1
|
Volpe DA. Application of transporter assays for drug discovery and development: an update of the literature. Expert Opin Drug Discov 2024; 19:1247-1257. [PMID: 39105537 DOI: 10.1080/17460441.2024.2387790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 07/30/2024] [Indexed: 08/07/2024]
Abstract
INTRODUCTION Determining whether a new drug is a substrate, inhibitor or inducer of efflux or uptake membrane transporters has become a routine process during drug discovery and development. In vitro assays are utilized to establish whether a new drug has the potential to be an object (substrate) or precipitant (inhibitor, inducer) in transporter-mediated clinical drug-drug interactions. The findings from these in vitro experiments are then used to determine whether further in vivo drug interaction studies are necessary for a new drug. AREAS COVERED This article provides an update on in vitro transporter assays, focusing on new uses of transfected cells, time-dependent inhibition, transporter induction, and complex model systems. EXPERT OPINION The newer in vitro assays add to the toolbox in defining new drugs as transporter substrates, inhibitors, or inducers. Complex models such as spheroids, organoids, and microphysiological systems require standardization and further research with model transporter substrates and inhibitors. In drug discovery, the more traditional transporter assays may be employed as substrate and inhibitor screening assays. In drug development, more complex cell models can be employed in later drug development to better understand how transporter(s) are involved in the absorption, distribution, and excretion of new drugs.
Collapse
Affiliation(s)
- Donna A Volpe
- Division of Applied Regulatory Science, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
2
|
Chothe PP, Argikar UA, Mitra P, Nakakariya M, Ramsden D, Rotter CJ, Sandoval P, Tohyama K. Drug transporters in drug disposition - highlights from the year 2023. Drug Metab Rev 2024:1-31. [PMID: 39221672 DOI: 10.1080/03602532.2024.2399523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Drug transporter field is rapidly evolving with significant progress in in vitro and in vivo tools and, computational models to assess transporter-mediated drug disposition and drug-drug interactions (DDIs) in humans. On behalf of all coauthors, I am pleased to share the fourth annual review highlighting articles published and deemed influential in the field of drug transporters in the year 2023. Each coauthor independently selected peer-reviewed articles published or available online in the year 2023 and summarized them as shown previously (Chothe et al. 2021; Chothe et al. 2022, 2023) with unbiased perspectives. Based on selected articles, this review was categorized into four sections: (1) transporter structure and in vitro evaluation, (2) novel in vitro/ex vivo models, (3) endogenous biomarkers, and (4) PBPK modeling for evaluating transporter DDIs (Table 1). As the scope of this review is not to comprehensively review each article, readers are encouraged to consult original paper for specific details. Finally, I appreciate all the authors for their time and continued support in writing this review.
Collapse
Affiliation(s)
- Paresh P Chothe
- Drug Metabolism and Pharmacokinetics, Oncology Research and Development, AstraZeneca, Waltham, MA, USA
| | - Upendra A Argikar
- Non-clinical Development, Bill and Melinda Gates Medical Research Institute, Cambridge, MA, USA
| | - Pallabi Mitra
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, CT, USA
| | - Masanori Nakakariya
- Drug Metabolism and Pharmacokinetics Research Laboratories, Takeda irinote Pharmaceutical Company Limited, Fujisawa, Japan
| | - Diane Ramsden
- Preclinical Development, Korro Bio, Inc. One Kendall Square, Cambridge, MA, USA
| | - Charles J Rotter
- Global Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. (TDCA), San Diego, CA, USA
| | - Philip Sandoval
- Global Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. (TDCA), Lexington, MA, USA
| | - Kimio Tohyama
- Drug Metabolism and Pharmacokinetics Research Laboratories, Takeda irinote Pharmaceutical Company Limited, Fujisawa, Japan
| |
Collapse
|
3
|
Michiba K, Watanabe K, Imaoka T, Nakai D. Recent Advances in the Gastrointestinal Complex in Vitro Model for ADME Studies. Pharmaceutics 2023; 16:37. [PMID: 38258048 PMCID: PMC10819272 DOI: 10.3390/pharmaceutics16010037] [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: 10/30/2023] [Revised: 12/06/2023] [Accepted: 12/16/2023] [Indexed: 01/24/2024] Open
Abstract
Intestinal absorption is a complex process involving the permeability of the epithelial barrier, efflux transporter activity, and intestinal metabolism. Identifying the key factors that govern intestinal absorption for each investigational drug is crucial. To assess and predict intestinal absorption in humans, it is necessary to leverage appropriate in vitro systems. Traditionally, Caco-2 monolayer systems and intestinal Ussing chamber studies have been considered the 'gold standard' for studying intestinal absorption. However, these methods have limitations that hinder their universal use in drug discovery and development. Recently, there has been an increasing number of reports on complex in vitro models (CIVMs) using human intestinal organoids derived from intestinal tissue specimens or iPSC-derived enterocytes plated on 2D or 3D in microphysiological systems. These CIVMs provide a more physiologically relevant representation of key ADME-related proteins compared to conventional in vitro methods. They hold great promise for use in drug discovery and development due to their ability to replicate the expressions and functions of these proteins. This review highlights recent advances in gut CIVMs employing intestinal organoid model systems compared to conventional methods. It is important to note that each CIVM should be tailored to the investigational drug properties and research questions at hand.
Collapse
Affiliation(s)
- Kazuyoshi Michiba
- Drug Metabolism & Pharmacokinetics Research Laboratory, Daiichi Sankyo Co., Ltd., 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan; (K.W.); (T.I.); (D.N.)
| | | | | | | |
Collapse
|
4
|
Rajan SAP, Sherfey J, Ohri S, Nichols L, Smith JT, Parekh P, Kadar EP, Clark F, George BT, Gregory L, Tess D, Gosset JR, Liras J, Geishecker E, Obach RS, Cirit M. A Novel Milli-fluidic Liver Tissue Chip with Continuous Recirculation for Predictive Pharmacokinetics Applications. AAPS J 2023; 25:102. [PMID: 37891356 DOI: 10.1208/s12248-023-00870-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
A crucial step in lead selection during drug development is accurate estimation and optimization of hepatic clearance using in vitro methods. However, current methods are limited by factors such as lack of physiological relevance, short culture/incubation times that are not consistent with drug exposure patterns in patients, use of drug absorbing materials, and evaporation during long-term incubation. To address these technological needs, we developed a novel milli-fluidic human liver tissue chip (LTC) that was designed with continuous media recirculation and optimized for hepatic cultures using human primary hepatocytes. Here, we characterized the LTC using a series of physiologically relevant metrics and test compounds to demonstrate that we could accurately predict the PK of both low- and high-clearance compounds. The non-biological characterization indicated that the cyclic olefin copolymer (COC)-based LTC exhibited negligible evaporation and minimal non-specific binding of drugs of varying ionic states and lipophilicity. Biologically, the LTC exhibited functional and polarized hepatic culture with sustained metabolic CYP activity for at least 15 days. This long-term culture was then used for drug clearance studies for low- and high-clearance compounds for at least 12 days, and clearance was estimated for a range of compounds with high in vitro-in vivo correlation (IVIVC). We also demonstrated that LTC can be induced by rifampicin, and the culture age had insignificant effect on depletion kinetic and predicted clearance value. Thus, we used advances in bioengineering to develop a novel purpose-built platform with high reproducibility and minimal variability to address unmet needs for PK applications.
