1
|
O'Mahony ET, Arian CM, Aryeh KS, Wang K, Thummel KE, Kelly EJ. Human intestinal enteroids: Nonclinical applications for predicting oral drug disposition, toxicity, and efficacy. Pharmacol Ther 2025:108879. [PMID: 40398537 DOI: 10.1016/j.pharmthera.2025.108879] [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: 11/12/2024] [Revised: 02/19/2025] [Accepted: 05/15/2025] [Indexed: 05/23/2025]
Abstract
The application of human enteroid systems presents a significant opportunity within the drug development pipeline, highlighting considerable potential for advancements in the characterization and evaluation of new molecular entities. Derived from LGR5+ crypt-based columnar cells, enteroid systems more accurately recapitulate the microanatomy and physiological processes of the human intestinal mucosa compared to traditionally used systems. They contain the complement of major mucosal epithelial cell types, maintain the genetic identity of the donor and intestinal segment they were derived from, and exhibit biological functions and specific activities that are seen in vivo. In this review, we examine the applications of human enteroid systems in nonclinical drug development and compare findings to existing and emerging in vitro models of the small intestine. Specifically, we explore enteroid systems in the context of predicting oral drug disposition, focusing on apparent permeability, intestinal first-pass metabolism, and drug interactions, as well as their utility in assessing drug-induced gastrointestinal toxicity and screening therapeutic efficacy against enteric diseases. Additionally, we highlight aspects of enteroid systems that warrant further study.
Collapse
Affiliation(s)
- Eimear T O'Mahony
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, United States of America
| | - Christopher M Arian
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, United States of America
| | - Kayenat S Aryeh
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, United States of America
| | - Kai Wang
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, United States of America
| | - Kenneth E Thummel
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, United States of America; Center of Excellence for Natural Product Drug Interaction Research, Spokane, WA, United States of America
| | - Edward J Kelly
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, United States of America; Kidney Research Institute, University of Washington, Seattle, WA, United States of America.
| |
Collapse
|
2
|
Rodriguez-Fernandez K, Gómez-Mantilla JD, Shukla S, Stopfer P, Sieger P, Mangas-Sanjuán V, Peters SA. Evaluation of Solubility-Limited Absorption as a Surrogate to Predicting Positive Food Effect of BCS II/IV Drugs. Clin Pharmacokinet 2025; 64:373-385. [PMID: 39899201 PMCID: PMC11954708 DOI: 10.1007/s40262-025-01473-9] [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: 12/31/2024] [Indexed: 02/04/2025]
Abstract
INTRODUCTION AND OBJECTIVE Physiologically based pharmacokinetic (PBPK) models are increasingly used to predict food effect (FE) but model parameterization is challenged by in vitro-in vivo (IVIV) disconnect and/or parameter nonidentifiability. To overcome these issues, we propose a simplified PBPK model, in which all solubility-driven processes are lumped into a single parameter, solubility, which is optimized against observed concentration-time data. METHODS A set of commercially available biopharmaceutical classification system (BCS) II/IV compounds was selected to measure the solubility in a fasted state simulated intestinal fluid (FaSSIF) medium. The compounds were ranked from the lowest to the highest dose-adjusted FaSSIF solubility (FaSSIF/D) value and subdivided into three areas based on an upper and a lower limit: drugs with FaSSIF/D > upper limit having no FE, drugs with FaSSIF/D < lower limit having FE, and drugs between the limits said to be in the sensitivity range (SR), for which we tested the hypothesis that solubility-limited absorption (SLA) identified by simplified PBPK model can reliably predict positive FE if their exposures are not impacted by gut efflux or gut metabolism. RESULTS We demonstrate, using a subset of drugs within SR for which PBPK models were available, that drugs with SLA exhibited a positive FE, while those with no SLA did not show FE. CONCLUSIONS This proposal allows for a reliable binary prediction of FE to enable timely decisions on the need for pilot FE studies as well as the timing of pivotal FE studies.
