1
|
Zhou L, Huang J, Li C, Gu Q, Li G, Li ZA, Xu J, Zhou J, Tuan RS. Organoids and organs-on-chips: Recent advances, applications in drug development, and regulatory challenges. MED 2025; 6:100667. [PMID: 40220744 DOI: 10.1016/j.medj.2025.100667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 08/30/2024] [Accepted: 03/12/2025] [Indexed: 04/14/2025]
Abstract
Organoids and organs-on-chips (OoCs) are rapidly evolving technologies for creating miniature human tissue models. They can mimic complex physiological functions and pathological conditions, offering more realistic platforms for disease modeling, drug screening, precision medicine, and regenerative therapies. The passing of the FDA Modernization Act 2.0 has reduced animal testing requirements for drug trials, marking a significant milestone in using advanced in vitro models such as organoids and OoCs for therapeutic discovery. Apart from technical and ethical challenges, regulatory issues persist in ensuring the reliability, scientificity, and applicability of these models in drug development. This perspective explores the concept, advancements, pros and cons, and applications of organoids and OoCs, particularly in drug research and development. It also examines global regulatory agencies' policies and actions on using these models in drug evaluation, aiming to guide industry standard setting and advance regulatory science.
Collapse
Affiliation(s)
- Liangbin Zhou
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China; Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science and Technology Park, Hong Kong SAR, China; Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Innovative Orthopaedic Biomaterials and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jingjing Huang
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Cun Li
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Qi Gu
- Key Laboratory of Organ Regeneration and Reconstruction, Beijing Institute for Stem Cell and Regenerative Medicine, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Gang Li
- Department of Otolaryngology-Head and Neck Surgery, Nanfang Hospital of the Southern Medical University, Guangzhou, China
| | - Zhong Alan Li
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China; Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science and Technology Park, Hong Kong SAR, China; Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Innovative Orthopaedic Biomaterials and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Jie Zhou
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| | - Rocky S Tuan
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China; Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science and Technology Park, Hong Kong SAR, China; Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
2
|
Hermann NG, Ficek RA, Markov DA, McCawley LJ, Hutson MS. Toxicokinetics for organ-on-chip devices. LAB ON A CHIP 2025; 25:2017-2029. [PMID: 40062965 DOI: 10.1039/d4lc00840e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Organ-on-chip (OOC) devices are an emerging New Approach Method in both pharmacology and toxicology. Such devices use heterotypic combinations of human cells in a micro-fabricated device to mimic in vivo conditions and better predict organ-specific toxicological responses in humans. One drawback of these devices is that they are often made from polydimethylsiloxane (PDMS), a polymer known to interact with hydrophobic chemicals. Due to this interaction, the actual dose experienced by cells inside OOC devices can differ strongly from the nominal dose. To account for these effects, we have developed a comprehensive model to characterize chemical-PDMS interactions, including partitioning into and diffusion through PDMS. We use these methods to characterize PDMS interactions for 24 chemicals, ranging from fluorescent dyes to persistent organic pollutants to organophosphate pesticides. We further show that these methods return physical interaction parameters that can be used to accurately predict time-dependent doses under continuous-flow conditions, as would be present in an OOC device. These results demonstrate the validity of the methods and model across geometries and flow rates.
Collapse
Affiliation(s)
- Nathaniel G Hermann
- Department of Physics and Astronomy, Vanderbilt University, PMB 401807, Nashville, TN 37240, USA.
| | - Richard A Ficek
- Department of Physics and Astronomy, Vanderbilt University, PMB 401807, Nashville, TN 37240, USA.
| | - Dmitry A Markov
- Department of Biomedical Engineering, Vanderbilt University, USA
| | - Lisa J McCawley
- Department of Biomedical Engineering, Vanderbilt University, USA
| | - M Shane Hutson
- Department of Physics and Astronomy, Vanderbilt University, PMB 401807, Nashville, TN 37240, USA.
| |
Collapse
|
3
|
Zhang X, Wang Y, Han J, Zhao W, Zhang W, Li X, Chen J, Song W, Wang L. Cardiac-Focused Multi-Organ Chips: Advanced Disease Modeling, Drug Testing, and Inter-Organ Communication. Adv Biol (Weinh) 2025; 9:e2400512. [PMID: 39913111 DOI: 10.1002/adbi.202400512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 12/18/2024] [Indexed: 02/07/2025]
Abstract
Heart disease remains a leading cause of mortality worldwide, posing a significant challenge to global healthcare systems. Traditional animal models and cell culture techniques are instrumental in advancing the understanding of cardiac pathophysiology. However, these methods are limited in their ability to fully replicate the heart's intricate functions. This underscores the need for a deeper investigation into the fundamental mechanisms of heart disease. Notably, cardiac pathology is often influenced by systemic factors, with conditions in other organs contributing to disease onset and progression. Cardiac-focused multi-organ chip technology has emerged to better elucidate these complex inter-organ communications and address the limitations of current in vitro models. This technology offers a novel approach by recreating the cardiac microenvironment and integrating it with other organ systems, thereby enabling more precise disease modeling and drug toxicity assessment. This review provides a comprehensive overview of the heart's structure and function, explores the advancements in cardiac organ chip development, and highlights the applications of cardiac-focused multi-organ chips in medical research. Finally, the future potential of this technology in enhancing disease modeling and therapeutic evaluation is discussed.
Collapse
Affiliation(s)
- Xiaolong Zhang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250 353, China
- Shandong Institute of Mechanical Design and Research, Jinan, 250 353, China
| | - Yushen Wang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250 353, China
- Shandong Institute of Mechanical Design and Research, Jinan, 250 353, China
| | - Junlei Han
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250 353, China
- Shandong Institute of Mechanical Design and Research, Jinan, 250 353, China
| | - Weilong Zhao
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250 353, China
- Shandong Institute of Mechanical Design and Research, Jinan, 250 353, China
| | - Wenhong Zhang
- College of Mechanical Engineering, Donghua University, Shanghai, 201 620, China
| | - Xinyu Li
- Department of Minimally Invasive Comprehensive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250 021, China
| | - Jun Chen
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250 353, China
- Shandong Institute of Mechanical Design and Research, Jinan, 250 353, China
| | - Wei Song
- Department of Minimally Invasive Comprehensive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250 021, China
| | - Li Wang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250 353, China
- Shandong Institute of Mechanical Design and Research, Jinan, 250 353, China
| |
Collapse
|
4
|
Aggarwal S, Chakraborty A, Singh V, Lory S, Karalis K, Rahme LG. Revealing the impact of Pseudomonas aeruginosa quorum sensing molecule 2'-aminoacetophenone on human bronchial-airway epithelium and pulmonary endothelium using a human airway-on-a-chip. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.21.644589. [PMID: 40196568 PMCID: PMC11974707 DOI: 10.1101/2025.03.21.644589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Pseudomonas aeruginosa (PA) causes severe respiratory infections utilizing multiple virulence functions. Our previous findings on PA quorum sensing (QS)-regulated small molecule, 2'-aminoacetophenone (2-AA), secreted by the bacteria in infected tissues, revealed its effect on immune and metabolic functions favouring a long-term presence of PA in the host. However, studies on 2-AA's specific effects on bronchial-airway epithelium and pulmonary endothelium remain elusive. To evaluate 2AA's spatiotemporal changes in the human airway, considering endothelial cells as the first point of contact when the route of lung infection is hematogenic, we utilized the microfluidic airway-on-chip lined by polarized human bronchial-airway epithelium and pulmonary endothelium. Using this platform, we performed RNA-sequencing to analyse responses of 2-AA-treated primary human pulmonary microvascular endothelium (HPMEC) and adjacent primary normal human bronchial epithelial (NHBE) cells from healthy female donors and potential cross-talk between these cells. Analyses unveiled specific signaling and biosynthesis pathways to be differentially regulated by 2-AA in epithelial cells, including HIF-1 and pyrimidine signaling, glycosaminoglycan, and glycosphingolipid biosynthesis, while in endothelial cells were fatty acid metabolism, phosphatidylinositol and estrogen receptor signaling, and proinflammatory signaling pathways. Significant overlap in both cell types in response to 2-AA was found in genes implicated in immune response and cellular functions. In contrast, we found that genes related to barrier permeability, cholesterol metabolism, and oxidative phosphorylation were differentially regulated upon exposure to 2-AA in the cell types studied. Murine in-vivo and additional in vitro cell culture studies confirmed cholesterol accumulation in epithelial cells. Results also revealed specific biomarkers associated with cystic fibrosis and idiopathic pulmonary fibrosis to be modulated by 2-AA in both cell types, with the cystic fibrosis transmembrane regulator expression to be affected only in endothelial cells. The 2-AA-mediated effects on healthy epithelial and endothelial primary cells within a microphysiological dynamic environment mimicking the human lung airway enhance our understanding of this QS signaling molecule. This study provides novel insights into their functions and potential interactions, paving the way for innovative, cell-specific therapeutic strategies to combat PA lung infections.
Collapse
|
5
|
Soliman Y, Al-Khodor J, Yildirim Köken G, Mustafaoglu N. A guide for blood-brain barrier models. FEBS Lett 2025; 599:599-644. [PMID: 39533665 DOI: 10.1002/1873-3468.15053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 10/18/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024]
Abstract
Understanding the intricate mechanisms underlying brain-related diseases hinges on unraveling the pivotal role of the blood-brain barrier (BBB), an essential dynamic interface crucial for maintaining brain equilibrium. This review offers a comprehensive analysis of BBB physiology, delving into its cellular and molecular components while exploring a wide range of in vivo and in vitro BBB models. Notably, recent advancements in 3D cell culture techniques are explicitly discussed, as they have significantly improved the fidelity of BBB modeling by enabling the replication of physiologically relevant environments under flow conditions. Special attention is given to the cellular aspects of in vitro BBB models, alongside discussions on advances in stem cell technologies, providing valuable insights into generating robust cellular systems for BBB modeling. The diverse array of cell types used in BBB modeling, depending on their sources, is meticulously examined in this comprehensive review, scrutinizing their respective derivation protocols and implications. By synthesizing diverse approaches, this review sheds light on the improvements of BBB models to capture physiological conditions, aiding in understanding BBB interactions in health and disease conditions to foster clinical developments.
Collapse
Affiliation(s)
- Yomna Soliman
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul, Turkey
- Faculty of Pharmacy, Mansoura University, Egypt
| | - Jana Al-Khodor
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul, Turkey
| | | | - Nur Mustafaoglu
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul, Turkey
- Sabancı University Nanotechnology Research and Application Center, Istanbul, Turkey
| |
Collapse
|
6
|
Bokhout L, Campeiro JD, Dalm SU. Exploring the landscape of current in vitro and in vivo models and their relevance for targeted radionuclide theranostics. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07123-3. [PMID: 40016527 DOI: 10.1007/s00259-025-07123-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 01/28/2025] [Indexed: 03/01/2025]
Abstract
Cancer remains a leading cause of mortality globally, driving ongoing research into innovative treatment strategies. Preclinical research forms the base for developing these novel treatments, using both in vitro and in vivo model systems that are, ideally, as clinically representative as possible. Emerging as a promising approach for cancer management, targeted radionuclide theranostics (TRT) uses radiotracers to deliver (cytotoxic) radionuclides specifically to cancer cells. Since the field is relatively new, more advanced preclinical models are not yet regularly applied in TRT research. This narrative review examines the currently applied in vitro, ex vivo and in vivo models for oncological research, discusses if and how these models are now applied for TRT studies, and whether not yet applied models can be of benefit for the field. A selection of different models is discussed, ranging from in vitro two-dimensional (2D) and three-dimensional (3D) cell models, including spheroids, organoids and tissue slice cultures, to in vivo mouse cancer models, such as cellline-derived models, patient-derived xenograft models and humanized models. Each of the models has advantages and limitations for studying human cancer biology, radiopharmaceutical assessment and treatment efficacy. Overall, there is a need to apply more advanced models in TRT research that better address specific TRT phenomena, such as crossfire and abscopal effects, to enhance the clinical relevance and effectiveness of preclinical TRT evaluations.
Collapse
Affiliation(s)
- Lisa Bokhout
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Joana D Campeiro
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Simone U Dalm
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
7
|
Petit I, Faucher Q, Bernard JS, Giunchi P, Humeau A, Sauvage FL, Marquet P, Védrenne N, Di Meo F. Proximal tubule-on-chip as a model for predicting cation transport and drug transporter dynamics. Sci Rep 2025; 15:2580. [PMID: 39833212 PMCID: PMC11747318 DOI: 10.1038/s41598-025-85653-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 01/06/2025] [Indexed: 01/22/2025] Open
Abstract
Deciphering the sources of variability in drug responses requires to understand the processes modulating drug pharmacokinetics. However, pharmacological research suffers from poor reproducibility across clinical, animal, and experimental models. Predictivity can be improved by using Organs-on-Chips, which are more physiological, human-oriented, micro-engineered devices that include microfluidics. OoC are particularly relevant at the fundamental and preclinical stages of drug development by providing more accurate assessment of key pharmacokinetic events. We have developed a proximal tubule-on-a-chip model combining commercial microfluidic and chip technologies. Using the RPTEC/TERT1 cell line, we set up a dual-flow system with antiparallel flows to mimic the dynamics of blood and urine. We assessed transporters mRNA expression, cellular polarization and protein expression via immunofluorescence, and monitored the transcellular transport of prototypic xenobiotics by determining their efflux ratios. Our results show that flow exposure significantly modulate mRNA expression of drug membrane transporters. Dynamic conditions also enhance cell polarization, as evidenced by preferential basal and apical expressions of Na + /K + -ATPase, P-gp, OCT2, and MATE1 , as well as the cellular secretory profile. We demonstrated unidirectional transcellular transport of metformin with a higher efflux than influx ratio, inhibited with OCT2 inhibitor, thus confirming the relevance of our proximal tubule-on-a-chip set up for cation transport investigations. Our proximal tubule-on-a-chip can also be used to explore the interactions between transporters, xenobiotics, and endogenous metabolites, possibly involved in the variability of individual drug responses. This study provides additional evidence that OoC can help bridge the gaps between systemic and local pharmacokinetics.
