. [PMID: 33445709 PMCID: PMC7828188 DOI: 10.3390/cells10010141] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/07/2021] [Accepted: 01/07/2021] [Indexed: 12/12/2022] Open
Abstract
Hypoxia is the most common microenvironment feature of lung cancer tumors, which affects cancer progression, metastasis and metabolism. Oxygen induces both proteomic and genomic changes within tumor cells, which cause many alternations in the tumor microenvironment (TME). This review defines current knowledge in the field of tumor hypoxia in non-small cell lung cancer (NSCLC), including biology, biomarkers, in vitro and in vivo studies and also hypoxia imaging and detection. While classic two-dimensional (2D) in vitro research models reveal some hypoxia dependent manifestations, three-dimensional (3D) cell culture models more accurately replicate the hypoxic TME. In this study, a systematic review of the current NSCLC 3D models that have been able to mimic the hypoxic TME is presented. The multicellular tumor spheroid, organoids, scaffolds, microfluidic devices and 3D bioprinting currently being utilized in NSCLC hypoxia studies are reviewed. Additionally, the utilization of 3D in vitro models for exploring biological and therapeutic parameters in the future is described.
Collapse
|
106
|
Ejiugwo M, Rochev Y, Gethin G, O'Connor G. Toward Developing Immunocompetent Diabetic Foot Ulcer-on-a-Chip Models for Drug Testing. Tissue Eng Part C Methods 2021; 27:77-88. [PMID: 33406980 DOI: 10.1089/ten.tec.2020.0331] [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: 12/15/2022] Open
Abstract
Bioengineering of skin has been significantly explored, ranging from the use of traditional cell culture systems to the most recent organ-on-a-chip (OoC) technology that permits skin modeling on physiological scales among other benefits. This article presents key considerations for developing physiologically relevant immunocompetent diabetic foot ulcer (DFU) models. Diabetic foot ulceration affects hundreds of millions of individuals globally, especially the elderly, and constitutes a major socioeconomic burden. When DFUs are not treated and managed in a timely manner, 15-50% of patients tend to undergo partial or complete amputation of the affected limb. Consequently, at least 40% of such patients die within 5 years postamputation. Currently, therapeutic strategies are actively sought and developed. However, present-day preclinical platforms (animals and in vitro models) are not robust enough to provide reliable data for clinical trials. Insights from published works on immunocompetent skin-on-a-chip models and bioengineering considerations, presented in this article, can inform researchers on how to develop robust OoC models for testing topical therapies such as growth factor-based therapies for DFUs. We propose that immunocompetent DFU-on-a-chip models should be bioengineered using diseased cells derived from individuals; in particular, the pathophysiological contribution of macrophages in diabetic wound healing, along with the typical fibroblasts and keratinocytes, needs to be recapitulated. The ideal model should consist of the following components: diseased cells embedded in reproducible scaffolds, which permit endogenous "diseased" extracellular matrix deposition, and the integration of the derived immunocompetent DFU model onto a microfluidic platform. The proposed DFU platforms will eventually facilitate reliable and robust drug testing of wound healing therapeutics, coupled with reduced clinical trial failure rates. Impact statement Current animal and cell-based systems are not physiologically relevant enough to retrieve reliable results for clinical translation of diabetic foot ulcer (DFU) therapies. Organ-on-a-chip (OoC) technology offers desirable features that could finally enable the vision of modeling DFU for pathophysiological studies and drug testing at a microscale. This article brings together the significant recent findings relevant to developing a minimally functional immunocompetent DFU-on-a-chip model, as wound healing cannot occur without a proper functioning immune response. It looks feasible in the future to recapitulate the stagnant inflammation in DFU (thought to impede wound healing) using OoC, diseased cells, and an endogenously produced extracellular matrix.
Collapse
Affiliation(s)
- Mirella Ejiugwo
- SFI CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, Galway City, Ireland.,School of Physics, and National University of Ireland Galway, Galway City, Ireland
| | - Yury Rochev
- SFI CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, Galway City, Ireland.,School of Physics, and National University of Ireland Galway, Galway City, Ireland
| | - Georgina Gethin
- SFI CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, Galway City, Ireland.,School of Nursing and Midwifery, National University of Ireland Galway, Galway City, Ireland
| | - Gerard O'Connor
- SFI CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, Galway City, Ireland.,School of Physics, and National University of Ireland Galway, Galway City, Ireland
| |
Collapse
|
107
|
Pars S, Achberger K, Kleger A, Liebau S, Pashkovskaia N. Generation of Functional Vascular Endothelial Cells and Pericytes from Keratinocyte Derived Human Induced Pluripotent Stem Cells. Cells 2021; 10:E74. [PMID: 33466396 PMCID: PMC7824831 DOI: 10.3390/cells10010074] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/18/2020] [Accepted: 12/30/2020] [Indexed: 12/24/2022] Open
Abstract
Human induced pluripotent stem cell (hiPSC)-derived endothelial cells (ECs) and pericytes provide a powerful tool for cardiovascular disease modelling, personalized drug testing, translational medicine, and tissue engineering. Here, we report a novel differentiation protocol that results in the fast and efficient production of ECs and pericytes from keratinocyte-derived hiPSCs. We found that the implementation of a 3D embryoid body (EB) stage significantly improves the differentiation efficiency. Compared with the monolayer-based technique, our protocol yields a distinct EC population with higher levels of EC marker expression such as CD31 and vascular endothelial cadherin (VE-cadherin). Furthermore, the EB-based protocol allows the generation of functional EC and pericyte populations that can promote blood vessel-like structure formation upon co-culturing. Moreover, we demonstrate that the EB-based ECs and pericytes can be successfully used in a microfluidic chip model, forming a stable 3D microvascular network. Overall, the described protocol can be used to efficiently differentiate both ECs and pericytes with distinct and high marker expression from keratinocyte-derived hiPSCs, providing a potent source material for future cardiovascular disease studies.
Collapse
Affiliation(s)
- Selin Pars
- Institute of Neuroanatomy & Developmental Biology (INDB), Eberhard Karls University Tübingen, Österbergstrasse 3, 72074 Tübingen, Germany; (S.P.); (K.A.); (S.L.)
| | - Kevin Achberger
- Institute of Neuroanatomy & Developmental Biology (INDB), Eberhard Karls University Tübingen, Österbergstrasse 3, 72074 Tübingen, Germany; (S.P.); (K.A.); (S.L.)
| | - Alexander Kleger
- Department of Internal Medicine 1, Ulm University Hospital, 89081 Ulm, Germany;
| | - Stefan Liebau
- Institute of Neuroanatomy & Developmental Biology (INDB), Eberhard Karls University Tübingen, Österbergstrasse 3, 72074 Tübingen, Germany; (S.P.); (K.A.); (S.L.)
| | - Natalia Pashkovskaia
- Institute of Neuroanatomy & Developmental Biology (INDB), Eberhard Karls University Tübingen, Österbergstrasse 3, 72074 Tübingen, Germany; (S.P.); (K.A.); (S.L.)
| |
Collapse
|
108
|
Yang S, Chen Z, Cheng Y, Liu T, Pu Y, Liang G. Environmental toxicology wars: Organ-on-a-chip for assessing the toxicity of environmental pollutants. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 268:115861. [PMID: 33120150 DOI: 10.1016/j.envpol.2020.115861] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 10/13/2020] [Accepted: 10/14/2020] [Indexed: 05/07/2023]
Abstract
Environmental pollution is a widespread problem, which has seriously threatened human health and led to an increase of human diseases. Therefore, it is critical to evaluate environmental pollutants quickly and efficiently. Because of obvious inter-species differences between animals and humans, and lack of physiologically-relevant microenvironment, animal models and in vitro two-dimensional (2D) models can not accurately describe toxicological effects and predicting actual in vivo responses. To make up the limitations of conventional environmental toxicology screening, organ-on-a-chip (OOC) systems are increasingly developing. OOC systems can provide a well-organized architecture with comparable to the complex microenvironment in vivo and generate realistic responses to environmental pollutants. The feasibility, adjustability and reliability of OCC systems make it possible to offer new opportunities for environmental pollutants screening, which can study their metabolism, collective response, and fate in vivo. Further progress can address the challenges to make OCC systems better investigate and evaluate environmental pollutants with high predictive power.
Collapse
Affiliation(s)
- Sheng Yang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, PR China, 210009.
| | - Zaozao Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, PR China, 210096.
| | - Yanping Cheng
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, PR China, 210009.
| | - Tong Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, PR China, 210009.
| | - Yuepu Pu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, PR China, 210009.
| | - Geyu Liang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, PR China, 210009.
| |
Collapse
|
109
|
Guo QR, Zhang LL, Liu JF, Li Z, Li JJ, Zhou WM, Wang H, Li JQ, Liu DY, Yu XY, Zhang JY. Multifunctional microfluidic chip for cancer diagnosis and treatment. Nanotheranostics 2021; 5:73-89. [PMID: 33391976 PMCID: PMC7738943 DOI: 10.7150/ntno.49614] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 10/08/2020] [Indexed: 02/06/2023] Open
Abstract
Microfluidic chip is not a chip in the traditional sense. It is technologies that control fluids at the micro level. As a burgeoning biochip, microfluidic chips integrate multiple disciplines, including physiology, pathology, cell biology, biophysics, engineering mechanics, mechanical design, materials science, and so on. The application of microfluidic chip has shown tremendous promise in the field of cancer therapy in the past three decades. Various types of cell and tissue cultures, including 2D cell culture, 3D cell culture and tissue organoid culture could be performed on microfluidic chips. Patient-derived cancer cells and tissues can be cultured on microfluidic chips in a visible, controllable, and high-throughput manner, which greatly advances the process of personalized medicine. Moreover, the functionality of microfluidic chip is greatly expanding due to the customizable nature. In this review, we introduce its application in developing cancer preclinical models, detecting cancer biomarkers, screening anti-cancer drugs, exploring tumor heterogeneity and producing nano-drugs. We highlight the functions and recent development of microfluidic chip to provide references for advancing cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Qiao-ru Guo
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R.China
| | - Ling-ling Zhang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R.China
| | - Ji-fang Liu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R.China
| | - Zhen Li
- Department of Gastroenterology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, P.R.China
| | - Jia-jun Li
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R.China
| | - Wen-min Zhou
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R.China
| | - Hui Wang
- Guangzhou Institute of Pediatrics/Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, P.R.China
| | - Jing-quan Li
- The First Affiliated Hospital, Hainan Medical University, Haikou, P.R.China
| | - Da-yu Liu
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, P.R.China
| | - Xi-yong Yu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R.China
| | - Jian-ye Zhang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R.China
- The First Affiliated Hospital, Hainan Medical University, Haikou, P.R.China
| |
Collapse
|
110
|
Xu M, Wang Y, Duan W, Xia S, Wei S, Liu W, Wang Q. Proteomic Reveals Reasons for Acquired Drug Resistance in Lung Cancer Derived Brain Metastasis Based on a Newly Established Multi-Organ Microfluidic Chip Model. Front Bioeng Biotechnol 2020; 8:612091. [PMID: 33415100 PMCID: PMC7783320 DOI: 10.3389/fbioe.2020.612091] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 11/25/2020] [Indexed: 12/30/2022] Open
Abstract
Anti-tumor drugs can effectively shrink the lesions of primary lung cancer; however, it has limited therapeutic effect on patients with brain metastasis (BM). A BM preclinical model based on a multi-organ microfluidic chip has been established proficiently in our previous work. In this study, the BM subpopulation (PC9-Br) derived from the parental PC9 cell line was isolated from the chip model and found to develop obvious resistance to antineoplastic drugs including chemotherapeutic agents (cisplatin, carboplatin, pemetrexed) and tyrosine kinase inhibitors (TKIs) which target epidermal growth factor receptor (EGFR); this suggested that the acquisition of drug-resistance by brain metastatic cells was attributable to the intrinsic changes in PC9-Br. Hence, we performed proteomic and revealed a greatly altered spectrum of BM protein expression compared with primary lung cancer cells. We identified the hyperactive glutathione (GSH) metabolism pathway with the overexpression of various GSH metabolism-related enzymes (GPX4, RRM2, GCLC, GPX1, GSTM4, GSTM1). Aldehyde dehydrogenases (ALDH1A1, ALDH3A1) were also found to be upregulated in BM. What's more, loss of EGFR and phosphorylated EGFR in PC9-Br gave reasons for the TKIs resistance. Collectively, our findings indicated potential mechanisms for the acquirement of drug resistance occurred in BM, providing new strategies to overcome therapeutic resistance in lung cancer BM.
