1
|
Afewerki S, Stocco TD, Rosa da Silva AD, Aguiar Furtado AS, Fernandes de Sousa G, Ruiz-Esparza GU, Webster TJ, Marciano FR, Strømme M, Zhang YS, Lobo AO. In vitro high-content tissue models to address precision medicine challenges. Mol Aspects Med 2023; 91:101108. [PMID: 35987701 PMCID: PMC9384546 DOI: 10.1016/j.mam.2022.101108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/29/2022] [Accepted: 07/20/2022] [Indexed: 01/18/2023]
Abstract
The field of precision medicine allows for tailor-made treatments specific to a patient and thereby improve the efficiency and accuracy of disease prevention, diagnosis, and treatment and at the same time would reduce the cost, redundant treatment, and side effects of current treatments. Here, the combination of organ-on-a-chip and bioprinting into engineering high-content in vitro tissue models is envisioned to address some precision medicine challenges. This strategy could be employed to tackle the current coronavirus disease 2019 (COVID-19), which has made a significant impact and paradigm shift in our society. Nevertheless, despite that vaccines against COVID-19 have been successfully developed and vaccination programs are already being deployed worldwide, it will likely require some time before it is available to everyone. Furthermore, there are still some uncertainties and lack of a full understanding of the virus as demonstrated in the high number new mutations arising worldwide and reinfections of already vaccinated individuals. To this end, efficient diagnostic tools and treatments are still urgently needed. In this context, the convergence of bioprinting and organ-on-a-chip technologies, either used alone or in combination, could possibly function as a prominent tool in addressing the current pandemic. This could enable facile advances of important tools, diagnostics, and better physiologically representative in vitro models specific to individuals allowing for faster and more accurate screening of therapeutics evaluating their efficacy and toxicity. This review will cover such technological advances and highlight what is needed for the field to mature for tackling the various needs for current and future pandemics as well as their relevancy towards precision medicine.
Collapse
Affiliation(s)
- Samson Afewerki
- Division of Nanotechnology and Functional Materials, Department of Materials Science and Engineering, Ångström Laboratory, Uppsala University, BOX 35, 751 03, Uppsala, Sweden
| | - Thiago Domingues Stocco
- Bioengineering Program, Technological and Scientific Institute, Brazil University, 08230-030, São Paulo, SP, Brazil; Faculty of Medical Sciences, Unicamp - State University of Campinas, 13083-877, Campinas, SP, Brazil
| | | | - André Sales Aguiar Furtado
- Interdisciplinary Laboratory for Advanced Materials, BioMatLab, Department of Materials Engineering, Federal University of Piauí (UFPI), Teresina, PI, Brazil
| | - Gustavo Fernandes de Sousa
- Interdisciplinary Laboratory for Advanced Materials, BioMatLab, Department of Materials Engineering, Federal University of Piauí (UFPI), Teresina, PI, Brazil
| | - Guillermo U Ruiz-Esparza
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA; Division of Health Sciences and Technology, Harvard University ‑ Massachusetts Institute of Technology, Boston, MA, 02115, USA
| | - Thomas J Webster
- Interdisciplinary Laboratory for Advanced Materials, BioMatLab, Department of Materials Engineering, Federal University of Piauí (UFPI), Teresina, PI, Brazil; Hebei University of Technology, Tianjin, China
| | - Fernanda R Marciano
- Department of Physics, Federal University of Piauí (UFPI), Teresina, PI, Brazil
| | - Maria Strømme
- Division of Nanotechnology and Functional Materials, Department of Materials Science and Engineering, Ångström Laboratory, Uppsala University, BOX 35, 751 03, Uppsala, Sweden
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA; Division of Health Sciences and Technology, Harvard University ‑ Massachusetts Institute of Technology, Boston, MA, 02115, USA.
| | - Anderson Oliveira Lobo
- Interdisciplinary Laboratory for Advanced Materials, BioMatLab, Department of Materials Engineering, Federal University of Piauí (UFPI), Teresina, PI, Brazil.
| |
Collapse
|
2
|
Lin MW, Huang HL, Yu XR, Lin YX, Li XA, Tsui L, Young SS, Chan HL, Chou HC. Urban sediment pollutants alternate human cell essential behaviour through promoting oxidative damage. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 229:113065. [PMID: 34920185 DOI: 10.1016/j.ecoenv.2021.113065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 11/20/2021] [Accepted: 12/05/2021] [Indexed: 06/14/2023]
Abstract
The main objective of this study was to establish a human cell-based platform to assess the effects of sediment toxicity on oxidative damage and cell essential behaviour. Since sediment pollution has increased as a consequence of including but not limited to industrialisation, the contaminants accumulated in sediments have already led to human health concerns. The Hsinchu Science Park is one of the most prominent semiconductor manufacturing centres in the world, and the Ke-Ya River flows through Hsinchu Science Park and the Hsinchu urban district. Because semiconductor wastes potentially contribute to higher-than-normal rates of cancers, birth defects, and serious diseases, the quality assessment of the Ke-Ya River has prompted widespread concerns. While previous studies have shown an association between the degradation of fish populations and sediment pollutants, very little is known about the issues on human health. Herein, the effects of sediment from three sediment sampling sites of the Ke-Ya River on 11 different human cell lines were directly evaluated. The upstream represents the undeveloped zone, the middle-stream represents the household/industrial wastewater zone, and the downstream represents the accumulation zone. Our results indicated that the sediment pollution of the downstream Ke-Ya River was more cytotoxic than that of the middle stream and upstream. Downstream sediment extract (DSE) significantly increased reactive oxygen species (ROS) levels across all cell types. Accordingly, oxidative stress can trigger redox-sensitive pathways and alter essential biological processes such as cell viability, cell adhesion, and cell motility. Importantly, the MTT assay indicated that DSE significantly decreased the viability of brain, oral, lung, breast, liver, pancreatic, cervical, prostate, and colorectal cells. Furthermore, the adhesive ability and wound healing ability of most cells were greatly reduced in the presence of DSE compared to other conditions. Thus, this study shows the results of the first analyses completed on the sediment cytotoxicity in human cells, and stimulated ROS levels are crucial for cellular life. In future research, the detailed cause and effect mechanisms of the abundant ROS generated in DSE will be further investigated. We sincerely hope that our study provides a scientific basis for further investigations with a global perspective on public health challenges.
Collapse
Affiliation(s)
- Meng-Wei Lin
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Hsiao-Lan Huang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Xin-Ru Yu
- Institute of Analytical and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Yu-Xuan Lin
- Department of Safety, Health and Environmental Engineering, Ming Chi University of Technology, New Taipei City, Taiwan
| | - Xi-An Li
- Department of Safety, Health and Environmental Engineering, Ming Chi University of Technology, New Taipei City, Taiwan
| | - Lo Tsui
- Department of Safety, Health and Environmental Engineering, Ming Chi University of Technology, New Taipei City, Taiwan
| | - Shuh-Sen Young
- Institute of Analytical and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| | - Hong-Lin Chan
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan; Department of Medical Science and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan.
| | - Hsiu-Chuan Chou
- Institute of Analytical and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
3
|
Sonker AK, Bhateria M, Karsauliya K, Singh SP. Investigating the glucuronidation and sulfation pathways contribution and disposition kinetics of Bisphenol S and its metabolites using LC-MS/MS-based nonenzymatic hydrolysis method. CHEMOSPHERE 2021; 273:129624. [PMID: 33515962 DOI: 10.1016/j.chemosphere.2021.129624] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 01/07/2021] [Accepted: 01/08/2021] [Indexed: 06/12/2023]
Abstract
Despite showing serious health consequences and widespread exposure, the toxicokinetic information required to evaluate the health risks of BPS is insufficient. Thus, we aim to describe the comprehensive toxicokinetics of BPS and its glucuronide (BPS-G) and sulfate (BPS-S) metabolites in rats. Simultaneous quantification of BPS and its metabolites (authentic standards) was accomplished using UPLC-MS/MS method. BPS displayed rapid absorption, extensive metabolism and fast elimination after oral administration. Following intravenous administration, BPS exhibited CL (8.8 L/h/kg) higher than the rat hepatic blood flow rate suggesting the likelihood of extrahepatic clearance. The CL value differed from those reported previously (sheep and piglets) and the probable reason could be attributed to dose- and/or interspecies differences. BPS was extensively metabolized and excreted primarily through urine as BPS-G (∼56%). BPS and BPS-S exhibited a high protein binding capacity in comparison to BPS-G. In in vitro metabolic stability study, BPS was predominantly metabolized through glucuronidation. The predicted in vivo hepatic clearance of BPS suggested it to be a high and intermediate clearance chemical in rats and humans, respectively. The significant interspecies difference observed in the clearance of BPS between rats and humans indicated that toxicokinetics of BPS should be considered for health risk assessment in humans.
Collapse
Affiliation(s)
- Ashish Kumar Sonker
- Toxicokinetics Laboratory & Regulatory Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India; Analytical Chemistry Laboratory & Regulatory Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, India
| | - Manisha Bhateria
- Toxicokinetics Laboratory & Regulatory Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, India; Analytical Chemistry Laboratory & Regulatory Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, India
| | - Kajal Karsauliya
- Toxicokinetics Laboratory & Regulatory Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, India; Analytical Chemistry Laboratory & Regulatory Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, India
| | - Sheelendra Pratap Singh
- Toxicokinetics Laboratory & Regulatory Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India; Analytical Chemistry Laboratory & Regulatory Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, India.
| |
Collapse
|
4
|
Khalil AS, Jaenisch R, Mooney DJ. Engineered tissues and strategies to overcome challenges in drug development. Adv Drug Deliv Rev 2020; 158:116-139. [PMID: 32987094 PMCID: PMC7518978 DOI: 10.1016/j.addr.2020.09.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/29/2020] [Accepted: 09/23/2020] [Indexed: 12/16/2022]
Abstract
Current preclinical studies in drug development utilize high-throughput in vitro screens to identify drug leads, followed by both in vitro and in vivo models to predict lead candidates' pharmacokinetic and pharmacodynamic properties. The goal of these studies is to reduce the number of lead drug candidates down to the most likely to succeed in later human clinical trials. However, only 1 in 10 drug candidates that emerge from preclinical studies will succeed and become an approved therapeutic. Lack of efficacy or undetected toxicity represents roughly 75% of the causes for these failures, despite these parameters being the primary exclusion criteria in preclinical studies. Recently, advances in both biology and engineering have created new tools for constructing new preclinical models. These models can complement those used in current preclinical studies by helping to create more realistic representations of human tissues in vitro and in vivo. In this review, we describe current preclinical models to identify their value and limitations and then discuss select areas of research where improvements in preclinical models are particularly needed to advance drug development. Following this, we discuss design considerations for constructing preclinical models and then highlight recent advances in these efforts. Taken together, we aim to review the advances as of 2020 surrounding the prospect of biological and engineering tools for adding enhanced biological relevance to preclinical studies to aid in the challenges of failed drug candidates and the burden this poses on the drug development enterprise and thus healthcare.