Collapse
Affiliation(s)
- Isy Petit
- U1248 Pharmacology & Transplantation, Inserm, Univ. Limoges, Limoges, France
| | - Quentin Faucher
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | | | - Perrine Giunchi
- U1248 Pharmacology & Transplantation, Inserm, Univ. Limoges, Limoges, France
- Institut de Recherche en Santé Digestive, INSERM, INRAE, ENVT, Univ. Toulouse III, Toulouse, France
- Institut de Mécanique Des Fluides de Toulouse (IMFT), CNRS, Univ. Toulouse, Toulouse, France
| | - Antoine Humeau
- U1248 Pharmacology & Transplantation, Inserm, Univ. Limoges, Limoges, France
| | | | - Pierre Marquet
- U1248 Pharmacology & Transplantation, Inserm, Univ. Limoges, Limoges, France
- Department of Pharmacology, Toxicology and Pharmacovigilance, CHU Limoges, Limoges, France
| | - Nicolas Védrenne
- U1248 Pharmacology & Transplantation, Inserm, Univ. Limoges, Limoges, France.
| | - Florent Di Meo
- U1248 Pharmacology & Transplantation, Inserm, Univ. Limoges, Limoges, France.
- UAR2015/US42 Integrative Biology Health Chemistry and Environment BISCEm, CNRS, Inserm, CHU Limoges, Univ. Limoges, Limoges, France.
| |
Collapse
|
8
|
Özkan A, LoGrande NT, Feitor JF, Goyal G, Ingber DE. Intestinal organ chips for disease modelling and personalized medicine. Nat Rev Gastroenterol Hepatol 2024; 21:751-773. [PMID: 39192055 DOI: 10.1038/s41575-024-00968-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/10/2024] [Indexed: 08/29/2024]
Abstract
Alterations in intestinal structure, mechanics and physiology underlie acute and chronic intestinal conditions, many of which are influenced by dysregulation of microbiome, peristalsis, stroma or immune responses. Studying human intestinal physiology or pathophysiology is difficult in preclinical animal models because their microbiomes and immune systems differ from those of humans. Although advances in organoid culture partially overcome this challenge, intestinal organoids still lack crucial features that are necessary to study functions central to intestinal health and disease, such as digestion or fluid flow, as well as contributions from long-term effects of living microbiome, peristalsis and immune cells. Here, we review developments in organ-on-a-chip (organ chip) microfluidic culture models of the human intestine that are lined by epithelial cells and interfaced with other tissues (such as stroma or endothelium), which can experience both fluid flow and peristalsis-like motions. Organ chips offer powerful ways to model intestinal physiology and disease states for various human populations and individual patients, and can be used to gain new insight into underlying molecular and biophysical mechanisms of disease. They can also be used as preclinical tools to discover new drugs and then validate their therapeutic efficacy and safety in the same human-relevant model.
Collapse
Affiliation(s)
- Alican Özkan
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Nina Teresa LoGrande
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Jessica F Feitor
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Girija Goyal
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA.
- Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, MA, USA.
| |
Collapse
|
9
|
Modi PS, Singh A, Chaturvedi A, Agarwal S, Dutta R, Nayak R, Singh AK. Tissue chips as headway model and incitement technology. Synth Syst Biotechnol 2024; 10:86-101. [PMID: 39286054 PMCID: PMC11403008 DOI: 10.1016/j.synbio.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 08/12/2024] [Accepted: 08/26/2024] [Indexed: 09/19/2024] Open
Abstract
Tissue on a chip or organ-on-chip (OOC) is a technology that's dignified to form a transformation in drug discovery through the use of advanced platforms. These are 3D in-vitro cell culture models that mimic micro-environment of human organs or tissues on artificial microstructures built on a portable microfluidic chip without involving sacrificial humans or animals. This review article aims to offer readers a thorough and insightful understanding of technology. It begins with an in-depth understanding of chip design and instrumentation, underlining its pivotal role and the imperative need for its development in the modern scientific landscape. The review article explores into the myriad applications of OOC technology, showcasing its transformative impact on fields such as radiobiology, drug discovery and screening, and its pioneering use in space research. In addition to highlighting these diverse applications, the article provides a critical analysis of the current challenges that OOC technology faces. It examines both the biological and technical limitations that hinder its progress and efficacy and discusses the potential advancements and innovations that could drive the OOC technology forward. Through this comprehensive review, readers will gain a deep appreciation of the significance, capabilities, and evolving landscape of OOC technology.
Collapse
Affiliation(s)
- Prerna Suchitan Modi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Abhishek Singh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Awyang Chaturvedi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Shailly Agarwal
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Raghav Dutta
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Ranu Nayak
- Amity Institute of Nanotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Alok Kumar Singh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| |
Collapse
|
10
|
Mehta V, Karnam G, Madgula V. Liver-on-chips for drug discovery and development. Mater Today Bio 2024; 27:101143. [PMID: 39070097 PMCID: PMC11279310 DOI: 10.1016/j.mtbio.2024.101143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/07/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024] Open
Abstract
Recent FDA modernization act 2.0 has led to increasing industrial R&D investment in advanced in vitro 3D models such as organoids, spheroids, organ-on-chips, 3D bioprinting, and in silico approaches. Liver-related advanced in vitro models remain the prime area of interest, as liver plays a central role in drug clearance of compounds. Growing evidence indicates the importance of recapitulating the overall liver microenvironment to enhance hepatocyte maturity and culture longevity using liver-on-chips (LoC) in vitro. Hence, pharmaceutical industries have started exploring LoC assays in the two of the most challenging areas: accurate in vitro-in vivo extrapolation (IVIVE) of hepatic drug clearance and drug-induced liver injury. We examine the joint efforts of commercial chip manufacturers and pharmaceutical companies to present an up-to-date overview of the adoption of LoC technology in the drug discovery. Further, several roadblocks are identified to the rapid adoption of LoC assays in the current drug development framework. Finally, we discuss some of the underexplored application areas of LoC models, where conventional 2D hepatic models are deemed unsuitable. These include clearance prediction of metabolically stable compounds, immune-mediated drug-induced liver injury (DILI) predictions, bioavailability prediction with gut-liver systems, hepatic clearance prediction of drugs given during pregnancy, and dose adjustment studies in disease conditions. We conclude the review by discussing the importance of PBPK modeling with LoC, digital twins, and AI/ML integration with LoC.
Collapse
Affiliation(s)
- Viraj Mehta
- Organoid Technology Lab, DMPK Department, Sai Life Sciences, Hyderabad, 500078, India
| | - Guruswamy Karnam
- Organoid Technology Lab, DMPK Department, Sai Life Sciences, Hyderabad, 500078, India
| | - Vamsi Madgula
- Organoid Technology Lab, DMPK Department, Sai Life Sciences, Hyderabad, 500078, India
| |
Collapse
|
11
|
Wang M, Sasaki Y, Sakagami R, Minamikawa T, Tsuda M, Ueno R, Deguchi S, Negoro R, So K, Higuchi Y, Yokokawa R, Takayama K, Yamashita F. Perfluoropolyether-Based Gut-Liver-on-a-Chip for the Evaluation of First-Pass Metabolism and Oral Bioavailability of Drugs. ACS Biomater Sci Eng 2024; 10:4635-4644. [PMID: 38822812 DOI: 10.1021/acsbiomaterials.4c00605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2024]
Abstract
In the evolving field of drug discovery and development, multiorgans-on-a-chip and microphysiological systems are gaining popularity owing to their ability to emulate in vivo biological environments. Among the various gut-liver-on-a-chip systems for studying oral drug absorption, the chip developed in this study stands out with two distinct features: incorporation of perfluoropolyether (PFPE) to effectively mitigate drug sorption and a unique enterohepatic single-passage system, which simplifies the analysis of first-pass metabolism and oral bioavailability. By introducing a bolus drug injection into the liver compartment, hepatic extraction alone could be evaluated, further enhancing our estimation of intestinal availability. In a study on midazolam (MDZ), PFPE-based chips showed more than 20-times the appearance of intact MDZ in the liver compartment effluent compared to PDMS-based counterparts. Notably, saturation of hepatic metabolism at higher concentrations was confirmed by observations when the dose was reduced from 200 μM to 10 μM. This result was further emphasized when the metabolism was significantly inhibited by the coadministration of ketoconazole. Our chip, which is designed to minimize the dead volume between the gut and liver compartments, is adept at sensitively observing the saturation of metabolism and the effect of inhibitors. Using genome-edited CYP3A4/UGT1A1-expressing Caco-2 cells, the estimates for intestinal and hepatic availabilities were 0.96 and 0.82, respectively; these values are higher than the known human in vivo values. Although the metabolic activity in each compartment can be further improved, this gut-liver-on-a-chip can not only be used to evaluate oral bioavailability but also to carry out individual assessment of both intestinal and hepatic availability.
Collapse
Affiliation(s)
- Mengyang Wang
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Yuko Sasaki
- Department of Applied Pharmaceutics and Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Rena Sakagami
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Tomotaka Minamikawa
- Department of Applied Pharmaceutics and Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Masahiro Tsuda
- Department of Applied Pharmaceutics and Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Ryohei Ueno
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, Kyoto 615-8540, Japan
| | - Sayaka Deguchi
- Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Ryosuke Negoro
- Laboratory of Molecular Pharmacokinetics, College of Pharmaceutical Sciences, Ritsumeikan University, Shiga 525-8577, Japan
| | - Kanako So
- Department of Applied Pharmaceutics and Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Yuriko Higuchi
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Ryuji Yokokawa
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, Kyoto 615-8540, Japan
| | - Kazuo Takayama
- Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Fumiyoshi Yamashita
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
- Department of Applied Pharmaceutics and Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
12
|
Shin YC, Than N, Park SJ, Kim HJ. Bioengineered human gut-on-a-chip for advancing non-clinical pharmaco-toxicology. Expert Opin Drug Metab Toxicol 2024; 20:593-606. [PMID: 38849312 DOI: 10.1080/17425255.2024.2365254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
INTRODUCTION There is a growing need for alternative models to advance current non-clinical experimental models because they often fail to accurately predict drug responses in human clinical trials. Human organ-on-a-chip models have emerged as promising approaches for advancing the predictability of drug behaviors and responses. AREAS COVERED We summarize up-to-date human gut-on-a-chip models designed to demonstrate intricate interactions involving the host, microbiome, and pharmaceutical compounds since these models have been reported a decade ago. This overview covers recent advances in gut-on-a-chip models as a bridge technology between non-clinical and clinical assessments of drug toxicity and metabolism. We highlight the promising potential of gut-on-a-chip platforms, offering a reliable and valid framework for investigating reciprocal crosstalk between the host, gut microbiome, and drug compounds. EXPERT OPINION Gut-on-a-chip platforms can attract multiple end users as predictive, human-relevant, and non-clinical model. Notably, gut-on-a-chip platforms provide a unique opportunity to recreate a human intestinal microenvironment, including dynamic bowel movement, luminal flow, oxygen gradient, host-microbiome interactions, and disease-specific manipulations restricted in animal and in vitro cell culture models. Additionally, given the profound impact of the gut microbiome on pharmacological bioprocess, it is critical to leverage breakthroughs of gut-on-a-chip technology to address knowledge gaps and drive innovations in predictive drug toxicology and metabolism.
Collapse
Affiliation(s)
- Yong Cheol Shin
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Nam Than
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Soo Jin Park
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Hyun Jung Kim
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Inflammation and Immunity, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
13
|
Abady MM, Jeong JS, Kwon HJ, Assiri AM, Cho J, Saadeldin IM. The reprotoxic adverse side effects of neurogenic and neuroprotective drugs: current use of human organoid modeling as a potential alternative to preclinical models. Front Pharmacol 2024; 15:1412188. [PMID: 38948466 PMCID: PMC11211546 DOI: 10.3389/fphar.2024.1412188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 05/29/2024] [Indexed: 07/02/2024] Open
Abstract
The management of neurological disorders heavily relies on neurotherapeutic drugs, but notable concerns exist regarding their possible negative effects on reproductive health. Traditional preclinical models often fail to accurately predict reprotoxicity, highlighting the need for more physiologically relevant systems. Organoid models represent a promising approach for concurrently studying neurotoxicity and reprotoxicity, providing insights into the complex interplay between neurotherapeutic drugs and reproductive systems. Herein, we have examined the molecular mechanisms underlying neurotherapeutic drug-induced reprotoxicity and discussed experimental findings from case studies. Additionally, we explore the utility of organoid models in elucidating the reproductive complications of neurodrug exposure. Have discussed the principles of organoid models, highlighting their ability to recapitulate neurodevelopmental processes and simulate drug-induced toxicity in a controlled environment. Challenges and future perspectives in the field have been addressed with a focus on advancing organoid technologies to improve reprotoxicity assessment and enhance drug safety screening. This review underscores the importance of organoid models in unraveling the complex relationship between neurotherapeutic drugs and reproductive health.
