1
|
Aguado-Sierra J, Dominguez-Gomez P, Amar A, Butakoff C, Leitner M, Schaper S, Kriegl JM, Darpo B, Vazquez M, Rast G. Virtual clinical QT exposure-response studies - A translational computational approach. J Pharmacol Toxicol Methods 2024; 126:107498. [PMID: 38432528 DOI: 10.1016/j.vascn.2024.107498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 12/13/2023] [Accepted: 02/29/2024] [Indexed: 03/05/2024]
Abstract
BACKGROUND AND PURPOSE A recent paradigm shift in proarrhythmic risk assessment suggests that the integration of clinical, non-clinical, and computational evidence can be used to reach a comprehensive understanding of the proarrhythmic potential of drug candidates. While current computational methodologies focus on predicting the incidence of proarrhythmic events after drug administration, the objective of this study is to predict concentration-response relationships of QTc as a clinical endpoint. EXPERIMENTAL APPROACH Full heart computational models reproducing human cardiac populations were created to predict the concentration-response relationship of changes in the QT interval as recommended for clinical trials. The concentration-response relationship of the QT-interval prolongation obtained from the computational cardiac population was compared against the relationship from clinical trial data for a set of well-characterized compounds: moxifloxacin, dofetilide, verapamil, and ondansetron. KEY RESULTS Computationally derived concentration-response relationships of QT interval changes for three of the four drugs had slopes within the confidence interval of clinical trials (dofetilide, moxifloxacin and verapamil) when compared to placebo-corrected concentration-ΔQT and concentration-ΔQT regressions. Moxifloxacin showed a higher intercept, outside the confidence interval of the clinical data, demonstrating that in this example, the standard linear regression does not appropriately capture the concentration-response results at very low concentrations. The concentrations corresponding to a mean QTc prolongation of 10 ms were consistently lower in the computational model than in clinical data. The critical concentration varied within an approximate ratio of 0.5 (moxifloxacin and ondansetron) and 1 times (dofetilide, verapamil) the critical concentration observed in human clinical trials. Notably, no other in silico methodology can approximate the human critical concentration values for a QT interval prolongation of 10 ms. CONCLUSION AND IMPLICATIONS Computational concentration-response modelling of a virtual population of high-resolution, 3-dimensional cardiac models can provide comparable information to clinical data and could be used to complement pre-clinical and clinical safety packages. It provides access to an unlimited exposure range to support trial design and can improve the understanding of pre-clinical-clinical translation.
Collapse
Affiliation(s)
- Jazmin Aguado-Sierra
- Elem Biotech, Barcelona, Spain; Barcelona Supercomputing Center, Barcelona, Spain.
| | | | | | | | - Michael Leitner
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach, Germany.
| | - Stefan Schaper
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach, Germany.
| | - Jan M Kriegl
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach, Germany.
| | | | - Mariano Vazquez
- Elem Biotech, Barcelona, Spain; Barcelona Supercomputing Center, Barcelona, Spain.
| | - Georg Rast
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach, Germany.
| |
Collapse
|
2
|
Bhateria M, Taneja I, Karsauliya K, Sonker AK, Shibata Y, Sato H, Singh SP, Hisaka A. Predicting the in vivo developmental toxicity of fenarimol from in vitro toxicity data using PBTK modelling-facilitated reverse dosimetry approach. Toxicol Appl Pharmacol 2024; 484:116879. [PMID: 38431230 DOI: 10.1016/j.taap.2024.116879] [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: 10/04/2023] [Revised: 02/21/2024] [Accepted: 02/27/2024] [Indexed: 03/05/2024]
Abstract
In vitro methods are widely used in modern toxicological testing; however, the data cannot be directly employed for risk assessment. In vivo toxicity of chemicals can be predicted from in vitro data using physiologically based toxicokinetic (PBTK) modelling-facilitated reverse dosimetry (PBTK-RD). In this study, a minimal-PBTK model was constructed to predict the in-vivo kinetic profile of fenarimol (FNL) in rats and humans. The model was verified by comparing the observed and predicted pharmacokinetics of FNL for rats (calibrator) and further applied to humans. Using the PBTK-RD approach, the reported in vitro developmental toxicity data for FNL was translated to in vivo dose-response data to predict the assay equivalent oral dose in rats and humans. The predicted assay equivalent rat oral dose (36.46 mg/kg) was comparable to the literature reported in vivo BMD10 value (22.8 mg/kg). The model was also employed to derive the chemical-specific adjustment factor (CSAF) for interspecies toxicokinetics variability of FNL. Further, Monte Carlo simulations were performed to predict the population variability in the plasma concentration of FNL and to derive CSAF for intersubject human kinetic differences. The comparison of CSAF values for interspecies and intersubject toxicokinetic variability with their respective default values revealed that the applied uncertainty factors were adequately protective.