Collapse
Affiliation(s)
- Andrew S Khalil
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02115, USA; Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02115, USA.
| |
Collapse
|
5
|
Inselman A, Liu F, Wang C, Shi Q, Pang L, Mattes W, White M, Lyn-Cook B, Rosas-Hernandez H, Cuevas E, Lantz S, Imam S, Ali S, Petibone DM, Shemansky JM, Xiong R, Wang Y, Tripathi P, Cao X, Heflich RH, Slikker W. Dr. Daniel Acosta and In Vitro toxicology at the U.S. Food and Drug Administration's National Center for Toxicological Research. Toxicol In Vitro 2019; 64:104471. [PMID: 31628011 DOI: 10.1016/j.tiv.2019.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 03/01/2019] [Accepted: 03/04/2019] [Indexed: 10/25/2022]
Abstract
For the past five years, Dr. Daniel Acosta has served as the Deputy Director of Research at the National Center for Toxicological Research (NCTR), a principle research laboratory of the U.S. Food and Drug Administration (FDA). Over his career at NCTR, Dr. Acosta has had a major impact on developing and promoting the use of in vitro assays in regulatory toxicity and product safety assessments. As Dr. Acosta nears his retirement we have dedicated this paper to his many accomplishments at the NCTR. Described within this paper are some of the in vitro studies that have been conducted under Dr. Acosta's leadership. These studies include toxicological assessments involving developmental effects, and the development and application of in vitro reproductive, heart, liver, neurological and airway cell and tissue models.
Collapse
Affiliation(s)
- Amy Inselman
- Division of Systems Biology, NCTR, FDA, Jefferson, AR 72079, USA
| | - Fang Liu
- Division of Neurotoxicology, NCTR, FDA, Jefferson, AR 72079, USA
| | - Cheng Wang
- Division of Neurotoxicology, NCTR, FDA, Jefferson, AR 72079, USA
| | - Qiang Shi
- Division of Systems Biology, NCTR, FDA, Jefferson, AR 72079, USA
| | - Li Pang
- Division of Systems Biology, NCTR, FDA, Jefferson, AR 72079, USA
| | - William Mattes
- Division of Systems Biology, NCTR, FDA, Jefferson, AR 72079, USA
| | - Matthew White
- Arkansas College of Osteopathic Medicine, Fort Smith, AR 72916, USA
| | - Beverly Lyn-Cook
- Division of Biochemical Toxicology, NCTR, FDA, Jefferson, AR 72079, USA
| | | | - Elvis Cuevas
- Division of Neurotoxicology, NCTR, FDA, Jefferson, AR 72079, USA
| | - Susan Lantz
- Division of Neurotoxicology, NCTR, FDA, Jefferson, AR 72079, USA
| | - Syed Imam
- Division of Neurotoxicology, NCTR, FDA, Jefferson, AR 72079, USA
| | - Syed Ali
- Division of Neurotoxicology, NCTR, FDA, Jefferson, AR 72079, USA
| | - Dayton M Petibone
- Division of Genetic and Molecular Toxicology, NCTR, FDA, Jefferson, AR 72079, USA
| | - Jennifer M Shemansky
- Division of Genetic and Molecular Toxicology, NCTR, FDA, Jefferson, AR 72079, USA
| | - Rui Xiong
- Division of Genetic and Molecular Toxicology, NCTR, FDA, Jefferson, AR 72079, USA
| | - Yiying Wang
- Division of Genetic and Molecular Toxicology, NCTR, FDA, Jefferson, AR 72079, USA
| | - Priya Tripathi
- Division of Genetic and Molecular Toxicology, NCTR, FDA, Jefferson, AR 72079, USA
| | - Xuefei Cao
- Division of Genetic and Molecular Toxicology, NCTR, FDA, Jefferson, AR 72079, USA
| | - Robert H Heflich
- Division of Genetic and Molecular Toxicology, NCTR, FDA, Jefferson, AR 72079, USA
| | | |
Collapse
|
6
|
Abstract
The poor pharmacokinetic parameters and low solubility of many anticancer therapeutics have warranted the use of drug-delivery systems such as liposomes. Overcoming some drawbacks of the conventional liposomes, targeted liposomal delivery by longer circulation time by addition of poly(ethylene glycol) to the liposomal surface and further adding specific ligands to achieve ligand selective retention and uptake has been introduced. PEGylated liposomes are the only second-generation liposomal formulations in clinical use and are now being challenged with the allergenic response they pose even in the treatment of naive patients. This article will review the challenges and hindrances in the use of long circulating liposomes and explore the opportunities to overcome this issue.
Collapse
|
7
|
Coecke S, Ahr H, Blaauboer BJ, Bremer S, Casati S, Castell J, Combes R, Corvi R, Crespi CL, Cunningham ML, Elaut G, Eletti B, Freidig A, Gennari A, Ghersi-Egea JF, Guillouzo A, Hartung T, Hoet P, Ingelman-Sundberg M, Munn S, Janssens W, Ladstetter B, Leahy D, Long A, Meneguz A, Monshouwer M, Morath S, Nagelkerke F, Pelkonen O, Ponti J, Prieto P, Richert L, Sabbioni E, Schaack B, Steiling W, Testai E, Vericat JA, Worth A. Metabolism: A Bottleneck in In Vitro Toxicological Test Development. Altern Lab Anim 2019; 34:49-84. [PMID: 16522150 DOI: 10.1177/026119290603400113] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Sandra Coecke
- ECVAM, Institute for Health and Consumer Protection, European Commission Joint Research Centre, Ispra, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Chebekoue SF, Krishnan K. A framework for application of quantitative property-property relationships (QPPRs) in physiologically based pharmacokinetic (PBPK) models for high-throughput prediction of internal dose of inhaled organic chemicals. CHEMOSPHERE 2019; 215:634-646. [PMID: 30347358 DOI: 10.1016/j.chemosphere.2018.10.041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 10/03/2018] [Accepted: 10/06/2018] [Indexed: 06/08/2023]
Abstract
New generation of toxicological tests and assessment strategies require validated toxicokinetic data or models that are lacking for most chemicals. This study aimed at developing a quantitative property-property relationship (QPPR)-based human physiologically based pharmacokinetic (PBPK) modeling framework for high-throughput predictions of inhalation toxicokinetics of organic chemicals. A PBPK model was parameterized with QPPR-derived values for hepatic clearance (CLh) and partition coefficients (P) [blood:air (Pba) and tissue:air (Pta) and tissue:blood (Ptb)]. The model was initially applied to an evaluation dataset of 40 organic chemicals in the applicability domain, and then to an expanded dataset of 249 organic chemicals from diverse chemical classes. 'Batch' analyses were performed for rapid assessments of hundreds of chemicals. The simulations of inhalation toxicokinetics following an 8-h exposure to 1 ppm of each chemical were successful. The mean ratios of their predicted-to-experimental values were within a factor of 1.36-2.36 for Ptb and 1.18 for CLh, for 80% of the chemicals in the evaluation dataset. The predicted 24-h area under the venous blood concentration-time curve (AUC24) values were within the predicted envelopes obtained while using experimental values of Pba and considering either no or maximal hepatic extraction. The reliability analysis (based on combined sensitivity and uncertainty analyses) indicated that AUC24 predictions for 55% of the expanded dataset were moderately to highly reliable, with 46% exhibiting highly reliable values. Overall, the modeling framework suggests that molecular structure and chemical properties can together be effectively used to obtain first-cut estimates of the toxicokinetics of data-poor organic chemicals for screening and prioritization purposes.
Collapse
Affiliation(s)
- Sandrine F Chebekoue
- École de Santé Publique de l'Université de Montréal (ESPUM), Montréal, Québec, Canada.
| | - Kannan Krishnan
- École de Santé Publique de l'Université de Montréal (ESPUM), Montréal, Québec, Canada; Institut de recherche Robert-Sauvé en santé et en sécurité du travail (IRSST), Montréal, Québec, Canada.
| |
Collapse
|
9
|
Bradai M, Han J, Omri AE, Funamizu N, Sayadi S, Isoda H. Effect of linear alkylbenzene sulfonate (LAS) on human intestinal Caco-2 cells at non cytotoxic concentrations. Cytotechnology 2016; 68:1267-75. [PMID: 25999174 PMCID: PMC4960175 DOI: 10.1007/s10616-015-9887-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 05/08/2015] [Indexed: 01/07/2023] Open
Abstract
Linear alkylbenzene sulfonate (LAS) is a cytotoxic synthetic anionic surfactant widely present in the environment due to its large-scale production and intensive use in the detergency field. In this study, we investigated the effect of LAS (CAS No. 25155-30-0) at non cytotoxic concentrations on human intestinal Caco-2 cells using different in vitro bioassays. As results, LAS increased Caco-2 cell proliferation at concentrations ranging from 1 to 15 ppm, more significantly for shorter exposure time (24 h), confirmed using flow cytometry and trypan blue exclusion methods. Moreover, proteomics analysis revealed that this effect was associated with an over-expression of elongation factor 2 and dipeptidyl peptidase 3, and a down-regulation of 14-3-3 protein theta, confirmed at mRNA level using real-time PCR. These findings suggest that LAS at non cytotoxic concentrations, similar to those observed at wastewater treatment plants outlets, increases the growth rate of colon cancer cells, raising thereby its tumor promotion effect potential.
