Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Clin Pharmacol Ther ; 115(3): 525-534, 2024 03.
Article in English | MEDLINE | ID: mdl-38065572

ABSTRACT

Clinical investigation of emvododstat for the treatment of solid tumors was halted after two patients who were heavily treated with other anticancer therapies experienced drug-induced liver failure. However, preclinical investigations supported that emvododstat at lower doses might be effective in treating acute myeloid leukemia (AML) and against severe acute respiratory syndrome-coronavirus 2 as a dihydroorotate dehydrogenase inhibitor. Therefore, a quantitative systems toxicology model, DILIsym, was used to predict liver safety of the proposed dosing of emvododstat in AML clinical trials. In vitro mechanistic toxicity data of emvododstat and its desmethyl metabolite were integrated with in vivo exposure within DILIsym to predict hepatotoxicity responses in a simulated human population. DILIsym simulations predicted alanine aminotransferase elevations observed in prior emvododstat clinical trials in patients with solid tumors, but not in the prospective AML clinical trial with the proposed dosing regimens. Exposure predictions based on physiologically-based pharmacokinetic modeling suggested that reduced doses of emvododstat would produce clinical exposures that would be efficacious to treat AML. In the AML clinical trial, only eight patients experienced aminotransferase elevations, all of which were mild (grade 1), all resolving within a short period of time, and no patient showed symptoms of hepatotoxicity, confirming the prospective prediction of liver safety. Overall, retrospective DILIsym simulations adequately predicted the liver safety liabilities of emvododstat in solid tumor trials and prospective simulations predicted the liver safety of reduced doses in an AML clinical trial. The modeling was critical to enabling regulatory approval to proceed with the AML clinical trial wherein the predicted liver safety was confirmed.


Subject(s)
Carbamates , Carbazoles , Chemical and Drug Induced Liver Injury , Drug-Related Side Effects and Adverse Reactions , Leukemia, Myeloid, Acute , Humans , Retrospective Studies , Leukemia, Myeloid, Acute/drug therapy , Chemical and Drug Induced Liver Injury/etiology
2.
Int J Mol Sci ; 24(11)2023 Jun 02.
Article in English | MEDLINE | ID: mdl-37298645

ABSTRACT

Biologics address a range of unmet clinical needs, but the occurrence of biologics-induced liver injury remains a major challenge. Development of cimaglermin alfa (GGF2) was terminated due to transient elevations in serum aminotransferases and total bilirubin. Tocilizumab has been reported to induce transient aminotransferase elevations, requiring frequent monitoring. To evaluate the clinical risk of biologics-induced liver injury, a novel quantitative systems toxicology modeling platform, BIOLOGXsym™, representing relevant liver biochemistry and the mechanistic effects of biologics on liver pathophysiology, was developed in conjunction with clinically relevant data from a human biomimetic liver microphysiology system. Phenotypic and mechanistic toxicity data and metabolomics analysis from the Liver Acinus Microphysiology System showed that tocilizumab and GGF2 increased high mobility group box 1, indicating hepatic injury and stress. Tocilizumab exposure was associated with increased oxidative stress and extracellular/tissue remodeling, and GGF2 decreased bile acid secretion. BIOLOGXsym simulations, leveraging the in vivo exposure predicted by physiologically-based pharmacokinetic modeling and mechanistic toxicity data from the Liver Acinus Microphysiology System, reproduced the clinically observed liver signals of tocilizumab and GGF2, demonstrating that mechanistic toxicity data from microphysiology systems can be successfully integrated into a quantitative systems toxicology model to identify liabilities of biologics-induced liver injury and provide mechanistic insights into observed liver safety signals.


Subject(s)
Biological Products , Chemical and Drug Induced Liver Injury, Chronic , Chemical and Drug Induced Liver Injury , Humans , Biological Products/pharmacology , Biomimetics , Chemical and Drug Induced Liver Injury/etiology , Liver
3.
Clin Pharmacol Ther ; 113(2): 275-297, 2023 02.
Article in English | MEDLINE | ID: mdl-35429164

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD), representing a clinical spectrum ranging from nonalcoholic fatty liver (NAFL) to nonalcoholic steatohepatitis (NASH), is rapidly evolving into a global pandemic. Patients with NAFLD are burdened with high rates of metabolic syndrome-related comorbidities resulting in polypharmacy. Therefore, it is crucial to gain a better understanding of NAFLD-mediated changes in drug disposition and efficacy/toxicity. Despite extensive clinical pharmacokinetic data in cirrhosis, current knowledge concerning pharmacokinetic alterations in NAFLD, particularly at different stages of disease progression, is relatively limited. In vitro-to-in vivo extrapolation coupled with physiologically based pharmacokinetic and pharmacodynamic (IVIVE-PBPK/PD) modeling offers a promising approach for optimizing pharmacologic predictions while refining and reducing clinical studies in this population. Use of IVIVE-PBPK to predict intra-organ drug concentrations at pharmacologically relevant sites of action is particularly advantageous when it can be linked to pharmacodynamic effects. Quantitative systems pharmacology/toxicology (QSP/QST) modeling can be used to translate pharmacokinetic and pharmacodynamic data from PBPK/PD models into clinically relevant predictions of drug response and toxicity. In this review, a detailed summary of NAFLD-mediated alterations in human physiology relevant to drug absorption, distribution, metabolism, and excretion (ADME) is provided. The application of literature-derived physiologic parameters and ADME-associated protein abundance data to inform virtual NAFLD population development and facilitate PBPK/PD, QSP, and QST predictions is discussed along with current limitations of these methodologies and knowledge gaps. The proposed methodologic framework offers great potential for meaningful prediction of pharmacological outcomes in patients with NAFLD and can inform both drug development and clinical practice for this population.