Collapse
Affiliation(s)
- Karine Rodriguez-Fernandez
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Valencia, Spain
- Interuniversity Research Institute for Molecular Recognition and Technological Development, Polytechnic University of Valencia-University of Valencia, Valencia, Spain
| | - José David Gómez-Mantilla
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Binger Straße 173, 55216, Ingelheim am Rhein, Germany
| | - Suneet Shukla
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Binger Straße 173, 55216, Ingelheim am Rhein, Germany
| | - Peter Stopfer
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Binger Straße 173, 55216, Ingelheim am Rhein, Germany
| | - Peter Sieger
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397, Biberach a.d. Riss, Germany
| | - Victor Mangas-Sanjuán
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Valencia, Spain
- Interuniversity Research Institute for Molecular Recognition and Technological Development, Polytechnic University of Valencia-University of Valencia, Valencia, Spain
| | - Sheila Annie Peters
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Binger Straße 173, 55216, Ingelheim am Rhein, Germany.
| |
Collapse
|
3
|
Fan X, Hou K, Liu G, Shi R, Wang W, Liang G. Strategies to overcome the limitations of current organoid technology - engineered organoids. J Tissue Eng 2025; 16:20417314251319475. [PMID: 40290859 PMCID: PMC12033597 DOI: 10.1177/20417314251319475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 01/26/2025] [Indexed: 04/30/2025] Open
Abstract
Organoids, as 3D in vitro models derived from stem cells, have unparalleled advantages over traditional cell and animal models for studying organogenesis, disease mechanisms, drug screening, and personalized diagnosis and treatment. Despite the tremendous progress made in organoid technology, the translational application of organoids still presents enormous challenges due to the complex structure and function of human organs. In this review, the limitations of the translational application of traditional organoid technologies are first described. Next, we explore ways to address many of the limitations of traditional organoid cultures by engineering various dimensions of organoid systems. Finally, we discuss future directions in the field, including potential roles in drug screening, simulated microphysiology system and personalized diagnosis and treatment. We hope that this review inspires future research into organoids and microphysiology system.
Collapse
Affiliation(s)
- Xulong Fan
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, China
| | - Kun Hou
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, China
- Institute of Organoids on Chips Translational Research, Henan Academy of Sciences, Zhengzhou, China
| | - Gaojian Liu
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, China
| | - Ruolin Shi
- Institute of Organoids on Chips Translational Research, Henan Academy of Sciences, Zhengzhou, China
| | - Wenjie Wang
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, China
| | - Gaofeng Liang
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, China
- Institute of Organoids on Chips Translational Research, Henan Academy of Sciences, Zhengzhou, China
| |
Collapse
|
4
|
Tóth G, Golubova A, Falk A, Lind SB, Nicholas M, Lanekoff I. Interleukin-13 Treatment of Living Lung Tissue Model Alters the Metabolome and Proteome-A Nano-DESI MS Metabolomics and Shotgun Proteomics Study. Int J Mol Sci 2024; 25:5034. [PMID: 38732251 PMCID: PMC11084154 DOI: 10.3390/ijms25095034] [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: 03/06/2024] [Revised: 04/04/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024] Open
Abstract
Asthma is a chronic respiratory disease with one of the largest numbers of cases in the world; thus, constant investigation and technical development are needed to unravel the underlying biochemical mechanisms. In this study, we aimed to develop a nano-DESI MS method for the in vivo characterization of the cellular metabolome. Using air-liquid interface (ALI) cell layers, we studied the role of Interleukin-13 (IL-13) on differentiated lung epithelial cells acting as a lung tissue model. We demonstrate the feasibility of nano-DESI MS for the in vivo monitoring of basal-apical molecular transport, and the subsequent endogenous metabolic response, for the first time. Conserving the integrity of the ALI lung-cell layer enabled us to perform temporally resolved metabolomic characterization followed by "bottom-up" proteomics on the same population of cells. Metabolic remodeling was observed upon histamine and corticosteroid treatment of the IL-13-exposed lung cell monolayers, in correlation with alterations in the proteomic profile. This proof of principle study demonstrates the utility of in vivo nano-DESI MS for characterizing ALI tissue layers, and the new markers identified in our study provide a good starting point for future, larger-scale studies.
