1
|
Lauro G, Aliberti M, De Nisco M, Pedatella S, Pepe G, Basilicata MG, Chini MG, Fischer K, Hofstetter RK, Werz O, Ferraro MG, Piccolo M, Irace C, Saviano A, Campiglia P, Bertamino A, Ostacolo C, Ciaglia T, Manfra M, Bifulco G. Furazanopyrazine-based novel promising anticancer agents interfering with the eicosanoid biosynthesis pathways by dual mPGES-1 and sEH inhibition. Eur J Med Chem 2025; 289:117402. [PMID: 40010271 DOI: 10.1016/j.ejmech.2025.117402] [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: 12/16/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 02/28/2025]
Abstract
We report the identification of a new set of compounds based on the furazanopyrazine core interfering with eicosanoid biosynthesis and acting as potentially effective anti-inflammatory and anticancer agents. Based on our previous promising results on a set of furazanopyrazine-based compounds against the microsomal prostaglandin E2 synthase-1 (mPGES-1) enzyme, we here identified derivatives with improved pharmacokinetic properties by replacing the ester moiety with a more stable ether group. A focused virtual library of 1 × 104 molecules was built and screened against mPGES-1 through molecular docking experiments, leading to the selection of 10 candidates for synthesis and biological evaluation. Several molecules were found to inhibit mPGES-1 and, among them, two items featured IC50 values in the low micromolar range. Additional computational studies on the collection of synthesized compounds demonstrated that compound 3b, previously emerged as an mPGES-1 inhibitor, interfered with soluble epoxide hydrolase (sEH) activity, thus emerging as a valuable dual mPGES-1/sEH inhibitor. The pharmacokinetic features of the most potent compounds were accurately estimated. Unfortunately, poor outcomes were obtained for 3b; on the other hand, compound 7e exhibited promising mPGES-1 inhibition and excellent pharmacokinetic profile, demonstrating that the novel furazanopyrazine-based items with ether moiety possess improved pharmacokinetic properties compared to the ester-based compounds reported in our previous study. Additionally, the anticancer properties of 7e and 7d, the latter emerged as the most active mPGES-1 inhibitor, were evaluated and both compounds showed promising activities against HCT-116 human colorectal cancer (CRC) cells. These findings highlight the furazanopyrazine core as a promising scaffold for disclosing new anti-inflammatory drugs with the ability to inhibit targets belonging to arachidonic acid cascade.
Collapse
Affiliation(s)
- Gianluigi Lauro
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano, 84084, Italy
| | - Michela Aliberti
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano, 84084, Italy
| | - Mauro De Nisco
- Department of Health Sciences, University of Basilicata, Viale dell'Ateneo Lucano, Potenza, I-85100, Italy
| | - Silvana Pedatella
- Department of Chemical Sciences, University of Napoli Federico II, Via Cintia 4, I-80126, Napoli, Italy
| | - Giacomo Pepe
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano, 84084, Italy
| | - Manuela Giovanna Basilicata
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", P.zza L. Miraglia 2, 80138, Naples, Italy
| | - Maria Giovanna Chini
- Department of Biosciences and Territory, University of Molise, C.da Fonte Lappone, Pesche, 86090, Italy
| | - Katrin Fischer
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, Jena, 07743, Germany
| | - Robert K Hofstetter
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, Jena, 07743, Germany
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, Jena, 07743, Germany
| | - Maria Grazia Ferraro
- Department of Molecular Medicine and Medical Biotechnologies, School of Medicine and Surgery, University of Naples, Via Domenico Montesano 49, Naples, 80131, Italy
| | - Marialuisa Piccolo
- BioChem Lab, Department of Pharmacy, School of Medicine and Surgery, University of Naples, Via Domenico Montesano 49, Naples, 80131, Italy
| | - Carlo Irace
- BioChem Lab, Department of Pharmacy, School of Medicine and Surgery, University of Naples, Via Domenico Montesano 49, Naples, 80131, Italy
| | - Anella Saviano
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano, 84084, Italy
| | - Alessia Bertamino
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano, 84084, Italy
| | - Carmine Ostacolo
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano, 84084, Italy
| | - Tania Ciaglia
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano, 84084, Italy
| | - Michele Manfra
- Department of Health Sciences, University of Basilicata, Viale dell'Ateneo Lucano, Potenza, I-85100, Italy.
| | - Giuseppe Bifulco
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano, 84084, Italy.
| |
Collapse
|
2
|
Benet LZ, Sodhi JK, Tiitto MV, Xiang Y. Application of Kirchhoff's Laws to pharmacologic and pharmacokinetic analyses. Pharmacol Rev 2025; 77:100050. [PMID: 40120204 DOI: 10.1016/j.pharmr.2025.100050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 02/13/2025] [Indexed: 03/25/2025] Open
Abstract
Recently, we introduced a straightforward approach to derive clearance and rate constant equations, without relying on differential equations, utilizing Kirchhoff's Laws, a well known physics methodology used to describe rate-defining processes either in series or parallel. Manuscripts from our laboratory have re-examined published experimental data, demonstrating that the Kirchhoff's Laws methodology can explain data previously considered anomalous, such as the following: (1) all experimental perfused liver clearance data conforming to the equation once thought to represent the unphysiological well stirred model, (2) instances where linear pharmacokinetic systemic bioavailability determinations exceed unity, (3) renal clearance being influenced by drug input processes, (4) statistically significant differences in bioavailability measures between urinary excretion and systemic concentration measurements, and (5) how the long-accepted steady-state clearance approach used in pharmacokinetics for the past half-century leads to unrealistic conclusions about the relationship between liver-to-blood Kpuu and hepatic availability FH. These findings demonstrate the potential for errors in pharmacokinetic evaluations that rely on differential equations. The Kirchhoff's Laws approach is applicable to all pharmacokinetic analyses of quality experimental data, both those that align with present pharmacokinetic theory, and those that do not. Although 3 publications have attempted to rebut our position, they fail to address unexplained experimental data, and we detail here why these analyses are invalid. Our discoveries are ongoing. Additionally, we briefly discuss the application of Kirchoff's Laws to saturable nonlinear kinetics, explaining increased pharmacodynamic response for extended vs immediate release dosage forms, as well as the advantages of successfully formulating high hepatic extraction drugs. SIGNIFICANCE STATEMENT: The Kirchhoff's Laws approach to deriving clearance equations for linear systems in parallel or in series, independent of differential equations, successfully describes anomalous published pharmacokinetic data that have previously been unexplained. We review 9 experimental outcomes in humans that are newly explained using the Kirchhoff's Laws approach, including the extension to deriving nonlinear saturable clearance relationships.
Collapse
Affiliation(s)
- Leslie Z Benet
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California.
| | - Jasleen K Sodhi
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California
| | - Markus Ville Tiitto
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California
| | - Yue Xiang
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California
| |
Collapse
|
3
|
Schaller S, Michon I, Baier V, Martins FS, Nolain P, Taneja A. Evaluation of BCRP-Related DDIs Between Methotrexate and Cyclosporin A Using Physiologically Based Pharmacokinetic Modelling. Drugs R D 2025; 25:1-17. [PMID: 39715910 PMCID: PMC12011704 DOI: 10.1007/s40268-024-00495-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/22/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND AND OBJECTIVE This study provides a physiologically based pharmacokinetic (PBPK) model-based analysis of the potential drug-drug interaction (DDI) between cyclosporin A (CsA), a breast cancer resistance protein transporter (BCRP) inhibitor, and methotrexate (MTX), a putative BCRP substrate. METHODS PBPK models for CsA and MTX were built using open-source tools and published data for both model building and for model verification and validation. The MTX and CsA PBPK models were evaluated for their application in simulating BCRP-related DDIs. A qualification of an introduced empirical uniform in vitro scaling factor of Ki values for transporter inhibition by CsA was conducted by using a previously developed model of rosuvastatin (sensitive index BCRP substrate), and assessing if corresponding DDI ratios were well captured. RESULTS Within the simulated DDI scenarios for MTX in the presence of CsA, the developed models could capture the observed changes in PK parameters as changes in the area under the curve ratios (area under the curve during DDI/area under the curve control) of 1.30 versus 1.31 observed and the DDI peak plasma concentration ratios (peak plasma concentration during DDI/peak plasma concentration control) of 1.07 versus 1.28 observed. The originally reported in vitro Ki values of CsA were scaled with the uniform qualified scaling factor for their use in the in vivo DDI simulations to correct for the low intracellular unbound fraction of the CsA effector concentration. The resulting predicted versus observed ratios of peak plasma concentration and area under the curve DDI ratios with MTX were 0.82 and 0.99, respectively, indicating adequate model accuracy and choice of a scaling factor to capture the observed DDI. CONCLUSIONS All models have been comprehensively documented and made publicly available as tools to support the drug development and clinical research community and further community-driven model development.
Collapse
Affiliation(s)
| | | | | | | | | | - Amit Taneja
- Galapagos SASU, Romainville, France
- Simulations Plus, Inc., Lancaster, California, USA
| |
Collapse
|
4
|
Isoherranen N. Physiologically based pharmacokinetic modeling of small molecules: How much progress have we made? Drug Metab Dispos 2025; 53:100013. [PMID: 39884807 DOI: 10.1124/dmd.123.000960] [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: 11/01/2023] [Revised: 01/10/2024] [Accepted: 02/01/2024] [Indexed: 02/09/2024] Open
Abstract
Physiologically based pharmacokinetic (PBPK) models of small molecules have become mainstream in drug development and in academic research. The use of PBPK models is continuously expanding, with the majority of work now focusing on predictions of drug-drug interactions, drug-disease interactions, and changes in drug disposition across lifespan. Recently, publications that use PBPK modeling to predict drug disposition during pregnancy and in organ impairment have increased reflecting the advances in incorporating diverse physiologic changes into the models. Because of the expanding computational power and diversity of modeling platforms available, the complexity of PBPK models has also increased. Academic efforts have provided clear advances in better capturing human physiology in PBPK models and incorporating more complex mathematical concepts into PBPK models. Examples of such advances include the segregated gut model with a series of gut compartments allowing modeling of physiologic blood flow distribution within an organ and zonation of metabolic enzymes and series compartment liver models allowing simulations of hepatic clearance for high extraction drugs. Despite these advances in academic research, the progress in assessing model quality and defining model acceptance criteria based on the intended use of the models has not kept pace. This Minireview suggests that awareness of the need for predefined criteria for model acceptance has increased, but many manuscripts still lack description of scientific justification and/or rationale for chosen acceptance criteria. As artificial intelligence and machine learning approaches become more broadly accepted, these tools offer promise for development of comprehensive assessment for existing observed data and analysis of model performance. SIGNIFICANCE STATEMENT: Physiologically based pharmacokinetic (PBPK) modeling has become a mainstream application in academic literature and is broadly used for predictions, analysis, and evaluation of pharmacokinetic data. Significant progress has been made in developing advanced PBPK models that better capture human physiology, but oftentimes sufficient justification for the chosen model acceptance criterion and model structure is still missing. This Minireview provides a summary of the current landscape of PBPK applications used and highlights the need for advancing PBPK modeling science and training in academia.
Collapse
Affiliation(s)
- Nina Isoherranen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington.
| |
Collapse
|
5
|
Nagar S, Parise R, Korzekwa K. Predicting Clearance with Simple and Permeability-Limited Physiologically Based Pharmacokinetic Frameworks: Comparison of Well-Stirred, Dispersion, and Parallel-Tube Liver Models. Drug Metab Dispos 2024; 52:1060-1072. [PMID: 39084881 PMCID: PMC11409860 DOI: 10.1124/dmd.124.001782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/01/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024] Open
Abstract
One-compartment (1C) and permeability-limited models were used to evaluate the ability of microsomal and hepatocyte intrinsic clearances to predict hepatic clearance. Well-stirred (WSM), parallel-tube (PTM), and dispersion (DM) models were evaluated within the liver as well as within whole-body physiologically based pharmacokinetic frameworks. It was shown that a linear combination of well-stirred and parallel-tube average liver blood concentrations accurately approximates dispersion model blood concentrations. Using a flow/permeability-limited model, a large systematic error was observed for acids and no systematic error for bases. A scaling factor that reduced interstitial fluid (ISF) plasma protein binding could greatly decrease the absolute average fold error (AAFE) for acids. Using a 1C model, a scalar to reduce plasma protein binding decreased the microsomal clearance AAFE for both acids and bases. With a permeability-limited model, only acids required this scalar. The mechanism of the apparent increased cytosolic concentrations for acids remains unknown. We also show that for hepatocyte intrinsic clearance in vitro-in vivo correlations (IVIVCs), a 1C model is mechanistically appropriate since hepatocyte clearance should represent the net clearance from ISF to elimination. A relationship was derived that uses microsomal and hepatocyte intrinsic clearance to solve for an active hepatic uptake clearance, but the results were inconclusive. Finally, the PTM model generally performed better than the WSM or DM models, with no clear advantage between microsomes and hepatocytes. SIGNIFICANCE STATEMENT: Prediction of drug clearance from microsomes or hepatocytes remains challenging. Various liver models (e.g., well-stirred, parallel-tube, and dispersion) have been mathematically incorporated into liver as well as whole-body physiologically based pharmacokinetic frameworks. Although the resulting models allow incorporation of pH partitioning, permeability, and active uptake for prediction of drug clearance, including these processes did not improve clearance predictions for both microsomes and hepatocytes.