Collapse
Affiliation(s)
- Mariam M. Abady
- Organic Metrology Group, Division of Chemical and Material Metrology, Korea Research Institute of Standards and Science, Daejeon, Republic of Korea
- Department of Bio-Analytical Science, University of Science and Technology, Daejeon, Republic of Korea
- Department of Nutrition and Food Science, National Research Centre, Cairo, Egypt
| | - Ji-Seon Jeong
- Organic Metrology Group, Division of Chemical and Material Metrology, Korea Research Institute of Standards and Science, Daejeon, Republic of Korea
- Department of Bio-Analytical Science, University of Science and Technology, Daejeon, Republic of Korea
| | - Ha-Jeong Kwon
- Organic Metrology Group, Division of Chemical and Material Metrology, Korea Research Institute of Standards and Science, Daejeon, Republic of Korea
| | - Abdullah M. Assiri
- Deperament of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Jongki Cho
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Republic of Korea
| | - Islam M. Saadeldin
- Deperament of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| |
Collapse
|
14
|
Bosmans C, Ginés Rodriguez N, Karperien M, Malda J, Moreira Teixeira L, Levato R, Leijten J. Towards single-cell bioprinting: micropatterning tools for organ-on-chip development. Trends Biotechnol 2024; 42:739-759. [PMID: 38310021 DOI: 10.1016/j.tibtech.2023.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 02/05/2024]
Abstract
Organs-on-chips (OoCs) hold promise to engineer progressively more human-relevant in vitro models for pharmaceutical purposes. Recent developments have delivered increasingly sophisticated designs, yet OoCs still lack in reproducing the inner tissue physiology required to fully resemble the native human body. This review emphasizes the need to include microarchitectural and microstructural features, and discusses promising avenues to incorporate well-defined microarchitectures down to the single-cell level. We highlight how their integration will significantly contribute to the advancement of the field towards highly organized structural and hierarchical tissues-on-chip. We discuss the combination of state-of-the-art micropatterning technologies to achieve OoCs resembling human-intrinsic complexity. It is anticipated that these innovations will yield significant advances in realization of the next generation of OoC models.
Collapse
Affiliation(s)
- Cécile Bosmans
- Department of Developmental BioEngineering, University of Twente, Enschede, The Netherlands
| | - Núria Ginés Rodriguez
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marcel Karperien
- Department of Developmental BioEngineering, University of Twente, Enschede, The Netherlands
| | - Jos Malda
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Liliana Moreira Teixeira
- Department of Advanced Organ bioengineering and Therapeutics, University of Twente, Enschede, The Netherlands.
| | - Riccardo Levato
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| | - Jeroen Leijten
- Department of Developmental BioEngineering, University of Twente, Enschede, The Netherlands.
| |
Collapse
|
15
|
Liang CC, Chen PY, Liu NC, Lee IC. Comparison between dynamic versus static models and real-time monitoring of neuronal dysfunction in an amyloid-β induced neuronal toxic model on a chip platform. LAB ON A CHIP 2024; 24:1887-1902. [PMID: 38414410 DOI: 10.1039/d3lc00507k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
Microfluidics-based organs-on-a-chip offer a promising method for dynamic and 3-dimensional (3D) cell culture to evaluate the cell behaviors within the biomimetic environment. The purpose of this study was to establish neural network connections in a 3D neural stem cell (NSC)-based system with an interstitial level of flow for simulating the brain microenvironment toward a dynamic amyloid-β (Aβ) induced neuronal toxic model on a chip and to compare the biological effects and neurite dysfunction between static and dynamic systems. The brain-on-a-chip system consisted of an impedance analyzing layer, a structured well with a connected channel, and an interface coating with polypeptide films fabricated with modification based on our previous study. The cytotoxicity and percentage of neuron/astrocyte differentiation were all compared in both static and dynamic brain-on-a-chip systems. Reactive oxygen species production, neuron marker expression and neurotransmitter-acetylcholine release were all compared to evaluate functional neurite losses in both static and dynamic systems with/without Aβ addition. Moreover, real-time impedance recording was used to consecutively monitor the neurite connection/disconnection in both static and dynamic brain-on-a-chip systems. The NSC-based dynamic brain-on-a-chip may enable the application of different neurodegenerative disease in vitro models for pathogenesis studies, drug discovery and novel therapeutic method development.
Collapse
Affiliation(s)
- Chu-Chun Liang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, No. 101, Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan.
| | - Po-Yen Chen
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, Maryland, 20742, USA
| | - Nien-Che Liu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, No. 101, Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan.
| | - I-Chi Lee
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, No. 101, Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan.
| |
Collapse
|
16
|
Rao WF, Wang YW, Li AQ, Zhou SS, Zheng ZM. An electromechanical stimulation regulating model with flexoelectric effect of piezoelectric laminated micro-beam for cell bionic culture. Sci Rep 2024; 14:6130. [PMID: 38480822 PMCID: PMC11636837 DOI: 10.1038/s41598-024-56708-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 03/09/2024] [Indexed: 03/19/2024] Open
Abstract
Cell bionic culture requires the construction of cell growth microenvironments. In this paper, mechanical force and electrical stimulations are applied to the cells cultured on the surface of the piezoelectric laminated micro-beam driven by an excitation voltage. Based on the extended dielectric theory, the electromechanical microenvironment regulating model of the current piezoelectric laminated micro-beam is established. The variational principle is used to obtain the governing equations and boundary conditions. The differential quadrature method and the iterative method are used to solve two boundary value problems for cantilever beams and simply supported beams. In two cases, the mechanical force and electrical stimulations applied to the cells are analyzed in detail and the microscale effect is investigated. This study is meaningful for improving the quality of cell culture and promoting the cross-integration of mechanics and biomedicine.
Collapse
Affiliation(s)
- Wei-Feng Rao
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, People's Republic of China
- Shandong Institute of Mechanical Design and Research, Jinan, 250031, People's Republic of China
| | - Ya-Wen Wang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, People's Republic of China
- Shandong Institute of Mechanical Design and Research, Jinan, 250031, People's Republic of China
| | - An-Qing Li
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, People's Republic of China.
- Shandong Institute of Mechanical Design and Research, Jinan, 250031, People's Republic of China.
| | - Sha-Sha Zhou
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, People's Republic of China
- Shandong Institute of Mechanical Design and Research, Jinan, 250031, People's Republic of China
| | - Zu-Mei Zheng
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, People's Republic of China
- Shandong Institute of Mechanical Design and Research, Jinan, 250031, People's Republic of China
| |
Collapse
|
17
|
Aazmi A, Zhang D, Mazzaglia C, Yu M, Wang Z, Yang H, Huang YYS, Ma L. Biofabrication methods for reconstructing extracellular matrix mimetics. Bioact Mater 2024; 31:475-496. [PMID: 37719085 PMCID: PMC10500422 DOI: 10.1016/j.bioactmat.2023.08.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/19/2023] Open
Abstract
In the human body, almost all cells interact with extracellular matrices (ECMs), which have tissue and organ-specific compositions and architectures. These ECMs not only function as cellular scaffolds, providing structural support, but also play a crucial role in dynamically regulating various cellular functions. This comprehensive review delves into the examination of biofabrication strategies used to develop bioactive materials that accurately mimic one or more biophysical and biochemical properties of ECMs. We discuss the potential integration of these ECM-mimics into a range of physiological and pathological in vitro models, enhancing our understanding of cellular behavior and tissue organization. Lastly, we propose future research directions for ECM-mimics in the context of tissue engineering and organ-on-a-chip applications, offering potential advancements in therapeutic approaches and improved patient outcomes.
Collapse
Affiliation(s)
- Abdellah Aazmi
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Duo Zhang
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 51817, China
| | - Corrado Mazzaglia
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| | - Mengfei Yu
- The Affiliated Stomatologic Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Zhen Wang
- Center for Laboratory Medicine, Allergy Center, Department of Transfusion Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Huayong Yang
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Yan Yan Shery Huang
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| | - Liang Ma
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
18
|
Menon R, Muglia LJ, Levin LH. Review on new approach methods to gain insight into the feto-maternal interface physiology. Front Med (Lausanne) 2023; 10:1304002. [PMID: 38098843 PMCID: PMC10720461 DOI: 10.3389/fmed.2023.1304002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/13/2023] [Indexed: 12/17/2023] Open
Abstract
Non-human animals represent a large and important feature in the history of biomedical research. The validity of their use, in terms of reproducible outcomes and translational confidence to the human situation, as well as ethical concerns surrounding that use, have been and remain controversial topics. Over the last 10 years, the communities developing microphysiological systems (MPS) have produced new approach method (NAMs) such as organoids and organs-on-a-chip. These alternative methodologies have shown indications of greater reliability and translatability than animal use in some areas, represent more humane substitutions for animals in these settings, and - with continued scientific effort - may change the conduct of basic research, clinical studies, safety testing, and drug development. Here, we present an introduction to these more human-relevant methodologies and suggest how a suite of pregnancy associated feto-maternal interface system-oriented NAMs may be integrated as reliable partial-/full animal replacements for investigators, significantly aid animal-/environmental welfare, and improve healthcare outcomes.
Collapse
Affiliation(s)
- Ramkumar Menon
- Department of Obstetrics and Gynecology, Division of Basic Science and Translational Research, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Louis J. Muglia
- The Burroughs Wellcome Fund, Research Triangle Park, NC, United States
- Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | | |
Collapse
|
19
|
Floryanzia SD, Nance E. Applications and Considerations for Microfluidic Systems To Model the Blood-Brain Barrier. ACS APPLIED BIO MATERIALS 2023; 6:3617-3632. [PMID: 37582179 PMCID: PMC11646049 DOI: 10.1021/acsabm.3c00364] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2023]
Abstract
In a myriad of developmental and degenerative brain diseases, characteristic pathological biomarkers are often associated with cerebral blood flow and vasculature. However, the relationship between vascular dysfunction and markers of brain disease is not well-defined. Additionally, it is difficult to deliver effective therapeutics to the brain due to the highly regulated blood-brain barrier (BBB) at the microvasculature interface of the brain. This Review first covers the need for modeling the BBB and the challenges of modeling the BBB. In vitro models of the BBB enable the study of the relationship between vascular dysfunction, BBB function, and disease progression and can serve as a platform to screen therapeutics. In particular, microfluidic-based in vitro BBB models are useful for studying brain vasculature as they support cell culture within the presence of continuous perfusion, which mirrors the in vivo flow and associated stress conditions in the brain. Early microfluidic models of the BBB created the most simplistic models possible that still displayed some functional aspects of the in vivo BBB. Therefore, this Review also discusses the emerging unique ways in which microfluidics in tandem with recent advancements in cell culture, biomaterials, and in vitro modeling can be used to develop more complex and physiologically relevant models of the BBB. Finally, we discuss the current and future state-of-the-art application of microfluidic BBB models for drug development and disease modeling, and the ongoing areas of needed innovation in this field.
Collapse
Affiliation(s)
- Sydney D Floryanzia
- Department of Chemical Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Elizabeth Nance
- Department of Chemical Engineering, University of Washington, Seattle, Washington 98195, United States
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
20
|
Keuper-Navis M, Walles M, Poller B, Myszczyszyn A, van der Made TK, Donkers J, Eslami Amirabadi H, Wilmer MJ, Aan S, Spee B, Masereeuw R, van de Steeg E. The application of organ-on-chip models for the prediction of human pharmacokinetic profiles during drug development. Pharmacol Res 2023; 195:106853. [PMID: 37473876 DOI: 10.1016/j.phrs.2023.106853] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 07/11/2023] [Accepted: 07/14/2023] [Indexed: 07/22/2023]
Abstract
Organ-on-chip (OoC) technology has led to in vitro models with many new possibilities compared to conventional in vitro and in vivo models. In this review, the potential of OoC models to improve the prediction of human oral bioavailability and intrinsic clearance is discussed, with a focus on the functionality of the models and the application in current drug development practice. Multi-OoC models demonstrating the application for pharmacokinetic (PK) studies are summarized and existing challenges are identified. Physiological parameters for a minimal viable platform of a multi-OoC model to study PK are provided, together with PK specific read-outs and recommendations for relevant reference compounds to validate the model. Finally, the translation to in vivo PK profiles is discussed, which will be required to routinely apply OoC models during drug development.
