Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Toxicol Sci ; 196(1): 112-125, 2023 10 30.
Article in English | MEDLINE | ID: mdl-37647630

ABSTRACT

To minimize the occurrence of unexpected toxicities in early phase preclinical studies of new drugs, it is vital to understand fundamental similarities and differences between preclinical species and humans. Species differences in sensitivity to acetaminophen (APAP) liver injury have been related to differences in the fraction of the drug that is bioactivated to the reactive metabolite N-acetyl-p-benzoquinoneimine (NAPQI). We have used physiologically based pharmacokinetic modeling to identify oral doses of APAP (300 and 1000 mg/kg in mice and rats, respectively) yielding similar hepatic burdens of NAPQI to enable the comparison of temporal liver tissue responses under conditions of equivalent chemical insult. Despite pharmacokinetic and biochemical verification of the equivalent NAPQI insult, serum biomarker and tissue histopathology analyses revealed that mice still exhibited a greater degree of liver injury than rats. Transcriptomic and proteomic analyses highlighted the stronger activation of stress response pathways (including the Nrf2 oxidative stress response and autophagy) in the livers of rats, indicative of a more robust transcriptional adaptation to the equivalent insult. Components of these pathways were also found to be expressed at a higher basal level in the livers of rats compared with both mice and humans. Our findings exemplify a systems approach to understanding differential species sensitivity to hepatotoxicity. Multiomics analysis indicated that rats possess a greater basal and adaptive capacity for hepatic stress responses than mice and humans, with important implications for species selection and human translation in the safety testing of new drug candidates associated with reactive metabolite formation.


Subject(s)
Acetaminophen , Chemical and Drug Induced Liver Injury , Rats , Mice , Humans , Animals , Acetaminophen/toxicity , Acetaminophen/metabolism , Proteomics , Species Specificity , Chemical and Drug Induced Liver Injury/metabolism , Liver/metabolism , Oxidative Stress , Systems Analysis
2.
Regul Toxicol Pharmacol ; 142: 105415, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37257751

ABSTRACT

Low levels of N-nitrosamines (NAs) were detected in pharmaceuticals and, as a result, health authorities (HAs) have published acceptable intakes (AIs) in pharmaceuticals to limit potential carcinogenic risk. The rationales behind the AIs have not been provided to understand the process for selecting a TD50 or read-across analog. In this manuscript we evaluated the toxicity data for eleven common NAs in a comprehensive and transparent process consistent with ICH M7. This evaluation included substances which had datasets that were robust, limited but sufficient, and substances with insufficient experimental animal carcinogenicity data. In the case of robust or limited but sufficient carcinogenicity information, AIs were calculated based on published or derived TD50s from the most sensitive organ site. In the case of insufficient carcinogenicity information, available carcinogenicity data and structure activity relationships (SARs) were applied to categorical-based AIs of 1500 ng/day, 150 ng/day or 18 ng/day; however additional data (such as biological or additional computational modelling) could inform an alternative AI. This approach advances the methodology used to derive AIs for NAs.


Subject(s)
Nitrosamines , Animals , Nitrosamines/toxicity , Carcinogens , Structure-Activity Relationship , Pharmaceutical Preparations
3.
Nat Rev Drug Discov ; 22(4): 317-335, 2023 04.
Article in English | MEDLINE | ID: mdl-36781957

ABSTRACT

For decades, preclinical toxicology was essentially a descriptive discipline in which treatment-related effects were carefully reported and used as a basis to calculate safety margins for drug candidates. In recent years, however, technological advances have increasingly enabled researchers to gain insights into toxicity mechanisms, supporting greater understanding of species relevance and translatability to humans, prediction of safety events, mitigation of side effects and development of safety biomarkers. Consequently, investigative (or mechanistic) toxicology has been gaining momentum and is now a key capability in the pharmaceutical industry. Here, we provide an overview of the current status of the field using case studies and discuss the potential impact of ongoing technological developments, based on a survey of investigative toxicologists from 14 European-based medium-sized to large pharmaceutical companies.


