51
|
Qian W, Gong G, Su H, Zhao Y, Fu W, Wang Y, Ji W, Sun X, Zhang B, Ma L, Li J, Zhang X, Li S, Sheng E, Lu Y, Zhu D. Hepar-on-a-sensor-platform with hybridization chain reaction amplification strategy to intuitively monitor the hepatoxicity of natural compounds. Acta Biomater 2023; 160:73-86. [PMID: 36804823 DOI: 10.1016/j.actbio.2023.02.021] [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: 10/20/2022] [Revised: 02/09/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023]
Abstract
The irrational use of natural compounds in the treatment of diseases can lead to serious side effects, especially hepatoxicity, and its toxic effects are usually cumulative and imperceptible. Therefore, an accurate sensing platform is urgently needed to monitor the hepatotoxicity of natural compounds. Here, we deposited a thermo-responsive alginate-RGD/Pluronic hydrogel to construct an in vitro three-dimensional(3D) hepar-platform, and a thorough validation was adopted to evaluate the bioprinted hepatic constructs. The engineered hepar-platform was then employed to access its biological response toward Emodin (EM) and Triptolide (TP), two typical hepatotoxic natural compounds. Subsequently, we integrated it with a robust fluorescent sensor based on hybridization chain reaction amplification strategy (HCR) to monitor the early hepatotoxic biomarker - glutathione-S-transferase-alpha (GST-α) secreted by this 3D constructs. Our study was the first attempt to construct an accurate hepar-on-a-sensor platform that could effectively detect GST-α for monitoring the hepatoxic effects of natural compounds. The limit of detection of the platform was 0.3 ng ml-1 and the accuracy of this platform was verified by enzyme linked immunosorbent assay. Furthermore, the variation of GST-α induced by EM and TP was consistent with hepatotoxicity studies, thus providing an important application value for evaluating the hepatotoxicity of natural compounds. STATEMENT OF SIGNIFICANCE: 1. We deposited a thermo-responsive alginate-RGD/Pluronic hydrogel to construct an in vitro three-dimensional(3D) hepar-platform, and elucidated the essential reasons why hybrid bioinks more suitable for 3D extrusion from biomaterials itself. Also, a thorough validation associated with a series of important proteins and genes involved in liver cell metabolism was adopted to evaluate the bioprinted hepatic constructs accurately 2. Glutathione-S-transferase-alpha is a soluble trace biomarker for acute hepatotoxic injury, the hepatotoxic effects of natural compounds on the secretion of GST-α has not been reported to date. We integrated our 3D hepar-platform with recognition molecules-aptamers and HCR amplification strategy to monitor the variation of GST-α, aiming at developing a robust and stable fluorescent biosensing platform to monitor the hepatoxicity of natural compounds.
Collapse
Affiliation(s)
- Wenhui Qian
- School of Pharmacy, Nanjing University of Chinese Medicine,Nanjing, Jiangsu 210023, PR China; Department of Pharmacy, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210002, PR China
| | - Guangming Gong
- Department of Pharmacy, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210002, PR China
| | - Hua Su
- Department of Pharmacy, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210002, PR China
| | - Yang Zhao
- Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Wenjuan Fu
- School of Pharmacy, Nanjing University of Chinese Medicine,Nanjing, Jiangsu 210023, PR China
| | - Yuting Wang
- School of Pharmacy, Nanjing University of Chinese Medicine,Nanjing, Jiangsu 210023, PR China
| | - Wenwen Ji
- School of Pharmacy, Nanjing University of Chinese Medicine,Nanjing, Jiangsu 210023, PR China
| | - Xuetong Sun
- School of Pharmacy, Nanjing University of Chinese Medicine,Nanjing, Jiangsu 210023, PR China
| | - Bei Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine,Nanjing, Jiangsu 210023, PR China
| | - Lijuan Ma
- School of Pharmacy, Nanjing University of Chinese Medicine,Nanjing, Jiangsu 210023, PR China
| | - Jianting Li
- School of Pharmacy, Nanjing University of Chinese Medicine,Nanjing, Jiangsu 210023, PR China
| | - Xiangying Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine,Nanjing, Jiangsu 210023, PR China
| | - Su Li
- School of Pharmacy, Nanjing University of Chinese Medicine,Nanjing, Jiangsu 210023, PR China
| | - Enze Sheng
- School of Pharmacy, Nanjing University of Chinese Medicine,Nanjing, Jiangsu 210023, PR China
| | - Yin Lu
- School of Pharmacy, Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, PR China.
| | - Dong Zhu
- School of Pharmacy, Nanjing University of Chinese Medicine,Nanjing, Jiangsu 210023, PR China.
| |
Collapse
|
52
|
Pognan F, Beilmann M, Boonen HCM, Czich A, Dear G, Hewitt P, Mow T, Oinonen T, Roth A, Steger-Hartmann T, Valentin JP, Van Goethem F, Weaver RJ, Newham P. The evolving role of investigative toxicology in the pharmaceutical industry. Nat Rev Drug Discov 2023; 22:317-335. [PMID: 36781957 PMCID: PMC9924869 DOI: 10.1038/s41573-022-00633-x] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2022] [Indexed: 02/15/2023]
Abstract
For decades, preclinical toxicology was essentially a descriptive discipline in which treatment-related effects were carefully reported and used as a basis to calculate safety margins for drug candidates. In recent years, however, technological advances have increasingly enabled researchers to gain insights into toxicity mechanisms, supporting greater understanding of species relevance and translatability to humans, prediction of safety events, mitigation of side effects and development of safety biomarkers. Consequently, investigative (or mechanistic) toxicology has been gaining momentum and is now a key capability in the pharmaceutical industry. Here, we provide an overview of the current status of the field using case studies and discuss the potential impact of ongoing technological developments, based on a survey of investigative toxicologists from 14 European-based medium-sized to large pharmaceutical companies.
Collapse
Affiliation(s)
- Francois Pognan
- Discovery and Investigative Safety, Novartis Pharma AG, Basel, Switzerland.
| | - Mario Beilmann
- Nonclinical Drug Safety Germany, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Harrie C M Boonen
- Drug Safety, Dept of Exploratory Toxicology, Lundbeck A/S, Valby, Denmark
| | | | - Gordon Dear
- In Vitro In Vivo Translation, GlaxoSmithKline David Jack Centre for Research, Ware, UK
| | - Philip Hewitt
- Chemical and Preclinical Safety, Merck Healthcare KGaA, Darmstadt, Germany
| | - Tomas Mow
- Safety Pharmacology and Early Toxicology, Novo Nordisk A/S, Maaloev, Denmark
| | - Teija Oinonen
- Preclinical Safety, Orion Corporation, Espoo, Finland
| | - Adrian Roth
- Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | | | | | - Freddy Van Goethem
- Predictive, Investigative & Translational Toxicology, Nonclinical Safety, Janssen Research & Development, Beerse, Belgium
| | - Richard J Weaver
- Innovation Life Cycle Management, Institut de Recherches Internationales Servier, Suresnes, France
| | - Peter Newham
- Clinical Pharmacology and Safety Sciences, AstraZeneca R&D, Cambridge, UK.
| |
Collapse
|
53
|
Yang S, Ooka M, Margolis RJ, Xia M. Liver three-dimensional cellular models for high-throughput chemical testing. CELL REPORTS METHODS 2023; 3:100432. [PMID: 37056374 PMCID: PMC10088249 DOI: 10.1016/j.crmeth.2023.100432] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
Drug-induced hepatotoxicity is a leading cause of drug withdrawal from the market. High-throughput screening utilizing in vitro liver models is critical for early-stage liver toxicity testing. Traditionally, monolayer human hepatocytes or immortalized liver cell lines (e.g., HepG2, HepaRG) have been used to test compound liver toxicity. However, monolayer-cultured liver cells sometimes lack the metabolic competence to mimic the in vivo condition and are therefore largely appropriate for short-term toxicological testing. They may not, however, be adequate for identifying chronic and recurring liver damage caused by drugs. Recently, several three-dimensional (3D) liver models have been developed. These 3D liver models better recapitulate normal liver function and metabolic capacity. This review describes the current development of 3D liver models that can be used to test drugs/chemicals for their pharmacologic and toxicologic effects, as well as the advantages and limitations of using these 3D liver models for high-throughput screening.
Collapse
Affiliation(s)
- Shu Yang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, USA
| | - Masato Ooka
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ryan Jared Margolis
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, USA
| | - Menghang Xia
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
54
|
Kang SY, Kimura M, Shrestha S, Lewis P, Lee S, Cai Y, Joshi P, Acharya P, Liu J, Yang Y, Sanchez JG, Ayyagari S, Alsberg E, Wells JM, Takebe T, Lee MY. A Pillar and Perfusion Plate Platform for Robust Human Organoid Culture and Analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.11.532210. [PMID: 36993405 PMCID: PMC10055006 DOI: 10.1101/2023.03.11.532210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Human organoids have potential to revolutionize in vitro disease modeling by providing multicellular architecture and function that are similar to those in vivo . This innovative and evolving technology, however, still suffers from assay throughput and reproducibility to enable high-throughput screening (HTS) of compounds due to cumbersome organoid differentiation processes and difficulty in scale-up and quality control. Using organoids for HTS is further challenged by lack of easy-to-use fluidic systems that are compatible with relatively large organoids. Here, we overcome these challenges by engineering "microarray three-dimensional (3D) bioprinting" technology and associated pillar and perfusion plates for human organoid culture and analysis. High-precision, high-throughput stem cell printing and encapsulation techniques were demonstrated on a pillar plate, which was coupled with a complementary deep well plate and a perfusion well plate for static and dynamic organoid culture. Bioprinted cells and spheroids in hydrogels were differentiated into liver and intestine organoids for in situ functional assays. The pillar/perfusion plates are compatible with standard 384-well plates and HTS equipment, and thus may be easily adopted in current drug discovery efforts.
Collapse
Affiliation(s)
- Soo-Yeon Kang
- Department of Biomedical Engineering, University of North Texas
| | - Masaki Kimura
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center
| | - Sunil Shrestha
- Department of Biomedical Engineering, University of North Texas
| | - Phillip Lewis
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center
| | - Sangjoon Lee
- Department of Biomedical Engineering, University of North Texas
| | - Yuqi Cai
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center
| | | | - Prabha Acharya
- Department of Biomedical Engineering, University of North Texas
| | - Jiafeng Liu
- Department of Biomedical Engineering, University of North Texas
| | - Yong Yang
- Department of Biomedical Engineering, University of North Texas
| | - J Guillermo Sanchez
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center
| | - Sriramya Ayyagari
- Department of Biomedical Engineering, University of Illinois at Chicago
| | - Eben Alsberg
- Department of Biomedical Engineering, University of Illinois at Chicago
- Departments of Orthopedics, Pharmacology, and Mechanical and Industrial Engineering, University of Illinois at Chicago
| | - James M Wells
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center
| | - Takanori Takebe
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center
| | - Moo-Yeal Lee
- Department of Biomedical Engineering, University of North Texas
- Bioprinting Laboratories Inc
| |
Collapse
|
55
|
Carpentier N, Urbani L, Dubruel P, Van Vlierberghe S. The native liver as inspiration to create superior in vitro hepatic models. Biomater Sci 2023; 11:1091-1115. [PMID: 36594602 DOI: 10.1039/d2bm01646j] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Drug induced liver injury (DILI) is one of the major reasons of drug withdrawal during the different phases of drug development. The later in the drug development a drug is discovered to be toxic, the higher the economical as well as the ethical impact will be. In vitro models for early detection of drug liver toxicity are under constant development, however to date a superior model of the liver is still lacking. Ideally, a highly reliable model should be established to maintain the different hepatic cell functionalities to the greatest extent possible, during a period of time long enough to allow for tracking of the toxicity of compounds. In the case of DILI, toxicity can appear even after months of exposure. To reach this goal, an in vitro model should be developed that mimics the in vivo liver environment, function and response to external stimuli. The different approaches for the development of liver models currently used in the field of tissue engineering will be described in this review. Combining different technologies, leading to optimal materials, cells and 3D-constructs will ultimately lead to an ideal superior model that fully recapitulates the liver.
Collapse
Affiliation(s)
- Nathan Carpentier
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium.
| | - Luca Urbani
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London SE5 9NT, UK.,Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Peter Dubruel
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium.
| | - Sandra Van Vlierberghe
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium.
| |
Collapse
|
56
|
Bentwich I. Pharma's Bio-AI revolution. Drug Discov Today 2023; 28:103515. [PMID: 36736581 DOI: 10.1016/j.drudis.2023.103515] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 01/13/2023] [Accepted: 01/25/2023] [Indexed: 02/05/2023]
Abstract
Drug development has become unbearably slow and expensive. A key underlying problem is the clinical prediction challenge: the inability to predict which drug candidates will be safe in the human body and for whom. Recently, a dramatic regulatory change has removed FDA's mandated reliance on antiquated, ineffective animal studies. A new frontier is an integration of several disruptive technologies [machine learning (ML), patient-on-chip, real-time sensing, and stem cells], which when integrated, have the potential to address this challenge, drastically cutting the time and cost of developing drugs, and tailoring them to individual patients.
