1
|
Yang K, Beaudoin JJ, Howell BA, Mullin J, Amini E, Lai JCK, Gelotte CK, Sista S, Atillasoy E. Quantitative Systems Toxicology Modeling of Acetaminophen Pharmacokinetics and Hepatic Biomarkers After Overdoses of Extended-Release and Immediate-Release Formulations in Adults With Chronic Alcohol Use or Low Glutathione. CPT Pharmacometrics Syst Pharmacol 2025. [PMID: 40365931 DOI: 10.1002/psp4.70045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/02/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Acetaminophen (APAP), an over-the-counter analgesic and antipyretic, can cause hepatotoxicity when ingested in large overdoses. APAP has multiple formulations including immediate-release (IR) and extended-release (ER) preparations. A recently published consensus statement on the management of APAP poisoning indicated that management of APAP-ER overdose is the same as that for APAP-IR overdose. Consistent with this consensus, it was previously reported that quantitative systems toxicology (QST) modeling using DILIsym predicted similar pharmacokinetic (PK) and hepatic biomarker profiles for the APAP-ER and APAP-IR formulations after overdose in healthy adults. Hepatic injury from APAP is caused by the reactive metabolite, N-acetyl-ρ-benzoquinone imine (NAPQI), which is formed predominantly by CYP2E1-mediated metabolism and eliminated by hepatic glutathione. As such, conditions that can increase NAPQI production (e.g., CYP2E1 induction by alcohol) or decrease hepatic glutathione stores (e.g., underling liver disease) may impact PK and susceptibility to hepatotoxicity after overdose of APAP-IR and APAP-ER. In the current study, APAP-IR and APAP-ER models in chronic alcohol users and individuals with low hepatic glutathione were developed and verified within DILIsym. Simulations using verified models predicted similar PK and hepatic biomarker profiles for the APAP-ER and APAP-IR formulations in moderate and excessive chronic alcohol users and adults with low hepatic glutathione levels after single acute overdoses up to ~100 g and repeat supratherapeutic ingestions (up to 7.8 g/day for 10 days). These results further support that approaches to manage APAP-IR overdoses can be applied to manage APAP-ER overdoses in adults with chronic alcohol consumption or lower hepatic glutathione levels.
Collapse
Affiliation(s)
- Kyunghee Yang
- Quantitative Systems Pharmacology Solutions, Simulations Plus Inc., Research Triangle Park, North Carolina, USA
| | - James J Beaudoin
- Quantitative Systems Pharmacology Solutions, Simulations Plus Inc., Research Triangle Park, North Carolina, USA
| | - Brett A Howell
- Quantitative Systems Pharmacology Solutions, Simulations Plus Inc., Research Triangle Park, North Carolina, USA
| | - James Mullin
- Physiologically-Based Pharmacokinetics Solutions, Simulations Plus Inc., Research Triangle Park, North Carolina, USA
| | - Elham Amini
- Physiologically-Based Pharmacokinetics Solutions, Simulations Plus Inc., Research Triangle Park, North Carolina, USA
| | | | | | | | | |
Collapse
|
2
|
Beilmann M, Adkins K, Boonen HCM, Hewitt P, Hu W, Mader R, Moore S, Rana P, Steger-Hartmann T, Villenave R, van Vleet T. Application of new approach methodologies for nonclinical safety assessment of drug candidates. Nat Rev Drug Discov 2025:10.1038/s41573-025-01182-9. [PMID: 40316753 DOI: 10.1038/s41573-025-01182-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2025] [Indexed: 05/04/2025]
Abstract
The development of new approach methodologies (NAMs) and advances with in vitro testing systems have prompted revisions in regulatory guidelines and inspired dedicated in vitro/ex vivo studies for nonclinical safety assessment. This Review by a safety reflection initiative subgroup of the European Federation of Pharmaceutical Industries and Associations (EFPIA)/Preclinical Development Expert Group (PDEG) summarizes the current state and potential application of in vitro studies using human-derived material for safety assessment in drug development. It focuses on case studies from recent projects in which animal models alone proved to be limited or inadequate for safety testing. It further highlights four categories of drug candidates for which alternative in vitro approaches are applicable and discusses progress in using in vitro testing solutions for safety assessment in these categories. Finally, the article highlights new risk assessment strategies, initiatives and consortia promoting the advancement of NAMs. This collective work is meant to encourage the use of NAMs for more human-relevant safety assessment, which should ultimately result in reduced animal testing for drug development.
Collapse
Affiliation(s)
- Mario Beilmann
- Global Nonclinical Safety & DMPK, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany.
| | | | | | - Philip Hewitt
- Chemical and Preclinical Safety, Merck Healthcare KGaA, Darmstadt, Germany
| | - Wenyue Hu
- Vividion Therapeutics, San Diego, CA, USA
| | - Robert Mader
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | | - Payal Rana
- Drug Safety R&D, Pfizer Inc., Groton, CT, USA
| | - Thomas Steger-Hartmann
- Research & Development, Pharmaceuticals, Preclinical Development, Bayer AG, Berlin, Germany
| | - Remi Villenave
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | |
Collapse
|
3
|
Beaudoin JJ, Yang K, Howell BA, Kenz Z, Lakhani VV, Woodhead JL, Lai JCK, Gelotte CK, Sista S, Atillasoy E. Modeling and Simulation of Acetaminophen Pharmacokinetics and Hepatic Biomarkers After Overdoses of Extended-Release and Immediate-Release Formulations in Healthy Adults Using the Quantitative Systems Toxicology Software Platform DILIsym. CPT Pharmacometrics Syst Pharmacol 2025; 14:681-694. [PMID: 39899441 PMCID: PMC12001258 DOI: 10.1002/psp4.13304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/26/2024] [Accepted: 12/16/2024] [Indexed: 02/05/2025] Open
Abstract
Acetaminophen (APAP) has been formulated as immediate-, modified-, and extended-release tablets (APAP-IR, -MR, and -ER, respectively). However, there was concern that APAP-MR previously available in Europe could form a bezoar after a large overdose, leading to delayed absorption and atypical pharmacokinetics (PK) compared to APAP-IR, and that current treatment guidelines developed for APAP overdose to prevent severe hepatotoxicity are inappropriate for APAP-MR. In contrast, APAP-ER caplets available in the United States are designed with an IR layer and an erodible ER layer. Using modeling and simulation, predicted PK and hepatotoxicity biomarkers following various acute overdose and repeated supratherapeutic ingestion (RSTI) scenarios with APAP-IR and APAP-ER were compared to investigate the differences between these two formulations. The existing APAP-IR representation within DILIsym v8A, a quantitative systems toxicology model of drug-induced liver injury, was updated, and an APAP-ER model was developed, using newly acquired in vitro (e.g., tiny-TIMsg) and clinical data. The model and simulated populations (SimPops) representing healthy adults were extensively validated, before simulating PK and three clinically useful hepatic biomarkers after various overdose scenarios. On average, APAP exposure after acute overdose and RSTI in healthy adults was predicted to be slightly lower for APAP-ER compared to APAP-IR, partially due to lower APAP absorption for APAP-ER, while not markedly impacting the expected time course of APAP plasma concentrations. Similar hepatic biomarker profiles were predicted for both APAP formulations. Based on these results, the APAP overdose consensus treatment guidelines updated in 2023 are not further impacted by this report.
Collapse
Affiliation(s)
- James J. Beaudoin
- Quantitative Systems Pharmacology SolutionsSimulations Plus Inc.Research Triangle ParkNorth CarolinaUSA
| | - Kyunghee Yang
- Quantitative Systems Pharmacology SolutionsSimulations Plus Inc.Research Triangle ParkNorth CarolinaUSA
| | - Brett A. Howell
- Quantitative Systems Pharmacology SolutionsSimulations Plus Inc.Research Triangle ParkNorth CarolinaUSA
| | - Zackary Kenz
- Quantitative Systems Pharmacology SolutionsSimulations Plus Inc.Research Triangle ParkNorth CarolinaUSA
| | - Vinal V. Lakhani
- Quantitative Systems Pharmacology SolutionsSimulations Plus Inc.Research Triangle ParkNorth CarolinaUSA
| | - Jeffrey L. Woodhead
- Quantitative Systems Pharmacology SolutionsSimulations Plus Inc.Research Triangle ParkNorth CarolinaUSA
| | | | | | - Sury Sista
- Kenvue Inc.Montgomery TownshipNew JerseyUSA
| | | |
Collapse
|
4
|
He Q, Li M, Ji P, Zheng A, Yao L, Zhu X, Shin JG, Lauschke VM, Han B, Xiang X. Comparison of drug-induced liver injury risk between propylthiouracil and methimazole: A quantitative systems toxicology approach. Toxicol Appl Pharmacol 2024; 491:117064. [PMID: 39122118 DOI: 10.1016/j.taap.2024.117064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/23/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
Propylthiouracil (PTU) and methimazole (MMI), two classical antithyroid agents possess risk of drug-induced liver injury (DILI) with unknown mechanism of action. This study aimed to examine and compare their hepatic toxicity using a quantitative system toxicology approach. The impact of PTU and MMI on hepatocyte survival, oxidative stress, mitochondrial function and bile acid transporters were assessed in vitro. The physiologically based pharmacokinetic (PBPK) models of PTU and MMI were constructed while their risk of DILI was calculated by DILIsym, a quantitative systems toxicology (QST) model by integrating the results from in vitro toxicological studies and PBPK models. The simulated DILI (ALT >2 × ULN) incidence for PTU (300 mg/d) was 21.2%, which was within the range observed in clinical practice. Moreover, a threshold dose of 200 mg/d was predicted with oxidative stress proposed as an important toxic mechanism. However, DILIsym predicted a 0% incidence of hepatoxicity caused by MMI (30 mg/d), suggesting that the toxicity of MMI was not mediated through mechanism incorporated into DILIsym. In conclusion, DILIsym appears to be a practical tool to unveil hepatoxicity mechanism and predict clinical risk of DILI.
Collapse
Affiliation(s)
- Qingfeng He
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Min Li
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Peiying Ji
- Department of Pharmacy, Kong Jiang Hospital of Yangpu District, Shanghai 200093, China
| | - Aole Zheng
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Li Yao
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Xiao Zhu
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jae-Gook Shin
- Department of Pharmacology and Pharmacogenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart 70376, Germany
| | - Bing Han
- Department of Pharmacy, Minhang Hospital, Fudan University, Shanghai 201100, China.
| | - Xiaoqiang Xiang
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai 201203, China.
| |
Collapse
|
5
|
Sang L, Zhou Z, Luo S, Zhang Y, Qian H, Zhou Y, He H, Hao K. An In Silico Platform to Predict Cardiotoxicity Risk of Anti-tumor Drug Combination with hiPSC-CMs Based In Vitro Study. Pharm Res 2024; 41:247-262. [PMID: 38148384 PMCID: PMC10879352 DOI: 10.1007/s11095-023-03644-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 12/15/2023] [Indexed: 12/28/2023]
Abstract
OBJECTIVE Antineoplastic agent-induced systolic dysfunction is a major reason for interruption of anticancer treatment. Although targeted anticancer agents infrequently cause systolic dysfunction, their combinations with chemotherapies remarkably increase the incidence. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) provide a potent in vitro model to assess cardiovascular safety. However, quantitatively predicting the reduction of ejection fraction based on hiPSC-CMs is challenging due to the absence of the body's regulatory response to cardiomyocyte injury. METHODS Here, we developed and validated an in vitro-in vivo translational platform to assess the reduction of ejection fraction induced by antineoplastic drugs based on hiPSC-CMs. The translational platform integrates drug exposure, drug-cardiomyocyte interaction, and systemic response. The drug-cardiomyocyte interaction was implemented as a mechanism-based toxicodynamic (TD) model, which was then integrated into a quantitative system pharmacology-physiological-based pharmacokinetics (QSP-PBPK) model to form a complete translational platform. The platform was validated by comparing the model-predicted and clinically observed incidence of doxorubicin and trastuzumab-induced systolic dysfunction. RESULTS A total of 33,418 virtual patients were incorporated to receive doxorubicin and trastuzumab alone or in combination. For doxorubicin, the QSP-PBPK-TD model successfully captured the overall trend of systolic dysfunction incidences against the cumulative doses. For trastuzumab, the predicted incidence interval was 0.31-2.7% for single-agent treatment and 0.15-10% for trastuzumab-doxorubicin sequential treatment, covering the observations in clinical reports (0.50-1.0% and 1.5-8.3%, respectively). CONCLUSIONS In conclusion, the in vitro-in vivo translational platform is capable of predicting systolic dysfunction incidence almost merely depend on hiPSC-CMs, which could facilitate optimizing the treatment protocol of antineoplastic agents.