Collapse
Affiliation(s)
- Mingxin Xu
- Department of Respiratory Medicine, The Second Hospital, Dalian Medical University, Dalian, China
| | - Yingyan Wang
- Laboratory Center for Diagnostics, Dalian Medical University, Dalian, China
| | - Wenzhe Duan
- Department of Respiratory Medicine, The Second Hospital, Dalian Medical University, Dalian, China
| | - Shengkai Xia
- Department of Respiratory Medicine, The Second Hospital, Dalian Medical University, Dalian, China
| | - Song Wei
- Department of Respiratory Medicine, The Second Hospital, Dalian Medical University, Dalian, China
| | - Wenwen Liu
- Cancer Translational Medicine Research Center, The Second Hospital, Dalian Medical University, Dalian, China
| | - Qi Wang
- Department of Respiratory Medicine, The Second Hospital, Dalian Medical University, Dalian, China.,Cancer Translational Medicine Research Center, The Second Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
111
|
Ma C, Peng Y, Li H, Chen W. Organ-on-a-Chip: A New Paradigm for Drug Development. Trends Pharmacol Sci 2020; 42:119-133. [PMID: 33341248 DOI: 10.1016/j.tips.2020.11.009] [Citation(s) in RCA: 285] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 01/16/2023]
Abstract
The pharmaceutical industry has been desperately searching for efficient drug discovery methods. Organ-on-a-Chip, a cutting-edge technology that can emulate the physiological environment and functionality of human organs on a chip for disease modeling and drug testing, shows great potential for revolutionizing the drug development pipeline. However, successful translation of this novel engineering platform into routine pharmacological and medical scenarios remains to be realized. In this review, we discuss how the Organ-on-a-Chip technology can have critical roles in different preclinical stages of drug development and highlight the current challenges in translation and commercialization of this technology for the pharmacological and medical end-users. Moreover, this review sheds light on the future developmental trends and need for a next-generation Organ-on-a-Chip platform to bridge the gap between animal studies and clinical trials for the pharmaceutical industry.
Collapse
Affiliation(s)
- Chao Ma
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY 11201, USA; Department of Biomedical Engineering, New York University, Brooklyn, NY 11201, USA
| | - Yansong Peng
- Department of Biomedical Engineering, New York University, Brooklyn, NY 11201, USA
| | - Hongtong Li
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY 11201, USA
| | - Weiqiang Chen
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY 11201, USA; Department of Biomedical Engineering, New York University, Brooklyn, NY 11201, USA; Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA.
| |
Collapse
|
112
|
Nikolakopoulou P, Rauti R, Voulgaris D, Shlomy I, Maoz BM, Herland A. Recent progress in translational engineered in vitro models of the central nervous system. Brain 2020; 143:3181-3213. [PMID: 33020798 PMCID: PMC7719033 DOI: 10.1093/brain/awaa268] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 06/17/2020] [Accepted: 06/21/2020] [Indexed: 02/07/2023] Open
Abstract
The complexity of the human brain poses a substantial challenge for the development of models of the CNS. Current animal models lack many essential human characteristics (in addition to raising operational challenges and ethical concerns), and conventional in vitro models, in turn, are limited in their capacity to provide information regarding many functional and systemic responses. Indeed, these challenges may underlie the notoriously low success rates of CNS drug development efforts. During the past 5 years, there has been a leap in the complexity and functionality of in vitro systems of the CNS, which have the potential to overcome many of the limitations of traditional model systems. The availability of human-derived induced pluripotent stem cell technology has further increased the translational potential of these systems. Yet, the adoption of state-of-the-art in vitro platforms within the CNS research community is limited. This may be attributable to the high costs or the immaturity of the systems. Nevertheless, the costs of fabrication have decreased, and there are tremendous ongoing efforts to improve the quality of cell differentiation. Herein, we aim to raise awareness of the capabilities and accessibility of advanced in vitro CNS technologies. We provide an overview of some of the main recent developments (since 2015) in in vitro CNS models. In particular, we focus on engineered in vitro models based on cell culture systems combined with microfluidic platforms (e.g. 'organ-on-a-chip' systems). We delve into the fundamental principles underlying these systems and review several applications of these platforms for the study of the CNS in health and disease. Our discussion further addresses the challenges that hinder the implementation of advanced in vitro platforms in personalized medicine or in large-scale industrial settings, and outlines the existing differentiation protocols and industrial cell sources. We conclude by providing practical guidelines for laboratories that are considering adopting organ-on-a-chip technologies.
Collapse
Affiliation(s)
- Polyxeni Nikolakopoulou
- AIMES, Center for the Advancement of Integrated Medical and Engineering Sciences, Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Rossana Rauti
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Dimitrios Voulgaris
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Iftach Shlomy
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Ben M Maoz
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel
| | - Anna Herland
- AIMES, Center for the Advancement of Integrated Medical and Engineering Sciences, Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden
| |
Collapse
|
113
|
Rodrigues RO, Sousa PC, Gaspar J, Bañobre-López M, Lima R, Minas G. Organ-on-a-Chip: A Preclinical Microfluidic Platform for the Progress of Nanomedicine. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2003517. [PMID: 33236819 DOI: 10.1002/smll.202003517] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/13/2020] [Indexed: 06/11/2023]
Abstract
Despite the progress achieved in nanomedicine during the last decade, the translation of new nanotechnology-based therapeutic systems into clinical applications has been slow, especially due to the lack of robust preclinical tissue culture platforms able to mimic the in vivo conditions found in the human body and to predict the performance and biotoxicity of the developed nanomaterials. Organ-on-a-chip (OoC) platforms are novel microfluidic tools that mimic complex human organ functions at the microscale level. These integrated microfluidic networks, with 3D tissue engineered models, have been shown high potential to reduce the discrepancies between the results derived from preclinical and clinical trials. However, there are many challenges that still need to be addressed, such as the integration of biosensor modules for long-time monitoring of different physicochemical and biochemical parameters. In this review, recent advances on OoC platforms, particularly on the preclinical validation of nanomaterials designed for cancer, as well as the current challenges and possible future directions for an end-use perspective are discussed.
Collapse
Affiliation(s)
- Raquel O Rodrigues
- Center for MicroElectromechanical Systems (CMEMS-UMinho), University of Minho, Campus de Azurém, Guimarães, 4800-058, Portugal
- Microfabrication and Exploratory Nanotechnology, INL-International Iberian Nanotechnology Laboratory, Av. Mestre José Veiga, Braga, 4715-330, Portugal
| | - Patrícia C Sousa
- Microfabrication and Exploratory Nanotechnology, INL-International Iberian Nanotechnology Laboratory, Av. Mestre José Veiga, Braga, 4715-330, Portugal
| | - João Gaspar
- Microfabrication and Exploratory Nanotechnology, INL-International Iberian Nanotechnology Laboratory, Av. Mestre José Veiga, Braga, 4715-330, Portugal
| | - Manuel Bañobre-López
- Advanced (magnetic) Theranostic Nanostructures Lab, Nanomedicine Unit, INL-International Iberian Nanotechnology Laboratory, Av. Mestre José Veiga, Braga, 4715-330, Portugal
| | - Rui Lima
- Transport Phenomena Research Center (CEFT), Faculdade de Engenharia da Universidade do Porto (FEUP), R. Dr. Roberto Frias, Porto, 4200-465, Portugal
- Mechanical Engineering and Resource Sustainability Center (MEtRICs), Mechanical Engineering Department, University of Minho, Campus de Azurém, Guimarães, 4800-058, Portugal
| | - Graça Minas
- Center for MicroElectromechanical Systems (CMEMS-UMinho), University of Minho, Campus de Azurém, Guimarães, 4800-058, Portugal
| |
Collapse
|
114
|
Li W, Sun X, Ji B, Yang X, Zhou B, Lu Z, Gao X. PLGA Nanofiber/PDMS Microporous Composite Membrane-Sandwiched Microchip for Drug Testing. MICROMACHINES 2020; 11:mi11121054. [PMID: 33260653 PMCID: PMC7760955 DOI: 10.3390/mi11121054] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/13/2020] [Accepted: 11/24/2020] [Indexed: 02/06/2023]
Abstract
Lung-on-a-chip devices could provide new strategies for a biomimetic lung cell microenvironment and construction of lung disease models in vitro, and are expected to greatly promote the development of drug evaluation, toxicological detection, and disease model building. In this study, we developed a novel poly (lactic-co-glycolic acid) (PLGA) nanofiber/polydimethylsiloxane (PDMS) microporous composite membrane-sandwiched lung-on-a-chip to perform anti-tumor drug testing. The composite membrane was characterized, and the results showed that it was permeable to molecules and thus could be used to study small-molecule drug diffusion. In addition, the microchip could apply perfusion fluids to simulate blood flow under extremely low fluid shear stress, and could also simulate the spherical-like shape of the alveoli by deformation of the composite membrane. Using this chip, we evaluated the anti-tumor drug efficacy of gefitinib in two kinds of non-small cell lung cancer cells, the lung adenocarcinoma NCI-H1650 cell line and the large cell lung cancer NCI-H460 cell line. We further probed the resistance of NCI-H460 cells to gefitinib under normoxic and hypoxic conditions. The established composite membrane-sandwiched lung chip can simulate more biochemical and biophysical factors in the lung physiological and pathological microenvironment, and it has important applications in the personalized treatment of lung tumors. It is expected to play a potential role in clinical diagnosis and drug screening.
Collapse
Affiliation(s)
- Wei Li
- Materials Genome Institute, Shanghai University, Shanghai 200444, China; (W.L.); (X.S.); (X.Y.)
| | - Xindi Sun
- Materials Genome Institute, Shanghai University, Shanghai 200444, China; (W.L.); (X.S.); (X.Y.)
| | - Bing Ji
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Taipa, Macau 999078, China; (B.J.); (B.Z.)
| | - Xingyuan Yang
- Materials Genome Institute, Shanghai University, Shanghai 200444, China; (W.L.); (X.S.); (X.Y.)
| | - Bingpu Zhou
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Taipa, Macau 999078, China; (B.J.); (B.Z.)
| | - Zhanjun Lu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Correspondence: (Z.L.); (X.G.)
| | - Xinghua Gao
- Materials Genome Institute, Shanghai University, Shanghai 200444, China; (W.L.); (X.S.); (X.Y.)
