1
|
Poulin P. Which Unbound Fraction Should We Use in the Well-Stirred Model for More Accurately Predicting Hepatic Clearance of Drugs for Humans? J Pharm Sci 2025:103827. [PMID: 40414348 DOI: 10.1016/j.xphs.2025.103827] [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: 03/05/2025] [Revised: 05/07/2025] [Accepted: 05/07/2025] [Indexed: 05/27/2025]
Abstract
As the hepatic clearance (CLH) of drugs becomes independent of hepatic blood flow rate, CLH depends primarily on intrinsic clearance (CLint). Incubations of microsomes or hepatocytes are commonly used to generate CLint. Therefore, CLint estimates corrected for binding to the in vitro systems and scaled to whole liver, are applied to a well-stirred liver model to obtain CLH predictions for drugs. In other words, CLint is extrapolated with the ratio of unbound fraction between the plasma (fup) and incubation medium (fuinc). However, this binding correction resulted to an important underprediction bias of CLH. Therefore, the approach considering fup and fuinc needs to be better understood for more precisely scaling CLint. The objective of this study was to explain the underprediction bias of CLH based on physicochemical properties of drugs. Analysis-ready datasets have been collected so that evaluation of the data generates a mechanistic understanding of the impact of unbound fraction on the prediction of CLH of human for 128 drugs. Experimental values of fuinc for liver are quantifying solely the binding to lipids in microsomes or hepatocytes in the absence of plasma proteins in the incubations. However, the experimental values of fup for plasma can estimate the binding to lipids and plasma proteins. Therefore, drugs binding primarily to lipids in the liver and plasma showed a less pronounced underprediction bias of CLH by using the ratios of fup/fuinc in the well-stirred model. In contrast, drugs binding primarily to the plasma proteins in the liver and plasma showed a larger underprediction bias of CLH. Furthermore, for the ionized drugs, values of fup and fuinc are not covering the pH gradient effect between plasma and hepatocytes, which also impacted the CLH predictions. For these reasons, a mechanistic approach was proposed to replace the conventional fup value with an adjusted fup (fu-adjusted) that accounts for differences in proteins/lipids binding and pH gradient effect between the liver and plasma. Hence, replacing fup with fu-adjusted did provide a universal and mechanisms-based approach removing the underprediction bias for all datasets of drugs. Overall, this study indicates which drug properties generated the largest underprediction bias of CLH and suggests that the Poulin et al. method referring to fu-adjusted could be the most proper unbound fraction to reduce that bias with the well-stirred model.
Collapse
Affiliation(s)
- Patrick Poulin
- Consultant Patrick Poulin Inc., Québec City, Québec, Canada; School of Public Health, Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|
2
|
Werner S, Pamies D, Zurich MG, Suter-Dick L. Hepatic and extra-hepatic metabolism of propylene glycol ethers in the context of central nervous system toxicity. Toxicology 2025; 512:154081. [PMID: 39929342 DOI: 10.1016/j.tox.2025.154081] [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: 01/09/2025] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/14/2025]
Abstract
Propylene glycol ethers (PGEs) are mixtures of an α-isomer and a β-isomer (β-PGE) that is oxidized via alcohol dehydrogenase (ADH) and aldehyde dehydrogenase (ALDH) to potentially neurotoxic alkoxy propionic acids (β-metabolites). While the liver is the primary organ for ADH- and ALDH-mediated metabolism, the contribution to the metabolism of β-PGEs by the blood-brain barrier (BBB) and the brain remains unknown. Here, we aimed to assess the neurotoxic potential of PGEs after systemic exposure by (1) comparing 3D HepaRG and human liver subcellular fraction (S9) for the in vitro determination of the kinetics of hepatic metabolism for β-PGEs, (2) evaluating the BBB-permeability of PGEs and β-metabolites, (3) determining the presence of ADH1 and ALDH2 and the extent of metabolization of β-PGEs in the BBB and brain. The results show that 3D HepaRG and S9 served as competent systems to estimate the enzymatic kinetic (clearance) for β-metabolite formation. We observed that PGEs and the β-metabolites could cross the BBB, based on their permeance across a cellular barrier consisting of the hCMEC/D3 cell line. Metabolic enzymes were not exclusive to the liver, as expression of ADH1 and ALDH2 was demonstrated using RT-qPCR, Western blot, and immunostainings in the BBB in vitro models and in BrainSpheres. Furthermore, LC-MS/MS quantification of the β-metabolites in all in vitro models revealed that 3D HepaRG had a similar metabolic capacity to primary human hepatocytes and that the amount of β-metabolite formed per protein in the BBB was approximately 10-30 % of that in the liver. We also demonstrated active metabolism in the BrainSpheres. In conclusion, the hepatic in vitro models provided data that will help to refine toxicokinetic models and predict internal exposures, thereby supporting the risk assessment of PGEs. In addition, the high permeance of the PGEs and the β-metabolites across the BBB increases the plausibility of neurotoxicity upon systemic exposure. This is further supported by the presence of active ADH1 and ALDH2 enzymes in the BBB in vitro systems and in BrainSpheres, suggesting metabolite formation in the central nervous system. Hence, we suggest that BBB-permeance and extra-hepatic metabolism of the β-PGEs may contribute to the neurotoxicity of PGEs.
Collapse
Affiliation(s)
- Sophie Werner
- University of Applied Sciences and Arts Northwestern Switzerland, School of Life Sciences, Muttenz, Switzerland; Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland; Swiss Centre for Applied Human Toxicology (SCAHT), Basel, Switzerland.
| | - David Pamies
- Swiss Centre for Applied Human Toxicology (SCAHT), Basel, Switzerland; Department of Biomedical Science, University of Lausanne, Lausanne, Switzerland.
| | - Marie-Gabrielle Zurich
- Swiss Centre for Applied Human Toxicology (SCAHT), Basel, Switzerland; Department of Biomedical Science, University of Lausanne, Lausanne, Switzerland.
| | - Laura Suter-Dick
- University of Applied Sciences and Arts Northwestern Switzerland, School of Life Sciences, Muttenz, Switzerland; Swiss Centre for Applied Human Toxicology (SCAHT), Basel, Switzerland.
| |
Collapse
|
3
|
Kukla DA, Schulz Pauly JA, Lesniak PR, Sande E, Wang YT, Kalvass JC, Stresser DM. Clearance prediction with three novel plated human hepatocyte models compared to conventional suspension assays: Assessment with 50 compounds and multiple donors. Drug Metab Dispos 2025; 53:100032. [PMID: 40023578 DOI: 10.1016/j.dmd.2024.100032] [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: 09/27/2024] [Accepted: 12/06/2024] [Indexed: 03/04/2025] Open
Abstract
Incubation of drugs with suspension hepatocytes (SH) to determine intrinsic clearance is common in drug discovery. However, the limited duration of SH assays hampers clearance assessment of metabolically stable compounds. In turn, this has driven the development of alternative in vitro approaches to generate intrinsic clearance estimates. Culturing primary hepatocytes with supportive cells as co/tricultures has been shown to maintain morphology, viability, and drug-metabolizing enzyme function for weeks, permitting extended incubations. Another assay from our laboratory is the preloaded hepatocyte assay (preload assay), which involves preloading plated monoculture hepatocytes with compounds and measuring the loss from cells in drug-free media. This approach increases analytical sensitivity compared to assays that measure bulk compound loss in the cells plus medium. We conducted a systematic evaluation of the ability of coculture, triculture, and preload assay models to predict human in vivo clearance for 50 predominantly low-clearance compounds with a range of physicochemical properties, including equal numbers of compounds following or violating Lipinski's rule of 5, across 3 hepatocyte donors. The results were compared with SH. Co/tricultures exhibited lower inter-donor differences compared to the preload and SH assays, likely due to the blunting of environmental cues after 5 days in culture prior to compound introduction. All 3 plated models significantly reduced the number of compounds with insufficient turnover to calculate CLint,u compared to SH (SH: 40%; preload: 18%; cocultures: 8%; tricultures: 4%), exhibited strong interexperimental reproducibility and robust predictions of blood clearance (preload: 26/41; cocultures: 31/46; tricultures: 30/48 within 3-fold of observed). SIGNIFICANCE STATEMENT: Preloading plated hepatocytes with compounds and measuring the loss in drug-free media, or culturing hepatocytes with supportive cells as co/tricultures, facilitate quantitation of metabolically stable compounds in substrate depletion assays compared to suspension hepatocytes (SH). All 4 models exhibit robust estimates of CLint,u and CLb, but plated models allowed assessment of several compounds found to be too stable to evaluate in SH.
Collapse
Affiliation(s)
- David A Kukla
- Quantitative, Translational, & ADME Sciences (QTAS), AbbVie Inc, North Chicago, Illinois
| | - Julia A Schulz Pauly
- Quantitative, Translational, & ADME Sciences (QTAS), AbbVie Inc, North Chicago, Illinois
| | - Paul R Lesniak
- Quantitative, Translational, & ADME Sciences (QTAS), AbbVie Inc, North Chicago, Illinois
| | - Elizabeth Sande
- Quantitative, Translational, & ADME Sciences (QTAS), AbbVie Inc, North Chicago, Illinois
| | - Yue-Ting Wang
- Quantitative, Translational, & ADME Sciences (QTAS), AbbVie Inc, North Chicago, Illinois
| | - John Cory Kalvass
- Quantitative, Translational, & ADME Sciences (QTAS), AbbVie Inc, North Chicago, Illinois
| | - David M Stresser
- Quantitative, Translational, & ADME Sciences (QTAS), AbbVie Inc, North Chicago, Illinois.
| |
Collapse
|
4
|
Ohri S, Parekh P, Nichols L, Rajan SAP, Cirit M. Utilization of a human Liver Tissue Chip for drug-metabolizing enzyme induction studies of perpetrator and victim drugs. Drug Metab Dispos 2025; 53:100004. [PMID: 39884808 DOI: 10.1124/dmd.124.001497] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/30/2024] [Accepted: 09/30/2024] [Indexed: 10/09/2024] Open
Abstract
Polypharmacy-related drug-drug interactions (DDIs) are a significant and growing healthcare concern. An increasing number of therapeutic drugs on the market underscores the necessity to accurately assess new drug combinations during preclinical evaluation for DDIs. In vitro primary human hepatocytes (PHH) models are only applicable for short-term induction studies because of their rapid loss of metabolic function. Though coculturing nonhuman stromal cells with PHH has been shown to stabilize metabolic activity long-term, there are concerns about human specificity for accurate clinical assessment. In this study, we demonstrated a PHH-only liver microphysiological system in the Liver Tissue Chip is capable of maintaining long-term functional and metabolic activity of PHH from 3 individual donors and thus a suitable platform for long-term DDI induction studies. The responses to rifampicin induction of 3 PHH donors were assessed using cytochrome P450 activity and mRNA changes. Additionally, victim pharmacokinetic studies were conducted with midazolam (high clearance) and alprazolam (low clearance) following perpetrator drug treatment, rifampicin-mediated induction, which resulted in a 2-fold and a 2.6-fold increase in midazolam and alprazolam intrinsic clearance values, respectively, compared with the untreated liver microphysiological system. We also investigated the induction effects of different dosing regimens of the perpetrator drug (rifampicin) on cytochrome P450 activity levels, showing minimal variation in the intrinsic clearance of the victim drug (midazolam). This study illustrates the utility of the Liver Tissue Chip for in vitro liver-specific DDI induction studies, providing a translational experimental system to predict clinical clearance values of both perpetrator and victim drugs. SIGNIFICANCE STATEMENT: This study demonstrated the utility of the Liver Tissue Chip with a primary human hepatocyte-only liver microphysiological system for drug-drug interaction induction studies. This unique in vitro system with continuous recirculation maintains long-term functionality and metabolic activity for up to 4 weeks, enabling the study of perpetrator and victim drug pharmacokinetics, quantification of drug-induced cytochrome P450 mRNA and activity levels, investigation of patient variability, and ultimately clinical predictions.
Collapse
Affiliation(s)
| | | | | | | | - Murat Cirit
- Javelin Biotech, Inc, Woburn, Massachusetts.
| |
Collapse
|
5
|
Werner S, Hegg L, Hopf NB, Borgatta M, Suter‐Dick L. In Vitro Hepatic Metabolism Input Parameters Support Toxicokinetic Simulations for the Formation of Methoxy Propionic Acid From β-Isomer Propylene Glycol Methyl Ether. Pharmacol Res Perspect 2024; 12:e70037. [PMID: 39655648 PMCID: PMC11629119 DOI: 10.1002/prp2.70037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 11/01/2024] [Indexed: 12/13/2024] Open
Abstract
Propylene glycol ethers (PGEs) are organic solvents commonly found as technical grade on the commercial market, as mixtures of secondary (α-isomer) and primary (β-isomer, generally < 5%) alcohols. After handling products containing PGEs, they readily enter the human body where they are metabolized. The minor β-isomer is oxidized by alcohol dehydrogenase (ADH) followed by aldehyde dehydrogenase (ALDH) to a potentially harmful metabolite. Although the enzymatic rate is needed to estimate both parent and metabolite internal exposures, kinetic data for many PGEs are still scarce. Therefore, we generated in vitro hepatic intrinsic clearance data for propylene glycol methyl ether β-isomer (β-PGME) and its metabolite methoxy propionic acid (2-MPA) and integrated these data into an in silico toxicokinetic (TK) model. Hepatic clearance values for the model were generated using an established in vitro 3D culture of the human HepaRG cell line and human S9 liver fraction. Our results showed the presence of ADH and ALDH and consequently, the formation of 2-MPA in the 3D HepaRG and S9 fraction, which was slow to medium. We integrated the hepatic clearance values into the TK model to predict urinary 2-MPA concentrations. The simulated urinary 2-MPA concentrations fitted well (within twofold error from observed experimental data) for both liver systems, showing that they were both able to reliably predict the hepatic clearance of β-PGME. Although S9 is suitable for short-term studies, 3D cell culture models maintain metabolic competence over days and weeks. This opens the opportunity for long-term metabolism studies applying the 3D HepaRG model alone or in multi-organ systems.