Collapse
Affiliation(s)
- Swati Nagar
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, Pennsylvania
| | - Rachel Parise
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, Pennsylvania
| | - Ken Korzekwa
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, Pennsylvania
| |
Collapse
|
6
|
Schulz Pauly JA, Kalvass JC. How predictive are isolated perfused liver data of in vivo hepatic clearance? A meta-analysis of isolated perfused rat liver data. Xenobiotica 2024; 54:658-669. [PMID: 39279675 DOI: 10.1080/00498254.2024.2404170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/18/2024]
Abstract
Isolated perfused rat liver (IPRL) experiments have been used to answer clearance-related questions, including evaluating the impact of pathological and physiological processes on hepatic clearance (CLH). However, to date, IPRL data has not been evaluated for in vivo CLH prediction accuracy.In addition to a detailed overview of available IPRL literature, we present an in-depth analysis of the performance of IPRL in CLH prediction.While the entire dataset poorly predicted CLH (GAFE = 3.2; 64% within 3-fold), IPRL conducted under optimal experimental conditions, such as in the presence of plasma proteins and with a perfusion rate within 2-fold of physiological liver blood flow and corrected for unbound fraction in the presence of red blood cells, can accurately predict rat CLH (GAFE = 2.0; 78% within 3-fold). Careful consideration of experimental conditions is needed to allow proper data analysis.Further, isolated perfused liver experiments in other species, including human livers, may allow us to address the current in vitro-in vivo disconnects of hepatic metabolic clearance and improve our methodology for CLH predictions.
Collapse
Affiliation(s)
- Julia A Schulz Pauly
- Quantitative, Translational, & ADME Sciences (QTAS), Abbvie Inc., North Chicago, IL, USA
| | - J Cory Kalvass
- Quantitative, Translational, & ADME Sciences (QTAS), Abbvie Inc., North Chicago, IL, USA
| |
Collapse
|
7
|
Benet LZ, Sodhi JK. Commentary: Pharmacokinetic Theory Must Consider Published Experimental Data. Drug Metab Dispos 2024; 52:932-938. [PMID: 38942444 PMCID: PMC11331591 DOI: 10.1124/dmd.124.001735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/12/2024] [Accepted: 06/17/2024] [Indexed: 06/30/2024] Open
Abstract
Recently, we have proposed simple methodology to derive clearance and rate constant equations, independent of differential equations, based on Kirchhoff's Laws, a common methodology from physics used to describe rate-defining processes either in series or parallel. Our approach has been challenged in three recent publications, two published in this journal, but notably what is lacking is that none evaluate experimental pharmacokinetic data. As reviewed here, manuscripts from our laboratory have evaluated published experimental data, demonstrating that the Kirchhoff's Laws approach explains (1) why all of the experimental perfused liver clearance data appear to fit the equation that was previously believed to be the well-stirred model, (2) why linear pharmacokinetic systemic bioavailability determinations can be greater than 1, (3) why renal clearance can be a function of drug input processes, and (4) why statistically different bioavailability measures may be found for urinary excretion versus systemic concentration measurements. Our most recent paper demonstrates (5) how the universally accepted steady-state clearance approach used by the field for the past 50 years leads to unrealistic outcomes concerning the relationship between liver-to-blood Kpuu and hepatic availability FH , highlighting the potential for errors in pharmacokinetic evaluations based on differential equations. The Kirchhoff's Laws approach is applicable to all pharmacokinetic analyses of quality experimental data, those that were previously adequately explained with present pharmacokinetic theory, and those that were not. The publications that have attempted to rebut our position do not address unexplained experimental data, and we show here why their analyses are not valid. SIGNIFICANCE STATEMENT: The Kirchhoff's Laws approach to deriving clearance equations for linear systems in parallel or in series, independent of differential equations, successfully describes published pharmacokinetic data that has previously been unexplained. Three recent publications claim to refute our proposed methodology; these publications only make theoretical arguments, do not evaluate experimental data, and never demonstrate that the Kirchhoff methodology provides incorrect interpretations of experimental pharmacokinetic data, including statistically significant data not explained by present pharmacokinetic theory. We demonstrate why these analyses are invalid.
Collapse
Affiliation(s)
- Leslie Z Benet
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California
| | - Jasleen K Sodhi
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California
| |
Collapse
|
8
|
Tsutsui H, Kato M, Kuramoto S, Yoshinari K. Quantitative prediction of CYP3A induction-mediated drug-drug interactions in clinical practice. Drug Metab Pharmacokinet 2024; 57:101010. [PMID: 39043066 DOI: 10.1016/j.dmpk.2024.101010] [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: 12/25/2023] [Revised: 03/04/2024] [Accepted: 03/04/2024] [Indexed: 07/25/2024]
Abstract
There have been no reports on the quantitative prediction of CYP3A induction-mediated decreases in AUC and Cmax for drug candidates identified as a "victims" of CYP3A induction. Our previous study separately evaluated the fold-induction of hepatic and intestinal CYP3A by known inducers using clinical induction data and revealed that we were able to quantitatively predict the AUC ratio (AUCR) of a few CYP3A substrates in the presence and absence of CYP3A inducers. In the present study, we investigate the predictability of AUCR and also Cmax ratio (CmaxR) in additional 54 clinical studies. The fraction metabolized by CYP3A (fm), the intestinal bioavailability (Fg), and the hepatic intrinsic clearance (CLint) of substrates were determined by the in vitro experiments as well as clinical data used for calculating AUCR and CmaxR. The result showed that 65-69% and 65-67% of predictions were within 2-fold of observed AUCR and CmaxR, respectively. A simulation using multiple parameter combinations suggested that the variability of fm and Fg within a certain range might have a minimal impact on the calculation output. These findings suggest that clinical AUCR and CmaxR of CYP3A substrates can be quantitatively predicted from the preclinical stage.
Collapse
Affiliation(s)
- Haruka Tsutsui
- Chugai Pharmaceutical Co., Ltd., 216 Totsukacho, Totsuka-ku, Yokohama-shi, Kanagawa, 244-8602, Japan; Department of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan.
| | - Motohiro Kato
- Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20, Shinmachi, Nishitokyo, Tokyo, 202-8585, Japan
| | - Shino Kuramoto
- Chugai Pharmaceutical Co., Ltd., 216 Totsukacho, Totsuka-ku, Yokohama-shi, Kanagawa, 244-8602, Japan
| | - Kouichi Yoshinari
- Department of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| |
Collapse
|
9
|
Pang KS, Lu WI, Mulder GJ. After 50 Years of Hepatic Clearance Models, Where Should We Go from Here? Improvements and Implications for Physiologically Based Pharmacokinetic Modeling. Drug Metab Dispos 2024; 52:919-931. [PMID: 39013583 DOI: 10.1124/dmd.124.001649] [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: 01/09/2024] [Accepted: 04/25/2024] [Indexed: 07/18/2024] Open
Abstract
There is overwhelming preference for application of the unphysiologic, well-stirred model (WSM) over the parallel tube model (PTM) and dispersion model (DM) to predict hepatic drug clearance, CLH , despite that liver blood flow is dispersive and closer to the DM in nature. The reasoning is the ease in computation relating the hepatic intrinsic clearance ( CLint ), hepatic blood flow ( QH ), unbound fraction in blood ( fub ) and the transmembrane clearances ( CLin and CLef ) to CLH for the WSM. However, the WSM, being the least efficient liver model, predicts a lower EH that is associated with the in vitro CLint ( Vmax / Km ), therefore requiring scale-up to predict CLH in vivo. By contrast, the miniPTM, a three-subcompartment tank-in-series model of uniform enzymes, closely mimics the DM and yielded similar patterns for CLint versus EH , substrate concentration [S] , and KL / B , the tissue to outflow blood concentration ratio. We placed these liver models nested within physiologically based pharmacokinetic models to describe the kinetics of the flow-limited, phenolic substrate, harmol, using the WSM (single compartment) and the miniPTM and zonal liver models (ZLMs) of evenly and unevenly distributed glucuronidation and sulfation activities, respectively, to predict CLH For the same, given CLint ( Vmax and Km ), the WSM again furnished the lowest extraction ratio ( EH,WSM = 0.5) compared with the miniPTM and ZLM (>0.68). Values of EH,WSM were elevated to those for EH, PTM and EH, ZLM when the Vmax s for sulfation and glucuronidation were raised 5.7- to 1.15-fold. The miniPTM is easily manageable mathematically and should be the new normal for liver/physiologic modeling. SIGNIFICANCE STATEMENT: Selection of the proper liver clearance model impacts strongly on CLH predictions. The authors recommend use of the tank-in-series miniPTM (3 compartments mini-parallel tube model), which displays similar properties as the dispersion model (DM) in relating CLint and [ S ] to CLH as a stand-in for the DM, which best describes the liver microcirculation. The miniPTM is readily modified to accommodate enzyme and transporter zonation.
Collapse
Affiliation(s)
- K Sandy Pang
- Leslie Dan Faculty of Pharmacy, University of Toronto, Ontario, Canada (K.S.P., W.I.L.) and Department of Toxicology, Leiden Academic Centre for Drug Research, Leiden University, The Netherlands (G.J.M.)
| | - Weijia Ivy Lu
- Leslie Dan Faculty of Pharmacy, University of Toronto, Ontario, Canada (K.S.P., W.I.L.) and Department of Toxicology, Leiden Academic Centre for Drug Research, Leiden University, The Netherlands (G.J.M.)
| | - Gerard J Mulder
- Leslie Dan Faculty of Pharmacy, University of Toronto, Ontario, Canada (K.S.P., W.I.L.) and Department of Toxicology, Leiden Academic Centre for Drug Research, Leiden University, The Netherlands (G.J.M.)
| |
Collapse
|
10
|
Russell LE, Yadav J, Maldonato BJ, Chien HC, Zou L, Vergara AG, Villavicencio EG. Transporter-mediated drug-drug interactions: regulatory guidelines, in vitro and in vivo methodologies and translation, special populations, and the blood-brain barrier. Drug Metab Rev 2024:1-28. [PMID: 38967415 DOI: 10.1080/03602532.2024.2364591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 05/31/2024] [Indexed: 07/06/2024]
Abstract
This review, part of a special issue on drug-drug interactions (DDIs) spearheaded by the International Society for the Study of Xenobiotics (ISSX) New Investigators, explores the critical role of drug transporters in absorption, disposition, and clearance in the context of DDIs. Over the past two decades, significant advances have been made in understanding the clinical relevance of these transporters. Current knowledge on key uptake and efflux transporters that affect drug disposition and development is summarized. Regulatory guidelines from the FDA, EMA, and PMDA that inform the evaluation of potential transporter-mediated DDIs are discussed in detail. Methodologies for preclinical and clinical testing to assess potential DDIs are reviewed, with an emphasis on the utility of physiologically based pharmacokinetic (PBPK) modeling. This includes the application of relative abundance and expression factors to predict human pharmacokinetics (PK) using preclinical data, integrating the latest regulatory guidelines. Considerations for assessing transporter-mediated DDIs in special populations, including pediatric, hepatic, and renal impairment groups, are provided. Additionally, the impact of transporters at the blood-brain barrier (BBB) on the disposition of CNS-related drugs is explored. Enhancing the understanding of drug transporters and their role in drug disposition and toxicity can improve efficacy and reduce adverse effects. Continued research is essential to bridge remaining gaps in knowledge, particularly in comparison with cytochrome P450 (CYP) enzymes.