Subject(s)
Metabolic Syndrome , Non-alcoholic Fatty Liver Disease , Humans , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Liver Cirrhosis , Comorbidity , Disease Progression , Liver/metabolism
4.
Front Pharmacol ; 13: 1085621, 2022.
Article in English | MEDLINE | ID: mdl-36733378

ABSTRACT

Inhibition of the canalicular phospholipid floppase multidrug resistance protein 3 (MDR3) has been implicated in cholestatic drug-induced liver injury (DILI), which is clinically characterized by disrupted bile flow and damage to the biliary epithelium. Reduction in phospholipid excretion, as a consequence of MDR3 inhibition, decreases the formation of mixed micelles consisting of bile acids and phospholipids in the bile duct, resulting in a surplus of free bile acids that can damage the bile duct epithelial cells, i.e., cholangiocytes. Cholangiocytes may compensate for biliary increases in bile acid monomers via the cholehepatic shunt pathway or bicarbonate secretion, thereby influencing viability or progression to toxicity. To address the unmet need to predict drug-induced bile duct injury in humans, DILIsym, a quantitative systems toxicology model of DILI, was extended by representing key features of the bile duct, cholangiocyte functionality, bile acid and phospholipid disposition, and cholestatic hepatotoxicity. A virtual, healthy representative subject and population (n = 285) were calibrated and validated utilizing a variety of clinical data. Sensitivity analyses were performed for 1) the cholehepatic shunt pathway, 2) biliary bicarbonate concentrations and 3) modes of MDR3 inhibition. Simulations showed that an increase in shunting may decrease the biliary bile acid burden, but raise the hepatocellular concentrations of bile acids. Elevating the biliary concentration of bicarbonate may decrease bile acid shunting, but increase bile flow rate. In contrast to competitive inhibition, simulations demonstrated that non-competitive and mixed inhibition of MDR3 had a profound impact on phospholipid efflux, elevations in the biliary bile acid-to-phospholipid ratio, cholangiocyte toxicity, and adaptation pathways. The model with its extended bile acid homeostasis representation was furthermore able to predict DILI liability for compounds with previously studied interactions with bile acid transport. The cholestatic liver injury submodel in DILIsym accounts for several processes pertinent to bile duct viability and toxicity and hence, is useful for predictions of MDR3 inhibition-mediated cholestatic DILI in humans.

5.
Regul Toxicol Pharmacol ; 118: 104788, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33153971

ABSTRACT

In 2019, the California Office of Environmental Health Hazard Assessment (OEHHA) initiated a review of the carcinogenic hazard potential of acetaminophen. The objective of the analysis herein was to inform this review by assessing whether variability in patient baseline characteristics (e.g. baseline glutathione (GSH) levels, pharmacokinetics, and capacity of hepatic antioxidants) leads to potential differences in carcinogenic hazard potential at different dosing schemes: maximum labeled doses of 4 g/day, repeated doses above the maximum labeled dose (>4-12 g/day), and acute overdoses of acetaminophen (>15 g). This was achieved by performing simulations of acetaminophen exposure in thousands of diverse virtual patients scenarios using the DILIsym® Quantitative Systems Toxicology (QST) model. Simulations included assessments of the dose and exposure response for toxicity and mode of cell death based on evaluations of the kinetics of changes of: GSH, N-acetyl-p-benzoquinone-imine (NAPQI), protein adducts, mitochondrial dysfunction, and hepatic cell death. Results support that, at therapeutic doses, cellular GSH binds to NAPQI providing sufficient buffering capacity to limit protein adduct formation and subsequent oxidative stress. Simulations evaluating repeated high-level supratherapeutic exposures or acute overdoses indicate that cell death precedes DNA damage that could result in carcinogenicity and thus acetaminophen does not present a carcinogenicity hazard to humans at any dose.


Subject(s)
Acetaminophen/adverse effects , Analgesics, Non-Narcotic/administration & dosage , Carcinogenicity Tests , Chemical and Drug Induced Liver Injury/etiology , Computer Simulation , Liver Neoplasms/chemically induced , Liver/drug effects , Acetaminophen/pharmacokinetics , Analgesics, Non-Narcotic/pharmacokinetics , Antioxidants/metabolism , Cell Death/drug effects , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , DNA Damage , Dose-Response Relationship, Drug , Glutathione/metabolism , Humans , Liver/metabolism , Liver/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Mitochondria, Liver/pathology , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Risk Assessment
6.
Regul Toxicol Pharmacol ; 118: 104801, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33039518

ABSTRACT

In 2019 the California Office of Environmental Health Hazard Assessment (OEHHA) initiated a review of the carcinogenic hazard potential of acetaminophen, including an assessment of the long-term rodent carcinogenicity and tumor initiation/promotion studies. The objective of the analysis herein was to inform this review process with a weight-of-evidence assessment of these studies and an assessment of the relevance of these models to humans. In most of the 14 studies, there were no increases in the incidences of tumors in any organ system. In the few studies in which an increase in tumor incidence was observed, there were factors such as absence of a dose response and a rodent-specific tumor supporting that these findings are not relevant to human hazard identification. In addition, we performed qualitative analysis and quantitative simulations of the exposures to acetaminophen and its metabolites and its toxicity profile; the data support that the rodent models are toxicologically relevant to humans. The preclinical carcinogenicity results are consistent with the broader weight of evidence assessment and evaluations of multiple international health authorities supporting that acetaminophen is not a carcinogenic hazard.