Collapse
Affiliation(s)
- Isaac Bentwich
- Quris-AI, 6 HaNatsiv Street, Tel Aviv-Yafo 6701033, Israel.
| |
Collapse
|
57
|
Tao F, Hanada S, Matsushima K, Arakawa H, Ishida N, Kato Y, Okimura S, Watanabe T, Kojima N. Enhancement and maintenance of hepatic metabolic functions by controlling 3D aggregation of cryopreserved human iPS cell-derived hepatocyte-like cells. J Biosci Bioeng 2023; 135:134-142. [PMID: 36481309 DOI: 10.1016/j.jbiosc.2022.10.006] [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: 07/31/2022] [Revised: 10/05/2022] [Accepted: 10/14/2022] [Indexed: 12/12/2022]
Abstract
Hepatocyte-like cells (HLCs) generated from human induced pluripotent stem cells are potent cells to study individual-specific hepatotoxicity for drug screening test. However, the functions of metabolic enzymes are practically low. Here, we reconstituted stable and compact 3D spheroids of commercially available cryopreserved HLCs by our original spheroid formation method with high viscous methylcellulose medium. 3D formation enhanced the hepatic functions and maintained the functions for 14 days. Especially, the expression of cytochrome P450s was 10- to 100-fold enhanced compared to conventional 2D culture, which is applicable to a typical drug-metabolizing test using liquid chromatograph-tandem mass spectrometer. In conclusion, we successfully formed human HLC spheroid from commercially available cryo-preserved cells, which realized remarkable hepatic maturation by prolonged 3D culture, especially in terms of drug-metabolizing enzymes. Our spheroid formation technology has the potential to make HLC spheroids a potent tool in aspects of pharmaceutical research, such as drug screening and pharmacokinetic studies.
Collapse
Affiliation(s)
- Fumiya Tao
- Department of Life and Environmental System Science, Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, Kanagawa 236-0027, Japan
| | - Sanshiro Hanada
- Department of Life and Environmental System Science, Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, Kanagawa 236-0027, Japan
| | - Kazuya Matsushima
- Department of Life and Environmental System Science, Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, Kanagawa 236-0027, Japan
| | - Hiroshi Arakawa
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan
| | - Naoki Ishida
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan
| | - Yukio Kato
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan
| | - Saya Okimura
- REPROCELL Inc., 3-8-11 Shin-yokohama, Kohoku-ku, Yokohama, Kanagawa 222-0033, Japan
| | - Tomohisa Watanabe
- Shonan Laboratory, REPROCELL Inc., 2-26-1 Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan
| | - Nobuhiko Kojima
- Department of Life and Environmental System Science, Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, Kanagawa 236-0027, Japan.
| |
Collapse
|
58
|
Horn G, Kranawetvogl T, John H, Weigel C, Rauen U, Worek F, Wille T. Human HepaRG liver spheroids: cold storage protocol and study on pyridinium oxime-induced hepatotoxicity in vitro. Chem Biol Interact 2023; 369:110285. [PMID: 36442613 DOI: 10.1016/j.cbi.2022.110285] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/28/2022] [Accepted: 11/22/2022] [Indexed: 11/27/2022]
Abstract
Oximes play an essential role in the therapy of organophosphorus compound (OP) poisoning by reactivating inhibited acetylcholinesterase. Impairment of liver function was observed in OP poisoning and associated with obidoxime treatment by some reports. In this study human three-dimensional HepaRG spheroids were used as complex in vitro model to investigate oxime-induced liver toxicity. In this context, cold storage of liver spheroids at 4 °C in standard culture medium and in optimized tissue preservation solutions of up to 72 h was assessed. Cold storage in standard culture medium resulted in a complete loss of viability whereas an optimized tissue preservation solution preserved viability. Separately from that liver spheroids were exposed to the four oximes pralidoxime, obidoxime, HI-6, MMB-4 and cytotoxicity (effective concentration, EC50) was determined with an ATP-based assay at several time points. The release of aspartate aminotransferase (AST), alanine aminotransferase (ALT) and albumin secretion was measured in supernatants. The same parameters were assessed with diclofenac as positive hepatotoxic control and with the OP pesticides malathion and malaoxon alone or in the presence of obidoxime. All individual tested oximes and OP showed a low cytotoxicity with effective concentrations mostly >2,000 μM. In contrast, the exposure to malaoxon in the presence of 1,000 μM obidoxime resulted in a marked decrease of viability and an increased release of AST indicating risk of liver injury only if oxime antidotes are strongly overdosed.
Collapse
Affiliation(s)
- Gabriele Horn
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstrasse 11, 80937, Munich, Germany
| | - Tamara Kranawetvogl
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstrasse 11, 80937, Munich, Germany
| | - Harald John
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstrasse 11, 80937, Munich, Germany
| | - Carlotta Weigel
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstrasse 11, 80937, Munich, Germany
| | - Ursula Rauen
- Institut für Physiologische Chemie, Universitätsklinikum, Hufelandstrasse 55, 45122, Essen, Germany
| | - Franz Worek
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstrasse 11, 80937, Munich, Germany
| | - Timo Wille
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstrasse 11, 80937, Munich, Germany.
| |
Collapse
|
59
|
Chipangura JK, Ntamo Y, Mohr B, Chellan N. A review of challenges and prospects of 3D cell-based culture models used for studying drug induced liver injury during early phases of drug development. Hum Exp Toxicol 2023; 42:9603271221147884. [PMID: 36879529 DOI: 10.1177/09603271221147884] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
Drug-induced liver injury (DILI) is the leading cause of compound attrition during drug development. Over the years, a battery of in-vitro cell culture toxicity tests is being conducted to evaluate the toxicity of compounds prior to testing in laboratory animals. Two-dimensional (2D) in-vitro cell culture models are commonly used and have provided a great deal of knowledge; however, these models often fall short in mimicking natural structures of tissues in-vivo. Testing in humans is the most logical method, but unfortunately there are ethical limitations associated with human tests. To overcome these limitations better human-relevant, predictive models are required. The past decade has witnessed significant efforts towards the development of three-dimensional (3D) in-vitro cell culture models better mimicking in-vivo physiology. 3D cell culture has advantages in being representative of the interactions of cells in-vivo and when validated can act as an interphase between 2D cell culture models and in-vivo animal models. The current review seeks to provide an overview of the challenges that make biomarkers used for detection of DILI not to be sensitive enough during drug development and explore how 3D cell culture models can be used to address the gap with the current models.
Collapse
Affiliation(s)
- John K Chipangura
- Faculty of Health Science, University of Cape Town Research Animal Facility, South Africa
| | - Yonela Ntamo
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
| | - Bert Mohr
- Faculty of Health Science, University of Cape Town Research Animal Facility, South Africa
| | - Nireshni Chellan
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- Centre for Cardio-metabolic Research in Africa (CARMA), Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, South Africa
| |
Collapse
|
60
|
Karabicici M, Akbari S, Ertem O, Gumustekin M, Erdal E. Human Liver Organoid Models for Assessment of Drug Toxicity at the Preclinical Stage. Endocr Metab Immune Disord Drug Targets 2023; 23:1713-1724. [PMID: 37055905 DOI: 10.2174/1871530323666230411100121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 01/14/2023] [Accepted: 02/23/2023] [Indexed: 04/15/2023]
Abstract
The hepatotoxicity of drugs is one of the leading causes of drug withdrawal from the pharmaceutical market and high drug attrition rates. Currently, the commonly used hepatocyte models include conventional hepatic cell lines and animal models, which cannot mimic human drug-induced liver injury (DILI) due to poorly defined dose-response relationships and/or lack of human-specific mechanisms of toxicity. In comparison to 2D culture systems from different cell sources such as primary human hepatocytes and hepatomas, 3D organoids derived from an inducible pluripotent stem cell (iPSC) or adult stem cells are promising accurate models to mimic organ behavior with a higher level of complexity and functionality owing to their ability to self-renewal. Meanwhile, the heterogeneous cell composition of the organoids enables metabolic and functional zonation of hepatic lobule important in drug detoxification and has the ability to mimic idiosyncratic DILI as well. Organoids having higher drug-metabolizing enzyme capacities can culture long-term and be combined with microfluidic-based technologies such as organ-on-chips for a more precise representation of human susceptibility to drug response in a high-throughput manner. However, there are numerous limitations to be considered about this technology, such as enough maturation, differences between protocols and high cost. Herein, we first reviewed the current preclinical DILI assessment tools and looked at the organoid technology with respect to in vitro detoxification capacities. Then we discussed the clinically applicable DILI assessment markers and the importance of liver zonation in the next generation organoid- based DILI models.
Collapse
Affiliation(s)
- Mustafa Karabicici
- Izmir Biomedicine and Genome Center (IBG-Izmir), Dokuz Eylul University Health Campus, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, CA, USA
| | - Soheil Akbari
- Izmir Biomedicine and Genome Center (IBG-Izmir), Dokuz Eylul University Health Campus, Izmir, Turkey
| | - Ozge Ertem
- Department of Pharmacology, Izmir Bozyaka Training and Research Hospital, Izmir, Turkey
| | - Mukaddes Gumustekin
- Department of Pharmacology, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Esra Erdal
- Izmir Biomedicine and Genome Center (IBG-Izmir), Dokuz Eylul University Health Campus, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
- Department of Medical Biology and Genetics, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| |
Collapse
|
61
|
Performance assessment and economic analysis of a human Liver-Chip for predictive toxicology. COMMUNICATIONS MEDICINE 2022; 2:154. [PMID: 36473994 PMCID: PMC9727064 DOI: 10.1038/s43856-022-00209-1] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 10/28/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Conventional preclinical models often miss drug toxicities, meaning the harm these drugs pose to humans is only realized in clinical trials or when they make it to market. This has caused the pharmaceutical industry to waste considerable time and resources developing drugs destined to fail. Organ-on-a-Chip technology has the potential improve success in drug development pipelines, as it can recapitulate organ-level pathophysiology and clinical responses; however, systematic and quantitative evaluations of Organ-Chips' predictive value have not yet been reported. METHODS 870 Liver-Chips were analyzed to determine their ability to predict drug-induced liver injury caused by small molecules identified as benchmarks by the Innovation and Quality consortium, who has published guidelines defining criteria for qualifying preclinical models. An economic analysis was also performed to measure the value Liver-Chips could offer if they were broadly adopted in supporting toxicity-related decisions as part of preclinical development workflows. RESULTS Here, we show that the Liver-Chip met the qualification guidelines across a blinded set of 27 known hepatotoxic and non-toxic drugs with a sensitivity of 87% and a specificity of 100%. We also show that this level of performance could generate over $3 billion annually for the pharmaceutical industry through increased small-molecule R&D productivity. CONCLUSIONS The results of this study show how incorporating predictive Organ-Chips into drug development workflows could substantially improve drug discovery and development, allowing manufacturers to bring safer, more effective medicines to market in less time and at lower costs.
Collapse
|
62
|
Rana P, Khan S, Arat S, Potter D, Khan N. Nonclinical Safety Signals in PharmaPendium Improve the Predictability of Human Drug-Induced Liver Injury. Chem Res Toxicol 2022; 35:2133-2144. [PMID: 36287557 DOI: 10.1021/acs.chemrestox.2c00243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Drug-induced liver injury (DILI) is a leading cause of candidate attrition during drug development in the pharmaceutical industry. This study evaluated liver toxicity signals for 249 approved drugs (114 of "most-DILI concern" and 135 of "no-DILI concern") using PharmaPendium and assessed the association between nonclinical and clinical injuries using contingency table analysis. All animal liver findings were combined into eight toxicity categories based on nature and severity. Together, these analyses revealed that cholestasis [odds ratio (OR): 5.02; 95% confidence interval (CI) 1.04-24.03] or liver aminotransferase increases (OR: 1.86; 95% CI 1.09-3.09) in rats and steatosis (OR-1.9; 95% CI 1.03-3.49) or liver aminotransferase increases (OR-2.57; 95% CI 1.4-4.7) in dogs were significant predictors of human liver injury. The predictive value further improved when the liver injury categories were combined into less severe (steatosis, cholestasis, liver aminotransferase increase, hyperbilirubinemia, or jaundice) and more-severe (liver necrosis, acute liver failure, or hepatotoxicity) injuries. In particular, less-severe liver injuries in the following pairs of species predicted human hepatotoxicity {[dog and mouse] (OR: 2.70; 95% CI 1.25-5.84), [dog and rat] (OR-2.61; 95% CI 1.48-4.59), [monkey and mouse] (OR-4.22; 95% CI 1.33-13.32), and [monkey and rat] (OR-2.45; 95% CI 1.15-5.21)} were predictive of human hepatotoxicity. Meanwhile, severe liver injuries in both [dog and rat] (OR-1.9; 95% CI 1.04-3.49) were significant predictors of human liver toxicity. Therefore, we concluded that the occurrence of DILI in humans is highly likely if liver injuries are observed in one rodent and one nonrodent species and that liver aminotransferase increases in dogs and rats can predict DILI in humans. Together, these findings indicate that the liver safety signals observed in animal toxicity studies indicate potential DILI risk in humans and could therefore be used to prioritize small molecules with less potential to cause DILI in humans.
Collapse
Affiliation(s)
- Payal Rana
- Drug Safety Research and Development, Pfizer, Groton, Connecticut 06340, United States
| | - Sanaa Khan
- Drug Safety Research and Development, Pfizer, Groton, Connecticut 06340, United States
| | - Seda Arat
- Drug Safety Research and Development, Pfizer, Groton, Connecticut 06340, United States
| | - David Potter
- Early Clinical Development Biostatistics, Pfizer, Inc., Cambridge, Massachusetts 02139, United States
| | - Nasir Khan
- Drug Safety Research and Development, Pfizer, Groton, Connecticut 06340, United States
| |
Collapse
|
63
|
Predictive validity in drug discovery: what it is, why it matters and how to improve it. Nat Rev Drug Discov 2022; 21:915-931. [PMID: 36195754 DOI: 10.1038/s41573-022-00552-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2022] [Indexed: 11/08/2022]
Abstract
Successful drug discovery is like finding oases of safety and efficacy in chemical and biological deserts. Screens in disease models, and other decision tools used in drug research and development (R&D), point towards oases when they score therapeutic candidates in a way that correlates with clinical utility in humans. Otherwise, they probably lead in the wrong direction. This line of thought can be quantified by using decision theory, in which 'predictive validity' is the correlation coefficient between the output of a decision tool and clinical utility across therapeutic candidates. Analyses based on this approach reveal that the detectability of good candidates is extremely sensitive to predictive validity, because the deserts are big and oases small. Both history and decision theory suggest that predictive validity is under-managed in drug R&D, not least because it is so hard to measure before projects succeed or fail later in the process. This article explains the influence of predictive validity on R&D productivity and discusses methods to evaluate and improve it, with the aim of supporting the application of more effective decision tools and catalysing investment in their creation.