Collapse
Affiliation(s)
- Laura E Russell
- Department of Quantitative, Translational, and ADME Sciences, AbbVie Inc, North Chicago, IL, USA
| | - Jaydeep Yadav
- Department of Pharmacokinetics, Dynamics, Metabolism, and Bioanalytics, Merck & Co., Inc, Boston, MA, USA
| | - Benjamin J Maldonato
- Department of Nonclinical Development and Clinical Pharmacology, Revolution Medicines, Inc, Redwood City, CA, USA
| | - Huan-Chieh Chien
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc, South San Francisco, CA, USA
| | - Ling Zou
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc, South San Francisco, CA, USA
| | - Ana G Vergara
- Department of Pharmacokinetics, Dynamics, Metabolism, and Bioanalytics, Merck & Co., Inc, Rahway, NJ, USA
| | - Erick G Villavicencio
- Department of Biology-Discovery, Imaging and Functional Genomics, Merck & Co., Inc, Rahway, NJ, USA
| |
Collapse
|
11
|
Iraci N, Carotenuto L, Ciaglia T, Belperio G, Di Matteo F, Mosca I, Carleo G, Giovanna Basilicata M, Ambrosino P, Turcio R, Puzo D, Pepe G, Gomez-Monterrey I, Soldovieri MV, Di Sarno V, Campiglia P, Miceli F, Bertamino A, Ostacolo C, Taglialatela M. In Silico Assisted Identification, Synthesis, and In Vitro Pharmacological Characterization of Potent and Selective Blockers of the Epilepsy-Associated KCNT1 Channel. J Med Chem 2024; 67:9124-9149. [PMID: 38782404 PMCID: PMC11181338 DOI: 10.1021/acs.jmedchem.4c00268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/28/2024] [Accepted: 04/09/2024] [Indexed: 05/25/2024]
Abstract
Gain-of-function (GoF) variants in KCNT1 channels cause severe, drug-resistant forms of epilepsy. Quinidine is a known KCNT1 blocker, but its clinical use is limited due to severe drawbacks. To identify novel KCNT1 blockers, a homology model of human KCNT1 was built and used to screen an in-house library of compounds. Among the 20 molecules selected, five (CPK4, 13, 16, 18, and 20) showed strong KCNT1-blocking ability in an in vitro fluorescence-based assay. Patch-clamp experiments confirmed a higher KCNT1-blocking potency of these compounds when compared to quinidine, and their selectivity for KCNT1 over hERG and Kv7.2 channels. Among identified molecules, CPK20 displayed the highest metabolic stability; this compound also blocked KCNT2 currents, although with a lower potency, and counteracted GoF effects prompted by 2 recurrent epilepsy-causing KCNT1 variants (G288S and A934T). The present results provide solid rational basis for future design of novel compounds to counteract KCNT1-related neurological disorders.
Collapse
Affiliation(s)
- Nunzio Iraci
- Department
of Chemical, Biological, Pharmaceutical and Environmental Sciences
(CHIBIOFARAM), University of Messina, Viale F. Stagno d’Alcontres
31, 98166 Messina, Italy
| | - Lidia Carotenuto
- Department
of Neuroscience, Reproductive Sciences and Dentistry, University Federico II of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Tania Ciaglia
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, SA, Italy
| | - Giorgio Belperio
- Department
of Science and Technology, University of
Sannio, Via F. De Sanctis, 82100 Benevento, Italy
| | - Francesca Di Matteo
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, SA, Italy
| | - Ilaria Mosca
- Department
of Medicine and Health Science Vincenzo Tiberio, University of Molise, Via C. Gazzani, 86100 Campobasso, Italy
| | - Giusy Carleo
- Department
of Neuroscience, Reproductive Sciences and Dentistry, University Federico II of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Manuela Giovanna Basilicata
- Department
of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, P.zza L. Miraglia 2, 80138 Naples, Italy
| | - Paolo Ambrosino
- Department
of Science and Technology, University of
Sannio, Via F. De Sanctis, 82100 Benevento, Italy
| | - Rita Turcio
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, SA, Italy
| | - Deborah Puzo
- Department
of Medicine and Health Science Vincenzo Tiberio, University of Molise, Via C. Gazzani, 86100 Campobasso, Italy
| | - Giacomo Pepe
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, SA, Italy
| | - Isabel Gomez-Monterrey
- Department
of Pharmacy, University Federico II of Naples, Via D. Montesano 49, 80131 Naples, Italy
| | - Maria Virginia Soldovieri
- Department
of Medicine and Health Science Vincenzo Tiberio, University of Molise, Via C. Gazzani, 86100 Campobasso, Italy
| | - Veronica Di Sarno
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, SA, Italy
| | - Pietro Campiglia
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, SA, Italy
| | - Francesco Miceli
- Department
of Neuroscience, Reproductive Sciences and Dentistry, University Federico II of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Alessia Bertamino
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, SA, Italy
| | - Carmine Ostacolo
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, SA, Italy
| | - Maurizio Taglialatela
- Department
of Neuroscience, Reproductive Sciences and Dentistry, University Federico II of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| |
Collapse
|
12
|
Benet LZ, Sodhi JK. Are all measures of liver Kp uu a function of F H, as determined following oral dosing, or have we made a critical error in defining hepatic drug clearance? Eur J Pharm Sci 2024; 196:106753. [PMID: 38522769 PMCID: PMC11813737 DOI: 10.1016/j.ejps.2024.106753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/05/2024] [Accepted: 03/21/2024] [Indexed: 03/26/2024]
Abstract
Here we present, utilizing universally accepted relationships for hepatic clearance at steady state, that for all models of hepatic elimination the ratio of unbound liver drug concentration to unbound systemic blood concentration, Kpuu, is a function of or related to the hepatic bioavailability for that drug, FH. According to the derivation for the well-stirred model, Kpuu can never exceed unity, can frequently be a function of hepatic blood flow, and is equivalent to the value of FH as determined following oral dosing. For the parallel tube model, Kpuu will not equal FH but will be a function of FH and will also never be a value greater than 1. When hepatic clearance is rate limited by basolateral transporters, Kpuu will be less than 1, and less than FH. We believe that such outcomes are highly unlikely, and that the error arises from a basic assumption concerning hepatic clearance that leads to the mechanistic models of hepatic elimination, the well-stirred, parallel tube and dispersion models. That basic assumption is that the steady-state systemic concentration multiplied by the hepatic systemic clearance is equal to the product of the average unbound liver steady-state concentration and the intrinsic hepatic clearance (Css · CL = CH,u · CLint). Calculations of Kpuu and FH based on present methods of analysis provide a strong argument as to why this universally accepted relationship is not correct. Alternatively, we have shown in recent publications that hepatic clearance may be adequately determined based on Kirchhoff's Laws where no assumption of the above equality concerning hepatic intrinsic clearance is required, and where Kpuu is independent of hepatic extraction ratio and FH.
Collapse
Affiliation(s)
- L Z Benet
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, CA, USA.
| | - J K Sodhi
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
13
|
Yan Z, Ma L, Carione P, Huang J, Hwang N, Kenny JR, Hop CECA. Introducing the Dynamic Well-Stirred Model for Predicting Hepatic Clearance and Extraction Ratio. J Pharm Sci 2024; 113:1094-1112. [PMID: 38220087 DOI: 10.1016/j.xphs.2023.12.020] [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: 09/01/2023] [Revised: 12/21/2023] [Accepted: 12/21/2023] [Indexed: 01/16/2024]
Abstract
The well-stirred model (WSM) incorporating the fraction of unbound drug (fu) to account for the effect of plasma binding on intrinsic clearance has been widely used for predicting hepatic clearance under the assumption that drug protein binding reaches equilibrium instantaneously. Our theoretical analysis reveals that the effect of protein binding on intrinsic clearance is better accounted for with the dynamic free fraction (fD), a measure of drug protein binding affinity, which leads to a putative dynamic well-stirred model (dWSM) without the instantaneous equilibrium assumption. Using recombinant CYP3A4 as the in vitro clearance system, we demonstrate that the binding effect of albumin on the intrinsic clearance of both highly bound midazolam and highly free verapamil is fully corrected by their corresponding fD values, respectively. On the other hand, fu only corrects the binding effect of albumin on the intrinsic clearance of verapamil, and yields severe over-correction of the intrinsic clearance of midazolam. The results suggest that the traditional WSM is suitable for highly free drugs like verapamil but not necessarily for highly bound drugs such as midazolam due to the violation of the instantaneous equilibrium assumption or under-estimating the true free drug concentration. In comparison, the dWSM incorporating fD holds true as long as drug elimination follows steady-state kinetics, and hence, it is more broadly applicable to drugs with different protein binding characteristics. Here we demonstrate with 36 diverse drugs, that the dWSM significantly improves the accuracy of predicting human hepatic clearance and liver extraction ratio from in vitro microsomal clearance data, highlighting the importance of drug plasma protein binding kinetics in addressing the under-prediction of hepatic clearance by the WSM.
Collapse
Affiliation(s)
- Zhengyin Yan
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, CA, 94080, USA.
| | - Li Ma
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, CA, 94080, USA
| | - Pasquale Carione
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, CA, 94080, USA
| | - Julie Huang
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, CA, 94080, USA
| | - Nicky Hwang
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, CA, 94080, USA
| | - Jane R Kenny
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, CA, 94080, USA
| | - Cornelis E C A Hop
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, CA, 94080, USA
| |
Collapse
|
14
|
Ciaglia T, Vestuto V, Di Sarno V, Musella S, Smaldone G, Di Matteo F, Napolitano V, Miranda MR, Pepe G, Basilicata MG, Novi S, Capolupo I, Bifulco G, Campiglia P, Gomez-Monterrey I, Snoeck R, Andrei G, Manfra M, Ostacolo C, Lauro G, Bertamino A. Peptidomimetics as potent dual SARS-CoV-2 cathepsin-L and main protease inhibitors: In silico design, synthesis and pharmacological characterization. Eur J Med Chem 2024; 266:116128. [PMID: 38232463 DOI: 10.1016/j.ejmech.2024.116128] [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: 11/06/2023] [Revised: 12/11/2023] [Accepted: 01/04/2024] [Indexed: 01/19/2024]
Abstract
In this paper we present the design, synthesis, and biological evaluation of a new series of peptidomimetics acting as potent anti-SARS-CoV-2 agents. Starting from our previously described Main Protease (MPro) and Papain Like Protease (PLPro) dual inhibitor, CV11, here we disclose its high inhibitory activity against cathepsin L (CTSL) (IC50 = 19.80 ± 4.44 nM), an emerging target in SARS-CoV-2 infection machinery. An in silico design, inspired by the structure of CV11, led to the development of a library of peptidomimetics showing interesting activities against CTSL and Mpro, allowing us to trace the chemical requirements for the binding to both enzymes. The screening in Vero cells infected with 5 different SARS-CoV-2 variants of concerns, highlighted sub-micromolar activities for most of the synthesized compounds (13, 15, 16, 17 and 31) in agreement with the enzymatic inhibition assays results. The compounds showed lack of activity against several different RNA viruses except for the 229E and OC43 human coronavirus strains, also characterized by a cathepsin-L dependent release into the host cells. The most promising derivatives were also evaluated for their chemical and metabolic in-vitro stability, with derivatives 15 and 17 showing a suitable profile for further preclinical characterization.
Collapse
Affiliation(s)
- Tania Ciaglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084, Fisciano, Salerno, Italy
| | - Vincenzo Vestuto
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084, Fisciano, Salerno, Italy
| | - Veronica Di Sarno
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084, Fisciano, Salerno, Italy
| | - Simona Musella
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084, Fisciano, Salerno, Italy
| | - Gerardina Smaldone
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084, Fisciano, Salerno, Italy
| | - Francesca Di Matteo
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084, Fisciano, Salerno, Italy
| | - Valeria Napolitano
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084, Fisciano, Salerno, Italy
| | - Maria Rosaria Miranda
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084, Fisciano, Salerno, Italy
| | - Giacomo Pepe
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084, Fisciano, Salerno, Italy
| | | | - Sara Novi
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084, Fisciano, Salerno, Italy
| | - Ilaria Capolupo
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084, Fisciano, Salerno, Italy
| | - Giuseppe Bifulco
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084, Fisciano, Salerno, Italy
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084, Fisciano, Salerno, Italy; European Biomedical Research Institute (EBRIS), Via S. De Renzi 50, 84125, Salerno, Italy
| | - Isabel Gomez-Monterrey
- Department of Pharmacy, University Federico II of Naples, Via D. Montesano 49, 80131, Naples, Italy
| | - Robert Snoeck
- Rega Institute for Medical Research, Department of Microbiology, Immunology, and Transplantation, KU Leuven, BE-3000, Leuven, Belgium
| | - Graciela Andrei
- Rega Institute for Medical Research, Department of Microbiology, Immunology, and Transplantation, KU Leuven, BE-3000, Leuven, Belgium
| | - Michele Manfra
- Department of Science, University of Basilicata, Via Dell'Ateneo Lucano 10, 85100, Potenza, Italy
| | - Carmine Ostacolo
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084, Fisciano, Salerno, Italy
| | - Gianluigi Lauro
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084, Fisciano, Salerno, Italy.
| | - Alessia Bertamino
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084, Fisciano, Salerno, Italy.
| |
Collapse
|
15
|
Jeong YS, Jusko WJ. A Complete Extension of Classical Hepatic Clearance Models Using Fractional Distribution Parameter f d in Physiologically Based Pharmacokinetics. J Pharm Sci 2024; 113:95-117. [PMID: 37279835 PMCID: PMC10902797 DOI: 10.1016/j.xphs.2023.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/30/2023] [Accepted: 05/30/2023] [Indexed: 06/08/2023]
Abstract
The classical organ clearance models have been proposed to relate the plasma clearance CLp to probable mechanism(s) of hepatic clearance. However, the classical models assume the intrinsic capability of drug elimination (CLu,int) that is physically segregated from the vascular blood but directly acts upon the unbound drug concentration in the blood (fubCavg), and do not handle the transit-time delay between the inlet/outlet concentrations in their closed-form clearance equations. Therefore, we propose unified model structures that can address the internal blood concentration patterns of clearance organs in a more mechanistic/physiological manner, based on the fractional distribution parameter fd operative in PBPK. The basic partial/ordinary differential equations for four classical models are revisited/modified to yield a more complete set of extended clearance models, i.e., the Rattle, Sieve, Tube, and Jar models, which are the counterparts of the dispersion, series-compartment, parallel-tube, and well-stirred models. We demonstrate the feasibility of applying the resulting extended models to isolated perfused rat liver data for 11 compounds and an example dataset for in vitro-in vivo extrapolation of the intrinsic to the systemic clearances. Based on their feasibilities to handle such real data, these models may serve as an improved basis for applying clearance models in the future.