Subject(s)
Drug Industry , Drug-Related Side Effects and Adverse Reactions , Humans , Drug-Related Side Effects and Adverse Reactions/prevention & control , Biomarkers , Technology , Drug Evaluation, Preclinical
4.
Toxicol Appl Pharmacol ; 391: 114915, 2020 03 15.
Article in English | MEDLINE | ID: mdl-32035082

ABSTRACT

Idiosyncratic drug-induced liver injury (IDILI) is a severe disease that cannot be detected during drug development. It has been shown that hepatotoxicity of some compounds associated with IDILI becomes apparent when these are combined in vivo and in vitro with LPS or TNF. Among these compounds trovafloxacin (TVX) induced apoptosis in the liver and increased pro-inflammatory cytokines in mice exposed to LPS/TNF. The hepatocyte survival and the cytokine release after TNF/LPS stimulation relies on a pulsatile activation of NF-κB. We set out to evaluate the dynamic activation of NF-κB in response to TVX + TNF or LPS models, both in mouse and human cells. Remarkably, TVX prolonged the first translocation of NF-κB induced by TNF both in vivo and in vitro. The prolonged p65 translocation caused by TVX was associated with an increased phosphorylation of IKK and MAPKs and accumulation of inhibitors of NF-κB such as IκBα and A20 in HepG2. Coherently, TVX suppressed further TNF-induced NF-κB translocations in HepG2 leading to decreased transcription of ICAM-1 and inhibitors of apoptosis. TVX prolonged LPS-induced NF-κB translocation in RAW264.7 macrophages increasing the secretion of TNF. In summary, this study presents new, relevant insights into the mechanism of TVX-induced liver injury underlining the resemblance between mouse and human models. In this study we convincingly show that regularly used toxicity models provide a coherent view of relevant pathways for IDILI. We propose that assessment of the kinetics of activation of NF-κB and MAPKs is an appropriate tool for the identification of hepatotoxic compounds during drug development.


Subject(s)
Anti-Bacterial Agents/toxicity , Chemical and Drug Induced Liver Injury/pathology , Fluoroquinolones/toxicity , Lipopolysaccharides/pharmacology , Naphthyridines/toxicity , Transcription Factor RelA/drug effects , Transcription Factor RelA/genetics , Translocation, Genetic/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Animals , Apoptosis/drug effects , Chemical and Drug Induced Liver Injury/genetics , Cytokines/metabolism , Humans , I-kappa B Proteins/metabolism , Mice , Mitogen-Activated Protein Kinases/metabolism , RAW 264.7 Cells , Tumor Necrosis Factor-alpha/metabolism
5.
Nat Rev Drug Discov ; 19(2): 131-148, 2020 02.
Article in English | MEDLINE | ID: mdl-31748707

ABSTRACT

Drug-induced liver injury (DILI) is a patient-specific, temporal, multifactorial pathophysiological process that cannot yet be recapitulated in a single in vitro model. Current preclinical testing regimes for the detection of human DILI thus remain inadequate. A systematic and concerted research effort is required to address the deficiencies in current models and to present a defined approach towards the development of new or adapted model systems for DILI prediction. This Perspective defines the current status of available models and the mechanistic understanding of DILI, and proposes our vision of a roadmap for the development of predictive preclinical models of human DILI.


Subject(s)
Chemical and Drug Induced Liver Injury/diagnosis , Disease Models, Animal , Drug-Related Side Effects and Adverse Reactions/prevention & control , Animals , Chemical and Drug Induced Liver Injury/etiology , Humans , Predictive Value of Tests
6.
Toxicol In Vitro ; 61: 104595, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31288073

ABSTRACT

Bile acids (BAs) are recognised as the causative agents of toxicity in drug-induced cholestasis (DIC). Research in isolated mitochondria and HepG2 cells have demonstrated BA-mediated mitochondrial dysfunction as a key mechanism of toxicity in DIC. However, HepG2 cells are of limited suitability for DIC studies as they do not express the necessary physiological characteristics. In this study, the mitotoxic potentials of BA mixtures were assessed in isolated mitochondria and a better-suited hepatic model, HepaRG cells. BAs induced structural alterations and a loss of mitochondrial membrane potential (MMP) in isolated mitochondria however, this toxicity did not translate to HepaRG cells. There were no changes in oxygen consumption rate, MMP or ATP levels in glucose and galactose media, indicating that there was no direct mitochondrial toxicity mediated via electron transport chain dysfunction in HepaRG cells. Assessment of key biliary transporters revealed that there was a time-dependent reduction in the expression and activity of multi-drug resistance protein 2 (MRP2), which was consistent with the induction of cytotoxicity in HepaRG cells. Overall, the findings from this study have demonstrated that mitochondrial dysfunction is not a mechanism of BA-induced toxicity in HepaRG cells.