Collapse
Affiliation(s)
- Marit Keuper-Navis
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Leiden, the Netherlands; Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, the Netherlands
| | - Markus Walles
- Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Birk Poller
- Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Adam Myszczyszyn
- Faculty of Veterinary Medicine & Regenerative Medicine Center Utrecht (RMCU), Utrecht University, Utrecht, the Netherlands
| | - Thomas K van der Made
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, the Netherlands
| | - Joanne Donkers
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Leiden, the Netherlands
| | | | | | - Saskia Aan
- Stichting Proefdiervrij, Den Haag, the Netherlands
| | - Bart Spee
- Faculty of Veterinary Medicine & Regenerative Medicine Center Utrecht (RMCU), Utrecht University, Utrecht, the Netherlands
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, the Netherlands
| | - Evita van de Steeg
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Leiden, the Netherlands.
| |
Collapse
|
21
|
German C, Chen Z, Przekwas A, Walenga R, Babiskin A, Zhao L, Fan J, Tan ML. Computational Model of In Vivo Corneal Pharmacokinetics and Pharmacodynamics of Topically Administered Ophthalmic Drug Products. Pharm Res 2023; 40:961-975. [PMID: 36959411 DOI: 10.1007/s11095-023-03480-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 02/09/2023] [Indexed: 03/25/2023]
Abstract
INTRODUCTION Although the eye is directly accessible on the surface of the human body, drug delivery can be extremely challenging due to the presence of multiple protective barriers in eye tissues. Researchers have developed complex formulation strategies to overcome these barriers to ophthalmic drug delivery. Current development strategies rely heavily on in vitro experiments and animal testing to predict human pharmacokinetics (PK) and pharmacodynamics (PD). OBJECTIVE The primary objective of the study was to develop a high-fidelity PK/PD model of the anterior eye for topical application of ophthalmic drug products. METHODS Here, we present a physiologically-based in silico approach to predicting PK and PD in rabbits after topical administration of ophthalmic products. A first-principles based approach was used to describe timolol dissolution, transport, and distribution, including consideration of ionized transport, following topical instillation of a timolol suspension. RESULTS Using literature transport and response parameters, the computational model described well the concentration-time and response-time profiles in rabbit. Comparison of validated rabbit model results and extrapolated human model results demonstrate observable differences in the distribution of timolol at multiple time points. CONCLUSION This modeling framework provides a tool for model-based prediction of PK in eye tissues and PD after topical ophthalmic drug administration to the eyes.
Collapse
Affiliation(s)
- Carrie German
- CFD Research Corporation, Computational Biology Division, 6820 Moquin Dr NW, Huntsville, AL, 35806, USA.
| | - Zhijian Chen
- CFD Research Corporation, Computational Biology Division, 6820 Moquin Dr NW, Huntsville, AL, 35806, USA
| | - Andrzej Przekwas
- CFD Research Corporation, Computational Biology Division, 6820 Moquin Dr NW, Huntsville, AL, 35806, USA
| | - Ross Walenga
- Division of Quantitative Methods and Modeling, Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA
| | - Andrew Babiskin
- Division of Quantitative Methods and Modeling, Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA
| | - Liang Zhao
- Division of Quantitative Methods and Modeling, Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA
| | - Jianghong Fan
- Division of Pharmacometrics, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA
| | - Ming-Liang Tan
- Division of Quantitative Methods and Modeling, Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA
| |
Collapse
|
22
|
Akalın AA, Dedekargınoğlu B, Choi SR, Han B, Ozcelikkale A. Predictive Design and Analysis of Drug Transport by Multiscale Computational Models Under Uncertainty. Pharm Res 2023; 40:501-523. [PMID: 35650448 PMCID: PMC9712595 DOI: 10.1007/s11095-022-03298-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 05/17/2022] [Indexed: 01/18/2023]
Abstract
Computational modeling of drug delivery is becoming an indispensable tool for advancing drug development pipeline, particularly in nanomedicine where a rational design strategy is ultimately sought. While numerous in silico models have been developed that can accurately describe nanoparticle interactions with the bioenvironment within prescribed length and time scales, predictive design of these drug carriers, dosages and treatment schemes will require advanced models that can simulate transport processes across multiple length and time scales from genomic to population levels. In order to address this problem, multiscale modeling efforts that integrate existing discrete and continuum modeling strategies have recently emerged. These multiscale approaches provide a promising direction for bottom-up in silico pipelines of drug design for delivery. However, there are remaining challenges in terms of model parametrization and validation in the presence of variability, introduced by multiple levels of heterogeneities in disease state. Parametrization based on physiologically relevant in vitro data from microphysiological systems as well as widespread adoption of uncertainty quantification and sensitivity analysis will help address these challenges.
Collapse
Affiliation(s)
- Ali Aykut Akalın
- Department of Mechanical Engineering, Middle East Technical University, 06531, Ankara, Turkey
| | - Barış Dedekargınoğlu
- Department of Mechanical Engineering, Middle East Technical University, 06531, Ankara, Turkey
| | - Sae Rome Choi
- School of Mechanical Engineering, Purdue University, 585 Purdue Mall, West Lafayette, Indiana, 47907, USA
| | - Bumsoo Han
- School of Mechanical Engineering, Purdue University, 585 Purdue Mall, West Lafayette, Indiana, 47907, USA.
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA.
- Center for Cancer Research, Purdue University, 585 Purdue Mall, West Lafayette, Indiana, 47907, USA.
| | - Altug Ozcelikkale
- Department of Mechanical Engineering, Middle East Technical University, 06531, Ankara, Turkey.
| |
Collapse
|
23
|
MacRae CA, Peterson RT. Zebrafish as a Mainstream Model for In Vivo Systems Pharmacology and Toxicology. Annu Rev Pharmacol Toxicol 2023; 63:43-64. [PMID: 36151053 DOI: 10.1146/annurev-pharmtox-051421-105617] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Pharmacology and toxicology are part of a much broader effort to understand the relationship between chemistry and biology. While biomedicine has necessarily focused on specific cases, typically of direct human relevance, there are real advantages in pursuing more systematic approaches to characterizing how health and disease are influenced by small molecules and other interventions. In this context, the zebrafish is now established as the representative screenable vertebrate and, through ongoing advances in the available scale of genome editing and automated phenotyping, is beginning to address systems-level solutions to some biomedical problems. The addition of broader efforts to integrate information content across preclinical model organisms and the incorporation of rigorous analytics, including closed-loop deep learning, will facilitate efforts to create systems pharmacology and toxicology with the ability to continuously optimize chemical biological interactions around societal needs. In this review, we outline progress toward this goal.
Collapse
Affiliation(s)
- Calum A MacRae
- Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, USA;
| | | |
Collapse
|
24
|
Jiang L, Li Q, Liang W, Du X, Yang Y, Zhang Z, Xu L, Zhang J, Li J, Chen Z, Gu Z. Organ-On-A-Chip Database Revealed-Achieving the Human Avatar in Silicon. Bioengineering (Basel) 2022; 9:685. [PMID: 36421086 PMCID: PMC9687773 DOI: 10.3390/bioengineering9110685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Organ-on-a-chip (OOC) provides microphysiological conditions on a microfluidic chip, which makes up for the shortcomings of traditional in vitro cellular culture models and animal models. It has broad application prospects in drug development and screening, toxicological mechanism research, and precision medicine. A large amount of data could be generated through its applications, including image data, measurement data from sensors, ~omics data, etc. A database with proper architecture is required to help scholars in this field design experiments, organize inputted data, perform analysis, and promote the future development of novel OOC systems. In this review, we overview existing OOC databases that have been developed, including the BioSystics Analytics Platform (BAP) developed by the University of Pittsburgh, which supports study design as well as data uploading, storage, visualization, analysis, etc., and the organ-on-a-chip database (Ocdb) developed by Southeast University, which has collected a large amount of literature and patents as well as relevant toxicological and pharmaceutical data and provides other major functions. We used examples to overview how the BAP database has contributed to the development and applications of OOC technology in the United States for the MPS consortium and how the Ocdb has supported researchers in the Chinese Organoid and Organs-On-A-Chip society. Lastly, the characteristics, advantages, and limitations of these two databases were discussed.
Collapse
Affiliation(s)
- Lincao Jiang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, SiPaiLou # 2, Nanjing 210096, China
| | - Qiwei Li
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, SiPaiLou # 2, Nanjing 210096, China
| | - Weicheng Liang
- School of Life Science and Technology, Southeast University, Nanjing 210096, China
| | - Xuan Du
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, SiPaiLou # 2, Nanjing 210096, China
| | - Yi Yang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, SiPaiLou # 2, Nanjing 210096, China
| | - Zilin Zhang
- Jiangsu Avartarget Biotechnology Corp., Suzhou 215163, China
| | - Lili Xu
- Jiangsu Avartarget Biotechnology Corp., Suzhou 215163, China
| | - Jing Zhang
- Jiangsu Avartarget Biotechnology Corp., Suzhou 215163, China
| | - Jian Li
- School of Life Science and Technology, Southeast University, Nanjing 210096, China
| | - Zaozao Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, SiPaiLou # 2, Nanjing 210096, China
| | - Zhongze Gu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, SiPaiLou # 2, Nanjing 210096, China
| |
Collapse
|
25
|
Sung B. In silico modeling of endocrine organ-on-a-chip systems. Math Biosci 2022; 352:108900. [PMID: 36075288 DOI: 10.1016/j.mbs.2022.108900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 08/30/2022] [Accepted: 08/31/2022] [Indexed: 10/14/2022]
Abstract
The organ-on-a-chip (OoC) is an artificially reconstructed microphysiological system that is implemented using tissue mimics integrated into miniaturized perfusion devices. OoCs emulate dynamic and physiologically relevant features of the body, which are not available in standard in vitro methods. Furthermore, OoCs provide highly sophisticated multi-organ connectivity and biomechanical cues based on microfluidic platforms. Consequently, they are often considered ideal in vitro systems for mimicking self-regulating biophysical and biochemical networks in vivo where multiple tissues and organs crosstalk through the blood flow, similar to the human endocrine system. Therefore, OoCs have been extensively applied to simulate complex hormone dynamics and endocrine signaling pathways in a mechanistic and fully controlled manner. Mathematical and computational modeling approaches are critical for quantitatively analyzing an OoC and predicting its complex responses. In this review article, recently developed in silico modeling concepts of endocrine OoC systems are summarized, including the mathematical models of tissue-level transport phenomena, microscale fluid dynamics, distant hormone signaling, and heterogeneous cell-cell communication. From this background, whole chip-level analytic approaches in pharmacokinetics and pharmacodynamics will be described with a focus on the spatial and temporal behaviors of absorption, distribution, metabolism, and excretion in endocrine biochips. Finally, quantitative design frameworks for endocrine OoCs are reviewed with respect to support parameter calibration/scaling and enable predictive in vitro-in vivo extrapolations. In particular, we highlight the analytical and numerical modeling strategies of the nonlinear phenomena in endocrine systems on-chip, which are of particular importance in drug screening and environmental health applications.
Collapse
Affiliation(s)
- Baeckkyoung Sung
- Biosensor Group, KIST Europe Forschungsgesellschaft mbH, 66123 Saarbrücken, Germany; Division of Energy & Environment Technology, University of Science & Technology, 34113 Daejeon, Republic of Korea.
| |
Collapse
|
26
|
Bai H, Ingber DE. What Can an Organ-on-a-Chip Teach Us About Human Lung Pathophysiology? Physiology (Bethesda) 2022; 37:0. [PMID: 35658627 PMCID: PMC9394778 DOI: 10.1152/physiol.00012.2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/31/2022] [Accepted: 06/01/2022] [Indexed: 12/25/2022] Open
Abstract
The intertwined relationship between structure and function has been key to understanding human organ physiology and disease pathogenesis. An organ-on-a-chip (organ chip) is a bioengineered microfluidic cell culture device lined by living cells and tissues that recapitulates organ-level functions in vitro. This is accomplished by recreating organ-specific tissue-tissue interfaces and microenvironmental biochemical and mechanical cues while providing dynamic perfusion through endothelium-lined vascular channels. In this review, we discuss how this emerging technology has contributed to the understanding of human lung structure-function relationships at the cell, tissue, and organ levels.
Collapse
Affiliation(s)
- Haiqing Bai
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts
- Vascular Biology Program, Boston Children's Hospital and Department of Surgery, Harvard Medical School, Boston, Massachusetts
- Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, Massachusetts
| |
Collapse
|
27
|
Liu J, Feng C, Zhang M, Song F, Liu H. Design and Fabrication of a Liver-on-a-chip Reconstructing Tissue-tissue Interfaces. Front Oncol 2022; 12:959299. [PMID: 35992870 PMCID: PMC9389071 DOI: 10.3389/fonc.2022.959299] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 06/24/2022] [Indexed: 11/25/2022] Open
Abstract
Despite the rapid advances in the liver-on-a-chip platforms, it remains a daunting challenge to construct a biomimetic liver-on-a-chip for in vitro research. This study aimed to reconstruct the tissue-tissue interfaces based on bilayer microspheres and form vascularized liver tissue. Firstly, we designed a tri-vascular liver-on-a-chip (TVLOC) comprising a hepatic artery, a portal vein and a central vein, and theoretically analyzed the distribution of velocity and concentration fields in the culture area. Secondly, we designed a bilayer microsphere generating microsystem based on the coaxial confocal principle, which is primarily used to produce bilayer microspheres containing different kinds of cells. Finally, the bilayer microspheres were co-cultured with endothelial cells in the cell culture area of the TVLOC to form vascularized liver tissue, and the cell viability and vascular network growth were analyzed. The results revealed that the TVLOC designed in this study can provide a substance concentration gradient similar to that of the liver microenvironment, and the bilayer microspheres can form a three-dimensional (3D) orderly liver structure with endothelial cells. Such a liver-on-a-chip is capable of maintaining the function of hepatocytes (HCs) pretty well. This work provides full insights into further simulation of the liver-on-a-chip.
