1
|
Balls M, Bailey J. Ethics and Controversies in Animal Subjects Research and Impact on Clinical Decision-Making. Anesthesiol Clin 2024; 42:593-606. [PMID: 39443032 DOI: 10.1016/j.anclin.2024.03.003] [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: 10/25/2024]
Abstract
The harm inflicted on animals in biomedical research is justified, by those who accept it, by weighing it against proposed benefits for humans, but these are underappreciated and overstated, respectively. Clinical translation is poor due to interspecies and intraspecies differences, hindering the understanding of diseases and the development of safe and effective new human drugs. An urgent shift away from animal use toward human-focused new approach methodologies would enable human diversity to be factored more effectively into clinical practice. From an ethical perspective, more needs to be done to facilitate and encourage biomedical research to have a human focus throughout.
Collapse
Affiliation(s)
- Michael Balls
- Faculty of Medicine and Health Sciences, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK.
| | - Jarrod Bailey
- Research and Regulatory Affairs, Physicians Committee for Responsible Medicine (PCRM), 5100 Wisconsin Avenue, Northwest, Suite 400, Washington, DC 20016-4131, USA
| |
Collapse
|
2
|
Shrestha S, Acharya P, Kang SY, Vanga MG, Lekkala VKR, Liu J, Yang Y, Joshi P, Lee MY. Regenerative human liver organoids (HLOs) in a pillar/perfusion plate for hepatotoxicity assays. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.25.586638. [PMID: 38586058 PMCID: PMC10996672 DOI: 10.1101/2024.03.25.586638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Human liver organoids (HLOs) differentiated from embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), and adult stem cells (ASCs) can recapitulate the structure and function of human fetal liver tissues, thus being considered as a promising tissue model for liver diseases and predictive compound screening. However, the adoption of HLOs in drug discovery faces several technical challenges, which include the lengthy differentiation process with multiple culture media leading to batch-to-batch variation, short-term maintenance of hepatic functions post-maturation, low assay throughput due to Matrigel dissociation and HLO transfer to a microtiter well plate, and insufficient maturity levels compared to primary hepatocytes. To address these issues, expandable HLOs (Exp-HLOs) derived from human iPSCs were generated by optimizing differentiation protocols, which were rapidly printed on a 144-pillar plate with sidewalls and slits (144PillarPlate) and dynamically cultured for up to 20 days into differentiated HLOs (Diff-HLOs) in a 144-perfusion plate with perfusion wells and reservoirs (144PerfusionPlate) for in situ organoid culture and analysis. The dynamically cultured Diff-HLOs exhibited greater maturity and reproducibility than those cultured statically, especially after a 10-day differentiation period. In addition, Diff-HLOs in the pillar/perfusion plate were tested with acetaminophen and troglitazone for 3 days to assess drug-induced liver injury (DILI) and then incubated in an expansion medium for 10 days to evaluate liver recovery from DILI. The assessment of liver regeneration post-injury is critical to understanding the mechanism of recovery and determining the threshold drug concentration beyond which there will be a sharp decrease in the liver's regenerative capacity. We envision that bioprinted Diff-HLOs in the pillar/perfusion plate could be used for high-throughput screening (HTS) of hepatotoxic compounds due to the short-term differentiation of passage-able Exp-HLOs, stable hepatic function post-maturation, high reproducibility, and high throughput with capability of in situ organoid culture, testing, staining, imaging, and analysis.
Collapse
|
3
|
Rafsanjani Nejad P, Lamichhane A, Guragain P, Luker G, Tavana H. A gravity-driven tissue chip to study the efficacy and toxicity of cancer therapeutics. LAB ON A CHIP 2024. [PMID: 39485368 PMCID: PMC11529822 DOI: 10.1039/d4lc00404c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 10/23/2024] [Indexed: 11/03/2024]
Abstract
Tissue chip and organs-on-chip technologies have emerged as promising tools in preclinical studies. In oncology, this is driven by the high failure rates of candidate drugs in clinical trials mainly due to inadequate efficacy or intolerable toxicity and the need for better predictive preclinical models than those traditionally used. However, the intricate design, fabrication, operation, and limited compatibility with automation limit the utility of tissue chips. To tackle these issues, we designed a novel 32-unit tissue chip in the format of standard 96-well plates to streamline automation, fabricated it using 3D printing, and leveraged gravity-driven flow to bypass the need for external flow devices. Each unit includes three interconnected tissue compartments that model liver, tumor, and bone marrow stroma. The focus on liver and bone marrow stroma was due to their respective roles in drug metabolism and disturbances to the bone marrow niche from off-target toxicity of chemotherapies. We analyzed flow patterns, mixing, and oxygen transport among and within the compartments through finite element simulations and demonstrated the utility of the tissue chip to study the efficacy of commonly-used cytotoxic cancer drugs against tumor cells and their toxicity toward liver and bone marrow cells. The ability to simultaneously study drug efficacy and toxicity in high throughput can help select promising therapeutics in early stages of drug discovery in preclinical studies.
Collapse
Affiliation(s)
| | - Astha Lamichhane
- Department of Biomedical Engineering, University of Akron, Akron, OH, USA.
| | - Prasiddha Guragain
- Department of Biomedical Engineering, University of Akron, Akron, OH, USA.
| | - Gary Luker
- Departments of Radiology, Microbiology and Immunology, Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Hossein Tavana
- Department of Biomedical Engineering, University of Akron, Akron, OH, USA.
| |
Collapse
|
4
|
Dhummakupt ES, Jenkins CC, Rizzo GM, Clay AE, Horsmon JR, Goralski TDP, Renner JA, Angelini DJ. Multiomic analysis of Lewisite exposed human dermal equivalent tissues. Chem Biol Interact 2024; 405:111295. [PMID: 39486569 DOI: 10.1016/j.cbi.2024.111295] [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: 08/15/2024] [Revised: 10/11/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024]
Abstract
Lewisite (Military Code: L) is an arsenical vesicant chemical warfare agent (CWA) that was developed in the United States during World War I. Even though its use has not been documented in warfare, large stockpiles were created and still exist in various locations around the world. Given that large quantities exist as well as the relative straightforward process for its creation, Lewisite still presents itself as a serious threat agent. In this study, we examined the effects of Lewisite on human dermal equivalent tissues (EpiDerm™/EpiDerm™-FT) through the evaluation of cellular viability, histology, and multiomic analysis.
Collapse
Affiliation(s)
- Elizabeth S Dhummakupt
- U.S. Army, Combat Capabilities Development Command (DEVCOM) Chemical Biological Center, Aberdeen Proving Ground, MD, 21010, USA.
| | - Conor C Jenkins
- U.S. Army, Combat Capabilities Development Command (DEVCOM) Chemical Biological Center, Aberdeen Proving Ground, MD, 21010, USA
| | - Gabrielle M Rizzo
- U.S. Army, Combat Capabilities Development Command (DEVCOM) Chemical Biological Center, Aberdeen Proving Ground, MD, 21010, USA
| | - Allison E Clay
- U.S. Army, Combat Capabilities Development Command (DEVCOM) Chemical Biological Center, Aberdeen Proving Ground, MD, 21010, USA
| | - Jennifer R Horsmon
- U.S. Army, Combat Capabilities Development Command (DEVCOM) Chemical Biological Center, Aberdeen Proving Ground, MD, 21010, USA
| | - Tyler D P Goralski
- U.S. Army, Combat Capabilities Development Command (DEVCOM) Chemical Biological Center, Aberdeen Proving Ground, MD, 21010, USA
| | - Julie A Renner
- U.S. Army, Combat Capabilities Development Command (DEVCOM) Chemical Biological Center, Aberdeen Proving Ground, MD, 21010, USA
| | - Daniel J Angelini
- U.S. Army, Combat Capabilities Development Command (DEVCOM) Chemical Biological Center, Aberdeen Proving Ground, MD, 21010, USA.
| |
Collapse
|
5
|
Chim SM, Howell K, Kokkosis A, Zambrowicz B, Karalis K, Pavlopoulos E. A Human Brain-Chip for Modeling Brain Pathologies and Screening Blood-Brain Barrier Crossing Therapeutic Strategies. Pharmaceutics 2024; 16:1314. [PMID: 39458643 PMCID: PMC11510380 DOI: 10.3390/pharmaceutics16101314] [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: 08/12/2024] [Revised: 09/17/2024] [Accepted: 10/06/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: The limited translatability of preclinical experimental findings to patients remains an obstacle for successful treatment of brain diseases. Relevant models to elucidate mechanisms behind brain pathogenesis, including cell-specific contributions and cell-cell interactions, and support successful targeting and prediction of drug responses in humans are urgently needed, given the species differences in brain and blood-brain barrier (BBB) functions. Human microphysiological systems (MPS), such as Organ-Chips, are emerging as a promising approach to address these challenges. Here, we examined and advanced a Brain-Chip that recapitulates aspects of the human cortical parenchyma and the BBB in one model. Methods: We utilized human primary astrocytes and pericytes, human induced pluripotent stem cell (hiPSC)-derived cortical neurons, and hiPSC-derived brain microvascular endothelial-like cells and included for the first time on-chip hiPSC-derived microglia. Results: Using Tumor necrosis factor alpha (TNFα) to emulate neuroinflammation, we demonstrate that our model recapitulates in vivo-relevant responses. Importantly, we show microglia-derived responses, highlighting the Brain-Chip's sensitivity to capture cell-specific contributions in human disease-associated pathology. We then tested BBB crossing of human transferrin receptor antibodies and conjugated adeno-associated viruses. We demonstrate successful in vitro/in vivo correlation in identifying crossing differences, underscoring the model's capacity as a screening platform for BBB crossing therapeutic strategies and ability to predict in vivo responses. Conclusions: These findings highlight the potential of the Brain-Chip as a reliable and time-efficient model to support therapeutic development and provide mechanistic insights into brain diseases, adding to the growing evidence supporting the value of MPS in translational research and drug discovery.
Collapse
Affiliation(s)
- Shek Man Chim
- Human Systems, Regeneron Pharmaceuticals, Tarrytown, NY 10591, USA; (K.H.); (A.K.); (K.K.)
- Velocigene, Regeneron Pharmaceuticals, Tarrytown, NY 10591, USA;
| | - Kristen Howell
- Human Systems, Regeneron Pharmaceuticals, Tarrytown, NY 10591, USA; (K.H.); (A.K.); (K.K.)
- Velocigene, Regeneron Pharmaceuticals, Tarrytown, NY 10591, USA;
| | - Alexandros Kokkosis
- Human Systems, Regeneron Pharmaceuticals, Tarrytown, NY 10591, USA; (K.H.); (A.K.); (K.K.)
- Velocigene, Regeneron Pharmaceuticals, Tarrytown, NY 10591, USA;
| | - Brian Zambrowicz
- Velocigene, Regeneron Pharmaceuticals, Tarrytown, NY 10591, USA;
| | - Katia Karalis
- Human Systems, Regeneron Pharmaceuticals, Tarrytown, NY 10591, USA; (K.H.); (A.K.); (K.K.)
- Velocigene, Regeneron Pharmaceuticals, Tarrytown, NY 10591, USA;
| | - Elias Pavlopoulos
- Human Systems, Regeneron Pharmaceuticals, Tarrytown, NY 10591, USA; (K.H.); (A.K.); (K.K.)