Collapse
Affiliation(s)
| | - Jason Sherfey
- Javelin Biotech Inc, 299 Washington street, Woburn, Massachusetts, 01801, USA
| | - Shivam Ohri
- Javelin Biotech Inc, 299 Washington street, Woburn, Massachusetts, 01801, USA
| | - Lauren Nichols
- Javelin Biotech Inc, 299 Washington street, Woburn, Massachusetts, 01801, USA
| | - J Tyler Smith
- Javelin Biotech Inc, 299 Washington street, Woburn, Massachusetts, 01801, USA
| | - Paarth Parekh
- Javelin Biotech Inc, 299 Washington street, Woburn, Massachusetts, 01801, USA
| | - Eugene P Kadar
- Pfizer Global Research and Development, Groton Laboratories, Eastern Point Road, Groton, Connecticut, 06340, USA
| | - Frances Clark
- Pfizer Global Research and Development, Groton Laboratories, Eastern Point Road, Groton, Connecticut, 06340, USA
| | - Billy T George
- Pfizer Global Research and Development, Groton Laboratories, Eastern Point Road, Groton, Connecticut, 06340, USA
| | - Lauren Gregory
- Pfizer Global Research and Development, Groton Laboratories, Eastern Point Road, Groton, Connecticut, 06340, USA
| | - David Tess
- Pfizer Global Research and Development, Groton Laboratories, Eastern Point Road, Groton, Connecticut, 06340, USA
| | - James R Gosset
- Pfizer Worldwide Research and Development, 610 Main Street, Cambridge, Massachusetts, 02139, USA
| | - Jennifer Liras
- Pfizer Worldwide Research and Development, 610 Main Street, Cambridge, Massachusetts, 02139, USA
| | - Emily Geishecker
- Javelin Biotech Inc, 299 Washington street, Woburn, Massachusetts, 01801, USA
| | - R Scott Obach
- Pfizer Global Research and Development, Groton Laboratories, Eastern Point Road, Groton, Connecticut, 06340, USA
| | - Murat Cirit
- Javelin Biotech Inc, 299 Washington street, Woburn, Massachusetts, 01801, USA.
| |
Collapse
|
5
|
Arakawa H, Nakazono Y, Matsuoka N, Hayashi M, Shirasaka Y, Hirao A, Tamai I. Induction of open-form bile canaliculus formation by hepatocytes for evaluation of biliary drug excretion. Commun Biol 2023; 6:866. [PMID: 37608051 PMCID: PMC10444810 DOI: 10.1038/s42003-023-05216-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 08/04/2023] [Indexed: 08/24/2023] Open
Abstract
Biliary excretion is a major drug elimination pathway that affects their efficacy and safety. The currently available in vitro sandwich-cultured hepatocyte method is cumbersome because drugs accumulate in the closed bile canalicular lumen formed between hepatocytes and their amounts cannot be mealsured directly. This study proposes a hepatocyte culture model for the rapid evaluation of drug biliary excretion using permeation assays. When hepatocytes are cultured on a permeable support coated with the cell adhesion protein claudins, an open-form bile canalicular lumen is formed at the surface of the permeable support. Upon application to the basolateral (blood) side, drugs appear on the bile canalicular side. The biliary excretion clearance of several drugs, as estimated from the obtained permeabilities, correlates well with the reported in vivo biliary excretion clearance in humans. Thus, the established model is useful for applications in the efficient evaluation of biliary excretion during drug discovery and development.
Collapse
Affiliation(s)
- Hiroshi Arakawa
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Yuya Nakazono
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Natsumi Matsuoka
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Momoka Hayashi
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Yoshiyuki Shirasaka
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Atsushi Hirao
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, 920-1192, Japan
- WPI Nano Life Science Institute (WPI-Nano LSI), Kanazawa University, Kanazawa, 920-1192, Japan
| | - Ikumi Tamai
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan.
| |
Collapse
|
6
|
Jeon HJ, Kim C, Kim K, Lee SE. Piperlongumine treatment impacts heart and liver development and causes developmental delay in zebrafish (Danio rerio) embryos. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 258:114995. [PMID: 37167734 DOI: 10.1016/j.ecoenv.2023.114995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 05/02/2023] [Accepted: 05/06/2023] [Indexed: 05/13/2023]
Abstract
Piperlongumine (PL) and piperine (PP) are alkaloids presented in long pepper (Piper longum), and they exhibit various biological activities, especially anti-cancer properties. With these regards, they are considered as future medicines with high potential. Even they are exposed to humans such a long time, their potential toxicities in the environment have not been studied. Therefore, their ecological toxicities were assessed using zebrafish embryos. PP showed low mortality and no abnormal phenotype up to 10 µM. However, PL exhibited strong acute toxicity at the concentration of 5-10 µM ranges, and abnormal development were frequently found in the range of 1-2.5 µM with pericardial and yolk sac edemas. In transgenic zebrafish embryos, PL induced an increase in the number of intersegmental vessels and delayed the early-stage development. PL treatment affected heart formation and heart rate. The presence of PL induced the expression of cytokines, inflammatory markers, and inflammasome in the embryos. The PL treatment changed the mRNA levels of the ER stress and apoptosis-related genes. In addition, ROS production was observed during early-stage development of PL-treated zebrafish embryos. These results indicate that developing PL as a medicine would require extremely meticulous strategies to prevent potential toxicity.
Collapse
Affiliation(s)
- Hwang-Ju Jeon
- Red River Research Station, Louisiana State University Agricultural Center, Bossier City, LA, USA
| | - Chaeeun Kim
- Department of Applied Biosciences, Kyungpook National University, Daegu 41566, Republic of Korea; Department of Integrative Biology, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Kyeongnam Kim
- Department of Applied Biosciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Sung-Eun Lee
- Department of Applied Biosciences, Kyungpook National University, Daegu 41566, Republic of Korea; Department of Integrative Biology, Kyungpook National University, Daegu 41566, Republic of Korea.
| |
Collapse
|
7
|
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: 0.5] [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
|
8
|
Sung JH. Multi-organ-on-a-chip for pharmacokinetics and toxicokinetic study of drugs. Expert Opin Drug Metab Toxicol 2021; 17:969-986. [PMID: 33764248 DOI: 10.1080/17425255.2021.1908996] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Accurate prediction of pharmacokinetic (PK) and toxicokinetics (TK) of drugs is imperative for successful development of new pharmaceutics. Although conventional in vitro methods for predicting the PK and TK of drugs are well established, limitations still exist and more advanced chip-based in vitro platforms combined with mathematical models can help researchers overcome the limitations. Areas covered: We will review recent progress in the development of multi-organ-on-a-chip platforms for predicting PK and TK of drugs, as well as mathematical approaches that can be combined with these platforms for experiment design, data analysis and in vitro-in vivo extrapolation (IVIVE) for application to humans. Expert opinion: Although there remain some challenges to be addressed, the remarkable progress in the area of multi-organ-on-a-chip in recent years indicate that we will see tangible outcomes that can be utilized in the pharmaceutical industry in near future.
