1
|
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
|
2
|
Samantasinghar A, Sunildutt N, Ahmed F, Memon FH, Kang C, Choi KH. Revolutionizing Biomedical Research: Unveiling the Power of Microphysiological Systems with Advanced Assays, Integrated Sensor Technologies, and Real-Time Monitoring. ACS OMEGA 2025; 10:9869-9889. [PMID: 40124012 PMCID: PMC11923667 DOI: 10.1021/acsomega.4c11227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 03/25/2025]
Abstract
The limitation of animal models to imitate a therapeutic response in humans is a key problem that challenges their use in fundamental research. Organ-on-a-chip (OOC) devices, also called microphysiological systems (MPS), are devices containing a lining of living cells grown under dynamic flow to recapitulate the important features of human physiology and pathophysiology with high precision. Recent advances in microfabrication and tissue engineering techniques have led to the wide adoption of OOC in next-generation experimental platforms. This review presents a comprehensive analysis of the OOC systems, categorizing them by flow types (single-pass and multipass), operational mechanisms (pumpless and pump-driven), and configurations (single-organ and multiorgan systems), along with their respective advantages and limitations. Furthermore, it explores the integration of qualitative and quantitative assay techniques, providing a comparative evaluation of systems with and without sensor integration. This review aims to fill essential knowledge gaps, driving the progress of the development of OOC systems and paving the way for breakthroughs in biomedical research, pharmaceutical innovation, and tissue engineering.
Collapse
Affiliation(s)
- Anupama Samantasinghar
- Department
of Mechatronics Engineering, Jeju National
University, Jeju 63243, Republic
of Korea
| | - Naina Sunildutt
- Department
of Mechatronics Engineering, Jeju National
University, Jeju 63243, Republic
of Korea
| | - Faheem Ahmed
- Department
of Mechatronics Engineering, Jeju National
University, Jeju 63243, Republic
of Korea
| | - Fida Hussain Memon
- Department
of Mechatronics Engineering, Jeju National
University, Jeju 63243, Republic
of Korea
- Department
of Electrical Engineering, Sukkur IBA University, Sindh 65200, Pakistan
| | - Chulung Kang
- Department
of Mechatronics Engineering, Jeju National
University, Jeju 63243, Republic
of Korea
| | - Kyung Hyun Choi
- Department
of Mechatronics Engineering, Jeju National
University, Jeju 63243, Republic
of Korea
| |
Collapse
|
3
|
Brandauer K, Schweinitzer S, Lorenz A, Krauß J, Schobesberger S, Frauenlob M, Ertl P. Advances of dual-organ and multi-organ systems for gut, lung, skin and liver models in absorption and metabolism studies. LAB ON A CHIP 2025; 25:1384-1403. [PMID: 39973270 DOI: 10.1039/d4lc01011f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Drug development is a costly and timely process with high risks of failure during clinical trials. Although in vitro tissue models have significantly advanced over the years, thus fostering a transition from animal-derived models towards human-derived models, failure rates still remain high. Current cell-based assays are still not able to provide an accurate prediction of the clinical success or failure of a drug candidate. To overcome the limitations of current methods, a variety of microfluidic systems have been developed as powerful tools that are capable of mimicking (micro)physiological conditions more closely by integrating physiological fluid flow conditions, mechanobiological cues and concentration gradients, to name only a few. One major advantage of these biochip-based tissue cultures, however, is their ability to seamlessly connect different organ models, thereby allowing the study of organ-crosstalk and metabolic byproduct effects. This is especially important when assessing absorption, distribution, metabolism, and excretion (ADME) processes of drug candidates, where an interplay between various organs is a prerequisite. In the current review, a number of in vitro models as well as microfluidic dual- and multi-organ systems are summarized with a focus on absorption (skin, lung, gut) and metabolism (liver). Additionally, the advantage of multi-organ chips in identifying a drug's on and off-target toxicity is discussed. Finally, the potential high-throughput implementation and modular chip design of multi-organ-on-a-chip systems within the pharmaceutical industry is highlighted, outlining the necessity of reducing handling complexity.
Collapse
Affiliation(s)
- Konstanze Brandauer
- Faculty of Technical Chemistry, TU Wien, Getreidemarkt 9, 1060 Vienna, Austria.
| | - Sophie Schweinitzer
- Faculty of Technical Chemistry, TU Wien, Getreidemarkt 9, 1060 Vienna, Austria.
| | - Alexandra Lorenz
- Faculty of Technical Chemistry, TU Wien, Getreidemarkt 9, 1060 Vienna, Austria.
| | - Judith Krauß
- Faculty of Technical Chemistry, TU Wien, Getreidemarkt 9, 1060 Vienna, Austria.
| | | | - Martin Frauenlob
- Faculty of Technical Chemistry, TU Wien, Getreidemarkt 9, 1060 Vienna, Austria.
| | - Peter Ertl
- Faculty of Technical Chemistry, TU Wien, Getreidemarkt 9, 1060 Vienna, Austria.
| |
Collapse
|
4
|
Mallya D, Gadre MA, Varadharajan S, Vasanthan KS. 3D bioprinting for the construction of drug testing models-development strategies and regulatory concerns. Front Bioeng Biotechnol 2025; 13:1457872. [PMID: 40028291 PMCID: PMC11868281 DOI: 10.3389/fbioe.2025.1457872] [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: 07/01/2024] [Accepted: 01/14/2025] [Indexed: 03/05/2025] Open
Abstract
A drug to be successfully launched in the market requires a significant amount of capital, resources and time, where the unsuccessful results in the last stages lead to catastrophic failure for discovering drugs. This is the very reason which calls for the invention of innovative models that can closely mimic the human in vivo model for producing reliable results. Throughout the innovation line, there has been improvement in the rationale in silico designing but yet there is requirement for in vitro-in vivo correlations. During the evolving of the drug testing models, the 3D models produced by different methods have been proven to produce better results than the traditional 2D models. However, the in vitro fabrications of live tissues are still bottleneck in realizing their complete potential. There is an urgent need for the development of single, standard and simplified in vitro 3D tissue models that can be reliable for investigating the biological and pathological aspects of drug discovery, which is yet to be achieved. The existing pre-clinical models have considerable drawbacks despite being the gold standard in pre-clinical research. The major drawback being the interspecies differences and low reliability on the generated results. This gap could be overcome by the fabrication of bioengineered human disease models for drug screening. The advancement in the fabrication of 3D models will provide a valuable tool in screening drugs at different stages as they are one step closer to bio-mimic human tissues. In this review, we have discussed on the evolution of preclinical studies, and different models, including mini tissues, spheroids, organoids, bioengineered three dimensional models and organs on chips. Furthermore, we provide details of different disease models fabricated across various organs and their applications. In addition to this, the review also focuses on the limitations and the current prospects of the role of three dimensionally bioprinted models in drug screening and development.
Collapse
Affiliation(s)
- Divya Mallya
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Mrunmayi Ashish Gadre
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - S. Varadharajan
- Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Kirthanashri S. Vasanthan
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
5
|
Graf K, Murrieta-Coxca JM, Vogt T, Besser S, Geilen D, Kaden T, Bothe AK, Morales-Prieto DM, Amiri B, Schaller S, Kaufmann L, Raasch M, Ammar RM, Maass C. Digital twin-enhanced three-organ microphysiological system for studying drug pharmacokinetics in pregnant women. Front Pharmacol 2025; 16:1528748. [PMID: 40034823 PMCID: PMC11873563 DOI: 10.3389/fphar.2025.1528748] [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: 11/15/2024] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
Background Pregnant women represent a vulnerable group in pharmaceutical research due to limited knowledge about drug metabolism and safety of commonly used corticosteroids like prednisone due to ethical and practical constraints. Current preclinical models, including animal studies, fail to accurately replicate human pregnancy conditions, resulting in gaps in drug safety and pharmacokinetics predictions. To address this issue, we used a three-organ microphysiological system (MPS) combined with a digital twin framework, to predict pharmacokinetics and fetal drug exposure. Methods The here shown human MPS integrated gut, liver, and placenta models, interconnected via the corresponding vasculature. Using prednisone as a model compound, we simulate oral drug administration and track its metabolism and transplacental transfer. To translate the generated data from MPS to human physiology, computational modelling techniques were developed. Results Our results demonstrate that the system maintains cellular integrity and accurately mimics in vivo drug dynamics, with predictions closely matching clinical data from pregnant women. Digital twinning closely aligned with the generated experimental data. Long-term exposure simulations confirmed the value of this integrated system for predicting the non-toxic metabolization of prednisone. Conclusion This approach may provide a potential non-animal alternative that could contribute to our understanding of drug behavior during pregnancy and may support early-stage drug safety assessment for vulnerable populations.
Collapse
Affiliation(s)
| | | | | | | | | | - Tim Kaden
- Dynamic42 GmbH, Jena, Germany
- Institute of Biochemistry II, Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | | | | | - Behnam Amiri
- MPSlabs, ESQlabs GmbH, Saterland, Germany
- ESQlabs GmbH, Saterland, Germany
| | | | - Ligaya Kaufmann
- Global Medical Affairs, Bayer Consumer Care AG, Basel, Switzerland
| | | | - Ramy M. Ammar
- Global R&D, Bayer Consumer Health, Steigerwald Arzneimittelwerk GmbH, Darmstadt, Germany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafr-El Sheikh, Egypt
| | - Christian Maass
- MPSlabs, ESQlabs GmbH, Saterland, Germany
- ESQlabs GmbH, Saterland, Germany
| |
Collapse
|
6
|
Parekh P, Sherfey J, Alaybeyoglu B, Cirit M. Pathway-Based Similarity Measurement to Quantify Transcriptomics Similarity Between Human Tissues and Preclinical Models. Clin Pharmacol Ther 2025; 117:485-494. [PMID: 39377352 PMCID: PMC11747893 DOI: 10.1002/cpt.3465] [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: 04/10/2024] [Accepted: 09/14/2024] [Indexed: 10/09/2024]
Abstract
Accurate clinical translation of preclinical research remains challenging, primarily due to species-specific differences and disease and patient heterogeneity. An important recent advancement has been development of microphysiological systems that consist of multiple human cell types that recapitulate key characteristics of their respective human systems, allowing essential physiologic processes to be accurately assessed during drug development. However, an unmet need remains regarding a quantitative method to evaluate the similarity between diverse sample types for various contexts of use (CoU)-specific pathways. To address this gap, this study describes the development of pathway-based similarity measurement (PBSM), which leverages RNA-seq data and pathway-based information to assess the human relevance of preclinical models for specific CoU. PBSM offers a quantitative method to compare the transcriptomic similarity of preclinical models to human tissues, shown here as proof of concept for liver and cardiac tissues, enabling improved model selection and validation. Thus, PBSM can successfully support CoU selection for preclinical models, assess the impact of different gene sets on similarity calculations, and differentiate among various in vitro and in vivo models. PBSM has the potential to reduce the translational gap in drug development by allowing quantitative evaluation of the similarity of preclinical models to human tissues, facilitating model selection, and improving understanding of context-specific applications. PBSM can serve as a foundation for enhancing the physiological relevance of in vitro models and supporting the development of more effective therapeutic interventions.
Collapse
Affiliation(s)
- Paarth Parekh
- Javelin Biotech, Inc., 299 Washington Street, Woburn, Massachusetts 01801, USA
| | - Jason Sherfey
- Javelin Biotech, Inc., 299 Washington Street, Woburn, Massachusetts 01801, USA
| | - Begum Alaybeyoglu
- Javelin Biotech, Inc., 299 Washington Street, Woburn, Massachusetts 01801, USA
| | - Murat Cirit
- Javelin Biotech, Inc., 299 Washington Street, Woburn, Massachusetts 01801, USA
| |
Collapse
|
7
|
Kim Y, Kang M, Mamo MG, Adisasmita M, Huch M, Choi D. Liver organoids: Current advances and future applications for hepatology. Clin Mol Hepatol 2025; 31:S327-S348. [PMID: 39722609 PMCID: PMC11925438 DOI: 10.3350/cmh.2024.1040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 12/24/2024] [Indexed: 12/28/2024] Open
Abstract
The creation of self-organizing liver organoids represents a significant, although modest, step toward addressing the ongoing organ shortage crisis in allogeneic liver transplantation. However, researchers have recognized that achieving a fully functional whole liver remains a distant goal, and the original ambition of organoid-based liver generation has been temporarily put on hold. Instead, liver organoids have revolutionized the field of hepatology, extending their influence into various domains of precision and molecular medicine. These 3D cultures, capable of replicating key features of human liver function and pathology, have opened new avenues for human-relevant disease modeling, CRISPR gene editing, and high-throughput drug screening that animal models cannot accomplish. Moreover, advancements in creating more complex systems have led to the development of multicellular assembloids, dynamic organoid-on-chip systems, and 3D bioprinting technologies. These innovations enable detailed modeling of liver microenvironments and complex tissue interactions. Progress in regenerative medicine and transplantation applications continues to evolve and strives to overcome the obstacles of biocompatibility and tumorigenecity. In this review, we examine the current state of liver organoid research by offering insights into where the field currently stands, and the pivotal developments that are shaping its future.