- Velocigene, Regeneron Pharmaceuticals, Tarrytown, NY 10591, USA;
| |
Collapse
|
6
|
Haber LT, Bradley MA, Buerger AN, Behrsing H, Burla S, Clapp PW, Dotson S, Fisher C, Genco KR, Kruszewski FH, McCullough SD, Page KE, Patel V, Pechacek N, Roper C, Sharma M, Jarabek AM. New approach methodologies (NAMs) for the in vitro assessment of cleaning products for respiratory irritation: workshop report. FRONTIERS IN TOXICOLOGY 2024; 6:1431790. [PMID: 39439531 PMCID: PMC11493779 DOI: 10.3389/ftox.2024.1431790] [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: 05/12/2024] [Accepted: 09/19/2024] [Indexed: 10/25/2024] Open
Abstract
The use of in vitro new approach methodologies (NAMs) to assess respiratory irritation depends on several factors, including the specifics of exposure methods and cell/tissue-based test systems. This topic was examined in the context of human health risk assessment for cleaning products at a 1-day public workshop held on 2 March 2023, organized by the American Cleaning Institute® (ACI). The goals of this workshop were to (1) review in vitro NAMs for evaluation of respiratory irritation, (2) examine different perspectives on current challenges and suggested solutions, and (3) publish a manuscript of the proceedings. Targeted sessions focused on exposure methods, in vitro cell/tissue test systems, and application to human health risk assessment. The importance of characterization of assays and development of reporting standards was noted throughout the workshop. The exposure methods session emphasized that the appropriate exposure system design depends on the purpose of the assessment. This is particularly important given the many dosimetry and technical considerations affecting relevance and translation of results to human exposure scenarios. Discussion in the in vitro cell/tissue test systems session focused on the wide variety of cell systems with varying suitability for evaluating key mechanistic steps, such as molecular initiating events (MIEs) and key events (KEs) likely present in any putative respiratory irritation adverse outcome pathway (AOP). This suggests the opportunity to further develop guidance around in vitro cell/tissue test system endpoint selection, assay design, characterization and validation, and analytics that provide information about a given assay's utility. The session on applications for human health protection emphasized using mechanistic understanding to inform the choice of test systems and integration of NAMs-derived data with other data sources (e.g., physicochemical properties, exposure information, and existing in vivo data) as the basis for in vitro to in vivo extrapolation. In addition, this group noted a need to develop procedures to align NAMs-based points of departure (PODs) and uncertainty factor selection with current human health risk assessment methods, together with consideration of elements unique to in vitro data. Current approaches are described and priorities for future characterization of in vitro NAMs to assess respiratory irritation are noted.
Collapse
Affiliation(s)
- Lynne T. Haber
- Risk Science Center, Department of Environmental and Public Health Sciences, University of Cincinnati, Cincinnati, OH, United States
| | - Mark A. Bradley
- Risk Science Center, Department of Environmental and Public Health Sciences, University of Cincinnati, Cincinnati, OH, United States
| | | | - Holger Behrsing
- Institute for In Vitro Sciences, Inc., Gaithersburg, MD, United States
| | | | - Phillip W. Clapp
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, United States
| | - Scott Dotson
- Insight Exposure and Risk Sciences Group, Cincinnati, OH, United States
| | | | | | | | - Shaun D. McCullough
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, Office of Research and Development, U.S. EPA, Chapel Hill, NC, United States
| | | | - Vivek Patel
- Institute for In Vitro Sciences, Inc., Gaithersburg, MD, United States
| | | | - Clive Roper
- Roper Toxicology Consulting Limited, Edinburgh, United Kingdom
| | - Monita Sharma
- PETA Science Consortium International e.V, Stuttgart, Germany
| | - Annie M. Jarabek
- Health and Environmental Effects Assessment Division, Center for Public Health and Environmental Assessment, Office of Research and Development, U.S. EPA, Chapel Hill, NC, United States
| |
Collapse
|
7
|
Deceuninck P, Gastaldello A, Mennecozzi M, Pistollato F. Exploring the connection between EU-funded research and methodological approaches: insights from a retrospective analysis. J Transl Med 2024; 22:891. [PMID: 39363357 PMCID: PMC11447993 DOI: 10.1186/s12967-024-05557-1] [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: 07/04/2024] [Accepted: 07/29/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND Over the last two decades, substantial investments have been directed towards supporting fundamental and applied research in Alzheimer's disease (AD), breast cancer (BC), and prostate cancer (PC), which continue to pose significant health challenges. Recently, the Joint Research Centre (JRC) of the European Commission (EC) conducted a retrospective analysis to examine the major scientific advancements resulting from EU-funded research in these disease areas and their impact on society. METHODS Building upon this analysis, our subsequent investigation delves into the methodological approaches-both animal and non-animal models and methods-employed in AD, BC, and PC research funded under past EU framework programs (FP5, FP6, FP7, and H2020), and explored the notable research outputs associated with these approaches. RESULTS Our findings indicate a prevalent use of animal-based methodologies in AD research, particularly evident in projects funded under H2020. Notably, projects focused on drug development, testing, or repurposing heavily relied on animal models. Conversely, research aimed at clinical trial design, patient stratification, diagnosis and diagnostic tool development, lifestyle interventions, and prevention-outputs with potential societal impact-more frequently utilised non-animal methods. Advanced investigations leveraging imaging, computational tools, biomarker discovery and organ/tissue chip technologies predominantly favoured non-animal strategies. CONCLUSIONS These insights highlight a correlation between methodological choices and the translational potential of research outcomes, suggesting the need for a reconsideration of research strategy planning in future framework programs.
Collapse
|
8
|
Dey S, Bhat A, Janani G, Shandilya V, Gupta R, Mandal BB. Microfluidic human physiomimetic liver model as a screening platform for drug induced liver injury. Biomaterials 2024; 310:122627. [PMID: 38823194 DOI: 10.1016/j.biomaterials.2024.122627] [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: 01/18/2024] [Revised: 05/02/2024] [Accepted: 05/19/2024] [Indexed: 06/03/2024]
Abstract
The pre-clinical animal models often fail to predict intrinsic and idiosyncratic drug induced liver injury (DILI), thus contributing to drug failures in clinical trials, black box warnings and withdrawal of marketed drugs. This suggests a critical need for human-relevant in vitro models to predict diverse DILI phenotypes. In this study, a porcine liver extracellular matrix (ECM) based biomaterial ink with high printing fidelity, biocompatibility and tunable rheological and mechanical properties is formulated for supporting both parenchymal and non-parenchymal cells. Further, we applied 3D printing and microfluidic technology to bioengineer a human physiomimetic liver acinus model (HPLAM), recapitulating the radial hepatic cord-like structure with functional sinusoidal microvasculature network, biochemical and biophysical properties of native liver acinus. Intriguingly, the human derived hepatic cells incorporated HPLAM cultured under physiologically relevant microenvironment, acts as metabolic biofactories manifesting enhanced hepatic functionality, secretome levels and biomarkers expression over several weeks. We also report that the matured HPLAM reproduces dose- and time-dependent hepatotoxic response of human clinical relevance to drugs typically recognized for inducing diverse DILI phenotypes as compared to conventional static culture. Overall, the developed HPLAM emulates in vivo like functions and may provide a useful platform for DILI risk assessment to better determine safety and human risk.
Collapse
Affiliation(s)
- Souradeep Dey
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Amritha Bhat
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - G Janani
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Vartik Shandilya
- Department of Chemical Engineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Raghvendra Gupta
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India; Department of Chemical Engineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India; Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Biman B Mandal
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India; Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India; Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India.
| |
Collapse
|
9
|
Kiaris H. Nontraditional models as research tools: the road not taken. Trends Mol Med 2024; 30:924-931. [PMID: 39069395 PMCID: PMC11466687 DOI: 10.1016/j.molmed.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/07/2024] [Accepted: 07/10/2024] [Indexed: 07/30/2024]
Abstract
Historical reasons resulted in the almost exclusive use of a few species, most prominently Mus musculus, as the mainstream models in biomedical research. This selection was not based on Mus's distinctive relevance to human disease but rather to the pre-existing availability of resources and tools for the species that were used as models, which has enabled their adoption for research in health sciences. Unless the utilization and range of nontraditional research models expand considerably, progress in biomedical research will remain restricted within the trajectory that has been set by the existing models and their ability to provide clinically relevant information.
Collapse
Affiliation(s)
- Hippokratis Kiaris
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy and Peromyscus Genetic Stock Center, University of South Carolina, Columbia, SC, USA.
| |
Collapse
|
10
|
Szewczyk K, Jiang L, Khawaja H, Miranti CK, Zohar Y. Microfluidic Applications in Prostate Cancer Research. MICROMACHINES 2024; 15:1195. [PMID: 39459070 PMCID: PMC11509716 DOI: 10.3390/mi15101195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/13/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024]
Abstract
Prostate cancer is a disease in which cells in the prostate, a gland in the male reproductive system below the bladder, grow out of control and, among men, it is the second-most frequently diagnosed cancer (other than skin cancer). In recent years, prostate cancer death rate has stabilized and, currently, it is the second-most frequent cause of cancer death in men (after lung cancer). Most deaths occur due to metastasis, as cancer cells from the original tumor establish secondary tumors in distant organs. For a long time, classical cell cultures and animal models have been utilized in basic and applied scientific research, including clinical applications for many diseases, such as prostate cancer, since no better alternatives were available. Although helpful in dissecting cellular mechanisms, these models are poor predictors of physiological behavior mainly because of the lack of appropriate microenvironments. Microfluidics has emerged in the last two decades as a technology that could lead to a paradigm shift in life sciences and, in particular, controlling cancer. Microfluidic systems, such as organ-on-chips, have been assembled to mimic the critical functions of human organs. These microphysiological systems enable the long-term maintenance of cellular co-cultures in vitro to reconstitute in vivo tissue-level microenvironments, bridging the gap between traditional cell cultures and animal models. Several reviews on microfluidics for prostate cancer studies have been published focusing on technology advancement and disease progression. As metastatic castration-resistant prostate cancer remains a clinically challenging late-stage cancer, with no curative treatments, we expanded this review to cover recent microfluidic applications related to prostate cancer research. The review includes discussions of the roles of microfluidics in modeling the human prostate, prostate cancer initiation and development, as well as prostate cancer detection and therapy, highlighting potentially major contributions of microfluidics in the continuous march toward eradicating prostate cancer.
Collapse
Affiliation(s)
- Kailie Szewczyk
- Department of Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ 85721, USA; (K.S.); (L.J.)
| | - Linan Jiang
- Department of Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ 85721, USA; (K.S.); (L.J.)
| | - Hunain Khawaja
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ 85724, USA;
| | - Cindy K. Miranti
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA;
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA
| | - Yitshak Zohar
- Department of Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ 85721, USA; (K.S.); (L.J.)
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA
| |
Collapse
|
11
|
Benam KH. Multidisciplinary approaches in electronic nicotine delivery systems pulmonary toxicology: emergence of living and non-living bioinspired engineered systems. COMMUNICATIONS ENGINEERING 2024; 3:123. [PMID: 39227652 PMCID: PMC11372223 DOI: 10.1038/s44172-024-00276-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 08/27/2024] [Indexed: 09/05/2024]
Abstract
Technology-based platforms offer crucial support for regulatory agencies in overseeing tobacco products to enhance public health protection. The use of electronic nicotine delivery systems (ENDS), such as electronic cigarettes, has surged exponentially over the past decade. However, the understanding of the impact of ENDS on lung health remains incomplete due to scarcity of physiologically relevant technologies for evaluating their toxicity. This review examines the societal and public health impacts of ENDS, prevalent preclinical approaches in pulmonary space, and the application of emerging Organ-on-Chip technologies and bioinspired robotics for assessing ENDS respiratory toxicity. It highlights challenges in ENDS inhalation toxicology and the value of multidisciplinary bioengineering approaches for generating reliable, human-relevant regulatory data at an accelerated pace.
Collapse
Affiliation(s)
- Kambez H Benam
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
12
|
Walocha R, Kim M, Wong-Ng J, Gobaa S, Sauvonnet N. Organoids and organ-on-chip technology for investigating host-microorganism interactions. Microbes Infect 2024; 26:105319. [PMID: 38447861 DOI: 10.1016/j.micinf.2024.105319] [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: 10/03/2023] [Revised: 02/26/2024] [Accepted: 03/01/2024] [Indexed: 03/08/2024]
Abstract
Recent advances in organoid and organ-on-chip (OoC) technologies offer an unprecedented level of tissue mimicry. These models can recapitulate the diversity of cellular composition, 3D organization, and mechanical stimulation. These approaches are intensively used to understand complex diseases. This review focuses on the latest advances in this field to study host-microorganism interactions.