Collapse
Affiliation(s)
- Yoo-Seong Jeong
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY, 14214, USA
| | - William J Jusko
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY, 14214, USA.
| |
Collapse
|
16
|
Chen X, Yu G, Li GF. Use of Clearance Concepts to Simulate Impact of Interleukin-6 on Drug Elimination Governed by Cytochromes P450 3A4 and Glomerular Filtration Rate. Eur J Drug Metab Pharmacokinet 2023; 48:619-621. [PMID: 37792131 DOI: 10.1007/s13318-023-00859-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2023] [Indexed: 10/05/2023]
Affiliation(s)
- Xiang Chen
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, China
| | - Guo Yu
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, China
| | - Guo-Fu Li
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, China.
- Subei People's Hospital, Yangzhou, China.
| |
Collapse
|
17
|
Rowland M, Weiss M, Pang KS. Kirchhoff's Laws and Hepatic Clearance, Well-Stirred Model - Is There Common Ground? Drug Metab Dispos 2023; 51:1451-1454. [PMID: 37562956 DOI: 10.1124/dmd.123.001300] [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: 02/15/2023] [Revised: 07/24/2023] [Accepted: 07/28/2023] [Indexed: 08/12/2023] Open
Abstract
Clearance concepts are extensively applied in drug development and drug therapy. The well-stirred model (WSM) of hepatic elimination is the most widely adopted physiologic model in pharmacokinetics owing to its simplicity. A common feature of this organ model is its use to relate hepatic clearance of a compound to the physiologic variables: organ blood flow rate, binding within blood, and hepatocellular metabolic and excretory activities. Recently, Kirchhoff's laws of electrical network have been applied to organ clearance (Pachter et al., 2022; Benet and Sodhi, 2023) with the claim that they yield the same equation for hepatic clearance as the WSM, and that the equation is independent of a mechanistic model. This commentary analyzes this claim and shows that implicit in the application of Kirchhoff's approaches are the same assumptions as those of the WSM. Concern is also expressed in the interpretation of permeability or transport parameters and related equations, as well as the inappropriateness of the corresponding equation defining hepatic clearance. There is no value, and some dangers, in applying Kirchhoff's electrical laws to organ clearance. SIGNIFICANCE STATEMENT: This commentary refutes this claim by Pachter et al. (2022), and Benet and Sodhi, (2023), who suggest that the well-stirred model (WSM) of hepatic elimination, the most widely applied physiologic model of hepatic clearance, provides the same equation as Kirchhoff's laws of electrical network that is independent of a physiologic model. A careful review shows that the claim is groundless and fraught with errors. We conclude that there is no place for the application of Kirchhoff's laws to organ clearance models.
Collapse
Affiliation(s)
- Malcolm Rowland
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (M.R.); Department of Pharmacology, Martin Luther University Halle-Wittenberg, Halle, Germany (M.W.); and Leslie Dan Faculty of Pharmacy, University of Toronto, Ontario, Canada (K.S.P.)
| | - Michael Weiss
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (M.R.); Department of Pharmacology, Martin Luther University Halle-Wittenberg, Halle, Germany (M.W.); and Leslie Dan Faculty of Pharmacy, University of Toronto, Ontario, Canada (K.S.P.)
| | - K Sandy Pang
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (M.R.); Department of Pharmacology, Martin Luther University Halle-Wittenberg, Halle, Germany (M.W.); and Leslie Dan Faculty of Pharmacy, University of Toronto, Ontario, Canada (K.S.P.)
| |
Collapse
|
18
|
Manevski N, Umehara K, Parrott N. Drug Design and Success of Prospective Mouse In Vitro-In Vivo Extrapolation (IVIVE) for Predictions of Plasma Clearance (CL p) from Hepatocyte Intrinsic Clearance (CL int). Mol Pharm 2023. [PMID: 37235687 DOI: 10.1021/acs.molpharmaceut.2c01001] [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: 05/28/2023]
Abstract
Hepatocyte intrinsic clearance (CLint) and methods of in vitro-in vivo extrapolation (IVIVE) are often used to predict plasma clearance (CLp) in drug discovery. While the prediction success of this approach is dependent on the chemotype, specific molecular properties and drug design features that govern these outcomes are poorly understood. To address this challenge, we investigated the success of prospective mouse CLp IVIVE across 2142 chemically diverse compounds. Dilution scaling, which assumes that the free fraction in hepatocyte incubations (fu,inc) is governed by binding to the 10% of serum in the incubation medium, was used as our default CLp IVIVE approach. Results show that predictions of CLp are better for smaller (molecular weight (MW) < 500 Da), less polar (total polar surface area (TPSA) < 100 Å2, hydrogen bond donor (HBD) ≤1, hydrogen bond acceptor (HBA) ≤ 6), lipophilic (log D > 3), and neutral compounds, with low HBD count playing the key role. If compounds are classified according to their chemical space, predictions were good for compounds resembling central nervous system (CNS) drugs [average absolute fold error (AAFE) of 2.05, average fold error (AFE) of 0.90], moderate for classical druglike compounds (according to Lipinski, Veber, and Ghose guidelines; AAFE of 2.55; AFE of 0.68), and poor for nonclassical "beyond the rule of 5" compounds (AAFE of 3.31; AFE of 0.41). From the perspective of measured druglike properties, predictions of CLp were better for compounds with moderate-to-high hepatocyte CLint (>10 μL/min/106 cells), high passive cellular permeability (Papp > 100 nm/s), and moderate observed CLp (5-50 mL/min/kg). Influences of plasma protein binding (fu,p) and P-glycoprotein (Pgp) apical efflux ratio (AP-ER) were less pronounced. If the extended clearance classification system (ECCS) is applied, predictions were good for class 2 (Papp > 50 nm/s; neutral or basic; AAFE of 2.35; AFE of 0.70) and acceptable for class 1A compounds (AAFE of 2.98; AFE of 0.70). Classes 1B, 3 A/B, and 4 showed poor outcomes (AAFE > 3.80; AFE < 0.60). Functional groups trending toward weaker CLp IVIVE were esters, carbamates, sulfonamides, carboxylic acids, ketones, primary and secondary amines, primary alcohols, oxetanes, and compounds liable to aldehyde oxidase metabolism, likely due to multifactorial reasons. Multivariate analysis showed that multiple properties are relevant, combining together to define the overall success of CLp IVIVE. Our results indicate that the current practice of prospective CLp IVIVE is suitable only for CNS-like compounds and well-behaved classical druglike space (e.g., high permeability or ECCS class 2) without challenging functional groups. Unfortunately, based on existing mouse data, prospective CLp IVIVE for complex and nonclassical chemotypes is poor and hardly better than random guessing. This is likely due to complexities such as extrahepatic metabolism and transporter-mediated disposition which are poorly captured by this methodology. With small-molecule drug discovery increasingly evolving toward nonclassical and complex chemotypes, existing CLp IVIVE methodology will require improvement. While empirical correction factors may bridge the gap in the near future, improved and new in vitro assays, data integration models, and machine learning (ML) methods are increasingly needed to address this challenge and reduce the number of nonclinical pharmacokinetic (PK) studies.
Collapse
Affiliation(s)
- Nenad Manevski
- Roche Pharmaceutical Research and Early Development (pRED), Roche Innovation Center Basel, 4070 Basel, Switzerland
| | - Kenichi Umehara
- Roche Pharmaceutical Research and Early Development (pRED), Roche Innovation Center Basel, 4070 Basel, Switzerland
| | - Neil Parrott
- Roche Pharmaceutical Research and Early Development (pRED), Roche Innovation Center Basel, 4070 Basel, Switzerland
| |
Collapse
|
19
|
Li X, Jusko WJ. Exploring the Pharmacokinetic Mysteries of the Liver: Application of Series Compartment Models of Hepatic Elimination. Drug Metab Dispos 2023; 51:618-628. [PMID: 36732075 PMCID: PMC10158499 DOI: 10.1124/dmd.122.001190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/23/2022] [Accepted: 01/24/2023] [Indexed: 02/04/2023] Open
Abstract
Among the basic hepatic clearance models, the dispersion model (DM) is the most physiologically sound compared with the well-stirred model and the parallel tube model. However, its application in physiologically-based pharmacokinetic (PBPK) modeling has been limited due to computational complexities. The series compartment models (SCM) of hepatic elimination that treats the liver as a cascade of well-stirred compartments connected by hepatic blood flow exhibits some mathematical similarities to the DM but is easier to operate. This work assesses the quantitative correlation between the SCM and DM and demonstrates the operation of the SCM in PBPK with the published single-dose blood and liver concentration-time data of six flow-limited compounds. The predicted liver concentrations and the estimated intrinsic clearance (CLint ) and PBPK-operative tissue-to-plasma partition coefficient (Kp ) values were shown to depend on the number of liver sub-compartments (n) and hepatic enzyme zonation in the SCM. The CLint and Kp decreased with increasing n, with more remarkable differences for drugs with higher hepatic extraction ratios. Given the same total CLint , the SCM yields a higher Kp when the liver perivenous region exhibits a lower CLint as compared with a high CLint at this region. Overall, the SCM nicely approximates the DM in characterizing hepatic elimination and offers an alternative flexible approach as well as providing some insights regarding sequential drug concentrations in the liver. SIGNIFICANCE STATEMENT: The SCM nicely approximates the DM when applied in PBPK for characterizing hepatic elimination. The number of liver sub-compartments and hepatic enzyme zonation are influencing factors for the SCM resulting in model-dependent predictions of total/internal liver concentrations and estimates of CLint and the PBPK-operative Kp . Such model-dependency may have an impact when the SCM is used for in vitro-to-in vivo extrapolation (IVIVE) and may also be relevant for PK/PD/toxicological effects when it is the driving force for such responses.
Collapse
Affiliation(s)
- Xiaonan Li
- Department of Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York
| | - William J Jusko
- Department of Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York
| |
Collapse
|
20
|
Korzekwa K, Nagar S. Process and System Clearances in Pharmacokinetic Models: Our Basic Clearance Concepts Are Correct. Drug Metab Dispos 2023; 51:532-542. [PMID: 36623886 PMCID: PMC10043942 DOI: 10.1124/dmd.122.001060] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 12/07/2022] [Accepted: 12/15/2022] [Indexed: 01/11/2023] Open
Abstract
Clearances are important parameters in pharmacokinetic (PK) models. All clearances in PK models are either process clearances that include diffusion, transport, and metabolism clearances or system clearances that include organ and systemic clearance. Clearance and volume of distribution are two independent parameters that characterize drug disposition in both individual compartments and systems of compartments. In this minireview, we show that systemic and organ clearances are net clearances that can be easily derived by partition analysis. When drugs are eliminated from the central compartment by first-order processes, systemic clearance is constant. When drugs are eliminated from a peripheral compartment, instantaneous systemic clearance will vary with time. However, average clearance and clearance at steady state will be constant and will equal dose divided by area under the curve. We show that peripheral elimination will not have a large impact on most pharmacokinetic analyses and that standard models of organ and systemic clearance are useful and appropriate. SIGNIFICANCE STATEMENT: There are two basic kinds of clearances used in pharmacokinetic models, process and system clearances. We show that organ and systemic clearances are net clearances with blood or plasma as the driving concentration. For linear pharmacokinetics, clearance is constant for elimination from the central compartment but varies with time for peripheral elimination. Despite the different kinds of clearance parameters and models, standard clearance models and concepts remain valid.