Collapse
Affiliation(s)
- Gábor Tóth
- Department of Chemistry—BMC, Uppsala University, 75237 Uppsala, Sweden
| | | | - Alexander Falk
- Department of Chemistry—BMC, Uppsala University, 75237 Uppsala, Sweden
| | | | | | - Ingela Lanekoff
- Department of Chemistry—BMC, Uppsala University, 75237 Uppsala, Sweden
| |
Collapse
|
5
|
Scheidecker B, Poulain S, Sugimoto M, Arakawa H, Kim SH, Kawanishi T, Kato Y, Danoy M, Nishikawa M, Sakai Y. Mechanobiological stimulation in organ-on-a-chip systems reduces hepatic drug metabolic capacity in favor of regenerative specialization. Biotechnol Bioeng 2024; 121:1435-1452. [PMID: 38184801 DOI: 10.1002/bit.28653] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 01/08/2024]
Abstract
Hepatic physiology depends on the liver's complex structural composition which among others, provides high oxygen supply rates, locally differential oxygen tension, endothelial paracrine signaling, as well as residual hemodynamic shear stress to resident hepatocytes. While functional improvements were shown by implementing these factors into hepatic culture systems, direct cause-effect relationships are often not well characterized-obfuscating their individual contribution in more complex microphysiological systems. By comparing increasingly complex hepatic in vitro culture systems that gradually implement these parameters, we investigate the influence of the cellular microenvironment to overall hepatic functionality in pharmacological applications. Here, hepatocytes were modulated in terms of oxygen tension and supplementation, endothelial coculture, and exposure to fluid shear stress delineated from oxygen influx. Results from transcriptomic and metabolomic evaluation indicate that particularly oxygen supply rates are critical to enhance cellular functionality-with cellular drug metabolism remaining comparable to physiological conditions after prolonged static culture. Endothelial signaling was found to be a major contributor to differential phenotype formation known as metabolic zonation, indicated by WNT pathway activity. Lastly, oxygen-delineated shear stress was identified to direct cellular fate towards increased hepatic plasticity and regenerative phenotypes at the cost of drug metabolic functionality - in line with regenerative effects observed in vivo. With these results, we provide a systematic evaluation of critical parameters and their impact in hepatic systems. Given their adherence to physiological effects in vivo, this highlights the importance of their implementation in biomimetic devices, such as organ-on-a-chip systems. Considering recent advances in basic liver biology, direct translation of physiological structures into in vitro models is a promising strategy to expand the capabilities of pharmacological models.
Collapse
Affiliation(s)
| | - Stéphane Poulain
- Institute of Industrial Science, University of Tokyo, Tokyo, Japan
| | - Masahiro Sugimoto
- Institute for Advanced Biosciences, Keio University, Yamagata, Japan
- Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Hiroshi Arakawa
- Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Soo H Kim
- Institute of Industrial Science, University of Tokyo, Tokyo, Japan
| | - Takumi Kawanishi
- Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Yukio Kato
- Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Mathieu Danoy
- Department of Chemical System Engineering, University of Tokyo, Tokyo, Japan
| | - Masaki Nishikawa
- Department of Chemical System Engineering, University of Tokyo, Tokyo, Japan
| | - Yasuyuki Sakai
- Department of Chemical System Engineering, University of Tokyo, Tokyo, Japan
| |
Collapse
|
6
|
Yasugi Y, Shirasaka Y, Tamai I. Quantitative analysis of the impact of membrane permeability on intestinal first-pass metabolism of CYP3A substrates. Biopharm Drug Dispos 2024; 45:3-14. [PMID: 38085672 DOI: 10.1002/bdd.2379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/30/2023] [Accepted: 11/08/2023] [Indexed: 02/15/2024]
Abstract
The aim of this study was firstly to investigate the effect of membrane permeability on the intestinal availability (Fg ) of 10 cytochrome P450 3A4 substrates with differing permeability (Papp ) and metabolic activity (CLint ) using Madin-Darby canine kidney II (MDCKII) cells expressing human CYP3A4 (MDCKII/CYP3A4 cells), and secondly to confirm the essential factors by simulations. A membrane permeation assay using MDCKII/CYP3A4 cells showed a significant correlation between human intestinal extraction ratio (ER) (Eg (=1 - Fg )) and in vitro cellular ER (r = 0.834). This relationship afforded better predictability of Eg values than the relationship between Eg and CLint,HIM values obtained from human intestinal microsomes (r = 0.598). An even stronger correlation was observed between 1 - Fa ·Fg and ER (r = 0.874). Simulation with a cellular kinetic model indicated that ER is sensitive to changes of PSpassive and CLint values, but not to the intracellular unbound fraction (fu,cell ) or P-gp-mediated efflux (PSP - gp ). It may be concluded that, based on the concentration-time profile of drugs in epithelial cells, transmembrane permeability influences Fg (or ER) and drug exposure time to metabolizing enzymes for P450 substrate.