Collapse
Affiliation(s)
- Mohamed Bradai
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, 305-8572, Japan
| | - Junkyu Han
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, 305-8572, Japan
- Alliance of Research on North Africa, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, 305-8572, Japan
| | - Abdelfatteh El Omri
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, 305-8572, Japan
| | - Naoyuki Funamizu
- Graduate School of Engineering, Hokkaido University, Sapporo, 060-0808, Japan
| | - Sami Sayadi
- Laboratory of Environmental Bioprocesses, Biotechnology Center of Sfax, Sfax, PB 3018, Tunisia
| | - Hiroko Isoda
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, 305-8572, Japan.
- Alliance of Research on North Africa, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, 305-8572, Japan.
| |
Collapse
|
10
|
Quantin P, Thélu A, Catoire S, Ficheux H. Perspectives and strategies of alternative methods used in the risk assessment of personal care products. ANNALES PHARMACEUTIQUES FRANÇAISES 2015; 73:422-35. [DOI: 10.1016/j.pharma.2015.06.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 06/02/2015] [Accepted: 06/11/2015] [Indexed: 10/23/2022]
|
11
|
Moulin F, Flint O. In VitroModels for the Prediction of Drug-Induced Liver Injury in Lead Discovery. ACTA ACUST UNITED AC 2015. [DOI: 10.1002/9783527673643.ch07] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
12
|
Bradai M, Han J, El Omri A, Funamizu N, Sayadi S, Isoda H. Cytotoxic effect of linear alkylbenzene sulfonate on human intestinal Caco-2 cells: associated biomarkers for risk assessment. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2014; 21:10840-10851. [PMID: 24878558 DOI: 10.1007/s11356-014-3074-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 05/20/2014] [Indexed: 06/03/2023]
Abstract
Linear alkylbenzene sulfonate (LAS) is a synthetic anionic surfactant widely present in the environment due to its intensive production and use in the detergency field. Admitting that current procedure of risk assessment has limits in providing realistic risk assessment data and predicting the cumulative effect of the toxicant mixtures, the incorporation of information regarding the mode of action and cell response mechanism seems to be a potential solution to overcome these limits. In this regard, we investigated in this study the LAS cytotoxicity on human intestinal Caco-2 cells, trying to unveil the protein actors implicated in the cell response using proteomics approach in order to give a better understanding of the toxicological effect and allow the identification of appropriate biomarkers reflecting the mode of action associated with LAS. As results, we demonstrated that LAS induces a time- and dose-dependent cytotoxicity in Caco-2 cells accompanied by an induction of oxidative stress followed by an excessive increase of intracellular calcium level. Proteomics approach helped in discovering three informative biomarkers of effect associated with LAS cytotoxic effect, reported for the first time: calreticulin, thioredoxin, and heat shock cognate 71 (HSP7C), confirmed by real-time PCR and western blot analysis. These biomarkers could serve for more reliable future risk assessment studies that consider the toxicants mode of action in order to help in the prediction of potential cumulative effects of environmentally coexisting contaminants.
Collapse
Affiliation(s)
- Mohamed Bradai
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, 305-8572, Japan
| | | | | | | | | | | |
Collapse
|
13
|
Pinto M, Costa PM, Louro H, Costa MH, Lavinha J, Caeiro S, Silva MJ. Determining oxidative and non-oxidative genotoxic effects driven by estuarine sediment contaminants on a human hepatoma cell line. THE SCIENCE OF THE TOTAL ENVIRONMENT 2014; 478:25-35. [PMID: 24530582 DOI: 10.1016/j.scitotenv.2014.01.084] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 01/03/2014] [Accepted: 01/22/2014] [Indexed: 06/03/2023]
Abstract
Estuarine sediments may be reservoirs of hydrophilic and hydrophobic pollutants, many of which are acknowledged genotoxicants, pro-mutagens and even potential carcinogens for humans. Still, studies aiming at narrowing the gap between ecological and human health risk of sediment-bound contaminant mixtures are scarce. Taking an impacted estuary as a case study (the Sado, SW Portugal), HepG2 (human hepatoma) cells were exposed in vitro for 48 h to extracts of sediments collected from two areas (urban/industrial and Triverine/agricultural), both contaminated by distinct mixtures of organic and inorganic toxicants, among which are found priority mutagens such as benzo[a]pyrene. Comparatively to a control test, extracts of sediments from both impacted areas produced deleterious effects in a dose-response manner. However, sediment extracts from the industrial area caused lower replication index plus higher cytotoxicity and genotoxicity (concerning total DNA strand breakage and clastogenesis), with emphasis on micronucleus induction. On the other hand, extracts from the rural area induced the highest oxidative damage to DNA, as revealed by the FPG (formamidopyrimidine-DNA glycosylase) enzyme in the Comet assay. Although the estuary, on its whole, has been classified as moderately contaminated, the results suggest that the sediments from the industrial area are significantly genotoxic and, furthermore, elicit permanent chromosome damage, thus potentially being more mutagenic than those from the rural area. The results are consistent with contamination by pro-mutagens like polycyclic aromatic hydrocarbons (PAHs), potentiated by metals. The sediments from the agriculture-influenced area likely owe their genotoxic effects to metals and other toxicants, probably pesticides and fertilizers, and able to induce reactive oxygen species without the formation of DNA strand breakage. The findings suggest that the mixtures of contaminants present in the assayed sediments are genotoxic to HepG2 cells, ultimately providing a useful approach to hazard identification and an effective line-of-evidence in the environmental monitoring of anthropogenically-impacted coastal ecosystems.
Collapse
Affiliation(s)
- M Pinto
- Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge, I.P., Av. Padre Cruz, 1649-016 Lisboa, Portugal
| | - P M Costa
- Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge, I.P., Av. Padre Cruz, 1649-016 Lisboa, Portugal; IMAR - Instituto do Mar, Departamento de Ciências e Engenharia do Ambiente, Faculdade de Ciências e Tecnologia da Universidade Nova de Lisboa, 2829-516 Caparica, Portugal.
| | - H Louro
- Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge, I.P., Av. Padre Cruz, 1649-016 Lisboa, Portugal
| | - M H Costa
- IMAR - Instituto do Mar, Departamento de Ciências e Engenharia do Ambiente, Faculdade de Ciências e Tecnologia da Universidade Nova de Lisboa, 2829-516 Caparica, Portugal
| | - J Lavinha
- Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge, I.P., Av. Padre Cruz, 1649-016 Lisboa, Portugal
| | - S Caeiro
- IMAR - Instituto do Mar, Departamento de Ciências e Engenharia do Ambiente, Faculdade de Ciências e Tecnologia da Universidade Nova de Lisboa, 2829-516 Caparica, Portugal; Departamento de Ciências e Tecnologia, Universidade Aberta, Rua da Escola Politécnica, 141, 1269-001 Lisboa, Portugal; CENSE - Centre for Environmental and Sustainability Research, Departamento de Ciências e Engenharia do Ambiente, Faculdade de Ciências e Tecnologia da Universidade Nova de Lisboa, 2829-516 Caparica, Portugal
| | - M J Silva
- Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge, I.P., Av. Padre Cruz, 1649-016 Lisboa, Portugal
| |
Collapse
|
14
|
Defining and characterizing drug/compound function. Biochem Pharmacol 2014; 87:40-63. [DOI: 10.1016/j.bcp.2013.07.033] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 07/22/2013] [Indexed: 12/25/2022]
|
15
|
Elaut G, Török G, Papeleu P, Vanhaecke T, Laus G, Tourwé D, Rogiers V. Rat hepatocyte suspensions as a suitable in vitro model for studying the biotransformation of histone deacetylase inhibitors. Altern Lab Anim 2013; 32 Suppl 1A:105-12. [PMID: 23577441 DOI: 10.1177/026119290403201s16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
This paper focuses on the use of liver-derived in vitro systems for biotransformation studies during early drug development, as exemplified by the two molecules recently studied in our laboratory: Trichostatin A (TSA) and its structural analogue 5-(4-dimethylaminobenzoyl)aminovaleric acid hydroxamide (4-Me2N-BAVAH). Phase I biotransformation of TSA, a histone deacetylase inhibitor with promising antifibrotic and antitumoural properties, was investigated in liver microsomal (rat and human) and in hepatocyte (rat) suspensions. Within 40 minutes, 50 microM of TSA was completely metabolised by 2 x 10(6) hepatocytes/ml. Reduction of the hydroxamic acid function to its corresponding amide and N-demethylation were the two major phase I biotransformation pathways, while hydrolysis products of TSA were minor metabolites. Lower concentrations of TSA (5 microM and 25 microM) were N-demethylated faster. Liver microsomes, however, metabolised TSA incompletely with the formation of two major metabolites, N-mono- and N-didemethylated TSA. Unlike TSA, 4-Me2N-BAVAH (50 microM) could still be detected after 3 hours of incubation with 2 x 10(6) rat hepatocytes/ml suspension. Hydrolysis and reduction of the hydroxamic acid function to its corresponding acid and amide, respectively, were shown to be the major phase I biotransformation pathways. Lower concentrations of 4-Me2N-BAVAH were hydrolysed more readily. 4-Me2N-BAVAH and its metabolites were less subjected to N-demethylation than TSA.
Collapse
Affiliation(s)
- Greetje Elaut
- Department of Toxicology, Vrije Universiteit Brussel, Brussels, Belgium.
| | | | | | | | | | | | | |
Collapse
|
16
|
Physiologically-based pharmacokinetic (PBPK) models in toxicity testing and risk assessment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 745:76-95. [PMID: 22437814 DOI: 10.1007/978-1-4614-3055-1_6] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Physiologically-based pharmacokinetic (PBPK) modeling offers a scientifically-sound framework for integrating mechanistic data on absorption, distribution, metabolism and elimination to predict the time-course of parent chemical, metabolite(s) or biomarkers in the exposed organism. A major advantage of PBPK models is their ability to forecast the impact of specific mechanistic processes and determinants on the tissue dose. In this regard, they facilitate integration of data obtained with in vitro and in silico methods, for making predictions of the tissue dosimetry in the whole animal, thus reducing and/or refining the use of animals in pharmacokinetic and toxicity studies. This chapter presents the principles and practice of PBPK modeling, as well as the application of these models in toxicity testing and health risk assessments.