Collapse
Affiliation(s)
- Ken Korzekwa
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, Pennsylvania
| | - Swati Nagar
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, Pennsylvania
| |
Collapse
|
21
|
Schulz JA, Stresser DM, Kalvass JC. Plasma Protein-Mediated Uptake and Contradictions to the Free Drug Hypothesis: A Critical Review. Drug Metab Rev 2023:1-34. [PMID: 36971325 DOI: 10.1080/03602532.2023.2195133] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
According to the free drug hypothesis (FDH), only free, unbound drug is available to interact with biological targets. This hypothesis is the fundamental principle that continues to explain the vast majority of all pharmacokinetic and pharmacodynamic processes. Under the FDH, the free drug concentration at the target site is considered the driver of pharmacodynamic activity and pharmacokinetic processes. However, deviations from the FDH are observed in hepatic uptake and clearance predictions, where observed unbound intrinsic hepatic clearance (CLint,u) is larger than expected. Such deviations are commonly observed when plasma proteins are present and form the basis of the so-called plasma protein-mediated uptake effect (PMUE). This review will discuss the basis of plasma protein binding as it pertains to hepatic clearance based on the FDH, as well as several hypotheses that may explain the underlying mechanisms of PMUE. Notably, some, but not all, potential mechanisms remained aligned with the FDH. Finally, we will outline possible experimental strategies to elucidate PMUE mechanisms. Understanding the mechanisms of PMUE and its potential contribution to clearance underprediction is vital to improving the drug development process.
Collapse
|
22
|
Patel M, Riede J, Bednarczyk D, Poller B, Deshmukh SV. Simplifying the Extended Clearance Concept Classification System (EC3S) to Guide Clearance Prediction in Drug Discovery. Pharm Res 2023; 40:937-949. [PMID: 36859748 DOI: 10.1007/s11095-023-03482-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 02/10/2023] [Indexed: 03/03/2023]
Abstract
PURPOSE The Extended Clearance Concept Classification System was established as a development-stage tool to provide a framework for identifying fundamental mechanism(s) governing drug disposition in humans. In the present study, the applicability of the EC3S in drug discovery has been investigated. In its current format, the EC3S relies on low-throughput hepatocyte uptake data, which are not frequently generated in a discovery setting. METHODS A relationship between hepatocyte uptake clearance and MDCK permeability was first established along with intrinsic clearance from human liver microsomes. The performance of this approach was examined by categorizing 64 drugs into EC3S classes and comparing the predicted major elimination pathway(s) to that observed in humans. As an extension of the work, the ability of the simplified EC3S to predict human systemic clearance based on intrinsic clearance generated using in-vitro metabolic systems was evaluated. RESULTS The assessment enabled the use of MDCK permeability and unscaled unbound intrinsic clearance to generate cut-off criteria to categorize compounds into four EC3S classes: Class 12ab, 2cd, 34ab, and 34cd, with major elimination mechanism(s) assigned to each class. The predictivity analysis suggested that systemic clearance could generally be predicted within threefold for EC3S class 12ab and 34ab compounds. For classes 2cd and 34cd, systemic clearance was poorly predicted using in-vitro systems explored in this study. CONCLUSION Collectively, our simplified classification approach is expected to facilitate the identification of mechanism(s) involved in drug elimination, faster resolution of in-vitro to in-vivo disconnects, and better design of mechanistic pharmacokinetic studies in drug discovery.
Collapse
Affiliation(s)
- Mitesh Patel
- Pharmacokinetic Sciences, Novartis Institutes for BioMedical Research, Inc., 250 Massachusetts Avenue 2A/242, Cambridge, MA, 02139, USA
| | - Julia Riede
- Pharmacokinetic Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Dallas Bednarczyk
- Pharmacokinetic Sciences, Novartis Institutes for BioMedical Research, Inc., 250 Massachusetts Avenue 2A/242, Cambridge, MA, 02139, USA
| | - Birk Poller
- Pharmacokinetic Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Sujal V Deshmukh
- Pharmacokinetic Sciences, Novartis Institutes for BioMedical Research, Inc., 250 Massachusetts Avenue 2A/242, Cambridge, MA, 02139, USA.
| |
Collapse
|
23
|
Yau E, Olivares-Morales A, Ogungbenro K, Aarons L, Gertz M. Investigation of simplified physiologically-based pharmacokinetic models in rat and human. CPT Pharmacometrics Syst Pharmacol 2023; 12:333-345. [PMID: 36754967 PMCID: PMC10014059 DOI: 10.1002/psp4.12911] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/03/2022] [Accepted: 12/12/2022] [Indexed: 02/10/2023] Open
Abstract
Whole-body physiologically-based pharmacokinetic (PBPK) models have many applications in drug research and development. It is often necessary to inform these models with animal or clinical data, updating model parameters, and making the model more predictive for future applications. This provides an opportunity and a challenge given the large number of parameters of such models. The aim of this work was to propose new mechanistic model structures with reduced complexity allowing for parameter optimization. These models were evaluated for their ability to estimate realistic values for unbound tissue to plasma partition coefficients (Kpu) and simulate observed pharmacokinetic (PK) data. Two approaches are presented: using either established kinetic lumping methods based on tissue time constants (drug-dependent) or a novel clustering analysis to identify tissues sharing common Kpu values or Kpu scalars based on similarities of tissue composition (drug-independent). PBPK models derived from these approaches were assessed using PK data of diazepam in rats and humans. Although the clustering analysis produced minor differences in tissue grouping depending on the method used, two larger groups of tissues emerged. One including the kidneys, liver, spleen, gut, heart, and lungs, and another including bone, brain, muscle, and pancreas whereas adipose and skin were generally considered distinct. Overall, a subdivision into four tissue groups appeared most physiologically relevant in terms of tissue composition. Several models were found to have similar abilities to describe diazepam i.v. data as empirical models. Comparability of estimated Kpus to experimental Kpu values for diazepam was one criterion for selecting the appropriate PK model structure.
Collapse
Affiliation(s)
- Estelle Yau
- Centre for Applied Pharmacokinetic Research (CAPKR), The University of Manchester, Manchester, UK.,Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, Basel, Switzerland.,Sanofi R&D, DMPK France, Paris, France
| | - Andrés Olivares-Morales
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, Basel, Switzerland
| | - Kayode Ogungbenro
- Centre for Applied Pharmacokinetic Research (CAPKR), The University of Manchester, Manchester, UK
| | - Leon Aarons
- Centre for Applied Pharmacokinetic Research (CAPKR), The University of Manchester, Manchester, UK
| | - Michael Gertz
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, Basel, Switzerland
| |
Collapse
|
24
|
Le Louedec F, Puisset F, Chatelut E, Tod M. Considering the Oral Bioavailability of Protein Kinase Inhibitors: Essential in Assessing the Extent of Drug-Drug Interaction and Improving Clinical Practice. Clin Pharmacokinet 2023; 62:55-66. [PMID: 36631685 DOI: 10.1007/s40262-022-01200-8] [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] [Accepted: 12/11/2022] [Indexed: 01/13/2023]
Abstract
Protein kinase inhibitors share pharmacokinetic (PK) pathways among themselves. They are all metabolized by several cytochromes P450 (CYP). For most of them, CYP3A4 is the predominant metabolic pathway. However, their oral bioavailability differs. For example, the oral bioavailability of imatinib has been estimated at nearly 100%, but that of ibrutinib averages 3% due to its high hepatic first-pass effect. Overall, the smaller the oral bioavailability, the larger its interindividual PK variability. Indeed, for drugs with low oral bioavailability, the extent of their absorption is an additional cause (along with elimination variability) of differences in drug exposure among patients. The impact of drug-drug interaction (DDI) also differs between drugs with low or high oral bioavailability. We describe and explain why the impact of CYP3A4 inhibitors and inducers is much greater for protein kinase inhibitors with low oral bioavailability. The effect of food on protein kinase inhibitors and DDIs corresponding to plasma protein binding will also be considered. Finally, the benefits of these concepts in clinical practice (including therapeutic drug monitoring) will be discussed. Overall, our main objective was to apply fundamental PK concepts to understanding the main clinical issues of these oral anticancer drugs.
Collapse
Affiliation(s)
- Félicien Le Louedec
- Institut Claudius-Regaud, Institut Universitaire du Cancer Toulouse, Oncopole, 31059, Toulouse, France
- CRCT, Cancer Research Center of Toulouse, Inserm U1037, Université Paul Sabatier, Toulouse, France
| | - Florent Puisset
- Institut Claudius-Regaud, Institut Universitaire du Cancer Toulouse, Oncopole, 31059, Toulouse, France
- CRCT, Cancer Research Center of Toulouse, Inserm U1037, Université Paul Sabatier, Toulouse, France
| | - Etienne Chatelut
- Institut Claudius-Regaud, Institut Universitaire du Cancer Toulouse, Oncopole, 31059, Toulouse, France.
- CRCT, Cancer Research Center of Toulouse, Inserm U1037, Université Paul Sabatier, Toulouse, France.
| | - Michel Tod
- Hospices Civils de Lyon, GH Nord, Service de Pharmacie, 69004, Lyon, France
- Université Claude Bernard Lyon 1, UMR CNRS 5558, LBBE-Laboratoire de Biométrie et Biologie Évolutive, 69622, Villeurbanne, France
| |
Collapse
|
25
|
Ladumor MK, Storelli F, Liang X, Lai Y, Enogieru OJ, Chothe PP, Evers R, Unadkat J. Predicting changes in the pharmacokinetics of CYP3A-metabolized drugs in hepatic impairment and insights into factors driving these changes. CPT Pharmacometrics Syst Pharmacol 2022; 12:261-273. [PMID: 36540952 PMCID: PMC9931433 DOI: 10.1002/psp4.12901] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/07/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022] Open
Abstract
Physiologically based pharmacokinetic models, populated with drug-metabolizing enzyme and transporter (DMET) abundance, can be used to predict the impact of hepatic impairment (HI) on the pharmacokinetics (PK) of drugs. To increase confidence in the predictive power of such models, they must be validated by comparing the predicted and observed PK of drugs in HI obtained by phenotyping (or probe drug) studies. Therefore, we first predicted the effect of all stages of HI (mild to severe) on the PK of drugs primarily metabolized by cytochrome P450 (CYP) 3A enzymes using the default HI module of Simcyp Version 21, populated with hepatic and intestinal CYP3A abundance data. Then, we validated the predictions using CYP3A probe drug phenotyping studies conducted in HI. Seven CYP3A substrates, metabolized primarily via CYP3A (fraction metabolized, 0.7-0.95), with low to high hepatic availability, were studied. For all stages of HI, the predicted PK parameters of drugs were within twofold of the observed data. This successful validation increases confidence in using the DMET abundance data in HI to predict the changes in the PK of drugs cleared by DMET for which phenotyping studies in HI are not available or cannot be conducted. In addition, using CYP3A drugs as an example, through simulations, we identified the salient PK factors that drive the major changes in exposure (area under the plasma concentration-time profile curve) to drugs in HI. This theoretical framework can be applied to any drug and DMET to quickly determine the likely magnitude of change in drug PK due to HI.
Collapse
Affiliation(s)
- Mayur K. Ladumor
- Department of PharmaceuticsUniversity of Washington School of PharmacySeattleWashingtonUSA
| | - Flavia Storelli
- Department of PharmaceuticsUniversity of Washington School of PharmacySeattleWashingtonUSA
| | - Xiaomin Liang
- Drug MetabolismGilead Sciences Inc.Foster CityCaliforniaUSA
| | - Yurong Lai
- Drug MetabolismGilead Sciences Inc.Foster CityCaliforniaUSA
| | | | - Paresh P. Chothe
- Global Drug Metabolism and PharmacokineticsTakeda Development Center USA, Inc.LexingtonMassachusettsUSA
| | - Raymond Evers
- Preclinical Sciences and Translational SafetyJanssen Research & Development, LLCSpring HousePennsylvaniaUSA
| | - Jashvant D. Unadkat
- Department of PharmaceuticsUniversity of Washington School of PharmacySeattleWashingtonUSA
| |
Collapse
|
26
|
Fragki S, Piersma AH, Westerhout J, Kienhuis A, Kramer NI, Zeilmaker MJ. Applicability of generic PBK modelling in chemical hazard assessment: A case study with IndusChemFate. Regul Toxicol Pharmacol 2022; 136:105267. [DOI: 10.1016/j.yrtph.2022.105267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 08/20/2022] [Accepted: 09/26/2022] [Indexed: 11/09/2022]
|
27
|
Pachter JA, Dill KA, Sodhi JK, Benet LZ. Review of the application of Kirchhoff's Laws of series and parallel flows to pharmacology: Defining organ clearance. Pharmacol Ther 2022; 239:108278. [PMID: 36075300 PMCID: PMC10832328 DOI: 10.1016/j.pharmthera.2022.108278] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/29/2022] [Accepted: 08/31/2022] [Indexed: 11/26/2022]
Abstract
Dosing rate decisions for drugs and changes in dosing in a patient due to disease states, drug interactions and pharmacogenomics are all based on clearance, a measure of the body's ability to eliminate drug. The primary organs of elimination are the liver and the kidney. Clearance for each of these organs is a summative composition of biologic processes. In 1857, Gustav Kirchhoff first developed his laws to describe the "motion of electricity in conductors... [and] ...in wires", recognizing that summative processes occur either in parallel or in series. Since then, Kirchhoff's Laws have also been applied to heat transfer, diffusion and drag force on falling objects, but not to pharmacology. Although not previously recognized, renal clearance always follow Kirchhoff's Laws, as does hepatic clearance for drugs where basolateral transporters are not clinically relevant. However, when basolateral transporters are clinically relevant, we demonstrate that the present accepted approach is inconsistent with recognized drug disposition processes. However, this clearance relationship can be easily corrected using Kirchhoff's Laws. The purpose of this review is to demonstrate that Kirchhoff's Laws, which define how to approach rate processes that occur in parallel versus processes that occur in series, can be applicable to pharmacology in addition to the over 160-year recognition of their use in physical sciences. We anticipate that the application to clearance will be only the first of many such pharmacological analyses.