Collapse
Affiliation(s)
- Remigiusz Walocha
- Tissue Homeostasis Group, Biomaterials and Microfluidics Core Facility, Institut Pasteur, Université Paris Cité, Paris, France; Biomaterials and Microfluidics Core Facility, Institut Pasteur, Université Paris Cité, Paris, France
| | - MinHee Kim
- Biomaterials and Microfluidics Core Facility, Institut Pasteur, Université Paris Cité, Paris, France
| | - Jérôme Wong-Ng
- Biomaterials and Microfluidics Core Facility, Institut Pasteur, Université Paris Cité, Paris, France
| | - Samy Gobaa
- Biomaterials and Microfluidics Core Facility, Institut Pasteur, Université Paris Cité, Paris, France
| | - Nathalie Sauvonnet
- Tissue Homeostasis Group, Biomaterials and Microfluidics Core Facility, Institut Pasteur, Université Paris Cité, Paris, France; Biomaterials and Microfluidics Core Facility, Institut Pasteur, Université Paris Cité, Paris, France.
| |
Collapse
|
13
|
Yang JW, Khorsandi D, Trabucco L, Ahmed M, Khademhosseini A, Dokmeci MR, Ye JY, Jucaud V. Liver-on-a-Chip Integrated with Label-Free Optical Biosensors for Rapid and Continuous Monitoring of Drug-Induced Toxicity. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2403560. [PMID: 39212623 DOI: 10.1002/smll.202403560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/06/2024] [Indexed: 09/04/2024]
Abstract
Drug toxicity assays using conventional 2D static cultures and animal studies have limitations preventing the translation of potential drugs to the clinic. The recent development of organs-on-a-chip platforms provides promising alternatives for drug toxicity/screening assays. However, most studies conducted with these platforms only utilize single endpoint results, which do not provide real-time/ near real-time information. Here, a versatile technology is presented that integrates a 3D liver-on-a-chip with a label-free photonic crystal-total internal reflection (PC-TIR) biosensor for rapid and continuous monitoring of the status of cells. This technology can detect drug-induced liver toxicity by continuously monitoring the secretion rates and levels of albumin and glutathione S-transferase α (GST-α) of a 3D liver on-a-chip model treated with Doxorubicin. The PC-TIR biosensor is based on a one-step antibody functionalization with high specificity and a detection range of 21.7 ng mL-1 to 7.83 x 103 ng mL-1 for albumin and 2.20 ng mL-1 to 7.94 x 102 ng mL-1 for GST-α. This approach provides critical advantages for the early detection of drug toxicity and improved temporal resolution to capture transient drug effects. The proposed proof-of-concept study introduces a scalable and efficient plug-in solution for organ-on-a-chip technologies, advancing drug development and in vitro testing methods by enabling timely and accurate toxicity assessments.
Collapse
Affiliation(s)
- Jia-Wei Yang
- Terasaki Institute for Biomedical Innovation, 21100 Erwin St, Woodland Hills, CA, 91367, USA
| | - Danial Khorsandi
- Terasaki Institute for Biomedical Innovation, 21100 Erwin St, Woodland Hills, CA, 91367, USA
| | - Luis Trabucco
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA
| | - Maisha Ahmed
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, 21100 Erwin St, Woodland Hills, CA, 91367, USA
| | - Mehmet Remzi Dokmeci
- Terasaki Institute for Biomedical Innovation, 21100 Erwin St, Woodland Hills, CA, 91367, USA
| | - Jing Yong Ye
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, 21100 Erwin St, Woodland Hills, CA, 91367, USA
| |
Collapse
|
14
|
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
|
15
|
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:e2400833. [PMID: 39101627 DOI: 10.1002/adhm.202400833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 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 Engineering, American University of Sharjah, Sharjah, UAE
| | - Remya Radha
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah, UAE
| | - Shabana Anjum
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah, UAE
| | - Vinod Paul
- Materials Science and Engineering PhD program, College of Arts and Sciences, American University of Sharjah, Sharjah, UAE
| | - Nour AlSawaftah
- Materials Science and Engineering PhD program, College of Arts and Sciences, American University of Sharjah, Sharjah, UAE
| | - William G Pitt
- Department of Chemical Engineering, Brigham Young University, Provo, UT, 84602, USA
| | - Nureddin Ashammakhi
- Institute for Quantitative Health Science and Engineering (IQ) and Department of Biomedical Engineering (BME), Michigan State University, East Lansing, MI, 48824, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095-1600, USA
| | - Ghaleb A Husseini
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah, UAE
- Materials Science and Engineering PhD program, College of Arts and Sciences, American University of Sharjah, Sharjah, UAE
| |
Collapse
|
16
|
Pun S, Prakash A, Demaree D, Krummel DP, Sciumè G, Sengupta S, Barrile R. Rapid Biofabrication of an Advanced Microphysiological System Mimicking Phenotypical Heterogeneity and Drug Resistance in Glioblastoma. Adv Healthc Mater 2024:e2401876. [PMID: 39101329 DOI: 10.1002/adhm.202401876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/10/2024] [Indexed: 08/06/2024]
Abstract
Microphysiological systems (MPSs) reconstitute tissue interfaces and organ functions, presenting a promising alternative to animal models in drug development. However, traditional materials like polydimethylsiloxane (PDMS) often interfere by absorbing hydrophobic molecules, affecting drug testing accuracy. Additive manufacturing, including 3D bioprinting, offers viable solutions. GlioFlow3D, a novel microfluidic platform combining extrusion bioprinting and stereolithography (SLA) is introduced. GlioFlow3D integrates primary human cells and glioblastoma (GBM) lines in hydrogel-based microchannels mimicking vasculature, within an SLA resin framework using cost-effective materials. The study introduces a robust protocol to mitigate SLA resin cytotoxicity. Compared to PDMS, GlioFlow3D demonstrated lower small molecule absorption, which is relevant for accurate testing of small molecules like Temozolomide (TMZ). Computational modeling is used to optimize a pumpless setup simulating interstitial fluid flow dynamics in tissues. Co-culturing GBM with brain endothelial cells in GlioFlow3D showed enhanced CD133 expression and TMZ resistance near vascular interfaces, highlighting spatial drug resistance mechanisms. This PDMS-free platform promises advanced drug testing, improving preclinical research and personalized therapy by elucidating complex GBM drug resistance mechanisms influenced by the tissue microenvironment.
Collapse
Affiliation(s)
- Sirjana Pun
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, 45221, USA
| | - Anusha Prakash
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, 45221, USA
- Abbvie, Worcester, Massachusetts, 01605, USA
| | - Dalee Demaree
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, 45221, USA
- Thermo Fisher Scientific, Waltham, Massachusetts, 02451, USA
| | - Daniel Pomeranz Krummel
- Department of Neurology, University of Cincinnati, Cincinnati, OH, 45219, USA
- Department of Neurosurgery, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Giuseppe Sciumè
- Institute of Mechanics and Engineering-12 M, University of Bordeaux, Bordeaux, 33607, France
| | - Soma Sengupta
- Department of Neurology, University of Cincinnati, Cincinnati, OH, 45219, USA
- Department of Neurosurgery, University of North Carolina, Chapel Hill, NC, 27599, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, 27599-7025, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599-7295, USA
| | - Riccardo Barrile
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, 45221, USA
- Center for Stem Cells and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| |
Collapse
|
17
|
Pang PD, Ahmed SM, Nishiga M, Stockbridge NL, Wu JC. Tackling the challenges of new approach methods for predicting drug effects from model systems. Nat Rev Drug Discov 2024; 23:565-566. [PMID: 38750208 PMCID: PMC11482555 DOI: 10.1038/d41573-024-00081-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2024]
Abstract
The passage of the FDA Modernization Act 2.0 in 2022 has provided additional impetus to develop new approach methods for predicting the effects of drug candidates in humans from models such as microphysiological systems based on human-derived induced pluripotent stem cells. Here, we highlight progress in the field and strategies to address various challenges, including the application of artificial intelligence tools.
Collapse
|
18
|
Liu H, Yin G, Kohlhepp MS, Schumacher F, Hundertmark J, Hassan MIA, Heymann F, Puengel T, Kleuser B, Mosig AS, Tacke F, Guillot A. Dissecting Acute Drug-Induced Hepatotoxicity and Therapeutic Responses of Steatotic Liver Disease Using Primary Mouse Liver and Blood Cells in a Liver-On-A-Chip Model. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403516. [PMID: 38868948 PMCID: PMC11321671 DOI: 10.1002/advs.202403516] [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: 04/03/2024] [Revised: 04/19/2024] [Indexed: 06/14/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is hallmarked by hepatic steatosis, cell injury, inflammation, and fibrosis. This study elaborates on a multicellular biochip-based liver sinusoid model to mimic MASLD pathomechanisms and investigate the therapeutic effects of drug candidates lanifibranor and resmetirom. Mouse liver primary hepatocytes, hepatic stellate cells, Kupffer cells, and endothelial cells are seeded in a dual-chamber biocompatible liver-on-a-chip (LoC). The LoC is then perfused with circulating immune cells (CICs). Acetaminophen (APAP) and free fatty acids (FFAs) treatment recapitulate acute drug-induced liver injury and MASLD, respectively. As a benchmark for the LoC, multiplex immunofluorescence on livers from APAP-injected and dietary MASLD-induced mice reveals characteristic changes on parenchymal and immune cell populations. APAP exposure induces cell death in the LoC, and increased inflammatory cytokine levels in the circulating perfusate. Under FFA stimulation, lipid accumulation, cellular damage, inflammatory secretome, and fibrogenesis are increased in the LoC, reflecting MASLD. Both injury conditions potentiate CIC migration from the perfusate to the LoC cellular layers. Lanifibranor prevents the onset of inflammation, while resmetirom decreases lipid accumulation in hepatocytes and increases the generation of FFA metabolites in the LoC. This study demonstrates the LoC potential for functional and molecular evaluation of liver disease drug candidates.
Collapse
Affiliation(s)
- Hanyang Liu
- Department of Hepatology & GastroenterologyCampus Virchow‐Klinikum and Campus Charité MitteCharité – Universitätsmedizin Berlin13353BerlinGermany
| | - Guo Yin
- Department of Hepatology & GastroenterologyCampus Virchow‐Klinikum and Campus Charité MitteCharité – Universitätsmedizin Berlin13353BerlinGermany
| | - Marlene Sophia Kohlhepp
- Department of Hepatology & GastroenterologyCampus Virchow‐Klinikum and Campus Charité MitteCharité – Universitätsmedizin Berlin13353BerlinGermany
| | - Fabian Schumacher
- Institute of PharmacyFreie Universität BerlinKönigin‐Luise‐Str. 2+414195BerlinGermany
| | - Jana Hundertmark
- Department of Hepatology & GastroenterologyCampus Virchow‐Klinikum and Campus Charité MitteCharité – Universitätsmedizin Berlin13353BerlinGermany
| | | | - Felix Heymann
- Department of Hepatology & GastroenterologyCampus Virchow‐Klinikum and Campus Charité MitteCharité – Universitätsmedizin Berlin13353BerlinGermany
| | - Tobias Puengel
- Department of Hepatology & GastroenterologyCampus Virchow‐Klinikum and Campus Charité MitteCharité – Universitätsmedizin Berlin13353BerlinGermany
| | - Burkhard Kleuser
- Institute of PharmacyFreie Universität BerlinKönigin‐Luise‐Str. 2+414195BerlinGermany
| | - Alexander Sandy Mosig
- Institute of Biochemistry IICenter for Sepsis Control and CareJena University Hospital07747JenaGermany
| | - Frank Tacke
- Department of Hepatology & GastroenterologyCampus Virchow‐Klinikum and Campus Charité MitteCharité – Universitätsmedizin Berlin13353BerlinGermany
| | - Adrien Guillot
- Department of Hepatology & GastroenterologyCampus Virchow‐Klinikum and Campus Charité MitteCharité – Universitätsmedizin Berlin13353BerlinGermany
| |
Collapse
|
19
|
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: 0] [Impact Index Per Article: 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
|
20
|
Mehta V, Karnam G, Madgula V. Liver-on-chips for drug discovery and development. Mater Today Bio 2024; 27:101143. [PMID: 39070097 PMCID: PMC11279310 DOI: 10.1016/j.mtbio.2024.101143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/07/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024] Open
Abstract
Recent FDA modernization act 2.0 has led to increasing industrial R&D investment in advanced in vitro 3D models such as organoids, spheroids, organ-on-chips, 3D bioprinting, and in silico approaches. Liver-related advanced in vitro models remain the prime area of interest, as liver plays a central role in drug clearance of compounds. Growing evidence indicates the importance of recapitulating the overall liver microenvironment to enhance hepatocyte maturity and culture longevity using liver-on-chips (LoC) in vitro. Hence, pharmaceutical industries have started exploring LoC assays in the two of the most challenging areas: accurate in vitro-in vivo extrapolation (IVIVE) of hepatic drug clearance and drug-induced liver injury. We examine the joint efforts of commercial chip manufacturers and pharmaceutical companies to present an up-to-date overview of the adoption of LoC technology in the drug discovery. Further, several roadblocks are identified to the rapid adoption of LoC assays in the current drug development framework. Finally, we discuss some of the underexplored application areas of LoC models, where conventional 2D hepatic models are deemed unsuitable. These include clearance prediction of metabolically stable compounds, immune-mediated drug-induced liver injury (DILI) predictions, bioavailability prediction with gut-liver systems, hepatic clearance prediction of drugs given during pregnancy, and dose adjustment studies in disease conditions. We conclude the review by discussing the importance of PBPK modeling with LoC, digital twins, and AI/ML integration with LoC.