Collapse
|
17
|
Shah AK, Agnihotri SA. Recent advances and novel strategies in pre-clinical formulation development: An overview. J Control Release 2011; 156:281-96. [DOI: 10.1016/j.jconrel.2011.07.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2011] [Accepted: 05/15/2011] [Indexed: 10/18/2022]
|
18
|
Moeller TA, Shukla SJ, Xia M. Assessment of compound hepatotoxicity using human plateable cryopreserved hepatocytes in a 1536-well-plate format. Assay Drug Dev Technol 2011; 10:78-87. [PMID: 22053711 DOI: 10.1089/adt.2010.0365] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Hepatotoxicity is a major concern for both drug development and toxicological evaluation of environmental chemicals. The assessment of compound-induced hepatotoxicity has traditionally relied on in vivo testing; however, it is being replaced by human in vitro models due to an emphasis on the reduction of animal testing and species-specific differences. Since most cell lines and hybridomas lack the full complement of enzymes at physiological levels found in the liver, primary hepatocytes are the gold standard to study liver toxicities in vitro due to the retention of most of their in vivo activities. Here, we optimized a cell viability assay using plateable cryopreserved human hepatocytes in a 1536-well-plate format. The assay was validated by deriving inhibitory concentration at 50% values for 12 known compounds, including tamoxifen, staurosporine, and phenylmercuric acetate, with regard to hepatotoxicity and general cytotoxicity using multiple hepatocyte donors. The assay performed well, and the cytotoxicity of these compounds was confirmed in comparison to HepG2 cells. This is the first study to report the reliability of using plateable cryopreserved human hepatocytes for cytotoxicity studies in a 1536-well-plate format. These results suggest that plateable cryopreserved human hepatocytes can be scaled up for screening a large compound library and may be amenable to other hepatocytic assays such as metabolic or drug safety studies.
Collapse
|
19
|
Myers JN, Rekhadevi PV, Ramesh A. Comparative evaluation of different cell lysis and extraction methods for studying benzo(a)pyrene metabolism in HT-29 colon cancer cell cultures. Cell Physiol Biochem 2011; 28:209-18. [PMID: 21865728 DOI: 10.1159/000331732] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2011] [Indexed: 01/26/2023] Open
Abstract
Lysis and extraction of cells are essential sample processing steps for investigations pertaining to metabolism of xenobiotics in cell culture studies. Of particular importance to these procedures are maintaining high lysis efficiency and analyte integrity as they influence the qualitative and quantitative distribution of drug and toxicant metabolites in the intra- and extracellular milieus. In this study we have compared the efficiency of different procedures viz. homogenization, sonication, bead beating, and molecular grinding resin treatment for disruption of HT-29 colon cells exposed to benzo(a)pyrene (BaP), a polycyclic aromatic hydrocarbon (PAH) compound and a suspected colon carcinogen. Also, we have evaluated the efficiency of various procedures for extracting BaP parent compound/metabolites from colon cells and culture media prior to High Performance Liquid Chromatography (HPLC) analyses. The extraction procedures include solid phase extraction, solid-supported liquid- liquid extraction, liquid-liquid extraction, and homogeneous liquid- liquid extraction. Our findings showed that bead-beating in combination with detergent treatment of cell pellet coupled with liquid-liquid extraction yielded greater concentrations of BaP metabolites compared to the other methods employed. Our method optimization strategy revealed that disruption of HT-29 colon cells by a combination of mechanical and chemical lysis followed by liquid-liquid extraction is efficient and robust enough for analyzing BaP metabolites from cell culture studies.
Collapse
Affiliation(s)
- Jeremy N Myers
- Department of Biochemistry & Cancer Biology, Meharry Medical College, Nashville, USA
| | | | | |
Collapse
|
20
|
Schroeder K, Bremm K, Alépée N, Bessems J, Blaauboer B, Boehn S, Burek C, Coecke S, Gombau L, Hewitt N, Heylings J, Huwyler J, Jaeger M, Jagelavicius M, Jarrett N, Ketelslegers H, Kocina I, Koester J, Kreysa J, Note R, Poth A, Radtke M, Rogiers V, Scheel J, Schulz T, Steinkellner H, Toeroek M, Whelan M, Winkler P, Diembeck W. Report from the EPAA workshop: In vitro ADME in safety testing used by EPAA industry sectors. Toxicol In Vitro 2011; 25:589-604. [DOI: 10.1016/j.tiv.2010.12.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2010] [Revised: 11/05/2010] [Accepted: 12/06/2010] [Indexed: 10/18/2022]
|
21
|
Switalla S, Lauenstein L, Prenzler F, Knothe S, Förster C, Fieguth HG, Pfennig O, Schaumann F, Martin C, Guzman CA, Ebensen T, Müller M, Hohlfeld JM, Krug N, Braun A, Sewald K. Natural innate cytokine response to immunomodulators and adjuvants in human precision-cut lung slices. Toxicol Appl Pharmacol 2010; 246:107-15. [PMID: 20434477 DOI: 10.1016/j.taap.2010.04.010] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2009] [Revised: 04/13/2010] [Accepted: 04/14/2010] [Indexed: 11/16/2022]
Abstract
Prediction of lung innate immune responses is critical for developing new drugs. Well-established immune modulators like lipopolysaccharides (LPS) can elicit a wide range of immunological effects. They are involved in acute lung diseases such as infections or chronic airway diseases such as COPD. LPS has a strong adjuvant activity, but its pyrogenicity has precluded therapeutic use. The bacterial lipopeptide MALP-2 and its synthetic derivative BPPcysMPEG are better tolerated. We have compared the effects of LPS and BPPcysMPEG on the innate immune response in human precision-cut lung slices. Cytokine responses were quantified by ELISA, Luminex, and Meso Scale Discovery technology. The initial response to LPS and BPPcysMPEG was marked by coordinated and significant release of the mediators IL-1β, MIP-1β, and IL-10 in viable PCLS. Stimulation of lung tissue with BPPcysMPEG, however, induced a differential response. While LPS upregulated IFN-γ, BPPcysMPEG did not. This traces back to their signaling pathways via TLR4 and TLR2/6. The calculated exposure doses selected for LPS covered ranges occurring in clinical studies with human beings. Correlation of obtained data with data from human BAL fluid after segmental provocation with endotoxin showed highly comparable effects, resulting in a coefficient of correlation >0.9. Furthermore, we were interested in modulating the response to LPS. Using dexamethasone as an immunosuppressive drug for anti-inflammatory therapy, we found a significant reduction of GM-CSF, IL-1β, and IFN-γ. The PCLS-model offers the unique opportunity to test the efficacy and toxicity of biological agents intended for use by inhalation in a complex setting in humans.
Collapse
Affiliation(s)
- S Switalla
- Fraunhofer Institute for Toxicology and Experimental Medicine, Division of Immunology, Allergy and Airway Research, Nikolai-Fuchs-Str. 1, D-30625 Hannover, Germany
| | - L Lauenstein
- Fraunhofer Institute for Toxicology and Experimental Medicine, Division of Immunology, Allergy and Airway Research, Nikolai-Fuchs-Str. 1, D-30625 Hannover, Germany
| | - F Prenzler
- Fraunhofer Institute for Toxicology and Experimental Medicine, Division of Immunology, Allergy and Airway Research, Nikolai-Fuchs-Str. 1, D-30625 Hannover, Germany
| | - S Knothe
- Fraunhofer Institute for Toxicology and Experimental Medicine, Division of Immunology, Allergy and Airway Research, Nikolai-Fuchs-Str. 1, D-30625 Hannover, Germany
| | - C Förster
- Klinikum Region Hannover (KRH), Hannover, Germany
| | - H-G Fieguth
- Klinikum Region Hannover (KRH), Hannover, Germany
| | - O Pfennig
- Klinikum Region Hannover (KRH), Hannover, Germany
| | - F Schaumann
- Fraunhofer Institute for Toxicology and Experimental Medicine, Division of Immunology, Allergy and Airway Research, Nikolai-Fuchs-Str. 1, D-30625 Hannover, Germany
| | - C Martin
- Institute of Pharmacology and Toxicology, RWTH Aachen, Wendlingweg 2, 52074 Aachen, Germany
| | - C A Guzman
- Helmholtz Centre for Infection Research, Department of Vaccinology and Applied Microbiology, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - T Ebensen
- Helmholtz Centre for Infection Research, Department of Vaccinology and Applied Microbiology, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - M Müller
- Fraunhofer Institute for Toxicology and Experimental Medicine, Division of Immunology, Allergy and Airway Research, Nikolai-Fuchs-Str. 1, D-30625 Hannover, Germany
| | - J M Hohlfeld
- Fraunhofer Institute for Toxicology and Experimental Medicine, Division of Immunology, Allergy and Airway Research, Nikolai-Fuchs-Str. 1, D-30625 Hannover, Germany
| | - N Krug
- Fraunhofer Institute for Toxicology and Experimental Medicine, Division of Immunology, Allergy and Airway Research, Nikolai-Fuchs-Str. 1, D-30625 Hannover, Germany
| | - A Braun
- Fraunhofer Institute for Toxicology and Experimental Medicine, Division of Immunology, Allergy and Airway Research, Nikolai-Fuchs-Str. 1, D-30625 Hannover, Germany
| | - K Sewald
- Fraunhofer Institute for Toxicology and Experimental Medicine, Division of Immunology, Allergy and Airway Research, Nikolai-Fuchs-Str. 1, D-30625 Hannover, Germany
| |
Collapse
|
22
|
Fagerholm U. Prediction of human pharmacokinetics—evaluation of methods for prediction of hepatic metabolic clearance. J Pharm Pharmacol 2010; 59:803-28. [PMID: 17637173 DOI: 10.1211/jpp.59.6.0007] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Abstract
Methods for prediction of hepatic clearance (CLH) in man have been evaluated. A physiologically-based in-vitro to in-vivo (PB-IVIV) method with human unbound fraction in blood (fu,bl) and hepatocyte intrinsic clearance (CLint)-data has a good rationale and appears to give the best predictions (maximum ∼2-fold errors; < 25% errors for half of CL-predictions; appropriate ranking). Inclusion of an empirical scaling factor is, however, needed, and reasons include the use of cryopreserved hepatocytes with low activity, and inappropriate CLint- and fu,bl-estimation methods. Thus, an improvement of this methodology is possible and required. Neglect of fu,bl or incorporation of incubation binding does not seem appropriate. When microsome CLint-data are used with this approach, the CLH is underpredicted by 5- to 9-fold on average, and a 106-fold underprediction (attrition potential) has been observed. The poor performance could probably be related to permeation, binding and low metabolic activity. Inclusion of scaling factors and neglect of fu,bl for basic and neutral compounds improve microsome predictions. The performance is, however, still not satisfactory. Allometry incorrectly assumes that the determinants for CLH relate to body weight and overpredicts human liver blood flow rate. Consequently, allometric methods have poor predictability. Simple allometry has an average overprediction potential, > 2-fold errors for ∼1/3 of predictions, and 140-fold underprediction to 5800-fold overprediction (potential safety risk) range. In-silico methodologies are available, but these need further development. Acceptable prediction errors for compounds with low and high CLH should be ∼50 and ∼10%, respectively. In conclusion, it is recommended that PB-IVIV with human hepatocyte CLint and fu,bl is applied and improved, limits for acceptable errors are decreased, and that animal CLH-studies and allometry are avoided.