Collapse
Affiliation(s)
- Yohan Kim
- Department of MetaBioHealth, Sungkyunkwan University, Suwon, Korea
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon, Korea
- Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon, Korea
| | - Minseok Kang
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
| | - Michael Girma Mamo
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
- Research Institute of Regenerative Medicine and Stem Cells, Hanyang University, Seoul, Korea
| | - Michael Adisasmita
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
- Research Institute of Regenerative Medicine and Stem Cells, Hanyang University, Seoul, Korea
| | - Meritxell Huch
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Dongho Choi
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
- Research Institute of Regenerative Medicine and Stem Cells, Hanyang University, Seoul, Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Korea
- Department of HY-KIST Bio-convergence, Hanyang University, Seoul, Korea
| |
Collapse
|
8
|
Cook SR, Ball AG, Mohammad A, Pompano RR. A 3D-printed multi-compartment organ-on-chip platform with a tubing-free pump models communication with the lymph node. LAB ON A CHIP 2025; 25:155-174. [PMID: 39661075 PMCID: PMC11633827 DOI: 10.1039/d4lc00489b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 11/28/2024] [Indexed: 12/12/2024]
Abstract
Multi-organ-on-chip systems (MOOCs) have the potential to mimic communication between organ systems and reveal mechanisms of health and disease. However, many existing MOOCs are challenging for non-experts to implement due to complex tubing, electronics, or pump mechanisms. In addition, few MOOCs have incorporated immune organs such as the lymph node (LN), limiting their applicability to model critical events such as vaccination. Here we developed a 3D-printed, user-friendly device and companion tubing-free impeller pump with the capacity to co-culture two or more tissue samples, including a LN, under a recirculating common media. Native tissue structure and immune function were incorporated by maintaining slices of murine LN tissue ex vivo in 3D-printed mesh supports for at least 24 h. In a two-compartment model of a LN and an upstream injection site in mock tissue, vaccination of the multi-compartment chip was similar to in vivo vaccination in terms of locations of antigen accumulation and acute changes in activation markers and gene expression in the LN. We anticipate that in the future, this flexible platform will enable models of multi-organ immune responses throughout the body.
Collapse
Affiliation(s)
- Sophie R Cook
- Department of Chemistry, University of Virginia, Charlottesville, VA, USA.
| | - Alexander G Ball
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
- Carter Immunology Center and UVA Comprehensive Cancer Center, University of Virginia, Charlottesville, VA, USA
| | | | - Rebecca R Pompano
- Department of Chemistry, University of Virginia, Charlottesville, VA, USA.
- Carter Immunology Center and UVA Comprehensive Cancer Center, University of Virginia, Charlottesville, VA, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
9
|
Maulana TI, Wevers NR, Kristoforus T, Chandler M, Lanz HL, Joore J, Vulto P, Villenave R, Kustermann S, Loskill P, Bircsak KM. Opportunities for Microphysiological Systems in Toxicity Testing of New Drug Modalities. Annu Rev Pharmacol Toxicol 2025; 65:47-69. [PMID: 39227343 DOI: 10.1146/annurev-pharmtox-061724-080621] [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: 09/05/2024]
Abstract
New drug modalities offer life-saving benefits for patients through access to previously undruggable targets. Yet these modalities pose a challenge for the pharmaceutical industry, as side effects are complex, unpredictable, and often uniquely human. With animal studies having limited predictive value due to translatability challenges, the pharmaceutical industry seeks out new approach methodologies. Microphysiological systems (MPS) offer important features that enable complex toxicological processes to be modeled in vitro such as (a) an adjustable complexity of cellular components, including immune components; (b) a modifiable tissue architecture; (c) integration and monitoring of dynamic mechanisms; and (d) a multiorgan connection. Here we review MPS studies in the context of four clinical adverse events triggered by new drug modalities: peripheral neuropathy, thrombocytopenia, immune-mediated hepatotoxicity, and cytokine release syndrome. We conclude that while the use of MPS for testing new drug modality-induced toxicities is still in its infancy, we see strong potential going forward.
Collapse
Affiliation(s)
- Tengku Ibrahim Maulana
- Department for Microphysiological Systems, Institute of Biomedical Engineering, Faculty of Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | | | - Theodora Kristoforus
- Department for Microphysiological Systems, Institute of Biomedical Engineering, Faculty of Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
| | | | | | - Jos Joore
- MIMETAS BV, Oegstgeest, The Netherlands
| | | | - Remi Villenave
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Stefan Kustermann
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Peter Loskill
- Department for Microphysiological Systems, Institute of Biomedical Engineering, Faculty of Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | | |
Collapse
|
10
|
Ohri S, Parekh P, Nichols L, Rajan SAP, Cirit M. Utilization of a human Liver Tissue Chip for drug-metabolizing enzyme induction studies of perpetrator and victim drugs. Drug Metab Dispos 2025; 53:100004. [PMID: 39884808 DOI: 10.1124/dmd.124.001497] [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: 07/17/2024] [Revised: 09/30/2024] [Accepted: 09/30/2024] [Indexed: 10/09/2024] Open
Abstract
Polypharmacy-related drug-drug interactions (DDIs) are a significant and growing healthcare concern. An increasing number of therapeutic drugs on the market underscores the necessity to accurately assess new drug combinations during preclinical evaluation for DDIs. In vitro primary human hepatocytes (PHH) models are only applicable for short-term induction studies because of their rapid loss of metabolic function. Though coculturing nonhuman stromal cells with PHH has been shown to stabilize metabolic activity long-term, there are concerns about human specificity for accurate clinical assessment. In this study, we demonstrated a PHH-only liver microphysiological system in the Liver Tissue Chip is capable of maintaining long-term functional and metabolic activity of PHH from 3 individual donors and thus a suitable platform for long-term DDI induction studies. The responses to rifampicin induction of 3 PHH donors were assessed using cytochrome P450 activity and mRNA changes. Additionally, victim pharmacokinetic studies were conducted with midazolam (high clearance) and alprazolam (low clearance) following perpetrator drug treatment, rifampicin-mediated induction, which resulted in a 2-fold and a 2.6-fold increase in midazolam and alprazolam intrinsic clearance values, respectively, compared with the untreated liver microphysiological system. We also investigated the induction effects of different dosing regimens of the perpetrator drug (rifampicin) on cytochrome P450 activity levels, showing minimal variation in the intrinsic clearance of the victim drug (midazolam). This study illustrates the utility of the Liver Tissue Chip for in vitro liver-specific DDI induction studies, providing a translational experimental system to predict clinical clearance values of both perpetrator and victim drugs. SIGNIFICANCE STATEMENT: This study demonstrated the utility of the Liver Tissue Chip with a primary human hepatocyte-only liver microphysiological system for drug-drug interaction induction studies. This unique in vitro system with continuous recirculation maintains long-term functionality and metabolic activity for up to 4 weeks, enabling the study of perpetrator and victim drug pharmacokinetics, quantification of drug-induced cytochrome P450 mRNA and activity levels, investigation of patient variability, and ultimately clinical predictions.
Collapse
Affiliation(s)
| | | | | | | | - Murat Cirit
- Javelin Biotech, Inc, Woburn, Massachusetts.
| |
Collapse
|
11
|
Nair DG, Weiskirchen R. Advanced In Vitro Models for Preclinical Drug Safety: Recent Progress and Prospects. Curr Issues Mol Biol 2024; 47:7. [PMID: 39852122 PMCID: PMC11763796 DOI: 10.3390/cimb47010007] [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: 11/13/2024] [Revised: 12/17/2024] [Accepted: 12/24/2024] [Indexed: 01/26/2025] Open
Abstract
The majority of drugs are typically orally administered. The journey from drug discovery to approval is often long and expensive, involving multiple stages. A major challenge in the drug development process is drug-induced liver injury (DILI), a condition that affects the liver, the organ responsible for metabolizing most drugs. Traditionally, identifying DILI risk has been difficult due to the poor correlation between preclinical animal models and in vitro systems. Differences in physiology between humans and animals or cell lines contribute to the failure of many drug programs during clinical trials. The use of advanced in vitro systems that closely mimic human physiology, such as organ-on-a-chip models like gut-liver-on-a-chip, can be crucial in improving drug efficacy while minimizing toxicity. Additionally, the adaptation of these technologies has the potential to significantly reduce both the time and cost associated with obtaining safe drug approvals, all while adhering to the 3Rs principle (replacement, reduction, refinement). In this review, we discuss the significance, current status, and future prospects of advanced platforms, specifically organ-on-a-chip models, in supporting preclinical drug discovery.
Collapse
Affiliation(s)
- Dileep G. Nair
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), University Hospital Aachen, D-52074 Aachen, Germany;
- Keliomics Inc., 4640 S Macadam Ave #270, Portland, OR 97239, USA
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), University Hospital Aachen, D-52074 Aachen, Germany;
| |
Collapse
|
12
|
Horiuchi S, Koda N, Ikeda Y, Tanaka Y, Masuo Y, Kato Y, Yamazaki D. Examination of common culture medium for human hepatocytes and engineered heart tissue: Towards an evaluation of cardiotoxicity associated with hepatic drug metabolism in vitro. PLoS One 2024; 19:e0315997. [PMID: 39715174 DOI: 10.1371/journal.pone.0315997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 12/04/2024] [Indexed: 12/25/2024] Open
Abstract
Cardiotoxicity associated with hepatic metabolism and drug-drug interactions is a serious concern. Predicting drug toxicity using animals remains challenging due to species and ethical concerns, necessitating the need to develop alternative approaches. Drug cardiotoxicity associated with hepatic metabolism cannot be detected using a cardiomyocyte-only evaluation system. Therefore, we aimed to establish a system for evaluating cardiotoxicity via hepatic metabolism by co-culturing cryopreserved human hepatocytes (cryoheps) and human iPS cell-derived engineered heart tissues (hiPSC-EHTs) using a stirrer-based microphysiological system. We investigated candidate media to identify a medium that can be used commonly for hepatocytes and cardiomyocytes. We found that the contraction length was significantly greater in the HM Dex (-) medium, the medium used for cryohep culture without dexamethasone, than that in the EHT medium used for hiPSC-EHT culture. Additionally, the beating rate, contraction length, contraction speed, and relaxation speed of hiPSC-EHT cultured in the HM Dex (-) medium were stable throughout the culture period. Among the major CYPs, the expression of CYP3A4 alone was low in cryoheps cultured in the HM Dex (-) medium. However, improved oxygenation using the InnoCell plate increased CYP3A4 expression to levels comparable to those found in the human liver. In addition, CYP3A4 activity was also increased by the improved oxygenation. Furthermore, expression levels of hepatic function-related gene and nuclear receptors in cryoheps cultured in HM Dex (-) medium were comparable to those in the human liver. These results suggest that the HM Dex (-) medium can be applied to co-culture and may allow the evaluation of cardiotoxicity via hepatic metabolism. Moreover, CYP induction by typical inducers was confirmed in cryoheps cultured in the HM Dex (-) medium, suggesting that drug-drug interactions could also be evaluated using this medium. Our findings may facilitate the evaluation of cardiotoxicity via hepatic metabolism, potentially reducing animal testing, lowering costs, and expediting drug development.
Collapse
Affiliation(s)
- Shinichiro Horiuchi
- Division of Pharmacology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Nanae Koda
- Division of Pharmacology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Yui Ikeda
- Faculty of Pharmacy, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Yuto Tanaka
- Faculty of Pharmacy, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Yusuke Masuo
- Faculty of Pharmacy, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Yukio Kato
- Faculty of Pharmacy, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Daiju Yamazaki
- Division of Pharmacology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| |
Collapse
|
13
|
Kimura H, Nishikawa M, Kutsuzawa N, Tokito F, Kobayashi T, Kurniawan DA, Shioda H, Cao W, Shinha K, Nakamura H, Doi K, Sakai Y. Advancements in Microphysiological systems: Exploring organoids and organ-on-a-chip technologies in drug development -focus on pharmacokinetics related organs. Drug Metab Pharmacokinet 2024; 60:101046. [PMID: 39847980 DOI: 10.1016/j.dmpk.2024.101046] [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: 09/07/2024] [Revised: 12/05/2024] [Accepted: 12/14/2024] [Indexed: 01/25/2025]
Abstract
This study explored the evolving landscape of Microphysiological Systems (MPS), with a focus on organoids and organ-on-a-chip (OoC) technologies, which are promising alternatives to animal testing in drug discovery. MPS technology offers in vitro models with high physiological relevance, simulating organ function for pharmacokinetic studies. Organoids composed of 3D cell aggregates and OoCs mimicking in vivo environments based on microfluidic platforms represent the forefront of MPS. This paper provides a comprehensive overview of their application in studying the gut, liver, and kidney and their challenges in becoming reliable alternatives to in vivo models. Although MPS technology is not yet fully comparable to in vivo systems, its continued development, aided by in silico, automation, and AI approaches, is anticipated to bring about further advancements. Collaboration across multiple disciplines and ongoing regulatory discussions will be crucial in driving MPS toward practical and ethical applications in biomedical research and drug development.
Collapse
Affiliation(s)
- Hiroshi Kimura
- Micro/Nano Technology Center, Tokai University, 4-1-1 Kitakaname, Hiratsuka, Kanagawa, 259-1292, Japan.
| | - Masaki Nishikawa
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo, 113-8654, Japan
| | - Naokata Kutsuzawa
- Micro/Nano Technology Center, Tokai University, 4-1-1 Kitakaname, Hiratsuka, Kanagawa, 259-1292, Japan; Division of Pulmonary Medicine, Department of Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1143, Japan
| | - Fumiya Tokito
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo, 113-8654, Japan
| | - Takuma Kobayashi
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo, 113-8654, Japan
| | - Dhimas Agung Kurniawan
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo, 113-8654, Japan
| | - Hiroki Shioda
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo, 113-8654, Japan
| | - Wenxin Cao
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo, 113-8654, Japan
| | - Kenta Shinha
- Micro/Nano Technology Center, Tokai University, 4-1-1 Kitakaname, Hiratsuka, Kanagawa, 259-1292, Japan
| | - Hiroko Nakamura
- Micro/Nano Technology Center, Tokai University, 4-1-1 Kitakaname, Hiratsuka, Kanagawa, 259-1292, Japan
| | - Kotaro Doi
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo, Tokyo, 113-8655, Japan
| | - Yasuyuki Sakai
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo, 113-8654, Japan
| |
Collapse
|
14
|
Iwao T, Matsunaga T. Development of intestinal organoids and microphysiological systems and their application to drug discovery. Drug Metab Pharmacokinet 2024; 60:101045. [PMID: 39847977 DOI: 10.1016/j.dmpk.2024.101045] [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: 08/29/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 01/25/2025]
Abstract
The intestines are an important organ with a variety of functions. For drug discovery research, experimental animals and Caco-2 cells derived from a human colon carcinoma may be used to evaluate the absorption and safety of orally administered drugs. These systems have issues, such as species differences with humans in experimental animals, variations in gene expression patterns, very low drug-metabolizing activities in Caco-2 cells, and the recent trend toward reduced animal testing. Thus, there is a need for new evaluation systems. Intestinal organoid technology and microphysiological systems (MPS) have attracted attention as novel evaluation systems for predicting drug disposition, safety, and efficacy in humans in vitro. Intestinal organoids are three-dimensional structures that contain a variety of intestinal cells. They also contain crypt-villus structures similar to those of living bodies. Using MPS, it is possible to improve the functionality of cells and evaluate the linkage and crosstalk between the intestine and the liver. These systems are expected to be powerful tools for drug discovery research to predict efficacy and toxicity in humans. This review outlines the current status of intestinal organoids and MPS studies.