Collapse
Affiliation(s)
- Lan Sang
- State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, China
| | - Zhengying Zhou
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, China
| | - Shizheng Luo
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, China
| | - Yicui Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, China
| | - Hongjie Qian
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Ying Zhou
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Hua He
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, China.
| | - Kun Hao
- State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
6
|
Beaudoin JJ, Clemens L, Miedel MT, Gough A, Zaidi F, Ramamoorthy P, Wong KE, Sarangarajan R, Battista C, Shoda LKM, Siler SQ, Taylor DL, Howell BA, Vernetti LA, Yang K. The Combination of a Human Biomimetic Liver Microphysiology System with BIOLOGXsym, a Quantitative Systems Toxicology (QST) Modeling Platform for Macromolecules, Provides Mechanistic Understanding of Tocilizumab- and GGF2-Induced Liver Injury. Int J Mol Sci 2023; 24:9692. [PMID: 37298645 PMCID: PMC10253699 DOI: 10.3390/ijms24119692] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/25/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
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.
Collapse
Affiliation(s)
- James J. Beaudoin
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, Durham, NC 27709, USA (S.Q.S.)
| | - Lara Clemens
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, Durham, NC 27709, USA (S.Q.S.)
| | - Mark T. Miedel
- Department of Computational and Systems Biology, Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15219, USA (A.G.); (D.L.T.)
| | - Albert Gough
- Department of Computational and Systems Biology, Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15219, USA (A.G.); (D.L.T.)
| | - Fatima Zaidi
- Metabolon Inc., Durham, NC 27713, USA (P.R.); (K.E.W.); (R.S.)
| | | | - Kari E. Wong
- Metabolon Inc., Durham, NC 27713, USA (P.R.); (K.E.W.); (R.S.)
| | | | - Christina Battista
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, Durham, NC 27709, USA (S.Q.S.)
| | - Lisl K. M. Shoda
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, Durham, NC 27709, USA (S.Q.S.)
| | - Scott Q. Siler
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, Durham, NC 27709, USA (S.Q.S.)
| | - D. Lansing Taylor
- Department of Computational and Systems Biology, Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15219, USA (A.G.); (D.L.T.)
| | - Brett A. Howell
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, Durham, NC 27709, USA (S.Q.S.)
| | - Lawrence A. Vernetti
- Department of Computational and Systems Biology, Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15219, USA (A.G.); (D.L.T.)
| | - Kyunghee Yang
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, Durham, NC 27709, USA (S.Q.S.)
| |
Collapse
|
7
|
Etzion O, Hamid S, Lurie Y, Gane EJ, Yardeni D, Duehren S, Bader N, Nevo-Shor A, Channa SM, Cotler SJ, Mawani M, Parkash O, Dahari H, Choong I, Glenn JS. Treatment of chronic hepatitis D with peginterferon lambda-the phase 2 LIMT-1 clinical trial. Hepatology 2023; 77:2093-2103. [PMID: 36800850 PMCID: PMC10187621 DOI: 10.1097/hep.0000000000000309] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 12/13/2022] [Accepted: 01/06/2023] [Indexed: 02/22/2023]
Abstract
BACKGROUND AND AIMS HDV infection leads to the most aggressive form of human viral hepatitis for which there is no FDA-approved therapy. PEG IFN-lambda-1a (Lambda) has previously demonstrated a good tolerability profile in HBV and HCV patients compared to PEG IFN-alfa. The goal of Phase 2 LIMT-1 trial was to evaluate the safety and efficacy of Lambda monotherapy in patients with HDV. APPROACH AND RESULTS An open-label study of Lambda 120 or 180 mcg, administered once weekly by subcutaneous injections for 48 weeks, followed by 24 weeks of posttreatment follow-up. Thirty-three patients were allocated to Lambda 180 mcg (n=14) or 120 mcg (n=19). Baseline mean values: HDV RNA 4.1 log10 IU/mL (SD±1.4); ALT 106 IU/L (35-364); and bilirubin 0.5 mg/dL (0.2-1.2). Intention-to-treat rates of virologic response to Lambda 180 mcg and 120 mcg, 24 weeks following treatment cessation were 5 of 14(36%) and 3 of 19 (16%), respectively. The posttreatment response rate of 50% was seen in low BL viral load (≤4 log10) on 180 mcg. Common on-treatment adverse events included flu-like symptoms and elevated transaminase levels. Eight (24%) cases of hyperbilirubinemia with or without liver enzyme elevation, leading to drug discontinuation, were mainly observed in the Pakistani cohort. The clinical course was uneventful, and all responded favorably to dose reduction or discontinuation. CONCLUSIONS Treatment with Lambda in patients with chronic HDV may result in virologic response during and following treatment cessation. Clinical phase 3 development of Lambda for this rare and serious disease is ongoing.
Collapse
Affiliation(s)
- Ohad Etzion
- Department of Gastroenterology and Liver Diseases, Soroka University Medical Center, Beer-Sheva, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Saeed Hamid
- Aga Khan University and Hospital, Karachi, Pakistan
| | - Yoav Lurie
- Shaare Zedek Medical Center, Jerusalem, Israel
| | | | - David Yardeni
- Department of Gastroenterology and Liver Diseases, Soroka University Medical Center, Beer-Sheva, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Sarah Duehren
- Division of Hepatology, Department of Medicine, The Program for Experimental and Theoretical Modeling, Loyola University Medical Center, Maywood, Illinois, USA
| | - Nimrah Bader
- Aga Khan University and Hospital, Karachi, Pakistan
| | - Anat Nevo-Shor
- Department of Gastroenterology and Liver Diseases, Soroka University Medical Center, Beer-Sheva, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Saleh Muhammad Channa
- Department of Gastroenterology, Ghulam Muhammad Mahar Medical College, Sukkur, Pakistan
| | - Scott J. Cotler
- Division of Hepatology, Department of Medicine, The Program for Experimental and Theoretical Modeling, Loyola University Medical Center, Maywood, Illinois, USA
| | - Minaz Mawani
- Aga Khan University and Hospital, Karachi, Pakistan
| | - Om Parkash
- Aga Khan University and Hospital, Karachi, Pakistan
| | - Harel Dahari
- Division of Hepatology, Department of Medicine, The Program for Experimental and Theoretical Modeling, Loyola University Medical Center, Maywood, Illinois, USA
| | - Ingrid Choong
- Eiger BioPharmaceuticals, Inc., Palo Alto, California, USA
| | - Jeffrey S. Glenn
- Departments of Medicine (Division of Gastroenterology and Hepatology) and Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
8
|
Beaudoin JJ, Yang K, Adiwidjaja J, Taneja G, Watkins PB, Siler SQ, Howell BA, Woodhead JL. Investigating bile acid-mediated cholestatic drug-induced liver injury using a mechanistic model of multidrug resistance protein 3 (MDR3) inhibition. Front Pharmacol 2023; 13:1085621. [PMID: 36733378 PMCID: PMC9887159 DOI: 10.3389/fphar.2022.1085621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/23/2022] [Indexed: 01/18/2023] Open
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.
Collapse
Affiliation(s)
- James J. Beaudoin
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, NC, United States
| | - Kyunghee Yang
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, NC, United States
| | - Jeffry Adiwidjaja
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, NC, United States
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Guncha Taneja
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, NC, United States
| | - Paul B. Watkins
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Scott Q. Siler
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, NC, United States
| | - Brett A. Howell
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, NC, United States
| | - Jeffrey L. Woodhead
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, NC, United States
| |
Collapse
|
9
|
Duan M, Guo X, Chen X, Guo M, Zhang M, Xu H, Wang C, Yang Y. Transcriptome analysis reveals hepatotoxicity in zebrafish induced by cyhalofop‑butyl. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2022; 252:106322. [PMID: 36240591 DOI: 10.1016/j.aquatox.2022.106322] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 10/01/2022] [Indexed: 06/16/2023]
Abstract
Cyhalofop‑butyl is a highly effective aryloxyphenoxypropionate herbicide and widely used for weed control in paddy fields. With the increasing residue of cyhalofop‑butyl, it poses a threat to the survival of aquatic organisms. Here, we investigated the effect of cyhalofop‑butyl on zebrafish to explore its potential hepatotoxic mechanism. The results showed that cyhalofop‑butyl induced hepatocyte degeneration, vacuolation and necrosis of larvae after embryonic exposure for 4 days and caused liver atrophy after 5 days. Meanwhile, the activities of enzymes related to liver function were significantly increased by 0.2 mg/L cyhalofop‑butyl and higher, such as alanine transaminase (ALT) and aspartate transaminase (AST). And the contents of triglyceride (TG) involved in lipid metabolism were significantly decreased by 0.4 mg/L cyhalofop-buty. The expression of genes related to liver development was also significantly down-regulated. Furthermore, transcriptome results showed that the pathways involved in metabolism, immune system and endocrine system were significantly impacted, which may be related to hepatoxicity. To sum up, the present study demonstrated the hepatoxicity caused by cyhalofop-buty and its underlying mechanism. The results may provide new insights for the risk of cyhalofop‑butyl to aquatic organisms and new horizons for the pathogenesis of hepatotoxicity.
Collapse
Affiliation(s)
- Manman Duan
- Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Sciences, China Agricultural University, Beijing, 100193, China
| | - Xuanjun Guo
- Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Sciences, China Agricultural University, Beijing, 100193, China
| | - Xiangguang Chen
- Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Sciences, China Agricultural University, Beijing, 100193, China
| | - Mengyu Guo
- Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Sciences, China Agricultural University, Beijing, 100193, China
| | - Mengna Zhang
- Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Sciences, China Agricultural University, Beijing, 100193, China
| | - Hao Xu
- Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Sciences, China Agricultural University, Beijing, 100193, China
| | - Chengju Wang
- Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Sciences, China Agricultural University, Beijing, 100193, China.
| | - Yang Yang
- State Key Laboratory for Biology of Plant Disease and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| |
Collapse
|
10
|
Cheng Y, Straube R, Alnaif AE, Huang L, Leil TA, Schmidt BJ. Virtual Populations for Quantitative Systems Pharmacology Models. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2486:129-179. [PMID: 35437722 DOI: 10.1007/978-1-0716-2265-0_8] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Quantitative systems pharmacology (QSP) places an emphasis on dynamic systems modeling, incorporating considerations from systems biology modeling and pharmacodynamics. The goal of QSP is often to quantitatively predict the effects of clinical therapeutics, their combinations, and their doses on clinical biomarkers and endpoints. In order to achieve this goal, strategies for incorporating clinical data into model calibration are critical. Virtual population (VPop) approaches facilitate model calibration while faced with challenges encountered in QSP model application, including modeling a breadth of clinical therapies, biomarkers, endpoints, utilizing data of varying structure and source, capturing observed clinical variability, and simulating with models that may require more substantial computational time and resources than often found in pharmacometrics applications. VPops are frequently developed in a process that may involve parameterization of isolated pathway models, integration into a larger QSP model, incorporation of clinical data, calibration, and quantitative validation that the model with the accompanying, calibrated VPop is suitable to address the intended question or help with the intended decision. Here, we introduce previous strategies for developing VPops in the context of a variety of therapeutic and safety areas: metabolic disorders, drug-induced liver injury, autoimmune diseases, and cancer. We introduce methodological considerations, prior work for sensitivity analysis and VPop algorithm design, and potential areas for future advancement. Finally, we give a more detailed application example of a VPop calibration algorithm that illustrates recent progress and many of the methodological considerations. In conclusion, although methodologies have varied, VPop strategies have been successfully applied to give valid clinical insights and predictions with the assistance of carefully defined and designed calibration and validation strategies. While a uniform VPop approach for all potential QSP applications may be challenging given the heterogeneity in use considerations, we anticipate continued innovation will help to drive VPop application for more challenging cases of greater scale while developing new rigorous methodologies and metrics.