Subject(s)
Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Carcinogenicity Tests , Cell Transformation, Neoplastic/chemically induced , Neoplasms/chemically induced , Acetaminophen/pharmacokinetics , Analgesics, Non-Narcotic/pharmacokinetics , Animals , Biotransformation , Dose-Response Relationship, Drug , Female , Humans , Male , Mice , Rats , Risk Assessment , Species Specificity , Toxicokinetics
7.
Pharm Res ; 36(3): 48, 2019 Feb 07.
Article in English | MEDLINE | ID: mdl-30734107

ABSTRACT

PURPOSE: Macrolide antibiotics are commonly prescribed treatments for drug-resistant bacterial infections; however, many macrolides have been shown to cause liver enzyme elevations and one macrolide, telithromycin, has been pulled from the market by its provider due to liver toxicity. This work seeks to assess the mechanisms responsible for the toxicity of macrolide antibiotics. METHODS: Five macrolides were assessed in in vitro systems designed to test for bile acid transporter inhibition, mitochondrial dysfunction, and oxidative stress. The macrolides were then represented in DILIsym, a quantitative systems pharmacology (QST) model of drug-induced liver injury, placing the in vitro results in context with each compound's predicted liver exposure and known biochemistry. RESULTS: DILIsym results suggest that solithromycin and clarithromycin toxicity is primarily due to inhibition of the mitochondrial electron transport chain (ETC) while erythromycin toxicity is primarily due to bile acid transporter inhibition. Telithromycin and azithromycin toxicity was not predicted by DILIsym and may be caused by mechanisms not currently incorporated into DILIsym or by unknown metabolite effects. CONCLUSIONS: The mechanisms responsible for toxicity can be significantly different within a class of drugs, despite the structural similarity among the drugs. QST modeling can provide valuable insight into the nature of these mechanistic differences.


Subject(s)
Anti-Bacterial Agents/adverse effects , Chemical and Drug Induced Liver Injury/etiology , Liver/drug effects , Macrolides/adverse effects , Models, Biological , Animals , CHO Cells , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Cricetulus , Hep G2 Cells , Humans , Liver/metabolism , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Stress/drug effects
8.
Clin Pharmacol Ther ; 104(5): 916-932, 2018 11.
Article in English | MEDLINE | ID: mdl-30137645

ABSTRACT

Bile salt export pump (BSEP) inhibition has emerged as an important mechanism that may contribute to the initiation of human drug-induced liver injury (DILI). Proactive evaluation and understanding of BSEP inhibition is recommended in drug discovery and development to aid internal decision making on DILI risk. BSEP inhibition can be quantified using in vitro assays. When interpreting assay data, it is important to consider in vivo drug exposure. Currently, this can be undertaken most effectively by consideration of total plasma steady state drug concentrations (Css,plasma ). However, because total drug concentrations are not predictive of pharmacological effect, the relationship between total exposure and BSEP inhibition is not causal. Various follow-up studies can aid interpretation of in vitro BSEP inhibition data and may be undertaken on a case-by-case basis. BSEP inhibition is one of several mechanisms by which drugs may cause DILI, therefore, it should be considered alongside other mechanisms when evaluating possible DILI risk.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 11/antagonists & inhibitors , Bile/metabolism , Chemical and Drug Induced Liver Injury/etiology , Drug Development/methods , Drug Discovery/methods , Liver/drug effects , Membrane Transport Modulators/toxicity , ATP Binding Cassette Transporter, Subfamily B, Member 11/chemistry , ATP Binding Cassette Transporter, Subfamily B, Member 11/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 11/metabolism , Animals , Cells, Cultured , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/prevention & control , Computer Simulation , Computer-Aided Design , Drug Design , Humans , In Vitro Techniques , Liver/metabolism , Membrane Transport Modulators/chemistry , Models, Biological , Protein Conformation , Risk Assessment , Risk Factors , Structure-Activity Relationship
9.
Toxicol Sci ; 166(1): 123-130, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30060248

ABSTRACT

CKA, a chemokine receptor antagonist intended for treating inflammatory conditions, produced dose-dependent hepatotoxicity in rats but advanced into the clinic where single doses of CKA up to 600 mg appeared safe in humans. Because existing toxicological platforms used during drug development are not perfectly predictive, a quantitative systems toxicology model investigated the hepatotoxic potential of CKA in humans and rats through in vitro assessments of CKA on mitochondrial respiration, oxidative stress, and bile acid transporters. DILIsym predicted that single doses of CKA caused serum ALT >3xULN in a subset of the simulated rat population, while single doses in a simulated human population did not produce serum ALT elevations. Species differences were largely attributed to differences in liver exposure, but increased sensitivity to inhibition of mitochondrial respiration in the rat also contributed. We conclude that mechanistic modeling can elucidate species differences in the hepatotoxic potential of drug candidates.