Collapse
Affiliation(s)
- Manisha Bhateria
- Toxicokinetics Laboratory, ASSIST and REACT Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, India
| | - Isha Taneja
- Certara UK Limited, Simcyp Division, Acero, 1 Concourse Way, Sheffield S1 2BJ, UK
| | - Kajal Karsauliya
- Toxicokinetics Laboratory, ASSIST and REACT Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, India
| | - Ashish Kumar Sonker
- Toxicokinetics Laboratory, ASSIST and REACT Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Yukihiro Shibata
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8675, Japan
| | - Hiromi Sato
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8675, Japan
| | - Sheelendra Pratap Singh
- Toxicokinetics Laboratory, ASSIST and REACT Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| | - Akihiro Hisaka
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8675, Japan
| |
Collapse
|
3
|
Smeltz M, Wambaugh JF, Wetmore BA. Plasma Protein Binding Evaluations of Per- and Polyfluoroalkyl Substances for Category-Based Toxicokinetic Assessment. Chem Res Toxicol 2023; 36:870-881. [PMID: 37184865 PMCID: PMC10506455 DOI: 10.1021/acs.chemrestox.3c00003] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
New approach methodologies (NAMs) that make use of in vitro screening and in silico approaches to inform chemical evaluations rely on in vitro toxicokinetic (TK) data to translate in vitro bioactive concentrations to exposure metrics reflective of administered dose. With 1364 per- and polyfluoroalkyl substances (PFAS) identified as of interest under Section 8 of the U.S. Toxic Substances Control Act (TSCA) and concern over the lack of knowledge regarding environmental persistence, human health, and ecological effects, the utility of NAMs to understand potential toxicities and toxicokinetics across these data-poor compounds is being evaluated. To address the TK data deficiency, 71 PFAS selected to span a wide range of functional groups and physico-chemical properties were evaluated for in vitro human plasma protein binding (PPB) by ultracentrifugation with liquid chromatography-mass spectrometry analysis. For the 67 PFAS successfully evaluated by ultracentrifugation, fraction unbound in plasma (fup) ranged from less than 0.0001 (pentadecafluorooctanoyl chloride) to 0.7302 (tetrafluorosuccinic acid), with over half of the PFAS showing PPB exceeding 99.5% (fup < 0.005). Category-based evaluations revealed that perfluoroalkanoyl chlorides and perfluorinated carboxylates (PFCAs) with 6-10 carbons were the highest bound, with similar median values for alkyl, ether, and polyether PFCAs. Interestingly, binding was lower for the PFCAs with a carbon chain length of ≥11. Lower binding also was noted for fluorotelomer carboxylic acids when compared to their carbon-equivalent perfluoroalkyl acids. Comparisons of the fup value derived using two PPB methods, ultracentrifugation or rapid equilibrium dialysis (RED), revealed RED failure for a subset of PFAS of high mass and/or predicted octanol-water partition coefficients exceeding 4 due to failure to achieve equilibrium. Bayesian modeling was used to provide uncertainty bounds around fup point estimates for incorporation into TK modeling. This PFAS PPB evaluation and grouping exercise across 67 structures greatly expand our current knowledge and will aid in PFAS NAM development.