Collapse
Affiliation(s)
- Urban Fagerholm
- Clinical Pharmacology, AstraZeneca R&D Södertälje, S-151 85 Södertälje, Sweden.
| |
Collapse
|
23
|
Characterizing uncertainty and population variability in the toxicokinetics of trichloroethylene and metabolites in mice, rats, and humans using an updated database, physiologically based pharmacokinetic (PBPK) model, and Bayesian approach. Toxicol Appl Pharmacol 2009; 241:36-60. [DOI: 10.1016/j.taap.2009.07.032] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2009] [Revised: 07/21/2009] [Accepted: 07/27/2009] [Indexed: 11/24/2022]
|
24
|
Dash A, Inman W, Hoffmaster K, Sevidal S, Kelly J, Obach RS, Griffith LG, Tannenbaum SR. Liver tissue engineering in the evaluation of drug safety. Expert Opin Drug Metab Toxicol 2009; 5:1159-74. [PMID: 19637986 PMCID: PMC4110978 DOI: 10.1517/17425250903160664] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Assessment of drug-liver interactions is an integral part of predicting the safety profile of new drugs. Existing model systems range from in vitro cell culture models to FDA-mandated animal tests. Data from these models often fail, however, to predict human liver toxicity, resulting in costly failures of clinical trials. In vitro screens based on cultured hepatocytes are now commonly used in early stages of development, but many toxic responses in vivo seem to be mediated by a complex interplay among several different cell types. We discuss some of the evolving trends in liver cell culture systems applied to drug safety assessment and describe an experimental model that captures complex liver physiology through incorporation of heterotypic cell-cell interactions, 3D architecture and perfused flow. We demonstrate how heterotypic interactions in this system can be manipulated to recreate an inflammatory environment and apply the model to test compounds that potentially exhibit idiosyncratic drug toxicity. Finally, we provide a perspective on how the range of existing and emerging in vitro liver culture approaches, from simple to complex, might serve needs across the range of stages in drug discovery and development, including applications in molecular therapeutics.
Collapse
Affiliation(s)
- Ajit Dash
- Underwood-Prescott Professor of Toxicology and Chemistry, Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Office 56-731A, Cambridge, MA 02139, USA
| | - Walker Inman
- Underwood-Prescott Professor of Toxicology and Chemistry, Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Office 56-731A, Cambridge, MA 02139, USA
| | - Keith Hoffmaster
- Novartis Institute of Biomedical Research, 350 Massachusetts Avenue, Cambridge, Massachusetts, MA 02139, USA
| | - Samantha Sevidal
- Pfizer Research Technology Center, Cambridge, Massachusetts, MA 02139, USA
| | - Joan Kelly
- Pfizer Research Technology Center, Cambridge, Massachusetts, MA 02139, USA
| | - R Scott Obach
- Pfizer Research Technology Center, Cambridge, Massachusetts, MA 02139, USA
| | - Linda G Griffith
- Underwood-Prescott Professor of Toxicology and Chemistry, Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Office 56-731A, Cambridge, MA 02139, USA
| | - Steven R Tannenbaum
- Underwood-Prescott Professor of Toxicology and Chemistry, Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Office 56-731A, Cambridge, MA 02139, USA
| |
Collapse
|
25
|
Kim S, Dere E, Burgoon LD, Chang CC, Zacharewski TR. Comparative analysis of AhR-mediated TCDD-elicited gene expression in human liver adult stem cells. Toxicol Sci 2009; 112:229-44. [PMID: 19684285 DOI: 10.1093/toxsci/kfp189] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Time course and dose-response studies were conducted in HL1-1 cells, a human liver cell line with stem cell-like characteristics, to assess the differential gene expression elicited by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) compared with other established models. Cells were treated with 0.001, 0.01, 0.1, 1, 10, or 100nM TCDD or dimethyl sulfoxide vehicle control for 12 h for the dose-response study, or with 10nM TCDD or vehicle for 1, 2, 4, 8, 12, 24, or 48 h for the time course study. Elicited changes were monitored using a human cDNA microarray with 6995 represented genes. Empirical Bayes analysis identified 144 genes differentially expressed at one or more time points following treatment. Most genes exhibited dose-dependent responses including CYP1A1, CYP1B1, ALDH1A3, and SLC7A5 genes. Comparative analysis of HL1-1 differential gene expression to human HepG2 data identified 74 genes with comparable temporal expression profiles including 12 putative primary responses. HL1-1-specific changes were related to lipid metabolism and immune responses, consistent with effects elicited in vivo. Furthermore, comparative analysis of HL1-1 cells with mouse Hepa1c1c7 hepatoma cell lines and C57BL/6 hepatic tissue identified 18 and 32 commonly regulated orthologous genes, respectively, with functions associated with signal transduction, transcriptional regulation, metabolism and transport. Although some common pathways are affected, the results suggest that TCDD elicits species- and model-specific gene expression profiles.
Collapse
Affiliation(s)
- Suntae Kim
- Department of Biochemistry & Molecular Biology, 501 Biochemistry Building, Wilson Road, East Lansing, MI 48824-1319, USA
| | | | | | | | | |
Collapse
|
26
|
Fagerholm U. Presentation of a modified dispersion model (MDM) for hepatic drug extraction and a new methodology for the prediction of the rate-limiting step in hepatic metabolic clearance. Xenobiotica 2009; 39:57-71. [DOI: 10.1080/00498250802562652] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
27
|
Kramer MA, Tracy TS. Studying cytochrome P450 kinetics in drug metabolism. Expert Opin Drug Metab Toxicol 2008; 4:591-603. [PMID: 18484917 DOI: 10.1517/17425255.4.5.591] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Determination of cytochrome P450 enzyme-mediated kinetics in vitro can be useful for predicting drug dosing and clearance in humans. Expressed P450s, human liver microsomes, human hepatocytes (both fresh and cryopreserved), and human liver slices are used to estimate K(m) and V(max) values for determination of intrinsic clearance of the drug for scale-up to predict in vivo clearance. OBJECTIVE To describe the advantages and disadvantages of the various in vitro systems used to estimate kinetic parameters for disposition of drugs and the various kinetic profiles that can be observed. METHODS A review of the literature was conducted to evaluate the utility of the various in vitro preparations, the methods for determining kinetic parameters and the types of kinetic profiles that may be observed. RESULTS/CONCLUSIONS The choice of in vitro system for determining kinetic parameters will depend on the objective of the studies, as each system has advantages and disadvantages. Kinetic parameter determinations must be carefully assessed to assure that the correct kinetic model is applied and the most accurate kinetic parameters are determined.
Collapse
Affiliation(s)
- Melissa A Kramer
- University of Minnesota, College of Pharmacy, Department of Experimental and Clinical Pharmacology, 7-115B Weaver-Densford Hall, 308 Harvard Street SE, Minneapolis, MN 55455, USA
| | | |
Collapse
|
28
|
Metabolism Comparative Cytotoxicity Assay (MCCA) and Cytotoxic Metabolic Pathway Identification Assay (CMPIA) with cryopreserved human hepatocytes for the evaluation of metabolism-based cytotoxicity in vitro: proof-of-concept study with aflatoxin B1. Chem Biol Interact 2008; 179:4-8. [PMID: 18950609 DOI: 10.1016/j.cbi.2008.09.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2008] [Revised: 09/16/2008] [Accepted: 09/17/2008] [Indexed: 11/23/2022]
Abstract
Recently, we have improved the cryopreservation procedures for human hepatocytes, leading to cells that can be cultured after thawing ("plateable" cryopreserved human hepatocytes). The ability to culture cryopreserved human hepatocytes allows application of the cells for prolonged incubations such as long-term (days) metabolism studies, enzyme induction studies, and cytotoxicity studies. We report here the application of the plateable cryopreserved human hepatocytes to evaluate the relationship between xenobiotic metabolism and toxicity. Two assays were developed: The Metabolism Comparative Cytotoxicity Assay (MCCA) and the Cytotoxic Metabolic Pathway Identification Assay (CMPIA). The MCCA was designed for the initial identification of the role of metabolism in cytotoxicity by comparing the cytotoxic potential of a toxicant in a metabolically competent (primary human hepatocytes) and a metabolically incompetent (Chinese hamster ovary (CHO)) cell type, as well as the evaluation of the role of P450 metabolism by comparing the cytotoxicity of the toxicant in question in human hepatocytes in the presence and absence of a nonspecific, irreversible P450 inhibitor, 1-aminobenzotriazole (ABT). The CMPIA was designed for the identification of the P450 isoforms involved in metabolic activation via the evaluation of the cytotoxicity of the toxicant in the presence and absence of isoform-selective P450 inhibitors. Results of a proof-of-concept study with the MCCA and CMPIA with a known hepatotoxicant, aflatoxin B1 (AFB1), are reported. AFB1 is known to require P450 metabolism for its toxicity. In the MCCA, AFB1 was found to have significantly higher cytotoxicity in human hepatocytes than CHO cells, therefore confirming its requirement for biotransformation to be toxic. ABT was found to effectively attenuate AFB1 cytotoxicity, confirming that P450 metabolism was involved in its metabolic activation. In the CMPIA, AFB1 cytotoxicity was found to be attenuated by ketoconazole and diethyldithiocarbamate, but not by furafylline, quinidine, and sulfaphenazole. Results with the isoform-selective inhibitors suggest that the isoforms inhibited by ketoconazole (mainly CYP3A4) and diethyldithiocarbamate (mainly CYP2A6, and CYP2E1), but not the isoforms inhibited by furafylline (mainly CYP1A2), sulfaphenazole (mainly CYP2C9) and quinidine (mainly CYP2D6) are involved in the metabolic activation of AFB1. This proof-of-concept study suggests that MCCA and CMPIA with cryopreserved human hepatocytes are potentially useful for the evaluation of the relationship between human xenobiotic metabolism and toxicity.