Collapse
Affiliation(s)
- Jonathan Asher Pachter
- State University of New York Stony Brook, Laufer Center for Physical and Quantitative Biology and the Department of Physics & Astronomy, Stony Brook, NY, USA
| | - Ken A Dill
- State University of New York Stony Brook, Laufer Center for Physical and Quantitative Biology and the Department of Physics & Astronomy, Stony Brook, NY, USA
| | - Jasleen K Sodhi
- University of California San Francisco, Schools of Pharmacy and Medicine, Department of Bioengineering and Therapeutic Sciences, San Francisco, CA, USA
| | - Leslie Z Benet
- University of California San Francisco, Schools of Pharmacy and Medicine, Department of Bioengineering and Therapeutic Sciences, San Francisco, CA, USA.
| |
Collapse
|
28
|
Li X, Jusko WJ. Assessing Liver-to-Plasma Partition Coefficients and In Silico Calculation Methods: When Does the Hepatic Model Matter in PBPK?. Drug Metab Dispos 2022; 50:DMD-AR-2022-000994. [PMID: 36195337 DOI: 10.1124/dmd.122.000994] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/24/2022] [Accepted: 09/09/2022] [Indexed: 11/22/2022] Open
Abstract
The primary models used in pharmacokinetics (PK) to assess hepatic clearance (CLh ) are the well-stirred (WSM), parallel tube (PTM), and dispersion model (DM) that differ in their internal flow patterns and assumed unbound liver concentrations. Physiologically-Based Pharmacokinetic (PBPK) models require a hepatic intrinsic clearance (CLint ) and tissue-to-plasma partition coefficient (Kp ). Given measured systemic and liver concentration-time profiles, these hepatic models perform similarly but yield model-specific CLint and Kp estimates. This work provides mathematical relationships for the three basic hepatic models and assesses their corresponding PBPK-relevant Kp values with literature-reported single-dose blood and liver concentration-time data of 14 compounds. Model fittings were performed with an open-loop approach where the CLh and extraction ratio (ER) were first estimated from fitting the blood data yielding CLint values for the three hepatic models. The pre-fitted blood data served as forcing input functions to obtain PBPK-operative Kp estimates that were compared with those obtained by the tissue/plasma area ratio (AR), Chen & Gross (C&G) and published in silico methods. The CLint and Kp values for the hepatic models increased with the ER and both showed a rank order being WSM > DM > PTM. Drugs with low ER showed no differences as expected. With model-specific CLint and Kp values, all hepatic models predict the same steady-state Kp (Kp ss ) that is comparable to those from the AR and C&G methods and reported by direct measurement. All in silico methods performed poorly for most compounds. Hepatic model selection requires cautious application and interpretation in PBPK modeling. Significance Statement The three hepatic models generate different single-dose (non-steady-state) values of CLint and Kp in PBPK models especially for drugs with high ER; however, all Kp ss values expected from constant rate infusion studies were the same. These findings are relevant when using these models for IVIVE where a model-dependent CLint is used to correct measured tissue concentrations for depletion by metabolism. This model-dependency may also have an impact when assessing the PK/pharmacodynamic relationships when effects relate to assumed hepatic concentrations.
Collapse
Affiliation(s)
- Xiaonan Li
- Pharmaceutical Sciences, University at Buffalo, United States
| | - William J Jusko
- Pharmaceutical Sciences, University at Buffalo, United States
| |
Collapse
|
29
|
The next frontier in ADME science: Predicting transporter-based drug disposition, tissue concentrations and drug-drug interactions in humans. Pharmacol Ther 2022; 238:108271. [DOI: 10.1016/j.pharmthera.2022.108271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 08/05/2022] [Accepted: 08/17/2022] [Indexed: 12/25/2022]
|
30
|
Ahmed AN, Rostami-Hodjegan A, Barber J, Al-Majdoub ZM. Examining Physiologically Based Pharmacokinetic Model Assumptions for Cross-Tissue Similarity of Activity per Unit of Enzyme: The Case Example of Uridine 5'-Diphosphate Glucuronosyltransferase. Drug Metab Dispos 2022; 50:1119-1125. [PMID: 35636771 DOI: 10.1124/dmd.121.000813] [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: 12/12/2021] [Accepted: 05/03/2022] [Indexed: 11/22/2022] Open
Abstract
The default assumption during in vitro in vivo extrapolation (IVIVE) to predict metabolic clearance in physiologically based pharmacokinetics (PBPK) is that protein expression and activity have the same relationship in various tissues. This assumption is examined for uridine 5'-diphosphate glucuronosyltransferases (UGTs), a case example where expression and hence metabolic activity are distributed across various tissues. Our literature analysis presents overwhelming evidence of a greater UGT activity per unit of enzyme (higher kcat) in kidney and intestinal tissues relative to liver (greater than 200-fold for UGT2B7). This analysis is based on application of abundance values reported using similar proteomic techniques and within the same laboratory. Our findings call into question the practice of assuming similar kcat during IVIVE estimations as part of PBPK and call for a systematic assessment of the kcat of various enzymes across different organs. The analysis focused on compiling data for probe substrates that were common for two or more of the studied tissues to allow for reliable comparison of cross-tissue enzyme kinetics; this meant that UGT enzymes included in the study were limited to UGT1A1, 1A3, 1A6, 1A9, and 2B7. Significantly, UGT1A9 (n = 24) and the liver (n = 27) were each found to account for around half of the total dataset; these were found to correlate with hepatic UGT1A9 data found in 15 of the studies, highlighting the need for more research into extrahepatic tissues and other UGT isoforms. SIGNIFICANCE STATEMENT: During physiologically based pharmacokinetic modeling (in vitro in vivo extrapolation) of drug clearance, the default assumption is that the activity per unit of enzyme is the same in all tissues. The analysis provides preliminary evidence that this may not be the case and that renal and intestinal tissues may have almost 250-fold greater uridine 5'-diphosphate glucuronosyltransferase activity per unit of enzyme than liver tissues.
Collapse
Affiliation(s)
- Anika N Ahmed
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, UK (A.N.A., A.R.-H., J.B., Z.M.A.-M.) and Certara, Simcyp Division, Sheffield, UK (A.R.-H.)
| | - Amin Rostami-Hodjegan
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, UK (A.N.A., A.R.-H., J.B., Z.M.A.-M.) and Certara, Simcyp Division, Sheffield, UK (A.R.-H.)
| | - Jill Barber
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, UK (A.N.A., A.R.-H., J.B., Z.M.A.-M.) and Certara, Simcyp Division, Sheffield, UK (A.R.-H.)
| | - Zubida M Al-Majdoub
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, UK (A.N.A., A.R.-H., J.B., Z.M.A.-M.) and Certara, Simcyp Division, Sheffield, UK (A.R.-H.)
| |
Collapse
|
31
|
Sandoval P, Chuang BC, Cohen L, Yoneyama T, Pusalkar S, Yucha RW, Chowdhury SK, Chothe PP. Sinusoidal Uptake Determines the Hepatic Clearance of Pevonedistat (TAK-924) as Explained by Extended Clearance Model. Drug Metab Dispos 2022; 50:980-988. [PMID: 35545257 DOI: 10.1124/dmd.122.000836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/18/2021] [Indexed: 11/22/2022] Open
Abstract
Quantitative assessment of hepatic clearance (CLH) of drugs is critical to accurately predict human dose and drug-drug interaction (DDI) liabilities. This is challenging for drugs that involve complex transporter-enzyme interplay. In this study, we demonstrate this interplay in the CLH and DDI effect in the presence of CYP3A4 perpetrator for pevonedistat using both the conventional clearance model (CCM) and the extended clearance model (ECM). In vitro metabolism and hepatocyte uptake data showed that pevonedistat is actively transported into the liver via multiple uptake transporters and metabolized predominantly by CYP3A4 (88%). The active uptake clearance (CLact,inf) and passive diffusion clearance (CLdiff,inf) were 21 and 8.7 ml/min/kg, respectively. The CLact,inf was underpredicted as Empirical Scaling Factor of 13 was needed to recover the in vivo plasma clearance (CLplasma). Both CCM and ECM predicted CLplasma of pevonedistat reasonably well (predicted CLplasma of 30.8 (CCM) and 32.1 (ECM) versus observed CLplasma of 32.2 ml/min/kg). However, both systemic and liver exposures in the presence of itraconazole were well predicted by ECM but not by CCM (predicted pevonedistat plasma area under the concentration-time curve ratio (AUCR) 2.73 (CCM) and 1.23 (ECM))., The ECM prediction is in accordance with the observed clinical DDI data (observed plasma AUCR of 1.14) that showed CYP3A4 inhibition did not alter pevonedistat exposure systemically, although ECM predicted liver AUCR of 2.85. Collectively, these data indicated that the hepatic uptake is the rate-determining step in the CLH of pevonedistat and are consistent with the lack of systemic clinical DDI with itraconazole. SIGNIFICANCE STATEMENT: In this study, we successfully demonstrated that the hepatic uptake is the rate-determining step in the CLH of pevonedistat. Both the conventional and extended clearance models predict CLplasma of pevonedistat well however, only the ECM accurately predicted DDI effect in the presence of itraconazole, thus providing further evidence for the lack of DDI with CYP3A4 perpetrators for drugs that involve complex transporter-enzyme interplay as there are currently not many examples in the literature except prototypical OATP substrate drugs.
Collapse
Affiliation(s)
- Philip Sandoval
- Global Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts
| | - Bei-Ching Chuang
- Global Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts
| | - Lawrence Cohen
- Global Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts
| | - Tomoki Yoneyama
- Global Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts
| | - Sandeepraj Pusalkar
- Global Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts
| | - Robert W Yucha
- Global Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts
| | - Swapan K Chowdhury
- Global Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts
| | - Paresh P Chothe
- Global Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts
| |
Collapse
|
32
|
Zhang M, Fisher C, Gardner I, Pan X, Kilford P, Bois FY, Jamei M. Understanding Interindividual Variability in the Drug Interaction of a Highly Extracted CYP1A2 Substrate Tizanidine: Application of a Permeability-Limited Multicompartment Liver Model in a Population Based Physiologically Based Pharmacokinetic Framework. Drug Metab Dispos 2022; 50:957-967. [PMID: 35504655 DOI: 10.1124/dmd.121.000818] [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: 12/15/2021] [Accepted: 04/05/2022] [Indexed: 11/22/2022] Open
Abstract
Tizanidine, a centrally acting skeletal muscle relaxant, is predominantly metabolized by CYP1A2 and undergoes extensive hepatic first-pass metabolism after oral administration. As a highly extracted drug, the systemic exposure to tizanidine exhibits considerable interindividual variability and is altered substantially when coadministered with CYP1A2 inhibitors or inducers. The aim of the current study was to compare the performance of a permeability-limited multicompartment liver (PerMCL) model, which operates as an approximation of the dispersion model, and the well stirred model (WSM) for predicting tizanidine drug-drug interactions (DDIs). Physiologically based pharmacokinetic models were developed for tizanidine, incorporating the PerMCL model and the WSM, respectively, to simulate the interaction of tizanidine with a range of CYP1A2 inhibitors and inducers. Whereas the WSM showed a tendency to underpredict the fold change of tizanidine area under the plasma concentration-time curve (AUC ratio) in the presence of perpetrators, the use of PerMCL model increased precision (absolute average-fold error: 1.32-1.42 versus 1.58) and decreased bias (average-fold error: 0.97-1.25 versus 0.63) for the predictions of mean AUC ratios as compared with the WSM. The PerMCL model captured the observed range of individual AUC ratios of tizanidine as well as the correlation between individual AUC ratios and CYP1A2 activities without interactions, whereas the WSM was not able to capture these. The results demonstrate the advantage of using the PerMCL model over the WSM in predicting the magnitude and interindividual variability of DDIs for a highly extracted sensitive substrate tizanidine. SIGNIFICANCE STATEMENT: This study demonstrates the advantages of the PerMCL model, which operates as an approximation of the dispersion model, in mitigating the tendency of the WSM to underpredict the magnitude and variability of DDIs of a highly extracted CYP1A2 substrate tizanidine when it is administered with CYP1A2 inhibitors or inducers. The physiologically based pharmacokinetic modeling approach described herein is valuable to the understanding of drug interactions of highly extracted substrates and the source of its interindividual variability.