Collapse
Affiliation(s)
- Marci Smeltz
- Center for Computational Toxicology and Exposure, US EPA Office of Research and Development, Research Triangle Park, NC 27711, USA
- Current Affiliation: Center for Environmental Measurement and Modeling; Research Triangle Park, NC, 27711, USA
| | - John F. Wambaugh
- Center for Computational Toxicology and Exposure, US EPA Office of Research and Development, Research Triangle Park, NC 27711, USA
| | - Barbara A. Wetmore
- Center for Computational Toxicology and Exposure, US EPA Office of Research and Development, Research Triangle Park, NC 27711, USA
| |
Collapse
|
4
|
Pauli I, Rezende CDO, Slafer BW, Dessoy MA, de Souza ML, Ferreira LLG, Adjanohun ALM, Ferreira RS, Magalhães LG, Krogh R, Michelan-Duarte S, Del Pintor RV, da Silva FBR, Cruz FC, Dias LC, Andricopulo AD. Multiparameter Optimization of Trypanocidal Cruzain Inhibitors With In Vivo Activity and Favorable Pharmacokinetics. Front Pharmacol 2022; 12:774069. [PMID: 35069198 PMCID: PMC8767159 DOI: 10.3389/fphar.2021.774069] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/22/2021] [Indexed: 12/20/2022] Open
Abstract
Cruzain, the main cysteine protease of Trypanosoma cruzi, plays key roles in all stages of the parasite's life cycle, including nutrition acquisition, differentiation, evasion of the host immune system, and invasion of host cells. Thus, inhibition of this validated target may lead to the development of novel drugs for the treatment of Chagas disease. In this study, a multiparameter optimization (MPO) approach, molecular modeling, and structure-activity relationships (SARs) were employed for the identification of new benzimidazole derivatives as potent competitive inhibitors of cruzain with trypanocidal activity and suitable pharmacokinetics. Extensive pharmacokinetic studies enabled the identification of metabolically stable and permeable compounds with high selectivity indices. CYP3A4 was found to be involved in the main metabolic pathway, and the identification of metabolic soft spots provided insights into molecular optimization. Compound 28, which showed a promising trade-off between pharmacodynamics and pharmacokinetics, caused no acute toxicity and reduced parasite burden both in vitro and in vivo.
Collapse
Affiliation(s)
- Ivani Pauli
- Laboratório de Química Medicinal e Computacional, Instituto de Física de São Carlos, Universidade de São Paulo, São Carlos, Brazil
| | - Celso de O Rezende
- Instituto de Química, Universidade Estadual de Campinas, Campinas, Brazil
| | - Brian W Slafer
- Instituto de Química, Universidade Estadual de Campinas, Campinas, Brazil
| | - Marco A Dessoy
- Instituto de Química, Universidade Estadual de Campinas, Campinas, Brazil
| | - Mariana L de Souza
- Laboratório de Química Medicinal e Computacional, Instituto de Física de São Carlos, Universidade de São Paulo, São Carlos, Brazil
| | - Leonardo L G Ferreira
- Laboratório de Química Medicinal e Computacional, Instituto de Física de São Carlos, Universidade de São Paulo, São Carlos, Brazil
| | - Abraham L M Adjanohun
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Rafaela S Ferreira
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luma G Magalhães
- Laboratório de Química Medicinal e Computacional, Instituto de Física de São Carlos, Universidade de São Paulo, São Carlos, Brazil
| | - Renata Krogh
- Laboratório de Química Medicinal e Computacional, Instituto de Física de São Carlos, Universidade de São Paulo, São Carlos, Brazil
| | - Simone Michelan-Duarte
- Laboratório de Química Medicinal e Computacional, Instituto de Física de São Carlos, Universidade de São Paulo, São Carlos, Brazil
| | | | | | - Fabio C Cruz
- Departamento de Farmacologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Luiz C Dias
- Instituto de Química, Universidade Estadual de Campinas, Campinas, Brazil
| | - Adriano D Andricopulo
- Laboratório de Química Medicinal e Computacional, Instituto de Física de São Carlos, Universidade de São Paulo, São Carlos, Brazil
| |
Collapse
|
5
|