Collapse
Affiliation(s)
- Takahiro Iwao
- Graduate School of Pharmaceutical Sciences, Nagoya City University, Japan.
| | - Tamihide Matsunaga
- Graduate School of Pharmaceutical Sciences, Nagoya City University, Japan.
| |
Collapse
|
15
|
Agustina-Hernández M, Francés-Herrero E, Gómez-Álvarez M, Alonso-Frías P, Romeu M, Monzó A, Ferrero H, Bueno-Fernandez C, Cervelló I. Biotechnological progresses in modelling the human endometrium: the evolution of current in vitro techniques and emerging trends. Front Bioeng Biotechnol 2024; 12:1495338. [PMID: 39698187 PMCID: PMC11653193 DOI: 10.3389/fbioe.2024.1495338] [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: 09/12/2024] [Accepted: 11/22/2024] [Indexed: 12/20/2024] Open
Abstract
The endometrium plays a fundamental role in the reproductive system yet many etiologies of infertility-related endometrial diseases such as endometriosis, adenomyosis, Asherman's syndrome or endometrial cancer remain unknown. There are currently no treatments that minimize the effects of this devastating disorder. Appropriate model systems that closely mimic the architecture and function of the endometrium in healthy and pathological states are needed to understand the underlying molecular pathways and develop novel or more effective treatments. This review summarizes the key milestones of in vitro culture models of the human endometrium throughout history, as well as the applications of advanced bioengineering techniques in the modelling of both healthy and pathological endometrium. Opportunities for future approaches are also discussed.
Collapse
Affiliation(s)
- Marcos Agustina-Hernández
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Emilio Francés-Herrero
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - María Gómez-Álvarez
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Paula Alonso-Frías
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Mónica Romeu
- Assisted Human Reproduction Unit, La Fe University and Polytechnic Hospital, Valencia, Spain
| | - Ana Monzó
- Assisted Human Reproduction Unit, La Fe University and Polytechnic Hospital, Valencia, Spain
| | - Hortensia Ferrero
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Clara Bueno-Fernandez
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
- Department of Pediatrics, Obstetrics and Gynecology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Irene Cervelló
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| |
Collapse
|
16
|
Byrne CE, Martier AT, Kpeli GW, Conrad KM, Bralower W, Olsen E, Fortes G, Culp CC, Wendell M, Boone KA, Burow MR, Mondrinos MJ. Adaptable Manufacturing and Biofabrication of Milliscale Organ Chips With Perfusable Vascular Beds. Biotechnol J 2024; 19:e202400550. [PMID: 39711093 DOI: 10.1002/biot.202400550] [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: 09/09/2024] [Revised: 11/09/2024] [Accepted: 12/04/2024] [Indexed: 12/24/2024]
Abstract
Microphysiological systems (MPS) containing perfusable vascular beds unlock the ability to model tissue-scale elements of vascular physiology and disease in vitro. Access to inexpensive stereolithography (SLA) 3D printers now enables benchtop fabrication of polydimethylsiloxane (PDMS) organ chips, eliminating the need for cleanroom access and microfabrication expertise, and can facilitate broader adoption of MPS approaches in preclinical research. Rapid prototyping of organ chip mold designs accelerates the processes of design, testing, and iteration, but geometric distortion and surface roughness of SLA resin prints can impede the development of standardizable manufacturing workflows. This study reports postprocessing procedures for manufacturing SLA-printed molds that produce fully cured, flat, patently bonded, and optically clear polydimethyl siloxane (PDMS) organ chips. Injection loading tests were conducted to identify milliscale membrane-free organ chip (MFOC) designs that allowed reproducible device loading by target end-users, a key requirement for broad nonexpert adoption in preclinical research. The optimized milliscale MFOC design was used to develop tissue engineering protocols for (i) driving bulk tissue vasculogenesis in MFOC, and (ii) seeding the bulk tissue interfaces with a confluent endothelium to stimulate self-assembly of perfusable anastomoses with the internal vasculature. Comparison of rocker- and pump-based protocols for flow-conditioning of anastomosed vascular beds revealed that continuous pump-driven flow is required for reproducible barrier maturation throughout the 3D tissue bulk. Demonstrated applications include nanoparticle perfusion and engineering perfusable tumor vasculature. These easily adaptable methods for designing and fabricating vascularized microphysiological systems can accelerate their adoption in a diverse range of preclinical laboratory settings.
Collapse
Affiliation(s)
| | - Ashley T Martier
- Department of Biomedical Engineering, Tulane University, New Orleans, USA
| | - Gideon Wills Kpeli
- Department of Biomedical Engineering, Tulane University, New Orleans, USA
| | | | - William Bralower
- Department of Biomedical Engineering, Tulane University, New Orleans, USA
| | - Elisabet Olsen
- Bioinnovation Program, Tulane University, New Orleans, USA
| | - Gabrielle Fortes
- Department of Biomedical Engineering, Tulane University, New Orleans, USA
| | - Caroline C Culp
- Department of Biomedical Engineering, Tulane University, New Orleans, USA
| | - Max Wendell
- Department of Biomedical Engineering, Tulane University, New Orleans, USA
| | - Keefer A Boone
- Department of Biomedical Engineering, Tulane University, New Orleans, USA
| | - Matthew R Burow
- Section of Hematology and Oncology, Deming Department of Medicine, Tulane University School of Medicine, New Orleans, USA
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, USA
| | - Mark J Mondrinos
- Department of Biomedical Engineering, Tulane University, New Orleans, USA
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, USA
- Department of Physiology, Tulane University School of Medicine, New Orleans, USA
| |
Collapse
|
17
|
Wang Y, Chen H, Zhang Z, He Y, Liu J, Zhao B, Wang Q, Xu J, Mao S, Zhang W, Yao X, Li W. Precision Treatment of Metachronous Multiple Primary Malignancies Based on Constructing Patient Tumor-Derived Organoids. Biomedicines 2024; 12:2708. [PMID: 39767614 PMCID: PMC11672942 DOI: 10.3390/biomedicines12122708] [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: 07/26/2024] [Revised: 08/25/2024] [Accepted: 08/30/2024] [Indexed: 01/11/2025] Open
Abstract
When a patient has two or more primary tumors, excluding the possibility of diffuse, recurrent, or metastatic, they can be defined as having multiple primary malignant neoplasms (MPMNs). Moreover, cases of three primary urinary tract tumors are very rare. Here, we reported a patient of MPMNs with four primary tumors, including three urinary tract cancers (renal cancer, prostate cancer, and bladder cancer) and lung cancer. The four tumors appeared over 13 years, and pathological results confirmed that they were all primary tumors after different surgeries. In addition, we established patient-derived organoids (PDOs) by collecting tumor specimens. Hematoxylin-eosin (H&E) staining of PDOs showed that the organoids were histopathological consistent with parental tumor. Immunohistochemistry showed that PDOs can also reflect the expression of pathological markers in patients. At the same time, PDOs may also serve as "avatars" of patients to predict sensitivity to different drugs. In summary, we reported a case of MPMNs with four primary tumors and established PDOs from its tumor specimens. A personalized treatment strategy was established based on the histopathological characteristics of the organoids.
Collapse
Affiliation(s)
- Yicheng Wang
- Department of Urology, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China; (Y.W.); (H.C.); (Z.Z.); (Y.H.); (J.L.); (J.X.); (S.M.); (X.Y.)
- School of Medicine, Tongji University, Shanghai 200092, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai 200435, China
| | - Haotian Chen
- Department of Urology, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China; (Y.W.); (H.C.); (Z.Z.); (Y.H.); (J.L.); (J.X.); (S.M.); (X.Y.)
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai 200435, China
| | - Zhijin Zhang
- Department of Urology, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China; (Y.W.); (H.C.); (Z.Z.); (Y.H.); (J.L.); (J.X.); (S.M.); (X.Y.)
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai 200435, China
| | - Yanyan He
- Department of Urology, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China; (Y.W.); (H.C.); (Z.Z.); (Y.H.); (J.L.); (J.X.); (S.M.); (X.Y.)
- Department of Pathology, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Ji Liu
- Department of Urology, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China; (Y.W.); (H.C.); (Z.Z.); (Y.H.); (J.L.); (J.X.); (S.M.); (X.Y.)
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai 200435, China
| | - Baoshuang Zhao
- Department of Hepatobiliary Surgery, Yiwu Central Hospital, Yiwu 322000, China;
| | - Qinwan Wang
- Department of Central Laboratory, Clinical Medicine Scientific and Technical Innovation Park, Shanghai Tenth People’s Hospital, Shanghai 200435, China;
| | - Jiangmei Xu
- Department of Urology, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China; (Y.W.); (H.C.); (Z.Z.); (Y.H.); (J.L.); (J.X.); (S.M.); (X.Y.)
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai 200435, China
| | - Shiyu Mao
- Department of Urology, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China; (Y.W.); (H.C.); (Z.Z.); (Y.H.); (J.L.); (J.X.); (S.M.); (X.Y.)
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai 200435, China
| | - Wentao Zhang
- Department of Urology, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China; (Y.W.); (H.C.); (Z.Z.); (Y.H.); (J.L.); (J.X.); (S.M.); (X.Y.)
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai 200435, China
| | - Xudong Yao
- Department of Urology, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China; (Y.W.); (H.C.); (Z.Z.); (Y.H.); (J.L.); (J.X.); (S.M.); (X.Y.)
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai 200435, China
| | - Wei Li
- Department of Urology, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China; (Y.W.); (H.C.); (Z.Z.); (Y.H.); (J.L.); (J.X.); (S.M.); (X.Y.)
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai 200435, China
| |
Collapse
|
18
|
Cordeiro S, Oliveira BB, Valente R, Ferreira D, Luz A, Baptista PV, Fernandes AR. Breaking the mold: 3D cell cultures reshaping the future of cancer research. Front Cell Dev Biol 2024; 12:1507388. [PMID: 39659521 PMCID: PMC11628512 DOI: 10.3389/fcell.2024.1507388] [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: 10/07/2024] [Accepted: 11/13/2024] [Indexed: 12/12/2024] Open
Abstract
Despite extensive efforts to unravel tumor behavior and develop anticancer therapies, most treatments fail when advanced to clinical trials. The main challenge in cancer research has been the absence of predictive cancer models, accurately mimicking the tumoral processes and response to treatments. The tumor microenvironment (TME) shows several human-specific physical and chemical properties, which cannot be fully recapitulated by the conventional 2D cell cultures or the in vivo animal models. These limitations have driven the development of novel in vitro cancer models, that get one step closer to the typical features of in vivo systems while showing better species relevance. This review introduces the main considerations required for developing and exploiting tumor spheroids and organoids as cancer models. We also detailed their applications in drug screening and personalized medicine. Further, we show the transition of these models into novel microfluidic platforms, for improved control over physiological parameters and high-throughput screening. 3D culture models have provided key insights into tumor biology, more closely resembling the in vivo TME and tumor characteristics, while enabling the development of more reliable and precise anticancer therapies.
Collapse
Affiliation(s)
- Sandra Cordeiro
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Beatriz B. Oliveira
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Ruben Valente
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Daniela Ferreira
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - André Luz
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Pedro V. Baptista
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Alexandra R. Fernandes
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| |
Collapse
|
19
|
Abed H, Radha R, Anjum S, Paul V, AlSawaftah N, Pitt WG, Ashammakhi N, Husseini GA. Targeted Cancer Therapy-on-A-Chip. Adv Healthc Mater 2024; 13:e2400833. [PMID: 39101627 PMCID: PMC11582519 DOI: 10.1002/adhm.202400833] [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: 03/04/2024] [Revised: 06/15/2024] [Indexed: 08/06/2024]
Abstract
Targeted cancer therapy (TCT) is gaining increased interest because it reduces the risks of adverse side effects by specifically treating tumor cells. TCT testing has traditionally been performed using two-dimensional (2D) cell culture and animal studies. Organ-on-a-chip (OoC) platforms have been developed to recapitulate cancer in vitro, as cancer-on-a-chip (CoC), and used for chemotherapeutics development and testing. This review explores the use of CoCs to both develop and test TCTs, with a focus on three main aspects, the use of CoCs to identify target biomarkers for TCT development, the use of CoCs to test free, un-encapsulated TCTs, and the use of CoCs to test encapsulated TCTs. Despite current challenges such as system scaling, and testing externally triggered TCTs, TCToC shows a promising future to serve as a supportive, pre-clinical platform to expedite TCT development and bench-to-bedside translation.