Collapse
|
64
|
McDuffie D, Barr D, Agarwal A, Thomas E. Physiologically relevant microsystems to study viral infection in the human liver. Front Microbiol 2022; 13:999366. [PMID: 36246284 PMCID: PMC9555087 DOI: 10.3389/fmicb.2022.999366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Viral hepatitis is a leading cause of liver disease and mortality. Infection can occur acutely or chronically, but the mechanisms that govern the clearance of virus or lack thereof are poorly understood and merit further investigation. Though cures for viral hepatitis have been developed, they are expensive, not readily accessible in vulnerable populations and some patients may remain at an increased risk of developing hepatocellular carcinoma (HCC) even after viral clearance. To sustain infection in vitro, hepatocytes must be fully mature and remain in a differentiated state. However, primary hepatocytes rapidly dedifferentiate in conventional 2D in vitro platforms. Physiologically relevant or physiomimetic microsystems, are increasingly popular alternatives to traditional two-dimensional (2D) monocultures for in vitro studies. Physiomimetic systems reconstruct and incorporate elements of the native cellular microenvironment to improve biologic functionality in vitro. Multiple elements contribute to these models including ancillary tissue architecture, cell co-cultures, matrix proteins, chemical gradients and mechanical forces that contribute to increased viability, longevity and physiologic function for the tissue of interest. These microsystems are used in a wide variety of applications to study biological phenomena. Here, we explore the use of physiomimetic microsystems as tools for studying viral hepatitis infection in the liver and how the design of these platforms is tailored for enhanced investigation of the viral lifecycle when compared to conventional 2D cell culture models. Although liver-based physiomimetic microsystems are typically applied in the context of drug studies, the platforms developed for drug discovery purposes offer a solid foundation to support studies on viral hepatitis. Physiomimetic platforms may help prolong hepatocyte functionality in order to sustain chronic viral hepatitis infection in vitro for studying virus-host interactions for prolonged periods.
Collapse
Affiliation(s)
- Dennis McDuffie
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, United States
| | - David Barr
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Ashutosh Agarwal
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, United States
- Desai Sethi Urology Institute, University of Miami Miller School of Medicine, Miami, FL, United States
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Emmanuel Thomas
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, United States
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
- Schiff Center for Liver Diseases, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
65
|
Brecklinghaus T, Albrecht W, Duda J, Kappenberg F, Gründler L, Edlund K, Marchan R, Ghallab A, Cadenas C, Rieck A, Vartak N, Tolosa L, Castell JV, Gardner I, Halilbasic E, Trauner M, Ullrich A, Zeigerer A, Demirci Turgunbayer Ö, Damm G, Seehofer D, Rahnenführer J, Hengstler JG. In vitro/in silico prediction of drug induced steatosis in relation to oral doses and blood concentrations by the Nile Red assay. Toxicol Lett 2022; 368:33-46. [PMID: 35963427 DOI: 10.1016/j.toxlet.2022.08.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 08/01/2022] [Accepted: 08/09/2022] [Indexed: 11/26/2022]
Abstract
The accumulation of lipid droplets in hepatocytes is a key feature of drug-induced liver injury (DILI) and can be induced by a subset of hepatotoxic compounds. In the present study, we optimized and evaluated an in vitro technique based on the fluorescent dye Nile Red, further named Nile Red assay to quantify lipid droplets induced by the exposure to chemicals. The Nile Red assay and a cytotoxicity test (CTB assay) were then performed on cells exposed concentration-dependently to 60 different compounds. Of these, 31 were known to induce hepatotoxicity in humans, and 13 were reported to also cause steatosis. In order to compare in vivo relevant blood concentrations, pharmacokinetic models were established for all compounds to simulate the maximal blood concentrations (Cmax) at therapeutic doses. The results showed that several hepatotoxic compounds induced an increase in lipid droplets at sub-cytotoxic concentrations. To compare how well (1) the cytotoxicity test alone, (2) the Nile Red assay alone, and (3) the combination of the cytotoxicity test and the Nile Red assay (based on the lower EC10 of both assays) allow the differentiation between hepatotoxic and non-hepatotoxic compounds, a previously established performance metric, the Toxicity Separation Index (TSI) was calculated. In addition, the Toxicity Estimation Index (TEI) was calculated to determine how well blood concentrations that cause an increased DILI risk can be estimated for hepatotoxic compounds. Our findings indicate that the combination of both assays improved the TSI and TEI compared to each assay alone. In conclusion, the study demonstrates that inclusion of the Nile Red assay into in vitro test batteries may improve the prediction of DILI compounds.
Collapse
Affiliation(s)
- Tim Brecklinghaus
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany.
| | - Wiebke Albrecht
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany
| | - Julia Duda
- Department of Statistics, TU Dortmund University, Vogelpothsweg 87, 44227, Dortmund, Germany
| | - Franziska Kappenberg
- Department of Statistics, TU Dortmund University, Vogelpothsweg 87, 44227, Dortmund, Germany
| | - Lisa Gründler
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany
| | - Karolina Edlund
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany
| | - Rosemarie Marchan
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany
| | - Ahmed Ghallab
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany; Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, South Valley University, 83523 Qena, Egypt
| | - Cristina Cadenas
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany
| | - Adrian Rieck
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany
| | - Nachiket Vartak
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany
| | - Laia Tolosa
- Experimental Hepatology Unit, Health Research Institute La Fe, Valencia, Spain
| | - José V Castell
- Experimental Hepatology Unit, Health Research Institute La Fe, Valencia, Spain; Biochemistry Department, University of Valencia and CIBEREHD
| | | | - Emina Halilbasic
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Michael Trauner
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Anett Ullrich
- Primacyt Cell Culture Technology GmbH, Schwerin, Germany
| | - Anja Zeigerer
- Institute for Diabetes and Cancer, Helmholtz Center Munich, 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany
| | - Özlem Demirci Turgunbayer
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany; Department of Biology, Faculty of Science, Dicle University, 21280, Diyarbakır, Turkey
| | - Georg Damm
- Department of Hepatobiliary Surgery and Visceral Transplantation, University of Leipzig, 04103 Leipzig, Germany
| | - Daniel Seehofer
- Department of Hepatobiliary Surgery and Visceral Transplantation, University of Leipzig, 04103 Leipzig, Germany
| | - Jörg Rahnenführer
- Department of Statistics, TU Dortmund University, Vogelpothsweg 87, 44227, Dortmund, Germany
| | - Jan G Hengstler
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany.
| |
Collapse
|
66
|
Liu M, Xiang Y, Yang Y, Long X, Xiao Z, Nan Y, Jiang Y, Qiu Y, Huang Q, Ai K. State-of-the-art advancements in Liver-on-a-chip (LOC): Integrated biosensors for LOC. Biosens Bioelectron 2022; 218:114758. [DOI: 10.1016/j.bios.2022.114758] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/24/2022] [Accepted: 09/24/2022] [Indexed: 12/12/2022]
|
67
|
Yan L, Messner CJ, Tian M, Gou X, Suter-Dick L, Zhang X. Evaluation of dioxin induced transcriptomic responses in a 3D human liver microtissue model. ENVIRONMENTAL RESEARCH 2022; 210:112906. [PMID: 35181307 DOI: 10.1016/j.envres.2022.112906] [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: 10/29/2021] [Revised: 01/27/2022] [Accepted: 02/04/2022] [Indexed: 06/14/2023]
Abstract
Three-dimensional human liver microtissue model provides a promising method for predicting the human hepatotoxicity of environmental chemicals. However, the dynamics of transcriptional responses of 3D human liver microtissue model to dioxins exposure remain unclear. Herein, time-series transcriptomic analysis was used to characterize modulation of gene expression over 14 days in 3D human liver microtissues exposed to 2,3,7,8-tetra-chlorodibenzo-p-dioxin (TCDD, 31 nM, 10 ng/ml). Changes in gene expression and modulation of biological pathways were evaluated at several time points. The results showed that microtissues stably expressed genes related to toxicological pathways (e.g. highly of genes involved in external stimuli and maintenance of cell homeostasis pathways) during the 14-day culture period. Furthermore, a weekly phenomenon pattern was observed for the number of the differentially expressed genes in microtissues exposed to TCDD at each time point. TCDD led to an induction of genes involved in cell cycle regulation at day three. Metabolic pathways were the main significantly induced pathways during the subsequent days, with the immune/inflammatory response enriched on the fifth day, and the cellular response to DNA damage was identified at the end of the exposure. Finally, relevant transcription patterns identified in microtissues were compared with published data on rodent and human cell-line studies to elucidate potential species-specific responses to TCDD over time. Cell development and cytochrome P450 pathway were mainly affected after a 3-day exposure, with the DNA damage response identified at the end of exposure in the human microtissue system but not in mouse/rat primary hepatocytes models. Overall, the 3D human liver microtissue model is a valuable tool to predict the toxic effects of environmental chemicals with a relatively long exposure.
Collapse
Affiliation(s)
- Lu Yan
- State Key Laboratory of Pollution Control & Resource Reuse, School of the Environment, Nanjing University, Nanjing, 210023, PR China
| | - Catherine Jane Messner
- University of Applied Sciences Northwestern Switzerland, School of Life Sciences, Muttenz, Switzerland; Swiss Centre for Applied Human Toxicology (SCAHT), Switzerland
| | - Mingming Tian
- State Key Laboratory of Pollution Control & Resource Reuse, School of the Environment, Nanjing University, Nanjing, 210023, PR China
| | - Xiao Gou
- State Key Laboratory of Pollution Control & Resource Reuse, School of the Environment, Nanjing University, Nanjing, 210023, PR China
| | - Laura Suter-Dick
- University of Applied Sciences Northwestern Switzerland, School of Life Sciences, Muttenz, Switzerland; Swiss Centre for Applied Human Toxicology (SCAHT), Switzerland
| | - Xiaowei Zhang
- State Key Laboratory of Pollution Control & Resource Reuse, School of the Environment, Nanjing University, Nanjing, 210023, PR China.
| |
Collapse
|
68
|
Cuvellier M, Rose S, Ezan F, Jarry U, De Oliveira H, Bruyère A, Drieu La Rochelle C, Legagneux V, Langouet S, Baffet G. In vitro long term differentiation and functionality of three-dimensional bioprinted primary human hepatocytes: application for in vivo engraftment. Biofabrication 2022; 14. [PMID: 35696992 DOI: 10.1088/1758-5090/ac7825] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 06/13/2022] [Indexed: 11/12/2022]
Abstract
In recent decades, 3D in vitro cultures of primary human hepatocytes (PHH) have been increasingly developed to establish models capable of faithfully mimicking main liver functions. The use of 3D bioprinting, capable of recreating structures composed of cells embedded in matrix with controlled microarchitectures, is an emergent key feature for tissue engineering. In this work, we used an extrusion-based system to print PHH in a methacrylated gelatin matrix (GelMa). PHH bioprinted in GelMa rapidly organized into polarized hollow spheroids and were viable for at least 28 days of culture. These PHH were highly differentiated with maintenance of liver differentiation genes over time, as demonstrated by transcriptomic analysis and functional approaches. The cells were polarized with localization of apico/canalicular regions, and displayed activities of phase I and II biotransformation enzymes that could be regulated by inducers. Furthermore, the implantation of the bioprinted structures in mice demonstrated their capability to vascularize, and their ability to maintain human hepatic specific functions for at least 28 days was illustrated by albumin secretion and debrisoquine metabolism. This model could hold great promise for human liver tissue generation and its use in future biotechnological developments.
Collapse
Affiliation(s)
- Marie Cuvellier
- Irset (Institut de recherche en santé́ environnement et travail) - UMR_S 1085, 2 Av du Pr Léon Bernard, Rennes, 35000, FRANCE
| | - Sophie Rose
- Irset (Institut de recherche en santé́ environnement et travail) - UMR_S 1085, 2 Av du pr Léon Bernard, Rennes, 35000, FRANCE
| | - Frédéric Ezan
- Irset (Institut de recherche en santé́ environnement et travail) - UMR_S 1085, 2 Av du pr Léon Bernard, Rennes, 35000, FRANCE
| | - Ulrich Jarry
- Unité de Pharmacologie Préclinique, Rennes, France, Biotrial Pharmacology, 7-9 Rue Jean-Louis Bertrand, Rennes, 35000, FRANCE
| | - Hugo De Oliveira
- , Université de Bordeaux, Bioingénierie tissulaire, rue Léo Saignat, Bordeaux, 33076, FRANCE
| | - Arnaud Bruyère
- Irset (Institut de recherche en santé́ environnement et travail) - UMR_S 1085, 2 Av. du Pr Léon Bernard, Rennes, 35000, FRANCE
| | - Christophe Drieu La Rochelle
- Unité de Pharmacologie Préclinique, Rennes, France, Biotrial Pharmacology, 7-9 Rue Jean-Louis Bertrand, Rennes, 35000, FRANCE
| | - Vincent Legagneux
- Irset (Institut de recherche en santé́ environnement et travail) - UMR_S 1085, 2 Av. du Pr Léon Bernard, Rennes, 35000, FRANCE
| | - Sophie Langouet
- Irset (Institut de recherche en santé́ environnement et travail) - UMR_S 1085, 2 Av. du Pr Léon Bernard, Rennes, 35000, FRANCE
| | - Georges Baffet
- Irset (Institut de recherche en santé́ environnement et travail) - UMR_S 1085, 2 Av. du Pr Léon Bernard, Rennes, 35000, FRANCE
| |
Collapse
|
69
|
Cox B, Barton P, Class R, Coxhead H, Delatour C, Gillent E, Henshall J, Isin EM, King L, Valentin JP. Setup of human liver-chips integrating 3D models, microwells and a standardized microfluidic platform as proof-of-concept study to support drug evaluation. BIOMATERIALS AND BIOSYSTEMS 2022; 7:100054. [PMID: 36824483 PMCID: PMC9934436 DOI: 10.1016/j.bbiosy.2022.100054] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 05/13/2022] [Accepted: 05/29/2022] [Indexed: 10/18/2022] Open
Abstract
Human 3D liver microtissues/spheroids are powerful in vitro models to study drug-induced liver injury (DILI) but the small number of cells per spheroid limits the models' usefulness to study drug metabolism. In this work, we scale up the number of spheroids on both a plate and a standardized organ-chip platform by factor 100 using a basic method which requires only limited technical expertise. We successfully generated up to 100 spheroids using polymer-coated microwells in a 96-well plate (= liver-plate) or organ-chip (= liver-chip). Liver-chips display a comparable cellular CYP3A4 activity, viability, and biomarker expression as liver spheroids for at least one week, while liver-plate cultures display an overall reduced hepatic functionality. To prove its applicability to drug discovery and development, the liver-chip was used to test selected reference compounds. The test system could discriminate toxicity of the DILI-positive compound tolcapone from its less hepatotoxic structural analogue entacapone, using biochemical and morphological readouts. Following incubation with diclofenac, the liver-chips had an increased metabolite formation compared to standard spheroid cultures. In summary, we generated a human liver-chip model using a standardized organ-chip platform which combines up to 100 spheroids and can be used for the evaluation of both drug safety and metabolism.