Collapse
|
29
|
Grindon C, Combes R, Cronin MT, Roberts DW, Garrod JF. Integrated Decision-tree Testing Strategies for Developmental and Reproductive Toxicity with Respect to the Requirements of the EU REACH Legislation. Altern Lab Anim 2008; 36 Suppl 1:123-38. [DOI: 10.1177/026119290803601s10] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Liverpool John Moores University and FRAME conducted a research project, sponsored by Defra, on the status of alternatives to animal testing with regard to the European Union REACH (Registration, Evaluation and Authorisation of Chemicals) system for the safety testing and risk assessment of chemicals. The project covered all the main toxicity endpoints associated with the REACH system. This paper focuses on the prospects for the use of alternative methods (both in vitro and in silico) in developmental and reproductive toxicity testing. It considers many tests based on primary cells and cell lines, and the available expert systems and QSARs for developmental and reproductive toxicity, and also covers tests for endocrine disruption. Ways in which reduction and refinement measures can be used are also discussed, particularly the use of an enhanced one-generation reproductive study, which could potentially replace the two-generation study, and therefore considerably reduce the number of animals required in reproductive toxicity. Decision-tree style integrated testing strategies are also proposed for developmental and reproductive toxicity and for endocrine disruption, followed by a number of recommendations for the future facilitation of developmental and reproductive toxicity testing, with respect to human risk assessment.
Collapse
Affiliation(s)
| | | | - Mark T.D. Cronin
- School of Pharmacy and Chemistry, Liverpool John Moores University, Liverpool, UK
| | - David W. Roberts
- School of Pharmacy and Chemistry, Liverpool John Moores University, Liverpool, UK
| | - John F. Garrod
- Chemicals and Nanotechnologies Division, Defra, London, UK
| |
Collapse
|
30
|
Fröhlich AK, Girreser U, Clement B. Metabolism of benzamidoxime (N-hydroxyamidine) in human hepatocytes and role of UDP-glucuronosyltransferases. Xenobiotica 2008; 35:17-25. [PMID: 15788365 DOI: 10.1080/00498250400021895] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
N-Hydroxyamidines (amidoximes) can act as pro-drugs of amidines (e.g. ximelagatran, a novel direct thrombin inhibitor). This known pro-drug principle is based on the N-reduction of an oral bioavailable amidoxime to its active form. Previous study of the metabolism of the model substrate benzamidoxime by pig hepatocytes demonstrated the formation of benzamidoxime-O-glucuronide in addition to the well-established N-reduction. The objective of the present work was to investigate the glucuronidation of benzamidoxime by using cultivated cryopreserved human hepatocytes. Furthermore, the involvement of human UDP-glucuronosyltransferases (UGTs) was examined by incubating benzamidoxime in the presence of eight human hepatic recombinant UGT enzymes. Metabolites were analysed by liquid chromatography/mass spectrometry using electrospray ionization and compared with authentic synthetic compounds. For the first time, the O-glucuronidation of benzamidoxime was demonstrated in cultures of human hepatocytes. UGT1A9 is the most efficient enzyme conjugating benzamidoxime, whereas the conversion activities of UGT1A1 and UGT1A3 were 60-fold lower. Human hepatocytes form two non-mutagenic compounds: benzamidine, as the predominating metabolite, and benzamidoxime-O-glucuronide to a lesser extent. N-oxidation of benzamidine was not detected.
Collapse
Affiliation(s)
- A K Fröhlich
- Pharmaceutical Institute, Christian-Albrechts-University of Kiel, Kiel, Germany
| | | | | |
Collapse
|
31
|
Sahu SC, Wiesenfeld PL, Kim CS, Ross IA, Sapienza PP, Newell R, O'Donnell MW, Flynn TJ. Validation of an in vitro model for assessment of androstenedione hepatotoxicity using the rat liver cell line clone-9. J Appl Toxicol 2008; 28:703-9. [PMID: 18059068 DOI: 10.1002/jat.1325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Androstenedione, a naturally occurring steroid hormone, has been used to enhance athletic performance. Little is known, however, about its hepatotoxicity. Clone-9 cells, a non-transformed epithelial cell line that was originally isolated from normal liver of a 4-week old Sprague-Dawley rat, were used as an in vitro model to assess the hepatotoxic potential of androstenedione. The cultures were treated with androstenedione for 24 h at 37 degrees C in 5% CO(2) at concentrations of 0-100 microg ml(-1). After the treatment period, the cells and the culture supernatants were assayed for markers of cytotoxicity which included: release of liver enzymes, cell viability, cellular double-stranded DNA content, oxidative stress, steatosis, cellular ATP content, caspase-3 activity, the mitochondrial permeability transition and induction of cytochrome P450 activity. Significant concentration-dependent differences from control were observed in some endpoints at medium concentrations of 10 microg ml(-1) and above. These in vitro findings were compared with comparable endpoints obtained from an in vivo study of androstenedione toxicity in female Sprague-Dawley rats. Of the eight endpoints that could be compared between the two studies, only three (lipid accumulation, ATP depletion and P450 activity) appeared to be concordant. This suggests that, under the experimental conditions used, the clone-9 cells were not a good model for androstenedione hepatotoxicity.
Collapse
Affiliation(s)
- Saura C Sahu
- Division of Toxicology, Office of Applied Research and Safety Assessment, U. S. Food and Drug Administration, Laurel, MD 20708, USA.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Sahu SC, Garthoff LH, Robl MG, Chirtel SJ, Ruggles DI, Flynn TJ, Sobotka TJ. Rat liver clone-9 cells in culture as a model for screening hepatotoxic potential of food-related products: hepatotoxicity of deoxynivalenol. J Appl Toxicol 2008; 28:765-72. [DOI: 10.1002/jat.1337] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
33
|
Lohitnavy M, Lu Y, Lohitnavy O, Chubb LS, Hirono S, Yang RSH. A possible role of multidrug resistance-associated protein 2 (Mrp2) in hepatic excretion of PCB126, an environmental contaminant: PBPK/PD modeling. Toxicol Sci 2008; 104:27-39. [PMID: 18281255 DOI: 10.1093/toxsci/kfn026] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
3,3',4,4',5'-Pentachlorobiphenyl (PCB126) is a carcinogenic environmental pollutant and its toxicity is mediated through binding with aryl hydrocarbon receptor (AhR). Earlier, we found that PCB126 treated F344 rats had 110-400 times higher PCB126 concentration in the liver than in the fat. Protein binding was suspected to be a major factor for the high liver concentration of PCB126 despite its high lipophilicity. In this research, we conducted a combined pharmacokinetic/pharmacodynamic study in male F344 rats. In addition to blood and tissue pharmacokinetics, we use the development of hepatic preneoplastic foci (glutathione-S-transferase placental form [GSTP]) as a pharmacodynamic endpoint. Experimental data were utilized for building a physiologically based pharmacokinetic/pharmacodynamic (PBPK/PD) model. PBPK/PD modeling was consistent with the experimental PK and PD data. Salient features of this model include: (1) bindings between PCB126 and hepatic proteins, particularly the multidrug resistance-associated protein (Mrp2), a protein transporter; (2) Mrp2-mediated excretion; and (3) a relationship between area under the curve of PCB126 in the livers and % volume of GSTP foci. Mrp2 involvement in PCB126 pharmacokinetics is supported by computational chemistry calculation using a three-dimensional quantitative structure-activity relationship model of Mrp2 developed by S. Hirono et al. (2005, Pharm. Res. 22, 260-269). This work, for the first time, provided a plausible role of a versatile hepatic transporter for drugs, Mrp2, in the disposition of an important environmental pollutant, PCB126.
Collapse
Affiliation(s)
- Manupat Lohitnavy
- Quantitative and Computational Toxicology Group, Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado 80523-1680, USA
| | | | | | | | | | | |
Collapse
|
34
|
Grindon C, Combes R, Cronin MT, Roberts DW, Garrod JF. Integrated Decision-tree Testing Strategies for Developmental and Reproductive Toxicity with Respect to the Requirements of the EU REACH Legislation. Altern Lab Anim 2008. [DOI: 10.1177/026119290803600108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Liverpool John Moores University and FRAME conducted a research project, sponsored by Defra, on the status of alternatives to animal testing with regard to the European Union REACH (Registration, Evaluation and Authorisation of Chemicals) system for the safety testing and risk assessment of chemicals. The project covered all the main toxicity endpoints associated with the REACH system. This paper focuses on the prospects for the use of alternative methods (both in vitro and in silico) in developmental and reproductive toxicity testing. It considers many tests based on primary cells and cell lines, and the available expert systems and QSARs for developmental and reproductive toxicity, and also covers tests for endocrine disruption. Ways in which reduction and refinement measures can be used are also discussed, particularly the use of an enhanced one-generation reproductive study, which could potentially replace the two-generation study, and therefore considerably reduce the number of animals required in reproductive toxicity. Decision-tree style integrated testing strategies are also proposed for developmental and reproductive toxicity and for endocrine disruption, followed by a number of recommendations for the future facilitation of developmental and reproductive toxicity testing, with respect to human risk assessment.