Collapse
Affiliation(s)
- Yougan Cheng
- QSP and PBPK, Bristol Myers Squibb, Princeton, NJ, USA.,Daiichi Sankyo, Inc., Pennington, NJ, USA
| | - Ronny Straube
- QSP and PBPK, Bristol Myers Squibb, Princeton, NJ, USA
| | - Abed E Alnaif
- QSP and PBPK, Bristol Myers Squibb, Princeton, NJ, USA.,EMD Serono, Billerica, MA, USA
| | - Lu Huang
- QSP and PBPK, Bristol Myers Squibb, Princeton, NJ, USA
| | - Tarek A Leil
- QSP and PBPK, Bristol Myers Squibb, Princeton, NJ, USA.,Daiichi Sankyo, Inc., Pennington, NJ, USA
| | | |
Collapse
|
11
|
Segovia-Zafra A, Di Zeo-Sánchez DE, López-Gómez C, Pérez-Valdés Z, García-Fuentes E, Andrade RJ, Lucena MI, Villanueva-Paz M. Preclinical models of idiosyncratic drug-induced liver injury (iDILI): Moving towards prediction. Acta Pharm Sin B 2021; 11:3685-3726. [PMID: 35024301 PMCID: PMC8727925 DOI: 10.1016/j.apsb.2021.11.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/07/2021] [Accepted: 11/10/2021] [Indexed: 02/08/2023] Open
Abstract
Idiosyncratic drug-induced liver injury (iDILI) encompasses the unexpected harms that prescription and non-prescription drugs, herbal and dietary supplements can cause to the liver. iDILI remains a major public health problem and a major cause of drug attrition. Given the lack of biomarkers for iDILI prediction, diagnosis and prognosis, searching new models to predict and study mechanisms of iDILI is necessary. One of the major limitations of iDILI preclinical assessment has been the lack of correlation between the markers of hepatotoxicity in animal toxicological studies and clinically significant iDILI. Thus, major advances in the understanding of iDILI susceptibility and pathogenesis have come from the study of well-phenotyped iDILI patients. However, there are many gaps for explaining all the complexity of iDILI susceptibility and mechanisms. Therefore, there is a need to optimize preclinical human in vitro models to reduce the risk of iDILI during drug development. Here, the current experimental models and the future directions in iDILI modelling are thoroughly discussed, focusing on the human cellular models available to study the pathophysiological mechanisms of the disease and the most used in vivo animal iDILI models. We also comment about in silico approaches and the increasing relevance of patient-derived cellular models.
Collapse
Affiliation(s)
- Antonio Segovia-Zafra
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid 28029, Spain
| | - Daniel E. Di Zeo-Sánchez
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
| | - Carlos López-Gómez
- Unidad de Gestión Clínica de Aparato Digestivo, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga 29010, Spain
| | - Zeus Pérez-Valdés
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
| | - Eduardo García-Fuentes
- Unidad de Gestión Clínica de Aparato Digestivo, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga 29010, Spain
| | - Raúl J. Andrade
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid 28029, Spain
| | - M. Isabel Lucena
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid 28029, Spain
- Platform ISCIII de Ensayos Clínicos, UICEC-IBIMA, Málaga 29071, Spain
| | - Marina Villanueva-Paz
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
| |
Collapse
|
12
|
Tosca EM, Bartolucci R, Magni P, Poggesi I. Modeling approaches for reducing safety-related attrition in drug discovery and development: a review on myelotoxicity, immunotoxicity, cardiovascular toxicity, and liver toxicity. Expert Opin Drug Discov 2021; 16:1365-1390. [PMID: 34181496 DOI: 10.1080/17460441.2021.1931114] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Introduction:Safety and tolerability is a critical area where improvements are needed to decrease the attrition rates during development of new drug candidates. Modeling approaches, when smartly implemented, can contribute to this aim.Areas covered:The focus of this review was on modeling approaches applied to four kinds of drug-induced toxicities: hematological, immunological, cardiovascular (CV) and liver toxicity. Papers, mainly published in the last 10 years, reporting models in three main methodological categories - computational models (e.g., quantitative structure-property relationships, machine learning approaches, neural networks, etc.), pharmacokinetic-pharmacodynamic (PK-PD) models, and quantitative system pharmacology (QSP) models - have been considered.Expert opinion:The picture observed in the four examined toxicity areas appears heterogeneous. Computational models are typically used in all areas as screening tools in the early stages of development for hematological, cardiovascular and liver toxicity, with accuracies in the range of 70-90%. A limited number of computational models, based on the analysis of drug protein sequence, was instead proposed for immunotoxicity. In the later stages of development, toxicities are quantitatively predicted with reasonably good accuracy using either semi-mechanistic PK-PD models (hematological and cardiovascular toxicity), or fully exploited QSP models (immuno-toxicity and liver toxicity).
Collapse
Affiliation(s)
- Elena M Tosca
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy
| | - Roberta Bartolucci
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy
| | - Paolo Magni
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy
| | - Italo Poggesi
- Clinical Pharmacology & Pharmacometrics, Janssen Research & Development, Beerse, Belgium
| |
Collapse
|
13
|
Zhou Z, Zhu J, Jiang M, Sang L, Hao K, He H. The Combination of Cell Cultured Technology and In Silico Model to Inform the Drug Development. Pharmaceutics 2021; 13:pharmaceutics13050704. [PMID: 34065907 PMCID: PMC8151315 DOI: 10.3390/pharmaceutics13050704] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/29/2021] [Accepted: 05/05/2021] [Indexed: 12/12/2022] Open
Abstract
Human-derived in vitro models can provide high-throughput efficacy and toxicity data without a species gap in drug development. Challenges are still encountered regarding the full utilisation of massive data in clinical settings. The lack of translated methods hinders the reliable prediction of clinical outcomes. Therefore, in this study, in silico models were proposed to tackle these obstacles from in vitro to in vivo translation, and the current major cell culture methods were introduced, such as human-induced pluripotent stem cells (hiPSCs), 3D cells, organoids, and microphysiological systems (MPS). Furthermore, the role and applications of several in silico models were summarised, including the physiologically based pharmacokinetic model (PBPK), pharmacokinetic/pharmacodynamic model (PK/PD), quantitative systems pharmacology model (QSP), and virtual clinical trials. These credible translation cases will provide templates for subsequent in vitro to in vivo translation. We believe that synergising high-quality in vitro data with existing models can better guide drug development and clinical use.
Collapse
Affiliation(s)
- Zhengying Zhou
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; (Z.Z.); (M.J.)
| | - Jinwei Zhu
- State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; (J.Z.); (L.S.)
| | - Muhan Jiang
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; (Z.Z.); (M.J.)
| | - Lan Sang
- State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; (J.Z.); (L.S.)
| | - Kun Hao
- State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; (J.Z.); (L.S.)
- Correspondence: (K.H.); (H.H.)
| | - Hua He
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; (Z.Z.); (M.J.)
- Correspondence: (K.H.); (H.H.)
| |
Collapse
|
14
|
Physiologically Based Pharmacokinetic Modeling to Characterize Acetaminophen Pharmacokinetics and N-Acetyl-p-Benzoquinone Imine (NAPQI) Formation in Non-Pregnant and Pregnant Women. Clin Pharmacokinet 2021; 59:97-110. [PMID: 31347013 PMCID: PMC6994454 DOI: 10.1007/s40262-019-00799-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Background and Objective Little is known about acetaminophen (paracetamol) pharmacokinetics during pregnancy. The aim of this study was to develop a physiologically based pharmacokinetic (PBPK) model to predict acetaminophen pharmacokinetics throughout pregnancy. Methods PBPK models for acetaminophen and its metabolites were developed in non-pregnant and pregnant women. Physiological and enzymatic changes in pregnant women expected to impact acetaminophen pharmacokinetics were considered. Models were evaluated using goodness-of-fit plots and by comparing predicted pharmacokinetic profiles with in vivo pharmacokinetic data. Predictions were performed to illustrate the average concentration at steady state (Css,avg) values, used as an indicator for efficacy, of acetaminophen achieved following administration of 1000 mg every 6 h. Furthermore, as a measurement of potential hepatotoxicity, the molar dose fraction of acetaminophen converted to N-acetyl-p-benzoquinone imine (NAPQI) was estimated. Results PBPK models successfully predicted the pharmacokinetics of acetaminophen and its metabolites in non-pregnant and pregnant women. Predictions resulted in the lowest Css,avg in the third trimester (median [interquartile range]: 4.5 [3.8–5.1] mg/L), while Css,avg was 6.7 [5.9–7.4], 5.6 [4.7–6.3], and 4.9 [4.1–5.5] mg/L in non-pregnant, first trimester, and second trimester populations, respectively. Assuming a constant raised cytochrome P450 2E1 activity throughout pregnancy, the molar dose fraction of acetaminophen converted to NAPQI was highest during the first trimester (median [interquartile range]: 11.0% [9.1–13.4%]), followed by the second (9.0% [7.5–11.0%]) and third trimester (8.2% [6.8–10.1%]), compared with non-pregnant women (7.7% [6.4–9.4%]). Conclusion Acetaminophen exposure is lower in pregnant than in non-pregnant women, and is related to pregnancy duration. Despite these findings, higher dose adjustments cannot be advised yet as it is unknown whether pregnancy affects the toxicodynamics of NAPQI. Information on glutathione abundance during pregnancy and NAPQI in vivo data are required to further refine the presented model. Electronic supplementary material The online version of this article (10.1007/s40262-019-00799-5) contains supplementary material, which is available to authorized users.
Collapse
|
15
|
Sirois JE. Comprehensive investigation evaluating the carcinogenic hazard potential of acetaminophen. Regul Toxicol Pharmacol 2021; 123:104944. [PMID: 33933547 DOI: 10.1016/j.yrtph.2021.104944] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 03/26/2021] [Accepted: 04/26/2021] [Indexed: 11/25/2022]
Abstract
In 2019, the California Office of Environmental Health Hazard Assessment initiated a review of the carcinogenic hazard potential of acetaminophen under Proposition 65. In conjunction with this review, a multidisciplinary team of experts with significant experience in the fields of hazard assessment, acetaminophen mechanism of action, epidemiology, and preclinical and clinical safety performed comprehensive weight of evidence reviews. The reviews evaluate multiple sources of data, including results from preclinical carcinogenicity, genotoxicity, human epidemiology, and mechanistic studies examining biochemical pathways of acetaminophen metabolism. This introductory article summarizes the comprehensive weight of evidence reviews that were performed on the carcinogenicity hazard potential of acetaminophen which are contained in 6 separate companion articles in this issue of Regulatory Toxicology & Pharmacology. Collectively, these results confirm that acetaminophen is not a carcinogenic hazard at any dose level, consistent with previous conclusions of key scientific bodies.