- Correspondence: (Z.L.); (X.G.)
| |
Collapse
|
115
|
Salminen AT, Allahyari Z, Gholizadeh S, McCloskey MC, Ajalik R, Cottle RN, Gaborski TR, McGrath JL. In vitro Studies of Transendothelial Migration for Biological and Drug Discovery. FRONTIERS IN MEDICAL TECHNOLOGY 2020; 2:600616. [PMID: 35047883 PMCID: PMC8757899 DOI: 10.3389/fmedt.2020.600616] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022] Open
Abstract
Inflammatory diseases and cancer metastases lack concrete pharmaceuticals for their effective treatment despite great strides in advancing our understanding of disease progression. One feature of these disease pathogeneses that remains to be fully explored, both biologically and pharmaceutically, is the passage of cancer and immune cells from the blood to the underlying tissue in the process of extravasation. Regardless of migratory cell type, all steps in extravasation involve molecular interactions that serve as a rich landscape of targets for pharmaceutical inhibition or promotion. Transendothelial migration (TEM), or the migration of the cell through the vascular endothelium, is a particularly promising area of interest as it constitutes the final and most involved step in the extravasation cascade. While in vivo models of cancer metastasis and inflammatory diseases have contributed to our current understanding of TEM, the knowledge surrounding this phenomenon would be significantly lacking without the use of in vitro platforms. In addition to the ease of use, low cost, and high controllability, in vitro platforms permit the use of human cell lines to represent certain features of disease pathology better, as seen in the clinic. These benefits over traditional pre-clinical models for efficacy and toxicity testing are especially important in the modern pursuit of novel drug candidates. Here, we review the cellular and molecular events involved in leukocyte and cancer cell extravasation, with a keen focus on TEM, as discovered by seminal and progressive in vitro platforms. In vitro studies of TEM, specifically, showcase the great experimental progress at the lab bench and highlight the historical success of in vitro platforms for biological discovery. This success shows the potential for applying these platforms for pharmaceutical compound screening. In addition to immune and cancer cell TEM, we discuss the promise of hepatocyte transplantation, a process in which systemically delivered hepatocytes must transmigrate across the liver sinusoidal endothelium to successfully engraft and restore liver function. Lastly, we concisely summarize the evolving field of porous membranes for the study of TEM.
Collapse
Affiliation(s)
- Alec T. Salminen
- Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Zahra Allahyari
- Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, United States
| | - Shayan Gholizadeh
- Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, United States
| | - Molly C. McCloskey
- Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Raquel Ajalik
- Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Renee N. Cottle
- Bioengineering, Clemson University, Clemson, SC, United States
| | - Thomas R. Gaborski
- Biomedical Engineering, University of Rochester, Rochester, NY, United States
- Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, United States
| | - James L. McGrath
- Biomedical Engineering, University of Rochester, Rochester, NY, United States
| |
Collapse
|
116
|
Azadi S, Tafazzoli Shadpour M, Warkiani ME. Characterizing the effect of substrate stiffness on the extravasation potential of breast cancer cells using a 3D microfluidic model. Biotechnol Bioeng 2020; 118:823-835. [PMID: 33111314 DOI: 10.1002/bit.27612] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 10/02/2020] [Accepted: 10/20/2020] [Indexed: 12/14/2022]
Abstract
Different biochemical and biomechanical cues from tumor microenvironment affect the extravasation of cancer cells to distant organs; among them, the mechanical signals are poorly understood. Although the effect of substrate stiffness on the primary migration of cancer cells has been previously probed, its role in regulating the extravasation ability of cancer cells is still vague. Herein, we used a microfluidic device to mimic the extravasation of tumor cells in a 3D microenvironment containing cancer cells, endothelial cells, and the biological matrix. The microfluidic-based extravasation model was utilized to probe the effect of substrate stiffness on the invasion ability of breast cancer cells. MCF7 and MDA-MB-231 cancer cells were cultured among substrates with different stiffness which followed by monitoring their extravasation capability through the microfluidic device. Our results demonstrated that acidic collagen at a concentration of 2.5 mg/ml promotes migration of cancer cells. Additionally, the substrate softening resulted in up to 46% reduction in the invasion of breast cancer cells. The substrate softening not only affected the number of extravasated cells but also reduced their migration distance up to 53%. We further investigated the secreted level of matrix metalloproteinase 9 (MMP9) and identified that there is a positive correlation between substrate stiffening, MMP9 concentration, and extravasation of cancer cells. These findings suggest that the substrate stiffness mediates the cancer cells extravasation in a microfluidic model. Changes in MMP9 level could be one of the possible underlying mechanisms which need more investigations to be addressed thoroughly.
Collapse
Affiliation(s)
- Shohreh Azadi
- Faculty of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | | | - Majid E Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales, Australia.,Institute of Molecular Medicine, Sechenov University, Moscow, Russia
| |
Collapse
|
117
|
Yanagisawa K, Konno M, Liu H, Irie S, Mizushima T, Mori M, Doki Y, Eguchi H, Matsusaki M, Ishii H. A Four-Dimensional Organoid System to Visualize Cancer Cell Vascular Invasion. BIOLOGY 2020; 9:biology9110361. [PMID: 33120912 PMCID: PMC7692192 DOI: 10.3390/biology9110361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 10/08/2020] [Accepted: 10/22/2020] [Indexed: 12/24/2022]
Abstract
Simple Summary Using vascular organoid culture with collagen microfiber, we have established a method for culturing organoids that recapitulates the vascular invasion process of cancer cells. This culture model made it possible to four-dimensionally evaluate the dynamics of cancer cells infiltrating into blood vessels. Abstract Vascular invasion of cancer is a critical step in cancer progression, but no drug has been developed to inhibit vascular invasion. To achieve the eradication of cancer metastasis, elucidation of the mechanism for vascular invasion and the development of innovative treatment methods are required. Here, a simple and reproducible vascular invasion model is established using a vascular organoid culture in a fibrin gel with collagen microfibers. Using this model, it was possible to observe and evaluate the cell dynamics and histological positional relationship of invasive cancer cells in four dimensions. Cancer-derived exosomes promoted the vascular invasion of cancer cells and loosened tight junctions in the vascular endothelium. As a new evaluation method, research using this vascular invasion mimic model will be advanced, and applications to the evaluation of the vascular invasion suppression effect of a drug are expected.
Collapse
Affiliation(s)
- Kiminori Yanagisawa
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan; (K.Y.); (M.K.); (T.M.); (M.M.); (Y.D.); (H.E.)
- Center of Molecular Innovation and Translational Research, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Masamitsu Konno
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan; (K.Y.); (M.K.); (T.M.); (M.M.); (Y.D.); (H.E.)
- Center of Molecular Innovation and Translational Research, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Hao Liu
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka 565-0871, Japan;
| | - Shinji Irie
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduated School of Engineering, Osaka University, Suita, Osaka 565-0871, Japan;
| | - Tsunekazu Mizushima
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan; (K.Y.); (M.K.); (T.M.); (M.M.); (Y.D.); (H.E.)
| | - Masaki Mori
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan; (K.Y.); (M.K.); (T.M.); (M.M.); (Y.D.); (H.E.)
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 819-0395, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan; (K.Y.); (M.K.); (T.M.); (M.M.); (Y.D.); (H.E.)
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan; (K.Y.); (M.K.); (T.M.); (M.M.); (Y.D.); (H.E.)
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka 565-0871, Japan;
- Correspondence: (M.M.); (H.I.); Tel.: +81-(0)6-6879-7357 (M.M.); +81-(0)6-6879-3251 or +81-(0)6-6210-8406 (H.I.); Fax: +81-(0)6-6879-7359 (M.M.); +81-(0)6-6879-3259 or +81-(0)6-4703-2856 (H.I.)
| | - Hideshi Ishii
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan; (K.Y.); (M.K.); (T.M.); (M.M.); (Y.D.); (H.E.)
- Center of Molecular Innovation and Translational Research, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
- Correspondence: (M.M.); (H.I.); Tel.: +81-(0)6-6879-7357 (M.M.); +81-(0)6-6879-3251 or +81-(0)6-6210-8406 (H.I.); Fax: +81-(0)6-6879-7359 (M.M.); +81-(0)6-6879-3259 or +81-(0)6-4703-2856 (H.I.)
| |
Collapse
|
118
|
Porter RJ, Murray GI, McLean MH. Current concepts in tumour-derived organoids. Br J Cancer 2020; 123:1209-1218. [PMID: 32728094 PMCID: PMC7555542 DOI: 10.1038/s41416-020-0993-5] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 05/28/2020] [Accepted: 07/03/2020] [Indexed: 12/13/2022] Open
Abstract
Cancer comprises a collection of highly proliferative and heterogeneous cells growing within an adaptive and evolving tumour microenvironment. Cancer survival rates have significantly improved following decades of cancer research. However, many experimental and preclinical studies do not translate to the bedside, reflecting the challenges of modelling the complexities and multicellular basis of human disease. Organoids are novel, complex, three-dimensional ex vivo tissue cultures that are derived from embryonic stem cells, induced pluripotent stem cells or tissue-resident progenitor cells, and represent a near-physiological model for studying cancer. Organoids develop by self-organisation, and can accurately represent the diverse genetic, cellular and pathophysiological hallmarks of cancer. In addition, co-culture methods and the ability to genetically manipulate these organoids have widened their utility in cancer research. Organoids thus offer a new and exciting platform for studying cancer and directing personalised therapies. This review aims to highlight how organoids are shaping the future of cancer research.
Collapse
Affiliation(s)
- Ross J Porter
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Scotland, UK
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Scotland, UK
| | - Graeme I Murray
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Scotland, UK
| | - Mairi H McLean
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Scotland, UK.
| |
Collapse
|
119
|
Three-Dimensional Culture System of Cancer Cells Combined with Biomaterials for Drug Screening. Cancers (Basel) 2020; 12:cancers12102754. [PMID: 32987868 PMCID: PMC7601447 DOI: 10.3390/cancers12102754] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/17/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary For the research and development of drug discovery, it is of prime importance to construct the three-dimensional (3D) tissue models in vitro. To this end, the enhancement design of cell function and activity by making use of biomaterials is essential. In this review, 3D culture systems of cancer cells combined with several biomaterials for anticancer drug screening are introduced. Abstract Anticancer drug screening is one of the most important research and development processes to develop new drugs for cancer treatment. However, there is a problem resulting in gaps between the in vitro drug screening and preclinical or clinical study. This is mainly because the condition of cancer cell culture is quite different from that in vivo. As a trial to mimic the in vivo cancer environment, there has been some research on a three-dimensional (3D) culture system by making use of biomaterials. The 3D culture technologies enable us to give cancer cells an in vitro environment close to the in vivo condition. Cancer cells modified to replicate the in vivo cancer environment will promote the biological research or drug discovery of cancers. This review introduces the in vitro research of 3D cell culture systems with biomaterials in addition to a brief summary of the cancer environment.
Collapse
|
120
|
Maschmeyer I, Kakava S. Organ-on-a-Chip. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2020; 179:311-342. [PMID: 32948885 DOI: 10.1007/10_2020_135] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Limitations of the current tools used in the drug development process, cell cultures, and animal models have highlighted the need for a new powerful tool that can emulate the human physiology in vitro. Advances in the field of microfluidics have made the realization of this tool closer than ever. Organ-on-a-chip platforms have been the first step forward, leading to the combination and integration of multiple organ models in the same platform with human-on-a-chip being the ultimate goal. Despite the current progress and technological developments, there are still several unmet engineering and biological challenges curtailing their development and widespread application in the biomedical field. The potentials, challenges, and current work on this unprecedented tool are being discussed in this chapter.
Collapse
|
121
|
Cai X, Briggs RG, Homburg HB, Young IM, Davis EJ, Lin YH, Battiste JD, Sughrue ME. Application of microfluidic devices for glioblastoma study: current status and future directions. Biomed Microdevices 2020; 22:60. [PMID: 32870410 DOI: 10.1007/s10544-020-00516-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Glioblastoma (GBM) is one of the most malignant primary brain tumors. This neoplasm is the hardest to treat and has a bad prognosis. Because of the characteristics of genetic heterogeneity and frequent recurrence, a successful cure for the disease is unlikely. Increasing evidence has revealed that the GBM stem cell-like cells (GSCs) and microenvironment are key elements in GBM recurrence and treatment failure. To better understand the mechanisms underlying this disease and to develop more effective therapeutic strategies for treatment, suitable approaches, techniques, and model systems closely mimicking real GBM conditions are required. Microfluidic devices, a model system mimicking the in vivo brain microenvironment, provide a very useful tool to analyze GBM cell behavior, their correlation with tumor malignancy, and the efficacy of multiple drug treatment. This paper reviews the applications of microfluidic devices in GBM research and summarizes progress and perspectives in this field.