Collapse
Affiliation(s)
| | | | - Mark T.D. Cronin
- School of Pharmacy and Chemistry, Liverpool John Moores University, Liverpool, UK
| | - David W. Roberts
- School of Pharmacy and Chemistry, Liverpool John Moores University, Liverpool, UK
| | - John F. Garrod
- Chemicals and Nanotechnologies Division, Defra, London, UK
| |
Collapse
|
35
|
Kale VM, Miranda SR, Wilbanks MS, Meyer SA. Comparative cytotoxicity of alachlor, acetochlor, and metolachlor herbicides in isolated rat and cryopreserved human hepatocytes. J Biochem Mol Toxicol 2008; 22:41-50. [DOI: 10.1002/jbt.20213] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
36
|
Cermak R. Effect of dietary flavonoids on pathways involved in drug metabolism. Expert Opin Drug Metab Toxicol 2007; 4:17-35. [DOI: 10.1517/17425255.4.1.17] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
37
|
Sahu SC, Ruggles DI, O'Donnell MW. Prooxidant activity and toxicity of nordihydroguaiaretic acid in clone-9 rat hepatocyte cultures. Food Chem Toxicol 2006; 44:1751-7. [PMID: 16839654 DOI: 10.1016/j.fct.2006.05.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2005] [Revised: 03/31/2006] [Accepted: 05/24/2006] [Indexed: 11/21/2022]
Abstract
Nordihydroguaiaretic acid (NDGA) is a polyphenol. It is present at high concentrations in the leaves of the evergreen desert shrub, Larrea tridentate (Creosote bush), which has a long history of medicinal use traditionally by the native Americans and Mexicans. It is generally believed that the antioxidant properties of NDGA are responsible for the medicinal value of this desert shrub. The clone-9 rat hepatocyte cultures were used as an in vitro model to assess the hepatotoxic potential of NDGA and to determine whether it exhibits any prooxidant activity. The hepatocyte cultures were treated with NDGA for 2 h at 37 degrees C at concentrations of 0-100 microM. After the treatment period the cells, the culture supernatants and cell lysates were assayed for evaluation of prooxidant activity and toxicity of NDGA. Oxidative stress level and oxidative cell injury as measured by the peroxidation of membrane lipids and DNA double-strand breaks were used to index prooxidant activity. Cytotoxicity as measured by the leakage of the liver enzyme lactate dehydrogenase (LDH) into the culture medium, mitochondrial function and extent of cell proliferation were used as the endpoints of toxicity. Significant concentration-dependent differences were observed in these biomarkers over the concentration range examined demonstrating the prooxidant activity and toxicity of NDGA in clone-9 rat hepatocyte cultures.
Collapse
Affiliation(s)
- Saura C Sahu
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, US Food and Drug Administration, 8301 Muirkirk Road, Laurel, MD 20708, USA.
| | | | | |
Collapse
|
38
|
Combes R, Balls M. Intelligent testing strategies for chemicals testing -- a case of more haste, less speed? Altern Lab Anim 2005; 33:289-97. [PMID: 16180981 DOI: 10.1177/026119290503300302] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The prospects for using (Q)SAR modelling, read-across (chemical) and other non-animal approaches as part of integrated testing strategies for chemical risk assessment, within the framework of the EU REACH legislation, are considered. The potential advantages and limitations of (Q)SAR modelling and read-across methods for chemical regulatory risk assessment are reviewed. It is concluded that it would be premature to base a testing strategy on chemical-based computational modelling approaches, until such time as criteria to validate them for their reliability and relevance by using independent and transparent procedures, have been agreed. This is mainly because of inherent problems in validating and accepting (Q)SARs for regulatory use in ways that are analogous to those that have been developed and applied for in vitro tests. Until this issue has been resolved, it is recommended that testing strategies should be developed which comprise the integrated use of computational and read-across approaches. These should be applied in a cautious and judicious way, in association with available tissue culture methods, and in conjunction with metabolism and biokinetic studies. Such strategies should be intelligently applied by being driven by exposure information (based on bioavailability, not merely on production volume) and hazard information needs, in preference to a tick-box approach. In the meantime, there should be increased efforts to develop improved (Q)SARs, expert systems and new in vitro methods, and, in particular, ways to expedite their validation and acceptance must be found and prospectively agreed with all major stakeholders.
Collapse
|
39
|
Froehlich AK, Girreser U, Clement B. METABOLISM OF N-HYDROXYGUANIDINES (N-HYDROXYDEBRISOQUINE) IN HUMAN AND PORCINE HEPATOCYTES: REDUCTION AND FORMATION OF GLUCURONIDES. Drug Metab Dispos 2005; 33:1532-7. [PMID: 16033947 DOI: 10.1124/dmd.105.004572] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The biotransformation of N-hydroxydebrisoquine, a model substrate for N-hydroxyguanidines, was studied in vitro with cultured and characterized porcine and human hepatocytes. The objective of the present work was to compare the N-oxidative and N-reductive metabolism of this compound using a monolayer culture system with previously described microsomal studies and to investigate the phase 2 metabolism, in particular, the glucuronidation of this class of compounds. At the same time, the suitability of pig hepatocytes as a model system for the human metabolism could be investigated. Two glucuronides of the parent compound N-hydroxydebrisoquine were analyzed. For the first time, one of these phase 2 metabolites could be identified as an O-glucuronide of an N-hydroxyguanidine by comparing it to a synthesized authentic compound. The involvement of certain human UDP-glucuronosyltransferases (UGTs) was evaluated by incubating the substrate with eight human hepatic recombinant UGT enzymes. Metabolites were determined by a newly developed LC-MS (liquid chromatography/mass spectrometry) analysis using electrospray ionization (ESI). The known microsomal reduction of the N-hydroxylated compound was also demonstrated with hepatocytes. The N-hydroxylation of the corresponding reduced compound (debrisoquine), which was previously described with microsomes, could not be detected in hepatocytes. There was no qualitative difference in the formation of the described derivatives by human and porcine hepatocytes. All phase 2 metabolites identified in hepatocyte culture were also formed by glucuronosyltransferases. In culture, the N-reduction of the N-hydroxylated substrate is the dominating reaction, indicating a predominance of N-reduction in vivo.
Collapse
Affiliation(s)
- Anja Kristina Froehlich
- Pharmazeutisches Institut, Christian-Albrechts-Universität Kiel, Gutenbergstr. 76, D-24118 Kiel, Germany
| | | | | |
Collapse
|
40
|
Vermeir M, Annaert P, Mamidi RNVS, Roymans D, Meuldermans W, Mannens G. Cell-based models to study hepatic drug metabolism and enzyme induction in humans. Expert Opin Drug Metab Toxicol 2005; 1:75-90. [PMID: 16922654 DOI: 10.1517/17425255.1.1.75] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Cell-based in vitro models are invaluable tools in elucidating the pharmacokinetic profile of a drug candidate during its drug discovery and development process. As biotransformation is one of the key determinants of a drug's disposition in the body, many in vitro models to study drug metabolism have been established, and others are still being developed and validated. This review is aimed at providing the reader with a concise overview of the characteristics and optimal application of established and emerging in vitro cell-based models to study human drug metabolism and induction of drug metabolising enzymes in the liver. The strengths and weaknesses of liver-derived models, such as primary hepatocytes, either freshly isolated or cryopreserved, and from adult or fetal donors, precision-cut liver slices, and cell lines, including immortalised cells, reporter cell lines, hepatocarcinoma-derived cell lines and recombinant cell lines, are discussed. Relevant cell culture configuration aspects as well as other models such as stem cell-derived hepatocyte-like cells and humanised animal models are also reviewed. The status of model development, their acceptance by health authorities and recommendations for the most appropriate use of the models are presented.
Collapse
Affiliation(s)
- Marc Vermeir
- Johnson & Johnson Pharmaceutical Research & Development, Preclinical Pharmacokinetics, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | | | | | | | | | | |
Collapse
|
41
|
Anderson LW, Collins JM, Klecker RW, Katki AG, Parchment RE, Boinpally RR, LoRusso PM, Ivy SP. Metabolic profile of XK469 (2(R)-[4-(7-chloro-2-quinoxalinyl)oxyphenoxy]-propionic acid; NSC698215) in patients and in vitro: low potential for active or toxic metabolites or for drug–drug interactions. Cancer Chemother Pharmacol 2005; 56:351-7. [PMID: 15895233 DOI: 10.1007/s00280-004-0962-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2004] [Accepted: 10/18/2004] [Indexed: 10/25/2022]
Abstract
As part of an ongoing phase 1 study, we studied the excretion of XK469 and its metabolism in patients and in vitro. Five primary metabolites were identified by HPLC/MS/MS. An oxidized product formed by cytosolic aldehyde oxidase was the predominant species both in urine and human hepatocytes in vitro. Conjugates of XK469 with glycine, taurine, and glucuronic acid, as well as the microsomal product, 4-oxo-XK469, were also found in urine and in vitro, but none were major contributors to the mass balance for XK469 elimination. Based upon the relative concentrations circulating in plasma, systemic exposure to parent drug was 100-fold higher than for the metabolites. Thus, both toxicity and efficacy of XK469 are most likely to be produced by the parent molecule, rather than the metabolites. Urinary recovery of parent drug was low (2% of dose in 24 h), partly because of the long half-life of XK469 (approximately 3 days). In addition, the metabolite profile in urine indicates that only 25% of the XK469-derived material was unchanged drug. Thus, urinary excretion was not a major factor in XK469 elimination. Variations in systemic exposure to XK469 will be strongly influenced by factors that alter the activity of aldehyde oxidase, including pharmacogenetics, enzyme inhibition, and enzyme induction, but no specific modifiers have been reported. The multiday half-life of XK469 hampered our ability to obtain a complete mass balance, and the possibility exists that other routes, such as biliary excretion, may also play a substantial role in XK469 disposition.
Collapse
Affiliation(s)
- Lawrence W Anderson
- Food and Drug Administration, WO Bldg 64, Rm 2014, 10903 New Hampshire Avenue, HFD-902, Silver Spring, MD 20993, USA.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Li AP, Bode C, Sakai Y. A novel in vitro system, the integrated discrete multiple organ cell culture (IdMOC) system, for the evaluation of human drug toxicity: comparative cytotoxicity of tamoxifen towards normal human cells from five major organs and MCF-7 adenocarcinoma breast cancer cells. Chem Biol Interact 2005; 150:129-36. [PMID: 15522266 DOI: 10.1016/j.cbi.2004.09.010] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
In vitro assays involving primary cells are used routinely to evaluate organ-specific toxic effects, for instance, the use of primary hepatocytes to evaluate hepatotoxicity. A major drawback of an in vitro system is the lack of multiple organ interactions as observed in a whole organism. A novel cell culture system, the integrated discrete multiorgan cell culture system (IdMOC), is described here. The IdMOC is based on the "wells within a well" concept, consisting of a cell culture plate with larger, containing wells, within each of which are multiple smaller wells. Cells from multiple organs can be cultured initially in the small wells (one organ per well, each in its specialized medium). On the day of toxicity testing, a volume of drug-containing medium is added to the containing well to flood all inner wells, thereby interconnecting all the small wells. After testing, the overlying medium is removed and each cell type is evaluated for toxicity using appropriate endpoints. We report here the application of IdMOC in the evaluation of the cytotoxicity of tamoxifen, an anticancer agent with known human toxicity, on primary cells from multiple human organs: liver (hepatocytes), kidney (kidney cortical cells), lung (small airway epithelial cells), central nervous system (astrocytes), blood vessels (aortic endothelial cells) as well as the MCF-7 human breast adenocarcinoma cells. IdMOC produced results that can be used for the quantitative evaluation of its anticancer effects (i.e., cytotoxicity towards MCF-7 cells) versus its toxicity toward normal organs (i.e., liver, kidney, lung, CNS, blood vessels).