Evaluation of Quantitative Structure Property Relationship Algorithms for Predicting Plasma Protein Binding in Humans. ACTA ACUST UNITED AC 2021; 17:100142. [PMID: 34017929 DOI: 10.1016/j.comtox.2020.100142] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The extent of plasma protein binding is an important compound-specific property that influences a compound's pharmacokinetic behavior and is a critical input parameter for predicting exposure in physiologically based pharmacokinetic (PBPK) modeling. When experimentally determined fraction unbound in plasma (fup) data are not available, quantitative structure-property relationship (QSPR) models can be used for prediction. Because available QSPR models were developed based on training sets containing pharmaceutical-like compounds, we compared their prediction accuracy for environmentally relevant and pharmaceutical compounds. Fup values were calculated using Ingle et al., Watanabe et al. and ADMET Predictor (Simulation Plus). The test set included 818 pharmaceutical and environmentally relevant compounds with fup values ranging from 0.01 to 1. Overall, the three QSPR models resulted in over-prediction of fup for highly binding compounds and under-prediction for low or moderately binding compounds. For highly binding compounds (0.01≤ fup ≤ 0.25), Watanabe et al. performed better with a lower mean absolute error (MAE) of 6.7% and a lower mean absolute relative prediction error (RPE) of 171.7 % than other methods. For low to moderately binding compounds, both Ingle et al. and ADMET Predictor performed better than Watanabe et al. with superior MAE and RPE values. The positive polar surface area, the number of basic functional groups and lipophilicity were the most important chemical descriptors for predicting fup. This study demonstrated that the prediction of fup was the most uncertain for highly binding compounds. This suggested that QSPR-predicted fup values should be used with caution in PBPK modeling.
Collapse
|
6
|
Miyamoto M, Kosugi Y, Iwasaki S, Chisaki I, Nakagawa S, Amano N, Hirabayashi H. Characterization of plasma protein binding in two mouse models of humanized liver, PXB mouse and humanized TK-NOG mouse. Xenobiotica 2020; 51:51-60. [PMID: 32779988 DOI: 10.1080/00498254.2020.1808735] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The unbound fractions in plasma (f up) in two mouse models of humanized liver mice, PXB and humanized TK-NOG mice, were compared with human f up values using equilibrium dialysis method. A good relationship between f up values obtained from PXB mice and humans was observed; the f up of 34/39 compounds (87.2%) in PXB mice were within 3-fold of human f up. In contrast, a weak correlation was observed between human and humanized TK-NOG mouse f up values; the f up of 15/24 compounds (62.5%) in humanized TK-NOG mice were within 3-fold of human f up. As different profiles of plasma protein binding (PPB) profiles were observed between PXB and humanized TK-NOG mice, f up evaluation is necessary in each mouse model to utilize these humanized liver mice for pharmacological, drug-drug interaction (DDI), and toxicity studies. The unbound fraction in the mixed plasma of human and SCID mouse plasma (85:15) was well correlated with f up in PXB mice (38/39 compounds within a 3-fold). Thus, this artificial PXB mouse plasma could be used to evaluate PPB.
Collapse
Affiliation(s)
- Maki Miyamoto
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, Fujisawa city, Japan
| | - Yohei Kosugi
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, Fujisawa city, Japan
| | - Shinji Iwasaki
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, Fujisawa city, Japan
| | - Ikumi Chisaki
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, Fujisawa city, Japan
| | - Sayaka Nakagawa
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, Fujisawa city, Japan
| | - Nobuyuki Amano
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, Fujisawa city, Japan
| | - Hideki Hirabayashi
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, Fujisawa city, Japan
| |
Collapse
|
7
|