Subject(s)
Carboxylic Acids/toxicity , Carrier Proteins/antagonists & inhibitors , Chemical and Drug Induced Liver Injury/etiology , Indoles/toxicity , Membrane Glycoproteins/antagonists & inhibitors , Models, Biological , Oxidative Stress/drug effects , Receptors, Chemokine/antagonists & inhibitors , Adult , Animals , Carboxylic Acids/administration & dosage , Carboxylic Acids/pharmacokinetics , Chemical and Drug Induced Liver Injury/metabolism , Dose-Response Relationship, Drug , Double-Blind Method , Female , Hep G2 Cells , Humans , Indoles/administration & dosage , Indoles/pharmacokinetics , Liver Function Tests , Male , Middle Aged , Rats, Wistar , Species Specificity , Tissue Distribution
10.
J Pharm Sci ; 106(9): 2401-2406, 2017 09.
Article in English | MEDLINE | ID: mdl-28465154

ABSTRACT

The intracellular unbound inhibitor concentration ([I]unbound,cell) is the most relevant concentration for predicting the inhibition of hepatic efflux transporters. However, the intracellular unbound fraction of inhibitor in hepatocytes (fu,cell,inhibitor) is not routinely determined. Studies are needed to evaluate the benefit of measuring fu,cell,inhibitor and using [I]unbound,cell versus intracellular total inhibitor concentration ([I]total,cell) when predicting inhibitory effects. This study examined the benefit of using [I]unbound,cell to predict hepatocellular bile acid disposition. Cellular total concentrations of taurocholate ([TCA]total,cell), a prototypical bile acid, were simulated using pharmacokinetic parameters estimated from sandwich-cultured human hepatocytes. The effect of various theoretical inhibitors was simulated by varying ([I]total,cell/ half maximal inhibitory concentration [IC50]) values. In addition, the fold change was calculated as the simulated [TCA]total,cell when fu,cell,inhibitor = 1 divided by the simulated [TCA]total,cell when fu,cell,inhibitor = 0.5-0.01. The lowest ([I]total,cell/IC50) value leading to a >2-fold change in [TCA]total,cell was chosen as a cutoff, and a framework was developed to categorize risk inhibitors for which the measurement of fu,cell,inhibitor is optimal. Fifteen compounds were categorized, 5 of which were compared with experimental observations. Future work is needed to evaluate this framework based on additional experimental data. In conclusion, the benefit of measuring fu,cell,inhibitor to predict hepatic efflux transporter-mediated drug-bile acid interactions can be determined a priori.


Subject(s)
Bile Acids and Salts/metabolism , Hepatocytes/metabolism , Membrane Transport Proteins/metabolism , Pharmaceutical Preparations/metabolism , Biological Transport/drug effects , Computer Simulation , Drug Interactions , Hepatocytes/drug effects , Humans , Models, Biological , Protein Binding , Taurocholic Acid/metabolism
11.
J Pharmacol Exp Ther ; 358(2): 324-33, 2016 08.
Article in English | MEDLINE | ID: mdl-27233294

ABSTRACT

Transporter-mediated alterations in bile acid disposition may have significant toxicological implications. Current methods to predict interactions are limited by the interplay of multiple transporters, absence of protein in the experimental system, and inaccurate estimates of inhibitor concentrations. An integrated approach was developed to predict altered bile acid disposition due to inhibition of multiple transporters using the model bile acid taurocholate (TCA). TCA pharmacokinetic parameters were estimated by mechanistic modeling using sandwich-cultured human hepatocyte data with protein in the medium. Uptake, basolateral efflux, and biliary clearance estimates were 0.63, 0.034, and 0.074 mL/min/g liver, respectively. Cellular total TCA concentrations (Ct,Cells) were selected as the model output based on sensitivity analysis. Monte Carlo simulations of TCA Ct,Cells in the presence of model inhibitors (telmisartan and bosentan) were performed using inhibition constants for TCA transporters and inhibitor concentrations, including cellular total inhibitor concentrations ([I]t,cell) or unbound concentrations, and cytosolic total or unbound concentrations. For telmisartan, the model prediction was accurate with an average fold error (AFE) of 0.99-1.0 when unbound inhibitor concentration ([I]u) was used; accuracy dropped when total inhibitor concentration ([I]t) was used. For bosentan, AFE was 1.2-1.3 using either [I]u or [I]t This difference was evaluated by sensitivity analysis of the cellular unbound fraction of inhibitor (fu,cell,inhibitor), which revealed higher sensitivity of fu,cell,inhibitor for predicting TCA Ct,Cells when inhibitors exhibited larger ([I]t,cell/IC50) values. In conclusion, this study demonstrated the applicability of a framework to predict hepatocellular bile acid concentrations due to drug-mediated inhibition of transporters using mechanistic modeling and cytosolic or cellular unbound concentrations.