Collapse
Affiliation(s)
- Benoit Cox
- Development Science, UCB Biopharma SRL, Chemin du Foriest 1, B1420 Braine-l'Alleud, Belgium,Corresponding author.
| | - Patrick Barton
- Development Science, UCB Biopharma SRL, 216 Bath Rd, Slough, Berkshire SL1 3WE, UK
| | - Reiner Class
- Development Science, UCB Biopharma SRL, Chemin du Foriest 1, B1420 Braine-l'Alleud, Belgium
| | - Hannah Coxhead
- Development Science, UCB Biopharma SRL, Chemin du Foriest 1, B1420 Braine-l'Alleud, Belgium
| | - Claude Delatour
- Development Science, UCB Biopharma SRL, Chemin du Foriest 1, B1420 Braine-l'Alleud, Belgium
| | - Eric Gillent
- Development Science, UCB Biopharma SRL, Chemin du Foriest 1, B1420 Braine-l'Alleud, Belgium
| | - Jamie Henshall
- Development Science, UCB Biopharma SRL, 216 Bath Rd, Slough, Berkshire SL1 3WE, UK
| | - Emre M. Isin
- Development Science, UCB Biopharma SRL, Chemin du Foriest 1, B1420 Braine-l'Alleud, Belgium
| | - Lloyd King
- Development Science, UCB Biopharma SRL, 216 Bath Rd, Slough, Berkshire SL1 3WE, UK
| | - Jean-Pierre Valentin
- Development Science, UCB Biopharma SRL, Chemin du Foriest 1, B1420 Braine-l'Alleud, Belgium
| |
Collapse
|
70
|
Mai P, Hampl J, Baca M, Brauer D, Singh S, Weise F, Borowiec J, Schmidt A, Küstner JM, Klett M, Gebinoga M, Schroeder IS, Markert UR, Glahn F, Schumann B, Eckstein D, Schober A. MatriGrid® Based Biological Morphologies: Tools for 3D Cell Culturing. Bioengineering (Basel) 2022; 9:bioengineering9050220. [PMID: 35621498 PMCID: PMC9138054 DOI: 10.3390/bioengineering9050220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/06/2022] [Accepted: 05/11/2022] [Indexed: 02/06/2023] Open
Abstract
Recent trends in 3D cell culturing has placed organotypic tissue models at another level. Now, not only is the microenvironment at the cynosure of this research, but rather, microscopic geometrical parameters are also decisive for mimicking a tissue model. Over the years, technologies such as micromachining, 3D printing, and hydrogels are making the foundation of this field. However, mimicking the topography of a particular tissue-relevant substrate can be achieved relatively simply with so-called template or morphology transfer techniques. Over the last 15 years, in one such research venture, we have been investigating a micro thermoforming technique as a facile tool for generating bioinspired topographies. We call them MatriGrid®s. In this research account, we summarize our learning outcome from this technique in terms of the influence of 3D micro morphologies on different cell cultures that we have tested in our laboratory. An integral part of this research is the evolution of unavoidable aspects such as possible label-free sensing and fluidic automatization. The development in the research field is also documented in this account.
Collapse
Affiliation(s)
- Patrick Mai
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Jörg Hampl
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
- Correspondence: (J.H.); (A.S.); Tel.: +49-3677-6933387 (A.S.)
| | - Martin Baca
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Dana Brauer
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Sukhdeep Singh
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Frank Weise
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Justyna Borowiec
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - André Schmidt
- Placenta Lab, Department of Obstetrics, Jena University Hospital, 07747 Jena, Germany; (A.S.); (U.R.M.)
| | - Johanna Merle Küstner
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Maren Klett
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Michael Gebinoga
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Insa S. Schroeder
- Biophysics Division, GSI Helmholtzzentrum für Schwerionenforschung, 64291 Darmstadt, Germany;
| | - Udo R. Markert
- Placenta Lab, Department of Obstetrics, Jena University Hospital, 07747 Jena, Germany; (A.S.); (U.R.M.)
| | - Felix Glahn
- Institute of Environmental Toxicology, Martin-Luther-University Halle-Wittenberg, 06097 Halle, Germany; (F.G.); (B.S.); (D.E.)
| | - Berit Schumann
- Institute of Environmental Toxicology, Martin-Luther-University Halle-Wittenberg, 06097 Halle, Germany; (F.G.); (B.S.); (D.E.)
| | - Diana Eckstein
- Institute of Environmental Toxicology, Martin-Luther-University Halle-Wittenberg, 06097 Halle, Germany; (F.G.); (B.S.); (D.E.)
| | - Andreas Schober
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
- Correspondence: (J.H.); (A.S.); Tel.: +49-3677-6933387 (A.S.)
| |
Collapse
|
71
|
Evaluation of the Anti-Histoplasma capsulatum Activity of Indole and Nitrofuran Derivatives and Their Pharmacological Safety in Three-Dimensional Cell Cultures. Pharmaceutics 2022; 14:pharmaceutics14051043. [PMID: 35631629 PMCID: PMC9147190 DOI: 10.3390/pharmaceutics14051043] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/04/2022] [Accepted: 05/09/2022] [Indexed: 11/25/2022] Open
Abstract
Histoplasma capsulatum is a fungus that causes histoplasmosis. The increased evolution of microbial resistance and the adverse effects of current antifungals help new drugs to emerge. In this work, fifty-four nitrofurans and indoles were tested against the H. capsulatum EH-315 strain. Compounds with a minimum inhibitory concentration (MIC90) equal to or lower than 7.81 µg/mL were selected to evaluate their MIC90 on ATCC G217-B strain and their minimum fungicide concentration (MFC) on both strains. The quantification of membrane ergosterol, cell wall integrity, the production of reactive oxygen species, and the induction of death by necrosis–apoptosis was performed to investigate the mechanism of action of compounds 7, 11, and 32. These compounds could reduce the extracted sterol and induce necrotic cell death, similarly to itraconazole. Moreover, 7 and 11 damaged the cell wall, causing flaws in the contour (11), or changing the size and shape of the fungal cell wall (7). Furthermore, 7 and 32 induced reactive oxygen species (ROS) formation higher than 11 and control. Finally, the cytotoxicity was measured in two models of cell culture, i.e., monolayers (cells are flat) and a three-dimensional (3D) model, where they present a spheroidal conformation. Cytotoxicity assays in the 3D model showed a lower toxicity in the compounds than those performed on cell monolayers. Overall, these results suggest that derivatives of nitrofurans and indoles are promising compounds for the treatment of histoplasmosis.
Collapse
|
72
|
Automated Analysis of Acetaminophen Toxicity on 3D HepaRG Cell Culture in Microbioreactor. Bioengineering (Basel) 2022; 9:bioengineering9050196. [PMID: 35621474 PMCID: PMC9137798 DOI: 10.3390/bioengineering9050196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/26/2022] [Accepted: 04/29/2022] [Indexed: 11/16/2022] Open
Abstract
Real-time monitoring of bioanalytes in organotypic cell cultivation devices is a major research challenge in establishing stand-alone diagnostic systems. Presently, no general technical facility is available that offers a plug-in system for bioanalytics in diversely available organotypic culture models. Therefore, each analytical device has to be tuned according to the microfluidic and interface environment of the 3D in vitro system. Herein, we report the design and function of a 3D automated culture and analysis device (3D-ACAD) which actively perfuses a custom-made 3D microbioreactor, samples the culture medium and simultaneously performs capillary-based flow ELISA. A microstructured MatriGrid® has been explored as a 3D scaffold for culturing HepaRG cells, with albumin investigated as a bioanalytical marker using flow ELISA. We investigated the effect of acetaminophen (APAP) on the albumin secretion of HepaRG cells over 96 h and compared this with the albumin secretion of 2D monolayer HepaRG cultures. Automated on-line monitoring of albumin secretion in the 3D in vitro mode revealed that the application of hepatotoxic drug-like APAP results in decreased albumin secretion. Furthermore, a higher sensitivity of the HepaRG cell culture in the automated 3D-ACAD system to APAP was observed compared to HepaRG cells cultivated as a monolayer. The results support the use of the 3D-ACAD model as a stand-alone device, working in real time and capable of analyzing the condition of the cell culture by measuring a functional analyte. Information obtained from our system is compared with conventional cell culture and plate ELISA, the results of which are presented herein.
Collapse
|
73
|
Mirahmad M, Sabourian R, Mahdavi M, Larijani B, Safavi M. In vitro cell-based models of drug-induced hepatotoxicity screening: progress and limitation. Drug Metab Rev 2022; 54:161-193. [PMID: 35403528 DOI: 10.1080/03602532.2022.2064487] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Drug-induced liver injury (DILI) is one of the major causes of post-approval withdrawal of therapeutics. As a result, there is an increasing need for accurate predictive in vitro assays that reliably detect hepatotoxic drug candidates while reducing drug discovery time, costs, and the number of animal experiments. In vitro hepatocyte-based research has led to an improved comprehension of the underlying mechanisms of chemical toxicity and can assist the prioritization of therapeutic choices with low hepatotoxicity risk. Therefore, several in vitro systems have been generated over the last few decades. This review aims to comprehensively present the development and validation of 2D (two-dimensional) and 3D (three-dimensional) culture approaches on hepatotoxicity screening of compounds and highlight the main factors affecting predictive power of experiments. To this end, we first summarize some of the recognized hepatotoxicity mechanisms and related assays used to appraise DILI mechanisms and then discuss the challenges and limitations of in vitro models.
Collapse
Affiliation(s)
- Maryam Mirahmad
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Reyhaneh Sabourian
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Maliheh Safavi
- Department of Biotechnology, Iranian Research Organization for Science and Technology, Tehran, Iran
| |
Collapse
|
74
|
Antunes N, Kundu B, Kundu SC, Reis RL, Correlo V. In Vitro Cancer Models: A Closer Look at Limitations on Translation. Bioengineering (Basel) 2022; 9:166. [PMID: 35447726 PMCID: PMC9029854 DOI: 10.3390/bioengineering9040166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/02/2022] [Accepted: 04/05/2022] [Indexed: 12/18/2022] Open
Abstract
In vitro cancer models are envisioned as high-throughput screening platforms for potential new therapeutic discovery and/or validation. They also serve as tools to achieve personalized treatment strategies or real-time monitoring of disease propagation, providing effective treatments to patients. To battle the fatality of metastatic cancers, the development and commercialization of predictive and robust preclinical in vitro cancer models are of urgent need. In the past decades, the translation of cancer research from 2D to 3D platforms and the development of diverse in vitro cancer models have been well elaborated in an enormous number of reviews. However, the meagre clinical success rate of cancer therapeutics urges the critical introspection of currently available preclinical platforms, including patents, to hasten the development of precision medicine and commercialization of in vitro cancer models. Hence, the present article critically reflects the difficulty of translating cancer therapeutics from discovery to adoption and commercialization in the light of in vitro cancer models as predictive tools. The state of the art of in vitro cancer models is discussed first, followed by identifying the limitations of bench-to-bedside transition. This review tries to establish compatibility between the current findings and obstacles and indicates future directions to accelerate the market penetration, considering the niche market.
Collapse
Affiliation(s)
- Nina Antunes
- Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, AvePark, Zona Industrial da Gandra, 4805-017 Barco, Portugal; (N.A.); (B.K.); (S.C.K.); (R.L.R.)
- ICVS/3 B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Banani Kundu
- Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, AvePark, Zona Industrial da Gandra, 4805-017 Barco, Portugal; (N.A.); (B.K.); (S.C.K.); (R.L.R.)
- ICVS/3 B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Subhas C. Kundu
- Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, AvePark, Zona Industrial da Gandra, 4805-017 Barco, Portugal; (N.A.); (B.K.); (S.C.K.); (R.L.R.)
- ICVS/3 B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Rui L. Reis
- Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, AvePark, Zona Industrial da Gandra, 4805-017 Barco, Portugal; (N.A.); (B.K.); (S.C.K.); (R.L.R.)
- ICVS/3 B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Vítor Correlo
- Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, AvePark, Zona Industrial da Gandra, 4805-017 Barco, Portugal; (N.A.); (B.K.); (S.C.K.); (R.L.R.)