Subject(s)
Bile Acids and Salts/toxicity , Mitochondria/drug effects , Cell Line , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/physiology , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/metabolism
7.
J Biol Chem ; 294(10): 3720-3734, 2019 03 08.
Article in English | MEDLINE | ID: mdl-30598509

ABSTRACT

Peroxisome proliferator-activated receptor α (PPARα) is a transcriptional regulator of lipid metabolism. GW7647 is a potent PPARα agonist that must reach the nucleus to activate this receptor. In cells expressing human fatty acid-binding protein 1 (FABP1), GW7647 treatment increases FABP1's nuclear localization and potentiates GW7647-mediated PPARα activation; GW7647 is less effective in cells that do not express FABP1. To elucidate the underlying mechanism, here we substituted residues in FABP1 known to dictate lipid signaling by other intracellular lipid-binding proteins. Substitutions of Lys-20 and Lys-31 to Ala in the FABP1 helical cap affected neither its nuclear localization nor PPARα activation. In contrast, Ala substitution of Lys-57, Glu-77, and Lys-96, located in the loops adjacent to the ligand-binding portal region, abolished both FABP1 nuclear localization and GW7647-induced PPARα activation but had little effect on GW7647-FABP1 binding affinity. Using solution NMR spectroscopy, we determined the WT FABP1 structure and analyzed the dynamics in the apo and GW7647-bound structures of both the WT and the K57A/E77A/K96A triple mutant. We found that GW7647 binding causes little change in the FABP1 backbone, but solvent exposes several residues in the loops around the portal region, including Lys-57, Glu-77, and Lys-96. These residues also become more solvent-exposed upon binding of FABP1 with the endogenous PPARα agonist oleic acid. Together with previous observations, our findings suggest that GW7647 binding stabilizes a FABP1 conformation that promotes its interaction with PPARα. We conclude that full PPARα agonist activity of GW7647 requires FABP1-dependent transport and nuclear localization processes.


Subject(s)
Butyrates/pharmacology , Fatty Acid-Binding Proteins/chemistry , Fatty Acid-Binding Proteins/metabolism , PPAR alpha/agonists , Phenylurea Compounds/pharmacology , Butyrates/metabolism , Fatty Acid-Binding Proteins/genetics , Humans , Ligands , Models, Molecular , Mutation , Phenylurea Compounds/metabolism , Protein Conformation/drug effects
8.
Clin Exp Allergy ; 49(5): 636-643, 2019 05.
Article in English | MEDLINE | ID: mdl-30657219

ABSTRACT

BACKGROUND: Beta-lactams allergy is the most commonly reported drug allergy and constitutes an important health problem. We previously showed the pre-existence of a naïve CD4+ T cell repertoire for benzylpenicillin (BP) coupled to human serum albumin (HSA) but little is known about the naïve CD8+ T cell repertoire specific for BP. OBJECTIVE: The purpose of this work was to identify naïve CD8+ T cells specific for BP and to explore mechanisms dictating their activation. METHODS: Co-cultures were established with naïve CD8+ T cells and autologous dendritic cells (DCs) loaded with HSA-BP or free BP. T cells were restimulated once a week with autologous DCs loaded with HSA-BP or BP. The specific CD8+ T cell response was measured using an IFN-γ ELISpot assay. RESULTS: When using free BP, we were able to detect a naïve CD8+ T cell repertoire for BP in the 6 out of 7 tested healthy donors. However, our results showed that HSA-BP was recognized by naïve CD8+ T cells in only one donor out of five tested healthy donors. Using free BP, we evidenced its binding to cellular proteins in DCs that was concentration dependent and was correlated with BP-specific CD8+ T cell activation. Moreover, the BP-specific CD8+ cell response was MHC class I-dependent and required intracellular processing and proteasome activity. CONCLUSION AND CLINICAL RELEVANCE: This work showed the existence of a naïve CD8+ T cell repertoire for BP when DCs were treated with free BP suggesting that patients could be immunized by haptenated peptides from cellular proteins generated in antigen-presenting cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Disease Susceptibility , Drug Hypersensitivity/immunology , Penicillin G/adverse effects , CD8-Positive T-Lymphocytes/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Susceptibility/immunology , Drug Hypersensitivity/diagnosis , Enzyme-Linked Immunospot Assay , Epitopes, T-Lymphocyte/immunology , Haptens , Histocompatibility Antigens Class I/immunology , Humans , Lymphocyte Activation/immunology , Lymphocyte Count , Proteasome Endopeptidase Complex/metabolism
9.
Toxicol Sci ; 167(2): 307-321, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30371856