Collapse
Affiliation(s)
- Sophie Werner
- School of Life SciencesUniversity of Applied Sciences and Arts Northwestern SwitzerlandMuttenzSwitzerland
- Department of Pharmaceutical SciencesUniversity of BaselBaselSwitzerland
- Swiss Centre for Applied Human Toxicology (SCAHT)BaselSwitzerland
| | - Lucie Hegg
- Swiss Centre for Applied Human Toxicology (SCAHT)BaselSwitzerland
- Center for Primary Care and Public Health (Unisanté)University of LausanneLausanneSwitzerland
| | - Nancy B. Hopf
- Swiss Centre for Applied Human Toxicology (SCAHT)BaselSwitzerland
- Center for Primary Care and Public Health (Unisanté)University of LausanneLausanneSwitzerland
| | - Myriam Borgatta
- Swiss Centre for Applied Human Toxicology (SCAHT)BaselSwitzerland
- Center for Primary Care and Public Health (Unisanté)University of LausanneLausanneSwitzerland
| | - Laura Suter‐Dick
- School of Life SciencesUniversity of Applied Sciences and Arts Northwestern SwitzerlandMuttenzSwitzerland
- Swiss Centre for Applied Human Toxicology (SCAHT)BaselSwitzerland
| |
Collapse
|
6
|
Savaryn JP, Coe K, Cerny MA, Colizza K, Moliner P, King L, Ma B, Atherton J, Auclair A, Cancilla MT, Eno M, Jurva U, Yue Q, Zhu SX, Freiberger E, Zhong G, Barlock B, Nachtigall J, Laboureur L, Pusalkar S, Guo R, Niehues M, Hauri S, Carreras ET, Maurer C, Prakash C, Jenkins GJ. The Current State of Biotransformation Science - Industry Survey of In Vitro and In Vivo Practices, Clinical Translation, and Future Trends. Pharm Res 2024; 41:2079-2093. [PMID: 39496990 PMCID: PMC11599300 DOI: 10.1007/s11095-024-03787-y] [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: 08/01/2024] [Accepted: 10/17/2024] [Indexed: 11/06/2024]
Abstract
Embedded within the field of drug metabolism and pharmacokinetics (DMPK), biotransformation is a discipline that studies the origins, disposition, and structural identity of metabolites to provide a comprehensive safety assessment, including the assessment of exposure coverage in toxicological species. Spanning discovery and development, metabolite identification (metID) scientists employ various strategies and tools to address stage-specific questions aimed at guiding the maturation of early chemical matter into drug candidates. During this process, the identity of major (and minor) circulating human metabolites is ascertained to comply with the regulatory requirements such as the Metabolites in Safety Testing (MIST) guidance. Through the International Consortium for Innovation and Quality in Pharmaceutical Development (IQ), the "Translatability of MetID In Vitro Systems Working Group" was created within the Translational and ADME Sciences Leadership Group. The remit of this group was to objectively determine how accurate commonly employed in vitro systems have been with respect to prediction of circulating human metabolites, both qualitatively and quantitatively. A survey composed of 34 questions was conducted across 26 pharmaceutical companies to obtain a foundational understanding of current metID practices, preclinically and clinically, as well as to provide perspective on how successful these practices have been at predicting circulating human metabolites. The results of this survey are presented as an initial snapshot of current industry-based metID practices, including our perspective on how a harmonized framework for the conduct of in vitro metID studies could be established. Future perspectives from current practices to emerging advances with greater translational capability are also provided.
Collapse
Affiliation(s)
- John P Savaryn
- AbbVie, Quantitative, Translational & ADME Sciences, North Chicago, IL, USA.
| | - Kevin Coe
- J&J, Translational PKPD & Investigational Toxicology, San Diego, CA, USA
| | | | - Kevin Colizza
- GSK, DMPK Disposition and Biotransformation, Collegeville, PA, USA.
| | | | - Lloyd King
- UCB Biopharma, Dept. of DMPK, Slough, UK
| | - Bin Ma
- Genentech, Inc., Department of Drug Metabolism and Pharmacokinetics, South San Francisco, CA, USA
| | - Jim Atherton
- Incyte Research Institute, Translational Sciences, Wilmington, DE, USA
| | - Adam Auclair
- Boehringer Ingelheim Pharmaceuticals, Inc.,Drug Metabolism and Pharmacokinetics, Ridgefield, CT, USA
| | - Mark T Cancilla
- Merck & Co., Inc., Pharmacokinetics, Dynamics, Metabolism, and Bioanalysis, Rahway, NJ, USA
| | - Marsha Eno
- Eisai Inc., Global Drug Metabolism and Pharmacokinetics, Cambridge, MA, USA
| | - Ulrik Jurva
- AstraZeneca, Drug Metabolism and Pharmacokinetics (DMPK), Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, Gothenburg, Sweden
| | - Qin Yue
- Gilead Sciences, Inc., Drug Metabolism Dept, Foster City, CA, USA
| | - Sean Xiaochun Zhu
- Takeda Development Center Americas, Inc., Drug Metabolism and Pharmacokinetics & Modeling, Cambridge, MA, USA
| | - Elyse Freiberger
- AbbVie, Quantitative, Translational & ADME Sciences, North Chicago, IL, USA
| | - Guo Zhong
- Amgen, Pharmacokinetics and Drug Metabolism Department, South San Francisco, CA, USA
| | | | | | | | | | - Runcong Guo
- Beigene, DMPK, Department of Biology, Shanghai, China
| | - Michael Niehues
- Bayer AG, In Vitro ADME & Isotope Chemistry, Berlin, Germany
| | - Simon Hauri
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| | - Ester Tor Carreras
- Novartis Pharma AG, Novartis Institute for Biomedical Research, Basel, Switzerland
| | | | - Chandra Prakash
- DMPK/Clinical Pharmacology, Agios Pharmaceuticals, Cambridge, MA, USA
| | - Gary J Jenkins
- AbbVie, Quantitative, Translational & ADME Sciences, North Chicago, IL, USA
| |
Collapse
|
7
|
Schulz Pauly JA, Sande E, Feng M, Wang YT, Stresser DM, Kalvass JC. Proof of Concept of an All-in-One System for Measuring Hepatic Influx, Egress, and Metabolic Clearance Based on the Extended Clearance Concept. Drug Metab Dispos 2024; 52:1048-1059. [PMID: 39095207 DOI: 10.1124/dmd.124.001768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 08/04/2024] Open
Abstract
Hepatic clearance (CLH ) prediction is a critical parameter to estimate human dose. However, CLH underpredictions are common, especially for slowly metabolized drugs, and may be attributable to drug properties that pose challenges for conventional in vitro absorption, distribution, metabolism, and elimination (ADME) assays, resulting in nonvalid data, which prevents in vitro to in vivo extrapolation and CLH predictions. Other processes, including hepatocyte and biliary distribution via transporters, can also play significant roles in CLH Recent advances in understanding the interplay of metabolism and drug transport for clearance processes have aided in developing the extended clearance model. In this study, we demonstrate proof of concept of a novel two-step assay enabling the measurement of multiple kinetic parameters from a single experiment in plated human primary hepatocytes with and without transporter and cytochrome P450 inhibitors-the hepatocyte uptake and loss assay (HUpLA). HUpLA accurately predicted the CLH of eight of the nine drugs (within twofold of the observed CLH ). Distribution clearances were within threefold of observed literature values in standard uptake and efflux assays. In comparison, the conventional suspension hepatocyte stability assay poorly predicted the CLH The CLH of only two drugs was predicted within twofold of the observed CLH Therefore, HUpLA is advantageous by enabling the measurement of enzymatic and transport processes concurrently within the same system, alleviating the need for applying scaling factors independently. The use of primary human hepatocytes enables physiologically relevant exploration of transporter-enzyme interplay. Most importantly, HUpLA shows promise as a sensitive measure for low-turnover drugs. Further evaluation across different drug characteristics is needed to demonstrate method robustness. SIGNIFICANCE STATEMENT: The hepatocyte uptake and loss assay involves measuring four commonly derived in vitro hepatic clearance endpoints. Since endpoints are generated within a single test system, it blunts experimental error originating from assays otherwise conducted independently. A key advantage is the concept of removing drug-containing media following intracellular drug loading, enabling the measurement of drug reappearance rate in media as well as the measurement of loss of total drug in the test system unencumbered by background quantities of drug in media otherwise present in a conventional assay.
Collapse
Affiliation(s)
- Julia A Schulz Pauly
- Quantitative, Translational, and ADME Sciences, Abbvie Inc., North Chicago, Illinois
| | - Elizabeth Sande
- Quantitative, Translational, and ADME Sciences, Abbvie Inc., North Chicago, Illinois
| | - Mei Feng
- Quantitative, Translational, and ADME Sciences, Abbvie Inc., North Chicago, Illinois
| | - Yue-Ting Wang
- Quantitative, Translational, and ADME Sciences, Abbvie Inc., North Chicago, Illinois
| | - David M Stresser
- Quantitative, Translational, and ADME Sciences, Abbvie Inc., North Chicago, Illinois
| | - John Cory Kalvass
- Quantitative, Translational, and ADME Sciences, Abbvie Inc., North Chicago, Illinois
| |
Collapse
|
8
|
Preiss LC, Georgi K, Lauschke VM, Petersson C. Comparison of Human Long-Term Liver Models for Clearance Prediction of Slowly Metabolized Compounds. Drug Metab Dispos 2024; 52:539-547. [PMID: 38604730 DOI: 10.1124/dmd.123.001638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/06/2024] [Accepted: 03/19/2024] [Indexed: 04/13/2024] Open
Abstract
The accurate prediction of human clearance is an important task during drug development. The proportion of low clearance compounds has increased in drug development pipelines across the industry since such compounds may be dosed in lower amounts and at lower frequency. These type of compounds present new challenges to in vitro systems used for clearance extrapolation. In this study, we compared the accuracy of clearance predictions of suspension culture to four different long-term stable in vitro liver models, including HepaRG sandwich culture, the Hµrel stochastic co-culture, the Hepatopac micropatterned co-culture (MPCC), and a micro-array spheroid culture. Hepatocytes in long-term stable systems remained viable and active over several days of incubation. Although intrinsic clearance values were generally high in suspension culture, clearance of low turnover compounds could frequently not be determined using this method. Metabolic activity and intrinsic clearance values from HepaRG cultures were low and, consequently, many compounds with low turnover did not show significant decline despite long incubation times. Similarly, stochastic co-cultures occasionally failed to show significant turnover for multiple low and medium turnover compounds. Among the different methods, MPCCs and spheroids provided the most consistent measurements. Notably, all culture methods resulted in underprediction of clearance; this could, however, be compensated for by regression correction. Combined, the results indicate that spheroid culture as well as the MPCC system provide adequate in vitro tools for human extrapolation for compounds with low metabolic turnover. SIGNIFICANCE STATEMENT: In this study, we compared suspension cultures, HepaRG sandwich cultures, the Hµrel liver stochastic co-cultures, the Hepatopac micropatterned co-cultures (MPCC), and micro-array spheroid cultures for low clearance determination and prediction. Overall, HepaRG and suspension cultures showed modest value for the low determination and prediction of clearance compounds. The micro-array spheroid culture resulted in the most robust clearance measurements, whereas using the MPCC resulted in the most accurate prediction for low clearance compounds.
Collapse
Affiliation(s)
- Lena C Preiss
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (L.C.P., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), The Healthcare Business of Merck KGaA, Darmstadt, Germany (L.C.P., K.G., C.P.); Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.); and University of Tuebingen, Tuebingen, Germany (V.M.L.)
| | - Katrin Georgi
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (L.C.P., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), The Healthcare Business of Merck KGaA, Darmstadt, Germany (L.C.P., K.G., C.P.); Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.); and University of Tuebingen, Tuebingen, Germany (V.M.L.)