Collapse
Affiliation(s)
- Mian Zhang
- Certara UK Ltd, Simcyp Division, Level 2-Acero, Sheffield, United Kingdom
| | - Ciarán Fisher
- Certara UK Ltd, Simcyp Division, Level 2-Acero, Sheffield, United Kingdom
| | - Iain Gardner
- Certara UK Ltd, Simcyp Division, Level 2-Acero, Sheffield, United Kingdom
| | - Xian Pan
- Certara UK Ltd, Simcyp Division, Level 2-Acero, Sheffield, United Kingdom
| | - Peter Kilford
- Certara UK Ltd, Simcyp Division, Level 2-Acero, Sheffield, United Kingdom
| | - Frederic Y Bois
- Certara UK Ltd, Simcyp Division, Level 2-Acero, Sheffield, United Kingdom
| | - Masoud Jamei
- Certara UK Ltd, Simcyp Division, Level 2-Acero, Sheffield, United Kingdom
| |
Collapse
|
33
|
Rowland M, Pang KS. Hepatic clearance models and IVIVE predictions. Clin Pharmacol Ther 2022; 111:1205-1207. [PMID: 35234281 DOI: 10.1002/cpt.2525] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/07/2022] [Indexed: 11/12/2022]
Affiliation(s)
- Malcolm Rowland
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK
| | - K Sandy Pang
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
34
|
Jones RS, Leung C, Chang JH, Brown S, Liu N, Yan Z, Kenny JR, Broccatelli F. Application of empirical scalars to enable early prediction of human hepatic clearance using IVIVE in drug discovery: an evaluation of 173 drugs. Drug Metab Dispos 2022; 50:DMD-AR-2021-000784. [PMID: 35636770 DOI: 10.1124/dmd.121.000784] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 04/20/2022] [Accepted: 05/12/2022] [Indexed: 11/22/2022] Open
Abstract
The utilization of in vitro data to predict drug pharmacokinetics (PK) in vivo has been a consistent practice in early drug discovery for decades. However, its success is hampered by mispredictions attributed to uncharacterized biological phenomena/experimental artifacts. Predicted drug clearance (CL) from experimental data (i.e. hepatocyte intrinsic clearance: CLint, fraction unbound in plasma: fu,p) is often systematically underpredicted using the well-stirred model (WSM). The objective of this study was to evaluate using empirical scalars in the WSM to correct for CL mispredictions. Drugs (N=28) were used to generate numerical scalars on CLint (α), and fu,p (β) to minimize the error (AAFE) for CL predictions. These scalars were validated using an additional dataset (N=28 drugs) and applied to a non-redundant AstraZeneca (AZ) dataset available in the literature (N=117 drugs) for a total of 173 compounds. CL predictions using the WSM were improved for most compounds using an α value of 3.66 (~64%<2-fold) compared to no scaling (~46%<2-fold). Similarly, using a β value of 0.55 or combination of α and β scalars (values of 1.74 and 0.66, respectively) resulted in a similar improvement in predictions (~64%<2-fold and ~65%<2-fold, respectively). For highly bound compounds (fu,p{less than or equal to}0.01), AAFE was substantially reduced across all scaling methods. Using the β scalar alone or a combination of α and β appeared optimal; and produce larger magnitude corrections for highly-bound compounds. Some drugs are still disproportionally mispredicted, however the improvements in prediction error and simplicity of applying these scalars suggests its utility for early-stage CL predictions. Significance Statement In early drug discovery, prediction of human clearance using in vitro experimental data plays an essential role in triaging compounds prior to in vivo studies. These predictions have been systematically underestimated. Here we introduce empirical scalars calibrated on the extent of plasma protein binding that appear to improve clearance prediction across multiple datasets. This approach can be used in early phases of drug discovery prior to the availability of pre-clinical data for early quantitative predictions of human clearance.
Collapse
Affiliation(s)
| | | | - Jae H Chang
- Preclinical Development Sciences, ORIC Pharmaceuticals, United States
| | | | | | | | - Jane R Kenny
- Drug Metabolism & Pharmacokinetics, Genentech Inc, United States
| | | |
Collapse
|
35
|
Obrezanova O, Martinsson A, Whitehead T, Mahmoud S, Bender A, Miljković F, Grabowski P, Irwin B, Oprisiu I, Conduit G, Segall M, Smith GF, Williamson B, Winiwarter S, Greene N. Prediction of In Vivo Pharmacokinetic Parameters and Time-Exposure Curves in Rats Using Machine Learning from the Chemical Structure. Mol Pharm 2022; 19:1488-1504. [PMID: 35412314 DOI: 10.1021/acs.molpharmaceut.2c00027] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Animal pharmacokinetic (PK) data as well as human and animal in vitro systems are utilized in drug discovery to define the rate and route of drug elimination. Accurate prediction and mechanistic understanding of drug clearance and disposition in animals provide a degree of confidence for extrapolation to humans. In addition, prediction of in vivo properties can be used to improve design during drug discovery, help select compounds with better properties, and reduce the number of in vivo experiments. In this study, we generated machine learning models able to predict rat in vivo PK parameters and concentration-time PK profiles based on the molecular chemical structure and either measured or predicted in vitro parameters. The models were trained on internal in vivo rat PK data for over 3000 diverse compounds from multiple projects and therapeutic areas, and the predicted endpoints include clearance and oral bioavailability. We compared the performance of various traditional machine learning algorithms and deep learning approaches, including graph convolutional neural networks. The best models for PK parameters achieved R2 = 0.63 [root mean squared error (RMSE) = 0.26] for clearance and R2 = 0.55 (RMSE = 0.46) for bioavailability. The models provide a fast and cost-efficient way to guide the design of molecules with optimal PK profiles, to enable the prediction of virtual compounds at the point of design, and to drive prioritization of compounds for in vivo assays.
Collapse
Affiliation(s)
- Olga Obrezanova
- Imaging and Data Analytics, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge CB4 0FZ, U.K
| | - Anton Martinsson
- Imaging and Data Analytics, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Gothenburg SE-43183, Sweden
| | - Tom Whitehead
- Intellegens Ltd., Eagle Labs, Cambridge CB4 3AZ, U.K
| | - Samar Mahmoud
- Optibrium Ltd., Cambridge Innovation Park, Cambridge CB25 9PB, U.K
| | - Andreas Bender
- Imaging and Data Analytics, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge CB4 0FZ, U.K.,Department of Chemistry, Centre for Molecular Informatics, University of Cambridge, Cambridge CB2 1EW, U.K
| | - Filip Miljković
- Imaging and Data Analytics, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Gothenburg SE-43183, Sweden
| | - Piotr Grabowski
- Imaging and Data Analytics, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge CB4 0FZ, U.K
| | - Ben Irwin
- Optibrium Ltd., Cambridge Innovation Park, Cambridge CB25 9PB, U.K
| | - Ioana Oprisiu
- Imaging and Data Analytics, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Gothenburg SE-43183, Sweden
| | | | - Matthew Segall
- Optibrium Ltd., Cambridge Innovation Park, Cambridge CB25 9PB, U.K
| | - Graham F Smith
- Imaging and Data Analytics, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge CB4 0FZ, U.K
| | - Beth Williamson
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge CB10 1XL, U.K
| | - Susanne Winiwarter
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), Biopharmaceutical R&D, AstraZeneca, Gothenburg SE-43183, Sweden
| | - Nigel Greene
- Imaging and Data Analytics, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Waltham, Massachusetts 02451, United States
| |
Collapse
|
36
|
Chai XN, Ludwig FA, Müglitz A, Gong Y, Schaefer M, Regenthal R, Krügel U. A Pharmacokinetic and Metabolism Study of the TRPC6 Inhibitor SH045 in Mice by LC-MS/MS. Int J Mol Sci 2022; 23:ijms23073635. [PMID: 35408998 PMCID: PMC8998618 DOI: 10.3390/ijms23073635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/17/2022] [Accepted: 03/24/2022] [Indexed: 12/10/2022] Open
Abstract
TRPC6, the sixth member of the family of canonical transient receptor potential (TRP) channels, contributes to a variety of physiological processes and human pathologies. This study extends the knowledge on the newly developed TRPC6 blocker SH045 with respect to its main target organs beyond the description of plasma kinetics. According to the plasma concentration-time course in mice, SH045 is measurable up to 24 h after administration of 20 mg/kg BW (i.v.) and up to 6 h orally. The short plasma half-life and rather low oral bioavailability are contrasted by its reported high potency. Dosage limits were not worked out, but absence of safety concerns for 20 mg/kg BW supports further dose exploration. The disposition of SH045 is described. In particular, a high extravascular distribution, most prominent in lung, and a considerable renal elimination of SH045 were observed. SH045 is a substrate of CYP3A4 and CYP2A6. Hydroxylated and glucuronidated metabolites were identified under optimized LC-MS/MS conditions. The results guide a reasonable selection of dose and application route of SH045 for target-directed preclinical studies in vivo with one of the rare high potent and subtype-selective TRPC6 inhibitors available.
Collapse
Affiliation(s)
- Xiao-Ning Chai
- Rudolf Boehm Institute for Pharmacology and Toxicology, Leipzig University, 04107 Leipzig, Germany; (X.-N.C.); (A.M.); (Y.G.); (M.S.)
| | - Friedrich-Alexander Ludwig
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 04318 Leipzig, Germany;
| | - Anne Müglitz
- Rudolf Boehm Institute for Pharmacology and Toxicology, Leipzig University, 04107 Leipzig, Germany; (X.-N.C.); (A.M.); (Y.G.); (M.S.)
| | - Yuanyuan Gong
- Rudolf Boehm Institute for Pharmacology and Toxicology, Leipzig University, 04107 Leipzig, Germany; (X.-N.C.); (A.M.); (Y.G.); (M.S.)
| | - Michael Schaefer
- Rudolf Boehm Institute for Pharmacology and Toxicology, Leipzig University, 04107 Leipzig, Germany; (X.-N.C.); (A.M.); (Y.G.); (M.S.)
| | - Ralf Regenthal
- Clinical Pharmacology, Rudolf Boehm Institute for Pharmacology and Toxicology, Leipzig University, 04107 Leipzig, Germany;
| | - Ute Krügel
- Rudolf Boehm Institute for Pharmacology and Toxicology, Leipzig University, 04107 Leipzig, Germany; (X.-N.C.); (A.M.); (Y.G.); (M.S.)
- Correspondence:
| |
Collapse
|
37
|
Consideration of Fractional Distribution Parameter f d in the Chen and Gross Method for Tissue-to-Plasma Partition Coefficients: Comparison of Several Methods. Pharm Res 2022; 39:463-479. [PMID: 35288804 PMCID: PMC9014445 DOI: 10.1007/s11095-022-03211-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/17/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE The tissue-to-plasma partition coefficient (Kp) describes the extent of tissue distribution in physiologically-based pharmacokinetic (PBPK) models. Constant-rate infusion studies are common for experimental determination of the steady-state Kp,ss, while the tissue-plasma concentration ratio (CT/Cp) in the terminal phase after intravenous doses is often utilized. The Chen and Gross (C&G) method converts a terminal slope CT/Cp to Kp,ss based on assumptions of perfusion-limited distribution in tissue-plasma equilibration. However, considering blood flow (QT) and apparent tissue permeability (fupPSin) in the rate of tissue distribution, this report extends the C&G method by utilizing a fractional distribution parameter (fd). METHODS Relevant PBPK equations for non-eliminating and eliminating organs along with lung and liver were derived for the conversion of CT/Cp values to Kp,ss. The relationships were demonstrated in rats with measured CT/Cp and Kp,ss values and the model-dependent fd for 8 compounds with a range of permeability coefficients. Several methods of assessing Kp were compared. RESULTS Utilizing fd in an extended C&G method, our estimations of Kp,ss from CT/Cp were improved, particularly for lower permeability compounds. However, four in silico methods for estimating Kp performed poorly across tissues in comparison with measured Kp values. Mathematical relationships between Kp and Kp,ss that are generally applicable for eliminating organs with tissue permeability limitations necessitates inclusion of an extraction ratio (ER) and fd. CONCLUSION Since many different types/sources of Kp are present in the literature and used in PBPK models, these perspectives and equations should provide better insights in measuring and interpreting Kp values in PBPK.