Collapse
Affiliation(s)
- Jing Liu
- School of Biology, Food and Environment, Hefei University, Hefei, China
| | - Chong Feng
- School of Biology, Food and Environment, Hefei University, Hefei, China
| | - Min Zhang
- School of Biology, Food and Environment, Hefei University, Hefei, China
| | - Feng Song
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou, China
- *Correspondence: Haochen Liu, ; Feng Song,
| | - Haochen Liu
- Department of Cardiovascular Surgery, Xi’an Children’s Hospital, Xi’an, China
- *Correspondence: Haochen Liu, ; Feng Song,
| |
Collapse
|
28
|
Abstract
The failure of animal models to predict therapeutic responses in humans is a major problem that also brings into question their use for basic research. Organ-on-a-chip (organ chip) microfluidic devices lined with living cells cultured under fluid flow can recapitulate organ-level physiology and pathophysiology with high fidelity. Here, I review how single and multiple human organ chip systems have been used to model complex diseases and rare genetic disorders, to study host-microbiome interactions, to recapitulate whole-body inter-organ physiology and to reproduce human clinical responses to drugs, radiation, toxins and infectious pathogens. I also address the challenges that must be overcome for organ chips to be accepted by the pharmaceutical industry and regulatory agencies, as well as discuss recent advances in the field. It is evident that the use of human organ chips instead of animal models for drug development and as living avatars for personalized medicine is ever closer to realization.
Collapse
Affiliation(s)
- Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA.
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA.
- Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, MA, USA.
| |
Collapse
|
29
|
Rusyn I, Sakolish C, Kato Y, Stephan C, Vergara L, Hewitt P, Bhaskaran V, Davis M, Hardwick RN, Ferguson SS, Stanko JP, Bajaj P, Adkins K, Sipes NS, Hunter ES, Baltazar MT, Carmichael PL, Sadh K, Becker RA. Microphysiological Systems Evaluation: Experience of TEX-VAL Tissue Chip Testing Consortium. Toxicol Sci 2022; 188:143-152. [PMID: 35689632 PMCID: PMC9333404 DOI: 10.1093/toxsci/kfac061] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Much has been written and said about the promise and excitement of microphysiological systems, miniature devices that aim to recreate aspects of human physiology on a chip. The rapid explosion of the offerings and persistent publicity placed high expectations on both product manufacturers and regulatory agencies to adopt the data. Inevitably, discussions of where this technology fits in chemical testing paradigms are ongoing. Some end-users became early adopters, whereas others have taken a more cautious approach because of the high cost and uncertainties of their utility. Here, we detail the experience of a public-private collaboration established for testing of diverse microphysiological systems. Collectively, we present a number of considerations on practical aspects of using microphysiological systems in the context of their applications in decision-making. Specifically, future end-users need to be prepared for extensive on-site optimization and have access to a wide range of imaging and other equipment. We reason that cells, related reagents, and the technical skills of the research staff, not the devices themselves, are the most critical determinants of success. Extrapolation from concentration-response effects in microphysiological systems to human blood or oral exposures, difficulties with replicating the whole organ, and long-term functionality remain as critical challenges. Overall, we conclude that it is unlikely that a rodent- or human-equivalent model is achievable through a finite number of microphysiological systems in the near future; therefore, building consensus and promoting the gradual incorporation of these models into tiered approaches for safety assessment and decision-making is the sensible path to wide adoption.
Collapse
Affiliation(s)
- Ivan Rusyn
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843, USA
| | - Courtney Sakolish
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843, USA
| | - Yuki Kato
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843, USA
| | - Clifford Stephan
- Institute of Biosciences and Technology, Texas A&M University, Houston, Texas 77030, USA
| | - Leoncio Vergara
- Institute of Biosciences and Technology, Texas A&M University, Houston, Texas 77030, USA
| | - Philip Hewitt
- Chemical and Preclinical Safety, Merck Healthcare KGaA, Darmstadt, Germany
| | - Vasanthi Bhaskaran
- Discovery Toxicology, Bristol Myers Squibb, Princeton, New Jersey 08543, USA
| | - Myrtle Davis
- Discovery Toxicology, Bristol Myers Squibb, Princeton, New Jersey 08543, USA
| | - Rhiannon N Hardwick
- Discovery Toxicology, Bristol Myers Squibb, San Diego, California 92130, USA
| | - Stephen S Ferguson
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, USA
| | - Jason P Stanko
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, USA
| | - Piyush Bajaj
- Global Investigative Toxicology, Preclinical Safety, Sanofi, Framingham, Massachusetts 01701, USA
| | - Karissa Adkins
- Global Investigative Toxicology, Preclinical Safety, Sanofi, Framingham, Massachusetts 01701, USA
| | - Nisha S Sipes
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, 27711, USA
| | - E Sidney Hunter
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, 27711, USA
| | - Maria T Baltazar
- Unilever Safety and Environmental Assurance Centre, Bedfordshire, Sharnbrook MK44 1LQ, UK
| | - Paul L Carmichael
- Unilever Safety and Environmental Assurance Centre, Bedfordshire, Sharnbrook MK44 1LQ, UK
| | - Kritika Sadh
- Unilever Safety and Environmental Assurance Centre, Bedfordshire, Sharnbrook MK44 1LQ, UK
| | - Richard A Becker
- American Chemistry Council, Washington, District of Columbia 20002, USA
| |
Collapse
|
30
|
Singh VK, Seed TM. Acute radiation syndrome drug discovery using organ-on-chip platforms. Expert Opin Drug Discov 2022; 17:865-878. [PMID: 35838021 DOI: 10.1080/17460441.2022.2099833] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION : The high attrition rate during drug development remains a challenge that costs a significant amount of time and money. Improving the probabilities of success during the early stages of radiation medical countermeasure (MCM) development for approval by the United States Food and Drug Administration (US FDA) following the Animal Rule will reduce this burden. For optimal development of MCMs, we need suitable and efficient radiation injury models with high biological relevance for evaluating drug efficacy as well as biomarker discovery and validation. AREA COVERED This article focuses on new technologies involving various organs-on-chip platforms. Of late, there have been rapid development of these technologies, especially in terms of mimicking both normal and abnormal physiological conditions. Here, we suggest possible applications of these novel systems for the discovery and development of radiation MCMs for the acute radiation syndrome (ARS). We offer preliminary information on the utility of one such system for MCM research and discovery for the ARS condition. EXPERT OPINION : Each organ-on-a-chip system has its own strengths and shortcomings. As such, the system selected for MCM discovery, development, and regulatory approval should be carefully considered and optimized to the fullest extent in order to augment successful drug testing and the minimization of attrition rates of candidate agents. The recent encouraging progress with organ-on-a-chip technology will likely lead to additional radiation MCMs for ARS approved by the US FDA. The acceptance of organ-on-a-chip technology may be a promising step toward improving the success rate of pharmaceuticals in MCM development.
Collapse
Affiliation(s)
- Vijay K Singh
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Thomas M Seed
- Tech Micro Services, 4417 Maple Avenue, Bethesda, MD, USA
| |
Collapse
|
31
|
Guo T, He C, Venado A, Zhou Y. Extracellular Matrix Stiffness in Lung Health and Disease. Compr Physiol 2022; 12:3523-3558. [PMID: 35766837 PMCID: PMC10088466 DOI: 10.1002/cphy.c210032] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The extracellular matrix (ECM) provides structural support and imparts a wide variety of environmental cues to cells. In the past decade, a growing body of work revealed that the mechanical properties of the ECM, commonly known as matrix stiffness, regulate the fundamental cellular processes of the lung. There is growing appreciation that mechanical interplays between cells and associated ECM are essential to maintain lung homeostasis. Dysregulation of ECM-derived mechanical signaling via altered mechanosensing and mechanotransduction pathways is associated with many common lung diseases. Matrix stiffening is a hallmark of lung fibrosis. The stiffened ECM is not merely a sequelae of lung fibrosis but can actively drive the progression of fibrotic lung disease. In this article, we provide a comprehensive view on the role of matrix stiffness in lung health and disease. We begin by summarizing the effects of matrix stiffness on the function and behavior of various lung cell types and on regulation of biomolecule activity and key physiological processes, including host immune response and cellular metabolism. We discuss the potential mechanisms by which cells probe matrix stiffness and convert mechanical signals to regulate gene expression. We highlight the factors that govern matrix stiffness and outline the role of matrix stiffness in lung development and the pathogenesis of pulmonary fibrosis, pulmonary hypertension, asthma, chronic obstructive pulmonary disease (COPD), and lung cancer. We envision targeting of deleterious matrix mechanical cues for treatment of fibrotic lung disease. Advances in technologies for matrix stiffness measurements and design of stiffness-tunable matrix substrates are also explored. © 2022 American Physiological Society. Compr Physiol 12:3523-3558, 2022.
Collapse
Affiliation(s)
- Ting Guo
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Alabama, USA.,Department of Respiratory Medicine, the Second Xiangya Hospital, Central-South University, Changsha, Hunan, China
| | - Chao He
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Alabama, USA
| | - Aida Venado
- Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Yong Zhou
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Alabama, USA
| |
Collapse
|
32
|
Li Z, Hui J, Yang P, Mao H. Microfluidic Organ-on-a-Chip System for Disease Modeling and Drug Development. BIOSENSORS 2022; 12:bios12060370. [PMID: 35735518 PMCID: PMC9220862 DOI: 10.3390/bios12060370] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/15/2022] [Accepted: 05/24/2022] [Indexed: 05/05/2023]
Abstract
An organ-on-a-chip is a device that combines micro-manufacturing and tissue engineering to replicate the critical physiological environment and functions of the human organs. Therefore, it can be used to predict drug responses and environmental effects on organs. Microfluidic technology can control micro-scale reagents with high precision. Hence, microfluidics have been widely applied in organ-on-chip systems to mimic specific organ or multiple organs in vivo. These models integrated with various sensors show great potential in simulating the human environment. In this review, we mainly introduce the typical structures and recent research achievements of several organ-on-a-chip platforms. We also discuss innovations in models applied to the fields of pharmacokinetics/pharmacodynamics, nano-medicine, continuous dynamic monitoring in disease modeling, and their further applications in other fields.
Collapse
Affiliation(s)
- Zening Li
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China; (Z.L.); (J.H.); (P.Y.)
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jianan Hui
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China; (Z.L.); (J.H.); (P.Y.)
| | - Panhui Yang
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China; (Z.L.); (J.H.); (P.Y.)
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hongju Mao
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China; (Z.L.); (J.H.); (P.Y.)
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
- Correspondence: ; Tel.: +86-21-62511070-8707
| |
Collapse
|
33
|
Chang X, Tan YM, Allen DG, Bell S, Brown PC, Browning L, Ceger P, Gearhart J, Hakkinen PJ, Kabadi SV, Kleinstreuer NC, Lumen A, Matheson J, Paini A, Pangburn HA, Petersen EJ, Reinke EN, Ribeiro AJS, Sipes N, Sweeney LM, Wambaugh JF, Wange R, Wetmore BA, Mumtaz M. IVIVE: Facilitating the Use of In Vitro Toxicity Data in Risk Assessment and Decision Making. TOXICS 2022; 10:232. [PMID: 35622645 PMCID: PMC9143724 DOI: 10.3390/toxics10050232] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 04/24/2022] [Indexed: 02/04/2023]
Abstract
During the past few decades, the science of toxicology has been undergoing a transformation from observational to predictive science. New approach methodologies (NAMs), including in vitro assays, in silico models, read-across, and in vitro to in vivo extrapolation (IVIVE), are being developed to reduce, refine, or replace whole animal testing, encouraging the judicious use of time and resources. Some of these methods have advanced past the exploratory research stage and are beginning to gain acceptance for the risk assessment of chemicals. A review of the recent literature reveals a burst of IVIVE publications over the past decade. In this review, we propose operational definitions for IVIVE, present literature examples for several common toxicity endpoints, and highlight their implications in decision-making processes across various federal agencies, as well as international organizations, including those in the European Union (EU). The current challenges and future needs are also summarized for IVIVE. In addition to refining and reducing the number of animals in traditional toxicity testing protocols and being used for prioritizing chemical testing, the goal to use IVIVE to facilitate the replacement of animal models can be achieved through their continued evolution and development, including a strategic plan to qualify IVIVE methods for regulatory acceptance.