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (L.C.P., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), The Healthcare Business of Merck KGaA, Darmstadt, Germany (L.C.P., K.G., C.P.); Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.); and University of Tuebingen, Tuebingen, Germany (V.M.L.)
| | - Carl Petersson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (L.C.P., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), The Healthcare Business of Merck KGaA, Darmstadt, Germany (L.C.P., K.G., C.P.); Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.); and University of Tuebingen, Tuebingen, Germany (V.M.L.)
| |
Collapse
|
9
|
Aluri KC, Slavsky M, Tan Y, Whitcher‐Johnstone A, Zhang Z, Hariparsad N, Ramsden D. Aminobenzotriazole inhibits and induces several key drug metabolizing enzymes complicating its utility as a pan CYP inhibitor for reaction phenotyping. Clin Transl Sci 2024; 17:e13746. [PMID: 38501263 PMCID: PMC10949176 DOI: 10.1111/cts.13746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 03/20/2024] Open
Abstract
Aminobenzotriazole (ABT) is commonly used as a non-selective inhibitor of cytochrome P450 (CYP) enzymes to assign contributions of CYP versus non-CYP pathways to the metabolism of new chemical entities. Despite widespread use, a systematic review of the drug-drug interaction (DDI) potential for ABT has not been published nor have the implications for using it in plated hepatocyte models for low clearance reaction phenotyping. The goal being to investigate the utility of ABT as a pan-CYP inhibitor for reaction phenotyping of low clearance compounds by evaluating stability over the incubation period, inhibition potential against UGT and sulfotransferase enzymes, and interaction with nuclear receptors involved in the regulation of drug metabolizing enzymes and transporters. Induction potential for additional inhibitors used to ascribe fraction metabolism (fm ), pathway including erythromycin, ketoconazole, azamulin, atipamezole, ZY12201, and quinidine was also investigated. ABT significantly inhibited the clearance of a non-selective UGT substrate 4-methylumbelliferone, with several UGTs shown to be inhibited using selective probe substrates in human hepatocytes and rUGTs. The inhibitors screened in the induction assay were shown to induce enzymes regulated through Aryl Hydrocarbon Receptor, Constitutive Androstane Receptor, and Pregnane X Receptor. Lastly, a case study identifying the mechanisms of a clinical DDI between Palbociclib and ARV-471 is provided as an example of the potential consequences of using ABT to derive fm . This work demonstrates that ABT is not an ideal pan-CYP inhibitor for reaction phenotyping of low clearance compounds and establishes a workflow that can be used to enable robust characterization of other prospective inhibitors.
Collapse
Affiliation(s)
| | | | - Ying Tan
- AstraZenecaWalthamMassachusettsUSA
| | | | | | | | | |
Collapse
|
10
|
Izat N, Bolleddula J, Abbasi A, Cheruzel L, Jones RS, Moss D, Ortega-Muro F, Parmentier Y, Peterkin VC, Tian DD, Venkatakrishnan K, Zientek MA, Barber J, Houston JB, Galetin A, Scotcher D. Challenges and Opportunities for In Vitro-In Vivo Extrapolation of Aldehyde Oxidase-Mediated Clearance: Toward a Roadmap for Quantitative Translation. Drug Metab Dispos 2023; 51:1591-1606. [PMID: 37751998 DOI: 10.1124/dmd.123.001436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 09/28/2023] Open
Abstract
Underestimation of aldehyde oxidase (AO)-mediated clearance by current in vitro assays leads to uncertainty in human dose projections, thereby reducing the likelihood of success in drug development. In the present study we first evaluated the current drug development practices for AO substrates. Next, the overall predictive performance of in vitro-in vivo extrapolation of unbound hepatic intrinsic clearance (CLint,u) and unbound hepatic intrinsic clearance by AO (CLint,u,AO) was assessed using a comprehensive literature database of in vitro (human cytosol/S9/hepatocytes) and in vivo (intravenous/oral) data collated for 22 AO substrates (total of 100 datapoints from multiple studies). Correction for unbound fraction in the incubation was done by experimental data or in silico predictions. The fraction metabolized by AO (fmAO) determined via in vitro/in vivo approaches was found to be highly variable. The geometric mean fold errors (gmfe) for scaled CLint,u (mL/min/kg) were 10.4 for human hepatocytes, 5.6 for human liver cytosols, and 5.0 for human liver S9, respectively. Application of these gmfe's as empirical scaling factors improved predictions (45%-57% within twofold of observed) compared with no correction (11%-27% within twofold), with the scaling factors qualified by leave-one-out cross-validation. A road map for quantitative translation was then proposed following a critical evaluation on the in vitro and clinical methodology to estimate in vivo fmAO In conclusion, the study provides the most robust system-specific empirical scaling factors to date as a pragmatic approach for the prediction of in vivo CLint,u,AO in the early stages of drug development. SIGNIFICANCE STATEMENT: Confidence remains low when predicting in vivo clearance of AO substrates using in vitro systems, leading to de-prioritization of AO substrates from the drug development pipeline to mitigate risk of unexpected and costly in vivo impact. The current study establishes a set of empirical scaling factors as a pragmatic tool to improve predictability of in vivo AO clearance. Developing clinical pharmacology strategies for AO substrates by utilizing mass balance/clinical drug-drug interaction data will help build confidence in fmAO.
Collapse
Affiliation(s)
- Nihan Izat
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Jayaprakasam Bolleddula
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Armina Abbasi
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Lionel Cheruzel
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Robert S Jones
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Darren Moss
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Fatima Ortega-Muro
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Yannick Parmentier
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Vincent C Peterkin
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Dan-Dan Tian
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Karthik Venkatakrishnan
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Michael A Zientek
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Jill Barber
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - J Brian Houston
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Daniel Scotcher
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| |
Collapse
|
11
|
Francis L, Ogungbenro K, De Bruyn T, Houston JB, Hallifax D. Exploring the Boundaries for In Vitro-In Vivo Extrapolation: Use of Isolated Rat Hepatocytes in Co-culture and Impact of Albumin Binding Properties in the Prediction of Clearance of Various Drug Types. Drug Metab Dispos 2023; 51:1463-1473. [PMID: 37580106 DOI: 10.1124/dmd.123.001309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 07/15/2023] [Accepted: 08/08/2023] [Indexed: 08/16/2023] Open
Abstract
Prediction of hepatic clearance of drugs (via uptake or metabolism) from in vitro systems continues to be problematic, particularly when plasma protein binding is high. The following work explores simultaneous assessment of both clearance processes, focusing on a commercial hepatocyte-fibroblast co-culture system (HμREL) over a 24-hour period using six probe drugs (ranging in metabolic and transporter clearance and low-to-high plasma protein binding). A rat hepatocyte co-culture assay was established using drug depletion (measuring both medium and total concentrations) and cell uptake kinetic analysis, both in the presence and absence of plasma protein (1% bovine serum albumin). Secretion of endogenous albumin was monitored as a marker of viability, and this reached 0.004% in incubations (at a rate similar to in vivo synthesis). Binding to stromal cells was substantial and required appropriate correction factors. Drug concentration-time courses were analyzed both by conventional methods and a mechanistic cell model prior to in vivo extrapolation. Clearance assayed by drug depletion in conventional suspended rat hepatocytes provided a benchmark to evaluate co-culture value. Addition of albumin appeared to improve predictions for some compounds (where fraction unbound in the medium is less than 0.1); however, for high-binding drugs, albumin significantly limited quantification and thus predictions. Overall, these results highlight ongoing challenges concerning in vitro hepatocyte system complexity and limitations of practical expediency. Considering this, more reliable measurement of hepatically cleared compounds seems possible through judicious use of available hepatocyte systems, including co-culture systems, as described herein; this would include those compounds with low metabolic turnover but high active uptake clearance. SIGNIFICANCE STATEMENT: Co-culture systems offer a more advanced tool than standard hepatocytes, with the ability to be cultured for longer periods of time, yet their potential as an in vitro tool has not been extensively assessed. We evaluate the strengths and limitations of the HμREL system using six drugs representing various metabolic and transporter-mediated clearance pathways with various degrees of albumin binding. Studies in the presence/absence of albumin allow in vitro-in vivo extrapolation and a framework to maximize their utility.
Collapse
Affiliation(s)
- Laura Francis
- 1Centre of Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom (L.F., K.O., J.B.H., D.H.) and Genentech, Inc., South San Francisco, California (T.D.B.)
| | - Kayode Ogungbenro
- 1Centre of Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom (L.F., K.O., J.B.H., D.H.) and Genentech, Inc., South San Francisco, California (T.D.B.)
| | - Tom De Bruyn
- 1Centre of Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom (L.F., K.O., J.B.H., D.H.) and Genentech, Inc., South San Francisco, California (T.D.B.)
| | - J Brian Houston
- 1Centre of Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom (L.F., K.O., J.B.H., D.H.) and Genentech, Inc., South San Francisco, California (T.D.B.)
| | - David Hallifax
- 1Centre of Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom (L.F., K.O., J.B.H., D.H.) and Genentech, Inc., South San Francisco, California (T.D.B.)
| |
Collapse
|
12
|
Di L. Recent advances in measurement of metabolic clearance, metabolite profile and reaction phenotyping of low clearance compounds. Expert Opin Drug Discov 2023; 18:1209-1219. [PMID: 37526497 DOI: 10.1080/17460441.2023.2238606] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 07/17/2023] [Indexed: 08/02/2023]
Abstract
INTRODUCTION Low metabolic clearance is usually a highly desirable property of drug candidates in order to reduce dose and dosing frequency. However, measurement of low clearance can be challenging in drug discovery. A number of new tools have recently been developed to address the gaps in the measurement of intrinsic clearance, identification of metabolites, and reaction phenotyping of low clearance compounds. AREAS COVERED The new methodologies of low clearance measurements are discussed, including the hepatocyte relay, HepatoPac®, HμREL®, and spheroid systems. In addition, metabolite formation rate determination and in vivo allometric scaling approaches are covered as alternative methods for low clearance measurements. With these new methods, measurement of ~ 20-fold lower limit of intrinsic clearance can be achieved. The advantages and limitations of each approach are highlighted. EXPERT OPINION Although several novel methods have been developed in recent years to address the challenges of low clearance, these assays tend to be time and labor intensive and costly. Future innovations focusing on developing systems with high enzymatic activities, ultra-sensitive universal quantifiable detectors, and artificial intelligence will further enhance our ability to explore the low clearance space.
Collapse
Affiliation(s)
- Li Di
- Research Fellow, Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Groton, CT, USA
| |
Collapse
|
13
|
Isin EM. Unusual Biotransformation Reactions of Drugs and Drug Candidates. Drug Metab Dispos 2023; 51:413-426. [PMID: 36653118 DOI: 10.1124/dmd.121.000744] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/09/2022] [Accepted: 01/03/2023] [Indexed: 01/19/2023] Open
Abstract
Detailed assessment of the fate of drugs in nonclinical test species and humans is essential to ensure the safety and efficacy of medicines in patients. In this context, biotransformation of drugs and drug candidates has been an area of keen interest over many decades in the pharmaceutical industry as well as academia. Although many of the enzymes and biotransformation pathways involved in the metabolism of xenobiotics and more specifically drugs have been well characterized, each drug molecule is unique and constitutes specific challenges for the biotransformation scientist. In this mini-review written for the special issue on the occasion of the 50th Anniversary celebration of Drug Metabolism and Disposition and to celebrate contributions of F. Peter Guengerich, one of the pioneers of the drug metabolism field, recently reported "unusual" biotransformation reactions are presented. Scientific and technological advances in the "toolbox" of the biotransformation scientists are summarized. As the pharmaceutical industry continues to explore therapeutic modalities different from the traditional small molecule drugs, the new challenges confronting the biotransformation scientist as well as future opportunities are discussed. SIGNIFICANCE STATEMENT: For the biotransformation scientists, it is essential to share and be aware of unexpected biotransformation reactions so that they can increase their confidence in predicting metabolites of drugs in humans to ensure the safety and efficacy of these metabolites before the medicines reach large numbers of patients. The purpose of this review is to highlight recent observations of "unusual" metabolites so that the scientists working in the area of drug metabolism can strengthen their readiness in expecting the unexpected.
Collapse
Affiliation(s)
- Emre M Isin
- Translational Medicine, Servier, 25/27 Rue Eugène Vignat, 45000, Orléans, France
| |
Collapse
|
14
|
Carpentier N, Urbani L, Dubruel P, Van Vlierberghe S. The native liver as inspiration to create superior in vitro hepatic models. Biomater Sci 2023; 11:1091-1115. [PMID: 36594602 DOI: 10.1039/d2bm01646j] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Drug induced liver injury (DILI) is one of the major reasons of drug withdrawal during the different phases of drug development. The later in the drug development a drug is discovered to be toxic, the higher the economical as well as the ethical impact will be. In vitro models for early detection of drug liver toxicity are under constant development, however to date a superior model of the liver is still lacking. Ideally, a highly reliable model should be established to maintain the different hepatic cell functionalities to the greatest extent possible, during a period of time long enough to allow for tracking of the toxicity of compounds. In the case of DILI, toxicity can appear even after months of exposure. To reach this goal, an in vitro model should be developed that mimics the in vivo liver environment, function and response to external stimuli. The different approaches for the development of liver models currently used in the field of tissue engineering will be described in this review. Combining different technologies, leading to optimal materials, cells and 3D-constructs will ultimately lead to an ideal superior model that fully recapitulates the liver.