ABSTRACT

Current gaps in drug safety sciences can result from the inability (1) to identify hazard across multiple target organs, (2) to predict and risk assess with certainty against drug safety liabilities for the major target organs, (3) to optimally manage and mitigate against drug safety liabilities, and (4) to apply principles of governance on the generation, integration, and use of experimental data. Translational safety assessment to evaluate several target-organ drug toxicities can only be partially achieved by use of current in vitro and in vivo test systems. What remains to be tackled necessitates the deployment of in vitro-human-relevant test systems to address human specific or selective forms of toxicities. Nevertheless, such models may only address in part some of the requirements in today's armament of biomedical tools essential for improving the discovery of drug candidates. Refinement of in silico tools, Target Safety Assessment and a greater understanding of mechanistic insights of toxicities might provide future opportunities to better identify drug safety liabilities. The increasing diversity of drug modalities present further challenges for nonclinical and clinical development requiring further research to develop suitable test systems and technologies. Our ability to optimally manage and mitigate safety risk will come from the greater refinement of safety margin estimates, provision and use of human-relevant safety biomarkers, and understanding of the translation from in silico, in vitro, and in vivo studies to human. An improvement of governance frameworks and standards at all levels within organizations, national, and international, can only help facilitate drug discovery and development programs.


Subject(s)
Drug Discovery/trends , Drug-Related Side Effects and Adverse Reactions/prevention & control , Biomarkers , Computer Simulation , Humans , Risk Assessment , Risk Management
10.
Toxicol Sci ; 167(2): 385-396, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30247740

ABSTRACT

The fluoroquinolone trovafloxacin (TVX) is associated with a high risk of drug-induced liver injury (DILI). Although part of the liver damage by TVX+TNF relies on neutrophils, we have recently demonstrated that liver recruitment of monocytes and neutrophils is delayed by TVX. Here we show that the delayed leukocyte recruitment is caused by a combination of effects which are linked to the capacity of TVX to block the hemichannel pannexin 1. TVX inhibited find-me signal release in apoptotic HepG2 hepatocytes, decelerated freshly isolated human neutrophils toward IL-8 and f-MLF, and decreased the liver expression of ICAM-1. In blood of TVX+TNF-treated mice, we observed an accumulation of activated neutrophils despite an increased MIP-2 release by the liver. Depletion of monocytes and neutrophils caused increased serum concentrations of TNF, IL-6, and MIP-2 in TVX-treated mice as well as in mice treated with the fluoroquinolone levofloxacin, known to have a lower DILI-inducing profile. This supports the idea that early leukocyte recruitment regulates inflammation. In conclusion, disrupted regulation by leukocytes appears to constitute a fundamental step in the onset of TVX-induced liver injury, acting in concert with the capability of TVX to induce hepatocyte cell death. Interference of leukocyte-mediated regulation of inflammation represents a novel mechanism to explain the onset of DILI.


Subject(s)
Anti-Infective Agents/toxicity , Chemical and Drug Induced Liver Injury/immunology , Fluoroquinolones/toxicity , Naphthyridines/toxicity , Neutrophil Infiltration/drug effects , Neutrophils/drug effects , Nucleotides/metabolism , Tumor Necrosis Factor-alpha/toxicity , Animals , Chemical and Drug Induced Liver Injury/metabolism , Connexins/metabolism , Hep G2 Cells , Humans , Inflammation , Intercellular Adhesion Molecule-1/metabolism , Male , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Neutrophil Infiltration/immunology , Neutrophils/immunology
11.
ALTEX ; 36(2): 289-313, 2019.
Article in English | MEDLINE | ID: mdl-30570669