Collapse
Affiliation(s)
- Jong Hwan Sung
- Department of Chemical Engineering, Hongik University, Seoul, sejong, Republic of Korea
| |
Collapse
|
9
|
Zhong Y, Yu JS, Wang X, Binas B, Yoo HH. Chemical-based primary human hepatocyte monolayer culture for the study of drug metabolism and hepatotoxicity: Comparison with the spheroid model. FASEB J 2021; 35:e21379. [PMID: 33566373 DOI: 10.1096/fj.202001629rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 12/29/2020] [Accepted: 01/04/2021] [Indexed: 01/01/2023]
Abstract
Traditionally cultured monolayers of primary human hepatocytes (PHHs) deteriorate within days and thereby become unsuitable for drug-related studies. PHH spheroids (3D PHHs) maintain liver functions for weeks, but are considerably more demanding. Recently, a chemical-based approach (5C PHHs) succeeded in long-term culture of hepatocyte monolayers, but it remains unclear whether the drug-related functions are preserved. To clarify this, we compared the 5C and 3D PHHs in terms of gene expression analysis, proteomic analysis, functionality (basal and induced activities of representative CYP450 enzymes and urea and albumin secretions), survival in culture, and sensitivity to representative drugs. In all comparisons, which spanned culture durations of up to 4 weeks, the 5C PHHs performed at least as well as the 3D PHHs. Hence, the novel 5C PHH monolayer format combines the convenience of the traditional monolayer format with the functionality and maintainability of the spheroid format. Our results suggest that 5C PHH monolayers can be used more conveniently and efficiently for high-throughput drug screening, preclinical drug safety evaluations, and mechanistic studies.
Collapse
Affiliation(s)
- Yixiang Zhong
- Institute of Pharmaceutical Science and Technology and College of Pharmacy, Hanyang University, Ansan, Republic of Korea.,Department of Molecular & Life Science, Hanyang University, Ansan, Republic of Korea
| | - Jun Sang Yu
- Institute of Pharmaceutical Science and Technology and College of Pharmacy, Hanyang University, Ansan, Republic of Korea
| | - Xiaoqiong Wang
- Department of Molecular & Life Science, Hanyang University, Ansan, Republic of Korea
| | - Bert Binas
- Department of Molecular & Life Science, Hanyang University, Ansan, Republic of Korea
| | - Hye Hyun Yoo
- Institute of Pharmaceutical Science and Technology and College of Pharmacy, Hanyang University, Ansan, Republic of Korea
| |
Collapse
|
10
|
Inoue T, Iwazaki N, Araki T, Hitotsumachi H. Human-Induced Pluripotent Stem Cell-Derived Hepatocytes and their Culturing Methods to Maintain Liver Functions for Pharmacokinetics and Safety Evaluation of Pharmaceuticals. Curr Pharm Biotechnol 2020; 21:773-779. [PMID: 32003687 DOI: 10.2174/1389201021666200131123524] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 10/23/2019] [Accepted: 12/31/2019] [Indexed: 12/12/2022]
Abstract
Human hepatocytes are essential cell types for pharmacokinetics and the safety evaluation of pharmaceuticals. However, widely used primary hepatocytes with individual variations in liver function lose those functions rapidly in culture. Hepatic cell lines are convenient to use but have low liver functions. Human-Induced Pluripotent Stem (hiPS) cells can be expanded and potentially differentiated into any cell or tissue, including the liver. HiPS cell-derived Hepatocyte-Like Cells (hiPSHeps) are expected to be extensively used as consistent functional human hepatocytes. Many laboratories are investigating methods of using hiPS cells to differentiate hepatocytes, but the derived cells still have immature liver functions. In this paper, we describe the current uses and limitations of conventional hepatic cells, evaluating the suitability of hiPS-Heps to pharmacokinetics and the safety evaluation of pharmaceuticals, and discuss the potential future use of non-conventional non-monolayer culture methods to derive fully functional hiPS-Heps.
Collapse
Affiliation(s)
- Tomoaki Inoue
- Consortium for Safety Assessment using Human iPS Cells (CSAHi), Japan
| | - Norihiko Iwazaki
- Consortium for Safety Assessment using Human iPS Cells (CSAHi), Japan
| | - Tetsuro Araki
- Consortium for Safety Assessment using Human iPS Cells (CSAHi), Japan
| | | |
Collapse
|
11
|
Busche M, Tomilova O, Schütte J, Werner S, Beer M, Groll N, Hagmeyer B, Pawlak M, Jones PD, Schmees C, Becker H, Schnabel J, Gall K, Hemmler R, Matz-Soja M, Damm G, Beuck S, Klaassen T, Moer J, Ullrich A, Runge D, Schenke-Layland K, Gebhardt R, Stelzle M. HepaChip-MP - a twenty-four chamber microplate for a continuously perfused liver coculture model. LAB ON A CHIP 2020; 20:2911-2926. [PMID: 32662810 DOI: 10.1039/d0lc00357c] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
HepaChip microplate (HepaChip-MP) is a microfluidic platform comprised of 24 independent culture chambers with continuous, unidirectional perfusion. In the HepaChip-MP, an automated dielectrophoresis process selectively assembles viable cells into elongated micro tissues. Freshly isolated primary human hepatocytes (PHH) and primary human liver endothelial cells (HuLEC) were successfully assembled as cocultures aiming to mimic the liver sinusoid. Minimal quantities of primary human cells are required to establish micro tissues in the HepaChip-MP. Metabolic function including induction of CYP enzymes in PHH was successfully measured demonstrating a high degree of metabolic activity of cells in HepaChip-MP cultures and sufficient sensitivity of LC-MS analysis even for the relatively small number of cells per chamber. Further, parallelization realized in HepaChip-MP enabled the acquisition of dose-response toxicity data of diclofenac with a single device. Several unique technical features should enable a widespread application of this in vitro model. We have demonstrated fully automated preparation of cell cultures in HepaChip-MP using a pipetting robot. The tubeless unidirectional perfusion system based on gravity-driven flow can be operated within a standard incubator system. Overall, the system readily integrates in workflows common in cell culture labs. Further research will be directed towards optimization of media composition to further extend culture lifetime and study oxygen gradients and their effect on zonation within the sinusoid-like microorgans. In summary, we have established a novel parallelized and scalable microfluidic in vitro liver model showing hepatocyte function and anticipate future in-depth studies of liver biology and applications in pre-clinical drug development.