Collapse
Affiliation(s)
- Jay E Sirois
- Consumer Healthcare Products Association, 1625 Eye Street, NW, Suite 600 Washington, DC, 20006, USA.
| |
Collapse
|
16
|
Application of the DILIsym® Quantitative Systems Toxicology drug-induced liver injury model to evaluate the carcinogenic hazard potential of acetaminophen. Regul Toxicol Pharmacol 2020; 118:104788. [PMID: 33153971 DOI: 10.1016/j.yrtph.2020.104788] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/11/2020] [Accepted: 10/04/2020] [Indexed: 12/12/2022]
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.
Collapse
|
17
|
Murray FJ, Monnot AD, Jacobson-Kram D, Cohen SM, Hardisty JF, Bandara SB, Kovochich M, Deore M, Pitchaiyan SK, Gelotte CK, Lai JCK, Atillasoy E, Hermanowski-Vosatka A, Kuffner E, Unice KM, Yang K, Gebremichael Y, Howell BA, Eichenbaum G. A critical review of the acetaminophen preclinical carcinogenicity and tumor promotion data and their implications for its carcinogenic hazard potential. Regul Toxicol Pharmacol 2020; 118:104801. [PMID: 33039518 DOI: 10.1016/j.yrtph.2020.104801] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 09/11/2020] [Accepted: 10/06/2020] [Indexed: 12/22/2022]
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.
Collapse
Affiliation(s)
| | | | | | - Samuel M Cohen
- Havlik-Wall Professor of Oncology, Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | | | | | | | | | | | | | | | | | | | | | | | - Kyunghee Yang
- DILIsym Services Inc., Research Triangle Park, NC, USA
| | | | | | | |
Collapse
|
18
|
Zhang MQ, Chen B, Zhang JP, Chen N, Liu CZ, Hu CQ. Liver toxicity of macrolide antibiotics in zebrafish. Toxicology 2020; 441:152501. [PMID: 32454074 DOI: 10.1016/j.tox.2020.152501] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/01/2020] [Accepted: 05/15/2020] [Indexed: 12/14/2022]
Abstract
Macrolide antibiotics (macrolides) are among the most commonly prescribed antibiotics worldwide and are used for a wide range of infections, but macrolides also expose people to the risk of adverse events include hepatotoxicity. Here, we report the liver toxicity of macrolides with different structures in zebrafish. The absorption, distribution, metabolism, excretion and toxicology (ADMET) parameters of macrolide compounds were predicted and contrasted by utilizing in silico analysis. Fluorescence imaging and Oil Red O stain assays showed all the tested macrolide drugs induced liver degeneration, changed liver size and liver steatosis in larval zebrafish. Through RNA-seq analysis, we found seven co-regulated differentially expressed genes (co-DEGs) associated with metabolism, apoptosis and immune system biological processes, and two co-regulated significant pathways including amino sugar and nucleotide sugar metabolism and apoptosis signaling pathway. We found that only fosab of seven co-DEGs was in the two co-regulated significant pathways. fosab encoded proto-oncogene c-Fos, which was closely associated with liver diseases. The whole-mount in situ hybridization showed high transcription of c-Fos induced by macrolide compounds mainly in the liver region of zebrafish larvae. Cell Counting Kit-8 (CCK-8) and lactate dehydrogenase (LDH) leakage assays revealed that macrolides exerts significant cytotoxic effects on L02 cells. qRT-PCR and western blot analysis demonstrated macrolides also promoted human c-Fos expression in L02 cells. The c-Fos overexpression significantly reduced cell viability by using CCK-8 assay. These data indicate that hepatotoxicity induced by macrolides may be correlated with c-Fos expression activated by these compounds. This study may provide a biomarker for the further investigations on the mechanism of hepatotoxicity induced by macrolide drugs with different structures, and extend our understanding for improving rational clinical application of macrolides.
Collapse
Affiliation(s)
- Miao-Qing Zhang
- Postdoctoral Scientific Research Workstation, China Resources Sanjiu Medical & Pharmaceutical Co., Ltd., Shenzhen 518110, China; Postdoctoral Mobile Research Station, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences & School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100190, China; Shenzhen China Resources Gosun Pharmaceuticals Co., Ltd., Shenzhen 518049, China; Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Bo Chen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Jing-Pu Zhang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Ning Chen
- Shenzhen China Resources Gosun Pharmaceuticals Co., Ltd., Shenzhen 518049, China.
| | - Chun-Zhao Liu
- Postdoctoral Mobile Research Station, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences & School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100190, China.
| | - Chang-Qin Hu
- National Institutes for Food and Drug Control, Beijing 100050, China.
| |
Collapse
|
19
|
Watkins PB. The DILI-sim Initiative: Insights into Hepatotoxicity Mechanisms and Biomarker Interpretation. Clin Transl Sci 2020; 12:122-129. [PMID: 30762301 PMCID: PMC6440570 DOI: 10.1111/cts.12629] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/04/2019] [Accepted: 02/05/2019] [Indexed: 12/16/2022] Open
Abstract
The drug‐induced liver injury (DILI)‐sim Initiative is a public‐private partnership involving scientists from industry, academia, and the US Food and Drug Administration (FDA). The Initiative uses quantitative systems toxicology (QST) to build and refine a model (DILIsym) capable of understanding and predicting liver safety liabilities in new drug candidates and to optimize interpretation of liver safety biomarkers used in clinical studies. Insights gained to date include the observation that most dose‐dependent hepatotoxicity can be accounted for by combinations of just three mechanisms (oxidative stress, interference with mitochondrial respiration, and alterations in bile acid homeostasis) and the importance of noncompetitive inhibition of bile acid transporters. The effort has also provided novel insight into species and interpatient differences in susceptibility, structure‐activity relationships, and the role of nonimmune mechanisms in delayed idiosyncratic hepatotoxicity. The model is increasingly used to evaluate new drug candidates and several clinical trials are underway that will test the model's ability to prospectively predict liver safety. With more refinement, in the future, it may be possible to use the DILIsym predictions to justify reduction in the size of some clinical trials. The mature model could also potentially assist physicians in managing the liver safety of their patients as well as aid in the diagnosis of DILI.
Collapse
Affiliation(s)
- Paul B Watkins
- Institute for Drug Safety Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.,Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
20
|
Longo DM, Shoda LKM, Howell BA, Coric V, Berman RM, Qureshi IA. Assessing Effects of BHV-0223 40 mg Zydis Sublingual Formulation and Riluzole 50 mg Oral Tablet on Liver Function Test Parameters Utilizing DILIsym. Toxicol Sci 2020; 175:292-300. [PMID: 32040174 PMCID: PMC7253195 DOI: 10.1093/toxsci/kfaa019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
For patients with amyotrophic lateral sclerosis who take oral riluzole tablets, approximately 50% experience alanine transaminase (ALT) levels above upper limit of normal (ULN), 8% above 3× ULN, and 2% above 5× ULN. BHV-0223 is a novel 40 mg rapidly sublingually disintegrating (Zydis) formulation of riluzole, bioequivalent to conventional riluzole 50 mg oral tablets, that averts the need for swallowing tablets and mitigates first-pass hepatic metabolism, thereby potentially reducing risk of liver toxicity. DILIsym is a validated multiscale computational model that supports evaluation of liver toxicity risks. DILIsym was used to compare the hepatotoxicity potential of oral riluzole tablets (50 mg BID) versus BHV-0223 (40 mg BID) by integrating clinical data and in vitro toxicity data. In a simulated population (SimPops), ALT levels > 3× ULN were predicted in 3.9% (11/285) versus 1.4% (4/285) of individuals with oral riluzole tablets and sublingual BHV-0223, respectively. This represents a relative risk reduction of 64% associated with BHV-0223 versus conventional riluzole tablets. Mechanistic investigations revealed that oxidative stress was responsible for the predicted ALT elevations. The validity of the DILIsym representation of riluzole and assumptions is supported by its ability to predict rates of ALT elevations for riluzole oral tablets comparable with that observed in clinical data. Combining a mechanistic, quantitative representation of hepatotoxicity with interindividual variability in both susceptibility and liver exposure suggests that sublingual BHV-0223 confers diminished rates of liver toxicity compared with oral tablets of riluzole, consistent with having a lower overall dose of riluzole and bypassing first-pass liver metabolism.
Collapse
Affiliation(s)
- Diane M Longo
- DILIsym Services, Inc., Research Triangle Park, North Carolina 27709
- To whom correspondence should be addressed at 6 Davis Drive, PO Box 12317, Research Triangle Park, NC 27709. E-mail:
| | - Lisl K M Shoda
- DILIsym Services, Inc., Research Triangle Park, North Carolina 27709
| | - Brett A Howell
- DILIsym Services, Inc., Research Triangle Park, North Carolina 27709
| | - Vladimir Coric
- Biohaven Pharmaceuticals, Inc., New Haven, Connecticut 06510
| | - Robert M Berman
- Biohaven Pharmaceuticals, Inc., New Haven, Connecticut 06510
| | - Irfan A Qureshi
- Biohaven Pharmaceuticals, Inc., New Haven, Connecticut 06510
| |
Collapse
|
21
|
Herland A, Maoz BM, Das D, Somayaji MR, Prantil-Baun R, Novak R, Cronce M, Huffstater T, Jeanty SSF, Ingram M, Chalkiadaki A, Benson Chou D, Marquez S, Delahanty A, Jalili-Firoozinezhad S, Milton Y, Sontheimer-Phelps A, Swenor B, Levy O, Parker KK, Przekwas A, Ingber DE. Quantitative prediction of human pharmacokinetic responses to drugs via fluidically coupled vascularized organ chips. Nat Biomed Eng 2020; 4:421-436. [PMID: 31988459 PMCID: PMC8011576 DOI: 10.1038/s41551-019-0498-9] [Citation(s) in RCA: 284] [Impact Index Per Article: 56.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 11/25/2019] [Indexed: 01/15/2023]
Abstract
Analyses of drug pharmacokinetics (PKs) and pharmacodynamics (PDs) performed in animals are often not predictive of drug PKs and PDs in humans, and in vitro PK and PD modelling does not provide quantitative PK parameters. Here, we show that physiological PK modelling of first-pass drug absorption, metabolism and excretion in humans-using computationally scaled data from multiple fluidically linked two-channel organ chips-predicts PK parameters for orally administered nicotine (using gut, liver and kidney chips) and for intravenously injected cisplatin (using coupled bone marrow, liver and kidney chips). The chips are linked through sequential robotic liquid transfers of a common blood substitute by their endothelium-lined channels (as reported by Novak et al. in an associated Article) and share an arteriovenous fluid-mixing reservoir. We also show that predictions of cisplatin PDs match previously reported patient data. The quantitative in-vitro-to-in-vivo translation of PK and PD parameters and the prediction of drug absorption, distribution, metabolism, excretion and toxicity through fluidically coupled organ chips may improve the design of drug-administration regimens for phase-I clinical trials.
Collapse
Affiliation(s)
- Anna Herland
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden
- AIMES, Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Ben M Maoz
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Debarun Das
- CFD Research Corporation, Huntsville, AL, USA
| | | | - Rachelle Prantil-Baun
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Richard Novak
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Michael Cronce
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Tessa Huffstater
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Sauveur S F Jeanty
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Miles Ingram
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Angeliki Chalkiadaki
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - David Benson Chou
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Susan Marquez
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Aaron Delahanty
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Sasan Jalili-Firoozinezhad
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Portugal Graduate Program, Universidade de Lisboa, Lisbon, Portugal
| | - Yuka Milton
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Alexandra Sontheimer-Phelps
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Ben Swenor
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Oren Levy
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Kevin K Parker
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | | | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden.