Collapse
|
38
|
Towards the Elucidation of the Pharmacokinetics of Voriconazole: A Quantitative Characterization of Its Metabolism. Pharmaceutics 2022; 14:pharmaceutics14030477. [PMID: 35335853 PMCID: PMC8948939 DOI: 10.3390/pharmaceutics14030477] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 01/19/2022] [Accepted: 01/25/2022] [Indexed: 12/28/2022] Open
Abstract
The small-molecule drug voriconazole (VRC) shows a complex and not yet fully understood metabolism. Consequently, its in vivo pharmacokinetics are challenging to predict, leading to therapy failures or adverse events. Thus, a quantitative in vitro characterization of the metabolism and inhibition properties of VRC for human CYP enzymes was aimed for. The Michaelis-Menten kinetics of voriconazole N-oxide (NO) formation, the major circulating metabolite, by CYP2C19, CYP2C9 and CYP3A4, was determined in incubations of human recombinant CYP enzymes and liver and intestine microsomes. The contribution of the individual enzymes to NO formation was 63.1% CYP2C19, 13.4% CYP2C9 and 29.5% CYP3A4 as determined by specific CYP inhibition in microsomes and intersystem extrapolation factors. The type of inhibition and inhibitory potential of VRC, NO and hydroxyvoriconazole (OH-VRC), emerging to be formed independently of CYP enzymes, were evaluated by their effects on CYP marker reactions. Time-independent inhibition by VRC, NO and OH-VRC was observed on all three enzymes with NO being the weakest and VRC and OH-VRC being comparably strong inhibitors of CYP2C9 and CYP3A4. CYP2C19 was significantly inhibited by VRC only. Overall, the quantitative in vitro evaluations of the metabolism contributed to the elucidation of the pharmacokinetics of VRC and provided a basis for physiologically-based pharmacokinetic modeling and thus VRC treatment optimization.
Collapse
|
39
|
Storelli F, Li CY, Sachar M, Kumar V, Heyward S, Sáfár Z, Kis E, Unadkat JD. Prediction of Hepatobiliary Clearances and Hepatic Concentrations of Transported Drugs in Humans Using Rosuvastatin as a Model Drug. Clin Pharmacol Ther 2022; 112:593-604. [DOI: 10.1002/cpt.2556] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/31/2022] [Indexed: 11/08/2022]
Affiliation(s)
- Flavia Storelli
- Department of Pharmaceutics University of Washington Seattle WA USA
| | - Cindy Yanfei Li
- Department of Pharmaceutics University of Washington Seattle WA USA
| | - Madhav Sachar
- Department of Pharmaceutics University of Washington Seattle WA USA
| | - Vineet Kumar
- Department of Pharmaceutics University of Washington Seattle WA USA
| | | | | | | | | |
Collapse
|
40
|
Rowland M, Roberts MS, Pang KS. In Defense of Current Concepts and Applications of Clearance in Drug Development and Therapeutics. Drug Metab Dispos 2022; 50:187-190. [PMID: 34740891 DOI: 10.1124/dmd.121.000637] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/22/2021] [Indexed: 11/22/2022] Open
Abstract
Clearance is one of the most widely quoted and applied pharmacokinetic concepts in drug development and therapy. Its foundations and associated models of drug elimination are well embedded and accepted within the scientific community. Recently, however, the prevailing views that have held us in good stead for the past almost 50 years have been challenged with the argument that organ clearance should not be based on elimination rate, now defined by extraction across the liver divided by incoming or systemic concentration, as in current practice, but rather, by the mean concentration of drug within the blood in the organ, which is model-dependent. We argue that all needed parameters already exist, and that the proposed new approach to organ clearance is confusing and unnecessary. SIGNIFICANCE STATEMENT: Clearance concepts are widely applied in drug development and therapy. Historically, hepatic clearance has been defined as the ratio of rate of elimination divided by ingoing blood concentration. Recently, this approach has been challenged arguing that clearance should be referenced to blood concentration within the liver extrapolation (IVIVE). There is no need for additional, a feature that corresponds to intrinsic clearance of the chosen clearance model, a widely accepted parameter in physiologically based pharmacokinetic (PBPK) and in vitro to in vivo extrapolation (IVIVE). There is no need for additional, confusing clearance terms, which offer no material benefit.
Collapse
Affiliation(s)
- Malcolm Rowland
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (M.R.); Therapeutics Research Centre, The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Australia (M.S.R.); UniSA Clinical and Health Sciences, Basil Hetzel Institute for Translational Health Research, University of South Australia, Adelaide, Australia (M.S.R.); and Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Canada (K.S.P.)
| | - Michael S Roberts
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (M.R.); Therapeutics Research Centre, The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Australia (M.S.R.); UniSA Clinical and Health Sciences, Basil Hetzel Institute for Translational Health Research, University of South Australia, Adelaide, Australia (M.S.R.); and Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Canada (K.S.P.)
| | - K Sandy Pang
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (M.R.); Therapeutics Research Centre, The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Australia (M.S.R.); UniSA Clinical and Health Sciences, Basil Hetzel Institute for Translational Health Research, University of South Australia, Adelaide, Australia (M.S.R.); and Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Canada (K.S.P.)
| |
Collapse
|
41
|
Dong J, Park MS. A myth of the well-stirred model: is the well-stirred model good for high clearance drugs? Eur J Pharm Sci 2022; 172:106134. [DOI: 10.1016/j.ejps.2022.106134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 12/31/2021] [Accepted: 01/21/2022] [Indexed: 11/27/2022]
|
42
|
Jusko WJ, Li X. Assessment of the Kochak-Benet Equation for Hepatic Clearance for the Parallel-Tube Model: Relevance of Classic Clearance Concepts in PK and PBPK. AAPS J 2021; 24:5. [PMID: 34853928 PMCID: PMC9639621 DOI: 10.1208/s12248-021-00656-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 10/11/2021] [Indexed: 12/14/2022] Open
Abstract
This report reviews concepts related to operation of the classic parallel-tube model (PTM) for hepatic disposition and examines two recent proposals of a newly derived equation to describe hepatic clearance (CLH). It is demonstrated that the proposed equation is identical to a re-arrangement of an earlier relationship from Pang and Rowland and provides a means of calculation of intrinsic clearance (CLint,PTM) rather than CLH as posed. We further demonstrate how classic hepatic clearance models with an assumed CLint, while subject to numerous limitations, remain highly useful and necessary in both traditional pharmacokinetics (PK) and physiologically based pharmacokinetic (PBPK) modeling.
Collapse
Affiliation(s)
- William J. Jusko
- Department of Pharmaceutical Sciences, State University of New York at Buffalo, 404 Pharmacy Building, Buffalo, New York 14214, USA,To whom correspondence should be addressed. ()
| | - Xiaonan Li
- Department of Pharmaceutical Sciences, State University of New York at Buffalo, 404 Pharmacy Building, Buffalo, New York 14214, USA
| |
Collapse
|
43
|
Abstract
The use of artificial intelligence methods in drug safety began in the early 2000s with applications such as predicting bacterial mutagenicity and hERG inhibition. The field has been endlessly expanding ever since and the models have become more complex. These approaches are now integrated into molecule risk assessment processes along with in vitro and in vivo methods. Today, artificial intelligence can be used in every phase of drug discovery and development, from profiling chemical libraries in early discovery, to predicting off-target effects in the mid-discovery phase, to assessing potential mutagenic impurities in development and degradants as part of life cycle management. This chapter provides an overview of artificial intelligence in drug safety and describes its application throughout the entire discovery and development process.
Collapse
|
44
|
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
|
45
|
Novak JJ, Burchett W, Di L. Effects of low temperature on blood‐to‐plasma ratio measurement. Biopharm Drug Dispos 2021; 42:234-241. [DOI: 10.1002/bdd.2265] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 01/12/2021] [Accepted: 01/24/2021] [Indexed: 11/06/2022]
Affiliation(s)
- Jonathan J. Novak
- Pharmacokinetics, Dynamics and Metabolism Pfizer Worldwide Research and Development Groton Connecticut USA
| | - Woodrow Burchett
- Early Clinical Development Pfizer Worldwide Research and Development Groton Connecticut USA
| | - Li Di
- Pharmacokinetics, Dynamics and Metabolism Pfizer Worldwide Research and Development Groton Connecticut USA
| |
Collapse
|
46
|
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
|
47
|
Yim DS, Choi S, Bae SH. Predicting human pharmacokinetics from preclinical data: absorption. Transl Clin Pharmacol 2020; 28:126-135. [PMID: 33062626 PMCID: PMC7533162 DOI: 10.12793/tcp.2020.28.e14] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/15/2020] [Accepted: 09/16/2020] [Indexed: 11/19/2022] Open
Abstract
Predicting the rate and extent of oral absorption of drugs in humans has been a challenging task for new drug researchers. This tutorial reviews in vitro and PBPK methods reported in the past decades that are widely applied to predicting oral absorption in humans. The physicochemical property and permeability (typically obtained using Caco-2 system) data is the first necessity to predict the extent of absorption from the gut lumen to the intestinal epithelium (Fa). Intrinsic clearance measured using the human microsome or hepatocytes is also needed to predict the gut (Fg) and hepatic (Fh) bioavailability. However, there are many issues with the correction of the inter-laboratory variability, hepatic cell membrane permeability, CYP3A4 dependency, etc. The bioavailability is finally calculated as F = Fa × Fg × Fh. Although the rate of absorption differs by micro-environments and locations in the intestine, it may be simply represented by ka. The ka, the first-order absorption rate constant, is predicted from in vitro and in vivo data. However, human PK-predicting software based on these PBPK theories should be carefully used because there are many assumptions and variances. They include differences in laboratory methods, inter-laboratory variances, and theories behind the methods. Thus, the user's knowledge and experiences in PBPK and in vitro methods are necessary for proper human PK prediction.
Collapse
Affiliation(s)
- Dong-Seok Yim
- Department of Clinical Pharmacology and Therapeutics, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.,PIPET (Pharmacometrics Institute for Practical Education and Training), College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Suein Choi
- Department of Clinical Pharmacology and Therapeutics, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.,PIPET (Pharmacometrics Institute for Practical Education and Training), College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | | |
Collapse
|
48
|
The Segregated Intestinal Flow Model (SFM) for Drug Absorption and Drug Metabolism: Implications on Intestinal and Liver Metabolism and Drug-Drug Interactions. Pharmaceutics 2020; 12:pharmaceutics12040312. [PMID: 32244748 PMCID: PMC7238003 DOI: 10.3390/pharmaceutics12040312] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 03/26/2020] [Accepted: 03/27/2020] [Indexed: 12/13/2022] Open
Abstract
The properties of the segregated flow model (SFM), which considers split intestinal flow patterns perfusing an active enterocyte region that houses enzymes and transporters (<20% of the total intestinal blood flow) and an inactive serosal region (>80%), were compared to those of the traditional model (TM), wherein 100% of the flow perfuses the non-segregated intestine tissue. The appropriateness of the SFM model is important in terms of drug absorption and intestinal and liver drug metabolism. Model behaviors were examined with respect to intestinally (M1) versus hepatically (M2) formed metabolites and the availabilities in the intestine (FI) and liver (FH) and the route of drug administration. The %contribution of the intestine to total first-pass metabolism bears a reciprocal relation to that for the liver, since the intestine, a gateway tissue, regulates the flow of substrate to the liver. The SFM predicts the highest and lowest M1 formed with oral (po) and intravenous (iv) dosing, respectively, whereas the extent of M1 formation is similar for the drug administered po or iv according to the TM, and these values sit intermediate those of the SFM. The SFM is significant, as this drug metabolism model explains route-dependent intestinal metabolism, describing a higher extent of intestinal metabolism with po versus the much reduced or absence of intestinal metabolism with iv dosing. A similar pattern exists for drug–drug interactions (DDIs). The inhibitor or inducer exerts its greatest effect on victim drugs when both inhibitor/inducer and drug are given po. With po dosing, more drug or inhibitor/inducer is brought into the intestine for DDIs. The bypass of flow and drug to the enterocyte region of the intestine after intravenous administration adds complications to in vitro–in vivo extrapolations (IVIVE).
Collapse
|