Collapse
Affiliation(s)
- Heba Abed
- Department of Chemical and Biological EngineeringAmerican University of SharjahSharjahUAE
| | - Remya Radha
- Department of Chemical and Biological EngineeringAmerican University of SharjahSharjahUAE
| | - Shabana Anjum
- Department of Chemical and Biological EngineeringAmerican University of SharjahSharjahUAE
| | - Vinod Paul
- Materials Science and Engineering PhD programCollege of Arts and SciencesAmerican University of SharjahSharjahUAE
| | - Nour AlSawaftah
- Materials Science and Engineering PhD programCollege of Arts and SciencesAmerican University of SharjahSharjahUAE
| | - William G. Pitt
- Department of Chemical EngineeringBrigham Young UniversityProvoUT84602USA
| | - Nureddin Ashammakhi
- Institute for Quantitative Health Science and Engineering (IQ) and Department of Biomedical Engineering (BME)Michigan State UniversityEast LansingMI48824USA
- Department of BioengineeringUniversity of California, Los AngelesLos AngelesCA90095‐1600USA
| | - Ghaleb A. Husseini
- Department of Chemical and Biological EngineeringAmerican University of SharjahSharjahUAE
- Materials Science and Engineering PhD programCollege of Arts and SciencesAmerican University of SharjahSharjahUAE
| |
Collapse
|
20
|
Tavakol DN, Nash TR, Kim Y, Graney PL, Liberman M, Fleischer S, Lock RI, O'Donnell A, Andrews L, Ning D, Yeager K, Harken A, Deoli N, Amundson SA, Garty G, Leong KW, Brenner DJ, Vunjak‐Novakovic G. Modeling the Effects of Protracted Cosmic Radiation in a Human Organ-on-Chip Platform. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401415. [PMID: 38965824 PMCID: PMC11558103 DOI: 10.1002/advs.202401415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/18/2024] [Indexed: 07/06/2024]
Abstract
Galactic cosmic radiation (GCR) is one of the most serious risks posed to astronauts during missions to the Moon and Mars. Experimental models capable of recapitulating human physiology are critical to understanding the effects of radiation on human organs and developing radioprotective measures against space travel exposures. The effects of systemic radiation are studied using a multi-organ-on-a-chip (multi-OoC) platform containing engineered tissue models of human bone marrow (site of hematopoiesis and acute radiation damage), cardiac muscle (site of chronic radiation damage) and liver (site of metabolism), linked by vascular circulation with an endothelial barrier separating individual tissue chambers from the vascular perfusate. Following protracted neutron radiation, the most damaging radiation component in deep space, a greater deviation of tissue function is observed as compared to the same cumulative dose delivered acutely. Further, by characterizing engineered bone marrow (eBM)-derived immune cells in circulation, 58 unique genes specific to the effects of protracted neutron dosing are identified, as compared to acutely irradiated and healthy tissues. It propose that this bioengineered platform allows studies of human responses to extended radiation exposure in an "astronaut-on-a-chip" model that can inform measures for mitigating cosmic radiation injury.
Collapse
Affiliation(s)
| | - Trevor R. Nash
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10032USA
| | - Youngbin Kim
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10032USA
| | - Pamela L. Graney
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10032USA
| | - Martin Liberman
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10032USA
| | - Sharon Fleischer
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10032USA
| | - Roberta I. Lock
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10032USA
| | - Aaron O'Donnell
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10032USA
| | - Leah Andrews
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10032USA
| | - Derek Ning
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10032USA
| | - Keith Yeager
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10032USA
| | - Andrew Harken
- Center for Radiological ResearchColumbia UniversityNew YorkNY10032USA
| | - Naresh Deoli
- Center for Radiological ResearchColumbia UniversityNew YorkNY10032USA
| | - Sally A. Amundson
- Center for Radiological ResearchColumbia UniversityNew YorkNY10032USA
| | - Guy Garty
- Center for Radiological ResearchColumbia UniversityNew YorkNY10032USA
| | - Kam W. Leong
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10032USA
| | - David J. Brenner
- Center for Radiological ResearchColumbia UniversityNew YorkNY10032USA
| | - Gordana Vunjak‐Novakovic
- Department of Biomedical EngineeringDepartment of Medicine, and College of Dental MedicineColumbia UniversityNew YorkNY10032USA
| |
Collapse
|
21
|
Harriot AD, Ward CW, Kim DH. Microphysiological systems to advance human pathophysiology and translational medicine. J Appl Physiol (1985) 2024; 137:1494-1501. [PMID: 39417817 DOI: 10.1152/japplphysiol.00087.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 09/09/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
Microphysiological systems (MPS) or "organ-on-a-chip" models are sophisticated tools that harness techniques from cell biology, tissue engineering, and microengineering to recapitulate human physiology. Typically, MPS are biofabricated three-dimensional (3-D) tissue constructs integrated into platforms designed to mimic the tissue microenvironment and provide functional outputs. Over the past decade, researchers have endeavored to manufacture high-throughput, high-fidelity MPS models of all major human organs. By incorporating patient-derived cells, researchers have produced biomimetic models of tissues with disease-linked genetic mutations capable of exhibiting patient heterogeneity. This work has demonstrated that MPS more closely model organotypic function and pathophysiology than traditional two-dimensional (2-D) culture systems. Moreover, investigators have shown that human MPS are better predictors of drug efficacy and toxicity than animal models. Thus, MPS have emerged as a promising candidate to improve the efficacy and safety of preclinical trials. In this mini-review, we provide an overview of current advances in MPS models, their applications in mechanistic research, and relevance to drug screening. Finally, we discuss current investments in MPS development by the United States federal government and research institutions around the world to advance translational medicine.
Collapse
Affiliation(s)
- Anicca D Harriot
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States
- Division of Geriatric Medicine and Gerontology, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
- Center for Microphysiological Systems, Johns Hopkins University, Baltimore, Maryland, United States
| | - Christopher W Ward
- Department of Orthopedics, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Claude D. Pepper Older Americans Independence Center, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland, United States
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
- Institute for NanoBio Technology, Johns Hopkins University, Baltimore, Maryland, United States
- Center for Microphysiological Systems, Johns Hopkins University, Baltimore, Maryland, United States
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland, United States
| |
Collapse
|
22
|
Peng X, Wu L, Li Q, Ge Y, Xu T, Zhao J. An Easy-to-Use Arrayed Brain-Heart Chip. BIOSENSORS 2024; 14:517. [PMID: 39589976 PMCID: PMC11592345 DOI: 10.3390/bios14110517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 11/28/2024]
Abstract
Multi-organ chips are effective at emulating human tissue and organ functions and at replicating the interactions among tissues and organs. An arrayed brain-heart chip was introduced whose configuration comprises open culture chambers and closed biomimetic vascular channels distributed in a horizontal pattern, separated from each other by an endothelial barrier based on fibrin matrix. A 300 μm-high and 13.2 mm-long endothelial barrier surrounded each organoid culture chamber, thereby satisfying the material transport requirements. Numerical simulations were used to analyze the construction process of fibrin barriers in order to optimize the structural design and experimental manipulation, which exhibited a high degree of correlation with experiment results. In each interconnective unit, a cerebral organoid, a cardiac organoid, and endothelial cells were co-cultured stably for a minimum of one week. The permeability of the endothelial barrier and recirculating perfusion enabled cross talk between cerebral organoids and cardiac organoids, as well as between organoids and endothelial cells. This was corroborated by the presence of cardiac troponin I (cTnI) in the cerebral organoid culture chamber and the observation of cerebral organoid and endothelial cells invading the fibrin matrix after one week of co-culture. The arrayed chip was simple to manipulate, clearly visible under a microscope, and compatible with automated pipetting devices, and therefore had significant potential for application.
Collapse
Affiliation(s)
- Xiyao Peng
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China; (X.P.); (Q.L.); (Y.G.); (T.X.)
- College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lei Wu
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China; (X.P.); (Q.L.); (Y.G.); (T.X.)
- College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiushi Li
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China; (X.P.); (Q.L.); (Y.G.); (T.X.)
| | - Yuqing Ge
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China; (X.P.); (Q.L.); (Y.G.); (T.X.)
- College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tiegang Xu
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China; (X.P.); (Q.L.); (Y.G.); (T.X.)
| | - Jianlong Zhao
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China; (X.P.); (Q.L.); (Y.G.); (T.X.)
- College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
23
|
Taylor S, Mueller E, Jones LR, Makela AV, Ashammakhi N. Translational Aspects of 3D and 4D Printing and Bioprinting. Adv Healthc Mater 2024; 13:e2400463. [PMID: 38979857 DOI: 10.1002/adhm.202400463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/22/2024] [Indexed: 07/10/2024]
Abstract
Three-dimensional (3D) printed medical devices include orthopedic and craniofacial implants, surgical tools, and external prosthetics that have been directly used in patients. While the advances of additive manufacturing techniques in the production of medical devices have been on the rise, clinical translation of living cellular constructs face significant limitations in terms of regulatory affairs, process technology, and materials development. In this perspective, the current status-quo of 3D and four-dimensional (4D) (bio)printing is summarized, current advancements are discussed and the challenges that need to be addressed for improved industrial translation and clinical applications of bioprinting are highlighted. It is focused on a multidisciplinary approach in discussing the key translational considerations, from the perspective of industry, regulatory bodies, funding strategies, and future directions.
Collapse
Affiliation(s)
| | - Eva Mueller
- Ricoh 3D for Healthcare, Ricoh USA, Winston-Salem, NC 27101, USA
| | - Lamont R Jones
- Department of Otolaryngology, Henry Ford Heath, Detroit, MI 48322, USA
| | - Ashley V Makela
- Institute for Quantitative Health Science & Engineering and Department of Engineering, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Nureddin Ashammakhi
- Institute for Quantitative Health Science & Engineering and Department of Engineering, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
24
|
Gaebler D, Hachey SJ, Hughes CCW. Improving tumor microenvironment assessment in chip systems through next-generation technology integration. Front Bioeng Biotechnol 2024; 12:1462293. [PMID: 39386043 PMCID: PMC11461320 DOI: 10.3389/fbioe.2024.1462293] [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: 07/09/2024] [Accepted: 09/10/2024] [Indexed: 10/12/2024] Open
Abstract
The tumor microenvironment (TME) comprises a diverse array of cells, both cancerous and non-cancerous, including stromal cells and immune cells. Complex interactions among these cells play a central role in driving cancer progression, impacting critical aspects such as tumor initiation, growth, invasion, response to therapy, and the development of drug resistance. While targeting the TME has emerged as a promising therapeutic strategy, there is a critical need for innovative approaches that accurately replicate its complex cellular and non-cellular interactions; the goal being to develop targeted, personalized therapies that can effectively elicit anti-cancer responses in patients. Microfluidic systems present notable advantages over conventional in vitro 2D co-culture models and in vivo animal models, as they more accurately mimic crucial features of the TME and enable precise, controlled examination of the dynamic interactions among multiple human cell types at any time point. Combining these models with next-generation technologies, such as bioprinting, single cell sequencing and real-time biosensing, is a crucial next step in the advancement of microfluidic models. This review aims to emphasize the importance of this integrated approach to further our understanding of the TME by showcasing current microfluidic model systems that integrate next-generation technologies to dissect cellular intra-tumoral interactions across different tumor types. Carefully unraveling the complexity of the TME by leveraging next generation technologies will be pivotal for developing targeted therapies that can effectively enhance robust anti-tumoral responses in patients and address the limitations of current treatment modalities.
Collapse
Affiliation(s)
- Daniela Gaebler
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| | - Stephanie J. Hachey
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| | - Christopher C. W. Hughes
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
- Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
25
|
Yang Y, Gao X, Widdicombe B, Zhang X, Zielinski JL, Cheng T, Gunatilaka A, Leung KK, Plaxco KW, Rajasekharan Unnithan R, Stewart AG. Dual-Purpose Aptamer-Based Sensors for Real-Time, Multiplexable Monitoring of Metabolites in Cell Culture Media. ACS NANO 2024; 18. [PMID: 39255458 PMCID: PMC11441400 DOI: 10.1021/acsnano.4c06813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/12/2024]
Abstract
The availability of high-frequency, real-time measurements of the concentrations of specific metabolites in cell culture systems will enable a deeper understanding of cellular metabolism and facilitate the application of good laboratory practice standards in cell culture protocols. However, currently available approaches to this end either are constrained to single-time-point and single-parameter measurements or are limited in the range of detectable analytes. Electrochemical aptamer-based (EAB) biosensors have demonstrated utility in real-time monitoring of analytes in vivo in blood and tissues. Here, we characterize a pH-sensing capability of EAB sensors that is independent of the specific target analyte of the aptamer sequence. We applied this dual-purpose EAB to the continuous measurement of pH and phenylalanine in several in vitro cell culture settings. The miniature EAB sensor that we developed exhibits rapid response times, good stability, high repeatability, and biologically relevant sensitivity. We also developed and characterized a leak-free reference electrode that mitigates the potential cytotoxic effects of silver ions released from conventional reference electrodes. Using the resulting dual-purpose sensor, we performed hourly measurements of pH and phenylalanine concentrations in the medium superfusing cultured epithelial tumor cell lines (A549, MDA-MB-23) and a human fibroblast cell line (MRC-5) for periods of up to 72 h. Our scalable technology may be multiplexed for high-throughput monitoring of pH and multiple analytes in support of the broad metabolic qualification of microphysiological systems.