Collapse
Affiliation(s)
- Xiaoqing Chang
- Inotiv-RTP, 601 Keystone Park Drive, Suite 200, Morrisville, NC 27560, USA; (X.C.); (D.G.A.); (S.B.); (L.B.); (P.C.)
| | - Yu-Mei Tan
- U.S. Environmental Protection Agency, Office of Pesticide Programs, 109 T.W. Alexander Drive, Durham, NC 27709, USA;
| | - David G. Allen
- Inotiv-RTP, 601 Keystone Park Drive, Suite 200, Morrisville, NC 27560, USA; (X.C.); (D.G.A.); (S.B.); (L.B.); (P.C.)
| | - Shannon Bell
- Inotiv-RTP, 601 Keystone Park Drive, Suite 200, Morrisville, NC 27560, USA; (X.C.); (D.G.A.); (S.B.); (L.B.); (P.C.)
| | - Paul C. Brown
- U.S. Food and Drug Administration, Center for Drug Evaluation and Research, 10903 New Hampshire Avenue, Silver Spring, MD 20903, USA; (P.C.B.); (A.J.S.R.); (R.W.)
| | - Lauren Browning
- Inotiv-RTP, 601 Keystone Park Drive, Suite 200, Morrisville, NC 27560, USA; (X.C.); (D.G.A.); (S.B.); (L.B.); (P.C.)
| | - Patricia Ceger
- Inotiv-RTP, 601 Keystone Park Drive, Suite 200, Morrisville, NC 27560, USA; (X.C.); (D.G.A.); (S.B.); (L.B.); (P.C.)
| | - Jeffery Gearhart
- The Henry M. Jackson Foundation, Air Force Research Laboratory, 711 Human Performance Wing, Wright-Patterson Air Force Base, OH 45433, USA;
| | - Pertti J. Hakkinen
- National Library of Medicine, National Center for Biotechnology Information, 8600 Rockville Pike, Bethesda, MD 20894, USA;
| | - Shruti V. Kabadi
- U.S. Food and Drug Administration, Center for Food Safety and Applied Nutrition, Office of Food Additive Safety, 5001 Campus Drive, HFS-275, College Park, MD 20740, USA;
| | - Nicole C. Kleinstreuer
- National Institute of Environmental Health Sciences, National Toxicology Program Interagency Center for the Evaluation of Alternative Toxicological Methods, P.O. Box 12233, Research Triangle Park, NC 27709, USA;
| | - Annie Lumen
- U.S. Food and Drug Administration, National Center for Toxicological Research, 3900 NCTR Road, Jefferson, AR 72079, USA;
| | - Joanna Matheson
- U.S. Consumer Product Safety Commission, Division of Toxicology and Risk Assessment, 5 Research Place, Rockville, MD 20850, USA;
| | - Alicia Paini
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy;
| | - Heather A. Pangburn
- Air Force Research Laboratory, 711 Human Performance Wing, 2729 R Street, Area B, Building 837, Wright-Patterson Air Force Base, OH 45433, USA;
| | - Elijah J. Petersen
- U.S. Department of Commerce, National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, MD 20899, USA;
| | - Emily N. Reinke
- U.S. Army Public Health Center, 8252 Blackhawk Rd., Aberdeen Proving Ground, MD 21010, USA;
| | - Alexandre J. S. Ribeiro
- U.S. Food and Drug Administration, Center for Drug Evaluation and Research, 10903 New Hampshire Avenue, Silver Spring, MD 20903, USA; (P.C.B.); (A.J.S.R.); (R.W.)
| | - Nisha Sipes
- U.S. Environmental Protection Agency, Center for Computational Toxicology and Exposure, 109 TW Alexander Dr., Research Triangle Park, NC 27711, USA; (N.S.); (J.F.W.); (B.A.W.)
| | - Lisa M. Sweeney
- UES, Inc., 4401 Dayton-Xenia Road, Beavercreek, OH 45432, Assigned to Air Force Research Laboratory, 711 Human Performance Wing, Wright-Patterson Air Force Base, OH 45433, USA;
| | - John F. Wambaugh
- U.S. Environmental Protection Agency, Center for Computational Toxicology and Exposure, 109 TW Alexander Dr., Research Triangle Park, NC 27711, USA; (N.S.); (J.F.W.); (B.A.W.)
| | - Ronald Wange
- U.S. Food and Drug Administration, Center for Drug Evaluation and Research, 10903 New Hampshire Avenue, Silver Spring, MD 20903, USA; (P.C.B.); (A.J.S.R.); (R.W.)
| | - Barbara A. Wetmore
- U.S. Environmental Protection Agency, Center for Computational Toxicology and Exposure, 109 TW Alexander Dr., Research Triangle Park, NC 27711, USA; (N.S.); (J.F.W.); (B.A.W.)
| | - Moiz Mumtaz
- Agency for Toxic Substances and Disease Registry, Office of the Associate Director for Science, 1600 Clifton Road, S102-2, Atlanta, GA 30333, USA
| |
Collapse
|
34
|
Yang Y, Chen Y, Wang L, Xu S, Fang G, Guo X, Chen Z, Gu Z. PBPK Modeling on Organs-on-Chips: An Overview of Recent Advancements. Front Bioeng Biotechnol 2022; 10:900481. [PMID: 35497341 PMCID: PMC9046607 DOI: 10.3389/fbioe.2022.900481] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 03/29/2022] [Indexed: 12/31/2022] Open
Abstract
Organ-on-a-chip (OoC) is a new and promising technology, which aims to improve the efficiency of drug development and realize personalized medicine by simulating in vivo environment in vitro. Physiologically based pharmacokinetic (PBPK) modeling is believed to have the advantage of better reflecting the absorption, distribution, metabolism and excretion process of drugs in vivo than traditional compartmental or non-compartmental pharmacokinetic models. The combination of PBPK modeling and organ-on-a-chip is believed to provide a strong new tool for new drug development and have the potential to replace animal testing. This article provides the recent development of organ-on-a-chip technology and PBPK modeling including model construction, parameter estimation and validation strategies. Application of PBPK modeling on Organ-on-a-Chip (OoC) has been emphasized, and considerable progress has been made. PBPK modeling on OoC would become an essential part of new drug development, personalized medicine and other fields.
Collapse
Affiliation(s)
- Yi Yang
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
| | - Yin Chen
- Jiangsu Provincial Center for Disease Control and Prevention, Key Laboratory of Enteric Pathogenic Microbiology, Ministry Health, Institute of Pathogenic Microbiology Health, Nanjing, China
| | - Liang Wang
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
- *Correspondence: Liang Wang, ; Zaozao Chen, ; Zhongze Gu,
| | - Shihui Xu
- Institute of Medical Devices (Suzhou), Southeast University, Suzhou, China
| | - Guoqing Fang
- Institute of Medical Devices (Suzhou), Southeast University, Suzhou, China
| | - Xilin Guo
- Jiangsu Provincial Center for Disease Control and Prevention, Key Laboratory of Enteric Pathogenic Microbiology, Ministry Health, Institute of Pathogenic Microbiology Health, Nanjing, China
| | - Zaozao Chen
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
- Institute of Medical Devices (Suzhou), Southeast University, Suzhou, China
- *Correspondence: Liang Wang, ; Zaozao Chen, ; Zhongze Gu,
| | - Zhongze Gu
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
- Institute of Medical Devices (Suzhou), Southeast University, Suzhou, China
- *Correspondence: Liang Wang, ; Zaozao Chen, ; Zhongze Gu,
| |
Collapse
|
35
|
Bai H, Si L, Jiang A, Belgur C, Zhai Y, Plebani R, Oh CY, Rodas M, Patil A, Nurani A, Gilpin SE, Powers RK, Goyal G, Prantil-Baun R, Ingber DE. Mechanical control of innate immune responses against viral infection revealed in a human lung alveolus chip. Nat Commun 2022; 13:1928. [PMID: 35396513 PMCID: PMC8993817 DOI: 10.1038/s41467-022-29562-4] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 03/23/2022] [Indexed: 12/24/2022] Open
Abstract
Mechanical breathing motions have a fundamental function in lung development and disease, but little is known about how they contribute to host innate immunity. Here we use a human lung alveolus chip that experiences cyclic breathing-like deformations to investigate whether physical forces influence innate immune responses to viral infection. Influenza H3N2 infection of mechanically active chips induces a cascade of host responses including increased lung permeability, apoptosis, cell regeneration, cytokines production, and recruitment of circulating immune cells. Comparison with static chips reveals that breathing motions suppress viral replication by activating protective innate immune responses in epithelial and endothelial cells, which are mediated in part through activation of the mechanosensitive ion channel TRPV4 and signaling via receptor for advanced glycation end products (RAGE). RAGE inhibitors suppress cytokines induction, while TRPV4 inhibition attenuates both inflammation and viral burden, in infected chips with breathing motions. Therefore, TRPV4 and RAGE may serve as new targets for therapeutic intervention in patients infected with influenza and other potential pandemic viruses that cause life-threatening lung inflammation.
Collapse
Affiliation(s)
- Haiqing Bai
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Longlong Si
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Amanda Jiang
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Chaitra Belgur
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Yunhao Zhai
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Roberto Plebani
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
- Center on Advanced Studies and Technology (CAST), Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, 66023, Italy
| | - Crystal Yuri Oh
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Melissa Rodas
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Aditya Patil
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Atiq Nurani
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Sarah E Gilpin
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Rani K Powers
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Girija Goyal
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Rachelle Prantil-Baun
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA.
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
- Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, MA, 02138, USA.
| |
Collapse
|
36
|
Ronaldson-Bouchard K, Teles D, Yeager K, Tavakol DN, Zhao Y, Chramiec A, Tagore S, Summers M, Stylianos S, Tamargo M, Lee BM, Halligan SP, Abaci EH, Guo Z, Jacków J, Pappalardo A, Shih J, Soni RK, Sonar S, German C, Christiano AM, Califano A, Hirschi KK, Chen CS, Przekwas A, Vunjak-Novakovic G. A multi-organ chip with matured tissue niches linked by vascular flow. Nat Biomed Eng 2022; 6:351-371. [PMID: 35478225 PMCID: PMC9250010 DOI: 10.1038/s41551-022-00882-6] [Citation(s) in RCA: 211] [Impact Index Per Article: 70.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 03/11/2022] [Indexed: 02/07/2023]
Abstract
Engineered tissues can be used to model human pathophysiology and test the efficacy and safety of drugs. Yet, to model whole-body physiology and systemic diseases, engineered tissues with preserved phenotypes need to physiologically communicate. Here we report the development and applicability of a tissue-chip system in which matured human heart, liver, bone and skin tissue niches are linked by recirculating vascular flow to allow for the recapitulation of interdependent organ functions. Each tissue is cultured in its own optimized environment and is separated from the common vascular flow by a selectively permeable endothelial barrier. The interlinked tissues maintained their molecular, structural and functional phenotypes over 4 weeks of culture, recapitulated the pharmacokinetic and pharmacodynamic profiles of doxorubicin in humans, allowed for the identification of early miRNA biomarkers of cardiotoxicity, and increased the predictive values of clinically observed miRNA responses relative to tissues cultured in isolation and to fluidically interlinked tissues in the absence of endothelial barriers. Vascularly linked and phenotypically stable matured human tissues may facilitate the clinical applicability of tissue chips.
Collapse
Affiliation(s)
| | - Diogo Teles
- Department of Biomedical Engineering, Columbia University, New York City, NY, USA
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarāes, Braga, Portugal
| | - Keith Yeager
- Department of Biomedical Engineering, Columbia University, New York City, NY, USA
| | | | - Yimu Zhao
- Department of Biomedical Engineering, Columbia University, New York City, NY, USA
| | - Alan Chramiec
- Department of Biomedical Engineering, Columbia University, New York City, NY, USA
| | - Somnath Tagore
- Department of Systems Biology, Columbia University, New York City, NY, USA
| | - Max Summers
- Department of Biomedical Engineering, Columbia University, New York City, NY, USA
| | - Sophia Stylianos
- Department of Biomedical Engineering, Columbia University, New York City, NY, USA
| | - Manuel Tamargo
- Department of Biomedical Engineering, Columbia University, New York City, NY, USA
| | - Busub Marcus Lee
- Department of Biomedical Engineering, Columbia University, New York City, NY, USA
| | - Susan P Halligan
- Department of Biomedical Engineering, Columbia University, New York City, NY, USA
| | - Erbil Hasan Abaci
- Department of Dermatology, Columbia University, New York City, NY, USA
| | - Zongyou Guo
- Department of Dermatology, Columbia University, New York City, NY, USA
| | - Joanna Jacków
- Department of Dermatology, Columbia University, New York City, NY, USA
| | | | - Jerry Shih
- Department of Biomedical Engineering, Boston University, The Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Rajesh K Soni
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York City, NY, USA
| | | | | | - Angela M Christiano
- Department of Dermatology, Columbia University, New York City, NY, USA
- Department of Genetics and Development, Columbia University, New York City, NY, USA
| | - Andrea Califano
- Department of Systems Biology, Columbia University, New York City, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York City, NY, USA
- Department of Biomedical Informatics, Columbia University, New York City, NY, USA
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York City, NY, USA
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA
- J.P. Sulzberger Columbia Genome Center, New York, NY, USA
| | - Karen K Hirschi
- Department of Cell Biology, University of Virginia, Charlottesville, VA, USA
| | - Christopher S Chen
- Department of Biomedical Engineering, Boston University, The Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | | | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York City, NY, USA.
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA.