Collapse
Affiliation(s)
- Marius Busche
- NMI Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany.
| | - Olena Tomilova
- NMI Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany.
| | - Julia Schütte
- NMI Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany.
| | - Simon Werner
- NMI Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany.
| | - Meike Beer
- NMI Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany.
| | - Nicola Groll
- NMI Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany.
| | - Britta Hagmeyer
- NMI Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany.
| | - Michael Pawlak
- NMI Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany.
| | - Peter D Jones
- NMI Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany.
| | - Christian Schmees
- NMI Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany.
| | | | | | | | | | - Madlen Matz-Soja
- Section of Hepatology, Clinic and Polyclinic for Gastroenterology, Hepatology, Infectiology, Pneumology, University Clinic Leipzig, Leipzig, Germany and Rudolf-Schönheimer-Institute of Biochemistry, Leipzig University, Leipzig, Germany
| | - Georg Damm
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, Leipzig, Germany
| | - Simon Beuck
- A & M Labor fuer Analytik und Metabolismusforschung Service GmbH, Bergheim, Germany
| | - Tobias Klaassen
- A & M Labor fuer Analytik und Metabolismusforschung Service GmbH, Bergheim, Germany
| | - Jana Moer
- PRIMACYT Cell Culture Technology GmbH, Schwerin, Germany
| | - Anett Ullrich
- PRIMACYT Cell Culture Technology GmbH, Schwerin, Germany
| | - Dieter Runge
- PRIMACYT Cell Culture Technology GmbH, Schwerin, Germany
| | - Katja Schenke-Layland
- NMI Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany. and Department of Women's Health, Research Institute for Women's Health, Eberhard Karls University Tübingen, Tübingen, Germany and Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University Tübingen, Germany and Department of Medicine/Cardiology, Cardiovascular Research Laboratories (CVRL), University of California (UCLA), Los Angeles, CA, USA
| | - Rolf Gebhardt
- Section of Hepatology, Clinic and Polyclinic for Gastroenterology, Hepatology, Infectiology, Pneumology, University Clinic Leipzig, Leipzig, Germany and Rudolf-Schönheimer-Institute of Biochemistry, Leipzig University, Leipzig, Germany and InViSys-Tübingen GbR, Leipzig, Germany
| | - Martin Stelzle
- NMI Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany.
| |
Collapse
|
12
|
Del Favero G, Kraegeloh A. Integrating Biophysics in Toxicology. Cells 2020; 9:E1282. [PMID: 32455794 PMCID: PMC7290780 DOI: 10.3390/cells9051282] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/10/2020] [Accepted: 05/15/2020] [Indexed: 12/20/2022] Open
Abstract
Integration of biophysical stimulation in test systems is established in diverse branches of biomedical sciences including toxicology. This is largely motivated by the need to create novel experimental setups capable of reproducing more closely in vivo physiological conditions. Indeed, we face the need to increase predictive power and experimental output, albeit reducing the use of animals in toxicity testing. In vivo, mechanical stimulation is essential for cellular homeostasis. In vitro, diverse strategies can be used to model this crucial component. The compliance of the extracellular matrix can be tuned by modifying the stiffness or through the deformation of substrates hosting the cells via static or dynamic strain. Moreover, cells can be cultivated under shear stress deriving from the movement of the extracellular fluids. In turn, introduction of physical cues in the cell culture environment modulates differentiation, functional properties, and metabolic competence, thus influencing cellular capability to cope with toxic insults. This review summarizes the state of the art of integration of biophysical stimuli in model systems for toxicity testing, discusses future challenges, and provides perspectives for the further advancement of in vitro cytotoxicity studies.
Collapse
Affiliation(s)
- Giorgia Del Favero
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Währinger Straße 38-40, 1090 Vienna, Austria
- Core Facility Multimodal Imaging, Faculty of Chemistry, University of Vienna Währinger Straße 38-40, 1090 Vienna, Austria
| | - Annette Kraegeloh
- INM—Leibniz-Institut für Neue Materialien GmbH, Campus D2 2, 66123 Saarbrücken, Germany;
| |
Collapse
|
13
|
Hurrell T, Kastrinou-Lampou V, Fardellas A, Hendriks DFG, Nordling Å, Johansson I, Baze A, Parmentier C, Richert L, Ingelman-Sundberg M. Human Liver Spheroids as a Model to Study Aetiology and Treatment of Hepatic Fibrosis. Cells 2020; 9:cells9040964. [PMID: 32295224 PMCID: PMC7227007 DOI: 10.3390/cells9040964] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/02/2020] [Accepted: 04/11/2020] [Indexed: 12/16/2022] Open
Abstract
Non-alcoholic fatty liver disease affects approximately one billion adults worldwide. Non-alcoholic steatohepatitis (NASH) is a progressive disease and underlies the advancement to liver fibrosis, cirrhosis, and hepatocellular carcinoma, for which there are no FDA-approved drug therapies. We developed a hetero-cellular spheroid system comprised of primary human hepatocytes (PHH) co-cultured with crude fractions of primary human liver non-parenchymal cells (NPC) from several matched or non-matched donors, to identify phenotypes with utility in investigating NASH pathogenesis and drug screening. Co-culture spheroids displayed stable expression of hepatocyte markers (albumin, CYP3A4) with the integration of stellate (vimentin, PDGFRβ), endothelial (vWF, PECAM1), and CD68-positive cells. Several co-culture spheroids developed a fibrotic phenotype either spontaneously, primarily observed in PNPLA3 mutant donors, or after challenge with free fatty acids (FFA), as determined by COL1A1 and αSMA expression. This phenotype, as well as TGFβ1 expression, was attenuated with an ALK5 inhibitor. Furthermore, CYP2E1, which has a strong pro-oxidant effect, was induced by NPCs and FFA. This system was used to evaluate the effects of anti-NASH drug candidates, which inhibited fibrillary deposition following 7 days of exposure. In conclusion, we suggest that this system is suitable for the evaluation of NASH pathogenesis and screening of anti-NASH drug candidates.
Collapse
Affiliation(s)
- Tracey Hurrell
- Ingelman-Sundberg Group, Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, 171 65 Stockholm, Sweden; (T.H.); (V.K.-L.); (A.F.); (D.F.G.H.); (Å.N.); (I.J.)
| | - Vlasia Kastrinou-Lampou
- Ingelman-Sundberg Group, Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, 171 65 Stockholm, Sweden; (T.H.); (V.K.-L.); (A.F.); (D.F.G.H.); (Å.N.); (I.J.)
| | - Achilleas Fardellas
- Ingelman-Sundberg Group, Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, 171 65 Stockholm, Sweden; (T.H.); (V.K.-L.); (A.F.); (D.F.G.H.); (Å.N.); (I.J.)
| | - Delilah F. G. Hendriks
- Ingelman-Sundberg Group, Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, 171 65 Stockholm, Sweden; (T.H.); (V.K.-L.); (A.F.); (D.F.G.H.); (Å.N.); (I.J.)
| | - Åsa Nordling
- Ingelman-Sundberg Group, Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, 171 65 Stockholm, Sweden; (T.H.); (V.K.-L.); (A.F.); (D.F.G.H.); (Å.N.); (I.J.)
| | - Inger Johansson
- Ingelman-Sundberg Group, Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, 171 65 Stockholm, Sweden; (T.H.); (V.K.-L.); (A.F.); (D.F.G.H.); (Å.N.); (I.J.)
| | - Audrey Baze
- KaLy-Cell, 67115 Plobsheim, France; (A.B.); (C.P.); (L.R.)
| | | | | | - Magnus Ingelman-Sundberg
- Ingelman-Sundberg Group, Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, 171 65 Stockholm, Sweden; (T.H.); (V.K.-L.); (A.F.); (D.F.G.H.); (Å.N.); (I.J.)