Collapse
Affiliation(s)
- Yiling Yang
- Department
of Electrical and Electronic Engineering, University of Melbourne, Melbourne, Victoria 3010, Australia
- ARC
Centre for Personalised Therapeutics Technologies, Melbourne, Victoria 3010, Australia
| | - Xumei Gao
- Department
of Biochemistry and Pharmacology, University
of Melbourne, Melbourne, Victoria 3010, Australia
- ARC
Centre for Personalised Therapeutics Technologies, Melbourne, Victoria 3010, Australia
| | - Bryce Widdicombe
- Department
of Electrical and Electronic Engineering, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Xiaodan Zhang
- Department
of Biochemistry and Pharmacology, University
of Melbourne, Melbourne, Victoria 3010, Australia
| | - Jana Lorraine Zielinski
- Department
of Biochemistry and Pharmacology, University
of Melbourne, Melbourne, Victoria 3010, Australia
| | - Tianhong Cheng
- Department
of Biochemistry and Pharmacology, University
of Melbourne, Melbourne, Victoria 3010, Australia
| | - Avanka Gunatilaka
- Department
of Biochemistry and Pharmacology, University
of Melbourne, Melbourne, Victoria 3010, Australia
| | - Kaylyn K. Leung
- Department
of Chemistry and Biochemistry, University
of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Kevin W. Plaxco
- Department
of Chemistry and Biochemistry, University
of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Ranjith Rajasekharan Unnithan
- Department
of Electrical and Electronic Engineering, University of Melbourne, Melbourne, Victoria 3010, Australia
- ARC
Centre for Personalised Therapeutics Technologies, Melbourne, Victoria 3010, Australia
| | - Alastair G. Stewart
- Department
of Biochemistry and Pharmacology, University
of Melbourne, Melbourne, Victoria 3010, Australia
- ARC
Centre for Personalised Therapeutics Technologies, Melbourne, Victoria 3010, Australia
| |
Collapse
|
26
|
Brasino DSK, Speese SD, Schilling K, Schutt CE, Barton MC. A Linkable, Polycarbonate Gut Microbiome-Distal Tumor Chip Platform for Interrogating Cancer Promoting Mechanisms. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309220. [PMID: 39023197 PMCID: PMC11425222 DOI: 10.1002/advs.202309220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/31/2024] [Indexed: 07/20/2024]
Abstract
Gut microbiome composition is tied to diseases ranging from arthritis to cancer to depression. However, mechanisms of action are poorly understood, limiting development of relevant therapeutics. Organ-on-chip platforms, which model minimal functional units of tissues and can tightly control communication between them, are ideal platforms to study these relationships. Many gut microbiome models are published to date but devices are typically fabricated using oxygen permeable polydimethylsiloxane, requiring interventions to support anaerobic bacteria. To address this challenge, a platform is developed where the chips are fabricated entirely from gas-impermeable polycarbonate without tapes or gaskets. These chips replicate polarized villus-like structures of the native tissue. Further, they enable co-cultures of commensal anaerobic bacteria Blautia coccoides on the surface of gut epithelia for two days within a standard incubator. Another complication of commonly used materials in organ-on-chip devices is high ad-/absorption, limiting applications in high-resolution microscopy and biomolecule interaction studies. For future communication studies between gut microbiota and distal tumors, an additional polycarbonate chip design is developed to support hydrogel-embedded tissue culture. These chips enable high-resolution microscopy with all relevant processing done on-chip. Designed for facile linking, this platform will make a variety of mechanistic studies possible.
Collapse
Affiliation(s)
- Danielle S K Brasino
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Sean D Speese
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Kevin Schilling
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Carolyn E Schutt
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, 97201, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Michelle C Barton
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, 97201, USA
| |
Collapse
|
27
|
Kang S, Chen EC, Cifuentes H, Co JY, Cole G, Graham J, Hsia R, Kiyota T, Klein JA, Kroll KT, Nieves Lopez LM, Norona LM, Peiris H, Potla R, Romero-Lopez M, Roth JG, Tseng M, Fullerton AM, Homan KA. Complex in vitromodels positioned for impact to drug testing in pharma: a review. Biofabrication 2024; 16:042006. [PMID: 39189069 DOI: 10.1088/1758-5090/ad6933] [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: 12/22/2023] [Accepted: 07/30/2024] [Indexed: 08/28/2024]
Abstract
Recent years have seen the creation and popularization of various complexin vitromodels (CIVMs), such as organoids and organs-on-chip, as a technology with the potential to reduce animal usage in pharma while also enhancing our ability to create safe and efficacious drugs for patients. Public awareness of CIVMs has increased, in part, due to the recent passage of the FDA Modernization Act 2.0. This visibility is expected to spur deeper investment in and adoption of such models. Thus, end-users and model developers alike require a framework to both understand the readiness of current models to enter the drug development process, and to assess upcoming models for the same. This review presents such a framework for model selection based on comparative -omics data (which we term model-omics), and metrics for qualification of specific test assays that a model may support that we term context-of-use (COU) assays. We surveyed existing healthy tissue models and assays for ten drug development-critical organs of the body, and provide evaluations of readiness and suggestions for improving model-omics and COU assays for each. In whole, this review comes from a pharma perspective, and seeks to provide an evaluation of where CIVMs are poised for maximum impact in the drug development process, and a roadmap for realizing that potential.
Collapse
Affiliation(s)
- Serah Kang
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Eugene C Chen
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Helen Cifuentes
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Julia Y Co
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Gabrielle Cole
- Investigative Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Jessica Graham
- Product Quality & Occupational Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of Americaica
| | - Rebecca Hsia
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Tomomi Kiyota
- Investigative Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Jessica A Klein
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Katharina T Kroll
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Lenitza M Nieves Lopez
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Leah M Norona
- Investigative Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Heshan Peiris
- Human Genetics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Ratnakar Potla
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Monica Romero-Lopez
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Julien G Roth
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Min Tseng
- Investigative Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Aaron M Fullerton
- Investigative Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Kimberly A Homan
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| |
Collapse
|
28
|
Mulay AR, Hwang J, Kim DH. Microphysiological Blood-Brain Barrier Systems for Disease Modeling and Drug Development. Adv Healthc Mater 2024; 13:e2303180. [PMID: 38430211 PMCID: PMC11338747 DOI: 10.1002/adhm.202303180] [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: 09/20/2023] [Revised: 02/22/2024] [Indexed: 03/03/2024]
Abstract
The blood-brain barrier (BBB) is a highly controlled microenvironment that regulates the interactions between cerebral blood and brain tissue. Due to its selectivity, many therapeutics targeting various neurological disorders are not able to penetrate into brain tissue. Pre-clinical studies using animals and other in vitro platforms have not shown the ability to fully replicate the human BBB leading to the failure of a majority of therapeutics in clinical trials. However, recent innovations in vitro and ex vivo modeling called organs-on-chips have shown the potential to create more accurate disease models for improved drug development. These microfluidic platforms induce physiological stressors on cultured cells and are able to generate more physiologically accurate BBBs compared to previous in vitro models. In this review, different approaches to create BBBs-on-chips are explored alongside their application in modeling various neurological disorders and potential therapeutic efficacy. Additionally, organs-on-chips use in BBB drug delivery studies is discussed, and advances in linking brain organs-on-chips onto multiorgan platforms to mimic organ crosstalk are reviewed.
Collapse
Affiliation(s)
- Atharva R. Mulay
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland 21218
| | - Jihyun Hwang
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland 21218
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205
- Center for Microphysiological Systems, Johns Hopkins University School of Medicine, Baltimore, MD, 21205
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, 21218
| |
Collapse
|
29
|
Stresser DM, Kopec AK, Hewitt P, Hardwick RN, Van Vleet TR, Mahalingaiah PKS, O'Connell D, Jenkins GJ, David R, Graham J, Lee D, Ekert J, Fullerton A, Villenave R, Bajaj P, Gosset JR, Ralston SL, Guha M, Amador-Arjona A, Khan K, Agarwal S, Hasselgren C, Wang X, Adams K, Kaushik G, Raczynski A, Homan KA. Towards in vitro models for reducing or replacing the use of animals in drug testing. Nat Biomed Eng 2024; 8:930-935. [PMID: 38151640 DOI: 10.1038/s41551-023-01154-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Affiliation(s)
- David M Stresser
- Quantitative, Translational & ADME Sciences, AbbVie, North Chicago, IL, USA.
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ), .
- IQ Microphysiological Systems Affiliate (IQ-), .
| | - Anna K Kopec
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Drug Safety Research & Development, Pfizer, Inc., Groton, CT, USA
| | - Philip Hewitt
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Chemical and Preclinical Safety, Merck KGaA, Darmstadt, Germany
| | - Rhiannon N Hardwick
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Discovery Toxicology, Pharmaceutical Candidate Optimization, Bristol Myers Squibb, San Diego, CA, USA
| | - Terry R Van Vleet
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Investigative Toxicology and Pathology, AbbVie, North Chicago, IL, USA
| | - Prathap Kumar S Mahalingaiah
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Investigative Toxicology and Pathology, AbbVie, North Chicago, IL, USA
| | - Denice O'Connell
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- Global Animal Welfare, AbbVie, North Chicago, IL, USA
- IQ 3Rs (Replacement, Reduction, Refinement) Translational and Predictive Sciences Leadership Group
| | - Gary J Jenkins
- Quantitative, Translational & ADME Sciences, AbbVie, North Chicago, IL, USA
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Translational and ADME Sciences Leadership Group (TALG)
| | - Rhiannon David
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Clinical Pharmacology & Safety Sciences, AstraZeneca, Cambridge, UK
| | - Jessica Graham
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- Product Quality & Occupational Toxicology, Genentech, Inc., South San Francisco, CA, USA
- IQ DruSafe
- Safety Assessment, Genentech, Inc., South San Francisco, CA, USA
| | - Donna Lee
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ 3Rs (Replacement, Reduction, Refinement) Translational and Predictive Sciences Leadership Group
- Safety Assessment, Genentech, Inc., South San Francisco, CA, USA
| | - Jason Ekert
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- UCB Pharma, Cambridge, MA, USA
| | - Aaron Fullerton
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Investigative Toxicology, Genentech, Inc., South San Francisco, CA, USA
| | - Remi Villenave
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Piyush Bajaj
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Global Investigative Toxicology, Preclinical Safety, Sanofi, Cambridge, MA, USA
| | - James R Gosset
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc, Cambridge, MA, USA
| | - Sherry L Ralston
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ 3Rs (Replacement, Reduction, Refinement) Translational and Predictive Sciences Leadership Group
- Preclinical Safety, AbbVie, North Chicago, IL, USA
| | - Manti Guha
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Discovery Biology, Incyte, Wilmington, DE, USA
| | - Alejandro Amador-Arjona
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Discovery Biology, Incyte, Wilmington, DE, USA
| | - Kainat Khan
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Clinical Pharmacology & Safety Sciences, AstraZeneca, Cambridge, UK
| | - Saket Agarwal
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Investigative Toxicology, Early Development, Alnylam Pharmaceuticals, Cambridge, MA, USA
| | - Catrin Hasselgren
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ DruSafe
- Predictive Toxicology, Genentech, Inc., South San Francisco, CA, USA
| | - Xiaoting Wang
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Translational Safety & Bioanalytical Sciences, Amgen Research, Amgen Inc., South San Francisco, CA, USA
| | - Khary Adams
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ 3Rs (Replacement, Reduction, Refinement) Translational and Predictive Sciences Leadership Group
- Laboratory Animal Resources, Incyte, Wilmington, DE, USA
| | - Gaurav Kaushik
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Nonclinical Drug Safety, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, USA
| | - Arkadiusz Raczynski
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Preclinical Safety Assessment, Vertex Pharmaceuticals, Inc, Boston, MA, USA
| | - Kimberly A Homan
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ), .
- IQ Microphysiological Systems Affiliate (IQ-), .
- Complex in vitro Systems Group, Genentech, Inc., South San Francisco, CA, USA.
| |
Collapse
|
30
|
Kim R, Sung JH. Recent Advances in Gut- and Gut-Organ-Axis-on-a-Chip Models. Adv Healthc Mater 2024; 13:e2302777. [PMID: 38243887 DOI: 10.1002/adhm.202302777] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 12/21/2023] [Indexed: 01/22/2024]
Abstract
The human gut extracts nutrients from the diet while forming the largest barrier against the outer environment. In addition, the gut actively maintains homeostasis through intricate interactions with the gut microbes, the immune system, the enteric nervous system, and other organs. These interactions influence digestive health and, furthermore, play crucial roles in systemic health and disease. Given its primary role in absorbing and metabolizing orally administered drugs, there is significant interest in the development of preclinical in vitro model systems that can accurately emulate the intestine in vivo. A gut-on-a-chip system holds great potential as a testing and screening platform because of its ability to emulate the physiological aspects of in vivo tissues and expandability to incorporate and combine with other organs. This review aims to identify the key physiological features of the human gut that need to be incorporated to build more accurate preclinical models and highlights the recent progress in gut-on-a-chip systems and competing technologies toward building more physiologically relevant preclinical model systems. Furthermore, various efforts to construct multi-organ systems with the gut, called gut-organ-axis-on-a-chip models, are discussed. In vitro gut models with physiological relevance can provide valuable platforms for bridging the gap between preclinical and clinical studies.
Collapse
Affiliation(s)
- Raehyun Kim
- Department of Biological and Chemical Engineering, Hongik University, Sejong, 30016, Republic of Korea
| | - Jong Hwan Sung
- Department of Chemical Engineering, Hongik University, Seoul, 04066, Republic of Korea
| |
Collapse
|
31
|
Deng ZM, Dai FF, Wang RQ, Deng HB, Yin TL, Cheng YX, Chen GT. Organ-on-a-chip: future of female reproductive pathophysiological models. J Nanobiotechnology 2024; 22:455. [PMID: 39085921 PMCID: PMC11290169 DOI: 10.1186/s12951-024-02651-w] [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: 08/02/2023] [Accepted: 06/18/2024] [Indexed: 08/02/2024] Open
Abstract
The female reproductive system comprises the internal and external genitalia, which communicate through intricate endocrine pathways. Besides secreting hormones that maintain the female secondary sexual characteristics, it also produces follicles and offspring. However, the in vitro systems have been very limited in recapitulating the specific anatomy and pathophysiology of women. Organ-on-a-chip technology, based on microfluidics, can better simulate the cellular microenvironment in vivo, opening a new field for the basic and clinical research of female reproductive system diseases. This technology can not only reconstruct the organ structure but also emulate the organ function as much as possible. The precisely controlled fluidic microenvironment provided by microfluidics vividly mimics the complex endocrine hormone crosstalk among various organs of the female reproductive system, making it a powerful preclinical tool and the future of pathophysiological models of the female reproductive system. Here, we review the research on the application of organ-on-a-chip platforms in the female reproductive systems, focusing on the latest progress in developing models that reproduce the physiological functions or disease features of female reproductive organs and tissues, and highlighting the challenges and future directions in this field.