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
22
|
Zhang JD, Sach-Peltason L, Kramer C, Wang K, Ebeling M. Multiscale modelling of drug mechanism and safety. Drug Discov Today 2020; 25:519-534. [DOI: 10.1016/j.drudis.2019.12.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 12/06/2019] [Accepted: 12/23/2019] [Indexed: 12/19/2022]
|
23
|
Watkins PB. Quantitative Systems Toxicology Approaches to Understand and Predict Drug-Induced Liver Injury. Clin Liver Dis 2020; 24:49-60. [PMID: 31753250 DOI: 10.1016/j.cld.2019.09.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The DILI-sim Initiative is a public-private partnership using quantitative systems toxicology to build a model (DILIsym) capable of understanding and predicting liver safety liabilities in drug candidates. The effort has provided insights into mechanisms underlying dose-dependent drug-induced liver injury (DILI) and interpatient differences in susceptibility to dose-dependent DILI. DILIsym may be useful in identifying drugs capable of causing idiosyncratic hepatotoxicity. DILIsym is used to optimize interpretation of traditional and newer serum biomarkers of DILI. DILIsym results are considered in drug development decisions. In the future, it may be possible to use DILsym predictions to justify reduction in size of some clinical trials.
Collapse
Affiliation(s)
- Paul B Watkins
- Institute for Drug Safety Sciences, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, 6 Davis Drive, PO Box 12137, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
24
|
Woodhead JL, Pellegrini L, Shoda LKM, Howell BA. Comparison of the Hepatotoxic Potential of Two Treatments for Autosomal-Dominant Polycystic Kidney DiseaseUsing Quantitative Systems Toxicology Modeling. Pharm Res 2020; 37:24. [PMID: 31909447 PMCID: PMC6944674 DOI: 10.1007/s11095-019-2726-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 10/18/2019] [Indexed: 12/18/2022]
Abstract
Purpose Autosomal-dominant polycystic kidney disease (ADPKD) is an orphan disease with few current treatment options. The vasopressin V2 receptor antagonist tolvaptan is approved in multiple countries for the treatment of ADPKD, however its use is associated with clinically significant drug-induced liver injury. Methods In prior studies, the potential for hepatotoxicity of tolvaptan was correctly predicted using DILIsym®, a quantitative systems toxicology (QST) mathematical model of drug-induced liver injury. In the current study, we evaluated lixivaptan, another proposed ADPKD treatment and vasopressin V2 receptor antagonist, using DILIsym®. Simulations were conducted that assessed the potential for lixivaptan and its three main metabolites to cause hepatotoxicity due to three injury mechanisms: bile acid accumulation, mitochondrial dysfunction, and oxidative stress generation. Results of these simulations were compared to previously published DILIsym results for tolvaptan. Results No ALT elevations were predicted to occur at the proposed clinical dose for lixivaptan, in contrast to previously published simulation results for tolvaptan. As such, lixivaptan was predicted to have a markedly lower risk of hepatotoxicity compared to tolvaptan with respect to the hepatotoxicity mechanisms represented in DILIsym. Conclusions These results demonstrate the potential for using QST methods to differentiate drugs in the same class for their potential to cause hepatotoxicity. Electronic supplementary material The online version of this article (10.1007/s11095-019-2726-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- J L Woodhead
- DILIsym Services, Inc., a Simulations Plus Company, Research Triangle Park, North Carolina, USA.
| | - L Pellegrini
- Palladio Biosciences, Inc., Newtown, Pennsylvania, USA
| | - L K M Shoda
- DILIsym Services, Inc., a Simulations Plus Company, Research Triangle Park, North Carolina, USA
| | - B A Howell
- DILIsym Services, Inc., a Simulations Plus Company, Research Triangle Park, North Carolina, USA
| |
Collapse
|
25
|
Generaux G, Lakhani VV, Yang Y, Nadanaciva S, Qiu L, Riccardi K, Di L, Howell BA, Siler SQ, Watkins PB, Barton HA, Aleo MD, Shoda LKM. Quantitative systems toxicology (QST) reproduces species differences in PF-04895162 liver safety due to combined mitochondrial and bile acid toxicity. Pharmacol Res Perspect 2019; 7:e00523. [PMID: 31624633 PMCID: PMC6785660 DOI: 10.1002/prp2.523] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 08/17/2019] [Accepted: 08/19/2019] [Indexed: 01/15/2023] Open
Abstract
Many compounds that appear promising in preclinical species, fail in human clinical trials due to safety concerns. The FDA has strongly encouraged the application of modeling in drug development to improve product safety. This study illustrates how DILIsym, a computational representation of liver injury, was able to reproduce species differences in liver toxicity due to PF-04895162 (ICA-105665). PF-04895162, a drug in development for the treatment of epilepsy, was terminated after transaminase elevations were observed in healthy volunteers (NCT01691274). Liver safety concerns had not been raised in preclinical safety studies. DILIsym, which integrates in vitro data on mechanisms of hepatotoxicity with predicted in vivo liver exposure, reproduced clinical hepatotoxicity and the absence of hepatotoxicity observed in the rat. Simulated differences were multifactorial. Simulated liver exposure was greater in humans than rats. The simulated human hepatotoxicity was demonstrated to be due to the interaction between mitochondrial toxicity and bile acid transporter inhibition; elimination of either mechanism from the simulations abrogated injury. The bile acid contribution occurred despite the fact that the IC50 for bile salt export pump (BSEP) inhibition by PF-04895162 was higher (311 µmol/L) than that has been generally thought to contribute to hepatotoxicity. Modeling even higher PF-04895162 liver exposures than were measured in the rat safety studies aggravated mitochondrial toxicity but did not result in rat hepatotoxicity due to insufficient accumulation of cytotoxic bile acid species. This investigative study highlights the potential for combined in vitro and computational screening methods to identify latent hepatotoxic risks and paves the way for similar and prospective studies.
Collapse
Affiliation(s)
- Grant Generaux
- DILIsym Services Inc.Research Triangle ParkNorth Carolina
| | | | - Yuching Yang
- DILIsym Services Inc.Research Triangle ParkNorth Carolina
- Present address:
Division of PharmacometricsOffice of Clinical PharmacologyOffice of Translational SciencesCenter for Drug Evaluation and ResearchFood and Drug Administration Food and Drug AdministrationSilver SpringMaryland
| | - Sashi Nadanaciva
- Compound Safety PredictionWorldwide Medicinal ChemistryPfizer Inc.GrotonConnecticut
| | - Luping Qiu
- Investigative ToxicologyDrug Safety Research and DevelopmentPfizer Inc.GrotonConnecticut
| | - Keith Riccardi
- Pharmacokinetics, Dynamics and MetabolismMedicinal SciencesPfizer Inc.GrotonConnecticut
| | - Li Di
- Pharmacokinetics, Dynamics and MetabolismMedicinal SciencesPfizer Inc.GrotonConnecticut
| | | | - Scott Q. Siler
- DILIsym Services Inc.Research Triangle ParkNorth Carolina
| | - Paul B. Watkins
- UNC Eshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
- UNC Institute for Drug Safety SciencesUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
| | - Hugh A. Barton
- Translational Modeling and SimulationBiomedicine DesignPfizer, Inc.GrotonConnecticut
| | - Michael D. Aleo
- Investigative ToxicologyDrug Safety Research and DevelopmentPfizer Inc.GrotonConnecticut
| | | |
Collapse
|
26
|
Managing the challenge of drug-induced liver injury: a roadmap for the development and deployment of preclinical predictive models. Nat Rev Drug Discov 2019; 19:131-148. [DOI: 10.1038/s41573-019-0048-x] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/26/2019] [Indexed: 12/13/2022]
|
27
|
Analyzing the Mechanisms Behind Macrolide Antibiotic-Induced Liver Injury Using Quantitative Systems Toxicology Modeling. Pharm Res 2019; 36:48. [PMID: 30734107 PMCID: PMC6373306 DOI: 10.1007/s11095-019-2582-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 01/27/2019] [Indexed: 12/22/2022]
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.
Collapse
|
28
|
Kammerer S, Küpper JH. Human hepatocyte systems for in vitro toxicology analysis. ACTA ACUST UNITED AC 2018. [DOI: 10.3233/jcb-179012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Sarah Kammerer
- Institute of Biotechnology, Brandenburg University of Technology, Cottbus-Senftenberg, Germany
| | - Jan-Heiner Küpper
- Institute of Biotechnology, Brandenburg University of Technology, Cottbus-Senftenberg, Germany
| |
Collapse
|
29
|
Zhang Q, Li J, Middleton A, Bhattacharya S, Conolly RB. Bridging the Data Gap From in vitro Toxicity Testing to Chemical Safety Assessment Through Computational Modeling. Front Public Health 2018; 6:261. [PMID: 30255008 PMCID: PMC6141783 DOI: 10.3389/fpubh.2018.00261] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 08/21/2018] [Indexed: 12/18/2022] Open
Abstract
Chemical toxicity testing is moving steadily toward a human cell and organoid-based in vitro approach for reasons including scientific relevancy, efficiency, cost, and ethical rightfulness. Inferring human health risk from chemical exposure based on in vitro testing data is a challenging task, facing various data gaps along the way. This review identifies these gaps and makes a case for the in silico approach of computational dose-response and extrapolation modeling to address many of the challenges. Mathematical models that can mechanistically describe chemical toxicokinetics (TK) and toxicodynamics (TD), for both in vitro and in vivo conditions, are the founding pieces in this regard. Identifying toxicity pathways and in vitro point of departure (PoD) associated with adverse health outcomes requires an understanding of the molecular key events in the interacting transcriptome, proteome, and metabolome. Such an understanding will in turn help determine the sets of sensitive biomarkers to be measured in vitro and the scope of toxicity pathways to be modeled in silico. In vitro data reporting both pathway perturbation and chemical biokinetics in the culture medium serve to calibrate the toxicity pathway and virtual tissue models, which can then help predict PoDs in response to chemical dosimetry experienced by cells in vivo. Two types of in vitro to in vivo extrapolation (IVIVE) are needed. (1) For toxic effects involving systemic regulations, such as endocrine disruption, organism-level adverse outcome pathway (AOP) models are needed to extrapolate in vitro toxicity pathway perturbation to in vivo PoD. (2) Physiologically-based toxicokinetic (PBTK) modeling is needed to extrapolate in vitro PoD dose metrics into external doses for expected exposure scenarios. Linked PBTK and TD models can explore the parameter space to recapitulate human population variability in response to chemical insults. While challenges remain for applying these modeling tools to support in vitro toxicity testing, they open the door toward population-stratified and personalized risk assessment.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Jin Li
- Unilever, Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, United Kingdom
| | - Alistair Middleton
- Unilever, Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, United Kingdom
| | - Sudin Bhattacharya
- Biomedical Engineering, Michigan State University, East Lansing, MI, United States
| | - Rory B Conolly
- Integrated Systems Toxicology Division, National Health and Environmental Effects Research Laboratory, United States Environmental Protection Agency, Durham, NC, United States
| |
Collapse
|
30
|
Guyot L, Honorat M, Jacob G, Bardel C, Tod M, Puisieux A, Guitton J, Payen L. Toxicokinetics and tolerance of a high energy material 3,4,5-trinitropyrazole (TNP) in mice. Toxicol Appl Pharmacol 2018; 355:103-111. [PMID: 29959026 DOI: 10.1016/j.taap.2018.06.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 06/15/2018] [Accepted: 06/25/2018] [Indexed: 10/28/2022]
Abstract
The high-energy compound 3,4,5-trinitropyrazole (TNP) was developed as an alternative to other less energetic and more sensitive explosive materials, in particular 1-methyl-2,4,6-trinitrobenzene (TNT). However, the level of toxicity of TNP remains understudied. Here using an in vivo CD1 mouse model, we mimicked an acute exposure (24 h) to TNP, given either orally or intravenously, and determined the maximum administrable doses (190 mg/kg and 11 mg/kg, respectively), as well as the lethal dose for 50% (LD50) of female or male mice (390 mg/kg for both) treated intravenously with TNP alone. Several metabolites including nitroso-dinitro-pyrazole, hydroxylamino-dinitro-pyrazole, hydroxyl-dinitro-pyrazole and amino-dinitro-pyrazole were identified in urine. TNP is quickly metabolized and eliminated via urine as two main amino-dinitro-pyrazole metabolites. A comparison of the transcriptomic effects of TNP and TNT after 10 days exposure enabled us to demonstrate no major induction of transcripts involved both in cell death mechanisms (apoptosis, necrosis, autophagy) and physiological pathways (glycolysis, ATP production). Finally, subchronic exposure to TNP was replicated in female mice, fed 16.8-52.8 mg/kg/day of TNP for one month, to study the impact on cellular functions. Although blood TNP levels remained high, a lower rate of TNP accumulation in the liver and lungs were observed than during an acute exposure. Conversely, cellular stress functions explored using the RT2 Profiler™ PCR Array Mouse Molecular Toxicology PathwayFinder remained unaltered after this chronic exposure. These findings demonstrate that TNP can be rapidly eliminated in vivo without accumulating in vital organs.