Collapse
Affiliation(s)
- Viraj Mehta
- Organoid Technology Lab, DMPK Department, Sai Life Sciences, Hyderabad, 500078, India
| | - Guruswamy Karnam
- Organoid Technology Lab, DMPK Department, Sai Life Sciences, Hyderabad, 500078, India
| | - Vamsi Madgula
- Organoid Technology Lab, DMPK Department, Sai Life Sciences, Hyderabad, 500078, India
| |
Collapse
|
21
|
Gallegos-Martínez S, Choy-Buentello D, Pérez-Álvarez KA, Lara-Mayorga IM, Aceves-Colin AE, Zhang YS, Trujillo-de Santiago G, Álvarez MM. A 3D-printed tumor-on-chip: user-friendly platform for the culture of breast cancer spheroids and the evaluation of anti-cancer drugs. Biofabrication 2024; 16:045010. [PMID: 38866003 DOI: 10.1088/1758-5090/ad5765] [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: 11/04/2023] [Accepted: 06/12/2024] [Indexed: 06/14/2024]
Abstract
Tumor-on-chips (ToCs) are useful platforms for studying the physiology of tumors and evaluating the efficacy and toxicity of anti-cancer drugs. However, the design and fabrication of a ToC system is not a trivial venture. We introduce a user-friendly, flexible, 3D-printed microfluidic device that can be used to culture cancer cells or cancer-derived spheroids embedded in hydrogels under well-controlled environments. The system consists of two lateral flow compartments (left and right sides), each with two inlets and two outlets to deliver cell culture media as continuous liquid streams. The central compartment was designed to host a hydrogel in which cells and microtissues can be confined and cultured. We performed tracer experiments with colored inks and 40 kDa fluorescein isothiocyanate dextran to characterize the transport/mixing performances of the system. We also cultured homotypic (MCF7) and heterotypic (MCF7-BJ) spheroids embedded in gelatin methacryloyl hydrogels to illustrate the use of this microfluidic device in sustaining long-term micro-tissue culture experiments. We further demonstrated the use of this platform in anticancer drug testing by continuous perfusion of doxorubicin, a commonly used anti-cancer drug for breast cancer. In these experiments, we evaluated drug transport, viability, glucose consumption, cell death (apoptosis), and cytotoxicity. In summary, we introduce a robust and friendly ToC system capable of recapitulating relevant aspects of the tumor microenvironment for the study of cancer physiology, anti-cancer drug transport, efficacy, and safety. We anticipate that this flexible 3D-printed microfluidic device may facilitate cancer research and the development and screening of strategies for personalized medicine.
Collapse
Affiliation(s)
- Salvador Gallegos-Martínez
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, Campus Monterrey, CP 64849 Monterrey, Nuevo León, Mexico
- Departamento de Mecatrónica e Ingeniería Eléctrica, Escuela de Ingeniería y Ciencias, Tecnológico de Monterrey, Monterrey, Nuevo León, CP 64849, Mexico
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, United States of America
| | - David Choy-Buentello
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, Campus Monterrey, CP 64849 Monterrey, Nuevo León, Mexico
| | - Kristen Aideé Pérez-Álvarez
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, Campus Monterrey, CP 64849 Monterrey, Nuevo León, Mexico
| | | | | | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, United States of America
| | - Grissel Trujillo-de Santiago
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, Campus Monterrey, CP 64849 Monterrey, Nuevo León, Mexico
- Departamento de Mecatrónica e Ingeniería Eléctrica, Escuela de Ingeniería y Ciencias, Tecnológico de Monterrey, Monterrey, Nuevo León, CP 64849, Mexico
| | - Mario Moisés Álvarez
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, Campus Monterrey, CP 64849 Monterrey, Nuevo León, Mexico
- Departamento de Mecatrónica e Ingeniería Eléctrica, Escuela de Ingeniería y Ciencias, Tecnológico de Monterrey, Monterrey, Nuevo León, CP 64849, Mexico
| |
Collapse
|
22
|
Pound P, Ritskes-Hoitinga M. Will Labour help us transition away from pre-clinical animal research? BMJ 2024; 386:q1600. [PMID: 39029938 DOI: 10.1136/bmj.q1600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/21/2024]
Affiliation(s)
| | - Merel Ritskes-Hoitinga
- Institute for Risk Assessment Sciences, Veterinary Medicine, Utrecht University, Netherlands
| |
Collapse
|
23
|
Kim J, Yoon T, Lee S, Kim PJ, Kim Y. Reconstitution of human tissue barrier function for precision and personalized medicine. LAB ON A CHIP 2024; 24:3347-3366. [PMID: 38895863 DOI: 10.1039/d4lc00104d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Tissue barriers in a body, well known as tissue-to-tissue interfaces represented by endothelium of the blood vessels or epithelium of organs, are essential for maintaining physiological homeostasis by regulating molecular and cellular transports. It is crucial for predicting drug response to understand physiology of tissue barriers through which drugs are absorbed, distributed, metabolized and excreted. Since the FDA Modernization Act 2.0, which prompts the inception of alternative technologies for animal models, tissue barrier chips, one of the applications of organ-on-a-chip or microphysiological system (MPS), have only recently been utilized in the context of drug development. Recent advancements in stem cell technology have brightened the prospects for the application of tissue barrier chips in personalized medicine. In past decade, designing and engineering these microfluidic devices, and demonstrating the ability to reconstitute tissue functions were main focus of this field. However, the field is now advancing to the next level of challenges: validating their utility in drug evaluation and creating personalized models using patient-derived cells. In this review, we briefly introduce key design parameters to develop functional tissue barrier chip, explore the remarkable recent progress in the field of tissue barrier chips and discuss future perspectives on realizing personalized medicine through the utilization of tissue barrier chips.
Collapse
Affiliation(s)
- Jaehoon Kim
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA.
| | - Taehee Yoon
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA.
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Sungryeong Lee
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA.
| | - Paul J Kim
- Department of Psychiatry & Behavioral Sciences, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - YongTae Kim
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA.
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
24
|
Jadalannagari S, Ewart L. Beyond the hype and toward application: liver complex in vitro models in preclinical drug safety. Expert Opin Drug Metab Toxicol 2024; 20:607-619. [PMID: 38465923 DOI: 10.1080/17425255.2024.2328794] [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: 01/15/2024] [Accepted: 03/06/2024] [Indexed: 03/12/2024]
Abstract
INTRODUCTION Drug induced Liver-Injury (DILI) is a leading cause of drug attrition and complex in vitro models (CIVMs), including three dimensional (3D) spheroids, 3D bio printed tissues and flow-based systems, could improve preclinical prediction. Although CIVMs have demonstrated good sensitivity and specificity in DILI detection their adoption remains limited. AREAS COVERED This article describes DILI, the challenges with its prediction and the current strategies and models that are being used. It reviews data from industry-FDA collaborations and strategic partnerships and finishes with an outlook of CIVMs in preclinical toxicity testing. Literature searches were performed using PubMed and Google Scholar while product information was collected from manufacturer websites. EXPERT OPINION Liver CIVMs are promising models for predicting DILI although, a decade after their introduction, routine use by the pharmaceutical industry is limited. To accelerate their adoption, several industry-regulator-developer partnerships or consortia have been established to guide the development and qualification. Beyond this, liver CIVMs should continue evolving to capture greater immunological mimicry while partnering with computational approaches to deliver systems that change the paradigm of predicting DILI.
Collapse
Affiliation(s)
| | - Lorna Ewart
- Department of Bioinnovations, Emulate Inc, Boston, MA, USA
| |
Collapse
|
25
|
Rajagopal D, MacLeod E, Corogeanu D, Vessillier S. Immune-related adverse events of antibody-based biological medicines in cancer therapy. J Cell Mol Med 2024; 28:e18470. [PMID: 38963257 PMCID: PMC11223167 DOI: 10.1111/jcmm.18470] [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: 01/19/2024] [Revised: 05/03/2024] [Accepted: 05/22/2024] [Indexed: 07/05/2024] Open
Abstract
Recombinant antibodies (Abs) are an integral modality for the treatment of multiple tumour malignancies. Since the Food and Drug Administration (FDA) approval of rituximab as the first monoclonal antibody (mAb) for cancer treatment, several mAbs and antibody (Ab)-based therapies have been approved for the treatment of solid tumour malignancies and other cancers. These Abs function by either blocking oncogenic pathways or angiogenesis, modulating immune response, or by delivering a conjugated drug. The use of Ab-based therapy in cancer patients who could benefit from the treatment, however, is still limited by associated toxicity profiles which may stem from biological features and processes related to target binding, alongside biochemical and/or biophysical characteristics of the therapeutic Ab. A significant immune-related adverse event (irAE) associated with Ab-based therapies is cytokine release syndrome (CRS), characterized by the development of fever, rash and even marked, life-threatening hypotension, and acute inflammation with secondary to systemic uncontrolled increase in a range of pro-inflammatory cytokines. Here, we review irAEs associated with specific classes of approved, Ab-based novel cancer immunotherapeutics, namely immune checkpoint (IC)-targeting Abs, bispecific Abs (BsAbs) and Ab-drug-conjugates (ADCs), highlighting the significance of harmonization in preclinical assay development for safety assessment of Ab-based biotherapeutics as an approach to support and refine clinical translation.
Collapse
Affiliation(s)
- Deepa Rajagopal
- Immunotherapy, Biotherapeutics and Advanced Therapies Division, Science, Research, and Innovation Group, Medicines and Healthcare products Regulatory Agency (MHRA)HertfordshireUK
| | - Elliot MacLeod
- Immunotherapy, Biotherapeutics and Advanced Therapies Division, Science, Research, and Innovation Group, Medicines and Healthcare products Regulatory Agency (MHRA)HertfordshireUK
- Present address:
Gilead Sciences, Winchester HouseOxfordUK
| | - Diana Corogeanu
- Immunotherapy, Biotherapeutics and Advanced Therapies Division, Science, Research, and Innovation Group, Medicines and Healthcare products Regulatory Agency (MHRA)HertfordshireUK
- Present address:
East Sussex Healthcare NHS Trust, Conquest HospitalEast SussexUK
| | - Sandrine Vessillier
- Immunotherapy, Biotherapeutics and Advanced Therapies Division, Science, Research, and Innovation Group, Medicines and Healthcare products Regulatory Agency (MHRA)HertfordshireUK
| |
Collapse
|
26
|
Alver CG, Drabbe E, Ishahak M, Agarwal A. Roadblocks confronting widespread dissemination and deployment of Organs on Chips. Nat Commun 2024; 15:5118. [PMID: 38879554 PMCID: PMC11180125 DOI: 10.1038/s41467-024-48864-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 05/16/2024] [Indexed: 06/19/2024] Open
Abstract
Organ on Chip platforms hold significant promise as alternatives to animal models or traditional cell cultures, both of which poorly recapitulate human pathophysiology and human level responses. Within the last 15 years, we have witnessed seminal scientific developments from academic laboratories, a flurry of startups and investments, and a genuine interest from pharmaceutical industry as well as regulatory authorities to translate these platforms. This Perspective identifies several fundamental design and process features that may act as roadblocks that prevent widespread dissemination and deployment of these systems, and provides a roadmap to help position this technology in mainstream drug discovery.
Collapse
Affiliation(s)
- Charles G Alver
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA
- Medical Scientist Training Program, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Emma Drabbe
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA
| | - Matthew Ishahak
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA
| | - Ashutosh Agarwal
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
- Desai Sethi Urology Institute, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
27
|
Morrison AI, Sjoerds MJ, Vonk LA, Gibbs S, Koning JJ. In vitro immunity: an overview of immunocompetent organ-on-chip models. Front Immunol 2024; 15:1373186. [PMID: 38835750 PMCID: PMC11148285 DOI: 10.3389/fimmu.2024.1373186] [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: 01/19/2024] [Accepted: 04/30/2024] [Indexed: 06/06/2024] Open
Abstract
Impressive advances have been made to replicate human physiology in vitro over the last few years due to the growth of the organ-on-chip (OoC) field in both industrial and academic settings. OoCs are a type of microphysiological system (MPS) that imitates functional and dynamic aspects of native human organ biology on a microfluidic device. Organoids and organotypic models, ranging in their complexity from simple single-cell to complex multi-cell type constructs, are being incorporated into OoC microfluidic devices to better mimic human physiology. OoC technology has now progressed to the stage at which it has received official recognition by the Food and Drug Administration (FDA) for use as an alternative to standard procedures in drug development, such as animal studies and traditional in vitro assays. However, an area that is still lagging behind is the incorporation of the immune system, which is a critical element required to investigate human health and disease. In this review, we summarise the progress made to integrate human immunology into various OoC systems, specifically focusing on models related to organ barriers and lymphoid organs. These models utilise microfluidic devices that are either commercially available or custom-made. This review explores the difference between the use of innate and adaptive immune cells and their role for modelling organ-specific diseases in OoCs. Immunocompetent multi-OoC models are also highlighted and the extent to which they recapitulate systemic physiology is discussed. Together, the aim of this review is to describe the current state of immune-OoCs, the limitations and the future perspectives needed to improve the field.