ABSTRACT

Investigative Toxicology describes the de-risking and mechanistic elucidation of toxicities, supporting early safety decisions in the pharmaceutical industry. Recently, Investigative Toxicology has contributed to a shift in pharmaceutical toxicology, from a descriptive to an evidence-based, mechanistic discipline. This was triggered by high costs and low throughput of Good Laboratory Practice in vivo studies, and increasing demands for adhering to the 3R (Replacement, Reduction and Refinement) principles of animal welfare. Outside the boundaries of regulatory toxicology, Investigative Toxicology has the flexibility to embrace new technologies, enhancing translational steps from in silico, in vitro to in vivo mechanistic understanding to eventually predict human response. One major goal of Investigative Toxicology is improving preclinical decisions, which coincides with the concept of animal-free safety testing. Currently, compounds under preclinical development are being discarded due to the use of inappropriate animal models. Progress in Investigative Toxicology could lead to humanized in vitro test systems and the development of medicines less reliant on animal tests. To advance this field a group of 14 European-based leaders from the pharmaceutical industry founded the Investigative Toxicology Leaders Forum (ITLF), an open, non-exclusive and pre-competitive group that shares knowledge and experience. The ITLF collaborated with the Centre for Alternatives to Animal Testing Europe (CAAT-Europe) to organize an "Investigative Toxicology Think-Tank", which aimed to enhance the interaction with experts from academia and regulatory bodies in the field. Summarizing the topics and discussion of the workshop, this article highlights Investigative Toxicology's position by identifying key challenges and perspectives.


Subject(s)
Drug Discovery , Drug Evaluation, Preclinical/trends , Toxicology/trends , Animal Testing Alternatives , Animals , Computer Simulation , Drug Industry , Europe , Humans , In Vitro Techniques , Risk Assessment
12.
Toxicol In Vitro ; 52: 189-194, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29933103

ABSTRACT

Human OATP1B1 is highly expressed at the basolateral membrane of the hepatocyte. It plays an important role in the sodium-independent transport of bile acids and bile salts and contributes to the systemic clearance of many drugs. In this study, the interaction of at least one representative of all major chemical classes of bile acids and bile salts, which include the bile acid chenodeoxycholate (CDC), monovalent (amidated) bile salts glycochenodeoxycholate (GCDC), taurochenodeoxycholate (TCDC) and taurocholate (TC), a sulfated bile acid 3-sulfo-chenodeoxycholate (3S-CDC) and a divalent (amidated and sulfated) bile salt 3-sulfo-glycolithocholate (3S-GLC) were tested with OATP1B1 overexpressed in HEK293 cells. All bile acid derivatives except for CDC showed an efficient transport by OATP1B1. 3S-GLC gave the lowest KM (0.708 ±â€¯0.125 µM) and 3S-CDC showed the highest Vmax value (158 ±â€¯87.3 pmol/mg protein/min). The ranking of Clint values (3S-GLC > 3S-CDC > TCDC > GCDC > TC) also showed a preference for sulfated derivatives. In summary, human OATP1B1 transports sulfate esters of bile acids and bile salts more efficiently than monovalent bile salts.


Subject(s)
Bile Acids and Salts/metabolism , Liver-Specific Organic Anion Transporter 1/metabolism , HEK293 Cells , Humans
13.
J Appl Toxicol ; 38(5): 753-765, 2018 05.
Article in English | MEDLINE | ID: mdl-29377180

ABSTRACT

Idiosyncratic drug-induced liver injury (iDILI) has a poorly understood pathogenesis. However, iDILI is often associated with inflammatory stress signals in human patients as well as animal models. Tumor necrosis factor (TNF) and neutrophils play a key role in onset of trovafloxacin (TVX)-induced iDILI, but the exact role of neutrophils and other leukocytes remains to be defined. We therefore set out to study the kinetics of immunological changes during the development of TVX-induced iDILI in the established murine model of acute liver injury induced by administration of TVX and TNF. Initially, TNF stimulated the appearance of leukocytes, in particular neutrophils, into the liver of TVX-treated mice, but even more so in control mice treated with the non-DILI inducing analogue levofloxacin (LVX) or saline as vehicle (Veh). This difference was apparent at 2 hours after TNF administration, but at 4 hours, the relative neutrophil amounts were reduced again in Veh- and LVX-treated mice whereas the amounts in TVX-treated mice remained at the same increased level as at 2 hours. The influx of monocytes/macrophages, which was unaffected in Veh- and LVX-treated mice was markedly reduced or even absent in TVX-treated mice. Unlike controls, mice receiving TVX + TNF display severe hepatotoxicity with clear pathology and apoptosis, coagulated hepatic vessels and increased alanine aminotransferase levels and interleukin 6/10 ratios. Findings indicate that TVX delays the acute influx of neutrophils and monocytes/macrophages. Considering their known anti-inflammatory functions, the disruption of influx of these innate immune cells may hamper the resolution of initial cytotoxic effects of TVX and thus contribute to liver injury development.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Fluoroquinolones/toxicity , Monocytes/drug effects , Naphthyridines/toxicity , Neutrophils/drug effects , Tumor Necrosis Factor-alpha/toxicity , Alanine Transaminase/blood , Animals , Chemical and Drug Induced Liver Injury/immunology , Chemical and Drug Induced Liver Injury/pathology , Cytokines/blood , Flow Cytometry , Leukocytes/drug effects , Levofloxacin/pharmacology , Liver/drug effects , Liver/immunology , Liver/pathology , Male , Mice , Mice, Inbred C57BL
14.
Toxicol In Vitro ; 46: 189-193, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29024779