- Correspondence:
| |
Collapse
|
14
|
Agarwal T, Biswas P, Pal S, Maiti TK, Chakraborty S, Ghosh SK, Dhar R. Inexpensive and Versatile Paper-Based Platform for 3D Culture of Liver Cells and Related Bioassays. ACS APPLIED BIO MATERIALS 2020; 3:2522-2533. [PMID: 35025303 DOI: 10.1021/acsabm.0c00237] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Tarun Agarwal
- Department of Biotechnology, Indian Institute of Technology, Kharagpur, West Bengal 721302, India
| | - Pratik Biswas
- Department of Biotechnology, Indian Institute of Technology, Kharagpur, West Bengal 721302, India
| | - Sampriti Pal
- Department of Biotechnology, Indian Institute of Technology, Kharagpur, West Bengal 721302, India
| | - Tapas Kumar Maiti
- Department of Biotechnology, Indian Institute of Technology, Kharagpur, West Bengal 721302, India
| | - Suman Chakraborty
- Department of Mechanical Engineering, Indian Institute of Technology, Kharagpur, West Bengal 721302, India
| | - Sudip Kumar Ghosh
- Department of Biotechnology, Indian Institute of Technology, Kharagpur, West Bengal 721302, India
| | - Riddhiman Dhar
- Department of Biotechnology, Indian Institute of Technology, Kharagpur, West Bengal 721302, India
| |
Collapse
|
15
|
da Silva Morais A, Vieira S, Zhao X, Mao Z, Gao C, Oliveira JM, Reis RL. Advanced Biomaterials and Processing Methods for Liver Regeneration: State-of-the-Art and Future Trends. Adv Healthc Mater 2020; 9:e1901435. [PMID: 31977159 DOI: 10.1002/adhm.201901435] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/13/2019] [Indexed: 12/17/2022]
Abstract
Liver diseases contribute markedly to the global burden of mortality and disease. The limited organ disposal for orthotopic liver transplantation results in a continuing need for alternative strategies. Over the past years, important progress has been made in the field of tissue engineering (TE). Many of the early trials to improve the development of an engineered tissue construct are based on seeding cells onto biomaterial scaffolds. Nowadays, several TE approaches have been developed and are applied to one vital organ: the liver. Essential elements must be considered in liver TE-cells and culturing systems, bioactive agents or growth factors (GF), and biomaterials and processing methods. The potential of hepatocytes, mesenchymal stem cells, and others as cell sources is demonstrated. They need engineered biomaterial-based scaffolds with perfect biocompatibility and bioactivity to support cell proliferation and hepatic differentiation as well as allowing extracellular matrix deposition and vascularization. Moreover, they require a microenvironment provided using conventional or advanced processing technologies in order to supply oxygen, nutrients, and GF. Herein the biomaterials and the conventional and advanced processing technologies, including cell-sheets process, 3D bioprinting, and microfluidic systems, as well as the future trends in these major fields are discussed.
Collapse
Affiliation(s)
- Alain da Silva Morais
- 3B's Research GroupI3Bs – Research Institute on Biomaterials, Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine 4805‐017 Barco Guimarães Portugal
- ICVS/3B's–PT Government Associate Laboratory Braga/ Guimarães Portugal
| | - Sílvia Vieira
- 3B's Research GroupI3Bs – Research Institute on Biomaterials, Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine 4805‐017 Barco Guimarães Portugal
- ICVS/3B's–PT Government Associate Laboratory Braga/ Guimarães Portugal
| | - Xinlian Zhao
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationDepartment of Polymer Science and EngineeringZhejiang University Hangzhou 310027 China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationDepartment of Polymer Science and EngineeringZhejiang University Hangzhou 310027 China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationDepartment of Polymer Science and EngineeringZhejiang University Hangzhou 310027 China
| | - Joaquim M. Oliveira
- 3B's Research GroupI3Bs – Research Institute on Biomaterials, Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine 4805‐017 Barco Guimarães Portugal
- ICVS/3B's–PT Government Associate Laboratory Braga/ Guimarães Portugal
- The Discoveries Centre for Regenerative and Precision MedicineUniversity of Minho 4805‐017 Barco Guimarães Portugal
| | - Rui L. Reis
- 3B's Research GroupI3Bs – Research Institute on Biomaterials, Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine 4805‐017 Barco Guimarães Portugal
- ICVS/3B's–PT Government Associate Laboratory Braga/ Guimarães Portugal
- The Discoveries Centre for Regenerative and Precision MedicineUniversity of Minho 4805‐017 Barco Guimarães Portugal
| |
Collapse
|
16
|
Fowler S, Chen WLK, Duignan DB, Gupta A, Hariparsad N, Kenny JR, Lai WG, Liras J, Phillips JA, Gan J. Microphysiological systems for ADME-related applications: current status and recommendations for system development and characterization. LAB ON A CHIP 2020; 20:446-467. [PMID: 31932816 DOI: 10.1039/c9lc00857h] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Over the last decade, progress has been made on the development of microphysiological systems (MPS) for absorption, distribution, metabolism, and excretion (ADME) applications. Central to this progress has been proof of concept data generated by academic and industrial institutions followed by broader characterization studies, which provide evidence for scalability and applicability to drug discovery and development. In this review, we describe some of the advances made for specific tissue MPS and outline the desired functionality for such systems, which are likely to make them applicable for practical use in the pharmaceutical industry. Single organ MPS platforms will be valuable for modelling tissue-specific functions. However, dynamic organ crosstalk, especially in the context of disease or toxicity, can only be obtained with the use of inter-linked MPS models which will enable scientists to address questions at the intersection of pharmacokinetics (PK) and efficacy, or PK and toxicity. In the future, successful application of MPS platforms that closely mimic human physiology may ultimately reduce the need for animal models to predict ADME outcomes and decrease the overall risk and cost associated with drug development.
Collapse
Affiliation(s)
- Stephen Fowler
- Pharma Research and Early Development, F.Hoffmann-La Roche Ltd, Grenzacherstrasse 124, CH4070, Basel, Switzerland
| | | | - David B Duignan
- Department of Drug Metabolism, Pharmacokinetics & Bioanalysis, AbbVie Bioresearch Center, Worcester, Massachusetts 01605, USA
| | - Anshul Gupta
- Amgen Research, 360 Binney St, Cambridge, MA 02141, USA
| | - Niresh Hariparsad
- Department of Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals, 50 Northern Ave, Boston, MA, USA
| | - Jane R Kenny
- DMPK, Genentech, 1 DNA Way, South San Francisco 94080, USA
| | | | - Jennifer Liras
- Medicine Design, Pfizer Inc, 1 Portland Ave, Cambridge, MA 02139, USA
| | | | - Jinping Gan
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb R&D, PO Box 4000, Princeton, NJ 08543-4000, USA.
| |
Collapse
|
17
|
Baudy AR, Otieno MA, Hewitt P, Gan J, Roth A, Keller D, Sura R, Van Vleet TR, Proctor WR. Liver microphysiological systems development guidelines for safety risk assessment in the pharmaceutical industry. LAB ON A CHIP 2020; 20:215-225. [PMID: 31799979 DOI: 10.1039/c9lc00768g] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The liver is critical to consider during drug development because of its central role in the handling of xenobiotics, a process which often leads to localized and/or downstream tissue injury. Our ability to predict human clinical safety outcomes with animal testing is limited due to species differences in drug metabolism and disposition, while traditional human in vitro liver models often lack the necessary in vivo physiological fidelity. To address this, increasing numbers of liver microphysiological systems (MPS) are being developed, however the inconsistency in their optimization and characterization often leads to models that do not possess critical levels of baseline performance that is required for many pharmaceutical industry applications. Herein we provide a guidance on best approaches to benchmark liver MPS based on 3 stages of characterization that includes key performance metrics and a 20 compound safety test set. Additionally, we give an overview of frequently used liver injury safety assays, describe the ideal MPS model, and provide a perspective on currently best suited MPS contexts of use. This pharmaceutical industry guidance has been written to help MPS developers and end users identify what could be the most valuable models for safety risk assessment.