Collapse
Affiliation(s)
- Xue Cai
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Robert G Briggs
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Hannah B Homburg
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | | | | | - Yueh-Hsin Lin
- Centre for Minimally Invasive Neurosurgery, Prince of Wales Private Hospital, Sydney, Australia
| | - James D Battiste
- Department of Neurology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Michael E Sughrue
- Cingulum Health, Sydney, Australia.
- Centre for Minimally Invasive Neurosurgery, Prince of Wales Private Hospital, Suite 19, Level 7, Barker Street, Randwick, New South Wales, 2031, Australia.
| |
Collapse
|
122
|
Hou W, Hu S, Yong KT, Zhang J, Ma H. Cigarette smoke-induced malignant transformation via STAT3 signalling in pulmonary epithelial cells in a lung-on-a-chip model. Biodes Manuf 2020. [DOI: 10.1007/s42242-020-00092-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
123
|
Laranga R, Duchi S, Ibrahim T, Guerrieri AN, Donati DM, Lucarelli E. Trends in Bone Metastasis Modeling. Cancers (Basel) 2020; 12:E2315. [PMID: 32824479 PMCID: PMC7464021 DOI: 10.3390/cancers12082315] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/11/2020] [Accepted: 08/14/2020] [Indexed: 12/12/2022] Open
Abstract
Bone is one of the most common sites for cancer metastasis. Bone tissue is composed by different kinds of cells that coexist in a coordinated balance. Due to the complexity of bone, it is impossible to capture the intricate interactions between cells under either physiological or pathological conditions. Hence, a variety of in vivo and in vitro approaches have been developed. Various models of tumor-bone diseases are routinely used to provide valuable information on the relationship between metastatic cancer cells and the bone tissue. Ideally, when modeling the metastasis of human cancers to bone, models would replicate the intra-tumor heterogeneity, as well as the genetic and phenotypic changes that occur with human cancers; such models would be scalable and reproducible to allow high-throughput investigation. Despite the continuous progress, there is still a lack of solid, amenable, and affordable models that are able to fully recapitulate the biological processes happening in vivo, permitting a correct interpretation of results. In the last decades, researchers have demonstrated that three-dimensional (3D) methods could be an innovative approach that lies between bi-dimensional (2D) models and animal models. Scientific evidence supports that the tumor microenvironment can be better reproduced in a 3D system than a 2D cell culture, and the 3D systems can be scaled up for drug screening in the same way as the 2D systems thanks to the current technologies developed. However, 3D models cannot completely recapitulate the inter- and intra-tumor heterogeneity found in patients. In contrast, ex vivo cultures of fragments of bone preserve key cell-cell and cell-matrix interactions and allow the study of bone cells in their natural 3D environment. Moreover, ex vivo bone organ cultures could be a better model to resemble the human pathogenic metastasis condition and useful tools to predict in vivo response to therapies. The aim of our review is to provide an overview of the current trends in bone metastasis modeling. By showing the existing in vitro and ex vivo systems, we aspire to contribute to broaden the knowledge on bone metastasis models and make these tools more appealing for further translational studies.
Collapse
Affiliation(s)
- Roberta Laranga
- Unit of Orthopaedic Pathology and Osteoarticular Tissue Regeneration, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (R.L.); (D.M.D.); (E.L.)
| | - Serena Duchi
- BioFab3D@ACMD, St Vincent’s Hospital, Melbourne, VIC 3065, Australia;
- Department of Surgery, St Vincent’s Hospital, University of Melbourne, Melbourne, VIC 3065, Australia
| | - Toni Ibrahim
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy;
| | - Ania Naila Guerrieri
- Unit of Orthopaedic Pathology and Osteoarticular Tissue Regeneration, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (R.L.); (D.M.D.); (E.L.)
| | - Davide Maria Donati
- Unit of Orthopaedic Pathology and Osteoarticular Tissue Regeneration, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (R.L.); (D.M.D.); (E.L.)
- Rizzoli Laboratory Unit, Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum University of Bologna, Via di Barbiano 1/10, 40136 Bologna, Italy
- 3rd Orthopaedic and Traumatologic Clinic Prevalently Oncologic, IRCCS Istituto Ortopedico Rizzoli, Via Pupilli 1, 40136 Bologna, Italy
| | - Enrico Lucarelli
- Unit of Orthopaedic Pathology and Osteoarticular Tissue Regeneration, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (R.L.); (D.M.D.); (E.L.)
| |
Collapse
|
124
|
Zanoni M, Cortesi M, Zamagni A, Arienti C, Pignatta S, Tesei A. Modeling neoplastic disease with spheroids and organoids. J Hematol Oncol 2020; 13:97. [PMID: 32677979 PMCID: PMC7364537 DOI: 10.1186/s13045-020-00931-0] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 07/02/2020] [Indexed: 12/15/2022] Open
Abstract
Cancer is a complex disease in which both genetic defects and microenvironmental components contribute to the development, progression, and metastasization of disease, representing major hurdles in the identification of more effective and safer treatment regimens for patients. Three-dimensional (3D) models are changing the paradigm of preclinical cancer research as they more closely resemble the complex tissue environment and architecture found in clinical tumors than in bidimensional (2D) cell cultures. Among 3D models, spheroids and organoids represent the most versatile and promising models in that they are capable of recapitulating the heterogeneity and pathophysiology of human cancers and of filling the gap between conventional 2D in vitro testing and animal models. Such 3D systems represent a powerful tool for studying cancer biology, enabling us to model the dynamic evolution of neoplastic disease from the early stages to metastatic dissemination and the interactions with the microenvironment. Spheroids and organoids have recently been used in the field of drug discovery and personalized medicine. The combined use of 3D models could potentially improve the robustness and reliability of preclinical research data, reducing the need for animal testing and favoring their transition to clinical practice. In this review, we summarize the recent advances in the use of these 3D systems for cancer modeling, focusing on their innovative translational applications, looking at future challenges, and comparing them with most widely used animal models.
Collapse
Affiliation(s)
- Michele Zanoni
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy.
| | - Michela Cortesi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Alice Zamagni
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Chiara Arienti
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Sara Pignatta
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Anna Tesei
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy.
| |
Collapse
|
125
|
Molina ER, Chim LK, Barrios S, Ludwig JA, Mikos AG. Modeling the Tumor Microenvironment and Pathogenic Signaling in Bone Sarcoma. TISSUE ENGINEERING. PART B, REVIEWS 2020; 26:249-271. [PMID: 32057288 PMCID: PMC7310212 DOI: 10.1089/ten.teb.2019.0302] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 01/07/2020] [Indexed: 12/17/2022]
Abstract
Investigations of cancer biology and screening of potential therapeutics for efficacy and safety begin in the preclinical laboratory setting. A staple of most basic research in cancer involves the use of tissue culture plates, on which immortalized cell lines are grown in monolayers. However, this practice has been in use for over six decades and does not account for vital elements of the tumor microenvironment that are thought to aid in initiation, propagation, and ultimately, metastasis of cancer. Furthermore, information gleaned from these techniques does not always translate to animal models or, more crucially, clinical trials in cancer patients. Osteosarcoma (OS) and Ewing sarcoma (ES) are the most common primary tumors of bone, but outcomes for patients with metastatic or recurrent disease have stagnated in recent decades. The unique elements of the bone tumor microenvironment have been shown to play critical roles in the pathogenesis of these tumors and thus should be incorporated in the preclinical models of these diseases. In recent years, the field of tissue engineering has leveraged techniques used in designing scaffolds for regenerative medicine to engineer preclinical tumor models that incorporate spatiotemporal control of physical and biological elements. We herein review the clinical aspects of OS and ES, critical elements present in the sarcoma microenvironment, and engineering approaches to model the bone tumor microenvironment. Impact statement The current paradigm of cancer biology investigation and therapeutic testing relies heavily on monolayer, monoculture methods developed over half a century ago. However, these methods often lack essential hallmarks of the cancer microenvironment that contribute to tumor pathogenesis. Tissue engineers incorporate scaffolds, mechanical forces, cells, and bioactive signals into biological environments to drive cell phenotype. Investigators of bone sarcomas, aggressive tumors that often rob patients of decades of life, have begun to use tissue engineering techniques to devise in vitro models for these diseases. Their efforts highlight how critical elements of the cancer microenvironment directly affect tumor signaling and pathogenesis.
Collapse
Affiliation(s)
- Eric R. Molina
- Department of Bioengineering, Rice University, Houston, Texas
| | - Letitia K. Chim
- Department of Bioengineering, Rice University, Houston, Texas
| | - Sergio Barrios
- Department of Bioengineering, Rice University, Houston, Texas
| | - Joseph A. Ludwig
- Division of Cancer Medicine, Department of Sarcoma Medical Oncology, MD Anderson Cancer Center, The University of Texas, Houston, Texas
| | | |
Collapse
|
126
|
Radhakrishnan J, Varadaraj S, Dash SK, Sharma A, Verma RS. Organotypic cancer tissue models for drug screening: 3D constructs, bioprinting and microfluidic chips. Drug Discov Today 2020; 25:879-890. [DOI: 10.1016/j.drudis.2020.03.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 02/09/2020] [Accepted: 03/03/2020] [Indexed: 12/20/2022]
|
127
|
Dynamic Culture Systems and 3D Interfaces Models for Cancer Drugs Testing. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1230:137-159. [PMID: 32285369 DOI: 10.1007/978-3-030-36588-2_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
Abstract
The mass use of biological agents for pharmaceutical purposes started with the development and distribution of vaccines, followed by the industrial production of antibiotics. The use of dynamic systems, such as bioreactors, had been already applied in the food industry in fermentation processes and started being used for the development of pharmaceutical agents from this point on. In the last decades, the use of bioreactors and microfluidic systems has been expanded in different fields. The emergence of the tissue engineering led to the development of in vitro models cultured in dynamic systems. This is particularly relevant considering the urgent reduction of the total dependence on animal disease models that is undermining the development of novel drugs, using alternatively human-based models to make the drug discovery process more reliable. The failure out coming from animal models has been more prevalent in certain types of cancer, such as glioblastoma multiform and in high-grade metastatic cancers like bone metastasis of breast or prostatic cancer. The difficulty in obtaining novel drugs for these purposes is mostly linked to the barriers around the tumors, which these bioactive molecules have to overcome to become effective. For that reason, the individualized study of each interface is paramount and is only realistic once applying human-based samples (e.g. cells or tissues) in three-dimensions for in vitro modeling under dynamic conditions. In this chapter, the most recent approaches to model these interfaces in 3D systems will be explored, highlighting their major contributions to the field. In this section, these systems' impact on increased knowledge in relevant aspects of cancer aggressiveness as invasive or motile cellular capacity, or even resistance to chemotherapeutic agents will have particular focus. The last section of this chapter will focus on the integration of the tumor interfaces in dynamic systems, particularly its application on high-throughput drug screening. The industrial translation of such platforms will be discussed, as well as the main upcoming challenges and future perspectives.