Collapse
Affiliation(s)
- Zhi-Min Deng
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Fang-Fang Dai
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Rui-Qi Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Hong-Bing Deng
- Hubei International Scientific and Technological Cooperation Base of Sustainable Resource and Energy, Hubei Key Laboratory of Biomass Resource Chemistry and Environmental Biotechnology, School of Resource and Environmental Science, Wuhan University, Wuhan, Hubei, 430060, China
| | - Tai-Lang Yin
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China.
| | - Yan-Xiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China.
| | - Gan-Tao Chen
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China.
| |
Collapse
|
32
|
Martier A, Chen Z, Schaps H, Mondrinos MJ, Fang JS. Capturing physiological hemodynamic flow and mechanosensitive cell signaling in vessel-on-a-chip platforms. Front Physiol 2024; 15:1425618. [PMID: 39135710 PMCID: PMC11317428 DOI: 10.3389/fphys.2024.1425618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 07/10/2024] [Indexed: 08/15/2024] Open
Abstract
Recent advances in organ chip (or, "organ-on-a-chip") technologies and microphysiological systems (MPS) have enabled in vitro investigation of endothelial cell function in biomimetic three-dimensional environments under controlled fluid flow conditions. Many current organ chip models include a vascular compartment; however, the design and implementation of these vessel-on-a-chip components varies, with consequently varied impact on their ability to capture and reproduce hemodynamic flow and associated mechanosensitive signaling that regulates key characteristics of healthy, intact vasculature. In this review, we introduce organ chip and vessel-on-a-chip technology in the context of existing in vitro and in vivo vascular models. We then briefly discuss the importance of mechanosensitive signaling for vascular development and function, with focus on the major mechanosensitive signaling pathways involved. Next, we summarize recent advances in MPS and organ chips with an integrated vascular component, with an emphasis on comparing both the biomimicry and adaptability of the diverse approaches used for supporting and integrating intravascular flow. We review current data showing how intravascular flow and fluid shear stress impacts vessel development and function in MPS platforms and relate this to existing work in cell culture and animal models. Lastly, we highlight new insights obtained from MPS and organ chip models of mechanosensitive signaling in endothelial cells, and how this contributes to a deeper understanding of vessel growth and function in vivo. We expect this review will be of broad interest to vascular biologists, physiologists, and cardiovascular physicians as an introduction to organ chip platforms that can serve as viable model systems for investigating mechanosensitive signaling and other aspects of vascular physiology.
Collapse
Affiliation(s)
- A. Martier
- Department of Biomedical Engineering, School of Science and Engineering, Tulane University, New Orleans, LA, United States
| | - Z. Chen
- Department of Cell and Molecular Biology, School of Science and Engineering, Tulane University, New Orleans, LA, United States
| | - H. Schaps
- Department of Cell and Molecular Biology, School of Science and Engineering, Tulane University, New Orleans, LA, United States
| | - M. J. Mondrinos
- Department of Biomedical Engineering, School of Science and Engineering, Tulane University, New Orleans, LA, United States
- Department of Physiology, School of Medicine, Tulane University, New Orleans, LA, United States
| | - J. S. Fang
- Department of Cell and Molecular Biology, School of Science and Engineering, Tulane University, New Orleans, LA, United States
- Department of Physiology, School of Medicine, Tulane University, New Orleans, LA, United States
| |
Collapse
|
33
|
Huang Y, Liu T, Huang Q, Wang Y. From Organ-on-a-Chip to Human-on-a-Chip: A Review of Research Progress and Latest Applications. ACS Sens 2024; 9:3466-3488. [PMID: 38991227 DOI: 10.1021/acssensors.4c00004] [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: 07/13/2024]
Abstract
Organ-on-a-Chip (OOC) technology, which emulates the physiological environment and functionality of human organs on a microfluidic chip, is undergoing significant technological advancements. Despite its rapid evolution, this technology is also facing notable challenges, such as the lack of vascularization, the development of multiorgan-on-a-chip systems, and the replication of the human body on a single chip. The progress of microfluidic technology has played a crucial role in steering OOC toward mimicking the human microenvironment, including vascularization, microenvironment replication, and the development of multiorgan microphysiological systems. Additionally, advancements in detection, analysis, and organoid imaging technologies have enhanced the functionality and efficiency of Organs-on-Chips (OOCs). In particular, the integration of artificial intelligence has revolutionized organoid imaging, significantly enhancing high-throughput drug screening. Consequently, this review covers the research progress of OOC toward Human-on-a-chip, the integration of sensors in OOCs, and the latest applications of organoid imaging technologies in the biomedical field.
Collapse
Affiliation(s)
- Yisha Huang
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, Sichuan 610212, China
| | - Tong Liu
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qi Huang
- School of Information Engineering, Shanghai Maritime University, Shanghai 201306, China
| | - Yuxi Wang
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
34
|
Ohri S, Parekh P, Nichols L, Rajan SAP, Cirit M. Utilization of a Human Liver Tissue Chip for Drug-Metabolizing Enzyme Induction Studies of Perpetrator and Victim Drugs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.17.603946. [PMID: 39091853 PMCID: PMC11291003 DOI: 10.1101/2024.07.17.603946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Polypharmacy-related drug-drug interactions (DDIs) are a significant and growing healthcare concern. Increasing number of therapeutic drugs on the market underscores the necessity to accurately assess the new drug combinations during pre-clinical evaluation for DDIs. In vitro primary human hepatocyte (PHH)-only models are commonly used only for perpetrator DDI studies due to their rapid loss of metabolic function. But co-culturing non-human cells with human PHHs can stabilize metabolic activity and be utilized for both perpetrator and victim studies, though this raises concerns about human specificity for accurate clinical assessment. In this study, we evaluated Liver Tissue Chip (LTC) with PHH-only liver microphysiological system (MPS) for DDI induction studies. Liver MPS from three individual donors maintained their functional and metabolic activity for up to 4 weeks demonstrating suitability for long-term pharmacokinetics (PK) studies. The responses to rifampicin induction of three PHH donors were assessed using CYP activity and mRNA changes. Additionally, victim PK studies were conducted with midazolam (high clearance) and alprazolam (low clearance) following rifampicin-mediated induction which resulted in a 2-fold and a 2.6-fold increase in midazolam and alprazolam intrinsic clearance values respectively compared to the untreated liver MPS. We also investigated the induction effects of different dosing regimens of the perpetrator drug (rifampicin) on CYP activity levels, showing minimal variation in the intrinsic clearance of the victim drug (midazolam). This study demonstrates the utility of the LTC for in vitro liver-specific DDI induction studies, providing a translational experimental system to predict clinical clearance values of both perpetrator and victim drugs.
Collapse
Affiliation(s)
- Shivam Ohri
- Javelin Biotech, Inc., 299 Washington Street, Woburn, Massachusetts 01801, USA
| | - Paarth Parekh
- Javelin Biotech, Inc., 299 Washington Street, Woburn, Massachusetts 01801, USA
| | - Lauren Nichols
- Javelin Biotech, Inc., 299 Washington Street, Woburn, Massachusetts 01801, USA
| | | | - Murat Cirit
- Javelin Biotech, Inc., 299 Washington Street, Woburn, Massachusetts 01801, USA
| |
Collapse
|
35
|
Kumar D, Nadda R, Repaka R. Advances and challenges in organ-on-chip technology: toward mimicking human physiology and disease in vitro. Med Biol Eng Comput 2024; 62:1925-1957. [PMID: 38436835 DOI: 10.1007/s11517-024-03062-7] [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: 10/19/2023] [Accepted: 02/23/2024] [Indexed: 03/05/2024]
Abstract
Organs-on-chips have been tissues or three-dimensional (3D) mini-organs that comprise numerous cell types and have been produced on microfluidic chips to imitate the complicated structures and interactions of diverse cell types and organs under controlled circumstances. Several morphological and physiological distinctions exist between traditional 2D cultures, animal models, and the growing popular 3D cultures. On the other hand, animal models might not accurately simulate human toxicity because of physiological variations and interspecies metabolic capability. The on-chip technique allows for observing and understanding the process and alterations occurring in metastases. The present study aimed to briefly overview single and multi-organ-on-chip techniques. The current study addresses each platform's essential benefits and characteristics and highlights recent developments in developing and utilizing technologies for single and multi-organs-on-chips. The study also discusses the drawbacks and constraints associated with these models, which include the requirement for standardized procedures and the difficulties of adding immune cells and other intricate biological elements. Finally, a comprehensive review demonstrated that the organs-on-chips approach has a potential way of investigating organ function and disease. The advancements in single and multi-organ-on-chip structures can potentially increase drug discovery and minimize dependency on animal models, resulting in improved therapies for human diseases.
Collapse
Affiliation(s)
- Dhiraj Kumar
- Department of Mechanical Engineering, Indian Institute of Technology Ropar, Punjab, 140001, India
| | - Rahul Nadda
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Punjab, 140001, India.
| | - Ramjee Repaka
- Department of Mechanical Engineering, Indian Institute of Technology Ropar, Punjab, 140001, India
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Punjab, 140001, India
| |
Collapse
|
36
|
Elblová P, Lunova M, Dejneka A, Jirsa M, Lunov O. Impact of mechanical cues on key cell functions and cell-nanoparticle interactions. DISCOVER NANO 2024; 19:106. [PMID: 38907808 PMCID: PMC11193707 DOI: 10.1186/s11671-024-04052-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 06/14/2024] [Indexed: 06/24/2024]
Abstract
In recent years, it has been recognized that mechanical forces play an important regulative role in living organisms and possess a direct impact on crucial cell functions, ranging from cell growth to maintenance of tissue homeostasis. Advancements in mechanobiology have revealed the profound impact of mechanical signals on diverse cellular responses that are cell type specific. Notably, numerous studies have elucidated the pivotal role of different mechanical cues as regulatory factors influencing various cellular processes, including cell spreading, locomotion, differentiation, and proliferation. Given these insights, it is unsurprising that the responses of cells regulated by physical forces are intricately linked to the modulation of nanoparticle uptake kinetics and processing. This complex interplay underscores the significance of understanding the mechanical microenvironment in shaping cellular behaviors and, consequently, influencing how cells interact with and process nanoparticles. Nevertheless, our knowledge on how localized physical forces affect the internalization and processing of nanoparticles by cells remains rather limited. A significant gap exists in the literature concerning a systematic analysis of how mechanical cues might bias the interactions between nanoparticles and cells. Hence, our aim in this review is to provide a comprehensive and critical analysis of the existing knowledge regarding the influence of mechanical cues on the complicated dynamics of cell-nanoparticle interactions. By addressing this gap, we would like to contribute to a detailed understanding of the role that mechanical forces play in shaping the complex interplay between cells and nanoparticles.
Collapse
Affiliation(s)
- Petra Elblová
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18200, Prague, Czech Republic
- Faculty of Mathematics and Physics, Charles University, Ke Karlovu 3, 121 16, Prague 2, Czech Republic
| | - Mariia Lunova
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18200, Prague, Czech Republic
- Institute for Clinical & Experimental Medicine (IKEM), 14021, Prague, Czech Republic
| | - Alexandr Dejneka
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18200, Prague, Czech Republic
| | - Milan Jirsa
- Institute for Clinical & Experimental Medicine (IKEM), 14021, Prague, Czech Republic
| | - Oleg Lunov
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18200, Prague, Czech Republic.
| |
Collapse
|
37
|
Nwokoye PN, Abilez OJ. Bioengineering methods for vascularizing organoids. CELL REPORTS METHODS 2024; 4:100779. [PMID: 38759654 PMCID: PMC11228284 DOI: 10.1016/j.crmeth.2024.100779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 03/01/2024] [Accepted: 04/24/2024] [Indexed: 05/19/2024]
Abstract
Organoids, self-organizing three-dimensional (3D) structures derived from stem cells, offer unique advantages for studying organ development, modeling diseases, and screening potential therapeutics. However, their translational potential and ability to mimic complex in vivo functions are often hindered by the lack of an integrated vascular network. To address this critical limitation, bioengineering strategies are rapidly advancing to enable efficient vascularization of organoids. These methods encompass co-culturing organoids with various vascular cell types, co-culturing lineage-specific organoids with vascular organoids, co-differentiating stem cells into organ-specific and vascular lineages, using organoid-on-a-chip technology to integrate perfusable vasculature within organoids, and using 3D bioprinting to also create perfusable organoids. This review explores the field of organoid vascularization, examining the biological principles that inform bioengineering approaches. Additionally, this review envisions how the converging disciplines of stem cell biology, biomaterials, and advanced fabrication technologies will propel the creation of increasingly sophisticated organoid models, ultimately accelerating biomedical discoveries and innovations.