Li Y, Evers R, Hafey MJ, Cheon K, Duong H, Lynch D, LaFranco-Scheuch L, Pacchione S, Tamburino AM, Tanis KQ, Geddes K, Holder D, Zhang NR, Kang W, Gonzalez RJ, Galijatovic-Idrizbegovic A, Pearson KM, Lebron JA, Glaab WE, Sistare FD. Use of a Bile Salt Export Pump Knockdown Rat Susceptibility Model to Interrogate Mechanism of Drug-Induced Liver Toxicity. Toxicol Sci 2020; 170:180-198. [PMID: 30903168 DOI: 10.1093/toxsci/kfz079] [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: 02/07/2023] Open
Abstract
Inhibition of the bile salt export pump (BSEP) may be associated with clinical drug-induced liver injury, but is poorly predicted by preclinical animal models. Here we present the development of a novel rat model using siRNA knockdown (KD) of Bsep that displayed differentially enhanced hepatotoxicity to 8 Bsep inhibitors and not to 3 Bsep noninhibitors when administered at maximally tolerated doses for 7 days. Bsep KD alone resulted in 3- and 4.5-fold increases in liver and plasma levels, respectively, of the sum of the 3 most prevalent taurine conjugated bile acids (T3-BA), approximately 90% decrease in plasma and liver glycocholic acid, and a distinct bile acid regulating gene expression pattern, without resulting in hepatotoxicity. Among the Bsep inhibitors, only asunaprevir and TAK-875 resulted in serum transaminase and total bilirubin increases associated with increases in plasma T3-BA that were enhanced by Bsep KD. Benzbromarone, lopinavir, and simeprevir caused smaller increases in plasma T3-BA, but did not result in hepatotoxicity in Bsep KD rats. Bosentan, cyclosporine A, and ritonavir, however, showed no enhancement of T3-BA in plasma in Bsep KD rats, as well as Bsep noninhibitors acetaminophen, MK-0974, or clarithromycin. T3-BA findings were further strengthened through monitoring TCA-d4 converted from cholic acid-d4 overcoming interanimal variability in endogenous bile acids. Bsep KD also altered liver and/or plasma levels of asunaprevir, TAK-875, TAK-875 acyl-glucuronide, benzbromarone, and bosentan. The Bsep KD rat model has revealed differences in the effects on bile acid homeostasis among Bsep inhibitors that can best be monitored using measures of T3-BA and TCA-d4 in plasma. However, the phenotype caused by Bsep inhibition is complex due to the involvement of several compensatory mechanisms.
Collapse
Affiliation(s)
- Yutai Li
- Safety Assessment and Laboratory Animal Resources
| | - Raymond Evers
- Pharmacokinetics, Pharmacodynamics and Drug Metabolism
| | | | | | - Hong Duong
- Safety Assessment and Laboratory Animal Resources
| | - Donna Lynch
- Safety Assessment and Laboratory Animal Resources
| | | | | | | | - Keith Q Tanis
- Genetics and Pharmacogenomics, MRL, West Point, PA 19486
| | | | | | | | - Wen Kang
- Safety Assessment and Laboratory Animal Resources
| | | | | | | | | | | | | |
Collapse
|
8
|
Shou WZ. Current status and future directions of high-throughput ADME screening in drug discovery. J Pharm Anal 2020; 10:201-208. [PMID: 32612866 PMCID: PMC7322755 DOI: 10.1016/j.jpha.2020.05.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/14/2020] [Accepted: 05/14/2020] [Indexed: 02/06/2023] Open
Abstract
During the last decade high-throughput in vitro absorption, distribution, metabolism and excretion (HT-ADME) screening has become an essential part of any drug discovery effort of synthetic molecules. The conduct of HT-ADME screening has been "industrialized" due to the extensive development of software and automation tools in cell culture, assay incubation, sample analysis and data analysis. The HT-ADME assay portfolio continues to expand in emerging areas such as drug-transporter interactions, early soft spot identification, and ADME screening of peptide drug candidates. Additionally, thanks to the very large and high-quality HT-ADME data sets available in many biopharma companies, in silico prediction of ADME properties using machine learning has also gained much momentum in recent years. In this review, we discuss the current state-of-the-art practices in HT-ADME screening including assay portfolio, assay automation, sample analysis, data processing, and prediction model building. In addition, we also offer perspectives in future development of this exciting field.