Collapse
|
128
|
Pisani P, Airoldi M, Allais A, Aluffi Valletti P, Battista M, Benazzo M, Briatore R, Cacciola S, Cocuzza S, Colombo A, Conti B, Costanzo A, della Vecchia L, Denaro N, Fantozzi C, Galizia D, Garzaro M, Genta I, Iasi GA, Krengli M, Landolfo V, Lanza GV, Magnano M, Mancuso M, Maroldi R, Masini L, Merlano MC, Piemonte M, Pisani S, Prina-Mello A, Prioglio L, Rugiu MG, Scasso F, Serra A, Valente G, Zannetti M, Zigliani A. Metastatic disease in head & neck oncology. ACTA OTORHINOLARYNGOLOGICA ITALICA : ORGANO UFFICIALE DELLA SOCIETA ITALIANA DI OTORINOLARINGOLOGIA E CHIRURGIA CERVICO-FACCIALE 2020; 40:S1-S86. [PMID: 32469009 PMCID: PMC7263073 DOI: 10.14639/0392-100x-suppl.1-40-2020] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The head and neck district represents one of the most frequent sites of cancer, and the percentage of metastases is very high in both loco-regional and distant areas. Prognosis refers to several factors: a) stage of disease; b) loco-regional relapses; c) distant metastasis. At diagnosis, distant metastases of head and neck cancers are present in about 10% of cases with an additional 20-30% developing metastases during the course of their disease. Diagnosis of distant metastases is associated with unfavorable prognosis, with a median survival of about 10 months. The aim of the present review is to provide an update on distant metastasis in head and neck oncology. Recent achievements in molecular profiling, interaction between neoplastic tissue and the tumor microenvironment, oligometastatic disease concepts, and the role of immunotherapy have all deeply changed the therapeutic approach and disease control. Firstly, we approach topics such as natural history, epidemiology of distant metastases and relevant pathological and radiological aspects. Focus is then placed on the most relevant clinical aspects; particular attention is reserved to tumours with distant metastasis and positive for EBV and HPV, and the oligometastatic concept. A substantial part of the review is dedicated to different therapeutic approaches. We highlight the role of immunotherapy and the potential effects of innovative technologies. Lastly, we present ethical and clinical perspectives related to frailty in oncological patients and emerging difficulties in sustainable socio-economical governance.
Collapse
Affiliation(s)
- Paolo Pisani
- ENT Unit, ASL AT, “Cardinal Massaja” Hospital, Asti, Italy
| | - Mario Airoldi
- Medical Oncology, Città della Salute e della Scienza, Torino, Italy
| | | | - Paolo Aluffi Valletti
- SCDU Otorinolaringoiatria, AOU Maggiore della Carità di Novara, Università del Piemonte Orientale, Italy
| | | | - Marco Benazzo
- SC Otorinolaringoiatria, Fondazione IRCCS Policlinico “S. Matteo”, Università di Pavia, Italy
| | | | | | - Salvatore Cocuzza
- Department of Medical, Surgical and Advanced Technologies “G.F. Ingrassia”, University of Catania, Italy
| | - Andrea Colombo
- ENT Unit, ASL AT, “Cardinal Massaja” Hospital, Asti, Italy
| | - Bice Conti
- Department of Drug Sciences, University of Pavia, Italy
- Polymerix S.r.L., Pavia, Italy
| | | | - Laura della Vecchia
- Unit of Otorhinolaryngology General Hospital “Macchi”, ASST dei Settelaghi, Varese, Italy
| | - Nerina Denaro
- Oncology Department A.O.S. Croce & Carle, Cuneo, Italy
| | | | - Danilo Galizia
- Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo,Italy
| | - Massimiliano Garzaro
- SCDU Otorinolaringoiatria, AOU Maggiore della Carità di Novara, Università del Piemonte Orientale, Italy
| | - Ida Genta
- Department of Drug Sciences, University of Pavia, Italy
- Polymerix S.r.L., Pavia, Italy
| | | | - Marco Krengli
- Dipartimento Medico Specialistico ed Oncologico, SC Radioterapia Oncologica, AOU Maggiore della Carità, Novara, Italy
- Dipartimento di Medicina Traslazionale, Università del Piemonte Orientale, Novara, Italy
| | | | - Giovanni Vittorio Lanza
- S.O.C. Chirurgia Toracica, Azienda Ospedaliera Nazionale “SS. Antonio e Biagio e Cesare Arrigo”, Alessandria, Italy
| | | | - Maurizio Mancuso
- S.O.C. Chirurgia Toracica, Azienda Ospedaliera Nazionale “SS. Antonio e Biagio e Cesare Arrigo”, Alessandria, Italy
| | - Roberto Maroldi
- Department of Radiology, University of Brescia, ASST Spedali Civili Brescia, Italy
| | - Laura Masini
- Dipartimento Medico Specialistico ed Oncologico, SC Radioterapia Oncologica, AOU Maggiore della Carità, Novara, Italy
| | - Marco Carlo Merlano
- Oncology Department A.O.S. Croce & Carle, Cuneo, Italy
- Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo,Italy
| | - Marco Piemonte
- ENT Unit, University Hospital “Santa Maria della Misericordia”, Udine, Italy
| | - Silvia Pisani
- Immunology and Transplantation Laboratory Fondazione IRCCS Policlinico “S. Matteo”, Pavia, Italy
| | - Adriele Prina-Mello
- LBCAM, Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin 8, Ireland
- Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN), Trinity College Dublin, Dublin 2, Ireland
| | - Luca Prioglio
- Department of Otorhinolaryngology, ASL 3 “Genovese”, “Padre Antero Micone” Hospital, Genoa, Italy
| | | | - Felice Scasso
- Department of Otorhinolaryngology, ASL 3 “Genovese”, “Padre Antero Micone” Hospital, Genoa, Italy
| | - Agostino Serra
- University of Catania, Italy
- G.B. Morgagni Foundation, Catania, Italy
| | - Guido Valente
- Dipartimento di Medicina Traslazionale, Università del Piemonte Orientale, Novara, Italy
| | - Micol Zannetti
- Dipartimento di Medicina Traslazionale, Università del Piemonte Orientale, Novara, Italy
| | - Angelo Zigliani
- Department of Radiology, University of Brescia, ASST Spedali Civili Brescia, Italy
| |
Collapse
|
129
|
Abstract
Cancer is a heterogeneous disease that requires a multimodal approach to diagnose, manage and treat. A better understanding of the disease biology can lead to identification of novel diagnostic/prognostic biomarkers and the discovery of the novel therapeutics with the goal of improving patient outcomes. Employing advanced technologies can facilitate this, enabling better diagnostic and treatment for cancer patients. In this regard, microfluidic technology has emerged as a promising tool in the studies of cancer, including single cancer cell analysis, modeling angiogenesis and metastasis, drug screening and liquid biopsy. Microfluidic technologies have opened new ways to study tumors in the preclinical and clinical settings. In this chapter, we highlight novel application of this technology in area of fundamental, translational and clinical cancer research.
Collapse
|
130
|
Abstract
The pulmonary blood-gas barrier represents a remarkable feat of engineering. It achieves the exquisite thinness needed for gas exchange by diffusion, the strength to withstand the stresses and strains of repetitive and changing ventilation, and the ability to actively maintain itself under varied demands. Understanding the design principles of this barrier is essential to understanding a variety of lung diseases, and to successfully regenerating or artificially recapitulating the barrier ex vivo. Many classical studies helped to elucidate the unique structure and morphology of the mammalian blood-gas barrier, and ongoing investigations have helped to refine these descriptions and to understand the biological aspects of blood-gas barrier function and regulation. This article reviews the key features of the blood-gas barrier that enable achievement of the necessary design criteria and describes the mechanical environment to which the barrier is exposed. It then focuses on the biological and mechanical components of the barrier that preserve integrity during homeostasis, but which may be compromised in certain pathophysiological states, leading to disease. Finally, this article summarizes recent key advances in efforts to engineer the blood-gas barrier ex vivo, using the platforms of lung-on-a-chip and tissue-engineered whole lungs. © 2020 American Physiological Society. Compr Physiol 10:415-452, 2020.
Collapse
Affiliation(s)
- Katherine L. Leiby
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
- Yale School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Micha Sam Brickman Raredon
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
- Yale School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Laura E. Niklason
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
- Yale School of Medicine, Yale University, New Haven, Connecticut, USA
- Department of Anesthesiology, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
131
|
Shafiee A. Design and Fabrication of Three-Dimensional Printed Scaffolds for Cancer Precision Medicine. Tissue Eng Part A 2020; 26:305-317. [PMID: 31992154 DOI: 10.1089/ten.tea.2019.0278] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Three-dimensional (3D)-engineered scaffolds have been widely investigated as drug delivery systems (DDS) or cancer models with the aim to develop effective cancer therapies. The in vitro and in vivo models developed via 3D printing (3DP) and tissue engineering concepts have significantly contributed to our understanding of cell-cell and cell-extracellular matrix interactions in the cancer microenvironment. Moreover, 3D tumor models were used to study the therapeutic efficiency of anticancer drugs. The present study aims to provide an overview of applying the 3DP and tissue engineering concepts for cancer studies with suggestions for future research directions. The 3DP technologies being used for the fabrication of personalized DDS have been highlighted and the potential technical approaches and challenges associated with the fused deposition modeling, the inkjet-powder bed, and stereolithography as the most promising 3DP techniques for drug delivery purposes are briefly described. Then, the advances, challenges, and future perspectives in tissue-engineered cancer models for precision medicine are discussed. Overall, future advances in this arena depend on the continuous integration of knowledge from cancer biology, biofabrication techniques, multiomics and patient data, and medical needs to develop effective treatments ultimately leading to improved clinical outcomes. Impact statement Three-dimensional printing (3DP) enables the fabrication of personalized medicines and drug delivery systems. The convergence of 3DP, tissue engineering concepts, and cancer biology could significantly improve our understanding of cancer biology and contribute to the development of new cancer therapies.
Collapse
Affiliation(s)
- Abbas Shafiee
- UQ Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, Australia.,Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Brisbane, Australia.,Royal Brisbane and Women's Hospital, Metro North Hospital and Health Service, Brisbane, Australia
| |
Collapse
|
132
|
Abstract
Recently, respiratory systems are increasingly threatened by high levels of environmental pollution. Organ-on-a-chip technology has the advantage of enabling more accurate preclinical experiments by reproducing in vivo organ physiology. To investigate disease mechanisms and treatment options, respiratory-physiology-on-a-chip systems have been studied for the last decade. Here, we delineate the strategic approaches to develop respiratory-physiology-on-a-chip that can recapitulate respiratory system in vitro. The state-of-the-art biofabrication methods and biomaterials are considered as key contributions to constructing the chips. We also explore the vascularization strategies to investigate complicated pathophysiological phenomena including inflammation and immune responses, which are the critical aggravating factors causing the complications in the respiratory diseases. In addition, challenges and future research directions are delineated to improve the mimicry of respiratory systems in terms of both structural and biological behaviors.
Collapse
|
133
|
Ashammakhi N, Darabi MA, Çelebi-Saltik B, Tutar R, Hartel MC, Lee J, Hussein S, Goudie MJ, Cornelius MB, Dokmeci MR, Khademhosseini A. Microphysiological Systems: Next Generation Systems for Assessing Toxicity and Therapeutic Effects of Nanomaterials. SMALL METHODS 2020; 4:1900589. [PMID: 33043130 PMCID: PMC7546538 DOI: 10.1002/smtd.201900589] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Indexed: 05/27/2023]
Abstract
Microphysiological systems, also known as organ-on-a-chip platforms, show promise for the development of new testing methods that can be more accurate than both conventional two-dimensional cultures and costly animal studies. The development of more intricate microphysiological systems can help to better mimic the human physiology and highlight the systemic effects of different drugs and materials. Nanomaterials are among a technologically important class of materials used for diagnostic, therapeutic, and monitoring purposes; all of which and can be tested using new organ-on-a-chip systems. In addition, the toxicity of nanomaterials which have entered the body from ambient air or diet can have deleterious effects on various body systems. This in turn can be studied in newly developed microphysiological systems. While organ-on-a-chip models can be useful, they cannot pick up secondary and systemic toxicity. Thus, the utilization of multi-organ-on-a-chip systems for advancing nanotechnology will largely be reflected in the future of drug development, toxicology studies and precision medicine. Various aspects of related studies, current challenges, and future perspectives are discussed in this paper.