Collapse
Affiliation(s)
- Andrew I. Morrison
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, Netherlands
| | - Mirthe J. Sjoerds
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Leander A. Vonk
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Susan Gibbs
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, Netherlands
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, Netherlands
| | - Jasper J. Koning
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, Netherlands
| |
Collapse
|
28
|
Ingelman-Sundberg M, Lauschke VM. Individualized Pharmacotherapy Utilizing Genetic Biomarkers and Novel In Vitro Systems As Predictive Tools for Optimal Drug Development and Treatment. Drug Metab Dispos 2024; 52:467-475. [PMID: 38575185 DOI: 10.1124/dmd.123.001302] [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: 12/22/2023] [Revised: 02/15/2024] [Accepted: 03/12/2024] [Indexed: 04/06/2024] Open
Abstract
In the area of drug development and clinical pharmacotherapy, a profound understanding of the pharmacokinetics and potential adverse reactions associated with the drug under investigation is paramount. Essential to this endeavor is a comprehensive understanding about interindividual variations in absorption, distribution, metabolism, and excretion (ADME) genetics and the predictive capabilities of in vitro systems, shedding light on metabolite formation and the risk of adverse drug reactions (ADRs). Both the domains of pharmacogenomics and the advancement of in vitro systems are experiencing rapid expansion. Here we present an update on these burgeoning fields, providing an overview of their current status and illuminating potential future directions. SIGNIFICANCE STATEMENT: There is very rapid development in the area of pharmacogenomics and in vitro systems for predicting drug pharmacokinetics and risk for adverse drug reactions. We provide an update of the current status of pharmacogenomics and developed in vitro systems on these aspects aimed to achieve a better personalized pharmacotherapy.
Collapse
Affiliation(s)
- Magnus Ingelman-Sundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (M.I.-S., V.M.L.); Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.); and University of Tübingen, Tübingen, Germany (V.M.L.)
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (M.I.-S., V.M.L.); Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.); and University of Tübingen, Tübingen, Germany (V.M.L.)
| |
Collapse
|
29
|
Shrestha S, Lekkala VKR, Acharya P, Kang SY, Vanga MG, Lee MY. Reproducible generation of human liver organoids (HLOs) on a pillar plate platform via microarray 3D bioprinting. LAB ON A CHIP 2024; 24:2747-2761. [PMID: 38660778 DOI: 10.1039/d4lc00149d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Human liver organoids (HLOs) hold significant potential for recapitulating the architecture and function of liver tissues in vivo. However, conventional culture methods of HLOs, forming Matrigel domes in 6-/24-well plates, have technical limitations such as high cost and low throughput in organoid-based assays for predictive assessment of compounds in clinical and pharmacological lab settings. To address these issues, we have developed a unique microarray 3D bioprinting protocol of progenitor cells in biomimetic hydrogels on a pillar plate with sidewalls and slits, coupled with a clear bottom, 384-deep well plate for scale-up production of HLOs. Microarray 3D bioprinting, a droplet-based printing technology, was used to generate a large number of small organoids on the pillar plate for predictive hepatotoxicity assays. Foregut cells, differentiated from human iPSCs, were mixed with Matrigel and then printed on the pillar plate rapidly and uniformly, resulting in coefficient of variation (CV) values in the range of 15-18%, without any detrimental effect on cell viability. Despite utilizing 10-50-fold smaller cell culture volume compared to their counterparts in Matrigel domes in 6-/24-well plates, HLOs differentiated on the pillar plate exhibited similar morphology and superior function, potentially due to rapid diffusion of nutrients and oxygen at the small scale. Day 25 HLOs were robust and functional on the pillar plate in terms of their viability, albumin secretion, CYP3A4 activity, and drug toxicity testing, all with low CV values. From three independent trials of in situ assessment, the IC50 values calculated for sorafenib and tamoxifen were 6.2 ± 1.6 μM and 25.4 ± 8.3 μM, respectively. Therefore, our unique 3D bioprinting and miniature organoid culture on the pillar plate could be used for scale-up, reproducible generation of HLOs with minimal manual intervention for high-throughput assessment of compound hepatotoxicity.
Collapse
Affiliation(s)
- Sunil Shrestha
- Department of Biomedical Engineering, University of North Texas, Denton, Texas, USA.
| | | | - Prabha Acharya
- Department of Biomedical Engineering, University of North Texas, Denton, Texas, USA.
| | - Soo-Yeon Kang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas, USA.
| | - Manav Goud Vanga
- Department of Biomedical Engineering, University of North Texas, Denton, Texas, USA.
| | - Moo-Yeal Lee
- Department of Biomedical Engineering, University of North Texas, Denton, Texas, USA.
- Bioprinting Laboratories Inc., Dallas, Texas, USA
| |
Collapse
|
30
|
Lekkala VKR, Kang SY, Liu J, Shrestha S, Acharya P, Joshi P, Zolfaghar M, Lee M, Vanga MG, Jamdagneya P, Pagnis S, Kundi A, Kabbur S, Kim UT, Yang Y, Lee MY. A Pillar/Perfusion Plate Enhances Cell Growth, Reproducibility, Throughput, and User Friendliness in Dynamic 3D Cell Culture. ACS Biomater Sci Eng 2024; 10:3478-3488. [PMID: 38695610 DOI: 10.1021/acsbiomaterials.4c00179] [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: 05/14/2024]
Abstract
Static three-dimensional (3D) cell culture has been demonstrated in ultralow attachment well plates, hanging droplet plates, and microtiter well plates with hydrogels or magnetic nanoparticles. Although it is simple, reproducible, and relatively inexpensive, thus potentially used for high-throughput screening, statically cultured 3D cells often suffer from a necrotic core due to limited nutrient and oxygen diffusion and waste removal and have a limited in vivo-like tissue structure. Here, we overcome these challenges by developing a pillar/perfusion plate platform and demonstrating high-throughput, dynamic 3D cell culture. Cell spheroids were loaded on the pillar plate with hydrogel by simple sandwiching and encapsulation and cultured dynamically in the perfusion plate on a digital rocker. Unlike traditional microfluidic devices, fast flow velocity was maintained within perfusion wells and the pillar plate was separated from the perfusion plate for cell-based assays. It was compatible with common lab equipment and allowed cell culture, testing, staining, and imaging in situ. The pillar/perfusion plate enhanced cell growth by rapid diffusion, reproducibility, assay throughput, and user friendliness in a dynamic 3D cell culture.
Collapse
Affiliation(s)
| | - Soo-Yeon Kang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Jiafeng Liu
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Sunil Shrestha
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Prabha Acharya
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Pranav Joshi
- Bioprinting Laboratories, Inc., Dallas, Texas 75234, United States
| | - Mona Zolfaghar
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Minseong Lee
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Manav Goud Vanga
- Bioprinting Laboratories, Inc., Dallas, Texas 75234, United States
| | - Paarth Jamdagneya
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Sohan Pagnis
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Arham Kundi
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Samarth Kabbur
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Ung Tae Kim
- Department of Civil and Environmental Engineering, Cleveland State University, Cleveland, Ohio 44115, United States
| | - Yong Yang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Moo-Yeal Lee
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
- Bioprinting Laboratories, Inc., Dallas, Texas 75234, United States
| |
Collapse
|
31
|
Yin DE, Palin AC, Lombo TB, Mahon RN, Poon B, Wu DY, Atala A, Brooks KM, Chen S, Coyne CB, D’Souza MP, Fackler OT, Furler O’Brien RL, Garcia-de-Alba C, Jean-Philippe P, Karn J, Majji S, Muotri AR, Ozulumba T, Sakatis MZ, Schlesinger LS, Singh A, Spiegel HM, Struble E, Sung K, Tagle DA, Thacker VV, Tidball AM, Varthakavi V, Vunjak-Novakovic G, Wagar LE, Yeung CK, Ndhlovu LC, Ott M. 3D human tissue models and microphysiological systems for HIV and related comorbidities. Trends Biotechnol 2024; 42:526-543. [PMID: 38071144 PMCID: PMC11065605 DOI: 10.1016/j.tibtech.2023.10.008] [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: 07/03/2023] [Revised: 10/22/2023] [Accepted: 10/24/2023] [Indexed: 03/03/2024]
Abstract
Three-dimensional (3D) human tissue models/microphysiological systems (e.g., organs-on-chips, organoids, and tissue explants) model HIV and related comorbidities and have potential to address critical questions, including characterization of viral reservoirs, insufficient innate and adaptive immune responses, biomarker discovery and evaluation, medical complexity with comorbidities (e.g., tuberculosis and SARS-CoV-2), and protection and transmission during pregnancy and birth. Composed of multiple primary or stem cell-derived cell types organized in a dedicated 3D space, these systems hold unique promise for better reproducing human physiology, advancing therapeutic development, and bridging the human-animal model translational gap. Here, we discuss the promises and achievements with 3D human tissue models in HIV and comorbidity research, along with remaining barriers with respect to cell biology, virology, immunology, and regulatory issues.
Collapse
|
32
|
Abstract
The Three Rs have become widely accepted and pursued, and are now the go-to framework that encourages the humane use of animals in science, where no other option is believed to exist. However, many people, including scientists, harbour varying degrees of concern about the value and impact of the Three Rs. This ranges from a continued adherence to the Three Rs principles in the belief that they have performed well, through a belief that there should be more emphasis (or indeed a sole focus) on replacement, to a view that the principles have hindered, rather than helped, a critical approach to animal research that should have resulted in replacement to a much greater extent. This critical review asks questions of the Three Rs and their implementation, and provides an overview of the current situation surrounding animal use in biomedical science (chiefly in research). It makes a case that it is time to move away from the Three Rs and that, while this happens, the principles need to be made more robust and enforced more efficiently. To expedite a shift from animal use in science, toward a much greater and quicker adoption of human-specific New Approach Methodologies (NAMs), some argue for a straightforward focus on the best available science.
Collapse
|
33
|
AlShmmari SK, Fardous RS, Shinwari Z, Cialla-May D, Popp J, Ramadan Q, Zourob M. Hepatic spheroid-on-a-chip: Fabrication and characterization of a spheroid-based in vitro model of the human liver for drug screening applications. BIOMICROFLUIDICS 2024; 18:034105. [PMID: 38817733 PMCID: PMC11136519 DOI: 10.1063/5.0210955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/06/2024] [Indexed: 06/01/2024]
Abstract
The integration of microfabrication and microfluidics techniques into cell culture technology has significantly transformed cell culture conditions, scaffold architecture, and tissue biofabrication. These tools offer precise control over cell positioning and enable high-resolution analysis and testing. Culturing cells in 3D systems, such as spheroids and organoids, enables recapitulating the interaction between cells and the extracellular matrix, thereby allowing the creation of human-based biomimetic tissue models that are well-suited for pre-clinical drug screening. Here, we demonstrate an innovative microfluidic device for the formation, culture, and testing of hepatocyte spheroids, which comprises a large array of patterned microwells for hosting hepatic spheroid culture in a reproducible and organized format in a dynamic fluidic environment. The device allows maintaining and characterizing different spheroid sizes as well as exposing to various drugs in parallel enabling high-throughput experimentation. These liver spheroids exhibit physiologically relevant hepatic functionality, as evidenced by their ability to produce albumin and urea at levels comparable to in vivo conditions and the capability to distinguish the toxic effects of selected drugs. This highlights the effectiveness of the microenvironment provided by the chip in maintaining the functionality of hepatocyte spheroids. These data support the notion that the liver-spheroid chip provides a favorable microenvironment for the maintenance of hepatocyte spheroid functionality.