Collapse
Affiliation(s)
- Wilson Z. Shou
- Bristol-Myers Squibb, PO Box 4000, Princeton, NJ, 08540, USA
| |
Collapse
|
9
|
Ridder BJ, Leishman DJ, Bridgland-Taylor M, Samieegohar M, Han X, Wu WW, Randolph A, Tran P, Sheng J, Danker T, Lindqvist A, Konrad D, Hebeisen S, Polonchuk L, Gissinger E, Renganathan M, Koci B, Wei H, Fan J, Levesque P, Kwagh J, Imredy J, Zhai J, Rogers M, Humphries E, Kirby R, Stoelzle-Feix S, Brinkwirth N, Rotordam MG, Becker N, Friis S, Rapedius M, Goetze TA, Strassmaier T, Okeyo G, Kramer J, Kuryshev Y, Wu C, Himmel H, Mirams GR, Strauss DG, Bardenet R, Li Z. A systematic strategy for estimating hERG block potency and its implications in a new cardiac safety paradigm. Toxicol Appl Pharmacol 2020; 394:114961. [PMID: 32209365 PMCID: PMC7166077 DOI: 10.1016/j.taap.2020.114961] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 02/14/2020] [Accepted: 03/19/2020] [Indexed: 12/13/2022]
Abstract
Introduction hERG block potency is widely used to calculate a drug's safety margin against its torsadogenic potential. Previous studies are confounded by use of different patch clamp electrophysiology protocols and a lack of statistical quantification of experimental variability. Since the new cardiac safety paradigm being discussed by the International Council for Harmonisation promotes a tighter integration of nonclinical and clinical data for torsadogenic risk assessment, a more systematic approach to estimate the hERG block potency and safety margin is needed. Methods A cross-industry study was performed to collect hERG data on 28 drugs with known torsadogenic risk using a standardized experimental protocol. A Bayesian hierarchical modeling (BHM) approach was used to assess the hERG block potency of these drugs by quantifying both the inter-site and intra-site variability. A modeling and simulation study was also done to evaluate protocol-dependent changes in hERG potency estimates. Results A systematic approach to estimate hERG block potency is established. The impact of choosing a safety margin threshold on torsadogenic risk evaluation is explored based on the posterior distributions of hERG potency estimated by this method. The modeling and simulation results suggest any potency estimate is specific to the protocol used. Discussion This methodology can estimate hERG block potency specific to a given voltage protocol. The relationship between safety margin thresholds and torsadogenic risk predictivity suggests the threshold should be tailored to each specific context of use, and safety margin evaluation may need to be integrated with other information to form a more comprehensive risk assessment. hERG potency/safety margin is a widely used nonclinical cardiac safety strategy. A new regulatory paradigm promotes the integration of nonclinical and clinical data. Lack of uncertainty quantification hindered using hERG potency in the new paradigm. A systematic method was established to address this limitation. Analysis supports using different safety margin thresholds in different context.
Collapse
Affiliation(s)
- Bradley J Ridder
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA
| | - Derek J Leishman
- Department of Toxicology and Pathology, Eli Lilly and Company, Indianapolis, IN, USA
| | | | - Mohammadreza Samieegohar
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA
| | - Xiaomei Han
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA
| | - Wendy W Wu
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA
| | - Aaron Randolph
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA
| | - Phu Tran
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA
| | - Jiansong Sheng
- CiPA LAB, 900 Clopper Rd, Suite 130, Gaithersburg, MD 20878, USA
| | - Timm Danker
- NMI-TT GmbH, Markwiesenstr. 55, 72770 Reutlingen, Germany
| | | | - Daniel Konrad
- B'SYS GmbH, The Ion Channel Company, Benkenstrasse 254, CH-4108, Witterswil, Switzerland
| | - Simon Hebeisen
- B'SYS GmbH, The Ion Channel Company, Benkenstrasse 254, CH-4108, Witterswil, Switzerland
| | - Liudmila Polonchuk
- F. Hoffmann-La Roche AG, F. Hoffmann-La Roche Ltd Bldg. 73/R. 103b Grenzacherstrasse, 124, CH-4070 Basel, Switzerland
| | - Evgenia Gissinger
- F. Hoffmann-La Roche AG, F. Hoffmann-La Roche Ltd Bldg. 73/R. 103b Grenzacherstrasse, 124, CH-4070 Basel, Switzerland
| | | | - Bryan Koci
- Eurofins Scientific, Eurofins Discovery, 6 Research Park Drive, St. Charles, MO 63304, USA
| | - Haiyang Wei
- Eurofins Scientific, Eurofins Discovery, 6 Research Park Drive, St. Charles, MO 63304, USA
| | - Jingsong Fan
- Bristol-Myers Squibb Company, Discovery Toxicology, Bristol-Myers Squibb, 3551 Lawrenceville, Princeton Rd, Lawrence Township, NJ 08648, USA
| | - Paul Levesque
- Bristol-Myers Squibb Company, Discovery Toxicology, Bristol-Myers Squibb, 3551 Lawrenceville, Princeton Rd, Lawrence Township, NJ 08648, USA
| | - Jae Kwagh
- Bristol-Myers Squibb Company, Discovery Toxicology, Bristol-Myers Squibb, 3551 Lawrenceville, Princeton Rd, Lawrence Township, NJ 08648, USA
| | | | - Jin Zhai
- Merck & Co., Inc, Kenilworth, NJ, USA
| | - Marc Rogers
- Metrion Biosciences Limited, Riverside 3, Suite 1, Granta Park, Great Abington, Cambridge CB21, 6AD, United Kingdom
| | - Edward Humphries
- Metrion Biosciences Limited, Riverside 3, Suite 1, Granta Park, Great Abington, Cambridge CB21, 6AD, United Kingdom
| | - Robert Kirby
- Metrion Biosciences Limited, Riverside 3, Suite 1, Granta Park, Great Abington, Cambridge CB21, 6AD, United Kingdom
| | | | - Nina Brinkwirth
- Nanion Technologies Munich, Ganghoferstrasse 70A, 80339 Munich, Germany
| | | | - Nadine Becker
- Nanion Technologies Munich, Ganghoferstrasse 70A, 80339 Munich, Germany
| | - Søren Friis
- Nanion Technologies Munich, Ganghoferstrasse 70A, 80339 Munich, Germany
| | - Markus Rapedius
- Nanion Technologies Munich, Ganghoferstrasse 70A, 80339 Munich, Germany
| | - Tom A Goetze
- Nanion Technologies Munich, Ganghoferstrasse 70A, 80339 Munich, Germany
| | - Tim Strassmaier
- Nanion Technologies, USA, 1 Naylon Place, Suite C, Livingston, NJ 07039, USA
| | - George Okeyo
- Nanion Technologies, USA, 1 Naylon Place, Suite C, Livingston, NJ 07039, USA
| | - James Kramer
- Charles River Laboratories, 14656 Neo Parkway, Cleveland, OH 44128, USA
| | - Yuri Kuryshev
- Charles River Laboratories, 14656 Neo Parkway, Cleveland, OH 44128, USA
| | - Caiyun Wu
- Charles River Laboratories, 14656 Neo Parkway, Cleveland, OH 44128, USA
| | - Herbert Himmel
- Bayer AG, RD-TS-TOX-SP-SPL1, Aprather Weg 18a, 42096 Wuppertal, Germany
| | - Gary R Mirams
- Centre for Mathematical Medicine & Biology, School of Mathematical Sciences, University of Nottingham, Nottingham, United Kingdom
| | - David G Strauss
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA
| | - Rémi Bardenet
- Université de Lille, CNRS, Centrale Lille, UMR 9189 - CRIStAL, Villeneuve d'Ascq, France
| | - Zhihua Li
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA.