ABSTRACT

The transport of bile acids facilitated by NTCP is an important factor in establishing bile flow. In this study, we examine the kinetics associated with human NTCP-dependent transport of two quantitatively important bile acids comprising the human bile acid pool, chenodeoxycholic acid and glycine-chenodeoxycholate, and secondary bile salt, 3-sulfo-glycolithocholate of potential toxicological significance. The study employed human NTCP overexpressing Chinese Hamster Ovary cells and results compared with taurocholate, a prototypical bile salt commonly used in transporter studies. GCDC and 3S-GLC but not CDCA were transported by NTCP. The efficient uptake of GCDC, TCA and 3S-GLC by NTCP enabled the determination of kinetics. GCDC displayed a lower KM (0.569±0.318µM) than TCA (6.44±3.83µM) and 3S-GLC (3.78±1.17µM). The apparent CLint value for GCDC was 20-fold greater (153±53µl/mg protein/min) than the apparent CLint for TCA (6.92±4.72µl/mg protein/min) and apparent CLint for 3S-GLC (8.05±1.33µl/mg protein/min). These kinetic results provide important complementary data on the substrate selectivity and specificity of NTCP to transport bile acids. NTCP transports GCDC with greater efficiency than TCA and has the same efficacy for 3S-GLC and TCA.


Subject(s)
Bile Acids and Salts/metabolism , Organic Anion Transporters, Sodium-Dependent/metabolism , Symporters/metabolism , Animals , Biological Transport , CHO Cells , Cricetinae , Cricetulus , Hepatocytes/metabolism , Humans , Kinetics , Organic Anion Transporters, Sodium-Dependent/genetics , Sodium , Symporters/genetics
15.
Toxicol Sci ; 161(1): 48-57, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29029322

ABSTRACT

A dynamic model based on ordinary differential equations that describes uptake, basolateral and canalicular export of taurocholic acid (TCA) in human HepaRG cells is presented. The highly reproducible inter-assay experimental data were used to reliably estimate model parameters. Primary human hepatocytes were similarly evaluated to establish a mathematical model, but with notably higher inter-assay differences in TCA clearance and bile canaliculi dynamics. By use of the HepaRG cell line, the simultaneous TCA clearance associated to basolateral uptake, canalicular and sinusoidal efflux, was predicted. The mathematical model accurately reproduced the dose-dependent inhibition of TCA clearance in the presence and absence of the prototypical cholestatic drugs cyclosporine A (CsA) and chlorpromazine. Rapid inhibition of TCA clearance and recovery were found to be major characteristics of CsA. Conversely, the action of chlorpromazine was described by slow onset of inhibition relative to inhibition of TCA clearance by CsA. The established mathematical model, validated by the use of these 2 prototypical cholestatic drugs and the integration of bile canalicular dynamics, provides an important development for the further study of human hepatobiliary function, through simultaneous temporal and vectorial membrane transport of bile acids in drug-induced cholestasis.