- ICVS/3 B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
| |
Collapse
|
75
|
Docci L, Milani N, Ramp T, Romeo AA, Godoy P, Franyuti DO, Krähenbühl S, Gertz M, Galetin A, Parrott N, Fowler S. Exploration and application of a liver-on-a-chip device in combination with modelling and simulation for quantitative drug metabolism studies. LAB ON A CHIP 2022; 22:1187-1205. [PMID: 35107462 DOI: 10.1039/d1lc01161h] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Microphysiological systems (MPS) are complex and more physiologically realistic cellular in vitro tools that aim to provide more relevant human in vitro data for quantitative prediction of clinical pharmacokinetics while also reducing the need for animal testing. The PhysioMimix liver-on-a-chip integrates medium flow with hepatocyte culture and has the potential to be adopted for in vitro studies investigating the hepatic disposition characteristics of drug candidates. The current study focusses on liver-on-a-chip system exploration for multiple drug metabolism applications. Characterization of cytochrome P450 (CYP), UDP-glucuronosyl transferase (UGT) and aldehyde oxidase (AO) activities was performed using 15 drugs and in vitro to in vivo extrapolation (IVIVE) was assessed for 12 of them. Next, the utility of the liver-on-a-chip for estimation of the fraction metabolized (fm) via specific biotransformation pathways of quinidine and diclofenac was established. Finally, the metabolite identification opportunities were also explored using efavirenz as an example drug with complex primary and secondary metabolism involving a combination of CYP, UGT and sulfotransferase enzymes. A key aspect of these investigations was the application of mathematical modelling for improved parameter calculation. Such approaches will be required for quantitative assessment of metabolism and/or transporter processes in systems where medium flow and system compartments result in non-homogeneous drug concentrations. In particular, modelling was used to explore the effect of evaporation from the medium and it was found that the intrinsic clearance (CLint) might be underestimated by up to 40% for low clearance compounds if evaporation is not accounted for. Modelling of liver-on-a-chip in vitro data also enhanced the approach to fm estimation allowing objective assessment of metabolism models of different complexity. The resultant diclofenac fm,UGT of 0.64 was highly comparable with values reported previously in the literature. The current study demonstrates the integration of mathematical modelling with experimental liver-on-a-chip studies and illustrates how this approach supports generation of high quality of data from complex in vitro cellular systems.
Collapse
Affiliation(s)
- Luca Docci
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070, Basel, Switzerland.
- Clinical Pharmacology & Toxicology, University Hospital, Schanzenstrasse 55, 4031, Basel, Switzerland
| | - Nicolò Milani
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070, Basel, Switzerland.
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, UK
| | - Thomas Ramp
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070, Basel, Switzerland.
| | - Andrea A Romeo
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070, Basel, Switzerland.
| | - Patricio Godoy
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070, Basel, Switzerland.
| | - Daniela Ortiz Franyuti
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070, Basel, Switzerland.
| | - Stephan Krähenbühl
- Clinical Pharmacology & Toxicology, University Hospital, Schanzenstrasse 55, 4031, Basel, Switzerland
| | - Michael Gertz
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070, Basel, Switzerland.
| | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, UK
| | - Neil Parrott
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070, Basel, Switzerland.
| | - Stephen Fowler
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070, Basel, Switzerland.
| |
Collapse
|
76
|
Rapid Detection of Direct Compound Toxicity and Trailing Detection of Indirect Cell Metabolite Toxicity in a 96-Well Fluidic Culture Device for Cell-Based Screening Environments: Tactics in Six Sigma Quality Control Charts. APPLIED SCIENCES-BASEL 2022. [PMID: 37502123 PMCID: PMC10374175 DOI: 10.3390/app12062786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Microfluidic screening tools, in vitro, evolve amid varied scientific disciplines. One emergent technique, simultaneously assessing cell toxicity from a primary compound and ensuing cell-generated metabolites (dual-toxicity screening), entails in-line systems having sequentially aligned culture chambers. To explore dual-tox screens, we probe the dissemination of nutrients involving 1-way transport with upstream compound dosing, midstream cascading flows, and downstream cessation. Distribution of flow gives rise to broad concentration ranges of dosing compound (0→ICcompound100) and wide-ranging concentration ranges of generated cell metabolites (0→ICmetabolites100). Innately, single-pass unidirectional flow retains 1st pass informative traits across the network, composed of nine interconnected culture wells, preserving both compound and cell-secreted byproducts as data indicators in each adjacent culture chamber. Thereafter, to assess effective compound hepatotoxicity (0→ECcompound100) and simultaneously classify for cell-metabolite toxicity (0→ECmetabolite100), we reveal utility by analyzing culture viability against ramping exposures of acetaminophen (APAP) and nefazodone (NEF), compounds of hepatic significance. We then discern metabolite generation with an emphasis on amplification across µchannel multiwell sites. Lastly, using conventional cell functions as indicator tools to assess dual toxicity, we investigate a non-drug induced liver injury (non-DILI) compound and DILI compound. The technology is for predictive evaluations of new compound formulations, new chemical entities (NCE), or drugs that have previously failed testing for unresolved reasons.
Collapse
|
77
|
Naito Y, Yoshinouchi Y, Sorayama Y, Kohara H, Kitano S, Irie S, Matsusaki M. Constructing vascularized hepatic tissue by cell-assembled viscous tissue sedimentation method and its application for vascular toxicity assessment. Acta Biomater 2022; 140:275-288. [PMID: 34826641 DOI: 10.1016/j.actbio.2021.11.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/29/2021] [Accepted: 11/17/2021] [Indexed: 01/09/2023]
Abstract
In vitro Construction of the liver sinusoidal structure using artificial tissue is an important but worthwhile challenge, particularly for assessing the risk of diseases such as sinusoidal obstruction syndrome (SOS). Current models are unsuitable for evaluating the toxicity because of lacking sinusoidal capillary. In this study, we developed a vascularized hepatic tissue (VHT) using a unique tissue engineering technique, the cell assembled viscous tissue by sedimentation (CAViTs) method. The "viscous bodies" created using the CAViTs method exhibited significant self-assembly within 6 h after seeding, promoting cell-cell interaction. The level of albumin secreted by the VHT was four times higher than that of 2D-coculture and maintained for 1 month. The gene expression pattern of the VHT was closer to that of total human liver, compared with the 2D system. Quantitative evaluations of the vascular structure of VHT treated with two typical SOS-inducing compounds, monocrotaline and retrorsine, revealed higher sensitivity (IC50 = 40.35 µM), 19.92 times higher than the cell-viability assay. Thus, VHT represents an innovative in vitro model that mimics the vessel network structure and could become a useful tool for the early screening of compounds associated with a risk of vascular toxicity. STATEMENT OF SIGNIFICANCE: Mimicking sinusoidal structures in in vitro liver model is important to consider from the perspective of predicting hepatotoxicity such like sinusoidal obstruction syndrome (SOS). However, it was difficult to reconstruct the vascular structure within the hepatocyte-rich environment. In this study, we constructed a vascularized hepatic tissue in a high-throughput manner by a unique method using collagen and heparin, and evaluated its applicability to toxicity assessment. Vessel morphology analysis of the model treated by monocrotaline, which is a well-known SOS-inducing compound, could predict the toxicity with higher sensitivity. To the best of our knowledge, this is the first report to provide vascularized hepatic tissues using sinusoidal endothelial cells at least for demonstrating applicability to the evaluation of SOS induction risk.
Collapse
|
78
|
Sasikumar S, Chameettachal S, Kingshott P, Cromer B, Pati F. Influence of Liver Extracellular Matrix in Predicting Drug-Induced Liver Injury: An Alternate Paradigm. ACS Biomater Sci Eng 2022; 8:834-846. [PMID: 34978414 DOI: 10.1021/acsbiomaterials.1c00994] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In vitro drug-induced liver injury (DILI) models are promising tools for drug development to predict adverse events during clinical usage. However, the currently available DILI models are not specific or not able to predict the injury accurately. This is believed to be mainly because of failure to conserve the hepatocyte phenotype, lack of longevity, and difficulty in maintaining the tissue-specific microenvironment. In this study, we have assessed the potential of decellularized liver extracellular matrix (DLM) in retaining the hepatic cellular phenotype and functionality in the presence of a tissue-specific microenvironment along with its role in influencing the effect of the drug on hepatic cells. We show that DLM helps maintain the phenotype of the hepatic cell line HepG2, a well-known cell line for secretion of human proteins that is easily available. Also, the DLM enhanced the expression of a metabolic marker carbamoyl phosphate synthetase I (CPS1), a regulator of urea cycle, and bile salt export pump (BSEP), a marker of hepatocyte polarity. We further validated the DLM for its influence on the sensitivity of cells toward different classes of drugs. Interestingly, the coculture model, in the presence of endothelial cells and stellate cells, exhibited a higher sensitivity for both acetaminophen and trovafloxacin, a toxic compound that does not show any toxicity on preclinical screening. Thus, our results demonstrate for the first time that a multicellular combination along with DLM can be a potential and reliable DILI model to screen multiple drugs.
Collapse
Affiliation(s)
- Shyama Sasikumar
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana 502284, India.,Department of Chemistry and Biotechnology, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
| | - Shibu Chameettachal
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana 502284, India
| | - Peter Kingshott
- Department of Chemistry and Biotechnology, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia.,ARC Training Centre Training Centre in Surface Engineering for Advanced Materials (SEAM), School of Engineering, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
| | - Brett Cromer
- Department of Chemistry and Biotechnology, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
| | - Falguni Pati
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana 502284, India
| |
Collapse
|
79
|
Friedman SL, Pinzani M. Hepatic fibrosis 2022: Unmet needs and a blueprint for the future. Hepatology 2022; 75:473-488. [PMID: 34923653 DOI: 10.1002/hep.32285] [Citation(s) in RCA: 246] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/12/2021] [Accepted: 11/15/2021] [Indexed: 12/12/2022]
Abstract
Steady progress over four decades toward understanding the pathogenesis and clinical consequences of hepatic fibrosis has led to the expectation of effective antifibrotic drugs, yet none has been approved. Thus, an assessment of the field is timely, to clarify priorities and accelerate progress. Here, we highlight the successes to date but, more importantly, identify gaps and unmet needs, both experimentally and clinically. These include the need to better define cell-cell interactions and etiology-specific elements of fibrogenesis and their link to disease-specific drivers of portal hypertension. Success in treating viral hepatitis has revealed the remarkable capacity of the liver to degrade scar in reversing fibrosis, yet we know little of the mechanisms underlying this response. Thus, there is an exigent need to clarify the cellular and molecular mechanisms of fibrosis regression in order for therapeutics to mimic the liver's endogenous capacity. Better refined and more predictive in vitro and animal models will hasten drug development. From a clinical perspective, current diagnostics are improving but not always biologically plausible or sufficiently accurate to supplant biopsy. More urgently, digital pathology methods that leverage machine learning and artificial intelligence must be validated in order to capture more prognostic information from liver biopsies and better quantify the response to therapies. For more refined treatment of NASH, orthogonal approaches that integrate genetic, clinical, and pathological data sets may yield treatments for specific subphenotypes of the disease. Collectively, these and other advances will strengthen and streamline clinical trials and better link histologic responses to clinical outcomes.
Collapse
Affiliation(s)
- Scott L Friedman
- Division of Liver DiseasesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Massimo Pinzani
- Institute for Liver and Digestive HealthUniversity College LondonLondonUK
| |
Collapse
|
80
|
Youhanna S, Kemas AM, Preiss L, Zhou Y, Shen JX, Cakal SD, Paqualini FS, Goparaju SK, Shafagh RZ, Lind JU, Sellgren CM, Lauschke VM. Organotypic and Microphysiological Human Tissue Models for Drug Discovery and Development-Current State-of-the-Art and Future Perspectives. Pharmacol Rev 2022; 74:141-206. [PMID: 35017176 DOI: 10.1124/pharmrev.120.000238] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 10/12/2021] [Indexed: 12/11/2022] Open
Abstract
The number of successful drug development projects has been stagnant for decades despite major breakthroughs in chemistry, molecular biology, and genetics. Unreliable target identification and poor translatability of preclinical models have been identified as major causes of failure. To improve predictions of clinical efficacy and safety, interest has shifted to three-dimensional culture methods in which human cells can retain many physiologically and functionally relevant phenotypes for extended periods of time. Here, we review the state of the art of available organotypic culture techniques and critically review emerging models of human tissues with key importance for pharmacokinetics, pharmacodynamics, and toxicity. In addition, developments in bioprinting and microfluidic multiorgan cultures to emulate systemic drug disposition are summarized. We close by highlighting important trends regarding the fabrication of organotypic culture platforms and the choice of platform material to limit drug absorption and polymer leaching while supporting the phenotypic maintenance of cultured cells and allowing for scalable device fabrication. We conclude that organotypic and microphysiological human tissue models constitute promising systems to promote drug discovery and development by facilitating drug target identification and improving the preclinical evaluation of drug toxicity and pharmacokinetics. There is, however, a critical need for further validation, benchmarking, and consolidation efforts ideally conducted in intersectoral multicenter settings to accelerate acceptance of these novel models as reliable tools for translational pharmacology and toxicology. SIGNIFICANCE STATEMENT: Organotypic and microphysiological culture of human cells has emerged as a promising tool for preclinical drug discovery and development that might be able to narrow the translation gap. This review discusses recent technological and methodological advancements and the use of these systems for hit discovery and the evaluation of toxicity, clearance, and absorption of lead compounds.
Collapse
Affiliation(s)
- Sonia Youhanna
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Aurino M Kemas
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Lena Preiss
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Yitian Zhou
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Joanne X Shen
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Selgin D Cakal
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Francesco S Paqualini
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Sravan K Goparaju
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Reza Zandi Shafagh
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Johan Ulrik Lind
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Carl M Sellgren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| |
Collapse
|
81
|
Zheng YB, Ma LD, Wu JL, Wang YM, Meng XS, Hu P, Liang QL, Xie YY, Luo GA. Design and fabrication of an integrated 3D dynamic multicellular liver-on-a-chip and its application in hepatotoxicity screening. Talanta 2022; 241:123262. [DOI: 10.1016/j.talanta.2022.123262] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 01/16/2022] [Accepted: 01/22/2022] [Indexed: 01/05/2023]
|
82
|
Liu J, Guo W, Sakkiah S, Ji Z, Yavas G, Zou W, Chen M, Tong W, Patterson TA, Hong H. Machine Learning Models for Predicting Liver Toxicity. Methods Mol Biol 2022; 2425:393-415. [PMID: 35188640 DOI: 10.1007/978-1-0716-1960-5_15] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Liver toxicity is a major adverse drug reaction that accounts for drug failure in clinical trials and withdrawal from the market. Therefore, predicting potential liver toxicity at an early stage in drug discovery is crucial to reduce costs and the potential for drug failure. However, current in vivo animal toxicity testing is very expensive and time consuming. As an alternative approach, various machine learning models have been developed to predict potential liver toxicity in humans. This chapter reviews current advances in the development and application of machine learning models for prediction of potential liver toxicity in humans and discusses possible improvements to liver toxicity prediction.