Collapse
Affiliation(s)
- Nathan Carpentier
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium.
| | - Luca Urbani
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London SE5 9NT, UK.,Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Peter Dubruel
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium.
| | - Sandra Van Vlierberghe
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium.
| |
Collapse
|
15
|
McDuffie D, Barr D, Agarwal A, Thomas E. Physiologically relevant microsystems to study viral infection in the human liver. Front Microbiol 2022; 13:999366. [PMID: 36246284 PMCID: PMC9555087 DOI: 10.3389/fmicb.2022.999366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Viral hepatitis is a leading cause of liver disease and mortality. Infection can occur acutely or chronically, but the mechanisms that govern the clearance of virus or lack thereof are poorly understood and merit further investigation. Though cures for viral hepatitis have been developed, they are expensive, not readily accessible in vulnerable populations and some patients may remain at an increased risk of developing hepatocellular carcinoma (HCC) even after viral clearance. To sustain infection in vitro, hepatocytes must be fully mature and remain in a differentiated state. However, primary hepatocytes rapidly dedifferentiate in conventional 2D in vitro platforms. Physiologically relevant or physiomimetic microsystems, are increasingly popular alternatives to traditional two-dimensional (2D) monocultures for in vitro studies. Physiomimetic systems reconstruct and incorporate elements of the native cellular microenvironment to improve biologic functionality in vitro. Multiple elements contribute to these models including ancillary tissue architecture, cell co-cultures, matrix proteins, chemical gradients and mechanical forces that contribute to increased viability, longevity and physiologic function for the tissue of interest. These microsystems are used in a wide variety of applications to study biological phenomena. Here, we explore the use of physiomimetic microsystems as tools for studying viral hepatitis infection in the liver and how the design of these platforms is tailored for enhanced investigation of the viral lifecycle when compared to conventional 2D cell culture models. Although liver-based physiomimetic microsystems are typically applied in the context of drug studies, the platforms developed for drug discovery purposes offer a solid foundation to support studies on viral hepatitis. Physiomimetic platforms may help prolong hepatocyte functionality in order to sustain chronic viral hepatitis infection in vitro for studying virus-host interactions for prolonged periods.
Collapse
Affiliation(s)
- Dennis McDuffie
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, United States
| | - David Barr
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Ashutosh Agarwal
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, United States
- Desai Sethi Urology Institute, University of Miami Miller School of Medicine, Miami, FL, United States
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Emmanuel Thomas
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, United States
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
- Schiff Center for Liver Diseases, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
16
|
Petersson C, Zhou X, Berghausen J, Cebrian D, Davies M, DeMent K, Eddershaw P, Riedmaier AE, Leblanc AF, Manveski N, Marathe P, Mavroudis PD, McDougall R, Parrott N, Reichel A, Rotter C, Tess D, Volak LP, Xiao G, Yang Z, Baker J. Current Approaches for Predicting Human PK for Small Molecule Development Candidates: Findings from the IQ Human PK Prediction Working Group Survey. AAPS J 2022; 24:85. [DOI: 10.1208/s12248-022-00735-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/05/2022] [Indexed: 11/30/2022] Open
|
17
|
Lai Y, Chu X, Di L, Gao W, Guo Y, Liu X, Lu C, Mao J, Shen H, Tang H, Xia CQ, Zhang L, Ding X. Recent advances in the translation of drug metabolism and pharmacokinetics science for drug discovery and development. Acta Pharm Sin B 2022; 12:2751-2777. [PMID: 35755285 PMCID: PMC9214059 DOI: 10.1016/j.apsb.2022.03.009] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [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
Drug metabolism and pharmacokinetics (DMPK) is an important branch of pharmaceutical sciences. The nature of ADME (absorption, distribution, metabolism, excretion) and PK (pharmacokinetics) inquiries during drug discovery and development has evolved in recent years from being largely descriptive to seeking a more quantitative and mechanistic understanding of the fate of drug candidates in biological systems. Tremendous progress has been made in the past decade, not only in the characterization of physiochemical properties of drugs that influence their ADME, target organ exposure, and toxicity, but also in the identification of design principles that can minimize drug-drug interaction (DDI) potentials and reduce the attritions. The importance of membrane transporters in drug disposition, efficacy, and safety, as well as the interplay with metabolic processes, has been increasingly recognized. Dramatic increases in investments on new modalities beyond traditional small and large molecule drugs, such as peptides, oligonucleotides, and antibody-drug conjugates, necessitated further innovations in bioanalytical and experimental tools for the characterization of their ADME properties. In this review, we highlight some of the most notable advances in the last decade, and provide future perspectives on potential major breakthroughs and innovations in the translation of DMPK science in various stages of drug discovery and development.
Collapse
Affiliation(s)
- Yurong Lai
- Drug Metabolism, Gilead Sciences Inc., Foster City, CA 94404, USA
| | - Xiaoyan Chu
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Li Di
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Groton, CT 06340, USA
| | - Wei Gao
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Yingying Guo
- Eli Lilly and Company, Indianapolis, IN 46221, USA
| | - Xingrong Liu
- Drug Metabolism and Pharmacokinetics, Biogen, Cambridge, MA 02142, USA
| | - Chuang Lu
- Drug Metabolism and Pharmacokinetics, Accent Therapeutics, Inc. Lexington, MA 02421, USA
| | - Jialin Mao
- Department of Drug Metabolism and Pharmacokinetics, Genentech, A Member of the Roche Group, South San Francisco, CA 94080, USA
| | - Hong Shen
- Drug Metabolism and Pharmacokinetics Department, Bristol-Myers Squibb Company, Princeton, NJ 08540, USA
| | - Huaping Tang
- Bioanalysis and Biomarkers, Glaxo Smith Kline, King of the Prussia, PA 19406, USA
| | - Cindy Q. Xia
- Department of Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Cambridge, MA 02139, USA
| | - Lei Zhang
- Office of Research and Standards, Office of Generic Drugs, CDER, FDA, Silver Spring, MD 20993, USA
| | - Xinxin Ding
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
18
|
Klammers F, Goetschi A, Ekiciler A, Walter I, Parrott N, Fowler S, Umehara K. Estimation of Fraction Metabolized by Cytochrome P450 Enzymes Using Long-Term Cocultured Human Hepatocytes. Drug Metab Dispos 2022; 50:566-575. [PMID: 35246464 DOI: 10.1124/dmd.121.000765] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 02/25/2022] [Indexed: 11/22/2022] Open
Abstract
Estimation of the fraction of a drug metabolized by individual hepatic CYP enzymes relative to hepatic metabolism (fm,CYP) or total clearance h as been challenging for low turnover compounds due to insufficient resolution of the intrinsic clearance (CLint) measurement in vitro and difficulties in quantifying the formation of low abundance metabolites. To overcome this gap, inhibition of drug depletion or selective metabolite formation for 7 marker CYP substrates was investigated using chemical inhibitors and a micro-patterned hepatocyte coculture system (HepatoPac). The use of 3 μM itraconazole was successfully validated for estimation of fm,CYP3A4 by demonstration of fm values within a 2-fold of in vivo estimates for 10 out of 13 CYP3A4 substrates in a reference set of marketed drugs. Other CYP3A4 inhibitors (ketoconazole and posaconazole) were not optimal for estimation of fm,CYP3A4 for low turnover compounds due to their high CLint. The current study also demonstrated that selective inhibition sufficient for fm calculation was achieved by inhibitors of CYP1A2 (20 μM furafylline), CYP2C8 (40 μM montelukast), CYP2C9 (40 μM sulfaphenazole), CYP2C19 [3 μM (-)N-3-benzyl-phenobarbital], and CYP2D6 (5 μM quinidine). Good estimation of fm,CYP2B6 was not possible in this study due to the poor selectivity of the tested inhibitor (20 μM ticlopidine). The approach verified in this study can result in an improved fm estimation that is aligned with the regulatory agencies' guidance and can support a victim drug-drug interaction risk assessment strategy for low clearance discovery and development drug candidates. SIGNIFICANCE STATEMENT: Successful qualification of a chemical inhibition assay for estimation of fraction metabolized requires chemical inhibitors that retain sufficient unbound concentrations over time in the incubates. The current cocultured hepatocyte assay enabled estimation of fraction metabolized, especially by CYP3A4, during the drug discovery phase where metabolite quantification methods may not be available. The method enables the assessment of pharmacokinetic variability and victim drug-drug interaction risks due to enzyme polymorphism or inhibition/induction with more confidence, especially for low clearance drug candidates.
Collapse
Affiliation(s)
- Florian Klammers
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Andreas Goetschi
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Aynur Ekiciler
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Isabelle Walter
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Neil Parrott
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Stephen Fowler
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Kenichi Umehara
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland
| |
Collapse
|
19
|
Walles M, Pähler A, Isin EM, Weidolf L. Meeting report of the second European Biotransformation Workshop. Xenobiotica 2022; 52:426-431. [PMID: 35410573 DOI: 10.1080/00498254.2022.2064253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Challenges and opportunities in the field of biotransformation were presented and discussed at the 2nd European Biotransformation workshop which was conducted virtually in collaboration with the DMDG on November 24/25, 2021. Here we summarise the presentations and discussions from this workshop.The following topics were covered:Regulatory requirements and biotransformation studies for antibody drug conjugates (ADCs) and antisense oligonucleotides (ASOs)Solutions for mass spectral data processing of peptides and oligonucleotidesFuture outsourcing needs in biotransformation for new modalitiesEstablished quantitative and qualitative workflows for metabolite identificationNew in vitro systems to study new chemical entities (NCEs) with low metabolic turnoverNew strategies on the timing of the human ADME (absorption, distribution, metabolism, excretion) study and to investigate the impact of human microbiome on drug development.
Collapse
Affiliation(s)
- M Walles
- Department, a Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - A Pähler
- Pharma Research and Early Development, F. Hoffmann-La Roche
| | - E M Isin
- DMPK, Translational Medicine, Servier, Orléans, France
| | - L Weidolf
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
20
|
Preiss LC, Lauschke VM, Georgi K, Petersson C. Multi-Well Array Culture of Primary Human Hepatocyte Spheroids for Clearance Extrapolation of Slowly Metabolized Compounds. AAPS J 2022; 24:41. [PMID: 35277751 DOI: 10.1208/s12248-022-00689-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 02/04/2022] [Indexed: 12/13/2022] Open
Abstract
Accurate prediction of human pharmacokinetics using in vitro tools is an important task during drug development. Albeit, currently used in vitro systems for clearance extrapolation such as microsomes and primary human hepatocytes in suspension culture show reproducible turnover, the utility of these systems is limited by a rapid decline of activity of drug metabolizing enzymes. In this study, a multi-well array culture of primary human hepatocyte spheroids was compared to suspension and single spheroid cultures from the same donor. Multi-well spheroids remained viable and functional over the incubation time of 3 days, showing physiological excretion of albumin and α-AGP. Their metabolic activity was similar compared to suspension and single spheroid cultures. This physiological activity, the high cell concentration, and the prolonged incubation time resulted in significant turnover of all tested low clearance compounds (n = 8). In stark contrast, only one or none of the compounds showed significant turnover when single spheroid or suspension cultures were used. Using multi-well spheroids and a regression offset approach (log(CLint) = 1.1 × + 0.85), clearance was predicted within 3-fold for 93% (13/14) of the tested compounds. Thus, multi-well spheroids represent a novel and valuable addition to the ADME in vitro tool kit for the determination of low clearance and overall clearance prediction. Graphical Abstract.
Collapse
Affiliation(s)
- Lena C Preiss
- Department of Drug Metabolism and Pharmacokinetics (DMPK), The Healthcare Business of Merck KGaA, Frankfurter Str. 250, 64293, Darmstadt, Germany.,Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.,Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tuebingen, Tuebingen, Germany
| | - Katrin Georgi
- Department of Drug Metabolism and Pharmacokinetics (DMPK), The Healthcare Business of Merck KGaA, Frankfurter Str. 250, 64293, Darmstadt, Germany
| | - Carl Petersson
- Department of Drug Metabolism and Pharmacokinetics (DMPK), The Healthcare Business of Merck KGaA, Frankfurter Str. 250, 64293, Darmstadt, Germany.
| |
Collapse
|
21
|
Taft BR, Yokokawa F, Kirrane T, Mata AC, Huang R, Blaquiere N, Waldron G, Zou B, Simon O, Vankadara S, Chan WL, Ding M, Sim S, Straimer J, Guiguemde A, Lakshminarayana SB, Jain JP, Bodenreider C, Thompson C, Lanshoeft C, Shu W, Fang E, Qumber J, Chan K, Pei L, Chen YL, Schulz H, Lim J, Abas SN, Ang X, Liu Y, Angulo-Barturen I, Jiménez-Díaz MB, Gamo FJ, Crespo-Fernandez B, Rosenthal PJ, Cooper RA, Tumwebaze P, Aguiar ACC, Campo B, Campbell S, Wagner J, Diagana TT, Sarko C. Discovery and Preclinical Pharmacology of INE963, a Potent and Fast-Acting Blood-Stage Antimalarial with a High Barrier to Resistance and Potential for Single-Dose Cures in Uncomplicated Malaria. J Med Chem 2022; 65:3798-3813. [PMID: 35229610 PMCID: PMC9278664 DOI: 10.1021/acs.jmedchem.1c01995] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
A series of 5-aryl-2-amino-imidazothiadiazole (ITD) derivatives
were identified by a phenotype-based high-throughput screening using
a blood stage Plasmodium falciparum (Pf) growth inhibition assay. A lead optimization program focused on
improving antiplasmodium potency, selectivity against human kinases,
and absorption, distribution, metabolism, excretion, and toxicity
properties and extended pharmacological profiles culminated in the
identification of INE963 (1), which demonstrates
potent cellular activity against Pf 3D7 (EC50 = 0.006 μM) and achieves “artemisinin-like”
kill kinetics in vitro with a parasite clearance
time of <24 h. A single dose of 30 mg/kg is fully curative in the Pf-humanized severe combined immunodeficient mouse model. INE963 (1) also exhibits a high barrier to resistance
in drug selection studies and a long half-life (T1/2) across species. These properties suggest the significant
potential for INE963 (1) to provide a curative
therapy for uncomplicated malaria with short dosing regimens. For
these reasons, INE963 (1) was progressed
through GLP toxicology studies and is now undergoing Ph1 clinical
trials.