Collapse
Affiliation(s)
- Nureddin Ashammakhi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, California, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, California, USA
- Department of Bioengineering, University of California, Los Angeles, California, USA
| | - Mohammad Ali Darabi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, California, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, California, USA
- Department of Bioengineering, University of California, Los Angeles, California, USA
| | - Betül Çelebi-Saltik
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, California, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, California, USA
- Department of Bioengineering, University of California, Los Angeles, California, USA
- Department of Stem Cell Sciences, Hacettepe University Graduate School of Health Sciences, 06100, Sihhiye, Ankara, Turkey
| | - Rumeysa Tutar
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, California, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, California, USA
- Department of Bioengineering, University of California, Los Angeles, California, USA
- Department of Chemistry, Faculty of Engineering, Istanbul University Cerrahpasa, Avcilar-Istanbul, Turkey
| | - Martin C. Hartel
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, California, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, California, USA
| | - Junmin Lee
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, California, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, California, USA
- Department of Bioengineering, University of California, Los Angeles, California, USA
| | - Saber Hussein
- Wright State University, Boonshoft School of Medicine, 3640 Colonel Glenn Hwy, Dayton, OH 45435, Ohio, USA
| | - Marcus J. Goudie
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, California, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, California, USA
- Department of Bioengineering, University of California, Los Angeles, California, USA
| | - Mercedes Brianna Cornelius
- California NanoSystems Institute (CNSI), University of California, Los Angeles, California, USA
- Department of Chemistry, University of California, Los Angeles, California, USA
| | - Mehmet R. Dokmeci
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, California, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, California, USA
| | - Ali Khademhosseini
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, California, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, California, USA
- Department of Bioengineering, University of California, Los Angeles, California, USA
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
134
|
Wang Y, Wu D, Wu G, Wu J, Lu S, Lo J, He Y, Zhao C, Zhao X, Zhang H, Wang S. Metastasis-on-a-chip mimicking the progression of kidney cancer in the liver for predicting treatment efficacy. Theranostics 2020; 10:300-311. [PMID: 31903121 PMCID: PMC6929630 DOI: 10.7150/thno.38736] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 08/29/2019] [Indexed: 12/21/2022] Open
Abstract
Metastasis is one of the most important factors that lead to poor prognosis in cancer patients, and effective suppression of the growth of primary cancer cells in a metastatic site is paramount in averting cancer progression. However, there is a lack of biomimetic three-dimensional (3D) in vitro models that can closely mimic the continuous growth of metastatic cancer cells in an organ-specific extracellular microenvironment (ECM) for assessing effective therapeutic strategies. Methods: In this metastatic tumor progression model, kidney cancer cells (Caki-1) and hepatocytes (i.e., HepLL cells) were co-cultured at an increasing ratio from 1:9 to 9:1 in a decellularized liver matrix (DLM)/gelatin methacryloyl (GelMA)-based biomimetic liver microtissue in a microfluidic device. Results:Via this model, we successfully demonstrated a linear anti-cancer relationship between the concentration of anti-cancer drug 5-Fluorouracil (5-FU) and the percentage of Caki-1 cells in the co-culture system (R2 = 0.89). Furthermore, the Poly(lactide-co-glycolide) (PLGA)-poly(ethylene glycol) (PEG)-based delivery system showed superior efficacy to free 5-FU in killing Caki-1 cells. Conclusions: In this study, we present a novel 3D metastasis-on-a-chip model mimicking the progression of kidney cancer cells metastasized to the liver for predicting treatment efficacy. Taken together, our study proved that the tumor progression model based on metastasis-on-a-chip with organ-specific ECM would provide a valuable tool for rapidly assessing treatment regimens and developing new chemotherapeutic agents.
Collapse
Affiliation(s)
- Yimin Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, China
- Institute for Translational Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310029, China
| | - Di Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, China
- Institute for Translational Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310029, China
| | - Guohua Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, China
- Institute for Translational Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310029, China
| | - Jianguo Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, China
- Institute for Translational Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310029, China
| | - Siming Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, China
- Institute for Translational Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310029, China
| | - James Lo
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, China
- Institute for Translational Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310029, China
- Department of Bioengineering, University of California Berkeley, Berkeley, CA, 94720, United States of America
| | - Yong He
- State Key Laboratory of Fluid Power and Mechatronic Systems, Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province College of Mechanical Engineering, Zhejiang University, Hangzhou, Zhejiang Province, 310029, China
| | - Chao Zhao
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0AH, United Kingdom
| | - Xin Zhao
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Hongbo Zhang
- Department of Pharmaceutical Science, Åbo Akademic University, FI-20520, Turku, Finland
| | - ShuQi Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, China
- Institute for Translational Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310029, China
| |
Collapse
|
135
|
Caballero D, Reis RL, Kundu SC. Engineering Patient-on-a-Chip Models for Personalized Cancer Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1230:43-64. [PMID: 32285364 DOI: 10.1007/978-3-030-36588-2_4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Traditional in vitro and in vivo models typically used in cancer research have demonstrated a low predictive power for human response. This leads to high attrition rates of new drugs in clinical trials, which threaten cancer patient prognosis. Tremendous efforts have been directed towards the development of a new generation of highly predictable pre-clinical models capable to reproduce in vitro the biological complexity of the human body. Recent advances in nanotechnology and tissue engineering have enabled the development of predictive organs-on-a-chip models of cancer with advanced capabilities. These models can reproduce in vitro the complex three-dimensional physiology and interactions that occur between organs and tissues in vivo, offering multiple advantages when compared to traditional models. Importantly, these models can be tailored to the biological complexity of individual cancer patients resulting into biomimetic and personalized cancer patient-on-a-chip platforms. The individualized models provide a more accurate and physiological environment to predict tumor progression on patients and their response to drugs. In this chapter, we describe the latest advances in the field of cancer patient-on-a-chip, and discuss about their main applications and current challenges. Overall, we anticipate that this new paradigm in cancer in vitro models may open up new avenues in the field of personalized - cancer - medicine, which may allow pharmaceutical companies to develop more efficient drugs, and clinicians to apply patient-specific therapies.
Collapse
Affiliation(s)
- David Caballero
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, Guimarães, Portugal. .,ICVS 3Bs PT Government Associate Lab, Braga, Guimarães, Portugal.
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, Guimarães, Portugal.,ICVS 3Bs PT Government Associate Lab, Braga, Guimarães, Portugal.,The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Guimarães, Portugal
| | - Subhas C Kundu
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, Guimarães, Portugal.,ICVS 3Bs PT Government Associate Lab, Braga, Guimarães, Portugal
| |
Collapse
|
136
|
Andrei L, Kasas S, Ochoa Garrido I, Stanković T, Suárez Korsnes M, Vaclavikova R, Assaraf YG, Pešić M. Advanced technological tools to study multidrug resistance in cancer. Drug Resist Updat 2020; 48:100658. [DOI: 10.1016/j.drup.2019.100658] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 02/06/2023]
|
137
|
Fan H, Demirci U, Chen P. Emerging organoid models: leaping forward in cancer research. J Hematol Oncol 2019; 12:142. [PMID: 31884964 PMCID: PMC6936115 DOI: 10.1186/s13045-019-0832-4] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 11/18/2019] [Indexed: 02/07/2023] Open
Abstract
Cancer heterogeneity is regarded as the main reason for the failure of conventional cancer therapy. The ability to reconstruct intra- and interpatient heterogeneity in cancer models is crucial for understanding cancer biology as well as for developing personalized anti-cancer therapy. Cancer organoids represent an emerging approach for creating patient-derived in vitro cancer models that closely recapitulate the pathophysiological features of natural tumorigenesis and metastasis. Meanwhile, cancer organoids have recently been utilized in the discovery of personalized anti-cancer therapy and prognostic biomarkers. Further, the synergistic combination of cancer organoids with organ-on-a-chip and 3D bioprinting presents a new avenue in the development of more sophisticated and optimized model systems to recapitulate complex cancer-stroma or multiorgan metastasis. Here, we summarize the recent advances in cancer organoids from a perspective of the in vitro emulation of natural cancer evolution and the applications in personalized cancer theranostics. We also discuss the challenges and trends in reconstructing more comprehensive cancer models for basic and clinical cancer research.
Collapse
Affiliation(s)
- Han Fan
- Department of Biomedical Engineering, Wuhan University School of Basic Medical Sciences, 115 Donghu Road, Wuhan, 430071, Hubei, China
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan, 430071, Hubei, China
| | - Utkan Demirci
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, 3155 Porter Drive, Palo Alto, CA, 94304, USA.
| | - Pu Chen
- Department of Biomedical Engineering, Wuhan University School of Basic Medical Sciences, 115 Donghu Road, Wuhan, 430071, Hubei, China.
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan, 430071, Hubei, China.
| |
Collapse
|
138
|
Ni BS, Tzao C, Huang JH. Plug-and-Play In Vitro Metastasis System toward Recapitulating the Metastatic Cascade. Sci Rep 2019; 9:18110. [PMID: 31792319 PMCID: PMC6889311 DOI: 10.1038/s41598-019-54711-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 11/11/2019] [Indexed: 02/07/2023] Open
Abstract
Microfluidic-based tumor models that mimic tumor culture environment have been developed to understand the cancer metastasis mechanism and discover effective antimetastatic drugs. These models successfully recapitulated key steps of metastatic cascades, yet still limited to few metastatic steps, operation difficulty, and small molecule absorption. In this study, we developed a metastasis system made of biocompatible and drug resistance plastics to recapitulate each metastasis stage in three-dimensional (3D) mono- and co-cultures formats, enabling the investigation of the metastatic responses of cancer cells (A549-GFP). The plug-and-play feature enhances the efficiency of the experimental setup and avoids initial culture failures. The results demonstrate that cancer cells tended to proliferate and migrate with circulating flow and intravasated across the porous membrane after a period of 3 d when they were treated with transforming growth factor-beta 1 (TGF-β1) or co-cultured with human pulmonary microvascular endothelial cells (HPMECs). The cells were also observed to detach and migrate into the circulating flow after a period of 20 d, indicating that they transformed into circulating tumor cells for the next metastasis stage. We envision this metastasis system can provide novel insights that would aid in fully understanding the entire mechanism of tumor invasion.
Collapse
Affiliation(s)
- Bing-Syuan Ni
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Ching Tzao
- Kuang Tien General Hospital, Taichung, 43303, Taiwan
| | - Jen-Huang Huang
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan.
| |
Collapse
|
139
|
Fetah KL, DiPardo BJ, Kongadzem EM, Tomlinson JS, Elzagheid A, Elmusrati M, Khademhosseini A, Ashammakhi N. Cancer Modeling-on-a-Chip with Future Artificial Intelligence Integration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1901985. [PMID: 31724305 PMCID: PMC6929691 DOI: 10.1002/smll.201901985] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 07/22/2019] [Indexed: 05/15/2023]
Abstract
Cancer is one of the leading causes of death worldwide, despite the large efforts to improve the understanding of cancer biology and development of treatments. The attempts to improve cancer treatment are limited by the complexity of the local milieu in which cancer cells exist. The tumor microenvironment (TME) consists of a diverse population of tumor cells and stromal cells with immune constituents, microvasculature, extracellular matrix components, and gradients of oxygen, nutrients, and growth factors. The TME is not recapitulated in traditional models used in cancer investigation, limiting the translation of preliminary findings to clinical practice. Advances in 3D cell culture, tissue engineering, and microfluidics have led to the development of "cancer-on-a-chip" platforms that expand the ability to model the TME in vitro and allow for high-throughput analysis. The advances in the development of cancer-on-a-chip platforms, implications for drug development, challenges to leveraging this technology for improved cancer treatment, and future integration with artificial intelligence for improved predictive drug screening models are discussed.