Collapse
Affiliation(s)
- Laetitia Guyot
- Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Laboratoire de biochimie-toxicologie, F-69495 Pierre Bénite, France; EMR 3738, Université Lyon 1, Faculté de Médecin Lyon-Sud-Charles Mérieux, F-69921 Oullins, France
| | - Myléne Honorat
- Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Laboratoire de biochimie-toxicologie, F-69495 Pierre Bénite, France
| | - Guy Jacob
- Airbus Safran Launchers, Centre de Recherches du Bouchet, 9 Rue Lavoisier, 91710 Vert le Petit, France
| | - Claire Bardel
- Hospices Civils de Lyon, Department of biostatistics, Lyon, France
| | - Michel Tod
- EMR 3738, Université Lyon 1, Faculté de Médecin Lyon-Sud-Charles Mérieux, F-69921 Oullins, France
| | - Alain Puisieux
- UMR INSERM U1052/CNRS 5286, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, 69373 Lyon, France
| | - Jérôme Guitton
- Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Laboratoire de biochimie-toxicologie, F-69495 Pierre Bénite, France; EMR 3738, Université Lyon 1, Faculté de Médecin Lyon-Sud-Charles Mérieux, F-69921 Oullins, France; Université Lyon 1, ISPBL, Faculté de pharmacie, Laboratoire de Toxicologie, 69373 Lyon, France.
| | - Léa Payen
- Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Laboratoire de biochimie-toxicologie, F-69495 Pierre Bénite, France; UMR INSERM U1052/CNRS 5286, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, 69373 Lyon, France; Université Lyon 1, ISPBL, Faculté de pharmacie, Laboratoire de Toxicologie, 69373 Lyon, France
| |
Collapse
|
31
|
Cirit M, Stokes CL. Maximizing the impact of microphysiological systems with in vitro-in vivo translation. LAB ON A CHIP 2018; 18:1831-1837. [PMID: 29863727 PMCID: PMC6019627 DOI: 10.1039/c8lc00039e] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Microphysiological systems (MPS) hold promise for improving therapeutic drug approval rates by providing more physiological, human-based, in vitro assays for preclinical drug development activities compared to traditional in vitro and animal models. Here, we first summarize why MPSs are needed in pharmaceutical development, and examine how MPS technologies can be utilized to improve preclinical efforts. We then provide the perspective that the full impact of MPS technologies will be realized only when robust approaches for in vitro-in vivo (MPS-to-human) translation are developed and utilized, and explain how the burgeoning field of quantitative systems pharmacology (QSP) can fill that need.
Collapse
Affiliation(s)
- Murat Cirit
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | | |
Collapse
|
32
|
Woodhead JL, Paech F, Maurer M, Engelhardt M, Schmitt-Hoffmann AH, Spickermann J, Messner S, Wind M, Witschi AT, Krähenbühl S, Siler SQ, Watkins PB, Howell BA. Prediction of Safety Margin and Optimization of Dosing Protocol for a Novel Antibiotic using Quantitative Systems Pharmacology Modeling. Clin Transl Sci 2018; 11:498-505. [PMID: 29877622 PMCID: PMC6132362 DOI: 10.1111/cts.12560] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 04/06/2018] [Indexed: 01/09/2023] Open
Abstract
Elevations of liver enzymes have been observed in clinical trials with BAL30072, a novel antibiotic. In vitro assays have identified potential mechanisms for the observed hepatotoxicity, including electron transport chain (ETC) inhibition and reactive oxygen species (ROS) generation. DILIsym, a quantitative systems pharmacology (QSP) model of drug-induced liver injury, has been used to predict the likelihood that each mechanism explains the observed toxicity. DILIsym was also used to predict the safety margin for a novel BAL30072 dosing scheme; it was predicted to be low. DILIsym was then used to recommend potential modifications to this dosing scheme; weight-adjusted dosing and a requirement to assay plasma alanine aminotransferase (ALT) daily and stop dosing as soon as ALT increases were observed improved the predicted safety margin of BAL30072 and decreased the predicted likelihood of severe injury. This research demonstrates a potential application for QSP modeling in improving the safety profile of candidate drugs.
Collapse
Affiliation(s)
- Jeffrey L Woodhead
- DILIsym Services, Inc., a Simulations Plus company, Research Triangle Park, North Carolina, USA
| | | | - Martina Maurer
- Basilea Pharmaceutica International Ltd., Basel, Switzerland
| | - Marc Engelhardt
- Basilea Pharmaceutica International Ltd., Basel, Switzerland
| | | | | | | | - Mathias Wind
- Basilea Pharmaceutica International Ltd., Basel, Switzerland
| | | | | | - Scott Q Siler
- DILIsym Services, Inc., a Simulations Plus company, Research Triangle Park, North Carolina, USA
| | - Paul B Watkins
- DILIsym Services, Inc., a Simulations Plus company, Research Triangle Park, North Carolina, USA
| | - Brett A Howell
- DILIsym Services, Inc., a Simulations Plus company, Research Triangle Park, North Carolina, USA
| |
Collapse
|
33
|
Battista C, Howell BA, Siler SQ, Watkins PB. An Introduction to DILIsym® Software, a Mechanistic Mathematical Representation of Drug-Induced Liver Injury. ACTA ACUST UNITED AC 2018. [DOI: 10.1007/978-1-4939-7677-5_6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
34
|
Kiyosawa N, Manabe S. Data-intensive drug development in the information age: applications of Systems Biology/Pharmacology/Toxicology. J Toxicol Sci 2017; 41:SP15-SP25. [PMID: 28003636 DOI: 10.2131/jts.41.sp15] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Pharmaceutical companies continuously face challenges to deliver new drugs with true medical value. R&D productivity of drug development projects depends on 1) the value of the drug concept and 2) data and in-depth knowledge that are used rationally to evaluate the drug concept's validity. A model-based data-intensive drug development approach is a key competitive factor used by innovative pharmaceutical companies to reduce information bias and rationally demonstrate the value of drug concepts. Owing to the accumulation of publicly available biomedical information, our understanding of the pathophysiological mechanisms of diseases has developed considerably; it is the basis for identifying the right drug target and creating a drug concept with true medical value. Our understanding of the pathophysiological mechanisms of disease animal models can also be improved; it can thus support rational extrapolation of animal experiment results to clinical settings. The Systems Biology approach, which leverages publicly available transcriptome data, is useful for these purposes. Furthermore, applying Systems Pharmacology enables dynamic simulation of drug responses, from which key research questions to be addressed in the subsequent studies can be adequately informed. Application of Systems Biology/Pharmacology to toxicology research, namely Systems Toxicology, should considerably improve the predictability of drug-induced toxicities in clinical situations that are difficult to predict from conventional preclinical toxicology studies. Systems Biology/Pharmacology/Toxicology models can be continuously improved using iterative learn-confirm processes throughout preclinical and clinical drug discovery and development processes. Successful implementation of data-intensive drug development approaches requires cultivation of an adequate R&D culture to appreciate this approach.
Collapse
Affiliation(s)
- Naoki Kiyosawa
- Translational Medicine & Clinical Pharmacology Department, Daiichi Sankyo Co. Ltd
| | | |
Collapse
|
35
|
Bloomingdale P, Housand C, Apgar JF, Millard BL, Mager DE, Burke JM, Shah DK. Quantitative systems toxicology. CURRENT OPINION IN TOXICOLOGY 2017; 4:79-87. [PMID: 29308440 PMCID: PMC5754001 DOI: 10.1016/j.cotox.2017.07.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The overarching goal of modern drug development is to optimize therapeutic benefits while minimizing adverse effects. However, inadequate efficacy and safety concerns remain to be the major causes of drug attrition in clinical development. For the past 80 years, toxicity testing has consisted of evaluating the adverse effects of drugs in animals to predict human health risks. The U.S. Environmental Protection Agency recognized the need to develop innovative toxicity testing strategies and asked the National Research Council to develop a long-range vision and strategy for toxicity testing in the 21st century. The vision aims to reduce the use of animals and drug development costs through the integration of computational modeling and in vitro experimental methods that evaluates the perturbation of toxicity-related pathways. Towards this vision, collaborative quantitative systems pharmacology and toxicology modeling endeavors (QSP/QST) have been initiated amongst numerous organizations worldwide. In this article, we discuss how quantitative structure-activity relationship (QSAR), network-based, and pharmacokinetic/pharmacodynamic modeling approaches can be integrated into the framework of QST models. Additionally, we review the application of QST models to predict cardiotoxicity and hepatotoxicity of drugs throughout their development. Cell and organ specific QST models are likely to become an essential component of modern toxicity testing, and provides a solid foundation towards determining individualized therapeutic windows to improve patient safety.
Collapse
Affiliation(s)
- Peter Bloomingdale
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, USA
| | - Conrad Housand
- Applied BioMath, LLC, 55 Old Bedford Road, Suite 208, Lincoln, MA 01773, USA
| | - Joshua F Apgar
- Applied BioMath, LLC, 55 Old Bedford Road, Suite 208, Lincoln, MA 01773, USA
| | - Bjorn L Millard
- Applied BioMath, LLC, 55 Old Bedford Road, Suite 208, Lincoln, MA 01773, USA
| | - Donald E Mager
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, USA
| | - John M Burke
- Applied BioMath, LLC, 55 Old Bedford Road, Suite 208, Lincoln, MA 01773, USA
| | - Dhaval K Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, USA
| |
Collapse
|
36
|
Wu L, Liu Z, Auerbach S, Huang R, Chen M, McEuen K, Xu J, Fang H, Tong W. Integrating Drug's Mode of Action into Quantitative Structure-Activity Relationships for Improved Prediction of Drug-Induced Liver Injury. J Chem Inf Model 2017; 57:1000-1006. [PMID: 28350954 PMCID: PMC6233892 DOI: 10.1021/acs.jcim.6b00719] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Drug-induced liver injury (DILI) is complex in mechanism. Different drugs could undergo different mechanisms but result in the same DILI type, while the same drug could lead to different DILI types via different mechanisms. Therefore, predicting a drug's potential for DILI should take its underlying mechanisms into consideration. To achieve that, we constructed a novel approach by incorporating the drug's Mode of Action (MOA) into Quantitative Structure-Activity Relationship (QSAR) modeling. This MOA-DILI approach was examined using a data set of 333 drugs. The drugs were first grouped according to their MOA profiles (positive or negative in each MOA) based on the Tox21 qHTS assays. QSAR models for individual MOA assays were developed and subsequently combined to obtain the MOA-DILI model. A hold-out testing strategy (222 drugs for training and 111 drugs as a test set) was employed, which yielded a predictive accuracy of 0.711. The MOA-DILI model was directly compared with the standard QSAR approach using the same hold-out strategy, and the QSAR model yielded an accuracy of 0.662. To minimize the random chance in splitting training/test sets, the hold-out testing process was repeated 1000 times, and the observed difference in prediction accuracy between MOA-DILI and QSARs was statistically significant (P value <0.0001). Out of 17 MOAs used, four assays (i.e., antioxidant response elements, PPAR-gamma, estrogen receptor, and thyroid receptor assays) contributed most to the improved prediction of the MOA-DILI model over QSARs. In conclusion, the MOA-DILI approach has the potential to significantly improve predictive outcomes and to reveal complex relationships between MOAs and DILI, all of which would be helpful in developing DILI predictive models in drug screening and for risk assessment of industrial chemicals.