Collapse
Affiliation(s)
- Peter N Nwokoye
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Oscar J Abilez
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA; Division of Pediatric CT Surgery, Stanford University, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA; Maternal and Child Health Research Institute, Stanford University, Stanford, CA 94305, USA; Bio-X Program, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
38
|
Kim R, Sung JH. Microfluidic gut-axis-on-a-chip models for pharmacokinetic-based disease models. BIOMICROFLUIDICS 2024; 18:031507. [PMID: 38947281 PMCID: PMC11210976 DOI: 10.1063/5.0206271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/05/2024] [Indexed: 07/02/2024]
Abstract
The low success rate of new drugs transitioning from animal testing to human clinical trials necessitates the development of more accurate and representative in vitro models. Recent advances in multi-organ-on-a-chip technology offer promising avenues for studying complex organ-organ interactions. Gut-liver-on-a-chip systems hold particular promise for mimicking the intricate interplay between the gut and liver, which play crucial roles in nutrient absorption, drug metabolism, detoxification, and immune response. Here, we discuss the key components of the gut-liver axis, including the gut epithelium, liver cells, gut microbiota, and their roles in the organ functions. We then explore the potential of gut-liver-on-a-chip models to replicate the intricate interactions between the two organs for pharmacokinetic studies and their expansion to more complicated multi-organ models. Finally, we provide perspectives and future directions for developing more physiologically relevant gut-liver-axis models for more efficient drug development, studying liver diseases, and personalizing treatment strategies.
Collapse
Affiliation(s)
- Raehyun Kim
- Department of Biological and Chemical Engineering, Hongik University, Sejong 30016, Republic of Korea
| | - Jong Hwan Sung
- Department of Chemical Engineering, Hongik University, Seoul 04066, Republic of Korea
| |
Collapse
|
39
|
Somova M, Simm S, Padmyastuti A, Ehrhardt J, Schoon J, Wolff I, Burchardt M, Roennau C, Pinto PC. Integrating tumor and healthy epithelium in a micro-physiology multi-compartment approach to study renal cell carcinoma pathophysiology. Sci Rep 2024; 14:9357. [PMID: 38653823 PMCID: PMC11039668 DOI: 10.1038/s41598-024-60164-w] [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: 10/09/2023] [Accepted: 04/19/2024] [Indexed: 04/25/2024] Open
Abstract
The advent of micro-physiological systems (MPS) in biomedical research has enabled the introduction of more complex and relevant physiological into in vitro models. The recreation of complex morphological features in three-dimensional environments can recapitulate otherwise absent dynamic interactions in conventional models. In this study we developed an advanced in vitro Renal Cell Carcinoma (RCC) that mimics the interplay between healthy and malignant renal tissue. Based on the TissUse Humimic platform our model combines healthy renal proximal tubule epithelial cells (RPTEC) and RCC. Co-culturing reconstructed RPTEC tubules with RCC spheroids in a closed micro-perfused circuit resulted in significant phenotypical changes to the tubules. Expression of immune factors revealed that interleukin-8 (IL-8) and tumor necrosis factor-alfa (TNF-α) were upregulated in the non-malignant cells while neutrophil gelatinase-associated lipocalin (NGAL) was downregulated in both RCC and RPTEC. Metabolic analysis showed that RCC prompted a shift in the energy production of RPTEC tubules, inducing glycolysis, in a metabolic adaptation that likely supports RCC growth and immunogenicity. In contrast, RCC maintained stable metabolic activity, emphasizing their resilience to external factors. RNA-seq and biological process analysis of primary RTPTEC tubules demonstrated that the 3D tubular architecture and MPS conditions reverted cells to a predominant oxidative phosphorylate state, a departure from the glycolytic metabolism observed in 2D culture. This dynamic RCC co-culture model, approximates the physiology of healthy renal tubules to that of RCC, providing new insights into tumor-host interactions. Our approach can show that an RCC-MPS can expand the complexity and scope of pathophysiology and biomarker studies in kidney cancer research.
Collapse
Affiliation(s)
- Maryna Somova
- Department of Urology, University Medicine Greifswald, DZ7 J05.15, Fleischmannstraße 8, 17475, Greifswald, Germany
| | - Stefan Simm
- Institute of Bioinformatics, University Medicine Greifswald, Fleischmannstraße 8, 17475, Greifswald, Germany
- Institute for Bioanalysis, Coburg University of Applied Sciences and Arts, Friedrich-Streib-Str. 2, 96450, Coburg, Germany
| | - Adventina Padmyastuti
- Department of Urology, University Medicine Greifswald, DZ7 J05.15, Fleischmannstraße 8, 17475, Greifswald, Germany
| | - Jens Ehrhardt
- Department of Obstetrics and Gynecology, University Medicine Greifswald, Fleischmannstraße 8, 17475, Greifswald, Germany
| | - Janosch Schoon
- Center for Orthopaedics, Trauma Surgery and Rehabilitation Medicine, University Medicine Greifswald, Fleichmannstraße 8, 17475, Greifswald, Germany
| | - Ingmar Wolff
- Department of Urology, University Medicine Greifswald, DZ7 J05.15, Fleischmannstraße 8, 17475, Greifswald, Germany
| | - Martin Burchardt
- Department of Urology, University Medicine Greifswald, DZ7 J05.15, Fleischmannstraße 8, 17475, Greifswald, Germany
| | - Cindy Roennau
- Department of Urology, University Medicine Greifswald, DZ7 J05.15, Fleischmannstraße 8, 17475, Greifswald, Germany
| | - Pedro Caetano Pinto
- Department of Urology, University Medicine Greifswald, DZ7 J05.15, Fleischmannstraße 8, 17475, Greifswald, Germany.
| |
Collapse
|
40
|
Ponmozhi J, Dhinakaran S, Kocsis D, Iván K, Erdő F. Models for barrier understanding in health and disease in lab-on-a-chips. Tissue Barriers 2024; 12:2221632. [PMID: 37294075 PMCID: PMC11042069 DOI: 10.1080/21688370.2023.2221632] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/31/2023] [Indexed: 06/10/2023] Open
Abstract
The maintenance of body homeostasis relies heavily on physiological barriers. Dysfunction of these barriers can lead to various pathological processes, including increased exposure to toxic materials and microorganisms. Various methods exist to investigate barrier function in vivo and in vitro. To investigate barrier function in a highly reproducible manner, ethically, and high throughput, researchers have turned to non-animal techniques and micro-scale technologies. In this comprehensive review, the authors summarize the current applications of organ-on-a-chip microfluidic devices in the study of physiological barriers. The review covers the blood-brain barrier, ocular barriers, dermal barrier, respiratory barriers, intestinal, hepatobiliary, and renal/bladder barriers under both healthy and pathological conditions. The article then briefly presents placental/vaginal, and tumour/multi-organ barriers in organ-on-a-chip devices. Finally, the review discusses Computational Fluid Dynamics in microfluidic systems that integrate biological barriers. This article provides a concise yet informative overview of the current state-of-the-art in barrier studies using microfluidic devices.
Collapse
Affiliation(s)
- J. Ponmozhi
- Microfluidics Laboratory, Department of Mechanical Engineering, IPS Academy-Institute of Engineering Science, Indore, India
| | - S. Dhinakaran
- The Centre for Fluid Dynamics, Department of Mechanical Engineering, Indian Institute of Technology Indore, Indore, India
| | - Dorottya Kocsis
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Kristóf Iván
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Franciska Erdő
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| |
Collapse
|
41
|
Reyes DR, Esch MB, Ewart L, Nasiri R, Herland A, Sung K, Piergiovanni M, Lucchesi C, Shoemaker JT, Vukasinovic J, Nakae H, Hickman J, Pant K, Taylor A, Heinz N, Ashammakhi N. From animal testing to in vitro systems: advancing standardization in microphysiological systems. LAB ON A CHIP 2024; 24:1076-1087. [PMID: 38372151 DOI: 10.1039/d3lc00994g] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Limitations with cell cultures and experimental animal-based studies have had the scientific and industrial communities searching for new approaches that can provide reliable human models for applications such as drug development, toxicological assessment, and in vitro pre-clinical evaluation. This has resulted in the development of microfluidic-based cultures that may better represent organs and organ systems in vivo than conventional monolayer cell cultures. Although there is considerable interest from industry and regulatory bodies in this technology, several challenges need to be addressed for it to reach its full potential. Among those is a lack of guidelines and standards. Therefore, a multidisciplinary team of stakeholders was formed, with members from the US Food and Drug Administration (FDA), the National Institute of Standards and Technology (NIST), European Union, academia, and industry, to provide a framework for future development of guidelines/standards governing engineering concepts of organ-on-a-chip models. The result of this work is presented here for interested parties, stakeholders, and other standards development organizations (SDOs) to foster further discussion and enhance the impact and benefits of these efforts.
Collapse
Affiliation(s)
- Darwin R Reyes
- National Institute of Standards and Technology (NIST), Gaithersburg, MD, USA.
| | - Mandy B Esch
- National Institute of Standards and Technology (NIST), Gaithersburg, MD, USA.
| | | | | | - Anna Herland
- Royal Institute of Technology, Stockholm, Sweden
| | - Kyung Sung
- Food and Drug Administration (FDA), Silver Spring, Maryland, USA
| | | | | | | | | | - Hiroki Nakae
- JMAC Japan bio Measurement & Analysis Consortium, Tokyo, Japan
| | | | | | - Anne Taylor
- Xona Microfluidics, Inc., Research Triangle Park, North Carolina, USA
| | - Niki Heinz
- Altis Biosystems, Inc., Durham, North Carolina, USA
| | - Nureddin Ashammakhi
- Institute for Quantitative Health Science and Engineering, Department of Biomedical Engineering, College of Engineering, and College of Human Medicine, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
42
|
Wheeler AE, Stoeger V, Owens RM. Lab-on-chip technologies for exploring the gut-immune axis in metabolic disease. LAB ON A CHIP 2024; 24:1266-1292. [PMID: 38226866 DOI: 10.1039/d3lc00877k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
The continued rise in metabolic diseases such as obesity and type 2 diabetes mellitus poses a global health burden, necessitating further research into factors implicated in the onset and progression of these diseases. Recently, the gut-immune axis, with diet as a main regulator, has been identified as a possible role player in their development. Translation of conventional 2D in vitro and animal models is however limited, while human studies are expensive and preclude individual mechanisms from being investigated. Lab-on-chip technology therefore offers an attractive new avenue to study gut-immune interactions. This review provides an overview of the influence of diet on gut-immune interactions in metabolic diseases and a critical analysis of the current state of lab-on-chip technology to study this axis. While there has been progress in the development of "immuno-competent" intestinal lab-on-chip models, with studies showing the ability of the technology to provide mechanical cues, support longer-term co-culture of microbiota and maintain in vivo-like oxygen gradients, platforms which combine all three and include intestinal and immune cells are still lacking. Further, immune cell types and inclusion of microenvironment conditions which enable in vivo-like immune cell dynamics as well as host-microbiome interactions are limited. Future model development should focus on combining these conditions to create an environment capable of hosting more complex microbiota and immune cells to allow further study into the effects of diet and related metabolites on the gut-immune ecosystem and their role in the prevention and development of metabolic diseases in humans.
Collapse
Affiliation(s)
- Alexandra E Wheeler
- Department of Chemical Engineering and Biotechnology, University of Cambridge, UK.
| | - Verena Stoeger
- Department of Chemical Engineering and Biotechnology, University of Cambridge, UK.
| | - Róisín M Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, UK.
| |
Collapse
|
43
|
Baker TK, Van Vleet TR, Mahalingaiah PK, Grandhi TSP, Evers R, Ekert J, Gosset JR, Chacko SA, Kopec AK. The Current Status and Use of Microphysiological Systems by the Pharmaceutical Industry: The International Consortium for Innovation and Quality Microphysiological Systems Affiliate Survey and Commentary. Drug Metab Dispos 2024; 52:198-209. [PMID: 38123948 DOI: 10.1124/dmd.123.001510] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 11/21/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023] Open
Abstract
Microphysiological systems (MPS) are comprised of one or multiple cell types of human or animal origins that mimic the biochemical/electrical/mechanical responses and blood-tissue barrier properties of the cells observed within a complex organ. The goal of incorporating these in vitro systems is to expedite and advance the drug discovery and development paradigm with improved predictive and translational capabilities. Considering the industry need for improved efficiency and the broad challenges of model qualification and acceptance, the International Consortium for Innovation and Quality (IQ) founded an IQ MPS working group in 2014 and Affiliate in 2018. This group connects thought leaders and end users, provides a forum for crosspharma collaboration, and engages with regulators to qualify translationally relevant MPS models. To understand how pharmaceutical companies are using MPS, the IQ MPS Affiliate conducted two surveys in 2019, survey 1, and 2021, survey 2, which differed slightly in the scope of definition of the complex in vitro models under question. The surveys captured demographics, resourcing, rank order for organs of interest, compound modalities tested, and MPS organ-specific questions, including nonclinical species needs and cell types. The major focus of this manuscript is on results from survey 2, where we specifically highlight the context of use for MPS within safety, pharmacology, or absorption, disposition, metabolism, and excretion and discuss considerations for including MPS data in regulatory submissions. In summary, these data provide valuable insights for developers, regulators, and pharma, offering a view into current industry practices and future considerations while highlighting key challenges impacting MPS adoption. SIGNIFICANCE STATEMENT: The application of microphysiological systems (MPS) represents a growing area of interest in the drug discovery and development framework. This study surveyed 20+ pharma companies to understand resourcing, current areas of application, and the key challenges and barriers to internal MPS adoption. These results will provide regulators, tech providers, and pharma industry leaders a starting point to assess the current state of MPS applications along with key learnings to effectively realize the potential of MPS as an emerging technology.