Subject(s)
Hepatocytes/cytology , Hepatocytes/metabolism , Membrane Transport Modulators/pharmacology , Membrane Transport Proteins/metabolism , Models, Biological , Taurocholic Acid/metabolism , Benzimidazoles/pharmacology , Benzoates/pharmacology , Biliary Tract/drug effects , Biliary Tract/metabolism , Cells, Cultured , Cytosol/drug effects , Cytosol/metabolism , Dose-Response Relationship, Drug , Humans , Monte Carlo Method , Telmisartan
12.
J Pharm Sci ; 105(2): 443-459, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26869411

ABSTRACT

Sandwich-cultured hepatocytes (SCH) are metabolically competent and have proper localization of basolateral and canalicular transporters with functional bile networks. Therefore, this cellular model is a unique tool that can be used to estimate biliary excretion of compounds. SCH have been used widely to assess hepatobiliary disposition of endogenous and exogenous compounds and metabolites. Mechanistic modeling based on SCH data enables estimation of metabolic and transporter-mediated clearances, which can be used to construct physiologically based pharmacokinetic models for prediction of drug disposition and drug-drug interactions in humans. In addition to pharmacokinetic studies, SCH also have been used to study cytotoxicity and perturbation of biological processes by drugs and hepatically generated metabolites. Human SCH can provide mechanistic insights underlying clinical drug-induced liver injury (DILI). In addition, data generated in SCH can be integrated into systems pharmacology models to predict potential DILI in humans. In this review, applications of SCH in studying hepatobiliary drug disposition and bile acid-mediated DILI are discussed. An example is presented to show how data generated in the SCH model were used to establish a quantitative relationship between intracellular bile acids and cytotoxicity, and how this information was incorporated into a systems pharmacology model for DILI prediction.


Subject(s)
Cell Culture Techniques/methods , Chemical and Drug Induced Liver Injury/metabolism , Hepatocytes/metabolism , Pharmaceutical Preparations/metabolism , Animals , Biological Transport/physiology , Drug Liberation/physiology , Hepatocytes/drug effects , Humans , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/physiology , Pharmaceutical Preparations/administration & dosage , Tissue Distribution/drug effects , Tissue Distribution/physiology
13.
J Pharmacol Exp Ther ; 353(2): 415-23, 2015 May.
Article in English | MEDLINE | ID: mdl-25711339

ABSTRACT

The bile salt export pump (BSEP) plays an important role in bile acid excretion. Impaired BSEP function may result in liver injury. Bile acids also undergo basolateral efflux, but the relative contributions of biliary (CLBile) versus basolateral efflux (CLBL) clearance to hepatocellular bile acid excretion have not been determined. In the present study, taurocholic acid (TCA; a model bile acid) disposition was characterized in human and rat sandwich-cultured hepatocytes (SCH) combined with pharmacokinetic modeling. In human SCH, biliary excretion of TCA predominated (CLBile = 0.14 ± 0.04 ml/min per g liver; CLBL = 0.042 ± 0.019 ml/min per g liver), whereas CLBile and CLBL contributed approximately equally to TCA hepatocellular excretion in rat SCH (CLBile = 0.34 ± 0.07 ml/min per g liver; CLBL = 0.26 ± 0.07 ml/min per g liver). Troglitazone decreased TCA uptake, CLBile, and CLBL; membrane vesicle assays revealed for the first time that the major metabolite, troglitazone sulfate, was a noncompetitive inhibitor of multidrug resistance-associated protein 4, a basolateral bile acid efflux transporter. Simulations revealed that decreased CLBile led to a greater increase in hepatic TCA exposure in human than in rat SCH. A decrease in both excretory pathways (CLBile and CLBL) exponentially increased hepatic TCA in both species, suggesting that 1) drugs that inhibit both pathways may have a greater risk for hepatotoxicity, and 2) impaired function of an alternate excretory pathway may predispose patients to hepatotoxicity when drugs that inhibit one pathway are administered. Simulations confirmed the protective role of uptake inhibition, suggesting that a drug's inhibitory effects on bile acid uptake also should be considered when evaluating hepatotoxic potential. Overall, the current study precisely characterized basolateral efflux of TCA, revealed species differences in hepatocellular TCA efflux pathways, and provided insights about altered hepatic bile acid exposure when multiple transport pathways are impaired.


Subject(s)
Bile Ducts/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism , Taurocholic Acid/metabolism , Adult , Animals , Bile Ducts/drug effects , Biological Transport/drug effects , Cells, Cultured , Chemical and Drug Induced Liver Injury/pathology , Chromans/pharmacology , Dehydroepiandrosterone/metabolism , Female , Hepatocytes/drug effects , Humans , Male , Middle Aged , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Rats , Species Specificity , Sulfuric Acid Esters/pharmacology , Taurocholic Acid/pharmacokinetics , Thiazolidinediones/pharmacology , Troglitazone
14.
Front Pharmacol ; 5: 240, 2014.
Article in English | MEDLINE | ID: mdl-25426072