Collapse
Affiliation(s)
| | - Monicah A Otieno
- Janssen Pharmaceutical Research and Development, Spring House, PA, USA
| | | | - Jinping Gan
- Bristol-Myers Squibb, New York City, NY, USA
| | | | | | | | | | | |
Collapse
|
18
|
Fabrication and evaluation of modified poly(ethylene terephthalate) microfibrous scaffolds for hepatocyte growth and functionality maintenance. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 109:110523. [PMID: 32228959 DOI: 10.1016/j.msec.2019.110523] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 11/15/2019] [Accepted: 12/05/2019] [Indexed: 12/13/2022]
Abstract
For hepatocyte culture in vitro, the surface feature of utilized scaffolds exerts a direct impact on cell adhesion, growth and differentiated functionality. Herein, to regulate hepatocyte growth and differentiated functionality, modified microfibrous scaffolds were fabricated by surface grafting monoamine terminated lactobionic lactone (L-NH2) and gelatin onto non-woven poly(ethylene terephthalate) (PET) fibrous substrate (PET-Gal and PET-Gel), respectively. The physicochemical properties of PET scaffolds before and after modification were characterized. Upon 15-day culture, the effects of modified PET scaffolds on growth and differentiated functionality of human induced hepatocytes (hiHeps) were evaluated, compared with that of control without modification. Results demonstrated that both L-NH2 and gelatin modifications improved scaffold properties including hydrophilicity, water uptake ratio, stiffness and roughness, resulting in efficient cell adhesion, ~20-fold cell expansion and enhanced differentiated functionality. After culture for 15 days, PET-Gal cultured cells formed aggregates, displaying better cell viability and significantly higher differentiated functionality regarding albumin secretion, urea synthesis, phases I (cytochrome P450, CYP1A1/2 and CYP3A4) and II (uridine 5'-diphosphate glucuronosyltransferases, UGT) enzyme activity, biliary excretion and detoxification ability (ammonia elimination and bilirubin conjugation), compared with PET and PET-Gel cultured ones. Hence, as a three-dimensional (3D) microfibrous scaffold, PET-Gal promotes hiHeps growth and differentiated functionality maintenance, which is promisingly utilized in bioartificial liver (BAL) bioreactors.
Collapse
|
19
|
Hadida M, Marchat D. Strategy for achieving standardized bone models. Biotechnol Bioeng 2019; 117:251-271. [PMID: 31531968 PMCID: PMC6915912 DOI: 10.1002/bit.27171] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/05/2019] [Accepted: 09/06/2019] [Indexed: 12/24/2022]
Abstract
Reliably producing functional in vitro organ models, such as organ-on-chip systems, has the potential to considerably advance biology research, drug development time, and resource efficiency. However, despite the ongoing major progress in the field, three-dimensional bone tissue models remain elusive. In this review, we specifically investigate the control of perfusion flow effects as the missing link between isolated culture systems and scientifically exploitable bone models and propose a roadmap toward this goal.
Collapse
Affiliation(s)
- Mikhael Hadida
- Mines Saint-Etienne, Univ Lyon, Univ Jean Monnet, INSERM, U 1059 Sainbiose, Centre CIS, Saint-Etienne, France
| | - David Marchat
- Mines Saint-Etienne, Univ Lyon, Univ Jean Monnet, INSERM, U 1059 Sainbiose, Centre CIS, Saint-Etienne, France
| |
Collapse
|
20
|
Shen JX, Youhanna S, Zandi Shafagh R, Kele J, Lauschke VM. Organotypic and Microphysiological Models of Liver, Gut, and Kidney for Studies of Drug Metabolism, Pharmacokinetics, and Toxicity. Chem Res Toxicol 2019; 33:38-60. [DOI: 10.1021/acs.chemrestox.9b00245] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Joanne X. Shen
- Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Sonia Youhanna
- Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Reza Zandi Shafagh
- Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
- Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Julianna Kele
- Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Volker M. Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| |
Collapse
|
21
|
Ohbuchi M. [Application of novel 3D-bioprinted human liver tissue models in drug discovery research]. Nihon Yakurigaku Zasshi 2019; 153:284-288. [PMID: 31178534 DOI: 10.1254/fpj.153.284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Primary human hepatocytes are widely used to study drug metabolism and enzyme induction. However, primary hepatocytes rapidly lose their hepatic function in conventional 2D cultures. Recently, a microphysiological system that overcomes this drawback has been actively investigated and applied in drug discovery research. Such novel in vitro models are desirable for the evaluation of the metabolic clearance of drugs with low turnover, drug-induced liver injury, and chronic liver diseases like liver fibrosis. This article reviews the characteristics and recent advances in 3D-bioprinted human liver tissue models in drug discovery research.
Collapse
Affiliation(s)
- Masato Ohbuchi
- Analysis & Pharmacokinetic Research Labs., Drug Discovery Research, Astellas Pharma Inc
| |
Collapse
|
22
|
Evaluation of Drug Biliary Excretion Using Sandwich-Cultured Human Hepatocytes. Eur J Drug Metab Pharmacokinet 2019; 44:13-30. [PMID: 30167999 DOI: 10.1007/s13318-018-0502-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Evaluation of hepatobiliary transport of drugs is an important challenge, notably during the development of new molecular identities. In this context, sandwich-cultured human hepatocytes (SCHH) have been proposed as an interesting and integrated tool for predicting in vitro biliary excretion of drugs. The present review was therefore designed to summarize key findings about SCHH, including their establishment, their main functional features and their use for the determination of canalicular transport and the prediction of in vivo biliary clearance and hepatobiliary excretion-related drug-drug interactions. Reviewed data highlight the fact that SCHH represent an original and probably unique holistic in vitro approach to predict biliary clearance in humans, through taking into account sinusoidal drug uptake, passive drug diffusion, drug metabolism and sinusoidal and canalicular drug efflux. Limits and proposed refinements for SCHH-based analysis of drug biliary excretion, as well as putative human alternative in vitro models to SCHH are also discussed.