Collapse
Affiliation(s)
- Benjamin R Taft
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research, 5959 Horton Street, Emeryville, California 94608, United States
| | - Fumiaki Yokokawa
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research, 5959 Horton Street, Emeryville, California 94608, United States
| | - Tom Kirrane
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research, 5959 Horton Street, Emeryville, California 94608, United States
| | - Anne-Catherine Mata
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research, 5959 Horton Street, Emeryville, California 94608, United States
| | - Richard Huang
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research, 5959 Horton Street, Emeryville, California 94608, United States
| | - Nicole Blaquiere
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research, 5959 Horton Street, Emeryville, California 94608, United States
| | - Grace Waldron
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research, 5959 Horton Street, Emeryville, California 94608, United States
| | - Bin Zou
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, no. 05-01, Chromos, Singapore 138670, Singapore
| | - Oliver Simon
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, no. 05-01, Chromos, Singapore 138670, Singapore
| | - Subramanyam Vankadara
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, no. 05-01, Chromos, Singapore 138670, Singapore
| | - Wai Ling Chan
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, no. 05-01, Chromos, Singapore 138670, Singapore
| | - Mei Ding
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, no. 05-01, Chromos, Singapore 138670, Singapore
| | - Sandra Sim
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, no. 05-01, Chromos, Singapore 138670, Singapore
| | - Judith Straimer
- Novartis Institute for Tropical Diseases, 5959 Horton Street, Emeryville, California 94608, United States
| | - Armand Guiguemde
- Novartis Institute for Tropical Diseases, 5959 Horton Street, Emeryville, California 94608, United States
| | - Suresh B Lakshminarayana
- Novartis Institute for Tropical Diseases, 5959 Horton Street, Emeryville, California 94608, United States
| | - Jay Prakash Jain
- Novartis Institute for Tropical Diseases, 5959 Horton Street, Emeryville, California 94608, United States
| | - Christophe Bodenreider
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, no. 05-01, Chromos, Singapore 138670, Singapore
| | - Christopher Thompson
- Novartis Institutes for Biomedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Christian Lanshoeft
- Novartis Institutes for Biomedical Research, Fabrikstrasse 14, Basel CH-4056, Switzerland
| | - Wei Shu
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research, 5959 Horton Street, Emeryville, California 94608, United States
| | - Eric Fang
- Chemical Biology and Therapeutics, Novartis Institutes for Biomedical Research, 5959 Horton Street, Emeryville, California 94608, United States
| | - Jafri Qumber
- Novartis Institute for Tropical Diseases, 5959 Horton Street, Emeryville, California 94608, United States
| | - Katherine Chan
- Novartis Institute for Tropical Diseases, 5959 Horton Street, Emeryville, California 94608, United States
| | - Luying Pei
- Novartis Institute for Tropical Diseases, 5959 Horton Street, Emeryville, California 94608, United States
| | - Yen-Liang Chen
- Novartis Institute for Tropical Diseases, 5959 Horton Street, Emeryville, California 94608, United States
| | - Hanna Schulz
- Novartis Institute for Tropical Diseases, 5959 Horton Street, Emeryville, California 94608, United States
| | - Jessie Lim
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, no. 05-01, Chromos, Singapore 138670, Singapore
| | - Siti Nurdiana Abas
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, no. 05-01, Chromos, Singapore 138670, Singapore
| | - Xiaoman Ang
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, no. 05-01, Chromos, Singapore 138670, Singapore
| | - Yugang Liu
- Technical Research and Development, Global Drug Development, Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, New Jersey 07936, United States
| | - Iñigo Angulo-Barturen
- The Art of Discovery, Astondo Bidea, BIC Bizkaia building, no. 612 Derio 48160 Bizkaia, Basque Country, Spain
| | - María Belén Jiménez-Díaz
- The Art of Discovery, Astondo Bidea, BIC Bizkaia building, no. 612 Derio 48160 Bizkaia, Basque Country, Spain
| | - Francisco Javier Gamo
- Tres Cantos Medicines Development Campus, GlaxoSmithKline, Severo Ochoa 2, Tres Cantos, Madrid 28760, Spain
| | - Benigno Crespo-Fernandez
- Tres Cantos Medicines Development Campus, GlaxoSmithKline, Severo Ochoa 2, Tres Cantos, Madrid 28760, Spain
| | - Philip J Rosenthal
- Department of Medicine, University of California, 533 Parnassus Avenue, San Francisco, California 94143, Unites States
| | - Roland A Cooper
- Department of Natural Sciences and Mathematics, Dominican University of California, San Rafael, California 94901, United States
| | - Patrick Tumwebaze
- Infectious Diseases Research Collaboration, Plot 2C Nakasero Hill Road, P.O. Box 7475 Kampala, Uganda
| | | | - Brice Campo
- Medicines for Malaria Venture, 20 Route de Pre-Bois, 1215 Geneva 15, Switzerland
| | - Simon Campbell
- Medicines for Malaria Venture, 20 Route de Pre-Bois, 1215 Geneva 15, Switzerland
| | - Jürgen Wagner
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, no. 05-01, Chromos, Singapore 138670, Singapore
| | - Thierry T Diagana
- Novartis Institute for Tropical Diseases, 5959 Horton Street, Emeryville, California 94608, United States
| | - Christopher Sarko
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research, 5959 Horton Street, Emeryville, California 94608, United States
| |
Collapse
|
22
|
Youhanna S, Kemas AM, Preiss L, Zhou Y, Shen JX, Cakal SD, Paqualini FS, Goparaju SK, Shafagh RZ, Lind JU, Sellgren CM, Lauschke VM. Organotypic and Microphysiological Human Tissue Models for Drug Discovery and Development-Current State-of-the-Art and Future Perspectives. Pharmacol Rev 2022; 74:141-206. [PMID: 35017176 DOI: 10.1124/pharmrev.120.000238] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 10/12/2021] [Indexed: 12/11/2022] Open
Abstract
The number of successful drug development projects has been stagnant for decades despite major breakthroughs in chemistry, molecular biology, and genetics. Unreliable target identification and poor translatability of preclinical models have been identified as major causes of failure. To improve predictions of clinical efficacy and safety, interest has shifted to three-dimensional culture methods in which human cells can retain many physiologically and functionally relevant phenotypes for extended periods of time. Here, we review the state of the art of available organotypic culture techniques and critically review emerging models of human tissues with key importance for pharmacokinetics, pharmacodynamics, and toxicity. In addition, developments in bioprinting and microfluidic multiorgan cultures to emulate systemic drug disposition are summarized. We close by highlighting important trends regarding the fabrication of organotypic culture platforms and the choice of platform material to limit drug absorption and polymer leaching while supporting the phenotypic maintenance of cultured cells and allowing for scalable device fabrication. We conclude that organotypic and microphysiological human tissue models constitute promising systems to promote drug discovery and development by facilitating drug target identification and improving the preclinical evaluation of drug toxicity and pharmacokinetics. There is, however, a critical need for further validation, benchmarking, and consolidation efforts ideally conducted in intersectoral multicenter settings to accelerate acceptance of these novel models as reliable tools for translational pharmacology and toxicology. SIGNIFICANCE STATEMENT: Organotypic and microphysiological culture of human cells has emerged as a promising tool for preclinical drug discovery and development that might be able to narrow the translation gap. This review discusses recent technological and methodological advancements and the use of these systems for hit discovery and the evaluation of toxicity, clearance, and absorption of lead compounds.
Collapse
Affiliation(s)
- Sonia Youhanna
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Aurino M Kemas
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Lena Preiss
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Yitian Zhou
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Joanne X Shen
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Selgin D Cakal
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Francesco S Paqualini
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Sravan K Goparaju
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Reza Zandi Shafagh
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Johan Ulrik Lind
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Carl M Sellgren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| |
Collapse
|
23
|
Van Ness KP, Cesar F, Yeung CK, Himmelfarb J, Kelly EJ. Microphysiological systems in absorption, distribution, metabolism, and elimination sciences. Clin Transl Sci 2022; 15:9-42. [PMID: 34378335 PMCID: PMC8742652 DOI: 10.1111/cts.13132] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 12/11/2022] Open
Abstract
The use of microphysiological systems (MPS) to support absorption, distribution, metabolism, and elimination (ADME) sciences has grown substantially in the last decade, in part driven by regulatory demands to move away from traditional animal-based safety assessment studies and industry desires to develop methodologies to efficiently screen and characterize drugs in the development pipeline. The past decade of MPS development has yielded great user-driven technological advances with the collective fine-tuning of cell culture techniques, fluid delivery systems, materials engineering, and performance enhancing modifications. The rapid advances in MPS technology have now made it feasible to evaluate critical ADME parameters within a stand-alone organ system or through interconnected organ systems. This review surveys current MPS developed for liver, kidney, and intestinal systems as stand-alone or interconnected organ systems, and evaluates each system for specific performance criteria recommended by regulatory authorities and MPS leaders that would render each system suitable for evaluating drug ADME. Whereas some systems are more suitable for ADME type research than others, not all system designs were intended to meet the recently published desired performance criteria and are reported as a summary of initial proof-of-concept studies.
Collapse
Affiliation(s)
- Kirk P. Van Ness
- Department of PharmaceuticsUniversity of WashingtonSeattleWashingtonUSA
| | - Francine Cesar
- Department of PharmaceuticsUniversity of WashingtonSeattleWashingtonUSA
| | - Catherine K. Yeung
- Department of PharmacyUniversity of WashingtonSeattleWashingtonUSA
- Kidney Research InstituteUniversity of WashingtonSeattleWashingtonUSA
| | | | - Edward J. Kelly
- Department of PharmaceuticsUniversity of WashingtonSeattleWashingtonUSA
- Kidney Research InstituteUniversity of WashingtonSeattleWashingtonUSA
| |
Collapse
|
24
|
Yoshida K, Doi Y, Iwazaki N, Yasuhara H, Ikenaga Y, Shimizu H, Nakada T, Watanabe T, Tateno C, Sanoh S, Kotake Y. Prediction of human pharmacokinetics for low-clearance compounds using pharmacokinetic data from chimeric mice with humanized livers. Clin Transl Sci 2021; 15:79-91. [PMID: 34080287 PMCID: PMC8742647 DOI: 10.1111/cts.13070] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 04/02/2021] [Accepted: 04/28/2021] [Indexed: 11/25/2022] Open
Abstract
Development of low-clearance (CL) compounds that can be slowly metabolized is a major goal in the pharmaceutical industry. However, the pursuit of low intrinsic CL (CLint ) often leads to significant challenges in evaluating the pharmacokinetics of such compounds. Although in vitro-in vivo extrapolation is widely used to predict human CL, its application has been limited for low-CLint compounds because of the low turnover of parent compounds in metabolic stability assays. To address this issue, we focused on chimeric mice with humanized livers (PXB-mice), which have been increasingly reported to accurately predict human CL in recent years. The predictive accuracy for nine low-CLint compounds with no significant turnover in a human hepatocyte assay was investigated using PXB-mouse methods such as single-species allometric scaling (PXB-SSS) approach and a novel physiologically based scaling (PXB-PBS) approach that assumes that the CLint per hepatocyte is equal between humans and PXB-mice. The percentages of compounds with predicted CL within 2- and 3-fold ranges of the observed CL for low-CLint compounds were 89% and 100%, respectively, for both PXB-SSS and PXB-PBS approaches. Moreover, the predicted CL was mostly consistent among the methods. Conversely, percentages of compounds with predicted CL within 2- and 3-fold ranges of the observed CL for low-CLint compounds were 50% and 63%, respectively for multispecies allometric scaling (MA). Overall, these PXB-mouse methods were much more accurate than conventional MA approaches, suggesting that PXB-mice are useful tool for predicting the human CL of low-CLint compounds that are slowly metabolized.