Collapse
Affiliation(s)
- Jie Liu
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA
| | - Wenjing Guo
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA
| | - Sugunadevi Sakkiah
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA
| | - Zuowei Ji
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA
| | - Gokhan Yavas
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA
| | - Wen Zou
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA
| | - Minjun Chen
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA
| | - Weida Tong
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA
| | - Tucker A Patterson
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA
| | - Huixiao Hong
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA.
| |
Collapse
|
83
|
Segovia-Zafra A, Di Zeo-Sánchez DE, López-Gómez C, Pérez-Valdés Z, García-Fuentes E, Andrade RJ, Lucena MI, Villanueva-Paz M. Preclinical models of idiosyncratic drug-induced liver injury (iDILI): Moving towards prediction. Acta Pharm Sin B 2021; 11:3685-3726. [PMID: 35024301 PMCID: PMC8727925 DOI: 10.1016/j.apsb.2021.11.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/07/2021] [Accepted: 11/10/2021] [Indexed: 02/08/2023] Open
Abstract
Idiosyncratic drug-induced liver injury (iDILI) encompasses the unexpected harms that prescription and non-prescription drugs, herbal and dietary supplements can cause to the liver. iDILI remains a major public health problem and a major cause of drug attrition. Given the lack of biomarkers for iDILI prediction, diagnosis and prognosis, searching new models to predict and study mechanisms of iDILI is necessary. One of the major limitations of iDILI preclinical assessment has been the lack of correlation between the markers of hepatotoxicity in animal toxicological studies and clinically significant iDILI. Thus, major advances in the understanding of iDILI susceptibility and pathogenesis have come from the study of well-phenotyped iDILI patients. However, there are many gaps for explaining all the complexity of iDILI susceptibility and mechanisms. Therefore, there is a need to optimize preclinical human in vitro models to reduce the risk of iDILI during drug development. Here, the current experimental models and the future directions in iDILI modelling are thoroughly discussed, focusing on the human cellular models available to study the pathophysiological mechanisms of the disease and the most used in vivo animal iDILI models. We also comment about in silico approaches and the increasing relevance of patient-derived cellular models.
Collapse
Affiliation(s)
- Antonio Segovia-Zafra
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid 28029, Spain
| | - Daniel E. Di Zeo-Sánchez
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
| | - Carlos López-Gómez
- Unidad de Gestión Clínica de Aparato Digestivo, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga 29010, Spain
| | - Zeus Pérez-Valdés
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
| | - Eduardo García-Fuentes
- Unidad de Gestión Clínica de Aparato Digestivo, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga 29010, Spain
| | - Raúl J. Andrade
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid 28029, Spain
| | - M. Isabel Lucena
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid 28029, Spain
- Platform ISCIII de Ensayos Clínicos, UICEC-IBIMA, Málaga 29071, Spain
| | - Marina Villanueva-Paz
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
| |
Collapse
|
84
|
Ströbel S, Kostadinova R, Fiaschetti-Egli K, Rupp J, Bieri M, Pawlowska A, Busler D, Hofstetter T, Sanchez K, Grepper S, Thoma E. A 3D primary human cell-based in vitro model of non-alcoholic steatohepatitis for efficacy testing of clinical drug candidates. Sci Rep 2021; 11:22765. [PMID: 34815444 PMCID: PMC8611054 DOI: 10.1038/s41598-021-01951-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/03/2021] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is a progressive and severe liver disease, characterized by lipid accumulation, inflammation, and downstream fibrosis. Despite its increasing prevalence, there is no approved treatment yet available for patients. This has been at least partially due to the lack of predictive preclinical models for studying this complex disease. Here, we present a 3D in vitro microtissue model that uses spheroidal, scaffold free co-culture of primary human hepatocytes, Kupffer cells, liver endothelial cells and hepatic stellate cells. Upon exposure to defined and clinically relevant lipotoxic and inflammatory stimuli, these microtissues develop key pathophysiological features of NASH within 10 days, including an increase of intracellular triglyceride content and lipids, and release of pro-inflammatory cytokines. Furthermore, fibrosis was evident through release of procollagen type I, and increased deposition of extracellular collagen fibers. Whole transcriptome analysis revealed changes in the regulation of pathways associated with NASH, such as lipid metabolism, inflammation and collagen processing. Importantly, treatment with anti-NASH drug candidates (Selonsertib and Firsocostat) decreased the measured specific disease parameter, in accordance with clinical observations. These drug treatments also significantly changed the gene expression patterns of the microtissues, thus providing mechanisms of action and revealing therapeutic potential. In summary, this human NASH model represents a promising drug discovery tool for understanding the underlying complex mechanisms in NASH, evaluating efficacy of anti-NASH drug candidates and identifying new approaches for therapeutic interventions.
Collapse
Affiliation(s)
- Simon Ströbel
- InSphero AG, Wagistrasse 27A, 8952 Schlieren, CH, Switzerland.
| | | | | | - Jana Rupp
- InSphero AG, Wagistrasse 27A, 8952 Schlieren, CH Switzerland
| | - Manuela Bieri
- InSphero AG, Wagistrasse 27A, 8952 Schlieren, CH Switzerland
| | | | - Donna Busler
- InSphero AG, Wagistrasse 27A, 8952 Schlieren, CH Switzerland
| | | | | | - Sue Grepper
- InSphero AG, Wagistrasse 27A, 8952 Schlieren, CH Switzerland
| | - Eva Thoma
- InSphero AG, Wagistrasse 27A, 8952 Schlieren, CH Switzerland
| |
Collapse
|
85
|
Schofield CA, Walker TM, Taylor MA, Patel M, Vlachou DF, Macina JM, Vidgeon-Hart MP, Williams A, McGill PJ, Newman CF, Sakatis MZ. Evaluation of a Three-Dimensional Primary Human Hepatocyte Spheroid Model: Adoption and Industrialization for the Enhanced Detection of Drug-Induced Liver Injury. Chem Res Toxicol 2021; 34:2485-2499. [PMID: 34797640 DOI: 10.1021/acs.chemrestox.1c00227] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Drug-induced liver injury is a leading cause of compound attrition during both preclinical and clinical drug development, and early strategies are in place to tackle this recurring problem. Human-relevant in vitro models that are more predictive of hepatotoxicity hazard identification, and that could be employed earlier in the drug discovery process, would improve the quality of drug candidate selection and help reduce attrition. We present an evaluation of four human hepatocyte in vitro models of increasing culture complexity (i.e., two-dimensional (2D) HepG2 monolayers, hepatocyte sandwich cultures, three-dimensional (3D) hepatocyte spheroids, and precision-cut liver slices), using the same tool compounds, viability end points, and culture time points. Having established the improved prediction potential of the 3D hepatocyte spheroid model, we describe implementing this model into an industrial screening setting, where the challenge was matching the complexity of the culture system with the scale and throughput required. Following further qualification and miniaturization into a 384-well, high-throughput screening format, data was generated on 199 compounds. This clearly demonstrated the ability to capture a greater number of severe hepatotoxins versus the current routine 2D HepG2 monolayer assay while continuing to flag no false-positive compounds. The industrialization and miniaturization of the 3D hepatocyte spheroid complex in vitro model demonstrates a significant step toward reducing drug attrition and improving the quality and safety of drugs, while retaining the flexibility for future improvements, and has replaced the routine use of the 2D HepG2 monolayer assay at GlaxoSmithKline.
Collapse
Affiliation(s)
- Christopher A Schofield
- Functional Genomics, Medicinal Science and Technology, GlaxoSmithKline Research and Development, Stevenage, Herts SG1 2NY, United Kingdom
| | - Tracy M Walker
- Oncology Cell Therapy, Oncology Therapy Area, GlaxoSmithKline Research and Development, Stevenage, Herts SG1 2NY, United Kingdom
| | - Maxine A Taylor
- Drug Metabolism and Pharmacokinetics, In Vitro/In Vivo Translation, GlaxoSmithKline Research and Development, Ware, Herts SG12 0DP, United Kingdom
| | - Metul Patel
- Screening, Profiling and Mechanistic Biology, Medicinal Science and Technology, GlaxoSmithKline Research and Development, Stevenage, Herts SG1 2NY, United Kingdom
| | - Denise F Vlachou
- Molecular Design U.K., Medicinal Science and Technology, GlaxoSmithKline Research and Development, Stevenage, Herts SG1 2NY, United Kingdom
| | - Justyna M Macina
- Screening, Profiling and Mechanistic Biology, Medicinal Science and Technology, GlaxoSmithKline Research and Development, Stevenage, Herts SG1 2NY, United Kingdom
| | - Martin P Vidgeon-Hart
- Non Clinical Safety, In Vitro/In Vivo Translation, GlaxoSmithKline Research and Development, Ware, Herts SG12 0DP, United Kingdom
| | - Ann Williams
- Pathology U.K., In Vitro/In Vivo Translation, GlaxoSmithKline Research and Development, Ware, Herts SG12 0DP, United Kingdom
| | - Paul J McGill
- Bioimaging U.K., In Vitro/In Vivo Translation, GlaxoSmithKline Research and Development, Ware, Herts SG12 0DP, United Kingdom
| | - Carla F Newman
- Bioimaging U.K., In Vitro/In Vivo Translation, GlaxoSmithKline Research and Development, Stevenage, Herts SG1 2NY, United Kingdom
| | - Melanie Z Sakatis
- Non Clinical Safety, In Vitro/In Vivo Translation, GlaxoSmithKline Research and Development, Ware, Herts SG12 0DP, United Kingdom
| |
Collapse
|
86
|
Development of Breast Cancer Spheroids to Evaluate Cytotoxic Response to an Anticancer Peptide. Pharmaceutics 2021; 13:pharmaceutics13111863. [PMID: 34834277 PMCID: PMC8619419 DOI: 10.3390/pharmaceutics13111863] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 12/13/2022] Open
Abstract
Breast cancer (BC) is the most commonly diagnosed cancer in women and one of the most common causes of cancer-related deaths. Despite intense research efforts, BC treatment still remains challenging. Improved drug development strategies are needed for impactful benefit to patients. Current preclinical studies rely mostly on cell-based screenings, using two-dimensional (2D) cell monolayers that do not mimic in vivo tumors properly. Herein, we explored the development and characterization of three-dimensional (3D) models, named spheroids, of the most aggressive BC subtypes (triple-negative breast cancer-TNBC; and human-epidermal growth receptor-2-HER2+), using the liquid overlay technique with several selected cell lines. In these cell line-derived spheroids, we studied cell density, proliferation, ultrastructure, apoptosis, reactive oxygen species (ROS) production, and cell permeabilization (live/dead). The results showed a formation of compact and homogeneous spheroids on day 7 after seeding 2000 cells/well for MDA-MB-231 and 5000 cells/well for BT-20 and BT-474. Next, we compared the efficacy of a model anticancer peptide (ACP) in cell monolayers and spheroids. Overall, the results demonstrated spheroids to be less sensitive to treatment than cell monolayers, revealing the need for more robust models in drug development.
Collapse
|
87
|
Brecklinghaus T, Albrecht W, Kappenberg F, Duda J, Vartak N, Edlund K, Marchan R, Ghallab A, Cadenas C, Günther G, Leist M, Zhang M, Gardner I, Reinders J, Russel FG, Foster AJ, Williams DP, Damle-Vartak A, Grandits M, Ecker G, Kittana N, Rahnenführer J, Hengstler JG. The hepatocyte export carrier inhibition assay improves the separation of hepatotoxic from non-hepatotoxic compounds. Chem Biol Interact 2021; 351:109728. [PMID: 34717914 DOI: 10.1016/j.cbi.2021.109728] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 10/15/2021] [Accepted: 10/25/2021] [Indexed: 02/08/2023]
Abstract
An in vitro/in silico method that determines the risk of human drug induced liver injury in relation to oral doses and blood concentrations of drugs was recently introduced. This method utilizes information on the maximal blood concentration (Cmax) for a specific dose of a test compound, which can be estimated using physiologically-based pharmacokinetic modelling, and a cytotoxicity test in cultured human hepatocytes. In the present study, we analyzed if the addition of an assay that measures the inhibition of bile acid export carriers, like BSEP and/or MRP2, to the existing method improves the differentiation of hepatotoxic and non-hepatotoxic compounds. Therefore, an export assay for 5-chloromethylfluorescein diacetate (CMFDA) was established. We tested 36 compounds in a concentration-dependent manner for which the risk of hepatotoxicity for specific oral doses and the capacity to inhibit hepatocyte export carriers are known. Compared to the CTB cytotoxicity test, substantially lower EC10 values were obtained using the CMFDA assay for several known BSEP and/or MRP2 inhibitors. To quantify if the addition of the CMFDA assay to our test system improves the overall separation of hepatotoxic from non-hepatotoxic compounds, the toxicity separation index (TSI) was calculated. We obtained a better TSI using the lower alert concentration from either the CMFDA or the CTB test (TSI: 0.886) compared to considering the CTB test alone (TSI: 0.775). In conclusion, the data show that integration of the CMFDA assay with an in vitro test battery improves the differentiation of hepatotoxic and non-hepatotoxic compounds in a set of compounds that includes bile acid export carrier inhibitors.