Collapse
|
23
|
Legallais C, Kim D, Mihaila SM, Mihajlovic M, Figliuzzi M, Bonandrini B, Salerno S, Yousef Yengej FA, Rookmaaker MB, Sanchez Romero N, Sainz-Arnal P, Pereira U, Pasqua M, Gerritsen KGF, Verhaar MC, Remuzzi A, Baptista PM, De Bartolo L, Masereeuw R, Stamatialis D. Bioengineering Organs for Blood Detoxification. Adv Healthc Mater 2018; 7:e1800430. [PMID: 30230709 DOI: 10.1002/adhm.201800430] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 08/23/2018] [Indexed: 12/11/2022]
Abstract
For patients with severe kidney or liver failure the best solution is currently organ transplantation. However, not all patients are eligible for transplantation and due to limited organ availability, most patients are currently treated with therapies using artificial kidney and artificial liver devices. These therapies, despite their relative success in preserving the patients' life, have important limitations since they can only replace part of the natural kidney or liver functions. As blood detoxification (and other functions) in these highly perfused organs is achieved by specialized cells, it seems relevant to review the approaches leading to bioengineered organs fulfilling most of the native organ functions. There, the culture of cells of specific phenotypes on adapted scaffolds that can be perfused takes place. In this review paper, first the functions of kidney and liver organs are briefly described. Then artificial kidney/liver devices, bioartificial kidney devices, and bioartificial liver devices are focused on, as well as biohybrid constructs obtained by decellularization and recellularization of animal organs. For all organs, a thorough overview of the literature is given and the perspectives for their application in the clinic are discussed.
Collapse
Affiliation(s)
- Cécile Legallais
- UMR CNRS 7338 Biomechanics & Bioengineering; Université de technologie de Compiègne; Sorbonne Universités; 60203 Compiègne France
| | - Dooli Kim
- (Bio)artificial organs; Department of Biomaterials Science and Technology; Faculty of Science and Technology; TechMed Institute; University of Twente; P.O. Box 217 7500 AE Enschede The Netherlands
| | - Sylvia M. Mihaila
- Division of Pharmacology; Utrecht Institute for Pharmaceutical Sciences; Utrecht University; Universiteitsweg 99 3584 CG Utrecht The Netherlands
- Department of Nephrology and Hypertension; University Medical Center Utrecht and Regenerative Medicine Utrecht; Utrecht University; Heidelberglaan 100 3584 CX Utrecht The Netherlands
| | - Milos Mihajlovic
- Division of Pharmacology; Utrecht Institute for Pharmaceutical Sciences; Utrecht University; Universiteitsweg 99 3584 CG Utrecht The Netherlands
| | - Marina Figliuzzi
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri; via Stezzano 87 24126 Bergamo Italy
| | - Barbara Bonandrini
- Department of Chemistry; Materials and Chemical Engineering “Giulio Natta”; Politecnico di Milano; Piazza Leonardo da Vinci 32 20133 Milan Italy
| | - Simona Salerno
- Institute on Membrane Technology; National Research Council of Italy; ITM-CNR; Via Pietro BUCCI, Cubo 17C - 87036 Rende Italy
| | - Fjodor A. Yousef Yengej
- Department of Nephrology and Hypertension; University Medical Center Utrecht and Regenerative Medicine Utrecht; Utrecht University; Heidelberglaan 100 3584 CX Utrecht The Netherlands
| | - Maarten B. Rookmaaker
- Department of Nephrology and Hypertension; University Medical Center Utrecht and Regenerative Medicine Utrecht; Utrecht University; Heidelberglaan 100 3584 CX Utrecht The Netherlands
| | | | - Pilar Sainz-Arnal
- Instituto de Investigación Sanitaria de Aragón (IIS Aragon); 50009 Zaragoza Spain
- Instituto Aragonés de Ciencias de la Salud (IACS); 50009 Zaragoza Spain
| | - Ulysse Pereira
- UMR CNRS 7338 Biomechanics & Bioengineering; Université de technologie de Compiègne; Sorbonne Universités; 60203 Compiègne France
| | - Mattia Pasqua
- UMR CNRS 7338 Biomechanics & Bioengineering; Université de technologie de Compiègne; Sorbonne Universités; 60203 Compiègne France
| | - Karin G. F. Gerritsen
- Department of Nephrology and Hypertension; University Medical Center Utrecht and Regenerative Medicine Utrecht; Utrecht University; Heidelberglaan 100 3584 CX Utrecht The Netherlands
| | - Marianne C. Verhaar
- Department of Nephrology and Hypertension; University Medical Center Utrecht and Regenerative Medicine Utrecht; Utrecht University; Heidelberglaan 100 3584 CX Utrecht The Netherlands
| | - Andrea Remuzzi
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri; via Stezzano 87 24126 Bergamo Italy
- Department of Management; Information and Production Engineering; University of Bergamo; viale Marconi 5 24044 Dalmine Italy
| | - Pedro M. Baptista
- Instituto de Investigación Sanitaria de Aragón (IIS Aragon); 50009 Zaragoza Spain
- Department of Management; Information and Production Engineering; University of Bergamo; viale Marconi 5 24044 Dalmine Italy
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas (CIBERehd); 28029 Barcelona Spain
- Fundación ARAID; 50009 Zaragoza Spain
- Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz; 28040 Madrid Spain. Department of Biomedical and Aerospace Engineering; Universidad Carlos III de Madrid; 28911 Madrid Spain
| | - Loredana De Bartolo
- Institute on Membrane Technology; National Research Council of Italy; ITM-CNR; Via Pietro BUCCI, Cubo 17C - 87036 Rende Italy
| | - Rosalinde Masereeuw
- Division of Pharmacology; Utrecht Institute for Pharmaceutical Sciences; Utrecht University; Universiteitsweg 99 3584 CG Utrecht The Netherlands
| | - Dimitrios Stamatialis
- (Bio)artificial organs; Department of Biomaterials Science and Technology; Faculty of Science and Technology; TechMed Institute; University of Twente; P.O. Box 217 7500 AE Enschede The Netherlands
| |
Collapse
|
24
|
Use of hepatocytes isolated from a liver-humanized mouse for studies on the metabolism of drugs: application to the metabolism of fentanyl and acetylfentanyl. Forensic Toxicol 2018; 36:467-475. [PMID: 29963210 PMCID: PMC6002451 DOI: 10.1007/s11419-018-0425-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 05/07/2018] [Indexed: 01/03/2023]
Abstract
Purpose The usefulness of hepatocytes isolated from a liver-humanized mouse (PXB-cells) as a model in vitro system for the prediction of the in vivo metabolism of new drugs of abuse was evaluated. Methods For the drug metabolism study, fentanyl, a powerful synthetic opioid, and acetylfentanyl, an N-acetyl analog of fentanyl, were selected as model drugs. PXB-cells were cultured with the drug for 24–48 h and then the media were collected and analyzed by liquid chromatography/mass spectrometry after deproteinization with acetonitrile. Results The main metabolite formed from fentanyl by PXB-cells was the desphenethylated metabolite (nor-fentanyl), and the other major metabolites formed were 4′-hydroxy-fentanyl, β-hydroxy-fentanyl and (ω-1)-hydroxy-fentanyl. ω-Hydroxy-fentanyl and 4′-hydroxy-3′-methoxy-fentanyl were the minor metabolites. Similar results were obtained for acetylfentanyl. The metabolite profile of fentanyl in PXB-cells was consistent with the in vivo metabolite profile of fentanyl reported previously. Most of the 4′-hydroxy- and 4′-hydroxy-3′-methoxy-metabolites of fentanyl and acetylfentanyl were conjugated in PXB-cells, indicating that PXB-cells had high conjugation enzyme activities. From experiments using human liver microsomes and anti-CYP antibodies, it was revealed that CYP3A4 was involved in the production of nor-fentanyl, β-hydroxy-fentanyl and (ω-1)-hydroxy-fentanyl, while CYP2D6 was partially involved in the production of 4′-hydroxy-fentanyl. Conclusions Our results indicated that PXB-cells have high activities of phase I and phase II drug-metabolizing-enzymes, can be stably supplied, and are easy to use; thus, PXB-cells are highly useful for the prediction of the in vivo metabolism of drugs of abuse.