Collapse
Affiliation(s)
- Kosuke Yoshida
- DMPK Research Laboratories, Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Kanagawa, Japan.,Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yuki Doi
- DMPK Research Laboratories, Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Kanagawa, Japan
| | - Norihiko Iwazaki
- DMPK Research Laboratories, Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Kanagawa, Japan
| | - Hidenori Yasuhara
- DMPK Research Laboratories, Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Kanagawa, Japan
| | - Yuka Ikenaga
- DMPK Research Laboratories, Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Kanagawa, Japan
| | - Hidetoshi Shimizu
- DMPK Research Laboratories, Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Kanagawa, Japan
| | - Tomohisa Nakada
- DMPK Research Laboratories, Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Kanagawa, Japan
| | - Tomoko Watanabe
- DMPK Research Laboratories, Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Kanagawa, Japan
| | - Chise Tateno
- Research and Development Department, PhoenixBio Co., Ltd, Hiroshima, Japan
| | - Seigo Sanoh
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yaichiro Kotake
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
25
|
Riede J, Wollmann BM, Molden E, Ingelman-Sundberg M. Primary human hepatocyte spheroids as an in vitro tool for investigating drug compounds with low clearance. Drug Metab Dispos 2021; 49:DMD-AR-2020-000340. [PMID: 34074732 DOI: 10.1124/dmd.120.000340] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/24/2021] [Accepted: 04/15/2021] [Indexed: 11/22/2022] Open
Abstract
Characterizing the pharmacokinetic properties of drug candidates represents an essential task during drug development. In the past, liver microsomes and primary suspended hepatocytes have been extensively used for this purpose, but their relatively short stability limits the applicability of such in vitro systems for drug compounds with low metabolic turnover. In the present study, we used 3D primary human hepatocyte spheroids to predict the hepatic clearance of seven drugs with low to intermediate clearance in humans. Our results indicate that hepatocyte spheroids maintain their in vivo like phenotype during prolonged incubations allowing to monitor the depletion of parent drug for seven days. In contrast, attempts to increase the relative metabolic capacity by pooling hepatocyte spheroids resulted in an immediate fusion of the spheroids followed by hepatocellular de-differentiation processes, demonstrating limited applicability of the pooling approach for quantitative pharmacokinetic studies. The hepatic clearance values obtained from incubations with individual spheroids were in close correlation with the clinical reference data with six out of seven drug compounds being predicted within a three-fold deviation and average fold and absolute average fold errors of 0.57 and 1.74, respectively. In conclusion, the hepatocyte spheroid model enables accurate hepatic clearance predictions for slowly metabolized drug compounds and represents a valuable tool for determining the pharmacokinetic properties of new drug candidates as well as for mechanistic pharmacokinetic studies. Significance Statement Traditional in vitro systems often fail to predict the hepatic clearance of slowly metabolized drug compounds. The current study demonstrates the ability of primary human hepatocyte spheroids to provide accurate projections on the hepatic clearance of drug compounds with low and intermediate clearance.
Collapse
Affiliation(s)
- Julia Riede
- Drug Metabolism and Pharmacokinetics, Novartis Institutes for Biomedical Research, Switzerland
| | | | - Espen Molden
- Dept. of Pharmacology, University of Oslo, Norway
| | | |
Collapse
|
26
|
Yadav J, El Hassani M, Sodhi J, Lauschke VM, Hartman JH, Russell LE. Recent developments in in vitro and in vivo models for improved translation of preclinical pharmacokinetics and pharmacodynamics data. Drug Metab Rev 2021; 53:207-233. [PMID: 33989099 DOI: 10.1080/03602532.2021.1922435] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Improved pharmacokinetics/pharmacodynamics (PK/PD) prediction in the early stages of drug development is essential to inform lead optimization strategies and reduce attrition rates. Recently, there have been significant advancements in the development of new in vitro and in vivo strategies to better characterize pharmacokinetic properties and efficacy of drug leads. Herein, we review advances in experimental and mathematical models for clearance predictions, advancements in developing novel tools to capture slowly metabolized drugs, in vivo model developments to capture human etiology for supporting drug development, limitations and gaps in these efforts, and a perspective on the future in the field.
Collapse
Affiliation(s)
- Jaydeep Yadav
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck & Co., Inc., Boston, MA, USA
| | | | - Jasleen Sodhi
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jessica H Hartman
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | | |
Collapse
|
27
|
Sodhi JK, Benet LZ. Successful and Unsuccessful Prediction of Human Hepatic Clearance for Lead Optimization. J Med Chem 2021; 64:3546-3559. [PMID: 33765384 PMCID: PMC8504179 DOI: 10.1021/acs.jmedchem.0c01930] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Development of new chemical entities is costly, time-consuming, and has a low success rate. Accurate prediction of pharmacokinetic properties is critical to progress compounds with favorable drug-like characteristics in lead optimization. Of particular importance is the prediction of hepatic clearance, which determines drug exposure and contributes to projection of dose, half-life, and bioavailability. The most commonly employed methodology to predict hepatic clearance is termed in vitro to in vivo extrapolation (IVIVE) that involves measuring drug metabolism in vitro, scaling-up this in vitro intrinsic clearance to a prediction of in vivo intrinsic clearance by reconciling the enzymatic content between the incubation and an average human liver, and applying a model of hepatic disposition to account for limitations of protein binding and blood flow to predict in vivo clearance. This manuscript reviews common in vitro techniques used to predict hepatic clearance as well as current challenges and recent theoretical advancements in IVIVE.
Collapse
Affiliation(s)
- Jasleen K Sodhi
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California 94143, United States
| | - Leslie Z Benet
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California 94143, United States
| |
Collapse
|
28
|
Sung JH. Multi-organ-on-a-chip for pharmacokinetics and toxicokinetic study of drugs. Expert Opin Drug Metab Toxicol 2021; 17:969-986. [PMID: 33764248 DOI: 10.1080/17425255.2021.1908996] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Accurate prediction of pharmacokinetic (PK) and toxicokinetics (TK) of drugs is imperative for successful development of new pharmaceutics. Although conventional in vitro methods for predicting the PK and TK of drugs are well established, limitations still exist and more advanced chip-based in vitro platforms combined with mathematical models can help researchers overcome the limitations. Areas covered: We will review recent progress in the development of multi-organ-on-a-chip platforms for predicting PK and TK of drugs, as well as mathematical approaches that can be combined with these platforms for experiment design, data analysis and in vitro-in vivo extrapolation (IVIVE) for application to humans. Expert opinion: Although there remain some challenges to be addressed, the remarkable progress in the area of multi-organ-on-a-chip in recent years indicate that we will see tangible outcomes that can be utilized in the pharmaceutical industry in near future.
Collapse
Affiliation(s)
- Jong Hwan Sung
- Department of Chemical Engineering, Hongik University, Seoul, sejong, Republic of Korea
| |
Collapse
|
29
|
Serras AS, Rodrigues JS, Cipriano M, Rodrigues AV, Oliveira NG, Miranda JP. A Critical Perspective on 3D Liver Models for Drug Metabolism and Toxicology Studies. Front Cell Dev Biol 2021; 9:626805. [PMID: 33732695 PMCID: PMC7957963 DOI: 10.3389/fcell.2021.626805] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 01/21/2021] [Indexed: 12/12/2022] Open
Abstract
The poor predictability of human liver toxicity is still causing high attrition rates of drug candidates in the pharmaceutical industry at the non-clinical, clinical, and post-marketing authorization stages. This is in part caused by animal models that fail to predict various human adverse drug reactions (ADRs), resulting in undetected hepatotoxicity at the non-clinical phase of drug development. In an effort to increase the prediction of human hepatotoxicity, different approaches to enhance the physiological relevance of hepatic in vitro systems are being pursued. Three-dimensional (3D) or microfluidic technologies allow to better recapitulate hepatocyte organization and cell-matrix contacts, to include additional cell types, to incorporate fluid flow and to create gradients of oxygen and nutrients, which have led to improved differentiated cell phenotype and functionality. This comprehensive review addresses the drug-induced hepatotoxicity mechanisms and the currently available 3D liver in vitro models, their characteristics, as well as their advantages and limitations for human hepatotoxicity assessment. In addition, since toxic responses are greatly dependent on the culture model, a comparative analysis of the toxicity studies performed using two-dimensional (2D) and 3D in vitro strategies with recognized hepatotoxic compounds, such as paracetamol, diclofenac, and troglitazone is performed, further highlighting the need for harmonization of the respective characterization methods. Finally, taking a step forward, we propose a roadmap for the assessment of drugs hepatotoxicity based on fully characterized fit-for-purpose in vitro models, taking advantage of the best of each model, which will ultimately contribute to more informed decision-making in the drug development and risk assessment fields.
Collapse
Affiliation(s)
- Ana S. Serras
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Joana S. Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Madalena Cipriano
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart, Germany
| | - Armanda V. Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Nuno G. Oliveira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Joana P. Miranda
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
30
|
Abstract
Accurate estimation of in vivo clearance in human is pivotal to determine the dose and dosing regimen for drug development. In vitro-in vivo extrapolation (IVIVE) has been performed to predict drug clearance using empirical and physiological scalars. Multiple in vitro systems and mathematical modeling techniques have been employed to estimate in vivo clearance. The models for predicting clearance have significantly improved and have evolved to become more complex by integrating multiple processes such as drug metabolism and transport as well as passive diffusion. This chapter covers the use of conventional as well as recently developed methods to predict metabolic and transporter-mediated clearance along with the advantages and disadvantages of using these methods and the associated experimental considerations. The general approaches to improve IVIVE by use of appropriate scalars, incorporation of extrahepatic metabolism and transport and application of physiologically based pharmacokinetic (PBPK) models with proteomics data are also discussed. The chapter also provides an overview of the advantages of using such dynamic mechanistic models over static models for clearance predictions to improve IVIVE.
Collapse
|
31
|
Kanebratt KP, Janefeldt A, Vilén L, Vildhede A, Samuelsson K, Milton L, Björkbom A, Persson M, Leandersson C, Andersson TB, Hilgendorf C. Primary Human Hepatocyte Spheroid Model as a 3D In Vitro Platform for Metabolism Studies. J Pharm Sci 2020; 110:422-431. [PMID: 33122050 DOI: 10.1016/j.xphs.2020.10.043] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/19/2020] [Accepted: 10/19/2020] [Indexed: 12/12/2022]
Abstract
3D cultures of primary human hepatocytes (PHH) are emerging as a more in vivo-like culture system than previously available hepatic models. This work describes the characterisation of drug metabolism in 3D PHH spheroids. Spheroids were formed from three different donors of PHH and the expression and activities of important cytochrome P450 enzymes (CYP1A2, 2B6, 2C9, 2D6, and 3A4) were maintained for up to 21 days after seeding. The activity of CYP2B6 and 3A4 decreased, while the activity of CYP2C9 and 2D6 increased over time (P < 0.05). For six test compounds, that are metabolised by multiple enzymes, intrinsic clearance (CLint) values were comparable to standard in vitro hepatic models and successfully predicted in vivo CLint within 3-fold from observed values for low clearance compounds. Remarkably, the metabolic turnover of these low clearance compounds was reproducibly measured using only 1-3 spheroids, each composed of 2000 cells. Importantly, metabolites identified in the spheroid cultures reproduced the major metabolites observed in vivo, both primary and secondary metabolites were captured. In summary, the 3D PHH spheroid model shows promise to be used in drug discovery projects to study drug metabolism, including unknown mechanisms, over an extended period of time.
Collapse
Affiliation(s)
- Kajsa P Kanebratt
- DMPK, Research and Early Development Cardiovascular Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca Gothenburg, Sweden.
| | - Annika Janefeldt
- DMPK, Research and Early Development Cardiovascular Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca Gothenburg, Sweden
| | - Liisa Vilén
- DMPK, Research and Early Development Cardiovascular Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca Gothenburg, Sweden
| | - Anna Vildhede
- DMPK, Research and Early Development Cardiovascular Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca Gothenburg, Sweden
| | - Kristin Samuelsson
- DMPK, Research and Early Development Cardiovascular Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca Gothenburg, Sweden
| | - Lucas Milton
- DMPK, Research and Early Development Cardiovascular Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca Gothenburg, Sweden
| | - Anders Björkbom
- DMPK, Research and Early Development Cardiovascular Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca Gothenburg, Sweden
| | - Marie Persson
- DMPK, Research and Early Development Cardiovascular Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca Gothenburg, Sweden
| | - Carina Leandersson
- Physical & Analytical Chemistry, Research and Early Development Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca Gothenburg, Sweden
| | - Tommy B Andersson
- DMPK, Research and Early Development Cardiovascular Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca Gothenburg, Sweden
| | - Constanze Hilgendorf
- DMPK, Research and Early Development Cardiovascular Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca Gothenburg, Sweden
| |
Collapse
|
32
|
Docci L, Klammers F, Ekiciler A, Molitor B, Umehara K, Walter I, Krähenbühl S, Parrott N, Fowler S. In Vitro to In Vivo Extrapolation of Metabolic Clearance for UGT Substrates Using Short-Term Suspension and Long-Term Co-cultured Human Hepatocytes. AAPS JOURNAL 2020; 22:131. [DOI: 10.1208/s12248-020-00482-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 07/10/2020] [Indexed: 01/08/2023]
|
33
|
Umehara K, Cantrill C, Wittwer MB, Di Lenarda E, Klammers F, Ekiciler A, Parrott N, Fowler S, Ullah M. Application of the Extended Clearance Classification System (ECCS) in Drug Discovery and Development: Selection of Appropriate In Vitro Tools and Clearance Prediction. Drug Metab Dispos 2020; 48:849-860. [PMID: 32739889 DOI: 10.1124/dmd.120.000133] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/20/2020] [Indexed: 12/17/2022] Open
Abstract
In vitro to in vivo extrapolation (IVIVE) to predict human hepatic clearance, including metabolism and transport, requires extensive experimental resources. In addition, there may be technical challenges to measure low clearance values. Therefore, prospective identification of rate-determining step(s) in hepatic clearance through application of the Extended Clearance Classification System (ECCS) could be beneficial for optimal compound characterization. IVIVE for hepatic intrinsic clearance (CLint,h) prediction is conducted for a set of 36 marketed drugs with low-to-high in vivo clearance, which are substrates of metabolic enzymes and active uptake transporters in the liver. The compounds were assigned to the ECCS classes, and CLint,h, estimated with HepatoPac (a micropatterned hepatocyte coculture system), was compared with values calculated based on suspended hepatocyte incubates. An apparent permeability threshold (apical to basal) of 50 nm/s in LLC-PK1 cells proved optimal for ECCS classification. A reasonable performance of the IVIVE for compounds across multiple classes using HepatoPac was achieved (with 2-3-fold error), except for substrates of uptake transporters (class 3b), for which scaling of uptake clearance using plated hepatocytes is more appropriate. Irrespective of the ECCS assignment, metabolic clearance can be estimated well using HepatoPac. The validation and approach elaborated in the present study can result in proposed decision trees for the selection of the optimal in vitro assays guided by ECCS class assignment, to support compound optimization and candidate selection. SIGNIFICANCE STATEMENT: Characterization of the rate-determining step(s) in hepatic elimination could be on the critical path of compound optimization during drug discovery. This study demonstrated that HepatoPac and plated hepatocytes are suitable tools for the estimation of metabolic and active uptake clearance, respectively, for a larger set of marketed drugs, supporting a comprehensive strategy to select optimal in vitro tools and to achieve Extended Clearance Classification System-dependent in vitro to in vivo extrapolation for human clearance prediction.