Subject(s)
Bile Acids and Salts/metabolism , Hepatocytes/metabolism , Models, Theoretical , Biological Transport , Cell Line , Chlorpromazine/metabolism , Chlorpromazine/pharmacology , Cyclosporine/metabolism , Cyclosporine/pharmacology , Hepatobiliary Elimination , Hepatocytes/drug effects , Humans , Kinetics , Primary Cell Culture
16.
Expert Opin Drug Metab Toxicol ; 13(7): 767-782, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28604124

ABSTRACT

INTRODUCTION: The liver is an important target for drug-induced toxicities. Early detection of hepatotoxic drugs requires use of well-characterized test systems, yet current knowledge, gaps and limitations of tests employed remains an important issue for drug development. Areas Covered: The current state of the science, understanding and application of test systems in use for the detection of drug-induced cytotoxicity, mitochondrial toxicity, cholestasis and inflammation is summarized. The test systems highlighted herein cover mostly in vitro and some in vivo models and endpoint measurements used in the assessment of small molecule toxic liabilities. Opportunities for research efforts in areas necessitating the development of specific tests and improved mechanistic understanding are highlighted. Expert Opinion: Use of in vitro test systems for safety optimization will remain a core activity in drug discovery. Substantial inroads have been made with a number of assays established for human Drug-induced Liver Injury. There nevertheless remain significant gaps with a need for improved in vitro tools and novel tests to address specific mechanisms of human Drug-Induced Liver Injury. Progress in these areas will necessitate not only models fit for application, but also mechanistic understanding of how chemical insult on the liver occurs in order to identify translational and quantifiable readouts for decision-making.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Drug Discovery/methods , Toxicity Tests/methods , Animals , Chemical and Drug Induced Liver Injury/physiopathology , Drug Design , Drug-Related Side Effects and Adverse Reactions/diagnosis , Humans , Models, Biological , Risk Assessment/methods
17.
Arch Toxicol ; 91(3): 1385-1400, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27344343

ABSTRACT

Assessing the potential of a new drug to cause drug-induced liver injury (DILI) is a challenge for the pharmaceutical industry. We therefore determined whether cell models currently used in safety assessment (HepG2, HepaRG, Upcyte and primary human hepatocytes in conjunction with basic but commonly used endpoints) are actually able to distinguish between novel chemical entities (NCEs) with respect to their potential to cause DILI. A panel of thirteen compounds (nine DILI implicated and four non-DILI implicated in man) were selected for our study, which was conducted, for the first time, across multiple laboratories. None of the cell models could distinguish faithfully between DILI and non-DILI compounds. Only when nominal in vitro concentrations were adjusted for in vivo exposure levels were primary human hepatocytes (PHH) found to be the most accurate cell model, closely followed by HepG2. From a practical perspective, this study revealed significant inter-laboratory variation in the response of PHH, HepG2 and Upcyte cells, but not HepaRG cells. This variation was also observed to be compound dependent. Interestingly, differences between donors (hepatocytes), clones (HepG2) and the effect of cryopreservation (HepaRG and hepatocytes) were less important than differences between the cell models per se. In summary, these results demonstrate that basic cell health endpoints will not predict hepatotoxic risk in simple hepatic cells in the absence of pharmacokinetic data and that a multicenter assessment of more sophisticated signals of molecular initiating events is required to determine whether these cells can be incorporated in early safety assessment.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Toxicity Tests, Acute/methods , Cells, Cultured , Cryopreservation , Hep G2 Cells/drug effects , Hepatocytes/drug effects , Humans , Reproducibility of Results , Toxicity Tests, Acute/standards
18.
Hepatology ; 65(2): 710-721, 2017 02.
Article in English | MEDLINE | ID: mdl-27775817

ABSTRACT

Current preclinical drug testing does not predict some forms of adverse drug reactions in humans. Efforts at improving predictability of drug-induced tissue injury in humans include using stem cell technology to generate human cells for screening for adverse effects of drugs in humans. The advent of induced pluripotent stem cells means that it may ultimately be possible to develop personalized toxicology to determine interindividual susceptibility to adverse drug reactions. However, the complexity of idiosyncratic drug-induced liver injury means that no current single-cell model, whether of primary liver tissue origin, from liver cell lines, or derived from stem cells, adequately emulates what is believed to occur during human drug-induced liver injury. Nevertheless, a single-cell model of a human hepatocyte which emulates key features of a hepatocyte is likely to be valuable in assessing potential chemical risk; furthermore, understanding how to generate a relevant hepatocyte will also be critical to efforts to build complex multicellular models of the liver. Currently, hepatocyte-like cells differentiated from stem cells still fall short of recapitulating the full mature hepatocellular phenotype. Therefore, we convened a number of experts from the areas of preclinical and clinical hepatotoxicity and safety assessment, from industry, academia, and regulatory bodies, to specifically explore the application of stem cells in hepatotoxicity safety assessment and to make recommendations for the way forward. In this short review, we particularly discuss the importance of benchmarking stem cell-derived hepatocyte-like cells to their terminally differentiated human counterparts using defined phenotyping, to make sure the cells are relevant and comparable between labs, and outline why this process is essential before the cells are introduced into chemical safety assessment. (Hepatology 2017;65:710-721).