| |
Collapse
|
10
|
Industry Perspective on Contemporary Protein-Binding Methodologies: Considerations for Regulatory Drug-Drug Interaction and Related Guidelines on Highly Bound Drugs. J Pharm Sci 2017; 106:3442-3452. [DOI: 10.1016/j.xphs.2017.09.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 09/07/2017] [Indexed: 11/21/2022]
|
11
|
Miyamoto M, Iwasaki S, Chisaki I, Nakagawa S, Amano N, Hirabayashi H. Comparison of predictability for human pharmacokinetics parameters among monkeys, rats, and chimeric mice with humanised liver. Xenobiotica 2017; 47:1052-1063. [DOI: 10.1080/00498254.2016.1265160] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Maki Miyamoto
- Drug Metabolism and Pharmacokinetics Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Shinji Iwasaki
- Drug Metabolism and Pharmacokinetics Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Ikumi Chisaki
- Drug Metabolism and Pharmacokinetics Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Sayaka Nakagawa
- Drug Metabolism and Pharmacokinetics Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Nobuyuki Amano
- Drug Metabolism and Pharmacokinetics Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Hideki Hirabayashi
- Drug Metabolism and Pharmacokinetics Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| |
Collapse
|
12
|
Hecht ES, Oberg AL, Muddiman DC. Optimizing Mass Spectrometry Analyses: A Tailored Review on the Utility of Design of Experiments. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2016; 27:767-85. [PMID: 26951559 PMCID: PMC4841694 DOI: 10.1007/s13361-016-1344-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 01/14/2016] [Accepted: 01/16/2016] [Indexed: 05/07/2023]
Abstract
Mass spectrometry (MS) has emerged as a tool that can analyze nearly all classes of molecules, with its scope rapidly expanding in the areas of post-translational modifications, MS instrumentation, and many others. Yet integration of novel analyte preparatory and purification methods with existing or novel mass spectrometers can introduce new challenges for MS sensitivity. The mechanisms that govern detection by MS are particularly complex and interdependent, including ionization efficiency, ion suppression, and transmission. Performance of both off-line and MS methods can be optimized separately or, when appropriate, simultaneously through statistical designs, broadly referred to as "design of experiments" (DOE). The following review provides a tutorial-like guide into the selection of DOE for MS experiments, the practices for modeling and optimization of response variables, and the available software tools that support DOE implementation in any laboratory. This review comes 3 years after the latest DOE review (Hibbert DB, 2012), which provided a comprehensive overview on the types of designs available and their statistical construction. Since that time, new classes of DOE, such as the definitive screening design, have emerged and new calls have been made for mass spectrometrists to adopt the practice. Rather than exhaustively cover all possible designs, we have highlighted the three most practical DOE classes available to mass spectrometrists. This review further differentiates itself by providing expert recommendations for experimental setup and defining DOE entirely in the context of three case-studies that highlight the utility of different designs to achieve different goals. A step-by-step tutorial is also provided.
Collapse
Affiliation(s)
- Elizabeth S Hecht
- W. M. Keck FTMS Laboratory for Human Health Research, Department of Chemistry, North Carolina State University, Raleigh, NC, 27695, USA
| | - Ann L Oberg
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - David C Muddiman
- W. M. Keck FTMS Laboratory for Human Health Research, Department of Chemistry, North Carolina State University, Raleigh, NC, 27695, USA.
| |
Collapse
|
13
|
Amaral A, Saran C, Amin J, Hatsis P. A Comparison of LC-MS/MS and a Fully Integrated Autosampler/Solid-Phase Extraction System for the Analysis of Protein Binding Samples. ACTA ACUST UNITED AC 2016; 21:620-5. [DOI: 10.1177/1087057116630706] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
A new analysis approach was evaluated for measuring plasma protein binding (PPB) of small molecules using the Agilent RapidFire high-throughput system coupled with a Sciex API 4000 mass spectrometer (RF-MS/MS). Thirty-three proprietary and 12 literature compounds were subjected to rapid equilibrium dialysis (RED) and evaluated in parallel using RF-MS/MS at 16.4 s/sample and traditional liquid chromatography–tandem mass spectrometry (LC-MS/MS) at 3.5 min/sample, thus making the RF-MS/MS analysis over 12 times faster than LC-MS/MS. The high-throughput analysis method that was developed demonstrated excellent correlation with the traditional LC-MS/MS analysis method with an r2 value of 0.96. The RF-MS/MS analysis method was implemented to increase sample throughput, decrease turnaround time for PPB data, and decrease time burden on existing LC-MS/MS instruments.
Collapse
Affiliation(s)
- Adam Amaral
- Drug Metabolism & Pharmacokinetics, Biogen, Inc., Cambridge, MA, USA
| | - Chitra Saran
- Analytical Sciences & Imaging, Novartis Institutes for Biomedical Research, Inc., Cambridge, MA, USA
| | - Jakal Amin
- Drug Metabolism & Pharmacokinetics, Agios Pharmaceuticals, Cambridge, MA 02139 USA
| | - Panos Hatsis
- Drug Metabolism & Pharmacokinetics, Novartis Institutes for Biomedical Research, Inc., East Hanover, NJ, USA
| |
Collapse
|