Collapse
Affiliation(s)
| | | | - Zakia Shinwari
- Cell Therapy and Immunology Department, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| | | | | | - Qasem Ramadan
- College of Science & General Studies, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Mohammed Zourob
- College of Science & General Studies, Alfaisal University, Riyadh 11533, Saudi Arabia
| |
Collapse
|
34
|
Bissoyi A, Gao Y, Tomás RMF, Kinney NLH, Whale TF, Guo Q, Gibson MI. Cryopreservation and Rapid Recovery of Differentiated Intestinal Epithelial Barrier Cells at Complex Transwell Interfaces Is Enabled by Chemically Induced Ice Nucleation. ACS APPLIED MATERIALS & INTERFACES 2024; 16. [PMID: 38671549 PMCID: PMC11082836 DOI: 10.1021/acsami.4c03931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/07/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024]
Abstract
Cell-based models, such as organ-on-chips, can replace and inform in vivo (animal) studies for drug discovery, toxicology, and biomedical science, but most cannot be banked "ready to use" as they do not survive conventional cryopreservation with DMSO alone. Here, we demonstrate how macromolecular ice nucleators enable the successful cryopreservation of epithelial intestinal models supported upon the interface of transwells, allowing recovery of function in just 7 days post-thaw directly from the freezer, compared to 21 days from conventional suspension cryopreservation. Caco-2 cells and Caco-2/HT29-MTX cocultures are cryopreserved on transwell inserts, with chemically induced ice nucleation at warmer temperatures resulting in increased cell viability but crucially retaining the complex cellular adhesion on the transwell insert interfaces, which other cryoprotectants do not. Trans-epithelial electrical resistance measurements, confocal microscopy, histology, and whole-cell proteomics demonstrated the rapid recovery of differentiated cell function, including the formation of tight junctions. Lucifer yellow permeability assays confirmed that the barrier functions of the cells were intact. This work will help solve the long-standing problem of transwell tissue barrier model storage, facilitating access to advanced predictive cellular models. This is underpinned by precise control of the nucleation temperature, addressing a crucial biophysical mode of damage.
Collapse
Affiliation(s)
- Akalabya Bissoyi
- Department
of Chemistry, University of Manchester, Oxford Road, Manchester M13 9PL, U.K.
- Manchester
Institute of Biotechnology, University of
Manchester, 131 Princess Street, Manchester M1 7DN, U.K.
| | - Yanan Gao
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, U.K.
- Department
of Biomedical Engineering, Southern University
of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Ruben M. F. Tomás
- Cryologyx
Ltd, Venture Centre, University of Warwick
Science Park, Coventry CV4 7EZ, U.K.
| | - Nina L. H. Kinney
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, U.K.
- Royal
Botanic Gardens Kew, Ardingly, West Sussex RH17 6TN, U.K.
| | - Thomas F. Whale
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, U.K.
- School
of Earth and Environment, University of
Leeds, Leeds LS2 9JT, U.K.
| | - Qiongyu Guo
- Department
of Biomedical Engineering, Southern University
of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Matthew I. Gibson
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, U.K.
- Division
of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, U.K.
- Department
of Chemistry, University of Manchester, Oxford Road, Manchester M13 9PL, U.K.
- Manchester
Institute of Biotechnology, University of
Manchester, 131 Princess Street, Manchester M1 7DN, U.K.
| |
Collapse
|
35
|
Bitounis D, Jacquinet E, Rogers MA, Amiji MM. Strategies to reduce the risks of mRNA drug and vaccine toxicity. Nat Rev Drug Discov 2024; 23:281-300. [PMID: 38263456 DOI: 10.1038/s41573-023-00859-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2023] [Indexed: 01/25/2024]
Abstract
mRNA formulated with lipid nanoparticles is a transformative technology that has enabled the rapid development and administration of billions of coronavirus disease 2019 (COVID-19) vaccine doses worldwide. However, avoiding unacceptable toxicity with mRNA drugs and vaccines presents challenges. Lipid nanoparticle structural components, production methods, route of administration and proteins produced from complexed mRNAs all present toxicity concerns. Here, we discuss these concerns, specifically how cell tropism and tissue distribution of mRNA and lipid nanoparticles can lead to toxicity, and their possible reactogenicity. We focus on adverse events from mRNA applications for protein replacement and gene editing therapies as well as vaccines, tracing common biochemical and cellular pathways. The potential and limitations of existing models and tools used to screen for on-target efficacy and de-risk off-target toxicity, including in vivo and next-generation in vitro models, are also discussed.
Collapse
Affiliation(s)
- Dimitrios Bitounis
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
- Moderna, Inc., Cambridge, MA, USA
| | | | | | - Mansoor M Amiji
- Departments of Pharmaceutical Sciences and Chemical Engineering, Northeastern University, Boston, MA, USA.
| |
Collapse
|
36
|
Kavanagh O, Krebs CE. Mitigating animal methods bias to reduce animal use and improve biomedical translation. Sci Prog 2024; 107:368504241253693. [PMID: 38752259 PMCID: PMC11102665 DOI: 10.1177/00368504241253693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2024]
Abstract
Nonanimal biomedical research methods have advanced rapidly over the last decade making them the first-choice model for many researchers due to improved translatability and avoidance of ethical concerns. Yet confidence in novel nonanimal methods is still being established and they remain a small portion of nonclinical biomedical research, which can lead peer reviewers to evaluate animal-free studies or grant proposals in a biased manner. This "animal methods bias" is the preference for animal-based research methods where they are not necessary or where nonanimal-based methods are suitable. It affects the fair consideration of animal-free biomedical research, hampering the uptake and dissemination of these approaches by putting pressure on researchers to conduct animal experiments and potentially perpetuating the use of poorly translatable model systems. An international team of researchers and advocates called the Coalition to Illuminate and Address Animal Methods Bias (COLAAB) aims to provide concrete evidence of the existence and consequences of this bias and to develop and implement solutions towards overcoming it. The COLAAB recently developed the first of several mitigation tools: the Author Guide for Addressing Animal Methods Bias in Publishing, which is described herein along with broader implications and future directions of this work.
Collapse
|
37
|
Luan Q, Pulido I, Isagirre A, Carretero J, Zhou J, Shimamura T, Papautsky I. Deciphering fibroblast-induced drug resistance in non-small cell lung carcinoma through patient-derived organoids in agarose microwells. LAB ON A CHIP 2024; 24:2025-2038. [PMID: 38410967 PMCID: PMC11209828 DOI: 10.1039/d3lc01044a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Patient-derived organoids (PDOs) serve as invaluable 3D tumor models, retaining the histological complexity and genetic heterogeneity found in primary tumors. However, the limitation of small sample volumes and the lack of tailored platforms have hindered the research using PDOs. Within the tumor microenvironment, cancer-associated fibroblasts play a pivotal role in influencing drug sensitivity. In this study, we introduce an agarose microwell platform designed for PDO-based tumor and tumor microenvironment models, enabling rapid drug screening and resistance studies with small sample volumes. These microwells, constructed using 3D printing molds, feature a U-shaped bottom and 200 μm diameter. We successfully generated co-culture spheroids of non-small cell lung carcinoma (NSCLC) cells, including NCI-H358 or A549, and NSCLC PDOs F231 or F671 with fibroblast cell line, WI-38. Our results demonstrate the production of uniformly-sized spheroids (coefficient of variation <30%), high viability (>80% after 1 week), and fibroblast-induced drug resistance. The PDOs maintained their viability (>81% after 2 weeks) and continued to proliferate. Notably, when exposed to adagrasib, a KRASG12C inhibitor, we observed reduced cytotoxicity in KRASG12C-mutant spheroids when co-cultured with fibroblasts or their supernatant. The fibroblast supernatant sustained proliferative signals in tumor models. Taking into account the physical features, viability, and drug resistance acquired through supernatants from the fibroblasts, our platform emerges as a suitable platform for in vitro tumor modeling and the evaluation of drug efficacy using patient-derived tissues.
Collapse
Affiliation(s)
- Qiyue Luan
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, 851 S. Morgan Street, 218 SEO, Chicago, IL 60607, USA.
| | - Ines Pulido
- Department of Surgery, University of Illinois Chicago, Chicago, IL 60612, USA
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| | - Angelique Isagirre
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, 851 S. Morgan Street, 218 SEO, Chicago, IL 60607, USA.
| | - Julian Carretero
- Departament de Fisiologia, Facultat de Farmacia, Universitat de Valencia, Burjassot, 46010, Spain
| | - Jian Zhou
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, 851 S. Morgan Street, 218 SEO, Chicago, IL 60607, USA.
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| | - Takeshi Shimamura
- Department of Surgery, University of Illinois Chicago, Chicago, IL 60612, USA
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| | - Ian Papautsky
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, 851 S. Morgan Street, 218 SEO, Chicago, IL 60607, USA.
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| |
Collapse
|
38
|
Shrestha S, Lekkala VKR, Acharya P, Kang SY, Vanga MG, Lee MY. Reproducible generation of human liver organoids (HLOs) on a pillar plate platform via microarray 3D bioprinting. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.11.584478. [PMID: 38559126 PMCID: PMC10979895 DOI: 10.1101/2024.03.11.584478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Human liver organoids (HLOs) hold significant potential for recapitulating the architecture and function of liver tissues in vivo. However, conventional culture methods of HLOs, forming Matrigel domes in 6-/24-well plates, have technical limitations such as high cost and low throughput in organoid-based assays for predictive assessment of compounds in clinical and pharmacological lab settings. To address these issues, we have developed a unique microarray 3D bioprinting protocol of progenitor cells in biomimetic hydrogels on a pillar plate with sidewalls and slits, coupled with a clear bottom, 384-deep well plate for scale-up production of HLOs. Microarray 3D bioprinting, a droplet-based printing technology, was used to generate a large number of small organoids on the pillar plate for predictive hepatotoxicity assays. Foregut cells, differentiated from human iPSCs, were mixed with Matrigel and then printed on the pillar plate rapidly and uniformly, resulting in coefficient of variation (CV) values in the range of 15 - 18%, without any detrimental effect on cell viability. Despite utilizing 10 - 50-fold smaller cell culture volume compared to their counterparts in Matrigel domes in 6-/24-well plates, HLOs differentiated on the pillar plate exhibited similar morphology and superior function, potentially due to rapid diffusion of nutrients and oxygen at the small scale. Day 25 HLOs were robust and functional on the pillar plate in terms of their viability, albumin secretion, CYP3A4 activity, and drug toxicity testing, all with low CV values. From three independent trials of in situ assessment, the IC50 values calculated for sorafenib and tamoxifen were 6.2 ± 1.6 μM and 25.4 ± 8.3 μM, respectively. Therefore, our unique 3D bioprinting and miniature organoid culture on the pillar plate could be used for scale-up, reproducible generation of HLOs with minimal manual intervention for high-throughput assessment of compound hepatotoxicity.
Collapse
Affiliation(s)
- Sunil Shrestha
- Department of Biomedical Engineering, University of North Texas, Denton, Texas
| | | | - Prabha Acharya
- Department of Biomedical Engineering, University of North Texas, Denton, Texas
| | - Soo-Yeon Kang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas
| | - Manav Goud Vanga
- Department of Biomedical Engineering, University of North Texas, Denton, Texas
| | - Moo-Yeal Lee
- Department of Biomedical Engineering, University of North Texas, Denton, Texas
- Bioprinting Laboratories Inc., Dallas, Texas
| |
Collapse
|
39
|
Schueler J, Sjöman H, Kriesi C. Sensor extended imaging workflow for creating fit for purpose models in basic and applied cell biology. Commun Biol 2024; 7:170. [PMID: 38341479 PMCID: PMC10858951 DOI: 10.1038/s42003-024-05843-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
While various engineering disciplines spent years on developing methods and workflows to increase their R&D efficiency, the field of cell biology has seen limited evolution in the fundamental approaches to interact with living cells. Perturbations are mostly of chemical nature, and physiologically relevant contexts and stimuli are left with limited attention, resulting in a solution space constrained within the boundaries of presently manageable perturbations. To predict in the laboratory how a drug will work in a human patient, cell biology must have a closer look at life and strive to mimic the human being in all his complexity. By implementing an iterative process from perturbation to measurement and vice versa, the authors suggest using a sensor-extended imaging workflow to implement product development practices to cell biology, opening a physiologically relevant solution space for the development of truly translational and predictive fit for purpose in vitro cell models.