Collapse
Affiliation(s)
- Kirsten Lee Fetah
- Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA, 90095, USA
- California NanoSystems Institute (CNSI), University of California, 570 Westwood Plaza, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California, Los Angeles, CA, 90095, USA
| | - Benjamin J DiPardo
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Eve-Mary Kongadzem
- School of Technology and Innovations, University of Vaasa, FI-65101, Vaasa, Finland
| | - James S Tomlinson
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Adam Elzagheid
- Biotechnology Research Center, Libyan Authority for Research, Science and Technology, Tripoli, Libya
| | - Mohammed Elmusrati
- School of Technology and Innovations, University of Vaasa, FI-65101, Vaasa, Finland
| | - Ali Khademhosseini
- Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA, 90095, USA
- California NanoSystems Institute (CNSI), University of California, 570 Westwood Plaza, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California, Los Angeles, CA, 90095, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Nureddin Ashammakhi
- Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA, 90095, USA
- California NanoSystems Institute (CNSI), University of California, 570 Westwood Plaza, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California, Los Angeles, CA, 90095, USA
- School of Technology and Innovations, University of Vaasa, FI-65101, Vaasa, Finland
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
- Division of Plastic Surgery, Department of Surgery, Oulu University, FI-9001, Oulu, Finland
| |
Collapse
|
140
|
Trujillo-de Santiago G, Flores-Garza BG, Tavares-Negrete JA, Lara-Mayorga IM, González-Gamboa I, Zhang YS, Rojas-Martínez A, Ortiz-López R, Álvarez MM. The Tumor-on-Chip: Recent Advances in the Development of Microfluidic Systems to Recapitulate the Physiology of Solid Tumors. MATERIALS (BASEL, SWITZERLAND) 2019; 12:E2945. [PMID: 31514390 PMCID: PMC6766252 DOI: 10.3390/ma12182945] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/03/2019] [Accepted: 09/04/2019] [Indexed: 12/29/2022]
Abstract
The ideal in vitro recreation of the micro-tumor niche-although much needed for a better understanding of cancer etiology and development of better anticancer therapies-is highly challenging. Tumors are complex three-dimensional (3D) tissues that establish a dynamic cross-talk with the surrounding tissues through complex chemical signaling. An extensive body of experimental evidence has established that 3D culture systems more closely recapitulate the architecture and the physiology of human solid tumors when compared with traditional 2D systems. Moreover, conventional 3D culture systems fail to recreate the dynamics of the tumor niche. Tumor-on-chip systems, which are microfluidic devices that aim to recreate relevant features of the tumor physiology, have recently emerged as powerful tools in cancer research. In tumor-on-chip systems, the use of microfluidics adds another dimension of physiological mimicry by allowing a continuous feed of nutrients (and pharmaceutical compounds). Here, we discuss recently published literature related to the culture of solid tumor-like tissues in microfluidic systems (tumor-on-chip devices). Our aim is to provide the readers with an overview of the state of the art on this particular theme and to illustrate the toolbox available today for engineering tumor-like structures (and their environments) in microfluidic devices. The suitability of tumor-on-chip devices is increasing in many areas of cancer research, including the study of the physiology of solid tumors, the screening of novel anticancer pharmaceutical compounds before resourcing to animal models, and the development of personalized treatments. In the years to come, additive manufacturing (3D bioprinting and 3D printing), computational fluid dynamics, and medium- to high-throughput omics will become powerful enablers of a new wave of more sophisticated and effective tumor-on-chip devices.
Collapse
Affiliation(s)
- Grissel Trujillo-de Santiago
- Centro de Biotecnología-FEMSA, Tecnologico de Monterrey, Monterrey, Nuevo León CP 64849, Mexico.
- Departamento de Ingeniería Mecátrónica y Eléctrica, Tecnologico de Monterrey, Monterrey, Nuevo León CP 64849, Mexico.
| | | | | | - Itzel Montserrat Lara-Mayorga
- Centro de Biotecnología-FEMSA, Tecnologico de Monterrey, Monterrey, Nuevo León CP 64849, Mexico
- Departamento de Ingeniería Mecátrónica y Eléctrica, Tecnologico de Monterrey, Monterrey, Nuevo León CP 64849, Mexico
| | - Ivonne González-Gamboa
- Centro de Biotecnología-FEMSA, Tecnologico de Monterrey, Monterrey, Nuevo León CP 64849, Mexico
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Augusto Rojas-Martínez
- Centro de Investigación y Transferencia en Salud, Hospital San José, Tecnologico de Monterrey, Monterrey, Nuevo León CP 64849, Mexico
| | - Rocío Ortiz-López
- Centro de Investigación y Transferencia en Salud, Hospital San José, Tecnologico de Monterrey, Monterrey, Nuevo León CP 64849, Mexico
| | - Mario Moisés Álvarez
- Centro de Biotecnología-FEMSA, Tecnologico de Monterrey, Monterrey, Nuevo León CP 64849, Mexico.
| |
Collapse
|
141
|
Dang BV, Hassanzadeh-Barforoushi A, Syed MS, Yang D, Kim SJ, Taylor RA, Liu GJ, Liu G, Barber T. Microfluidic Actuation via 3D-Printed Molds toward Multiplex Biosensing of Cell Apoptosis. ACS Sens 2019; 4:2181-2189. [PMID: 31321976 DOI: 10.1021/acssensors.9b01057] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Multiplexed analysis of biochemical analytes such as proteins, enzymes, and immune products using a microfluidic device has the potential to cut assay time, reduce sample volume, realize high-throughput, and decrease experimental error without compromising sensitivity. Despite these huge benefits, the need for expensive specialized equipment and the complex photolithography fabrication process for the multiplexed devices have, to date, prevented widespread adoption of microfluidic systems. Here, we present a simple method to fabricate a new microfluidic-based multiplexed biosensing device by taking advantage of 3D-printing. The device is an integration of normally closed (NC) microfluidic valving units which offer superior operational flexibility by using PDMS membrane (E ∼ 1-2 MPa) and require minimized energy input (1-5 kPa). To systematically engineer the device, we first report on the geometrical and operational analysis of a single 3D-printed valving unit. Based on the characterization, we introduce a full prototype multiplexed chip comprising several microfluidic valves. The prototype offers-for the first time in a 3D-printed microfluidic device-the capability of on-demand performce of both a sequential and a parallel biochemical assay. As a proof of concept, our device has been used to simultaneously measure the apoptotic activity of 5 different members of the caspase protease enzyme family. In summary, the 3D-printed valving system showcased in this study overcomes traditional bottlenecks of microfabrication, enabling a new class of sophisticated liquid manipulation required in performing multiplexed sensing for biochemical assays.
Collapse
Affiliation(s)
- Bac Van Dang
- School of Mechanical and Manufacturing Engineering, Faculty of Engineering, The University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Amin Hassanzadeh-Barforoushi
- School of Mechanical and Manufacturing Engineering, Faculty of Engineering, The University of New South Wales, Sydney, New South Wales 2052, Australia
- Cancer Division, Garvan Institute of Medical Research/the Kinghorn Cancer Centre, Sydney, New South Wales 2010, Australia
| | - Maira Shakeel Syed
- School of Mechanical and Manufacturing Engineering, Faculty of Engineering, The University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Danting Yang
- Graduate School of Biomedical Engineering, ARC Centre of Excellence in Nanoscale BioPhotonics, Australian Centre for NanoMedicine, Faculty of Engineering, The University of New South Wales, Sydney, New South Wales 2052, Australia
- Department of Preventative Medicine, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Medical School of Ningbo University, Ningbo, Zhejiang 315211, China
| | - Sung-Jin Kim
- Department of Mechanical Engineering, Konkuk University, Seoul, 05029, Republic of Korea
| | - Robert A. Taylor
- School of Mechanical and Manufacturing Engineering, Faculty of Engineering, The University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Guo-Jun Liu
- Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, New South Wales 2234, Australia
- Discipline of Medical Imaging & Radiation Sciences, Faculty of Medicine and Health, Brain and Mind Centre, University of Sydney, 94 Mallett Street, Camperdown, New South Wales 2050, Australia
| | - Guozhen Liu
- Graduate School of Biomedical Engineering, ARC Centre of Excellence in Nanoscale BioPhotonics, Australian Centre for NanoMedicine, Faculty of Engineering, The University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Tracie Barber
- School of Mechanical and Manufacturing Engineering, Faculty of Engineering, The University of New South Wales, Sydney, New South Wales 2052, Australia
| |
Collapse
|
142
|
Ai Y, Zhang F, Wang C, Xie R, Liang Q. Recent progress in lab-on-a-chip for pharmaceutical analysis and pharmacological/toxicological test. Trends Analyt Chem 2019. [DOI: 10.1016/j.trac.2019.06.026] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
143
|
Fetah K, Tebon P, Goudie MJ, Eichenbaum J, Ren L, Barros N, Nasiri R, Ahadian S, Ashammakhi N, Dokmeci MR, Khademhosseini A. The emergence of 3D bioprinting in organ-on-chip systems. ACTA ACUST UNITED AC 2019. [DOI: 10.1088/2516-1091/ab23df] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
144
|
Liu W, Song J, Du X, Zhou Y, Li Y, Li R, Lyu L, He Y, Hao J, Ben J, Wang W, Shi H, Wang Q. AKR1B10 (Aldo-keto reductase family 1 B10) promotes brain metastasis of lung cancer cells in a multi-organ microfluidic chip model. Acta Biomater 2019; 91:195-208. [PMID: 31034948 DOI: 10.1016/j.actbio.2019.04.053] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 04/22/2019] [Accepted: 04/23/2019] [Indexed: 12/18/2022]
Abstract
Brain metastasis (BM) is a leading cause of mortality in patients with non-small cell lung cancer (NSCLC). However, the molecular mechanisms underlying BM of NSCLC remain largely unknown because of the lack of models to accurately investigate such a dynamic and complex process. Here we developed a multi-organ microfluidic chip as a new methodological platform to study BM. The chip consisted of two bionic organ units - an upstream "lung" and a downstream "brain" characterized by a functional "blood-brain barrier (BBB)" structure, allowing real-time visual monitoring of the entire BM process, from the growth of primary tumor to its breaking through the BBB, and finally reaching the brain parenchyma. The chip was verified by lung cancer cell lines with differing metastatic abilities and then applied for the BM research where we first demonstrated that the protein expression of Aldo-keto reductase family 1 B10 (AKR1B10) was significantly elevated in lung cancer BM. Silencing AKR1B10 in brain metastatic tumor cells suppressed their extravasation through the BBB in the in vitro Transwell model, in our ex vivo microfluidic chip, as well as the in vivo model of brain metastasis in nude mice. Moreover, AKR1B10 downregulated the expression of matrix metalloproteinase (MMP)-2 and MMP-9 via MEK/ERK signaling in metastatic lung cancers. These data suggest that our multi-organ microfluidic chip is a practical alternative to study BM pathogenesis, and AKR1B10 is a diagnostic biomarker and a prospective therapeutic target for NSCLC BM. STATEMENT OF SIGNIFICANCE: Brain metastasis (BM) of non-small cell lung cancer (NSCLC) is a complex cascade, and in particular, the process of lung cancer cells penetrating the blood-brain barrier (BBB) is very unique. However, due to the lack of reliable models that can faithfully mimic the dynamic process of BBB breaking, its molecular mechanisms have not well elucidated so far. In addition, although Aldo-keto reductase family 1 B10 (AKR1B10) has been implicated to the tumor development of liver cancer and many other cancers, little is known on its roles in the BM. Here, we established a multi-organ microfluidic bionic chip platform to recapitulate the entire BM process, and applied it to the BM pathology research, especially BBB extravasation. By using the chip and traditional models synergistically, we first demonstrated that AKR1B10 was significantly elevated in lung cancer BM, and defined the value of AKR1B10 as a diagnostic serum biomarker for lung cancer patients suffering from BM. Further, we investigated the role and mechanisms of AKR1B10 in BM that it promotes the extravasation of cancer cells through the BBB.