Collapse
Affiliation(s)
- Leihong Wu
- National Center for Toxicological Research, US Food and Drug Administration, 3900 NCTR RD, Jefferson, AR 72079, USA
| | - Zhichao Liu
- National Center for Toxicological Research, US Food and Drug Administration, 3900 NCTR RD, Jefferson, AR 72079, USA
| | - Scott Auerbach
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, 111 T.W. Alexander Drive, RTP, NC 27709, USA
| | - Ruili Huang
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, Maryland 20850, USA
| | - Minjun Chen
- National Center for Toxicological Research, US Food and Drug Administration, 3900 NCTR RD, Jefferson, AR 72079, USA
| | - Kristin McEuen
- University of Arkansas at Little Rock, 2801 S University Ave, Little Rock, AR 72204, USA
| | - Joshua Xu
- National Center for Toxicological Research, US Food and Drug Administration, 3900 NCTR RD, Jefferson, AR 72079, USA
| | - Hong Fang
- National Center for Toxicological Research, US Food and Drug Administration, 3900 NCTR RD, Jefferson, AR 72079, USA
| | - Weida Tong
- National Center for Toxicological Research, US Food and Drug Administration, 3900 NCTR RD, Jefferson, AR 72079, USA
| |
Collapse
|
37
|
Yucha RW, He K, Shi Q, Cai L, Nakashita Y, Xia CQ, Liao M. In Vitro Drug-Induced Liver Injury Prediction: Criteria Optimization of Efflux Transporter IC50 and Physicochemical Properties. Toxicol Sci 2017; 157:487-499. [DOI: 10.1093/toxsci/kfx060] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
38
|
Yang K, Battista C, Woodhead JL, Stahl SH, Mettetal JT, Watkins PB, Siler SQ, Howell BA. Systems pharmacology modeling of drug-induced hyperbilirubinemia: Differentiating hepatotoxicity and inhibition of enzymes/transporters. Clin Pharmacol Ther 2017; 101:501-509. [PMID: 28074467 PMCID: PMC5367379 DOI: 10.1002/cpt.619] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 10/31/2016] [Accepted: 01/04/2017] [Indexed: 12/20/2022]
Abstract
Elevations in serum bilirubin during drug treatment may indicate global liver dysfunction and a high risk of liver failure. However, drugs also can increase serum bilirubin in the absence of hepatic injury by inhibiting specific enzymes/transporters. We constructed a mechanistic model of bilirubin disposition based on known functional polymorphisms in bilirubin metabolism/transport. Using physiologically based pharmacokinetic (PBPK) model-predicted drug exposure and enzyme/transporter inhibition constants determined in vitro, our model correctly predicted indinavir-mediated hyperbilirubinemia in humans and rats. Nelfinavir was predicted not to cause hyperbilirubinemia, consistent with clinical observations. We next examined a new drug candidate that caused both elevations in serum bilirubin and biochemical evidence of liver injury in rats. Simulations suggest that bilirubin elevation primarily resulted from inhibition of transporters rather than global liver dysfunction. We conclude that mechanistic modeling of bilirubin can help elucidate underlying mechanisms of drug-induced hyperbilirubinemia, and thereby distinguish benign from clinically important elevations in serum bilirubin.
Collapse
Affiliation(s)
- K Yang
- DILIsym Services Inc, Research Triangle Park, North Carolina, USA
| | - C Battista
- DILIsym Services Inc, Research Triangle Park, North Carolina, USA.,University of North Carolina Institute for Drug Safety Sciences, The Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - J L Woodhead
- DILIsym Services Inc, Research Triangle Park, North Carolina, USA
| | - S H Stahl
- ADME Transporters, Drug Safety and Metabolism, Innovative Medicines and Early Development, AstraZeneca, Cambridge, United Kingdom
| | - J T Mettetal
- Drug Safety and Metabolism, AstraZeneca R&D, Waltham, Massachusetts, USA
| | - P B Watkins
- University of North Carolina Institute for Drug Safety Sciences, The Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - S Q Siler
- DILIsym Services Inc, Research Triangle Park, North Carolina, USA
| | - B A Howell
- DILIsym Services Inc, Research Triangle Park, North Carolina, USA
| |
Collapse
|
39
|
Human leukocyte antigen and idiosyncratic adverse drug reactions. Drug Metab Pharmacokinet 2017; 32:21-30. [DOI: 10.1016/j.dmpk.2016.11.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 11/08/2016] [Accepted: 11/09/2016] [Indexed: 12/17/2022]
|
40
|
Sistare FD, Mattes WB, LeCluyse EL. The Promise of New Technologies to Reduce, Refine, or Replace Animal Use while Reducing Risks of Drug Induced Liver Injury in Pharmaceutical Development. ILAR J 2017; 57:186-211. [DOI: 10.1093/ilar/ilw025] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 07/25/2016] [Accepted: 09/13/2016] [Indexed: 12/19/2022] Open
|
41
|
Pizzo F, Lombardo A, Manganaro A, Benfenati E. A New Structure-Activity Relationship (SAR) Model for Predicting Drug-Induced Liver Injury, Based on Statistical and Expert-Based Structural Alerts. Front Pharmacol 2016; 7:442. [PMID: 27920722 PMCID: PMC5118449 DOI: 10.3389/fphar.2016.00442] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Accepted: 11/04/2016] [Indexed: 12/20/2022] Open
Abstract
The prompt identification of chemical molecules with potential effects on liver may help in drug discovery and in raising the levels of protection for human health. Besides in vitro approaches, computational methods in toxicology are drawing attention. We built a structure-activity relationship (SAR) model for evaluating hepatotoxicity. After compiling a data set of 950 compounds using data from the literature, we randomly split it into training (80%) and test sets (20%). We also compiled an external validation set (101 compounds) for evaluating the performance of the model. To extract structural alerts (SAs) related to hepatotoxicity and non-hepatotoxicity we used SARpy, a statistical application that automatically identifies and extracts chemical fragments related to a specific activity. We also applied the chemical grouping approach for manually identifying other SAs. We calculated accuracy, specificity, sensitivity and Matthews correlation coefficient (MCC) on the training, test and external validation sets. Considering the complexity of the endpoint, the model performed well. In the training, test and external validation sets the accuracy was respectively 81, 63, and 68%, specificity 89, 33, and 33%, sensitivity 93, 88, and 80% and MCC 0.63, 0.27, and 0.13. Since it is preferable to overestimate hepatotoxicity rather than not to recognize unsafe compounds, the model's architecture followed a conservative approach. As it was built using human data, it might be applied without any need for extrapolation from other species. This model will be freely available in the VEGA platform.
Collapse
Affiliation(s)
- Fabiola Pizzo
- Department of Environmental Health Sciences, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri" Milan, Italy
| | - Anna Lombardo
- Department of Environmental Health Sciences, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri" Milan, Italy
| | - Alberto Manganaro
- Department of Environmental Health Sciences, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri" Milan, Italy
| | - Emilio Benfenati
- Department of Environmental Health Sciences, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri" Milan, Italy
| |
Collapse
|
42
|
Woodhead JL, Watkins PB, Howell BA, Siler SQ, Shoda LKM. The role of quantitative systems pharmacology modeling in the prediction and explanation of idiosyncratic drug-induced liver injury. Drug Metab Pharmacokinet 2016; 32:40-45. [PMID: 28129975 DOI: 10.1016/j.dmpk.2016.11.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 10/14/2016] [Accepted: 11/15/2016] [Indexed: 01/12/2023]
Abstract
Idiosyncratic drug-induced liver injury (iDILI) is a serious concern in drug development. The rarity and multifactorial nature of iDILI makes it difficult to predict and explain. Recently, human leukocyte antigen (HLA) allele associations have provided strong support for a role of an adaptive immune response in the pathogenesis of many iDILI cases; however, it is likely that an adaptive immune attack requires several preceding events. Quantitative systems pharmacology (QSP), an in silico modeling technique that leverages known physiology and the results of in vitro experiments in order to make predictions about how drugs affect biological processes, is proposed as a potentially useful tool for predicting and explaining critical events that likely precede immune-mediated iDILI, as well as the immune attack itself. DILIsym, a QSP platform for drug-induced liver injury, has demonstrated success in predicting the presence of delayed hepatocellular stress events that likely precede the iDILI cascade, and has successfully predicted hepatocellular stress likely underlying iDILI attributed to troglitazone and tolvaptan. The incorporation of a model of the adaptive immune system into DILIsym would represent and important advance. In summary, QSP methods can play a key role in the future prediction and understanding of both immune-mediated and non-immune-mediated iDILI.
Collapse
Affiliation(s)
- Jeffrey L Woodhead
- DILIsym Services, Inc., 6 Davis Drive, Research Triangle Park, NC 27709, USA.
| | - Paul B Watkins
- Institute for Drug Safety Sciences, UNC-Eshelman School of Pharmacy, 6 Davis Drive, Research Triangle Park, NC 27709, USA
| | - Brett A Howell
- DILIsym Services, Inc., 6 Davis Drive, Research Triangle Park, NC 27709, USA
| | - Scott Q Siler
- DILIsym Services, Inc., 6 Davis Drive, Research Triangle Park, NC 27709, USA
| | - Lisl K M Shoda
- DILIsym Services, Inc., 6 Davis Drive, Research Triangle Park, NC 27709, USA
| |
Collapse
|
43
|
Woodhead JL, Brock WJ, Roth SE, Shoaf SE, Brouwer KLR, Church R, Grammatopoulos TN, Stiles L, Siler SQ, Howell BA, Mosedale M, Watkins PB, Shoda LKM. Application of a Mechanistic Model to Evaluate Putative Mechanisms of Tolvaptan Drug-Induced Liver Injury and Identify Patient Susceptibility Factors. Toxicol Sci 2016; 155:61-74. [PMID: 27655350 PMCID: PMC5216653 DOI: 10.1093/toxsci/kfw193] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Tolvaptan is a selective vasopressin V2 receptor antagonist, approved in several countries for the treatment of hyponatremia and autosomal dominant polycystic kidney disease (ADPKD). No liver injury has been observed with tolvaptan treatment in healthy subjects and in non-ADPKD indications, but ADPKD clinical trials showed evidence of drug-induced liver injury (DILI). Although all DILI events resolved, additional monitoring in tolvaptan-treated ADPKD patients is required. In vitro assays identified alterations in bile acid disposition and inhibition of mitochondrial respiration as potential mechanisms underlying tolvaptan hepatotoxicity. This report details the application of DILIsym software to determine whether these mechanisms could account for the liver safety profile of tolvaptan observed in ADPKD clinical trials. DILIsym simulations included physiologically based pharmacokinetic estimates of hepatic exposure for tolvaptan and2 metabolites, and their effects on hepatocyte bile acid transporters and mitochondrial respiration. The frequency of predicted alanine aminotransferase (ALT) elevations, following simulated 90/30 mg split daily dosing, was 7.9% compared with clinical observations of 4.4% in ADPKD trials. Toxicity was multifactorial as inhibition of bile acid transporters and mitochondrial respiration contributed to the simulated DILI. Furthermore, simulation analysis identified both pre-treatment risk factors and on-treatment biomarkers predictive of simulated DILI. The simulations demonstrated that in vivo hepatic exposure to tolvaptan and the DM-4103 metabolite, combined with these 2 mechanisms of toxicity, were sufficient to account for the initiation of tolvaptan-mediated DILI. Identification of putative risk-factors and potential novel biomarkers provided insight for the development of mechanism-based tolvaptan risk-mitigation strategies.