Collapse
Affiliation(s)
- Julia Schueler
- Charles River Germany GmbH, Am Flughafen 12-14, 79111, Freiburg, Germany.
| | - Heikki Sjöman
- Vitroscope AS, Leirfossvegen 5d, 7037, Trondheim, Norway
| | - Carlo Kriesi
- Vitroscope AS, Leirfossvegen 5d, 7037, Trondheim, Norway
| |
Collapse
|
40
|
Ali ASM, Berg J, Roehrs V, Wu D, Hackethal J, Braeuning A, Woelken L, Rauh C, Kurreck J. Xeno-Free 3D Bioprinted Liver Model for Hepatotoxicity Assessment. Int J Mol Sci 2024; 25:1811. [PMID: 38339088 PMCID: PMC10855587 DOI: 10.3390/ijms25031811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 01/26/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
Three-dimensional (3D) bioprinting is one of the most promising methodologies that are currently in development for the replacement of animal experiments. Bioprinting and most alternative technologies rely on animal-derived materials, which compromises the intent of animal welfare and results in the generation of chimeric systems of limited value. The current study therefore presents the first bioprinted liver model that is entirely void of animal-derived constituents. Initially, HuH-7 cells underwent adaptation to a chemically defined medium (CDM). The adapted cells exhibited high survival rates (85-92%) after cryopreservation in chemically defined freezing media, comparable to those preserved in standard medium (86-92%). Xeno-free bioink for 3D bioprinting yielded liver models with high relative cell viability (97-101%), akin to a Matrigel-based liver model (83-102%) after 15 days of culture. The established xeno-free model was used for toxicity testing of a marine biotoxin, okadaic acid (OA). In 2D culture, OA toxicity was virtually identical for cells cultured under standard conditions and in CDM. In the xeno-free bioprinted liver model, 3-fold higher concentrations of OA than in the respective monolayer culture were needed to induce cytotoxicity. In conclusion, this study describes for the first time the development of a xeno-free 3D bioprinted liver model and its applicability for research purposes.
Collapse
Affiliation(s)
- Ahmed S. M. Ali
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, TIB 4/3-2, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Johanna Berg
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, TIB 4/3-2, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Viola Roehrs
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, TIB 4/3-2, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Dongwei Wu
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, TIB 4/3-2, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | | | - Albert Braeuning
- Department Food Safety, German Federal Institute for Risk Assessment (BfR), 10589 Berlin, Germany;
| | - Lisa Woelken
- Department of Food Biotechnology and Food Process Engineering, Technische Universität Berlin, 14195 Berlin, Germany (C.R.)
| | - Cornelia Rauh
- Department of Food Biotechnology and Food Process Engineering, Technische Universität Berlin, 14195 Berlin, Germany (C.R.)
| | - Jens Kurreck
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, TIB 4/3-2, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| |
Collapse
|
41
|
Bai H, Olson KNP, Pan M, Marshall T, Singh H, Ma J, Gilbride P, Yuan Y, McCormack J, Si L, Maharjan S, Huang D, Qian X, Livermore C, Zhang YS, Xie X. Rapid Prototyping of Thermoplastic Microfluidic 3D Cell Culture Devices by Creating Regional Hydrophilicity Discrepancy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304332. [PMID: 38032118 PMCID: PMC10870023 DOI: 10.1002/advs.202304332] [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: 06/28/2023] [Revised: 10/02/2023] [Indexed: 12/01/2023]
Abstract
Microfluidic 3D cell culture devices that enable the recapitulation of key aspects of organ structures and functions in vivo represent a promising preclinical platform to improve translational success during drug discovery. Essential to these engineered devices is the spatial patterning of cells from different tissue types within a confined microenvironment. Traditional fabrication strategies lack the scalability, cost-effectiveness, and rapid prototyping capabilities required for industrial applications, especially for processes involving thermoplastic materials. Here, an approach to pattern fluid guides inside microchannels is introduced by establishing differential hydrophilicity using pressure-sensitive adhesives as masks and a subsequent selective coating with a biocompatible polymer. Optimal coating conditions are identified using polyvinylpyrrolidone, which resulted in rapid and consistent hydrogel flow in both the open-chip prototype and the fully bonded device containing additional features for medium perfusion. The suitability of the device for dynamic 3D cell culture is tested by growing human hepatocytes in the device under controlled fluid flow for a 14-day period. Additionally, the study demonstrated the potential of using the device for pharmaceutical high-throughput screening applications, such as predicting drug-induced liver injury. The approach offers a facile strategy of rapid prototyping thermoplastic microfluidic organ chips with varying geometries, microstructures, and substrate materials.
Collapse
Affiliation(s)
| | | | - Ming Pan
- Xellar BiosystemsCambridgeMA02458USA
| | | | | | | | | | | | | | - Longlong Si
- CAS Key Laboratory of Quantitative Engineering BiologyShenzhen Institute of Synthetic BiologyShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhen518055P. R. China
- University of Chinese Academy of SciencesBeijing100049P. R. China
| | - Sushila Maharjan
- Division of Engineering in MedicineDepartment of MedicineBrigham and Women's HospitalHarvard Medical SchoolCambridgeMA02142USA
| | - Di Huang
- Research Center for Nano‐biomaterials & Regenerative MedicineCollege of Biomedical EngineeringTaiyuan University of TechnologyTaiyuan030024P. R. China
| | | | - Carol Livermore
- Department of Mechanical and Industrial EngineeringNortheastern UniversityBostonMA02115USA
| | - Yu Shrike Zhang
- Division of Engineering in MedicineDepartment of MedicineBrigham and Women's HospitalHarvard Medical SchoolCambridgeMA02142USA
| | - Xin Xie
- Xellar BiosystemsCambridgeMA02458USA
| |
Collapse
|
42
|
Fish A, Kulkarni A. Flow-Induced Shear Stress Primes NLRP3 Inflammasome Activation in Macrophages via Piezo1. ACS APPLIED MATERIALS & INTERFACES 2024; 16:4505-4518. [PMID: 38240257 DOI: 10.1021/acsami.3c18645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
The NLRP3 inflammasome is a crucial component of the innate immune system, playing a pivotal role in initiating and regulating the body's inflammatory response to various pathogens and cellular damage. Environmental stimuli, such as temperature, pH level, and nutrient availability, can influence the behavior and functions of innate immune cells, including immune cell activity, proliferation, and cytokine production. However, there is limited understanding regarding how mechanical forces, like shear stress, govern the intrinsic inflammatory reaction, particularly the activation of the NLRP3 inflammasome, and how shear stress impacts NLRP3 inflammasome activation through its capacity to induce alterations in gene expression and cytokine secretion. Here, we investigated how shear stress can act as a priming signal in NLRP3 inflammasome activation by exposing immortalized bone marrow-derived macrophages (iBMDMs) to numerous physiologically relevant magnitudes of shear stress before chemically inducing inflammasome activation. We demonstrated that shear stress of large magnitudes was able to prime iBMDMs more effectively for inflammasome activation compared to lower shear stress magnitudes, as quantified by the percentage of cells where ASC-CFP specks formed and IL-1β secretion, the hallmarks of inflammasome activation. Testing this in NLRP3 and caspase-1 knockout iBMDMs showed that the NLRP3 inflammasome was primarily primed for activation due to shear stress exposure. Quantitative polymerase chain reaction (qPCR) and a small-molecule inhibitor study mechanistically determined that shear stress regulates the NLRP3 inflammasome by upregulating Piezo1, IKKβ, and NLRP3. These findings offer insights into the mechanistic relationship among physiological shear stresses, inflammasome activation, and their impact on the progression of inflammatory diseases and their interconnected pathogenesis.
Collapse
Affiliation(s)
- Adam Fish
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Ashish Kulkarni
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| |
Collapse
|
43
|
Perry AS, Hadad N, Chatterjee E, Ramos MJ, Farber-Eger E, Roshani R, Stolze LK, Zhao S, Martens L, Kendall TJ, Thone T, Amancherla K, Bailin S, Gabriel CL, Koethe J, Carr JJ, Terry JG, Freedman J, Tanriverdi K, Alsop E, Keuren-Jensen KV, Sauld JFK, Mahajan G, Khan S, Colangelo L, Nayor M, Fisher-Hoch S, McCormick J, North KE, Below J, Wells Q, Abel D, Kalhan R, Scott C, Guilliams M, Fallowfield JA, Banovich NE, Das S, Shah R. A prognostic molecular signature of hepatic steatosis is spatially heterogeneous and dynamic in human liver. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.26.24301828. [PMID: 38352394 PMCID: PMC10863022 DOI: 10.1101/2024.01.26.24301828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) prevalence is increasing in parallel with an obesity pandemic, calling for novel strategies for prevention and treatment. We defined a circulating proteome of human MASLD across ≈7000 proteins in ≈5000 individuals from diverse, at-risk populations across the metabolic health spectrum, demonstrating reproducible diagnostic performance and specifying both known and novel metabolic pathways relevant to MASLD (central carbon and amino acid metabolism, hepatocyte regeneration, inflammation, fibrosis, insulin sensitivity). A parsimonious proteomic signature of MASLD was associated with a protection from MASLD and its related multi-system metabolic consequences in >26000 free-living individuals, with an additive effect to polygenic risk. The MASLD proteome was encoded by genes that demonstrated transcriptional enrichment in liver, with spatial transcriptional activity in areas of steatosis in human liver biopsy and dynamicity for select targets in human liver across stages of steatosis. We replicated several top relations from proteomics and spatial tissue transcriptomics in a humanized "liver-on-a-chip" model of MASLD, highlighting the power of a full translational approach to discovery in MASLD. Collectively, these results underscore utility of blood-based proteomics as a dynamic "liquid biopsy" of human liver relevant to clinical biomarker and mechanistic applications.
Collapse
|
44
|
Pierfelice TV, D'Amico E, Petrini M, Romano M, D'Arcangelo C, Sbordone L, Barone A, Plebani R, Iezzi G. A Systematic Review on Organ-on-a-Chip in PDMS or Hydrogel in Dentistry: An Update of the Literature. Gels 2024; 10:102. [PMID: 38391432 PMCID: PMC10887950 DOI: 10.3390/gels10020102] [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: 01/02/2024] [Revised: 01/19/2024] [Accepted: 01/24/2024] [Indexed: 02/24/2024] Open
Abstract
Organs-on-a-chip (OoCs) are microfluidic devices constituted by PDMS or hydrogel in which different layers of cells are separated by a semipermeable membrane. This technology can set many parameters, like fluid shear stress, chemical concentration gradient, tissue-organ interface, and cell interaction. The use of these devices in medical research permits the investigation of cell patterning, tissue-material interface, and organ-organ interaction, mimicking the complex structures and microenvironment of human and animal bodies. This technology allows us to reconstitute in vitro complex conditions that recapitulate in vivo environments. One of the main advantages of these systems is that they represent a very realistic model that, in many cases, can replace animal experimentation, eliminating costs and related ethical issues. Organ-on-a-chip can also contain bacteria or cancer cells. This technology could be beneficial in dentistry for testing novel antibacterial substances and biomaterials, performing studies on inflammatory disease, or planning preclinical studies. A significant number of publications and reviews have been published on this topic. Still, to our knowledge, they mainly focus on the materials used for fabrication and the different patterns of the chip applied to the experimentations. This review presents the most recent applications of organ-on-a-chip models in dentistry, starting from the reconstituted dental tissues to their clinical applications and future perspectives.