- College of Dental Medicine, Columbia University, New York, NY, USA.
| |
Collapse
|
37
|
Lohasz C, Loretan J, Sterker D, Görlach E, Renggli K, Argast P, Frey O, Wiesmann M, Wartmann M, Rausch M, Hierlemann A. A Microphysiological Cell-Culturing System for Pharmacokinetic Drug Exposure and High-Resolution Imaging of Arrays of 3D Microtissues. Front Pharmacol 2022; 12:785851. [PMID: 35342386 PMCID: PMC8954798 DOI: 10.3389/fphar.2021.785851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/06/2021] [Indexed: 12/17/2022] Open
Abstract
Understanding the pharmacokinetic/pharmacodynamic (PK/PD)-relationship of a drug candidate is key to determine effective, yet safe treatment regimens for patients. However, current testing strategies are inefficient in characterizing in vivo responses to fluctuating drug concentrations during multi-day treatment cycles. Methods based on animal models are resource-intensive and require time, while traditional in vitro cell-culturing methods usually do not provide temporally-resolved information on the effects of in vivo–like drug exposure scenarios. To address this issue, we developed a microfluidic system to 1) culture arrays of three-dimensional spheroids in vitro, to 2) apply specific dynamic drug exposure profiles, and to 3) in-situ analyze spheroid growth and the invoked drug effects in 3D by means of 2-photon microscopy at tissue and single-cell level. Spheroids of fluorescently-labeled T-47D breast cancer cells were monitored under perfusion-culture conditions at short time intervals over three days and exposed to either three 24 h-PK-cycles or a dose-matched constant concentration of the phosphatidylinositol 3-kinase inhibitor BYL719. While the overall efficacy of the two treatment regimens was similar, spheroids exposed to the PK profile displayed cycle-dependent oscillations between regression and regrowth. Spheroids treated with a constant BYL719 concentration regressed at a steady, albeit slower rate. At a single-cell level, the cell density in BYL719-treated spheroids oscillated in a concentration-dependent manner. Our system represents a versatile tool for in-depth preclinical characterization of PK/PD parameters, as it enables an evaluation of drug efficacy and/or toxicity under realistic exposure conditions.
Collapse
Affiliation(s)
- Christian Lohasz
- ETH Zürich, Department of Biosystems Science and Engineering, Basel, Switzerland
| | | | - Dario Sterker
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | | | - Kasper Renggli
- ETH Zürich, Department of Biosystems Science and Engineering, Basel, Switzerland
| | - Paul Argast
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | | | - Marion Wiesmann
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Markus Wartmann
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Martin Rausch
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Andreas Hierlemann
- ETH Zürich, Department of Biosystems Science and Engineering, Basel, Switzerland
| |
Collapse
|
38
|
Huang Y, Chen Y, Lu S, Zhao C. Recent advance of <i>in vitro</i> models in natural phytochemicals absorption and metabolism. EFOOD 2022. [DOI: 10.53365/efood.k/146945] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Natural phytochemicals absorption and metabolic process are mainly in the human gut. Simulating the absorption and metabolism of natural phytochemicals in vitro to predict the rate and degree of absorption of natural phytochemicals provides convenience for many researchers. However, in this process, many physiological factors <i>in vitro</i> are affected, such as stomach and intestinal juice composition, pH, intestinal transmission rate and so on. In recent years, the research methods have gradually improved to make these models more suitable for the natural phytochemicals absorption process, <i>in vitro</i> simulation models have become an essential means to study natural phytochemicals absorption. Therefore, this paper introduces the advantages and disadvantages of commonly used <i>in vitro</i> simulation models of natural phytochemicals absorption and metabolism, as well as briefly introduces the working principle of each model. To provide a theoretical basis for simulating natural phytochemicals absorption <i>in vitro</i> and development and utilization of natural phytochemicals.
Collapse
|
39
|
Moossavi S, Arrieta MC, Sanati-Nezhad A, Bishehsari F. Gut-on-chip for ecological and causal human gut microbiome research. Trends Microbiol 2022; 30:710-721. [DOI: 10.1016/j.tim.2022.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 01/16/2022] [Accepted: 01/20/2022] [Indexed: 10/19/2022]
|
40
|
Eslami Amirabadi H, Donkers JM, Wierenga E, Ingenhut B, Pieters L, Stevens L, Donkers T, Westerhout J, Masereeuw R, Bobeldijk-Pastorova I, Nooijen I, van de Steeg E. Intestinal explant barrier chip: long-term intestinal absorption screening in a novel microphysiological system using tissue explants. LAB ON A CHIP 2022; 22:326-342. [PMID: 34877953 DOI: 10.1039/d1lc00669j] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The majority of intestinal in vitro screening models use cell lines that do not reflect the complexity of the human intestinal tract and hence often fail to accurately predict intestinal drug absorption. Tissue explants have intact intestinal architecture and cell type diversity, but show short viability in static conditions. Here, we present a medium throughput microphysiological system, Intestinal Explant Barrier Chip (IEBC), that creates a dynamic microfluidic microenvironment and prolongs tissue viability. Using a snap fit mechanism, we successfully incorporated human and porcine colon tissue explants and studied tissue functionality, integrity and viability for 24 hours. With a proper distinction of transcellular over paracellular transport (ratio >2), tissue functionality was good at early and late timepoints. Low leakage of FITC-dextran and preserved intracellular lactate dehydrogenase levels indicate maintained tissue integrity and viability, respectively. From a selection of low to high permeability drugs, 6 out of 7 properly ranked according to their fraction absorbed. In conclusion, the IEBC is a novel screening platform benefitting from the complexity of tissue explants and the flow in microfluidic chips.
Collapse
Affiliation(s)
- Hossein Eslami Amirabadi
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, The Netherlands.
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Joanne M Donkers
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, The Netherlands.
| | - Esmée Wierenga
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, The Netherlands.
| | - Bastiaan Ingenhut
- Materials solution department, TNO, and Brightlands Materials Centre, Geleen, The Netherlands
| | - Lisanne Pieters
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, The Netherlands.
| | - Lianne Stevens
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, The Netherlands.
- Department of Surgery, Division of Transplantation, Leiden University Medical Centre, Leiden, The Netherlands
| | - Tim Donkers
- Division of Space systems engineering, TNO, Delft, the Netherlands
| | | | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Ivana Bobeldijk-Pastorova
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, The Netherlands.
| | - Irene Nooijen
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, The Netherlands.
| | - Evita van de Steeg
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, The Netherlands.
| |
Collapse
|
41
|
Kundu B, Caballero D, Abreu CM, Reis RL, Kundu SC. The Tumor Microenvironment: An Introduction to the Development of Microfluidic Devices. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1379:115-138. [DOI: 10.1007/978-3-031-04039-9_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
42
|
Three-Dimensional Culture Systems for Dissecting Notch Signalling in Health and Disease. Int J Mol Sci 2021; 22:ijms222212473. [PMID: 34830355 PMCID: PMC8618738 DOI: 10.3390/ijms222212473] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 12/12/2022] Open
Abstract
Three-dimensional (3D) culture systems opened up new horizons in studying the biology of tissues and organs, modelling various diseases, and screening drugs. Producing accurate in vitro models increases the possibilities for studying molecular control of cell–cell and cell–microenvironment interactions in detail. The Notch signalling is linked to cell fate determination, tissue definition, and maintenance in both physiological and pathological conditions. Hence, 3D cultures provide new accessible platforms for studying activation and modulation of the Notch pathway. In this review, we provide an overview of the recent advances in different 3D culture systems, including spheroids, organoids, and “organ-on-a-chip” models, and their use in analysing the crucial role of Notch signalling in the maintenance of tissue homeostasis, pathology, and regeneration.
Collapse
|
43
|
Lee Y, Kim MH, Alves DR, Kim S, Lee LP, Sung JH, Park S. Gut-Kidney Axis on Chip for Studying Effects of Antibiotics on Risk of Hemolytic Uremic Syndrome by Shiga Toxin-Producing Escherichia coli. Toxins (Basel) 2021; 13:toxins13110775. [PMID: 34822559 PMCID: PMC8622205 DOI: 10.3390/toxins13110775] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 10/23/2021] [Accepted: 10/30/2021] [Indexed: 12/30/2022] Open
Abstract
Shiga toxin-producing Escherichia coli (STEC) infects humans by colonizing the large intestine, and causes kidney damage by secreting Shiga toxins (Stxs). The increased secretion of Shiga toxin 2 (Stx2) by some antibiotics, such as ciprofloxacin (CIP), increases the risk of hemolytic–uremic syndrome (HUS), which can be life-threatening. However, previous studies evaluating this relationship have been conflicting, owing to the low frequency of EHEC infection, very small number of patients, and lack of an appropriate animal model. In this study, we developed gut–kidney axis (GKA) on chip for co-culturing gut (Caco-2) and kidney (HKC-8) cells, and observed both STEC O157:H7 (O157) infection and Stx intoxication in the gut and kidney cells on the chip, respectively. Without any antibiotic treatment, O157 killed both gut and kidney cells in GKA on the chip. CIP treatment reduced O157 infection in the gut cells, but increased Stx2-induced damage in the kidney cells, whereas the gentamycin treatment reduced both O157 infection in the gut cells and Stx2-induced damage in the kidney cells. This is the first report to recapitulate a clinically relevant situation, i.e., that CIP treatment causes more damage than gentamicin treatment. These results suggest that GKA on chip is very useful for simultaneous observation of O157 infections and Stx2 poisoning in gut and kidney cells, making it suitable for studying the effects of antibiotics on the risk of HUS.
Collapse
Affiliation(s)
- Yugyeong Lee
- Department of Biomedical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Korea;
| | - Min-Hyeok Kim
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Korea; (M.-H.K.); (D.R.A.)
- Department of Chemical Engineering, Hongik University, Seoul 04066, Korea
| | - David Rodrigues Alves
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Korea; (M.-H.K.); (D.R.A.)
- Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, 1362035 Lisboa, Portugal
| | - Sejoong Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea;
| | - Luke P. Lee
- Institute of Quantum Biophysics (IQB), Department of Biophysics, Sungkyunkwan University (SKKU), Suwon 16419, Korea;
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jong Hwan Sung
- Department of Chemical Engineering, Hongik University, Seoul 04066, Korea
- Correspondence: (J.H.S.); (S.P.); Tel.: +82-2-320-3067 (J.H.S.); +82-31-290-7431 (S.P.)
| | - Sungsu Park
- Department of Biomedical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Korea;
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Korea; (M.-H.K.); (D.R.A.)
- Institute of Quantum Biophysics (IQB), Department of Biophysics, Sungkyunkwan University (SKKU), Suwon 16419, Korea;
- Correspondence: (J.H.S.); (S.P.); Tel.: +82-2-320-3067 (J.H.S.); +82-31-290-7431 (S.P.)
| |
Collapse
|
44
|
Singh AV, Maharjan RS, Kromer C, Laux P, Luch A, Vats T, Chandrasekar V, Dakua SP, Park BW. Advances in Smoking Related In Vitro Inhalation Toxicology: A Perspective Case of Challenges and Opportunities from Progresses in Lung-on-Chip Technologies. Chem Res Toxicol 2021; 34:1984-2002. [PMID: 34397218 DOI: 10.1021/acs.chemrestox.1c00219] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The inhalation toxicology of multifaceted particulate matter from the environment, cigarette smoke, and e-cigarette liquid vapes is a major research topic concerning the adverse effect of these items on lung tissue. In vitro air-liquid interface (ALI) culture models hold more potential in an inhalation toxicity assessment. Apropos to e-cigarette toxicity, the multiflavor components of the vapes pose a complex experimental bottleneck. While an appropriate ALI setup has been one part of the focus to overcome this, parallel attention towards the development of an ideal exposure system has pushed the field forward. With the advent of microfluidic devices, lung-on-chip (LOC) technologies show enormous opportunities in in vitro smoke-related inhalation toxicity. In this review, we provide a framework, establish a paradigm about smoke-related inhalation toxicity testing in vitro, and give a brief overview of breathing LOC experimental design concepts. The capabilities with optimized bioengineering approaches and microfluidics and their fundamental pros and cons are presented with specific case studies. The LOC model can imitate the structural, functional, and mechanical properties of human alveolar-capillary interface and are more reliable than conventional in vitro models. Finally, we outline current perspective challenges as well as opportunities of future development to smoking lungs-on-chip technologies based on advances in soft robotics, machine learning, and bioengineering.