Collapse
Affiliation(s)
| | - William J Brock
- Otsuka Pharmaceutical Development & Commercialization, Brock Scientific Consulting, Montgomery Village, Rockville, Maryland
| | - Sharin E Roth
- Otsuka Pharmaceutical Development & Commercialization, Rockville, Maryland
| | - Susan E Shoaf
- Otsuka Pharmaceutical Development & Commercialization, Rockville, Maryland
| | - Kim L R Brouwer
- UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Rachel Church
- UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,UNC Institute for Drug Safety Sciences, University of North Carolina at Chapel Hill, Research Triangle Park, North Carolina
| | | | | | - Scott Q Siler
- DILIsym Services, Inc, Research Triangle Park, North Carolina
| | - Brett A Howell
- DILIsym Services, Inc, Research Triangle Park, North Carolina
| | - Merrie Mosedale
- UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,UNC Institute for Drug Safety Sciences, University of North Carolina at Chapel Hill, Research Triangle Park, North Carolina
| | - Paul B Watkins
- UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,UNC Institute for Drug Safety Sciences, University of North Carolina at Chapel Hill, Research Triangle Park, North Carolina
| | - Lisl K M Shoda
- DILIsym Services, Inc, Research Triangle Park, North Carolina;
| |
Collapse
|
44
|
Advances in Engineered Liver Models for Investigating Drug-Induced Liver Injury. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1829148. [PMID: 27725933 PMCID: PMC5048025 DOI: 10.1155/2016/1829148] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 07/19/2016] [Indexed: 12/17/2022]
Abstract
Drug-induced liver injury (DILI) is a major cause of drug attrition. Testing drugs on human liver models is essential to mitigate the risk of clinical DILI since animal studies do not always suffice due to species-specific differences in liver pathways. While primary human hepatocytes (PHHs) can be cultured on extracellular matrix proteins, a rapid decline in functions leads to low sensitivity (<50%) in DILI prediction. Semiconductor-driven engineering tools now allow precise control over the hepatocyte microenvironment to enhance and stabilize phenotypic functions. The latest platforms coculture PHHs with stromal cells to achieve hepatic stability and enable crosstalk between the various liver cell types towards capturing complex cellular mechanisms in DILI. The recent introduction of induced pluripotent stem cell-derived human hepatocyte-like cells can potentially allow a better understanding of interindividual differences in idiosyncratic DILI. Liver models are also being coupled to other tissue models via microfluidic perfusion to study the intertissue crosstalk upon drug exposure as in a live organism. Here, we review the major advances being made in the engineering of liver models and readouts as they pertain to DILI investigations. We anticipate that engineered human liver models will reduce drug attrition, animal usage, and cases of DILI in humans.
Collapse
|
45
|
Zurlinden TJ, Reisfeld B. Characterizing the Effects of Race/Ethnicity on Acetaminophen Pharmacokinetics Using Physiologically Based Pharmacokinetic Modeling. Eur J Drug Metab Pharmacokinet 2016; 42:143-153. [DOI: 10.1007/s13318-016-0329-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
46
|
Friedrich CM. A model qualification method for mechanistic physiological QSP models to support model-informed drug development. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2016; 5:43-53. [PMID: 26933515 PMCID: PMC4761232 DOI: 10.1002/psp4.12056] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 12/17/2015] [Indexed: 12/23/2022]
Abstract
Mechanistic physiological modeling is a scientific method that combines available data with scientific knowledge and engineering approaches to facilitate better understanding of biological systems, improve decision‐making, reduce risk, and increase efficiency in drug discovery and development. It is a type of quantitative systems pharmacology (QSP) approach that places drug‐specific properties in the context of disease biology. This tutorial provides a broadly applicable model qualification method (MQM) to ensure that mechanistic physiological models are fit for their intended purposes.
Collapse
|
47
|
Longo DM, Yang Y, Watkins PB, Howell BA, Siler SQ. Elucidating Differences in the Hepatotoxic Potential of Tolcapone and Entacapone With DILIsym(®), a Mechanistic Model of Drug-Induced Liver Injury. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2016; 5:31-9. [PMID: 26844013 PMCID: PMC4728295 DOI: 10.1002/psp4.12053] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 12/02/2015] [Indexed: 12/18/2022]
Abstract
Tolcapone and entacapone are catechol‐O‐methyltransferase (COMT) inhibitors developed as adjunct therapies for treating Parkinson's disease. While both drugs have been shown to cause mitochondrial dysfunction and inhibition of the bile salt export protein (BSEP), liver injury has only been associated with the use of tolcapone. Here we used a multiscale, mechanistic model (DILIsym®) to simulate the response to tolcapone and entacapone. In a simulated population (SimPops™) receiving recommended doses of tolcapone (200 mg t.i.d.), increases in serum alanine transaminase (ALT) >3× the upper limit of normal (ULN) were observed in 2.2% of the population. In contrast, no simulated patients receiving recommended doses of entacapone (200 mg 8× day) experienced serum ALT >3× ULN. Further, DILIsym® analyses revealed patient‐specific risk factors that may contribute to tolcapone‐mediated hepatotoxicity. In summary, the simulations demonstrated that differences in mitochondrial uncoupling potency and hepatic exposure primarily account for the difference in hepatotoxic potential for tolcapone and entacapone.
Collapse
Affiliation(s)
- D M Longo
- Hamner-UNC Institute for Drug Safety Sciences, Hamner Institutes for Health Sciences, Research Triangle Park North Carolina USA; DILIsym® Services, Research Triangle Park North Carolina USA
| | - Y Yang
- Hamner-UNC Institute for Drug Safety Sciences, Hamner Institutes for Health Sciences, Research Triangle Park North Carolina USA
| | - P B Watkins
- Hamner-UNC Institute for Drug Safety Sciences, Hamner Institutes for Health Sciences, Research Triangle Park North Carolina USA; DILIsym® Services, Research Triangle Park North Carolina USA
| | - B A Howell
- Hamner-UNC Institute for Drug Safety Sciences, Hamner Institutes for Health Sciences, Research Triangle Park North Carolina USA; DILIsym® Services, Research Triangle Park North Carolina USA
| | - S Q Siler
- Hamner-UNC Institute for Drug Safety Sciences, Hamner Institutes for Health Sciences, Research Triangle Park North Carolina USA; DILIsym® Services, Research Triangle Park North Carolina USA
| |
Collapse
|
48
|
Thompson RA, Isin EM, Ogese MO, Mettetal JT, Williams DP. Reactive Metabolites: Current and Emerging Risk and Hazard Assessments. Chem Res Toxicol 2016; 29:505-33. [DOI: 10.1021/acs.chemrestox.5b00410] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Richard A. Thompson
- DMPK, Respiratory, Inflammation & Autoimmunity iMed, AstraZeneca R&D, 431 83 Mölndal, Sweden
| | - Emre M. Isin
- DMPK, Cardiovascular & Metabolic Diseases iMed, AstraZeneca R&D, 431 83 Mölndal, Sweden
| | - Monday O. Ogese
- Translational Safety, Drug Safety and Metabolism, AstraZeneca R&D, Darwin Building 310, Cambridge Science Park, Milton Rd, Cambridge CB4 0FZ, United Kingdom
| | - Jerome T. Mettetal
- Translational Safety, Drug Safety and Metabolism, AstraZeneca R&D, 35 Gatehouse Dr, Waltham, Massachusetts 02451, United States
| | - Dominic P. Williams
- Translational Safety, Drug Safety and Metabolism, AstraZeneca R&D, Darwin Building 310, Cambridge Science Park, Milton Rd, Cambridge CB4 0FZ, United Kingdom
| |
Collapse
|
49
|
Zurlinden TJ, Heard K, Reisfeld B. A novel approach for estimating ingested dose associated with paracetamol overdose. Br J Clin Pharmacol 2015; 81:634-45. [PMID: 26441245 DOI: 10.1111/bcp.12796] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 09/04/2015] [Accepted: 10/01/2015] [Indexed: 11/28/2022] Open
Abstract
AIM In cases of paracetamol (acetaminophen, APAP) overdose, an accurate estimate of tissue-specific paracetamol pharmacokinetics (PK) and ingested dose can offer health care providers important information for the individualized treatment and follow-up of affected patients. Here a novel methodology is presented to make such estimates using a standard serum paracetamol measurement and a computational framework. METHODS The core component of the computational framework was a physiologically-based pharmacokinetic (PBPK) model developed and evaluated using an extensive set of human PK data. Bayesian inference was used for parameter and dose estimation, allowing the incorporation of inter-study variability, and facilitating the calculation of uncertainty in model outputs. RESULTS Simulations of paracetamol time course concentrations in the blood were in close agreement with experimental data under a wide range of dosing conditions. Also, predictions of administered dose showed good agreement with a large collection of clinical and emergency setting PK data over a broad dose range. In addition to dose estimation, the platform was applied for the determination of optimal blood sampling times for dose reconstruction and quantitation of the potential role of paracetamol conjugate measurement on dose estimation. CONCLUSIONS Current therapies for paracetamol overdose rely on a generic methodology involving the use of a clinical nomogram. By using the computational framework developed in this study, serum sample data, and the individual patient's anthropometric and physiological information, personalized serum and liver pharmacokinetic profiles and dose estimate could be generated to help inform an individualized overdose treatment and follow-up plan.
Collapse
Affiliation(s)
- Todd J Zurlinden
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, Colorado, 80523-1370
| | - Kennon Heard
- Department of Emergency Medicine, University of Colorado School of Medicine, 12401 E. 17th Avenue Campus Box B-215, Aurora, CO, 80045.,Rocky Mountain Poison and Drug Center, Denver, CO, 80204
| | - Brad Reisfeld
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, Colorado, 80523-1370.,School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado, 80523-1376, USA
| |
Collapse
|
50
|
Abstract
Attrition due to nonclinical safety represents a major issue for the productivity of pharmaceutical research and development (R&D) organizations, especially during the compound optimization stages of drug discovery and the early stages of clinical development. Focusing on decreasing nonclinical safety-related attrition is not a new concept, and various approaches have been experimented with over the last two decades. Front-loading testing funnels in Discovery with in vitro toxicity assays designed to rapidly identify unfavorable molecules was the approach adopted by most pharmaceutical R&D organizations a few years ago. However, this approach has also a non-negligible opportunity cost. Hence, significant refinements to the "fail early, fail often" paradigm have been proposed recently to reflect the complexity of accurately categorizing compounds with early data points without taking into account other important contextual aspects, in particular efficacious systemic and tissue exposures. This review provides an overview of toxicology approaches and models that can be used in pharmaceutical Discovery at the series/lead identification and lead optimization stages to guide and inform chemistry efforts, as well as a personal view on how to best use them to meet nonclinical safety-related attrition objectives consistent with a sustainable pharmaceutical R&D model. The scope of this review is limited to small molecules, as large molecules are associated with challenges that are quite different. Finally, a perspective on how several emerging technologies may impact toxicity evaluation is also provided.
Collapse
Affiliation(s)
- Eric A G Blomme
- Global Preclinical Safety, AbbVie Inc. , 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Yvonne Will
- Drug Safety Research and Development, Pfizer , Eastern Point Road, Groton, Connecticut 06340, United States
| |
Collapse
|