Collapse
Affiliation(s)
- Tania Vanessa Pierfelice
- Department of Medical, Oral and Biotechnological Sciences, University G. d'Annunzio of Chieti-Pescara, 66100 Chieti, Italy
| | - Emira D'Amico
- Department of Medical, Oral and Biotechnological Sciences, University G. d'Annunzio of Chieti-Pescara, 66100 Chieti, Italy
| | - Morena Petrini
- Department of Medical, Oral and Biotechnological Sciences, University G. d'Annunzio of Chieti-Pescara, 66100 Chieti, Italy
| | - Mario Romano
- Department of Medical, Oral and Biotechnological Sciences, University G. d'Annunzio of Chieti-Pescara, 66100 Chieti, Italy
| | - Camillo D'Arcangelo
- Department of Medical, Oral and Biotechnological Sciences, University G. d'Annunzio of Chieti-Pescara, 66100 Chieti, Italy
| | - Ludovico Sbordone
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, 86100 Campobasso, Italy
| | - Antonio Barone
- Department of Surgical, Medical, Molecular Pathologies and of the Critical Needs, School of Dentistry, University of Pisa, 56126 Pisa, Italy
- Complex Unit of Stomatology and Oral Surgery, University Hospital of Pisa, 56126 Pisa, Italy
| | - Roberto Plebani
- Department of Medical, Oral and Biotechnological Sciences, University G. d'Annunzio of Chieti-Pescara, 66100 Chieti, Italy
| | - Giovanna Iezzi
- Department of Medical, Oral and Biotechnological Sciences, University G. d'Annunzio of Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
45
|
Jiang L, Khawaja H, Tahsin S, Clarkson TA, Miranti CK, Zohar Y. Microfluidic-based human prostate-cancer-on-chip. Front Bioeng Biotechnol 2024; 12:1302223. [PMID: 38322789 PMCID: PMC10844564 DOI: 10.3389/fbioe.2024.1302223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 01/09/2024] [Indexed: 02/08/2024] Open
Abstract
Lack of adequate models significantly hinders advances in prostate cancer treatment, where resistance to androgen-deprivation therapies and bone metastasis remain as major challenges. Current in vitro models fail to faithfully mimic the complex prostate physiology. In vivo animal models can shed light on the oncogenes involved in prostate cancer development and progression; however, the animal prostate gland is fundamentally different from that of human, and the underlying genetic mechanisms are different. To address this problem, we developed the first in vitro microfluidic human Prostate-Cancer-on-Chip (PCoC) model, where human prostate cancer and stromal fibroblast cells were co-cultivated in two channels separated by a porous membrane under culture medium flow. The established microenvironment enables soluble signaling factors secreted by each culture to locally diffuse through the membrane pores affecting the neighboring culture. We particularly explored the conversion of the stromal fibroblasts into cancer-associated fibroblasts (CAFs) due to the interaction between the 2 cell types. Immunofluorescence microscopy revealed that tumor cells induced CAF biomarkers, αSMA and COL1A1, in stromal fibroblasts. The stromal CAF conversion level was observed to increase along the flow direction in response to diffusion agents, consistent with simulations of solute concentration gradients. The tumor cells also downregulated androgen receptor (AR) expression in stromal fibroblasts, while an adequate level of stromal AR expression is maintained in normal prostate homeostasis. We further investigated tumor invasion into the stroma, an early step in the metastatic cascade, in devices featuring a serpentine channel with orthogonal channel segments overlaying a straight channel and separated by an 8 µm-pore membrane. Both tumor cells and stromal CAFs were observed to cross over into their neighboring channel, and the stroma's role seemed to be proactive in promoting cell invasion. As control, normal epithelial cells neither induced CAF conversion nor promoted cell invasion. In summary, the developed PCoC model allows spatiotemporal analysis of the tumor-stroma dynamic interactions, due to bi-directional signaling and physical contact, recapitulating tissue-level multicellular responses associated with prostate cancer in vivo. Hence, it can serve as an in vitro model to dissect mechanisms in human prostate cancer development and seek advanced therapeutic strategies.
Collapse
Affiliation(s)
- Linan Jiang
- Department of Aerospace and Mechanical Engineering, Tucson, AZ, United States
| | - Hunain Khawaja
- Cancer Biology Graduate Interdisciplinary Program, Tucson, AZ, United States
| | - Shekha Tahsin
- Cancer Biology Graduate Interdisciplinary Program, Tucson, AZ, United States
| | | | - Cindy K Miranti
- Department of Molecular and Cellular Biology, Tucson, AZ, United States
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, United States
| | - Yitshak Zohar
- Department of Aerospace and Mechanical Engineering, Tucson, AZ, United States
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
46
|
Parvatam S, Pamies D, Pistollato F, Beken S, Mariappan I, Roth A, Piergiovanni M, G C Maisonneuve B, Ewart L, Majumder A, Dandekar P, Date R, Mahadik K, Thiyagarajan S, Coecke S. Taking the leap toward human-specific nonanimal methodologies: The need for harmonizing global policies for microphysiological systems. Stem Cell Reports 2024; 19:37-40. [PMID: 38134927 PMCID: PMC10828677 DOI: 10.1016/j.stemcr.2023.11.008] [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: 08/26/2023] [Revised: 11/20/2023] [Accepted: 11/20/2023] [Indexed: 12/24/2023] Open
Abstract
With a recent amendment, India joined other countries that have removed the legislative barrier toward the use of human-relevant methods in drug development. Here, global stakeholders weigh in on the urgent need to globally harmonize the guidelines toward the standardization of microphysiological systems. We discuss a possible framework for establishing scientific confidence and regulatory approval of these methods.
Collapse
Affiliation(s)
| | - David Pamies
- SCAHT - Swiss Centre for Applied Human Toxicology. University of Basel. Missionsstrasse 64, 4055 Basel, Switzerland
| | | | - Sonja Beken
- Federal Agency for Medicines and Health Products, Brussels, Belgium
| | | | | | | | | | - Lorna Ewart
- Emulate Inc, 27 Drydock Avenue, Boston, MA, USA
| | | | | | | | - Kasturi Mahadik
- Centre for Predictive Human Model Systems, Atal Incubation Centre-Centre for Cellular and Molecular Biology (AIC-CCMB), Hyderabad, India
| | | | - Sandra Coecke
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| |
Collapse
|
47
|
Perez-Araluce M, Jüngst T, Sanmartin C, Prosper F, Plano D, Mazo MM. Biomaterials-Based Antioxidant Strategies for the Treatment of Oxidative Stress Diseases. Biomimetics (Basel) 2024; 9:23. [PMID: 38248597 PMCID: PMC10813727 DOI: 10.3390/biomimetics9010023] [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: 11/17/2023] [Revised: 12/14/2023] [Accepted: 12/27/2023] [Indexed: 01/23/2024] Open
Abstract
Oxidative stress is characterized by an increase in reactive oxygen species or a decrease in antioxidants in the body. This imbalance leads to detrimental effects, including inflammation and multiple chronic diseases, ranging from impaired wound healing to highly impacting pathologies in the neural and cardiovascular systems, or the bone, amongst others. However, supplying compounds with antioxidant activity is hampered by their low bioavailability. The development of biomaterials with antioxidant capacity is poised to overcome this roadblock. Moreover, in the treatment of chronic inflammation, material-based strategies would allow the controlled and targeted release of antioxidants into the affected tissue. In this review, we revise the main causes and effects of oxidative stress, and survey antioxidant biomaterials used for the treatment of chronic wounds, neurodegenerative diseases, cardiovascular diseases (focusing on cardiac infarction, myocardial ischemia-reperfusion injury and atherosclerosis) and osteoporosis. We anticipate that these developments will lead to the emergence of new technologies for tissue engineering, control of oxidative stress and prevention of diseases associated with oxidative stress.
Collapse
Affiliation(s)
- Maria Perez-Araluce
- Biomedical Engineering Program, Enabling Technologies Division, CIMA Universidad de Navarra, 31008 Pamplona, Spain;
| | - Tomasz Jüngst
- Department for Functional Materials in Medicine and Dentistry, Institute of Functional Materials and Biofabrication, University of Würzburg, D-97070 Würzburg, Germany
- Bavarian Polymer Institute, University of Bayreuth, 95447 Bayreuth, Germany
| | - Carmen Sanmartin
- Department of Pharmaceutical Science, Universidad de Navarra, 31008 Pamplona, Spain;
| | - Felipe Prosper
- Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
- Centro de Investigacion Biomedica en Red de Cancer (CIBERONC) CB16/12/00489, 28029 Madrid, Spain
- Hemato-Oncology Program, Cancer Division, CIMA Universidad de Navarra, 31008 Pamplona, Spain
| | - Daniel Plano
- Department of Pharmaceutical Science, Universidad de Navarra, 31008 Pamplona, Spain;
| | - Manuel M. Mazo
- Biomedical Engineering Program, Enabling Technologies Division, CIMA Universidad de Navarra, 31008 Pamplona, Spain;
- Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
| |
Collapse
|
48
|
Kasturi M, Mathur V, Gadre M, Srinivasan V, Vasanthan KS. Three Dimensional Bioprinting for Hepatic Tissue Engineering: From In Vitro Models to Clinical Applications. Tissue Eng Regen Med 2024; 21:21-52. [PMID: 37882981 PMCID: PMC10764711 DOI: 10.1007/s13770-023-00576-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/07/2023] [Accepted: 07/11/2023] [Indexed: 10/27/2023] Open
Abstract
Fabrication of functional organs is the holy grail of tissue engineering and the possibilities of repairing a partial or complete liver to treat chronic liver disorders are discussed in this review. Liver is the largest gland in the human body and plays a responsible role in majority of metabolic function and processes. Chronic liver disease is one of the leading causes of death globally and the current treatment strategy of organ transplantation holds its own demerits. Hence there is a need to develop an in vitro liver model that mimics the native microenvironment. The developed model should be a reliable to understand the pathogenesis, screen drugs and assist to repair and replace the damaged liver. The three-dimensional bioprinting is a promising technology that recreates in vivo alike in vitro model for transplantation, which is the goal of tissue engineers. The technology has great potential due to its precise control and its ability to homogeneously distribute cells on all layers in a complex structure. This review gives an overview of liver tissue engineering with a special focus on 3D bioprinting and bioinks for liver disease modelling and drug screening.
Collapse
Affiliation(s)
- Meghana Kasturi
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Vidhi Mathur
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Mrunmayi Gadre
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Varadharajan Srinivasan
- Department of Civil Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Kirthanashri S Vasanthan
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
49
|
Wang L, Hu D, Xu J, Hu J, Wang Y. Complex in vitro Model: A Transformative Model in Drug Development and Precision Medicine. Clin Transl Sci 2023; 17:e13695. [PMID: 38062923 PMCID: PMC10828975 DOI: 10.1111/cts.13695] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/25/2023] [Accepted: 11/18/2023] [Indexed: 02/02/2024] Open
Abstract
In vitro and in vivo models play integral roles in preclinical drug research, evaluation, and precision medicine. In vitro models primarily involve research platforms based on cultured cells, typically in the form of two-dimensional (2D) cell models. However, notable disparities exist between 2D cultured cells and in vivo cells across various aspects, rendering the former inadequate for replicating the physiologically relevant functions of human or animal organs and tissues. Consequently, these models failed to accurately reflect real-life scenarios post-drug administration. Complex in vitro models (CIVMs) refer to in vitro models that integrate a multicellular environment and a three-dimensional (3D) structure using bio-polymer or tissue-derived matrices. These models seek to reconstruct the organ- or tissue-specific characteristics of the extracellular microenvironment. The utilization of CIVMs allows for enhanced physiological correlation of cultured cells, thereby better mimicking in vivo conditions without ethical concerns associated with animal experimentation. Consequently, CIVMs have gained prominence in disease research and drug development. This review aimed to comprehensively examine and analyze the various types, manufacturing techniques, and applications of CIVM in the domains of drug discovery, drug development, and precision medicine. The objective of this study was to provide a comprehensive understanding of the progress made in CIVMs and their potential future use in these fields.
Collapse
Affiliation(s)
- Luming Wang
- Department of Thoracic SurgeryThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Key Laboratory of Clinical Evaluation Technology for Medical Device of Zhejiang ProvinceHangzhouChina
| | - Danping Hu
- Hangzhou Chexmed Technology Co., Ltd.HangzhouChina
| | - Jinming Xu
- Department of Thoracic SurgeryThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Key Laboratory of Clinical Evaluation Technology for Medical Device of Zhejiang ProvinceHangzhouChina
| | - Jian Hu
- Department of Thoracic SurgeryThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Key Laboratory of Clinical Evaluation Technology for Medical Device of Zhejiang ProvinceHangzhouChina
| | - Yifei Wang
- Hangzhou Chexmed Technology Co., Ltd.HangzhouChina
| |
Collapse
|
50
|
McGill MR, Curry SC. The Evolution of Circulating Biomarkers for Use in Acetaminophen/Paracetamol-Induced Liver Injury in Humans: A Scoping Review. LIVERS 2023; 3:569-596. [PMID: 38434489 PMCID: PMC10906739 DOI: 10.3390/livers3040039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/05/2024] Open
Abstract
Acetaminophen (APAP) is a widely used drug, but overdose can cause severe acute liver injury. The first reports of APAP hepatotoxicity in humans were published in 1966, shortly after the development of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) as the first biomarkers of liver injury as opposed to liver function. Thus, the field of liver injury biomarkers has evolved alongside the growth in APAP hepatotoxicity incidence. Numerous biomarkers have been proposed for use in the management of APAP overdose patients in the intervening years. Here, we comprehensively review the development of these markers from the 1960s to the present day and briefly discuss possible future directions.
Collapse
Affiliation(s)
- Mitchell R McGill
- Dept. of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR 72212, USA
- Dept. of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72212, USA
- Dept. of Pathology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72212, USA
| | - Steven C Curry
- Division of Clinical Data Analytics and Decision Support, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85006, USA
- Department of Medical Toxicology, Banner-University Medical Center Phoenix, Phoenix, AZ 85006, USA
| |
Collapse
|