Collapse
Affiliation(s)
- Kenichi Umehara
- Pharmaceutical Sciences, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Carina Cantrill
- Pharmaceutical Sciences, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Matthias Beat Wittwer
- Pharmaceutical Sciences, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Elisa Di Lenarda
- Pharmaceutical Sciences, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Florian Klammers
- Pharmaceutical Sciences, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Aynur Ekiciler
- Pharmaceutical Sciences, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Neil Parrott
- Pharmaceutical Sciences, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Stephen Fowler
- Pharmaceutical Sciences, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Mohammed Ullah
- Pharmaceutical Sciences, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland
| |
Collapse
|
34
|
Ballard TE, Kratochwil N, Cox LM, Moen MA, Klammers F, Ekiciler A, Goetschi A, Walter I. Simplifying the Execution of HepatoPac MetID Experiments: Metabolite Profile and Intrinsic Clearance Comparisons. Drug Metab Dispos 2020; 48:804-810. [PMID: 32623369 DOI: 10.1124/dmd.120.000013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 06/12/2020] [Indexed: 02/02/2023] Open
Abstract
The HepatoPac micropatterned coculture (MPCC) hepatocyte system has been shown to be an effective tool to investigate the qualitative human and preclinical species' metabolite profiles of new drug candidates. However, additional improvements to the overall study conditions and execution, layout, and human-donor count could be made. To that end, we have evaluated several ways to increase the amount of data one can generate per MPCC plate and how to more efficiently execute a MPCC study for the purpose of metabolite generation. Herein, we compare a set of compounds using single- and 10-donor pooled human MPCC hepatocytes. Intrinsic clearance and mean metabolic activities assessed by diverse enzyme markers were comparable between the single- and 10-donor pool. We have confirmed that the generated metabolite profiles were indistinguishable between the single- and 10-donor pool and also that rat MPCC can be performed at 400 µl media volume, which greatly simplifies study execution. Additional tips for successful study execution are also described. SIGNIFICANCE STATEMENT: When using the HepatoPac micropatterned coculture (MPCC) system, sometimes simple experimental condition variables or problematic plate designs can hamper productive study execution. We evaluated conditions to increase the amount of data one can generate per MPCC plate and, perhaps more importantly, execute that study more efficiently with less likelihood of error. We describe some of our key learnings, provide an examination of enzyme activity levels and clearance values, and provide some recommendations to simplify the execution of a HepatoPac experiment.
Collapse
Affiliation(s)
- T Eric Ballard
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Groton, Connecticut (T.E.B., L.M.C., M.A.M.) and Drug Disposition and Safety, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland (N.K., F.K., A.E., A.G., I.W.)
| | - N Kratochwil
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Groton, Connecticut (T.E.B., L.M.C., M.A.M.) and Drug Disposition and Safety, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland (N.K., F.K., A.E., A.G., I.W.)
| | - Loretta M Cox
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Groton, Connecticut (T.E.B., L.M.C., M.A.M.) and Drug Disposition and Safety, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland (N.K., F.K., A.E., A.G., I.W.)
| | - Mark A Moen
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Groton, Connecticut (T.E.B., L.M.C., M.A.M.) and Drug Disposition and Safety, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland (N.K., F.K., A.E., A.G., I.W.)
| | - F Klammers
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Groton, Connecticut (T.E.B., L.M.C., M.A.M.) and Drug Disposition and Safety, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland (N.K., F.K., A.E., A.G., I.W.)
| | - A Ekiciler
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Groton, Connecticut (T.E.B., L.M.C., M.A.M.) and Drug Disposition and Safety, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland (N.K., F.K., A.E., A.G., I.W.)
| | - A Goetschi
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Groton, Connecticut (T.E.B., L.M.C., M.A.M.) and Drug Disposition and Safety, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland (N.K., F.K., A.E., A.G., I.W.)
| | - I Walter
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Groton, Connecticut (T.E.B., L.M.C., M.A.M.) and Drug Disposition and Safety, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland (N.K., F.K., A.E., A.G., I.W.)
| |
Collapse
|
35
|
Industrial Approach to Determine the Relative Contribution of Seven Major UGT Isoforms to Hepatic Glucuronidation. J Pharm Sci 2020; 109:2309-2320. [DOI: 10.1016/j.xphs.2020.03.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 02/28/2020] [Accepted: 03/23/2020] [Indexed: 01/19/2023]
|
36
|
Louisse J, Alewijn M, Peijnenburg AA, Cnubben NH, Heringa MB, Coecke S, Punt A. Towards harmonization of test methods for in vitro hepatic clearance studies. Toxicol In Vitro 2020; 63:104722. [DOI: 10.1016/j.tiv.2019.104722] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 11/07/2019] [Accepted: 11/13/2019] [Indexed: 12/26/2022]
|
37
|
Fowler S, Chen WLK, Duignan DB, Gupta A, Hariparsad N, Kenny JR, Lai WG, Liras J, Phillips JA, Gan J. Microphysiological systems for ADME-related applications: current status and recommendations for system development and characterization. LAB ON A CHIP 2020; 20:446-467. [PMID: 31932816 DOI: 10.1039/c9lc00857h] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Over the last decade, progress has been made on the development of microphysiological systems (MPS) for absorption, distribution, metabolism, and excretion (ADME) applications. Central to this progress has been proof of concept data generated by academic and industrial institutions followed by broader characterization studies, which provide evidence for scalability and applicability to drug discovery and development. In this review, we describe some of the advances made for specific tissue MPS and outline the desired functionality for such systems, which are likely to make them applicable for practical use in the pharmaceutical industry. Single organ MPS platforms will be valuable for modelling tissue-specific functions. However, dynamic organ crosstalk, especially in the context of disease or toxicity, can only be obtained with the use of inter-linked MPS models which will enable scientists to address questions at the intersection of pharmacokinetics (PK) and efficacy, or PK and toxicity. In the future, successful application of MPS platforms that closely mimic human physiology may ultimately reduce the need for animal models to predict ADME outcomes and decrease the overall risk and cost associated with drug development.
Collapse
Affiliation(s)
- Stephen Fowler
- Pharma Research and Early Development, F.Hoffmann-La Roche Ltd, Grenzacherstrasse 124, CH4070, Basel, Switzerland
| | | | - David B Duignan
- Department of Drug Metabolism, Pharmacokinetics & Bioanalysis, AbbVie Bioresearch Center, Worcester, Massachusetts 01605, USA
| | - Anshul Gupta
- Amgen Research, 360 Binney St, Cambridge, MA 02141, USA
| | - Niresh Hariparsad
- Department of Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals, 50 Northern Ave, Boston, MA, USA
| | - Jane R Kenny
- DMPK, Genentech, 1 DNA Way, South San Francisco 94080, USA
| | | | - Jennifer Liras
- Medicine Design, Pfizer Inc, 1 Portland Ave, Cambridge, MA 02139, USA
| | | | - Jinping Gan
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb R&D, PO Box 4000, Princeton, NJ 08543-4000, USA.
| |
Collapse
|
38
|
Zhou Y, Shen JX, Lauschke VM. Comprehensive Evaluation of Organotypic and Microphysiological Liver Models for Prediction of Drug-Induced Liver Injury. Front Pharmacol 2019; 10:1093. [PMID: 31616302 PMCID: PMC6769037 DOI: 10.3389/fphar.2019.01093] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 08/26/2019] [Indexed: 12/21/2022] Open
Abstract
Drug-induced liver injury (DILI) is a major concern for the pharmaceutical industry and constitutes one of the most important reasons for the termination of promising drug development projects. Reliable prediction of DILI liability in preclinical stages is difficult, as current experimental model systems do not accurately reflect the molecular phenotype and functionality of the human liver. As a result, multiple drugs that passed preclinical safety evaluations failed due to liver toxicity in clinical trials or postmarketing stages in recent years. To improve the selection of molecules that are taken forward into the clinics, the development of more predictive in vitro systems that enable high-throughput screening of hepatotoxic liabilities and allow for investigative studies into DILI mechanisms has gained growing interest. Specifically, it became increasingly clear that the choice of cell types and culture method both constitute important parameters that affect the predictive power of test systems. In this review, we present current 3D culture paradigms for hepatotoxicity tests and critically evaluate their utility and performance for DILI prediction. In addition, we highlight possibilities of these emerging platforms for mechanistic evaluations of selected drug candidates and present current research directions towards the further improvement of preclinical liver safety tests. We conclude that organotypic and microphysiological liver systems have provided an important step towards more reliable DILI prediction. Furthermore, we expect that the increasing availability of comprehensive benchmarking studies will facilitate model dissemination that might eventually result in their regulatory acceptance.
Collapse
Affiliation(s)
| | | | - Volker M. Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
39
|
Watari R, Kakiki M, Yamasaki C, Ishida Y, Tateno C, Kuroda Y, Ishida S, Kusano K. Prediction of Human Hepatic Clearance for Cytochrome P450 Substrates via a New Culture Method Using the Collagen Vitrigel Membrane Chamber and Fresh Hepatocytes Isolated from Liver Humanized Mice. Biol Pharm Bull 2019; 42:348-353. [PMID: 30828066 DOI: 10.1248/bpb.b18-00582] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In drug discovery, hepatocytes have been widely utilized as in vitro tools for predicting the in vivo hepatic clearance (CL) of drug candidates. However, conventional hepatocyte models do not always reproduce in vivo physiological function, and CYP activities in particular decrease quite rapidly during culture. Furthermore, conventional in vitro assays have limitations in their ability to predict hepatic CL of metabolically stable drug candidates. In order to accurately predict hepatic CL of candidate drugs, a new method of culturing hepatocytes that activates their functional properties, including CYP activities, is in high demand. In the previous study, we established a novel long-term culture method for PXB-cells® using a collagen vitrigel membrane (CVM) chamber, which can maintain CYP activity and liver specific functions at high levels for several weeks. In this study, the vitrigel culture method was applied to predictions of hepatic CL for 22 CYP typical substrates with low to middle CL, and the prediction accuracy by this method was assessed by comparing CL data between predicted (in vitro intrinsic CL using the dispersion model) and observed (in vivo clinical data) values. The results of this study showed that in vitro CL values for approximately 60% (13/22) and 80% (18/22) of the compounds were predicted within a 2- and 3-fold difference with in vivo CL, respectively. These results suggest that the new culture method using the CVM chamber and PXB-cells is a promising in vitro system for predicting human hepatic CL with high accuracy for CYP substrates, including metabolically stable drug candidates.