Subject(s)
Chemical and Drug Induced Liver Injury/diagnosis , Drug-Related Side Effects and Adverse Reactions/diagnosis , Hepatocytes/drug effects , Pluripotent Stem Cells/drug effects , Toxicity Tests , Cells, Cultured/drug effects , Hepatocytes/metabolism , Humans , In Vitro Techniques , Pluripotent Stem Cells/metabolism , Predictive Value of Tests , Sensitivity and Specificity
19.
Biomed Res Int ; 2016: 9737920, 2016.
Article in English | MEDLINE | ID: mdl-27689095

ABSTRACT

Drug-induced liver injury (DILI) is a major cause of late-stage clinical drug attrition, market withdrawal, black-box warnings, and acute liver failure. Consequently, it has been an area of focus for toxicologists and clinicians for several decades. In spite of considerable efforts, limited improvements in DILI prediction have been made and efforts to improve existing preclinical models or develop new test systems remain a high priority. While prediction of intrinsic DILI has improved, identifying compounds with a risk for idiosyncratic DILI (iDILI) remains extremely challenging because of the lack of a clear mechanistic understanding and the multifactorial pathogenesis of idiosyncratic drug reactions. Well-defined clinical diagnostic criteria and risk factors are also missing. This paper summarizes key data interpretation challenges, practical considerations, model limitations, and the need for an integrated risk assessment. As demonstrated through selected initiatives to address other types of toxicities, opportunities exist however for improvement, especially through better concerted efforts at harmonization of current, emerging and novel in vitro systems or through the establishment of strategies for implementation of preclinical DILI models across the pharmaceutical industry. Perspectives on the incorporation of newer technologies and the value of precompetitive consortia to identify useful practices are also discussed.

20.
Arch Toxicol ; 90(12): 2979-3003, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27659300

ABSTRACT

The current test systems employed by pharmaceutical industry are poorly predictive for drug-induced liver injury (DILI). The 'MIP-DILI' project addresses this situation by the development of innovative preclinical test systems which are both mechanism-based and of physiological, pharmacological and pathological relevance to DILI in humans. An iterative, tiered approach with respect to test compounds, test systems, bioanalysis and systems analysis is adopted to evaluate existing models and develop new models that can provide validated test systems with respect to the prediction of specific forms of DILI and further elucidation of mechanisms. An essential component of this effort is the choice of compound training set that will be used to inform refinement and/or development of new model systems that allow prediction based on knowledge of mechanisms, in a tiered fashion. In this review, we focus on the selection of MIP-DILI training compounds for mechanism-based evaluation of non-clinical prediction of DILI. The selected compounds address both hepatocellular and cholestatic DILI patterns in man, covering a broad range of pharmacologies and chemistries, and taking into account available data on potential DILI mechanisms (e.g. mitochondrial injury, reactive metabolites, biliary transport inhibition, and immune responses). Known mechanisms by which these compounds are believed to cause liver injury have been described, where many if not all drugs in this review appear to exhibit multiple toxicological mechanisms. Thus, the training compounds selection offered a valuable tool to profile DILI mechanisms and to interrogate existing and novel in vitro systems for the prediction of human DILI.


Subject(s)
Chemical and Drug Induced Liver Injury/prevention & control , Computational Biology/methods , Drugs, Investigational/adverse effects , Evidence-Based Medicine , Expert Systems , Liver/drug effects , Models, Biological , Adaptive Immunity/drug effects , Animals , Artificial Intelligence , Chemical and Drug Induced Liver Injury/immunology , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/physiopathology , Drugs, Investigational/chemistry , Drugs, Investigational/classification , Drugs, Investigational/pharmacology , Hepatobiliary Elimination/drug effects , Humans , Liver/immunology , Liver/metabolism , Liver/physiopathology , Lysosomes/drug effects , Lysosomes/metabolism , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Molecular Structure , Oxidative Stress/drug effects , Severity of Illness Index
SELECTION OF CITATIONS
SEARCH DETAIL
...