ABSTRACT

Inhibition of the bile salt export pump (BSEP) has been linked to incidence of drug-induced liver injury (DILI), presumably by the accumulation of toxic bile acids in the liver. We have previously constructed and validated a model of bile acid disposition within DILIsym®, a mechanistic model of DILI. In this paper, we use DILIsym® to simulate the DILI response of the hepatotoxic BSEP inhibitors bosentan and CP-724,714 and the non-hepatotoxic BSEP inhibitor telmisartan in humans in order to explore whether we can predict that hepatotoxic BSEP inhibitors can cause bile acid accumulation to reach toxic levels. We also simulate bosentan in rats in order to illuminate potential reasons behind the lack of toxicity in rats compared to the toxicity observed in humans. DILIsym® predicts that bosentan, but not telmisartan, will cause mild hepatocellular ATP decline and serum ALT elevation in a simulated population of humans. The difference in hepatotoxic potential between bosentan and telmisartan is consistent with clinical observations. However, DILIsym® underpredicts the incidence of bosentan toxicity. DILIsym® also predicts that bosentan will not cause toxicity in a simulated population of rats, and that the difference between the response to bosentan in rats and in humans is primarily due to the less toxic bile acid pool in rats. Our simulations also suggest a potential synergistic role for bile acid accumulation and mitochondrial electron transport chain (ETC) inhibition in producing the observed toxicity in CP-724,714, and suggest that CP-724,714 metabolites may also play a role in the observed toxicity. Our work also compares the impact of competitive and noncompetitive BSEP inhibition for CP-724,714 and demonstrates that noncompetitive inhibition leads to much greater bile acid accumulation and potential toxicity. Our research demonstrates the potential for mechanistic modeling to contribute to the understanding of how bile acid transport inhibitors cause DILI.

15.
Drug Metab Dispos ; 42(7): 1219-26, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24799397

ABSTRACT

Inhibition of bile acid transport by troglitazone (TGZ) and its major metabolite, TGZ sulfate (TS), may lead to hepatocellular accumulation of toxic bile acids; TS accumulation and hepatotoxicity may be associated with impaired TS biliary excretion. This study evaluated the impact of impaired transport of breast cancer resistance protein (Bcrp) and multidrug resistance-associated protein 2 (Mrp2) on the hepatobiliary disposition of generated metabolites, TS and TGZ glucuronide (TG). Sandwich-cultured hepatocytes (SCH) from Mrp2-deficient (TR(-)) rats in combination with Bcrp knockdown using RNA interference were employed. The biliary excretion index (BEI) of generated TS was not significantly altered by impaired Bcrp (20.9 to 21.1%) and/or Mrp2 function (24.4% and 17.5% in WT and TR(-) rat SCH, respectively). Thus, loss-of-function of Mrp2 and/or Bcrp do not appear to be risk factors for increased hepatocellular TS accumulation in rats, potentially because of a compensatory transporter(s) that excretes TS into bile. Further investigations revealed that the compensatory TS biliary transporter was not the bile salt export pump (Bsep) or P-glycoprotein (P-gp). Interestingly, TGZ sulfation was significantly decreased in TR(-) compared with WT rat SCH (total recovery: 2.8 versus 5.0% of TGZ dose), resulting in decreased hepatocellular TS accumulation, even though sulfotransferase activity in TR(-) rat hepatocyte S9 fraction was similar. Hepatocellular TG accumulation was significantly increased in TR(-) compared with WT rat SCH due to increased glucuronidation and negligible TG biliary excretion. These data emphasize that the interplay between metabolite formation and excretion determines hepatocellular exposure to generated metabolites such as TS and TG.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Chromans/metabolism , Hepatocytes/drug effects , Hypoglycemic Agents/metabolism , Thiazolidinediones/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Animals , Cells, Cultured , Hepatocytes/metabolism , Male , Rats , Rats, Wistar , Troglitazone
16.
Mol Pharm ; 11(3): 766-75, 2014 Mar 03.
Article in English | MEDLINE | ID: mdl-24410402

ABSTRACT

Breast cancer resistance protein (BCRP) and multidrug resistance-associated protein 2 (MRP2) are members of the ATP binding cassette (ABC) transporter family located in the canalicular membrane of hepatocytes that mediate biliary excretion of many drugs and endogenous compounds. BCRP and MRP2 have overlapping substrate profiles. Predicting drug disposition in the setting of altered transport function has important clinical significance. This investigation was designed to establish an in vitro model system to evaluate the impact of impaired Mrp2 and Bcrp function on hepatobiliary drug disposition. To achieve Bcrp knockdown by RNA interference (RNAi), sandwich-cultured hepatocytes (SCH) from Mrp2-deficient (TR(-)) and wild-type (WT) rats were infected with adenoviral vectors to express shRNA targeting Bcrp (Ad-siBcrp) at multiplicity of infection (MOI) of 1-10. MOI of 5 was identified as optimal. At MOI of 5, viral infection as well as WT or TR(-) status was statistically significant predictors of the rosuvastatin (RSV) biliary excretion index (BEI), consistent with the known role of Bcrp and Mrp2 in the biliary excretion of RSV in vivo in rats. Relative to WT rat SCH, marginal mean BEI (%) of RSV in TR(-) rat SCH decreased by 28.6 (95% CI: 5.8-51.3). Ad-siBcrp decreased marginal mean BEI (%) of RSV by 13.3 (7.5-9.1) relative to SCH infected with adenoviral vectors expressing a nontargeting shRNA (Ad-siNT). The BEI of RSV was almost ablated in TR(-) rat SCH with Bcrp knockdown (5.9 ± 3.0%) compared to Ad-siNT-infected WT rat SCH (45.4 ± 6.6%). These results demonstrated the feasibility of Bcrp knockdown in TR(-) rat SCH as an in vitro system to assess the impact of impaired Bcrp and Mrp2 function. At MOI of 5, viral infection had minimal effects on RSV total accumulation, but significantly decreased marginal mean taurocholate total accumulation (pmol/mg of protein) and BEI (%) by 9.9 (7.0-12.8) and 7.5 (3.7-11.3), respectively, relative to noninfected SCH. These findings may be due to off-target effects on hepatic bile acid transporters, even though no changes in protein expression levels of the hepatic bile acid transporters were observed. This study established a strategy for optimization of the knockdown system, and demonstrated the potential use of RNAi in SCH as an in vitro tool to predict altered hepatobiliary drug disposition when canalicular transporters are impaired.