Collapse
Affiliation(s)
- Tim Brecklinghaus
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany.
| | - Wiebke Albrecht
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - Franziska Kappenberg
- Department of Statistics, TU Dortmund University, Vogelpothsweg 87, 44227, Dortmund, Germany
| | - Julia Duda
- Department of Statistics, TU Dortmund University, Vogelpothsweg 87, 44227, Dortmund, Germany
| | - Nachiket Vartak
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - Karolina Edlund
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - Rosemarie Marchan
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - Ahmed Ghallab
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany; Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, South Valley University, 83523, Qena, Egypt
| | - Cristina Cadenas
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - Georgia Günther
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, Universitätsstr. 10, PO Box M657, 78457, Constance, Germany
| | - Mian Zhang
- Simcyp (A Certara Company), Sheffield, UK
| | | | - Jörg Reinders
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - Frans Gm Russel
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Alison J Foster
- Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, United Kingdom
| | - Dominic P Williams
- Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, United Kingdom
| | - Amruta Damle-Vartak
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany; Division of Signal Transduction and Growth Control, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Melanie Grandits
- University of Vienna, Department of Pharmaceutical Sciences, Althanstraße 14, Vienna, Austria
| | - Gerhard Ecker
- University of Vienna, Department of Pharmaceutical Sciences, Althanstraße 14, Vienna, Austria
| | - Naim Kittana
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, PO Box 7, Nablus, Palestine
| | - Jörg Rahnenführer
- Department of Statistics, TU Dortmund University, Vogelpothsweg 87, 44227, Dortmund, Germany
| | - Jan G Hengstler
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany.
| |
Collapse
|
88
|
Subbaiah MAM, Meanwell NA. Bioisosteres of the Phenyl Ring: Recent Strategic Applications in Lead Optimization and Drug Design. J Med Chem 2021; 64:14046-14128. [PMID: 34591488 DOI: 10.1021/acs.jmedchem.1c01215] [Citation(s) in RCA: 269] [Impact Index Per Article: 67.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The benzene moiety is the most prevalent ring system in marketed drugs, underscoring its historic popularity in drug design either as a pharmacophore or as a scaffold that projects pharmacophoric elements. However, introspective analyses of medicinal chemistry practices at the beginning of the 21st century highlighted the indiscriminate deployment of phenyl rings as an important contributor to the poor physicochemical properties of advanced molecules, which limited their prospects of being developed into effective drugs. This Perspective deliberates on the design and applications of bioisosteric replacements for a phenyl ring that have provided practical solutions to a range of developability problems frequently encountered in lead optimization campaigns. While the effect of phenyl ring replacements on compound properties is contextual in nature, bioisosteric substitution can lead to enhanced potency, solubility, and metabolic stability while reducing lipophilicity, plasma protein binding, phospholipidosis potential, and inhibition of cytochrome P450 enzymes and the hERG channel.
Collapse
Affiliation(s)
- Murugaiah A M Subbaiah
- Department of Medicinal Chemistry, Biocon-Bristol Myers Squibb Research and Development Centre, Biocon Park, Bommasandra IV Phase, Jigani Link Road, Bangalore, Karnataka 560099, India
| | - Nicholas A Meanwell
- Department of Small Molecule Drug Discovery, Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| |
Collapse
|
89
|
Macko P, Palosaari T, Whelan M. Extrapolating from acute to chronic toxicity in vitro. Toxicol In Vitro 2021; 76:105206. [PMID: 34186185 PMCID: PMC8434427 DOI: 10.1016/j.tiv.2021.105206] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/21/2021] [Accepted: 06/24/2021] [Indexed: 12/01/2022]
Abstract
Chemical safety assessment requires information on both chronic and acute effects of toxicants. Traditionally, such information has been provided by a set of animal studies conducted over different durations, ranging from a single dose with observation of effects over a few days, to repeat daily dosing and observations made over many months. With the advent of modern mechanistic approaches to toxicology, the role of in vitro studies within alternative approaches has never been more prominent. Typical in vitro experiments are conducted over short durations with measurements of response at a single time point, with a focus on providing effect and concentration-response information as input to hazard and risk assessment. This limits the usefulness of such data since potential chronic effects that cumulate over time are not usually considered. To address this, an experimental design is presented to characterise the toxicodynamics of a response not only in terms of concentration, but also as a function of time. Generation of concentration-time-effect responses allows both the extrapolation of points of departure from an acute to chronic exposure, and the determination of a chronicity index that provides a quantitative measure of a chemical's potential to cause cumulative effects over time. In addition, the approach provides a means to characterise the dynamics of key event relationships for the development of quantitative adverse outcome pathways.
Collapse
Affiliation(s)
- Peter Macko
- European Commission, Joint Research Centre (JRC), Ispra, Italy.
| | - Taina Palosaari
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Maurice Whelan
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| |
Collapse
|
90
|
Utoh R, Enomoto S, Yamada M, Yamanaka K, Yajima Y, Furusawa K, Seki M. Polyanion-induced, microfluidic engineering of fragmented collagen microfibers for reconstituting extracellular environments of 3D hepatocyte culture. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 129:112417. [PMID: 34579926 DOI: 10.1016/j.msec.2021.112417] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/23/2021] [Accepted: 08/30/2021] [Indexed: 12/11/2022]
Abstract
Artificial biological scaffolds made of extracellular matrix (ECM) components, such as type I collagen, provide ideal physicochemical cues to various cell culture platforms. However, it remains a challenge to fabricate micrometer-sized ECM materials with precisely controlled morphologies that could reconstitute the 3-dimensional (3D) microenvironments surrounding cells. In the present study, we proposed a unique process to fabricate fragmented collagen microfibers using a microfluidic laminar-flow system. The continuous flow of an acidic collagen solution was neutralized to generate solid fibers, which were subsequently fragmented by applying a gentle shear stress in a polyanion-containing phosphate buffer. The morphology of the fiber fragment was controllable in a wide range by changing the type and/or concentration of the polyanion and by tuning the applied shear stress. The biological benefits of the fragmented fibers were investigated through the formation of multicellular spheroids composed of primary rat hepatocytes and microfibers on non-cell-adhesive micro-vessels. The microfibers enhanced the survival and functions of the hepatocytes and reproduced proper cell polarity, because the fibers facilitated the formation of cell-cell and cell-matrix interactions while modulating the close packing of cells. These results clearly indicated that the microengineered fragmented collagen fibers have great potential to reconstitute extracellular microenvironments for hepatocytes in 3D culture, which will be of significant benefit for cell-based drug testing and bottom-up tissue engineering.
Collapse
Affiliation(s)
- Rie Utoh
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| | - Sakiko Enomoto
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| | - Masumi Yamada
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan.
| | - Keigo Yamanaka
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| | - Yuya Yajima
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| | - Kazuya Furusawa
- Department of Applied Chemistry and Food Science, Faculty of Environmental and Information Sciences, Fukui University of Technology, 3-6-1 Gakuen, Fukui 910-8505, Japan
| | - Minoru Seki
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| |
Collapse
|
91
|
Xu Q. Human Three-Dimensional Hepatic Models: Cell Type Variety and Corresponding Applications. Front Bioeng Biotechnol 2021; 9:730008. [PMID: 34631680 PMCID: PMC8497968 DOI: 10.3389/fbioe.2021.730008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/30/2021] [Indexed: 12/23/2022] Open
Abstract
Owing to retained hepatic phenotypes and functions, human three-dimensional (3D) hepatic models established with diverse hepatic cell types are thought to recoup the gaps in drug development and disease modeling limited by a conventional two-dimensional (2D) cell culture system and species-specific variability in drug metabolizing enzymes and transporters. Primary human hepatocytes, human hepatic cancer cell lines, and human stem cell-derived hepatocyte-like cells are three main hepatic cell types used in current models and exhibit divergent hepatic phenotypes. Primary human hepatocytes derived from healthy hepatic parenchyma resemble in vivo-like genetic and metabolic profiling. Human hepatic cancer cell lines are unlimitedly reproducible and tumorigenic. Stem cell-derived hepatocyte-like cells derived from patients are promising to retain the donor's genetic background. It has been suggested in some studies that unique properties of cell types endue them with benefits in different research fields of in vitro 3D modeling paradigm. For instance, the primary human hepatocyte was thought to be the gold standard for hepatotoxicity study, and stem cell-derived hepatocyte-like cells have taken a main role in personalized medicine and regenerative medicine. However, the comprehensive review focuses on the hepatic cell type variety, and corresponding applications in 3D models are sparse. Therefore, this review summarizes the characteristics of different cell types and discusses opportunities of different cell types in drug development, liver disease modeling, and liver transplantation.
Collapse
Affiliation(s)
- Qianqian Xu
- School of Chinese Medicine, and Centre for Cancer and Inflammation Research, Hong Kong Baptist University, Hong Kong, China
| |
Collapse
|
92
|
Three-Dimensional Liver Culture Systems to Maintain Primary Hepatic Properties for Toxicological Analysis In Vitro. Int J Mol Sci 2021; 22:ijms221910214. [PMID: 34638555 PMCID: PMC8508724 DOI: 10.3390/ijms221910214] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/15/2021] [Accepted: 09/19/2021] [Indexed: 12/13/2022] Open
Abstract
Drug-induced liver injury (DILI) is the major reason for failures in drug development and withdrawal of approved drugs from the market. Two-dimensional cultures of hepatocytes often fail to reliably predict DILI: hepatoma cell lines such as HepG2 do not reflect important primary-like hepatic properties and primary human hepatocytes (pHHs) dedifferentiate quickly in vitro and are, therefore, not suitable for long-term toxicity studies. More predictive liver in vitro models are urgently required in drug development and compound safety evaluation. This review discusses available human hepatic cell types for in vitro toxicology analysis and their usage in established and emerging three-dimensional (3D) culture systems. Generally, 3D cultures maintain or improve primary hepatic functions (including expression of drug-metabolizing enzymes) of different liver cells for several weeks of culture, thus allowing long-term and repeated-dose toxicity studies. Spheroid cultures of pHHs have been comprehensively tested, but also other cell types such as HepaRG benefit from 3D culture systems. Emerging 3D culture techniques include usage of induced pluripotent stem-cell-derived hepatocytes and primary-like upcyte cells, as well as advanced culture techniques such as microfluidic liver-on-a-chip models. In-depth characterization of existing and emerging 3D hepatocyte technologies is indispensable for successful implementation of such systems in toxicological analysis.
Collapse
|
93
|
Xiao RR, Lv T, Tu X, Li P, Wang T, Dong H, Tu P, Ai X. An integrated biomimetic array chip for establishment of collagen-based 3D primary human hepatocyte model for prediction of clinical drug-induced liver injury. Biotechnol Bioeng 2021; 118:4687-4698. [PMID: 34478150 DOI: 10.1002/bit.27931] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 07/05/2021] [Accepted: 08/28/2021] [Indexed: 12/19/2022]
Abstract
Drug-induced liver injury (DILI) is a leading cause of therapy failure in the clinic and also contributes much to acute liver failure cases. Investigations of predictive sensitivity in animal models have limitations due to interspecies differences. Previously reported in vitro models of liver injury based on primary human hepatocytes (PHHs) cannot meet the requirements of high physiological fidelity, low cost, simple operation, and high throughput with improved sensitivity. Herein, we developed an integrated biomimetic array chip (iBAC) for establishing extracellular matrix (ECM)-based models. A collagen-based 3D PHH model was constructed on the iBAC as a case for the prediction of clinical DILI at throughput. The iBAC has a three-layer structure with a core component of 3D implanting holes. At an initial cell seeding numbers of 5000-10,000, the collagen-based 3D PHH model was optimized with improved and stabilized liver functionality, including cell viability, albumin, and urea production. Moreover, basal activities of most metabolic enzymes on the iBAC were maintained for at least 12 days. Next, a small-scale hepatotoxicity screening indicated that the 3D PHH model on the iBAC was more sensitive for predicting hepatotoxicity than the 2D PHH model on the plate. Finally, a large-scale screening of liver toxicity using 122 clinical drugs further demonstrated that the collagen-based 3D PHH model on the iBAC had superior predictive sensitivity compared to all previously reported in vitro models. These results indicated the importance of 3D collagen for liver physiological functionality and hepatotoxicity prediction. We anticipant it being a promising tool for risk assessment of drug-induced hepatotoxicity with a widespread acceptance in drug industry.
Collapse
Affiliation(s)
| | - Tian Lv
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xia Tu
- Discovery Biology Unit, WuXi AppTec (Shanghai) Co., Ltd., Shanghai, China
| | - Peiwen Li
- R&D Department, Beijing Daxiang Biotech, Beijing, China
| | - Tiantian Wang
- Discovery Biology Unit, WuXi AppTec (Shanghai) Co., Ltd., Shanghai, China
| | - Haiheng Dong
- Discovery Biology Unit, WuXi AppTec (Shanghai) Co., Ltd., Shanghai, China
| | - Pengfei Tu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xiaoni Ai
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| |
Collapse
|
94
|
Wang H, Brown PC, Chow EC, Ewart L, Ferguson SS, Fitzpatrick S, Freedman BS, Guo GL, Hedrich W, Heyward S, Hickman J, Isoherranen N, Li AP, Liu Q, Mumenthaler SM, Polli J, Proctor WR, Ribeiro A, Wang J, Wange RL, Huang S. 3D cell culture models: Drug pharmacokinetics, safety assessment, and regulatory consideration. Clin Transl Sci 2021; 14:1659-1680. [PMID: 33982436 PMCID: PMC8504835 DOI: 10.1111/cts.13066] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/12/2022] Open
Abstract
Nonclinical testing has served as a foundation for evaluating potential risks and effectiveness of investigational new drugs in humans. However, the current two-dimensional (2D) in vitro cell culture systems cannot accurately depict and simulate the rich environment and complex processes observed in vivo, whereas animal studies present significant drawbacks with inherited species-specific differences and low throughput for increased demands. To improve the nonclinical prediction of drug safety and efficacy, researchers continue to develop novel models to evaluate and promote the use of improved cell- and organ-based assays for more accurate representation of human susceptibility to drug response. Among others, the three-dimensional (3D) cell culture models present physiologically relevant cellular microenvironment and offer great promise for assessing drug disposition and pharmacokinetics (PKs) that influence drug safety and efficacy from an early stage of drug development. Currently, there are numerous different types of 3D culture systems, from simple spheroids to more complicated organoids and organs-on-chips, and from single-cell type static 3D models to cell co-culture 3D models equipped with microfluidic flow control as well as hybrid 3D systems that combine 2D culture with biomedical microelectromechanical systems. This article reviews the current application and challenges of 3D culture systems in drug PKs, safety, and efficacy assessment, and provides a focused discussion and regulatory perspectives on the liver-, intestine-, kidney-, and neuron-based 3D cellular models.