Collapse
|
25
|
Smith AK, Xu Y, Ropella GEP, Hunt CA. A Model Mechanism-Based Explanation of an In Vitro-In Vivo Disconnect for Improving Extrapolation and Translation. J Pharmacol Exp Ther 2018; 365:127-138. [PMID: 29434053 DOI: 10.1124/jpet.117.245019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Accepted: 02/05/2018] [Indexed: 11/22/2022] Open
Abstract
An improved understanding of in vivo-to-in vitro hepatocyte changes is crucial to interpreting in vitro data correctly and further improving hepatocyte-based in vitro-to-in vivo extrapolations to human targets. We demonstrate using virtual experiments as a means of helping to untangle plausible causes of inaccurate extrapolations. We start with virtual mice that use biomimetic software livers. Previously, using these mice, we discovered model mechanisms that enabled achieving quantitative validation targets while also providing plausible causal explanations for temporal characteristics of acetaminophen hepatotoxicity. We isolated virtual hepatocytes, created a virtual culture, and then conducted dose-response experiments in both culture and mice. We expected to see differences between the two dose-response curves but were somewhat surprised that they crossed because it evidenced that simulated acetaminophen metabolism and toxicity are different for virtual culture and mouse contexts even though individual hepatocyte mechanisms were unchanged. Differences in dose-response curves provide a virtual example of an in vivo-to-in vitro disconnect. We use detailed results of experiments to explain this disconnect. Individual hepatocytes contribute differently to system-level phenomena. In liver, hepatocytes are exposed to acetaminophen sequentially. Relative production of the reactive acetaminophen metabolite is largest (smallest) in pericentral (periportal) hepatocytes. Because that sequential exposure is absent in culture, hepatocytes from different lobular locations do not respond the same. A virtual culture-to-mouse translation can stand as a scientifically challengeable hypothesis explaining an in vivo-to-in vitro disconnect. It provides a framework to develop more reliable interpretations of in vitro observations, which then may be used to improve extrapolations.
Collapse
Affiliation(s)
- Andrew K Smith
- Bioengineering and Therapeutic Sciences, University of California, San Francisco, California (A.K.S., Y.X., C.A.H.); and Tempus Dictum, Inc., Milwaukie, Oregon (G.E.P.R.)
| | - Yanli Xu
- Bioengineering and Therapeutic Sciences, University of California, San Francisco, California (A.K.S., Y.X., C.A.H.); and Tempus Dictum, Inc., Milwaukie, Oregon (G.E.P.R.)
| | - Glen E P Ropella
- Bioengineering and Therapeutic Sciences, University of California, San Francisco, California (A.K.S., Y.X., C.A.H.); and Tempus Dictum, Inc., Milwaukie, Oregon (G.E.P.R.)
| | - C Anthony Hunt
- Bioengineering and Therapeutic Sciences, University of California, San Francisco, California (A.K.S., Y.X., C.A.H.); and Tempus Dictum, Inc., Milwaukie, Oregon (G.E.P.R.)
| |
Collapse
|
26
|
Technical aspects of microphysiological systems (MPS) as a promising wet human-in-vivo simulator. Drug Metab Pharmacokinet 2018; 33:40-42. [DOI: 10.1016/j.dmpk.2017.11.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 10/24/2017] [Accepted: 11/17/2017] [Indexed: 11/23/2022]
|
27
|
Organs-on-a-chip: Current applications and consideration points for in vitro ADME-Tox studies. Drug Metab Pharmacokinet 2018; 33:49-54. [DOI: 10.1016/j.dmpk.2018.01.003] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 12/24/2017] [Accepted: 12/27/2017] [Indexed: 02/07/2023]
|
28
|
Matsunaga N, Fukuchi Y, Imawaka H, Tamai I. Sandwich-Cultured Hepatocytes for Mechanistic Understanding of Hepatic Disposition of Parent Drugs and Metabolites by Transporter-Enzyme Interplay. Drug Metab Dispos 2018; 46:680-691. [PMID: 29352067 DOI: 10.1124/dmd.117.079236] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 01/17/2018] [Indexed: 12/13/2022] Open
Abstract
Functional interplay between transporters and drug-metabolizing enzymes is currently one of the hottest topics in the field of drug metabolism and pharmacokinetics. Uptake transporter-enzyme interplay is important to determine intrinsic hepatic clearance based on the extended clearance concept. Enzyme and efflux transporter interplay, which includes both sinusoidal (basolateral) and canalicular efflux transporters, determines the fate of metabolites formed in the liver. As sandwich-cultured hepatocytes (SCHs) maintain metabolic activities and form a canalicular network, the whole interplay between uptake and efflux transporters and drug-metabolizing enzymes can be investigated simultaneously. In this article, we review the utility and applicability of SCHs for mechanistic understanding of hepatic disposition of both parent drugs and metabolites. In addition, the utility of SCHs for mimicking species-specific disposition of parent drugs and metabolites in vivo is described. We also review application of SCHs for clinically relevant prediction of drug-drug interactions caused by drugs and metabolites. The usefulness of mathematical modeling of hepatic disposition of parent drugs and metabolites in SCHs is described to allow a quantitative understanding of an event in vitro and to develop a more advanced model to predict in vivo disposition.
Collapse
Affiliation(s)
- Norikazu Matsunaga
- Pharmacokinetic Research Laboratories, Ono Pharmaceutical Co., Ltd., Tsukuba, Japan (N.M. Y.F., H.I.); Department of Membrane Transport and Biopharmaceutics, Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (I.T.)
| | - Yukina Fukuchi
- Pharmacokinetic Research Laboratories, Ono Pharmaceutical Co., Ltd., Tsukuba, Japan (N.M. Y.F., H.I.); Department of Membrane Transport and Biopharmaceutics, Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (I.T.)
| | - Haruo Imawaka
- Pharmacokinetic Research Laboratories, Ono Pharmaceutical Co., Ltd., Tsukuba, Japan (N.M. Y.F., H.I.); Department of Membrane Transport and Biopharmaceutics, Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (I.T.)
| | - Ikumi Tamai
- Pharmacokinetic Research Laboratories, Ono Pharmaceutical Co., Ltd., Tsukuba, Japan (N.M. Y.F., H.I.); Department of Membrane Transport and Biopharmaceutics, Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (I.T.)
| |
Collapse
|