Collapse
Affiliation(s)
- Ryuji Watari
- Drug Metabolism and Pharmacokinetics Tsukuba, Global Drug Metabolism and Pharmacokinetics, Biopharmaceutical Assessments Core Function Unit, Medicine Development Center, Eisai Co., Ltd.,Department of Genomics-Based Drug Discovery, Graduate School of Comprehensive Human Sciences, University of Tsukuba
| | - Motoharu Kakiki
- Drug Metabolism and Pharmacokinetics Tsukuba, Global Drug Metabolism and Pharmacokinetics, Biopharmaceutical Assessments Core Function Unit, Medicine Development Center, Eisai Co., Ltd
| | | | | | | | - Yukie Kuroda
- Division of Pharmacology, National Institute of Health Sciences
| | - Seiichi Ishida
- Division of Pharmacology, National Institute of Health Sciences
| | - Kazutomi Kusano
- Drug Metabolism and Pharmacokinetics Tsukuba, Global Drug Metabolism and Pharmacokinetics, Biopharmaceutical Assessments Core Function Unit, Medicine Development Center, Eisai Co., Ltd
| |
Collapse
|
40
|
Naritomi Y, Sanoh S, Ohta S. Utility of Chimeric Mice with Humanized Liver for Predicting Human Pharmacokinetics in Drug Discovery: Comparison with in Vitro– in Vivo Extrapolation and Allometric Scaling. Biol Pharm Bull 2019; 42:327-336. [DOI: 10.1248/bpb.b18-00754] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yoichi Naritomi
- Analysis & Pharmacokinetics Research Laboratories, Astellas Pharma Inc
| | - Seigo Sanoh
- Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Shigeru Ohta
- Graduate School of Biomedical and Health Sciences, Hiroshima University
| |
Collapse
|
41
|
Docci L, Parrott N, Krähenbühl S, Fowler S. Application of New Cellular and Microphysiological Systems to Drug Metabolism Optimization and Their Positioning Respective to In Silico Tools. SLAS DISCOVERY 2019; 24:523-536. [PMID: 30817893 DOI: 10.1177/2472555219831407] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
New cellular model systems for drug metabolism applications, such as advanced 2D liver co-cultures, spheroids, and microphysiological systems (MPSs), offer exciting opportunities to reproduce human biology more closely in vitro with the aim of improving predictions of pharmacokinetics, drug-drug interactions, and efficacy. These advanced cellular systems have quickly become established for human intrinsic clearance determination and have been validated for several other absorption, distribution, metabolism, and excretion (ADME) applications. Adoption will be driven through the demonstration of clear added value, for instance, by more accurate and precise clearance predictions and by more reliable extrapolation of drug interaction potential leading to faster progression to pivotal proof-of-concept studies. New experimental systems are attractive when they can (1) increase experimental capacity, removing optimization bottlenecks; (2) improve measurement quality of ADME properties that impact pharmacokinetics; and (3) enable measurements to be made that were not previously possible, reducing risk in ADME prediction and candidate selection. As new systems become established, they will find their place in the repository of tools used at different stages of the research and development process, depending on the balance of value, throughput, and cost. In this article, we give a perspective on the integration of these new methodologies into ADME optimization during drug discovery, and the likely applications and impacts on drug development.
Collapse
Affiliation(s)
- Luca Docci
- 1 Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland.,2 Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Neil Parrott
- 1 Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland
| | | | - Stephen Fowler
- 1 Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland
| |
Collapse
|
42
|
Gouliarmou V, Lostia AM, Coecke S, Bernasconi C, Bessems J, Dorne JL, Ferguson S, Testai E, Remy UG, Brian Houston J, Monshouwer M, Nong A, Pelkonen O, Morath S, Wetmore BA, Worth A, Zanelli U, Zorzoli MC, Whelan M. Establishing a systematic framework to characterise in vitro methods for human hepatic metabolic clearance. Toxicol In Vitro 2018; 53:233-244. [DOI: 10.1016/j.tiv.2018.08.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 07/17/2018] [Accepted: 08/08/2018] [Indexed: 12/26/2022]
|
43
|
Burton RD, Hieronymus T, Chamem T, Heim D, Anderson S, Zhu X, Hutzler JM. Assessment of the Biotransformation of Low-Turnover Drugs in the H µREL Human Hepatocyte Coculture Model. Drug Metab Dispos 2018; 46:1617-1625. [PMID: 30135244 DOI: 10.1124/dmd.118.082867] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 08/15/2018] [Indexed: 01/24/2023] Open
Abstract
Metabolic profiles of four drugs possessing diverse metabolic pathways (timolol, meloxicam, linezolid, and XK469) were compared following incubations in both suspended cryopreserved human hepatocytes and the HμREL hepatocyte coculture model. In general, minimal metabolism was observed following 4-hour incubations in both suspended hepatocytes and the HμREL model, whereas incubations conducted up to 7 days in the HμREL coculture model resulted in more robust metabolic turnover. In the case of timolol, in vivo human data suggest that 22% of the dose is transformed via multistep oxidative opening of the morpholine moiety. Only the first-step oxidation was detected in suspended hepatocytes, whereas the relevant downstream metabolites were produced in the HµREL model. For meloxicam, both the hydroxymethyl and subsequent carboxylic acid metabolites were abundant following incubation in the HμREL model, while only a trace amount of the hydroxymethyl metabolite was observed in suspension. Similar to timolol, linezolid generated substantially higher levels of morpholine ring-opened carboxylic acid metabolites in the HμREL model. Finally, while the major aldehyde oxidase-mediated mono-oxidative metabolite of XK469 was minimally produced in hepatocyte suspension, the HμREL model robustly produced this metabolite, consistent with a pathway reported to account for 54% of the total urinary excretion in human. In addition, low-level taurine and glycine conjugates were identified in the HµREL model. In summary, continuous metabolite production was observed for up to 7 days of incubation in the HµREL model, covering cytochrome P450, aldehyde oxidase, and numerous conjugative pathways, while predominant metabolites correlated with relevant metabolites reported in human in vivo studies.
Collapse
Affiliation(s)
- Richard D Burton
- Q Solutions, a Quintiles Quest Joint Venture, Bioanalytical and ADME Laboratories, Indianapolis, Indiana
| | - Todd Hieronymus
- Q Solutions, a Quintiles Quest Joint Venture, Bioanalytical and ADME Laboratories, Indianapolis, Indiana
| | - Taysir Chamem
- Q Solutions, a Quintiles Quest Joint Venture, Bioanalytical and ADME Laboratories, Indianapolis, Indiana
| | - David Heim
- Q Solutions, a Quintiles Quest Joint Venture, Bioanalytical and ADME Laboratories, Indianapolis, Indiana
| | - Shelby Anderson
- Q Solutions, a Quintiles Quest Joint Venture, Bioanalytical and ADME Laboratories, Indianapolis, Indiana
| | - Xiaochun Zhu
- Q Solutions, a Quintiles Quest Joint Venture, Bioanalytical and ADME Laboratories, Indianapolis, Indiana
| | - J Matthew Hutzler
- Q Solutions, a Quintiles Quest Joint Venture, Bioanalytical and ADME Laboratories, Indianapolis, Indiana
| |
Collapse
|
44
|
Kratochwil NA, Triyatni M, Mueller MB, Klammers F, Leonard B, Turley D, Schmaler J, Ekiciler A, Molitor B, Walter I, Gonsard PA, Tournillac CA, Durrwell A, Marschmann M, Jones R, Ullah M, Boess F, Ottaviani G, Jin Y, Parrott NJ, Fowler S. Simultaneous Assessment of Clearance, Metabolism, Induction, and Drug-Drug Interaction Potential Using a Long-Term In Vitro Liver Model for a Novel Hepatitis B Virus Inhibitor. J Pharmacol Exp Ther 2018; 365:237-248. [PMID: 29453199 DOI: 10.1124/jpet.117.245712] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 01/26/2018] [Indexed: 01/04/2023] Open
Abstract
Long-term in vitro liver models are now widely explored for human hepatic metabolic clearance prediction, enzyme phenotyping, cross-species metabolism, comparison of low clearance drugs, and induction studies. Here, we present studies using a long-term liver model, which show how metabolism and active transport, drug-drug interactions, and enzyme induction in healthy and diseased states, such as hepatitis B virus (HBV) infection, may be assessed in a single test system to enable effective data integration for physiologically based pharmacokinetic (PBPK) modeling. The approach is exemplified in the case of (3S)-4-[[(4R)-4-(2-Chloro-4-fluorophenyl)-5-methoxycarbonyl-2-thiazol-2-yl-1,4-dihydropyrimidin-6-yl]methyl]morpholine-3-carboxylic acid RO6889678, a novel inhibitor of HBV with a complex absorption, distribution, metabolism, and excretion (ADME) profile. RO6889678 showed an intracellular enrichment of 78-fold in hepatocytes, with an apparent intrinsic clearance of 5.2 µl/min per mg protein and uptake and biliary clearances of 2.6 and 1.6 µl/min per mg protein, respectively. When apparent intrinsic clearance was incorporated into a PBPK model, the simulated oral human profiles were in good agreement with observed data at low doses but were underestimated at high doses due to unexpected overproportional increases in exposure with dose. In addition, the induction potential of RO6889678 on cytochrome P450 (P450) enzymes and transporters at steady state was assessed and cotreatment with ritonavir revealed a complex drug-drug interaction with concurrent P450 inhibition and moderate UDP-glucuronosyltransferase induction. Furthermore, we report on the first evaluation of in vitro pharmacokinetics studies using HBV-infected HepatoPac cocultures. Thus, long-term liver models have great potential as translational research tools exploring pharmacokinetics of novel drugs in vitro in health and disease.
Collapse
Affiliation(s)
- Nicole A Kratochwil
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Miriam Triyatni
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Martina B Mueller
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Florian Klammers
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Brian Leonard
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Dan Turley
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Josephine Schmaler
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Aynur Ekiciler
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Birgit Molitor
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Isabelle Walter
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Pierre-Alexis Gonsard
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Charles A Tournillac
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Alexandre Durrwell
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Michaela Marschmann
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Russell Jones
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Mohammed Ullah
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Franziska Boess
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Giorgio Ottaviani
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Yuyan Jin
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Neil J Parrott
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Stephen Fowler
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| |
Collapse
|
45
|
Morgan P, Brown DG, Lennard S, Anderton MJ, Barrett JC, Eriksson U, Fidock M, Hamrén B, Johnson A, March RE, Matcham J, Mettetal J, Nicholls DJ, Platz S, Rees S, Snowden MA, Pangalos MN. Impact of a five-dimensional framework on R&D productivity at AstraZeneca. Nat Rev Drug Discov 2018; 17:167-181. [DOI: 10.1038/nrd.2017.244] [Citation(s) in RCA: 269] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
46
|
Watari R, Kakiki M, Oshikata A, Takezawa T, Yamasaki C, Ishida Y, Tateno C, Kuroda Y, Ishida S, Kusano K. A long-term culture system based on a collagen vitrigel membrane chamber that supports liver-specific functions of hepatocytes isolated from mice with humanized livers. J Toxicol Sci 2018; 43:521-529. [DOI: 10.2131/jts.43.521] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Ryuji Watari
- Drug Metabolism and Pharmacokinetics Tsukuba, Global Drug Metabolism and Pharmacokinetics, Biopharmaceutical Assessments Core Function Unit, Medicine Development Center, Eisai Co., Ltd
- Department of Genomics-Based Drug Discovery, Graduate School of Comprehensive Human Sciences, University of Tsukuba
| | - Motoharu Kakiki
- Drug Metabolism and Pharmacokinetics Tsukuba, Global Drug Metabolism and Pharmacokinetics, Biopharmaceutical Assessments Core Function Unit, Medicine Development Center, Eisai Co., Ltd
| | - Ayumi Oshikata
- Division of Biotechnology, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization
| | - Toshiaki Takezawa
- Division of Biotechnology, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization
| | | | | | | | - Yukie Kuroda
- Division of Pharmacology, National Institute of Health Sciences
| | - Seiichi Ishida
- Division of Pharmacology, National Institute of Health Sciences
| | - Kazutomi Kusano
- Drug Metabolism and Pharmacokinetics Tsukuba, Global Drug Metabolism and Pharmacokinetics, Biopharmaceutical Assessments Core Function Unit, Medicine Development Center, Eisai Co., Ltd
| |
Collapse
|
47
|
Cassidy KC, Yi P. Qualitative and quantitative prediction of human in vivo metabolic pathways in a human hepatocyte-murine stromal cell co-culture model. Xenobiotica 2017; 48:1192-1205. [DOI: 10.1080/00498254.2017.1395927] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
| | - Ping Yi
- Eli Lilly and Company, Lilly Corporate Centre , Indianapolis, IN , USA
| |
Collapse
|
48
|
Long-term hepatitis B infection in a scalable hepatic co-culture system. Nat Commun 2017; 8:125. [PMID: 28743900 PMCID: PMC5527081 DOI: 10.1038/s41467-017-00200-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 06/08/2017] [Indexed: 02/06/2023] Open
Abstract
Hepatitis B virus causes chronic infections in 250 million people worldwide. Chronic hepatitis B virus carriers are at risk of developing fibrosis, cirrhosis, and hepatocellular carcinoma. A prophylactic vaccine exists and currently available antivirals can suppress but rarely cure chronic infections. The study of hepatitis B virus and development of curative antivirals are hampered by a scarcity of models that mimic infection in a physiologically relevant, cellular context. Here, we show that cell-culture and patient-derived hepatitis B virus can establish persistent infection for over 30 days in a self-assembling, primary hepatocyte co-culture system. Importantly, infection can be established without antiviral immune suppression, and susceptibility is not donor dependent. The platform is scalable to microwell formats, and we provide proof-of-concept for its use in testing entry inhibitors and antiviral compounds. The lack of models that mimic hepatitis B virus (HBV) infection in a physiologically relevant context has hampered drug development. Here, Winer et al. establish a self-assembling, primary hepatocyte co-culture system that can be infected with patient-derived HBV without further modifications.
Collapse
|