Subject(s)
ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/physiology , Biliary Tract/drug effects , Fluorobenzenes/pharmacology , Hepatocytes/drug effects , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Taurocholic Acid/pharmacology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Biliary Tract/cytology , Biliary Tract/metabolism , Biological Transport , Blotting, Western , Cells, Cultured , Detergents/pharmacology , Hepatocytes/cytology , Hepatocytes/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Male , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Rosuvastatin Calcium
17.
Drug Metab Dispos ; 42(4): 665-74, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24154606

ABSTRACT

Impaired hepatic bile acid export may contribute to development of cholestatic drug-induced liver injury (DILI). The multidrug resistance-associated proteins (MRP) 3 and 4 are postulated to be compensatory hepatic basolateral bile acid efflux transporters when biliary excretion by the bile salt export pump (BSEP) is impaired. BSEP inhibition is a risk factor for cholestatic DILI. This study aimed to characterize the relationship between MRP3, MRP4, and BSEP inhibition and cholestatic potential of drugs. The inhibitory effect of 88 drugs (100 µM) on MRP3- and MRP4-mediated substrate transport was measured in membrane vesicles. Drugs selected for investigation included 50 BSEP non-inhibitors (24 non-cholestatic; 26 cholestatic) and 38 BSEP inhibitors (16 non-cholestatic; 22 cholestatic). MRP4 inhibition was associated with an increased risk of cholestatic potential among BSEP non-inhibitors. In this group, for each 1% increase in MRP4 inhibition, the odds of the drug being cholestatic increased by 3.1%. Using an inhibition cutoff of 21%, which predicted a 50% chance of cholestasis, 62% of cholestatic drugs inhibited MRP4 (P < 0.05); in contrast, only 17% of non-cholestatic drugs were MRP4 inhibitors. Among BSEP inhibitors, MRP4 inhibition did not provide additional predictive value of cholestatic potential; almost all BSEP inhibitors were also MRP4 inhibitors. Inclusion of pharmacokinetic predictor variables (e.g., maximal unbound concentration in plasma) in addition to percent MRP4 inhibition in logistic regression models did not improve cholestasis prediction. Association of cholestasis with percent MRP3 inhibition was not statistically significant, regardless of BSEP-inhibition status. Inhibition of MRP4, in addition to BSEP, may be a risk factor for the development of cholestatic DILI.


Subject(s)
ATP-Binding Cassette Transporters/antagonists & inhibitors , Chemical and Drug Induced Liver Injury/metabolism , Cholestasis/metabolism , Drug-Related Side Effects and Adverse Reactions/metabolism , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Pharmaceutical Preparations , ATP Binding Cassette Transporter, Subfamily B, Member 11 , ATP-Binding Cassette Transporters/genetics , Chemical and Drug Induced Liver Injury/etiology , Cholestasis/chemically induced , Drug-Related Side Effects and Adverse Reactions/etiology , HEK293 Cells , Humans , Liver/drug effects , Liver/metabolism , Logistic Models , Multidrug Resistance-Associated Proteins/genetics , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/chemistry , Predictive Value of Tests , Risk Factors , Transfection
18.
J Pharmacol Exp Ther ; 347(3): 727-36, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24023367

ABSTRACT

Transporters responsible for hepatic uptake and biliary clearance (CLBile) of rosuvastatin (RSV) have been well characterized. However, the contribution of basolateral efflux clearance (CLBL) to hepatic and systemic exposure of RSV is unknown. Additionally, the appropriate design of in vitro hepatocyte efflux experiments to estimate CLBile versus CLBL remains to be established. A novel uptake and efflux protocol was developed in sandwich-cultured hepatocytes (SCH) to achieve desired tight junction modulation while maintaining cell viability. Subsequently, studies were conducted to determine the role of CLBL in the hepatic disposition of RSV using SCH from wild-type (WT) and multidrug resistance-associated protein 2 (Mrp2)-deficient (TR(-)) rats in the absence and presence of the P-glycoprotein and breast cancer resistance protein (Bcrp) inhibitor elacridar (GF120918). RSV CLBile was nearly ablated by GF120918 in TR(-) SCH, confirming that Mrp2 and Bcrp are responsible for the majority of RSV CLBile. Pharmacokinetic modeling revealed that CLBL and CLBile represent alternative elimination routes with quantitatively similar contributions to the overall hepatocellular excretion of RSV in rat SCH under baseline conditions (WT SCH in the absence of GF120918) and also in human SCH. Membrane vesicle experiments revealed that RSV is a substrate of MRP4 (Km = 21 ± 7 µM, Vmax = 1140 ± 210 pmol/min per milligram of protein). Alterations in MRP4-mediated RSV CLBL due to drug-drug interactions, genetic polymorphisms, or disease states may lead to changes in hepatic and systemic exposure of RSV, with implications for the safety and efficacy of this commonly used medication.