Collapse
|
145
|
Ruzycka M, Cimpan MR, Rios-Mondragon I, Grudzinski IP. Microfluidics for studying metastatic patterns of lung cancer. J Nanobiotechnology 2019; 17:71. [PMID: 31133019 PMCID: PMC6537392 DOI: 10.1186/s12951-019-0492-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 05/04/2019] [Indexed: 01/09/2023] Open
Abstract
The incidence of lung cancer continues to rise worldwide. Because the aggressive metastasis of lung cancer cells is the major drawback of successful therapies, the crucial challenge of modern nanomedicine is to develop diagnostic tools to map the molecular mechanisms of metastasis in lung cancer patients. In recent years, microfluidic platforms have been given much attention as tools for novel point-of-care diagnostic, an important aspect being the reconstruction of the body organs and tissues mimicking the in vivo conditions in one simple microdevice. Herein, we present the first comprehensive overview of the microfluidic systems used as innovative tools in the studies of lung cancer metastasis including single cancer cell analysis, endothelial transmigration, distant niches migration and finally neoangiogenesis. The application of the microfluidic systems to study the intercellular crosstalk between lung cancer cells and surrounding tumor microenvironment and the connection with multiple molecular signals coming from the external cellular matrix are discussed. We also focus on recent breakthrough technologies regarding lab-on-chip devices that serve as tools for detecting circulating lung cancer cells. The superiority of microfluidic systems over traditional in vitro cell-based assays with regard to modern nanosafety studies and new cancer drug design and discovery is also addressed. Finally, the current progress and future challenges regarding printable and paper-based microfluidic devices for personalized nanomedicine are summarized.
Collapse
Affiliation(s)
- Monika Ruzycka
- Department of Applied Toxicology, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Street, 02-097, Warsaw, Poland
| | - Mihaela R Cimpan
- Biomaterials - Department for Clinical Dentistry, University of Bergen, Årstadveien 19, 5009, Bergen, Norway
| | - Ivan Rios-Mondragon
- Biomaterials - Department for Clinical Dentistry, University of Bergen, Årstadveien 19, 5009, Bergen, Norway
| | - Ireneusz P Grudzinski
- Department of Applied Toxicology, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Street, 02-097, Warsaw, Poland.
| |
Collapse
|
146
|
Roy Choudhury A, Gupta S, Chaturvedi PK, Kumar N, Pandey D. Mechanobiology of Cancer Stem Cells and Their Niche. CANCER MICROENVIRONMENT : OFFICIAL JOURNAL OF THE INTERNATIONAL CANCER MICROENVIRONMENT SOCIETY 2019; 12:17-27. [PMID: 31004332 PMCID: PMC6529500 DOI: 10.1007/s12307-019-00222-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 04/10/2019] [Indexed: 01/25/2023]
Abstract
Though the existence of cancer stem cells remained enigmatic initially, over the time their participation in tumorigenesis and tumor progression has become highly evident. Today, they are also appreciated as the causal element for tumor heterogeneity and drug-resistance. Cancer stem cells activate a set of molecular pathways some of which are triggered by the unique mechanical properties of the tumor tissue stroma. A relatively new field called mechanobiology has emerged, which aims to critically evaluate the mechanical properties associated with biological events like tissue morphogenesis, cell-cell or cell-matrix interactions, cellular migration and also the development and progression of cancer. Development of more realistic model systems and biophysical instrumentation for observation and manipulation of cell-dynamics in real-time has invoked a hope for some novel therapeutic modalities against cancer in the future. This review discusses the fundamental concepts of cancer stem cells from an intriguing viewpoint of mechanobiology and some important breakthroughs to date.
Collapse
Affiliation(s)
- Ankit Roy Choudhury
- Department of Reproductive Biology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Surabhi Gupta
- Department of Reproductive Biology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Pradeep Kumar Chaturvedi
- Department of Reproductive Biology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Neeraj Kumar
- Department of Reproductive Biology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Deepak Pandey
- Department of Reproductive Biology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India.
| |
Collapse
|
147
|
Cedillo-Alcantar DF, Han YD, Choi J, Garcia-Cordero JL, Revzin A. Automated Droplet-Based Microfluidic Platform for Multiplexed Analysis of Biochemical Markers in Small Volumes. Anal Chem 2019; 91:5133-5141. [PMID: 30834743 DOI: 10.1021/acs.analchem.8b05689] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The ability to detect multiple analytes in a small sample volume has significance for numerous areas of research, including organs-on-chip, small animal experiments, and neonatology. The objective of this study was to develop an automated microfluidics platform for multiplexed detection of analytes in microliter sample volumes. This platform employed computer-controlled microvalves to create laminar co-flows of sample and assay reagent solutions. It also contained valve-regulated cross-junction for discretizing sample/reagent mixtures into water-in-oil droplets. Microfluidic automation allowed us to control parameters related to frequency of droplet generation and the number of droplets of the same composition, as well as the size of droplets. Each droplet represented an individual enzymatic assay carried out in a sub-nanoliter (0.8 nL) volume reactor. An enzymatic reaction involving target analyte and assay reagents produced colorimetric or fluorescent signals in droplets. Importantly, intensity of optical signal was proportional to the concentration of analyte in question. This microfluidic bioanalysis platform was used in conjunction with commercial "mix-detect" assays for glucose, total bile acids, and lactate dehydrogenase (LDH). After characterizing these assays individually, we demonstrated sensitive multiplexed detection of three analytes from as little as 3 μL. In fact, this volume was sufficient to generate multiple repeat droplets for each of the three biochemical assays as well as positive control droplets, confirming the quality of assay reagents and negative control droplets to help with background subtraction. One potential application for this microfluidic bioanalysis platform involves sampling cell-conditioned media in organ-on-chip devices. To highlight this application, hepatocyte spheroids were established in microfluidic devices, injured on-chip by exposure to lipotoxic agent (palmitate), and then connected to the bioanalysis module for daily monitoring of changes in cytotoxicity (LDH), energy metabolism (glucose), and liver function (total bile acids). Microfluidic in-droplet assays revealed increased levels of LDH as well as reduction in bile acid synthesis-results that were consistent with hepatic injury. Importantly, these experiments highlighted the fact that in-droplet assays were sufficiently sensitive to detect changes in functional output of a relatively small (∼100) number of hepatocyte spheroids cultured in a microfluidic device. Moving forward, we foresee increasing the multiplexing capability of this technology and applying this platform to other biological/medical scenarios where detection of multiple analytes from a small sample volume is desired.
Collapse
Affiliation(s)
- Diana F Cedillo-Alcantar
- Unidad Monterrey , Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional , Vía del Conocimiento 201 , Parque PIIT, Apodaca , Nuevo León CP 66628 , México.,Mayo Clinic , Rochester , Minnesota 55905 , United States
| | - Yong Duk Han
- Mayo Clinic , Rochester , Minnesota 55905 , United States
| | - Jonghoon Choi
- Mayo Clinic , Rochester , Minnesota 55905 , United States
| | - Jose L Garcia-Cordero
- Unidad Monterrey , Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional , Vía del Conocimiento 201 , Parque PIIT, Apodaca , Nuevo León CP 66628 , México
| | | |
Collapse
|
148
|
Dhiman N, Kingshott P, Sumer H, Sharma CS, Rath SN. On-chip anticancer drug screening - Recent progress in microfluidic platforms to address challenges in chemotherapy. Biosens Bioelectron 2019; 137:236-254. [PMID: 31121461 DOI: 10.1016/j.bios.2019.02.070] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 02/27/2019] [Accepted: 02/27/2019] [Indexed: 12/18/2022]
Abstract
There is an increasing need for advanced and inexpensive preclinical models to accelerate the development of anticancer drugs. While costly animal models fail to predict human clinical outcomes, in vitro models such as microfluidic chips ('tumor-on-chip') are showing tremendous promise at predicting and providing meaningful preclinical drug screening outcomes. Research on 'tumor-on-chips' has grown enormously worldwide and is being widely accepted by pharmaceutical companies as a drug development tool. In light of this shift in philosophy, it is important to review the recent literature on microfluidic devices to determine how rapidly the technology has progressed as a promising model for drug screening and aiding cancer therapy. We review the past five years of successful developments and capabilities in microdevice technology (cancer models) for use in anticancer drug screening. Microfluidic devices that are being designed to address current challenges in chemotherapy, such as drug resistance, combinatorial drug therapy, personalized medicine, and cancer metastasis are also reviewed in detail. We provide a perspective on how personalized 'tumor-on-chip', as well as high-throughput microfluidic platforms based on patient-specific tumor cells, can potentially replace the more expensive and 'non-human' animal models in preclinical anticancer drug development.
Collapse
Affiliation(s)
- Nandini Dhiman
- Regenerative Medicine and Stem Cells Laboratory, Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Telangana, India; Department of Chemistry and Biotechnology, Faculty of Science and Engineering Technology, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Peter Kingshott
- Department of Chemistry and Biotechnology, Faculty of Science and Engineering Technology, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Huseyin Sumer
- Department of Chemistry and Biotechnology, Faculty of Science and Engineering Technology, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Chandra S Sharma
- Creative & Advanced Research Based On Nanomaterials Laboratory, Department of Chemical Engineering, Indian Institute of Technology Hyderabad, Kandi, Telangana, India
| | - Subha Narayan Rath
- Regenerative Medicine and Stem Cells Laboratory, Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Telangana, India.
| |
Collapse
|
149
|
Lee SH, Jun BH. Advances in dynamic microphysiological organ-on-a-chip: Design principle and its biomedical application. J IND ENG CHEM 2019. [DOI: 10.1016/j.jiec.2018.11.041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
150
|
Zhao Y, Kankala RK, Wang SB, Chen AZ. Multi-Organs-on-Chips: Towards Long-Term Biomedical Investigations. Molecules 2019; 24:E675. [PMID: 30769788 PMCID: PMC6412790 DOI: 10.3390/molecules24040675] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/06/2019] [Accepted: 02/11/2019] [Indexed: 12/12/2022] Open
Abstract
With advantageous features such as minimizing the cost, time, and sample size requirements, organ-on-a-chip (OOC) systems have garnered enormous interest from researchers for their ability for real-time monitoring of physical parameters by mimicking the in vivo microenvironment and the precise responses of xenobiotics, i.e., drug efficacy and toxicity over conventional two-dimensional (2D) and three-dimensional (3D) cell cultures, as well as animal models. Recent advancements of OOC systems have evidenced the fabrication of 'multi-organ-on-chip' (MOC) models, which connect separated organ chambers together to resemble an ideal pharmacokinetic and pharmacodynamic (PK-PD) model for monitoring the complex interactions between multiple organs and the resultant dynamic responses of multiple organs to pharmaceutical compounds. Numerous varieties of MOC systems have been proposed, mainly focusing on the construction of these multi-organ models, while there are only few studies on how to realize continual, automated, and stable testing, which still remains a significant challenge in the development process of MOCs. Herein, this review emphasizes the recent advancements in realizing long-term testing of MOCs to promote their capability for real-time monitoring of multi-organ interactions and chronic cellular reactions more accurately and steadily over the available chip models. Efforts in this field are still ongoing for better performance in the assessment of preclinical attributes for a new chemical entity. Further, we give a brief overview on the various biomedical applications of long-term testing in MOCs, including several proposed applications and their potential utilization in the future. Finally, we summarize with perspectives.
Collapse
Affiliation(s)
- Yi Zhao
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, China.
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen 361021, China.
| | - Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, China.
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen 361021, China.
| | - Shi-Bin Wang
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, China.
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen 361021, China.
| | - Ai-Zheng Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, China.
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen 361021, China.
| |
Collapse
|