Collapse
Affiliation(s)
- Hongbing Wang
- Department of Pharmaceutical SciencesUniversity of Maryland School of PharmacyBaltimoreMarylandUSA
| | - Paul C. Brown
- Center for Drug Evaluation and ResearchUS Food and Drug Administration (FDA)Silver SpringMarylandUSA
| | - Edwin C.Y. Chow
- Office of Clinical PharmacologyOffice of Translational SciencesCenter for Drug Evaluation and ResearchUS Food and Drug Administration (FDA)Silver SpringMarylandUSA
| | | | - Stephen S. Ferguson
- Division of the National Toxicology ProgramNational Institute of Environmental Health SciencesResearch Triangle ParkNorth CarolinaUSA
| | - Suzanne Fitzpatrick
- Office of the Center DirectorCenter for Food Safety and Applied NutritionUS Food and Drug Administration (FDA)Silver SpringMarylandUSA
| | - Benjamin S. Freedman
- Division of NephrologyDepartment of PathologyKidney Research Institute, and Institute for Stem Cell and Regenerative MedicineUniversity of WashingtonSeattleWashingtonUSA
- Department of MedicineUniversity of WashingtonSeattleWashingtonUSA
| | - Grace L. Guo
- Department of Pharmacology and ToxicologyErnest Mario School of PharmacyRutgers UniversityPiscatawayNew JerseyUSA
| | - William Hedrich
- Pharmaceutical Candidate Optimization, Metabolism and PharmacokineticsBristol‐Myers Squibb CompanyPrincetonNew JerseyUSA
| | | | - James Hickman
- NanoScience Technology CenterUniversity of Central FloridaOrlandoFloridaUSA
| | - Nina Isoherranen
- Department of PharmaceuticsSchool of PharmacyUniversity of WashingtonSeattleWashingtonUSA
| | - Albert P. Li
- In Vitro ADMET LaboratoriesColumbiaMarylandUSA
- In Vitro ADMET LaboratoriesMaldenMassachusettsUSA
| | - Qi Liu
- Office of Clinical PharmacologyOffice of Translational SciencesCenter for Drug Evaluation and ResearchUS Food and Drug Administration (FDA)Silver SpringMarylandUSA
| | - Shannon M. Mumenthaler
- Lawrence J. Ellison Institute for Transformative MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - James Polli
- Department of Pharmaceutical SciencesUniversity of Maryland School of PharmacyBaltimoreMarylandUSA
| | - William R. Proctor
- Predictive Toxicology, Safety AssessmentGenentech, IncSouth San FranciscoCaliforniaUSA
| | - Alexandre Ribeiro
- Office of Clinical PharmacologyOffice of Translational SciencesCenter for Drug Evaluation and ResearchUS Food and Drug Administration (FDA)Silver SpringMarylandUSA
| | - Jian‐Ying Wang
- Department of SurgeryCell Biology GroupUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Ronald L. Wange
- Center for Drug Evaluation and ResearchUS Food and Drug Administration (FDA)Silver SpringMarylandUSA
| | - Shiew‐Mei Huang
- Office of Clinical PharmacologyOffice of Translational SciencesCenter for Drug Evaluation and ResearchUS Food and Drug Administration (FDA)Silver SpringMarylandUSA
| |
Collapse
|
95
|
Lu S, Zhang J, Lin S, Zheng D, Shen Y, Qin J, Li Y, Wang S. Recent advances in the development of in vitro liver models for hepatotoxicity testing. Biodes Manuf 2021. [DOI: 10.1007/s42242-021-00142-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
96
|
Signore MA, De Pascali C, Giampetruzzi L, Siciliano PA, Francioso L. Gut-on-Chip microphysiological systems: Latest advances in the integration of sensing strategies and adoption of mature detection mechanisms. SENSING AND BIO-SENSING RESEARCH 2021. [DOI: 10.1016/j.sbsr.2021.100443] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
97
|
Kang W, Podtelezhnikov AA, Tanis KQ, Pacchione S, Su M, Bleicher KB, Wang Z, Laws GM, Griffiths TG, Kuhls MC, Chen Q, Knemeyer I, Marsh DJ, Mitra K, Lebron J, Sistare FD. Development and Application of a Transcriptomic Signature of Bioactivation in an Advanced In Vitro Liver Model to Reduce Drug-induced Liver Injury Risk Early in the Pharmaceutical Pipeline. Toxicol Sci 2021; 177:121-139. [PMID: 32559289 DOI: 10.1093/toxsci/kfaa094] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Early risk assessment of drug-induced liver injury (DILI) potential for drug candidates remains a major challenge for pharmaceutical development. We have previously developed a set of rat liver transcriptional biomarkers in short-term toxicity studies to inform the potential of drug candidates to generate a high burden of chemically reactive metabolites that presents higher risk for human DILI. Here, we describe translation of those NRF1-/NRF2-mediated liver tissue biomarkers to an in vitro assay using an advanced micropatterned coculture system (HEPATOPAC) with primary hepatocytes from male Wistar Han rats. A 9-day, resource-sparing and higher throughput approach designed to identify new chemical entities with lower reactive metabolite-forming potential was qualified for internal decision making using 93 DILI-positive and -negative drugs. This assay provides 81% sensitivity and 90% specificity in detecting hepatotoxicants when a positive test outcome is defined as the bioactivation signature score of a test drug exceeding the threshold value at an in vitro test concentration that falls within 3-fold of the estimated maximum drug concentration at the human liver inlet following highest recommended clinical dose administrations. Using paired examples of compounds from distinct chemical series and close structural analogs, we demonstrate that this assay can differentiate drugs with lower DILI risk. The utility of this in vitro transcriptomic approach was also examined using human HEPATOPAC from a single donor, yielding 68% sensitivity and 86% specificity when the aforementioned criteria are applied to the same 93-drug test set. Routine use of the rat model has been adopted with deployment of the human model as warranted on a case-by-case basis. This in vitro transcriptomic signature-based strategy can be used early in drug discovery to derisk DILI potential from chemically reactive metabolites by guiding structure-activity relationship hypotheses and candidate selection.
Collapse
Affiliation(s)
- Wen Kang
- Safety Assessment & Laboratory Animal Resources
| | | | | | | | - Ming Su
- Safety Assessment & Laboratory Animal Resources
| | | | - Zhibin Wang
- Safety Assessment & Laboratory Animal Resources
| | | | | | | | - Qing Chen
- Pharmacokinetics, Pharmacodynamics & Drug Metabolism, Merck & Co., Inc., West Point, Pennsylvania 19486
| | - Ian Knemeyer
- Pharmacokinetics, Pharmacodynamics & Drug Metabolism, Merck & Co., Inc., West Point, Pennsylvania 19486
| | | | | | - Jose Lebron
- Safety Assessment & Laboratory Animal Resources
| | | |
Collapse
|
98
|
Monckton CP, Brown GE, Khetani SR. Latest impact of engineered human liver platforms on drug development. APL Bioeng 2021; 5:031506. [PMID: 34286173 PMCID: PMC8286174 DOI: 10.1063/5.0051765] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 06/21/2021] [Indexed: 01/07/2023] Open
Abstract
Drug-induced liver injury (DILI) is a leading cause of drug attrition, which is partly due to differences between preclinical animals and humans in metabolic pathways. Therefore, in vitro human liver models are utilized in biopharmaceutical practice to mitigate DILI risk and assess related mechanisms of drug transport and metabolism. However, liver cells lose phenotypic functions within 1–3 days in two-dimensional monocultures on collagen-coated polystyrene/glass, which precludes their use to model the chronic effects of drugs and disease stimuli. To mitigate such a limitation, bioengineers have adapted tools from the semiconductor industry and additive manufacturing to precisely control the microenvironment of liver cells. Such tools have led to the fabrication of advanced two-dimensional and three-dimensional human liver platforms for different throughput needs and assay endpoints (e.g., micropatterned cocultures, spheroids, organoids, bioprinted tissues, and microfluidic devices); such platforms have significantly enhanced liver functions closer to physiologic levels and improved functional lifetime to >4 weeks, which has translated to higher sensitivity for predicting drug outcomes and enabling modeling of diseased phenotypes for novel drug discovery. Here, we focus on commercialized engineered liver platforms and case studies from the biopharmaceutical industry showcasing their impact on drug development. We also discuss emerging multi-organ microfluidic devices containing a liver compartment that allow modeling of inter-tissue crosstalk following drug exposure. Finally, we end with key requirements for engineered liver platforms to become routine fixtures in the biopharmaceutical industry toward reducing animal usage and providing patients with safe and efficacious drugs with unprecedented speed and reduced cost.
Collapse
Affiliation(s)
- Chase P Monckton
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| | - Grace E Brown
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| | - Salman R Khetani
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| |
Collapse
|
99
|
Lombardo FC, Ravaynia PS, Modena MM, Hierlemann A, Keiser J. Evaluation of Human Liver Microtissues for Drug Screening on Schistosoma mansoni Schistosomula. ACS Infect Dis 2021; 7:1894-1900. [PMID: 33105989 DOI: 10.1021/acsinfecdis.0c00614] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Schistosomiasis is a major neglected tropical disease with more than 200 million infections annually. Despite only one drug, praziquantel, being available, the drug pipeline against schistosomiasis is empty, and drug screening tools have limitations. We evaluated the potential of human liver microtissues (hLiMTs) in antischistosomal drug discovery. Because hLiMTs express all human P450 enzymes, they are an excellent tool to evaluate compounds' bioinactivation, bioactivation, and toxicity. To validate the metabolic conversion capacity of hLiMTs, we first quantified (R)- and (S)-praziquantel and the main metabolite trans-OH-praziquantel following incubation with 0.032-50 μM (0.01-15.62 μg/mL) praziquantel for up to 72 h by a validated LC-MS/MS method. We cocultured hLiMTs with newly transformed schistosomula (NTS) and evaluated the antischistosomal activity and cytotoxicity of three prodrugs terfenadine, tamoxifen citrate, and flutamide. HLiMTs converted 300-350 ng (R)-praziquantel within 24 h into trans-OH-praziquantel. We observed changes in the IC50 values for terfenadine, flutamide, and tamoxifen citrate in comparison to the standard NTS assay in vitro. Cytotoxicity was observed at high concentrations of flutamide and tamoxifen citrate. An in vitro platform containing hLiMTs could serve as an advanced drug screening tool for Schistosoma mansoni, providing information on reduced or increased activity and toxicity.
Collapse
Affiliation(s)
- Flavio C. Lombardo
- Swiss Tropical and Public Health Institute, Socinstrasse 57, CH-4002 Basel, Switzerland
- Universität Basel, Petersplatz 1, CH-4001 Basel, Switzerland
| | - Paolo S. Ravaynia
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Mario M. Modena
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Andreas Hierlemann
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Jennifer Keiser
- Swiss Tropical and Public Health Institute, Socinstrasse 57, CH-4002 Basel, Switzerland
- Universität Basel, Petersplatz 1, CH-4001 Basel, Switzerland
| |
Collapse
|
100
|
Khojasteh SC, Argikar UA, Driscoll JP, Heck CJS, King L, Jackson KD, Jian W, Kalgutkar AS, Miller GP, Kramlinger V, Rietjens IMCM, Teitelbaum AM, Wang K, Wei C. Novel advances in biotransformation and bioactivation research - 2020 year in review. Drug Metab Rev 2021; 53:384-433. [PMID: 33910427 PMCID: PMC8826528 DOI: 10.1080/03602532.2021.1916028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
This annual review is the sixth of its kind since 2016 (see references). Our objective is to explore and share articles which we deem influential and significant in the field of biotransformation and bioactivation. These fields are constantly evolving with new molecular structures and discoveries of corresponding pathways for metabolism that impact relevant drug development with respect to efficacy and safety. Based on the selected articles, we created three sections: (1) drug design, (2) metabolites and drug metabolizing enzymes, and (3) bioactivation and safety (Table 1). Unlike in years past, more biotransformation experts have joined and contributed to this effort while striving to maintain a balance of authors from academic and industry settings.
Collapse
Affiliation(s)
- S Cyrus Khojasteh
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, USA
| | - Upendra A Argikar
- Translational Medicine, Novartis Institutes for Biomedical Research, Inc., Cambridge, MA, USA
| | - James P Driscoll
- Department of Drug Metabolism and Pharmacokinetics, MyoKardia, Inc., South San Francisco, CA, USA
| | - Carley J S Heck
- Medicine Design, Pfizer Worldwide Research, Development and Medical, Groton, CT, USA
| | - Lloyd King
- Department of DMPK, UCB Biopharma, Slough, UK
| | - Klarissa D Jackson
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, Chapel Hill, NC, USA
| | - Wenying Jian
- Drug Metabolism and Pharmacokinetics, Janssen Research & Development, Spring House, PA, USA
| | - Amit S Kalgutkar
- Medicine Design, Pfizer Worldwide Research, Development and Medical, Cambridge, MA, USA
| | - Grover P Miller
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Valerie Kramlinger
- Translational Medicine, Novartis Institutes for Biomedical Research, Inc., Cambridge, MA, USA
| | | | - Aaron M Teitelbaum
- Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, USA
| | - Kai Wang
- Drug Metabolism and Pharmacokinetics, Janssen Research & Development, San Diego, CA, USA
| | - Cong Wei
- Drug Metabolism & Pharmacokinetics, Biogen Inc., Cambridge, MA, USA
| |
Collapse
|