Subject(s)
Bile/metabolism , Fluorobenzenes/pharmacokinetics , Hepatocytes/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Pyrimidines/pharmacokinetics , Sulfonamides/pharmacokinetics , Acridines/pharmacology , Adenosine Triphosphate/physiology , Algorithms , Animals , Carrier Proteins/metabolism , Cell Membrane/metabolism , Cells, Cultured , Hepatocytes/drug effects , Humans , L-Lactate Dehydrogenase/metabolism , Male , Models, Biological , Multidrug Resistance-Associated Proteins/metabolism , Plasmids , Rats , Rats, Wistar , Rosuvastatin Calcium , Tetrahydroisoquinolines/pharmacology , Tight Junctions/metabolism
19.
J Pharm Sci ; 102(9): 3037-57, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23653385

ABSTRACT

Drug-induced cholestasis is an important form of acquired liver disease and is associated with significant morbidity and mortality. Bile acids are key signaling molecules, but they can exert toxic responses when they accumulate in hepatocytes. This review focuses on the physiological mechanisms of drug-induced cholestasis associated with altered bile acid homeostasis due to direct (e.g., bile acid transporter inhibition) or indirect (e.g., activation of nuclear receptors, altered function/expression of bile acid transporters) processes. Mechanistic information about the effects of a drug on bile acid homeostasis is important when evaluating the cholestatic potential of a compound, but experimental data often are not available. The relationship between physicochemical properties, pharmacokinetic parameters, and inhibition of the bile salt export pump among 77 cholestatic drugs with different pathophysiological mechanisms of cholestasis (i.e., impaired formation of bile vs. physical obstruction of bile flow) was investigated. The utility of in silico models to obtain mechanistic information about the impact of compounds on bile acid homeostasis to aid in predicting the cholestatic potential of drugs is highlighted.


Subject(s)
Bile Acids and Salts/metabolism , Cholestasis/chemically induced , Cholestasis/metabolism , Animals , Bile Ducts/drug effects , Bile Ducts/metabolism , Bile Ducts/pathology , Biological Transport/drug effects , Carrier Proteins/metabolism , Cholestasis/diagnosis , Cholestasis/pathology , Humans , Liver/drug effects , Liver/metabolism , Liver/pathology , Pharmaceutical Preparations/chemistry , Pharmaceutical Preparations/metabolism , Pharmacokinetics
20.
Biochem Pharmacol ; 84(1): 93-103, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22484313

ABSTRACT

Pregnancy alters the rate and extent of drug metabolism, but little is known about the underlying molecular mechanism. We have found that 17ß-estradiol (E2) upregulates expression of the major drug-metabolizing enzyme CYP2B6 in primary human hepatocytes. Results from promoter reporter assays in HepG2 cells revealed that E2 activates constitutive androstane receptor (CAR) and enhances promoter activity of CYP2B6, for which high concentrations of E2 reached during pregnancy were required. E2 triggered nuclear translocation of CAR in primary rat hepatocytes that were transiently transfected with human CAR as well as in primary human hepatocytes, further confirming transactivation of CAR by E2. E2-activated estrogen receptor (ER) also enhanced CYP2B6 promoter activity. The DNA-binding domain of ER was not required for the induction of CYP2B6 promoter activity by E2, suggesting involvement of a non-classical mechanism of ER action. Results from deletion and mutation assays as well as electrophorectic mobility shift and supershift assays revealed that two AP-1 binding sites (-1782/-1776 and -1664/-1658 of CYP2B6) are critical for ER-mediated activation of the CYP2B6 promoter by E2. Concurrent activation of both ER and CAR by E2 enhanced CYP2B6 expression in a synergistic manner. Our data demonstrate that at high concentrations reached during pregnancy, E2 activates both CAR and ER that synergistically induce CYP2B6 expression. These results illustrate pharmacological activity of E2 that would likely become prominent during pregnancy.


Subject(s)
Aryl Hydrocarbon Hydroxylases/genetics , Estradiol/pharmacology , Estrogens/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Hepatocytes/drug effects , Oxidoreductases, N-Demethylating/genetics , Pregnancy/genetics , Adult , Aryl Hydrocarbon Hydroxylases/metabolism , Binding Sites , Cell Nucleus/metabolism , Chromatin Immunoprecipitation , Chromatography, High Pressure Liquid , Constitutive Androstane Receptor , Cytochrome P-450 CYP2B6 , Dose-Response Relationship, Drug , Electrophoretic Mobility Shift Assay , Estradiol/blood , Estrogens/blood , Female , Gene Expression Profiling , Genes, Reporter , Hep G2 Cells , Hepatocytes/enzymology , Humans , Luciferases/genetics , Middle Aged , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Oxidoreductases, N-Demethylating/metabolism , Pregnancy/blood , Promoter Regions, Genetic , Real-Time Polymerase Chain Reaction , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Tandem Mass Spectrometry , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...