Collapse
Affiliation(s)
- Albert P Li
- Advanced Pharmaceutical Sciences Inc., PMB 146, 6400 Baltimore National Pike, Baltimore, MD 21228, USA.
| | | | | |
Collapse
|
43
|
Combes RD. The case for taking account of metabolism when testing for potential endocrine disruptors in vitro. Altern Lab Anim 2004; 32:121-35. [PMID: 15601241 DOI: 10.1177/026119290403200210] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Legislation in the USA, Europe and Japan will require that chemicals are tested for their ability to disrupt the hormonal systems of mammals. Such chemicals are known as endocrine disruptors (EDs), and will require extensive testing as part of the new European Union Registration, Evaluation and Authorisation of Chemicals (REACH) system for the risk assessment of chemicals. Both in vivo and in vitro tests are proposed for this purpose, and there has been much discussion and action concerning the development and validation of such tests. However, to date, little interest has been shown in incorporating metabolism into in vitro tests for EDs, in sharp contrast to other areas of toxicity testing, such as genotoxicity, and, ironically, such in vitro tests are criticised for not modelling in vivo metabolism. This is despite the existence of much information showing that endogenous and exogenous steroids are extensively metabolised by Phase I and Phase II enzymes both in the liver and in hormonally active tissues. Such metabolism can lead to the activation or detoxification of steroids and EDs. The absence of metabolism from these tests could give rise to false-positive data (due to lack of detoxification) or false-negative data (lack of activation). This paper aims to explain why in vitro assays for EDs should incorporate mammalian metabolising systems. The background to ED testing, the test methods available, and the role of mammalian metabolism in the activation and detoxification of both endogenous and exogenous steroids, are described. The available types of metabolising systems are compared, and the potential problems in incorporating metabolising systems into in vitro tests for EDs, and how these might be overcome, are discussed. It is recommended that there should be: a) an assessment of the intrinsic metabolising capacity of cell systems used in tests for EDs; b) an investigation into the relevance of using the prostaglandin H synthase system for metabolising EDs; and c) a feasibility study into the generation of genetically engineered mammalian cell lines expressing specific metabolising enzymes, which could also be used to detect EDs.
Collapse
Affiliation(s)
- Robert D Combes
- FRAME, Russell & Burch House, 96-98 North Sherwood Street, Nottingham NG1 4EE, UK.
| |
Collapse
|
44
|
Keldenich J. Prediction of human clearance (CL) and volume of distribution (VD). DRUG DISCOVERY TODAY. TECHNOLOGIES 2004; 1:389-395. [PMID: 24981619 DOI: 10.1016/j.ddtec.2004.10.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The crucial pharmacokinetic parameters 'volume of distribution' and 'human clearance' determine the extent and duration a compound remains in an organism. Potential drug candidates will fail to become successful drugs on the market without favorable values for these parameters, even if they are most efficacious at the target in vitro.The prediction of volume of distribution and human clearance in drug research and development is a key technology to assess possible drug candidates.:
Collapse
Affiliation(s)
- Jörg Keldenich
- Bayer HealthCare AG, Pharmaceutical Research, D-42096 Wuppertal, Germany.
| |
Collapse
|
45
|
Abstract
A comprehensive, multidisciplinary approach is proposed here for the development of a drug with an acceptable safety profile. Key parameters to be considered for drug safety evaluation based on this comprehensive approach include the following: (1) Pharmacology: Possible toxicity due to drug-target interactions, including interactions with unintended molecular targets, or with molecular targets in unintended organs. (2) Chemistry: Chemical scaffolding and side-chains with safety concerns. (3) Toxicology: Toxicity in animals in vivo, and in relevant animal and human cells in culture. (4) Drug metabolism and pharmacokinetics: Safety concerns due to toxification or detoxification, organ distribution, clearance and pharmacokinetic drug-drug interactions. (5) RISK FACTORS: Physiological, environmental and genetic factors that may enhance a patient's susceptibility. It is proposed that this integrated, multidisciplinary approach to safety evaluation may enhance the accuracy of the prediction of drug safety and thereby the efficiency of drug development.
Collapse
Affiliation(s)
- Albert P Li
- Advanced Pharmaceutical Sciences, Inc., PMB #146, 6400 Baltimore National Pike, Baltimore, MD 21228, USA.
| |
Collapse
|
46
|
Affiliation(s)
- Albert P Li
- Advanced Pharmaceutical Sciences, PMB#146, 6400 Baltimore National Pike, Baltimore, MD 21228, USA
| |
Collapse
|
47
|
Willett KL, Roth RA, Walker L. Workshop Overview: Hepatotoxicity Assessment for Botanical Dietary Supplements. Toxicol Sci 2004; 79:4-9. [PMID: 14976355 DOI: 10.1093/toxsci/kfh075] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Botanical dietary supplements (herbal products) have flooded the market in the United States over the past decade, and studies show a significant percentage of Americans use them. With increasing frequency and duration of exposure, some serious adverse effects, though relatively uncommon, have been reported. Among the most troublesome is the association of some botanicals with serious hepatotoxicity. In some cases, hepatotoxicity has been linked to the consumption of botanicals with recognized hepatotoxic components (e.g., pyrrolizidine alkaloids). However, in other cases, the causative agent(s) is less clear and, overall, the mechanisms of hepatotoxicity are poorly understood. To help create a scientific basis for understanding botanical-induced hepatotoxicity and better tools for hepatotoxicity assessment and prediction, the National Center for Natural Product Research (NCNPR) hosted a workshop (September 8 and 9, 2003) in cooperation with the Center for Food Safety and Applied Nutrition (CFSAN) of the Food and Drug Administration (FDA). The workshop featured presentations by 22 experts and was attended by 65 individuals. The agenda can be found in the supplementary data at www.toxsci.oupjournals.org.
Collapse
Affiliation(s)
- Kristine L Willett
- Department of Pharmacology and Environmental Toxicology, School of Pharmacy, University of Mississippi, University, Mississippi 38677, USA
| | | | | |
Collapse
|
48
|
Lindhagen E, Vig Hjarnaa PJ, Friberg LE, Latini S, Larsson R. Pharmacodynamic differences between species exemplified by the novel anticancer agent CHS 828. Drug Dev Res 2004. [DOI: 10.1002/ddr.10353] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
49
|
Xu J, Ma M, Purcell WM. Biochemical and functional changes of rat liver spheroids during spheroid formation and maintenance in culture: II. nitric oxide synthesis and related changes. J Cell Biochem 2003; 90:1176-85. [PMID: 14635191 DOI: 10.1002/jcb.10731] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Liver cells isolated from intact tissue can reaggregate to form three-dimensional, multicellular spheroids in vitro. During this process, cells undergo a histological and environmental change. How cells respond biochemically to this change has not been studied in detail previously. We have investigated some biochemical changes in rat liver cells during the formation and maintenance of spheroids. Liver cells were isolated from male Sprague rats and spheroids cultured by a gyrotatory-mediated method. Liver cells were shown to respond to the isolation procedure and the formation of spheroids triggered histological environmental changes that increased arginine uptake, nitric oxide (NO) and urea syntheses, as well as raised levels of GSH, GSSG, glutamic acid and aspartic acid secretion within the first couple of days after cell isolation. Levels were maintained at a relatively stable level in the mature spheroids (>5 days) over the 3 week period of observation. P450 1A1 activity was lost in the first 2 days and gradually recovered thereafter. This study, for the first time, shows that liver cells after isolation and during spheroid formation actively uptake arginine and increase NO and urea syntheses. A high level of NO is likely to play an important role in modulating a series of biochemical changes in liver cells. It is considered that liver cells actively respond to the 'challenge' induced by the isolation procedure and subsequent histological environmental changes, and biochemical modulation and instability result. The stable cell-cell contacts and histological environment in mature spheroids permit and support functional recovery and maintenance in vitro. This period of stability permits the use of spheroids in toxicity studies to establish acute and chronic paradigms.
Collapse
Affiliation(s)
- Jinsheng Xu
- Centre for Research in Biomedicine, Faculty of Applied Sciences, University of the West of England, Bristol, United Kingdom.
| | | | | |
Collapse
|
50
|
Blaauboer BJ. Biokinetic and Toxicodynamic Modelling and its Role in Toxicological Research and Risk Assessment. Altern Lab Anim 2003; 31:277-81. [PMID: 15612871 DOI: 10.1177/026119290303100310] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Toxicological risk assessment for chemicals is still mainly based on highly standardised protocols for animal experimentation and exposure assessment. However, developments in our knowledge of general physiology, in chemicobiological interactions and in (computer-supported) modelling, have resulted in a tremendous change in our understanding of the molecular mechanisms underlying the toxicity of chemicals. This permits the development of biologically based models, in which the biokinetics as well as the toxicodynamics of compounds can be described. In this paper, the possibilities are discussed of developing systems in which the systemic (acute and chronic) toxicities of chemicals can be quantified without the heavy reliance on animal experiments. By integrating data derived from different sources, predictions of toxicity can be made. Key elements in this integrated approach are the evaluation of chemical functionalities representing structural alerts for toxic actions, the construction of biokinetic models on the basis of non-animal data (for example, tissue–blood partition coefficients, in vitro biotransformation parameters), tests or batteries of tests for determining basal cytotoxicity, and more-specific tests for evaluating tissue or organ toxicity. It is concluded that this approach is a useful tool for various steps in toxicological hazard and risk assessment, especially for those forms of toxicity for which validated in vitro and other non-animal tests have already been developed.
Collapse
Affiliation(s)
- Bas J Blaauboer
- Institute for Risk Assessment Sciences (IRAS), Division of Toxicology, Utrecht University, P.O. Box 80.176, 3508 TD Utrecht, The Netherlands
| |
Collapse
|