Collapse
Affiliation(s)
- Thomas K Baker
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.) baker_thomas_k@lilly
| | - Terry R Van Vleet
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.)
| | - Prathap Kumar Mahalingaiah
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.)
| | - Taraka Sai Pavan Grandhi
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.)
| | - Raymond Evers
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.)
| | - Jason Ekert
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.)
| | - James R Gosset
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.)
| | - Silvi A Chacko
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.)
| | - Anna K Kopec
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.)
| |
Collapse
|
44
|
Tawade P, Mastrangeli M. Integrated Electrochemical and Optical Biosensing in Organs-on-Chip. Chembiochem 2024; 25:e202300560. [PMID: 37966365 DOI: 10.1002/cbic.202300560] [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: 08/09/2023] [Revised: 11/13/2023] [Accepted: 11/13/2023] [Indexed: 11/16/2023]
Abstract
Demand for biocompatible, non-invasive, and continuous real-time monitoring of organs-on-chip has driven the development of a variety of novel sensors. However, highest accuracy and sensitivity can arguably be achieved by integrated biosensing, which enables in situ monitoring of the in vitro microenvironment and dynamic responses of tissues and miniature organs recapitulated in organs-on-chip. This paper reviews integrated electrical, electrochemical, and optical sensing methods within organ-on-chip devices and platforms. By affording precise detection of analytes and biochemical reactions, these methods expand and advance the monitoring capabilities and reproducibility of organ-on-chip technology. The integration of these sensing techniques allows a deeper understanding of organ functions, and paves the way for important applications such as drug testing, disease modeling, and personalized medicine. By consolidating recent advancements and highlighting challenges in the field, this review aims to foster further research and innovation in the integration of biosensing in organs-on-chip.
Collapse
Affiliation(s)
- Pratik Tawade
- Electronic Components, Technology and Materials, Department of Microelectronics, Delft University of Technology, Mekelweg 4, 2628CD, Delft, Netherlands
| | - Massimo Mastrangeli
- Electronic Components, Technology and Materials, Department of Microelectronics, Delft University of Technology, Mekelweg 4, 2628CD, Delft, Netherlands
| |
Collapse
|
45
|
Chen X, Li M, Wang Z, Zhao K, Gu J, Li Q, He JJ. A Label-Free Optical Biosensor Based on an Array of Microring Resonators for the Detection of Human Serum Albumin. SENSORS (BASEL, SWITZERLAND) 2024; 24:677. [PMID: 38276369 PMCID: PMC10818899 DOI: 10.3390/s24020677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/03/2024] [Accepted: 01/16/2024] [Indexed: 01/27/2024]
Abstract
We introduced a label-free sensing system based on an array of microring resonators (MRRs) which was successfully employed for human serum albumin (HSA) detection. The sensing-ring surface was functionalized to immobilize anti-HSA, facilitating HSA binding. Our refractive index sensing system demonstrates high sensitivity at 168 nm/RIU and a low limit of detection (LOD) of 63.54 ng/mL, closely comparable to current HSA detection methods. These findings confirm the potential of MRRs as biocompatible sensors for HSA detection. This system holds great promise as an innovative platform for the detection of HSA, carrying significant importance in medical diagnostics.
Collapse
Affiliation(s)
- Xin Chen
- Department of Optical Engineering, School of Opto-Electronic Engineering, Changchun University of Science and Technology, Changchun 130022, China; (X.C.); (Z.W.); (K.Z.)
| | - Mingyu Li
- Department of Optical Engineering, School of Opto-Electronic Engineering, Changchun University of Science and Technology, Changchun 130022, China; (X.C.); (Z.W.); (K.Z.)
| | - Zhaoyu Wang
- Department of Optical Engineering, School of Opto-Electronic Engineering, Changchun University of Science and Technology, Changchun 130022, China; (X.C.); (Z.W.); (K.Z.)
| | - Kaihao Zhao
- Department of Optical Engineering, School of Opto-Electronic Engineering, Changchun University of Science and Technology, Changchun 130022, China; (X.C.); (Z.W.); (K.Z.)
| | - Jiamei Gu
- State Key Laboratory of Modern Optical Instrumentation, College of Optical Science and Engineering, Zhejiang University, Hangzhou 310027, China; (J.G.); (J.-J.H.)
| | - Qiushun Li
- Dezhou Research Institute, Qilu University of Technology (Shandong Academy of Sciences), Dezhou 253084, China
| | - Jian-Jun He
- State Key Laboratory of Modern Optical Instrumentation, College of Optical Science and Engineering, Zhejiang University, Hangzhou 310027, China; (J.G.); (J.-J.H.)
| |
Collapse
|
46
|
Zhou Z, Pang Y, Ji J, He J, Liu T, Ouyang L, Zhang W, Zhang XL, Zhang ZG, Zhang K, Sun W. Harnessing 3D in vitro systems to model immune responses to solid tumours: a step towards improving and creating personalized immunotherapies. Nat Rev Immunol 2024; 24:18-32. [PMID: 37402992 DOI: 10.1038/s41577-023-00896-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2023] [Indexed: 07/06/2023]
Abstract
In vitro 3D models are advanced biological tools that have been established to overcome the shortcomings of oversimplified 2D cultures and mouse models. Various in vitro 3D immuno-oncology models have been developed to mimic and recapitulate the cancer-immunity cycle, evaluate immunotherapy regimens, and explore options for optimizing current immunotherapies, including for individual patient tumours. Here, we review recent developments in this field. We focus, first, on the limitations of existing immunotherapies for solid tumours, secondly, on how in vitro 3D immuno-oncology models are established using various technologies - including scaffolds, organoids, microfluidics and 3D bioprinting - and thirdly, on the applications of these 3D models for comprehending the cancer-immunity cycle as well as for assessing and improving immunotherapies for solid tumours.
Collapse
Affiliation(s)
- Zhenzhen Zhou
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Haidian District, Beijing, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, China
- Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Beijing, China
| | - Yuan Pang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Haidian District, Beijing, China.
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, China.
- Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Beijing, China.
| | - Jingyuan Ji
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Haidian District, Beijing, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, China
- Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Beijing, China
| | - Jianyu He
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Haidian District, Beijing, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, China
- Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Beijing, China
| | - Tiankun Liu
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Haidian District, Beijing, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, China
- Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Beijing, China
| | - Liliang Ouyang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Haidian District, Beijing, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, China
- Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Beijing, China
| | - Wen Zhang
- Department of Immunology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District, Beijing, China
| | - Xue-Li Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhi-Gang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kaitai Zhang
- State Key Laboratory of Molecular Oncology, Department of Aetiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District, Beijing, China
| | - Wei Sun
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Haidian District, Beijing, China.
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, China.
- Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Beijing, China.
- Department of Mechanical Engineering, Drexel University, Philadelphia, PA, USA.
| |
Collapse
|
47
|
Gupta B, Malviya R, Srivastava S, Ahmad I, Rab SO, Uniyal P. Construction, Features and Regulatory Aspects of Organ-chip for Drug Delivery Applications: Advances and Prospective. Curr Pharm Des 2024; 30:1952-1965. [PMID: 38859792 DOI: 10.2174/0113816128305296240523112043] [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: 01/18/2024] [Accepted: 04/25/2024] [Indexed: 06/12/2024]
Abstract
Organ-on-chip is an innovative technique that emerged from tissue engineering and microfluidic technologies. Organ-on-chip devices (OoCs) are anticipated to provide efficient explanations for dealing with challenges in pharmaceutical advancement and individualized illness therapies. Organ-on-chip is an advanced method that can replicate human organs' physiological conditions and functions on a small chip. It possesses the capacity to greatly transform the drug development process by enabling the simulation of diseases and the testing of drugs. Effective integration of this advanced technical platform with common pharmaceutical and medical contexts is still a challenge. Microfluidic technology, a micro-level technique, has become a potent tool for biomedical engineering research. As a result, it has revolutionized disciplines, including physiological material interpreting, compound detection, cell-based assay, tissue engineering, biological diagnostics, and pharmaceutical identification. This article aims to offer an overview of newly developed organ-on-a-chip systems. It includes single-organ platforms, emphasizing the most researched organs, including the heart, liver, blood arteries, and lungs. Subsequently, it provides a concise overview of tumor-on-a-chip systems and emphasizes their use in evaluating anti-cancer medications.
Collapse
Affiliation(s)
- Babita Gupta
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, U.P., India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, U.P., India
| | - Saurabh Srivastava
- School of Pharmacy, KPJ Healthcare University College (KPJUC), Nilai, Malaysia
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Prerna Uniyal
- School of Pharmacy, Graphic Era Hill University, Dehradun, India
| |
Collapse
|
48
|
Zhang S, Xu G, Wu J, Liu X, Fan Y, Chen J, Wallace G, Gu Q. Microphysiological Constructs and Systems: Biofabrication Tactics, Biomimetic Evaluation Approaches, and Biomedical Applications. SMALL METHODS 2024; 8:e2300685. [PMID: 37798902 DOI: 10.1002/smtd.202300685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/23/2023] [Indexed: 10/07/2023]
Abstract
In recent decades, microphysiological constructs and systems (MPCs and MPSs) have undergone significant development, ranging from self-organized organoids to high-throughput organ-on-a-chip platforms. Advances in biomaterials, bioinks, 3D bioprinting, micro/nanofabrication, and sensor technologies have contributed to diverse and innovative biofabrication tactics. MPCs and MPSs, particularly tissue chips relevant to absorption, distribution, metabolism, excretion, and toxicity, have demonstrated potential as precise, efficient, and economical alternatives to animal models for drug discovery and personalized medicine. However, current approaches mainly focus on the in vitro recapitulation of the human anatomical structure and physiological-biochemical indices at a single or a few simple levels. This review highlights the recent remarkable progress in MPC and MPS models and their applications. The challenges that must be addressed to assess the reliability, quantify the techniques, and utilize the fidelity of the models are also discussed.
Collapse
Affiliation(s)
- Shuyu Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, China
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine/Department of Fetal Medicine and Prenatal Diagnosis/BioResource Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Guoshi Xu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chaoyang District, Beijing, 100101, China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 100049, China
| | - Juan Wu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chaoyang District, Beijing, 100101, China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 100049, China
| | - Xiao Liu
- Department of Gastroenterology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yong Fan
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine/Department of Fetal Medicine and Prenatal Diagnosis/BioResource Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Jun Chen
- Intelligent Polymer Research Institute, Australian Institute for Innovative Materials, Innovation Campus, University of Wollongong, North Wollongong, NSW, 2500, Australia
| | - Gordon Wallace
- Intelligent Polymer Research Institute, Australian Institute for Innovative Materials, Innovation Campus, University of Wollongong, North Wollongong, NSW, 2500, Australia
| | - Qi Gu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chaoyang District, Beijing, 100101, China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 100049, China
| |
Collapse
|
49
|
Abdalkader RK, Fujita T. Corneal epithelium models for safety assessment in drug development: Present and future directions. Exp Eye Res 2023; 237:109697. [PMID: 37890755 DOI: 10.1016/j.exer.2023.109697] [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: 06/30/2022] [Revised: 10/18/2023] [Accepted: 10/24/2023] [Indexed: 10/29/2023]
Abstract
The human corneal epithelial barrier plays a crucial role in drug testing studies, including drug absorption, distribution, metabolism, and excretion (ADME), as well as toxicity testing during the preclinical stages of drug development. However, despite the valuable insights gained from animal and current in vitro models, there remains a significant discrepancy between preclinical drug predictions and actual clinical outcomes. Additionally, there is a growing emphasis on adhering to the 3R principles (refine, reduce, replace) to minimize the use of animals in testing. To tackle these challenges, there is a rising demand for alternative in vitro models that closely mimic the human corneal epithelium. Recently, remarkable advancements have been made in two key areas: microphysiological systems (MPS) or organs-on-chips (OoCs), and stem cell-derived organoids. These cutting-edge platforms integrate four major disciplines: stem cells, microfluidics, bioprinting, and biosensing technologies. This integration holds great promise in developing powerful and biomimetic models of the human cornea.
Collapse
Affiliation(s)
- Rodi Kado Abdalkader
- Ritsumeikan Global Innovation Research Organization (R-GIRO), Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan.
| | - Takuya Fujita
- Ritsumeikan Global Innovation Research Organization (R-GIRO), Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan; Department of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan
| |
Collapse
|
50
|
Borenstein JT, Cummins G, Dutta A, Hamad E, Hughes MP, Jiang X, Lee HH, Lei KF, Tang XS, Zheng Y, Chen J. Bionanotechnology and bioMEMS (BNM): state-of-the-art applications, opportunities, and challenges. LAB ON A CHIP 2023; 23:4928-4949. [PMID: 37916434 DOI: 10.1039/d3lc00296a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
The development of micro- and nanotechnology for biomedical applications has defined the cutting edge of medical technology for over three decades, as advancements in fabrication technology developed originally in the semiconductor industry have been applied to solving ever-more complex problems in medicine and biology. These technologies are ideally suited to interfacing with life sciences, since they are on the scale lengths as cells (microns) and biomacromolecules (nanometers). In this paper, we review the state of the art in bionanotechnology and bioMEMS (collectively BNM), including developments and challenges in the areas of BNM, such as microfluidic organ-on-chip devices, oral drug delivery, emerging technologies for managing infectious diseases, 3D printed microfluidic devices, AC electrokinetics, flexible MEMS devices, implantable microdevices, paper-based microfluidic platforms for cellular analysis, and wearable sensors for point-of-care testing.
Collapse
Affiliation(s)
| | - Gerard Cummins
- School of Engineering, University of Birmingham, Edgbaston, B15 2TT, UK.
| | - Abhishek Dutta
- Department of Electrical & Computer Engineering, University of Connecticut, USA.
| | - Eyad Hamad
- Biomedical Engineering Department, School of Applied Medical Sciences, German Jordanian University, Amman, Jordan.
| | - Michael Pycraft Hughes
- Department of Biomedical Engineering, Khalifa University, Abu Dhabi, United Arab Emirates.
| | - Xingyu Jiang
- Department of Biomedical Engineering, Southern University of Science and Technology, China.
| | - Hyowon Hugh Lee
- Weldon School of Biomedical Engineering, Center for Implantable Devices, Purdue University, West Lafayette, IN, USA.
| | | | | | | | - Jie Chen
- Department of Electrical and Computer Engineering, University of Alberta, Edmonton, AB T6G 2R3, Canada.
| |
Collapse
|