Collapse
Affiliation(s)
- Ajay Vikram Singh
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Strasse 8-10, Berlin 10589, Germany
| | - Romi Singh Maharjan
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Strasse 8-10, Berlin 10589, Germany
| | - Charlotte Kromer
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Strasse 8-10, Berlin 10589, Germany
| | - Peter Laux
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Strasse 8-10, Berlin 10589, Germany
| | - Andreas Luch
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Strasse 8-10, Berlin 10589, Germany
| | - Tanusri Vats
- KNIPSS Management Institute, Faridipur Campus, NH 96, Faizabad-Allahabad Road, Sultanpur 228119, Uttar Pradesh, India
| | | | | | - Byung-Wook Park
- Department of Chemical Engineering, Rayen School of Engineering, Youngstown State University, Youngstown 44555, Ohio, United States
| |
Collapse
|
45
|
Si L, Bai H, Rodas M, Cao W, Oh CY, Jiang A, Moller R, Hoagland D, Oishi K, Horiuchi S, Uhl S, Blanco-Melo D, Albrecht RA, Liu WC, Jordan T, Nilsson-Payant BE, Golynker I, Frere J, Logue J, Haupt R, McGrath M, Weston S, Zhang T, Plebani R, Soong M, Nurani A, Kim SM, Zhu DY, Benam KH, Goyal G, Gilpin SE, Prantil-Baun R, Gygi SP, Powers RK, Carlson KE, Frieman M, tenOever BR, Ingber DE. A human-airway-on-a-chip for the rapid identification of candidate antiviral therapeutics and prophylactics. Nat Biomed Eng 2021; 5:815-829. [PMID: 33941899 PMCID: PMC8387338 DOI: 10.1038/s41551-021-00718-9] [Citation(s) in RCA: 235] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 03/19/2021] [Indexed: 02/05/2023]
Abstract
The rapid repurposing of antivirals is particularly pressing during pandemics. However, rapid assays for assessing candidate drugs typically involve in vitro screens and cell lines that do not recapitulate human physiology at the tissue and organ levels. Here we show that a microfluidic bronchial-airway-on-a-chip lined by highly differentiated human bronchial-airway epithelium and pulmonary endothelium can model viral infection, strain-dependent virulence, cytokine production and the recruitment of circulating immune cells. In airway chips infected with influenza A, the co-administration of nafamostat with oseltamivir doubled the treatment-time window for oseltamivir. In chips infected with pseudotyped severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), clinically relevant doses of the antimalarial drug amodiaquine inhibited infection but clinical doses of hydroxychloroquine and other antiviral drugs that inhibit the entry of pseudotyped SARS-CoV-2 in cell lines under static conditions did not. We also show that amodiaquine showed substantial prophylactic and therapeutic activities in hamsters challenged with native SARS-CoV-2. The human airway-on-a-chip may accelerate the identification of therapeutics and prophylactics with repurposing potential.
Collapse
Affiliation(s)
- Longlong Si
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Haiqing Bai
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Melissa Rodas
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Wuji Cao
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Crystal Yuri Oh
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Amanda Jiang
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Rasmus Moller
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daisy Hoagland
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kohei Oishi
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shu Horiuchi
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Skyler Uhl
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniel Blanco-Melo
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Randy A Albrecht
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Wen-Chun Liu
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tristan Jordan
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Ilona Golynker
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Justin Frere
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - James Logue
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Robert Haupt
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Marisa McGrath
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Stuart Weston
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Tian Zhang
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Roberto Plebani
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Center on Advanced Studies and Technology (CAST), Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Mercy Soong
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Atiq Nurani
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Seong Min Kim
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Danni Y Zhu
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Kambez H Benam
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Girija Goyal
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Sarah E Gilpin
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Rachelle Prantil-Baun
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Rani K Powers
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Kenneth E Carlson
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Matthew Frieman
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Benjamin R tenOever
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA.
- Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, MA, USA.
| |
Collapse
|
46
|
Joseph X, Akhil V, Arathi A, Mohanan PV. Comprehensive Development in Organ-On-A-Chip Technology. J Pharm Sci 2021; 111:18-31. [PMID: 34324944 DOI: 10.1016/j.xphs.2021.07.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/21/2021] [Accepted: 07/21/2021] [Indexed: 12/19/2022]
Abstract
The expeditious advancement in the organ on chip technology provided a phase change to the conventional in vitro tests used to evaluate absorption, distribution, metabolism, excretion (ADME) studies and toxicity assessments. The demand for an accurate predictive model for assessing toxicity and reducing the potential risk factors became the prime area of any drug delivery process. Researchers around the globe are welcoming the incorporation of organ-on-a-chips for ADME and toxicity evaluation. Organ-on-a-chip (OOC) is an interdisciplinary technology that evolved as a contemporary in vitro model for the pharmacokinetics and pharmacodynamics (PK-PD) studies of a proposed drug candidate in the pre-clinical phases of drug development. The OOC provides a platform that mimics the physiological functions occurring in the human body. The precise flow control systems and the rapid sample processing makes OOC more advanced than the conventional two-dimensional (2D) culture systems. The integration of various organs as in the multi organs-on-a-chip provides more significant ideas about the time and dose dependant effects occurring in the body when a new drug molecule is administered as part of the pre-clinical times. This review outlines the comprehensive development in the organ-on-a-chip technology, various OOC models and its drug development applications, toxicity evaluation and efficacy studies.
Collapse
Affiliation(s)
- X Joseph
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum 695012, Kerala, India
| | - V Akhil
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum 695012, Kerala, India
| | - A Arathi
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum 695012, Kerala, India
| | - P V Mohanan
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum 695012, Kerala, India.
| |
Collapse
|
47
|
Vichare AS, Kamath SU, Leist M, Hayes AW, Mahadevan B. Application of the 3Rs principles in the development of pharmaceutical generics. Regul Toxicol Pharmacol 2021; 125:105016. [PMID: 34302895 DOI: 10.1016/j.yrtph.2021.105016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 07/06/2021] [Accepted: 07/19/2021] [Indexed: 10/20/2022]
Abstract
Although the 3Rs are broadly applied in nonclinical testing, a better appreciation of the 3Rs is needed in the field of differentiated or value-added pharmaceutical generics because the minor changes in formulation, dosage form, indication, and application route often do not require additional safety testing. The US FDA and the EU EMA have comprehensive regulations for such drugs based on quality, therapeutic equivalence, and safety guidelines. However, no scientific publications on how the concept of replacement and reduction from 3Rs principles can be applied in the safety assessment of differentiated generics were found in the public domain. In this review, we discuss the application of 3Rs in nonclinical testing requirements for differentiated generics. Practical examples are provided in the form of case studies from regulated markets. We highlight the need for utilization of existing data to establish equivalence (differentiated generic vs innovator) in efficacy and safety. The case studies indicate that data requirements from animal experiments have been reduced to a large extent in some major markets without compromising quality and safety. In this context, we also highlight the problem that on a global scale, a true reduction of animal experiments will only be achieved when all countries adopt similar practices.
Collapse
Affiliation(s)
- Abhijit S Vichare
- Global Preclinical & Product Safety, Abbott Healthcare Pvt Ltd., Mumbai, India.
| | - Sushant U Kamath
- Global Preclinical & Product Safety, Abbott Healthcare Pvt Ltd., Mumbai, India
| | - Marcel Leist
- In vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | - A Wallace Hayes
- The University of South Florida, College of Public Health, Tampa, FL, USA
| | | |
Collapse
|
48
|
Monckton CP, Brown GE, Khetani SR. Latest impact of engineered human liver platforms on drug development. APL Bioeng 2021; 5:031506. [PMID: 34286173 PMCID: PMC8286174 DOI: 10.1063/5.0051765] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 06/21/2021] [Indexed: 01/07/2023] Open
Abstract
Drug-induced liver injury (DILI) is a leading cause of drug attrition, which is partly due to differences between preclinical animals and humans in metabolic pathways. Therefore, in vitro human liver models are utilized in biopharmaceutical practice to mitigate DILI risk and assess related mechanisms of drug transport and metabolism. However, liver cells lose phenotypic functions within 1–3 days in two-dimensional monocultures on collagen-coated polystyrene/glass, which precludes their use to model the chronic effects of drugs and disease stimuli. To mitigate such a limitation, bioengineers have adapted tools from the semiconductor industry and additive manufacturing to precisely control the microenvironment of liver cells. Such tools have led to the fabrication of advanced two-dimensional and three-dimensional human liver platforms for different throughput needs and assay endpoints (e.g., micropatterned cocultures, spheroids, organoids, bioprinted tissues, and microfluidic devices); such platforms have significantly enhanced liver functions closer to physiologic levels and improved functional lifetime to >4 weeks, which has translated to higher sensitivity for predicting drug outcomes and enabling modeling of diseased phenotypes for novel drug discovery. Here, we focus on commercialized engineered liver platforms and case studies from the biopharmaceutical industry showcasing their impact on drug development. We also discuss emerging multi-organ microfluidic devices containing a liver compartment that allow modeling of inter-tissue crosstalk following drug exposure. Finally, we end with key requirements for engineered liver platforms to become routine fixtures in the biopharmaceutical industry toward reducing animal usage and providing patients with safe and efficacious drugs with unprecedented speed and reduced cost.
Collapse
Affiliation(s)
- Chase P Monckton
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| | - Grace E Brown
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| | - Salman R Khetani
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| |
Collapse
|
49
|
Hosic S, Bindas AJ, Puzan ML, Lake W, Soucy JR, Zhou F, Koppes RA, Breault DT, Murthy SK, Koppes AN. Rapid Prototyping of Multilayer Microphysiological Systems. ACS Biomater Sci Eng 2021; 7:2949-2963. [PMID: 34275297 PMCID: PMC8290094 DOI: 10.1021/acsbiomaterials.0c00190] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Microfluidic organs-on-chips aim to realize more biorelevant in vitro experiments compared to traditional two-dimensional (2D) static cell culture. Often such devices are fabricated via poly(dimethylsiloxane) (PDMS) soft lithography, which offers benefits (e.g., high feature resolution) along with drawbacks (e.g., prototyping time/costs). Here, we report benchtop fabrication of multilayer, PDMS-free, thermoplastic organs-on-chips via laser cut and assembly with double-sided adhesives that overcome some limitations of traditional PDMS lithography. Cut and assembled chips are economical to prototype ($2 per chip), can be fabricated in parallel within hours, and are Luer compatible. Biocompatibility was demonstrated with epithelial line Caco-2 cells and primary human small intestinal organoids. Comparable to control static Transwell cultures, Caco-2 and organoids cultured on chips formed confluent monolayers expressing tight junctions with low permeability. Caco-2 cells-on-chip differentiated ∼4 times faster, including increased mucus, compared to controls. To demonstrate the robustness of cut and assemble, we fabricated a dual membrane, trilayer chip integrating 2D and 3D compartments with accessible apical and basolateral flow chambers. As proof of concept, we cocultured a human, differentiated monolayer and intact 3D organoids within multilayered contacting compartments. The epithelium exhibited 3D tissue structure and organoids expanded close to the adjacent monolayer, retaining proliferative stem cells over 10 days. Taken together, cut and assemble offers the capability to rapidly and economically manufacture microfluidic devices, thereby presenting a compelling fabrication technique for developing organs-on-chips of various geometries to study multicellular tissues.
Collapse
Affiliation(s)
- Sanjin Hosic
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., 313 Snell Engineering, Boston, Massachusetts 02115, United States
| | - Adam J Bindas
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., 313 Snell Engineering, Boston, Massachusetts 02115, United States
| | - Marissa L Puzan
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., 313 Snell Engineering, Boston, Massachusetts 02115, United States
| | - Will Lake
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., 313 Snell Engineering, Boston, Massachusetts 02115, United States
| | - Jonathan R Soucy
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., 313 Snell Engineering, Boston, Massachusetts 02115, United States
| | - Fanny Zhou
- Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, United States
| | - Ryan A Koppes
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., 313 Snell Engineering, Boston, Massachusetts 02115, United States
| | - David T Breault
- Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, United States
- Department of Pediatrics, Harvard Medical School, 300 Longwood Avenue, Boston, Massachusetts 02115, United States
- Principal Faculty, Harvard Stem Cell Institute, 7 Divinity Ave, Cambridge, Massachusetts 02138, United States
| | - Shashi K Murthy
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., 313 Snell Engineering, Boston, Massachusetts 02115, United States
| | - Abigail N Koppes
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., 313 Snell Engineering, Boston, Massachusetts 02115, United States
- Department of Biology, Northeastern University, 360 Huntington Ave., 313 Snell Engineering, Boston, Massachusetts 02115, United States
| |
Collapse
|
50
|
Ahmed A, Joshi IM, Mansouri M, Ahamed NNN, Hsu MC, Gaborski TR, Abhyankar VV. Engineering fiber anisotropy within natural collagen hydrogels. Am J Physiol Cell Physiol 2021; 320:C1112-C1124. [PMID: 33852366 PMCID: PMC8285641 DOI: 10.1152/ajpcell.00036.2021] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 04/06/2021] [Accepted: 04/06/2021] [Indexed: 12/14/2022]
Abstract
It is well known that biophysical properties of the extracellular matrix (ECM), including stiffness, porosity, composition, and fiber alignment (anisotropy), play a crucial role in controlling cell behavior in vivo. Type I collagen (collagen I) is a ubiquitous structural component in the ECM and has become a popular hydrogel material that can be tuned to replicate the mechanical properties found in vivo. In this review article, we describe popular methods to create 2-D and 3-D collagen I hydrogels with anisotropic fiber architectures. We focus on methods that can be readily translated from engineering and materials science laboratories to the life-science community with the overall goal of helping to increase the physiological relevance of cell culture assays.
Collapse
Affiliation(s)
- Adeel Ahmed
- Department of Microsystems Engineering, Rochester Institute of Technology, Rochester, New York
| | - Indranil M Joshi
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, New York
| | - Mehran Mansouri
- Department of Microsystems Engineering, Rochester Institute of Technology, Rochester, New York
| | - Nuzhet N N Ahamed
- Department of Microsystems Engineering, Rochester Institute of Technology, Rochester, New York
| | - Meng-Chun Hsu
- Department of Microsystems Engineering, Rochester Institute of Technology, Rochester, New York
| | - Thomas R Gaborski
- Department of Microsystems Engineering, Rochester Institute of Technology, Rochester, New York
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, New York
| | - Vinay V Abhyankar
- Department of Microsystems Engineering, Rochester Institute of Technology, Rochester, New York
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, New York
| |
Collapse
|