Collapse
Affiliation(s)
- Yugo Yasugi
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Yoshiyuki Shirasaka
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Ikumi Tamai
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
7
|
Di L. Quantitative Translation of Substrate Intrinsic Clearance from Recombinant CYP1A1 to Humans. AAPS J 2023; 25:98. [PMID: 37798423 DOI: 10.1208/s12248-023-00863-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 09/15/2023] [Indexed: 10/07/2023] Open
Abstract
CYP1A1 is a cytochrome P450 family 1 enzyme that is mostly expressed in the extrahepatic tissues. To understand the CYP1A1 contribution to drug clearance in humans, we examined the in vitro-in vivo extrapolation (IVIVE) of intrinsic clearance (CLint) for a set of drugs that are in vitro CYP1A1 substrates. Despite being strong in vitro CYP1A1 substrates, 82% of drugs gave good IVIVE with predicted CLint within 2-3-fold of the observed values using human liver microsomes and hepatocytes, suggesting they were not in vivo CYP1A1 substrates due to the lack of extrahepatic contribution to CLint. Only three drugs (riluzole, melatonin and ramelteon) that are CYP1A2 substrates yielded significant underprediction of in vivo CLint up to 11-fold. The fold of CLint underprediction was linearly proportional to human recombinant CYP1A1 (rCYP1A1) CLint, indicating they were likely to be in vivo CYP1A1 substrates. Using these three substrates, a calibration curve can be developed to enable direct translation from in vitro rCYP1A1 CLint to in vivo extrahepatic contributions in humans. In vivo CYP1A1 substrates are planar and small, which is consistent with the structure of the active site. This is in contrast to the in vitro substrates, which include large and nonplanar molecules, suggesting rCYP1A1 is more accessible than what is in vivo. The impact of CYP1A1 on first-pass intestinal metabolism was also evaluated and shown to be minimal. This is the first study providing new insights on in vivo translation of CYP1A1 contributions to human clearance using in vitro rCYP1A1 data.
Collapse
Affiliation(s)
- Li Di
- Pharmacokinetic, Dynamics and Drug Metabolism, Pfizer Worldwide Research and Development, Groton, Connecticut, 06543, USA.
| |
Collapse
|
8
|
Maeda K. Quantitative Prediction of Intestinal Absorption of Drugs from In Vitro Study: Utilization of Differentiated Intestinal Epithelial Cells Derived from Intestinal Stem Cells at Crypts. Drug Metab Dispos 2023; 51:1136-1144. [PMID: 37142427 DOI: 10.1124/dmd.122.000966] [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: 05/30/2022] [Revised: 03/21/2023] [Accepted: 04/28/2023] [Indexed: 05/06/2023] Open
Abstract
Prediction of intestinal absorption of drugs in humans is one of the critical elements in the development process for oral drugs. However, it remains challenging, because intestinal absorption of drugs is influenced by multiple factors, including the function of various metabolic enzymes and transporters, and large species differences in drug bioavailability hinder the prediction of human bioavailability directly from in vivo animal experiments. For the screening of intestinal absorption properties of drugs, a transcellular transport assay with Caco-2 cells is still routinely used by pharmaceutical companies because of its convenience, but the predictability of the fraction of the oral dose that goes to the portal vein of metabolic enzyme/transporter substrate drugs was not always good because the cellular expression of metabolic enzymes and transporters is different from that in the human intestine. Recently, various novel in vitro experimental systems have been proposed such as the use of human-derived intestinal samples, transcellular transport assay with induced pluripotent stem-derived enterocyte-like cells, or differentiated intestinal epithelial cells derived from intestinal stem cells at crypts. Crypt-derived differentiated epithelial cells have an excellent potential to characterize species differences and regional differences in intestinal absorption of drugs because a unified protocol can be used for the proliferation of intestinal stem cells and their differentiation into intestinal absorptive epithelial cells regardless of the animal species and the gene expression pattern of differentiated cells is maintained at the site of original crypts. The advantages and disadvantages of novel in vitro experimental systems for characterizing intestinal absorption of drugs are also discussed. SIGNIFICANCE STATEMENT: Among novel in vitro tools for the prediction of human intestinal absorption of drugs, crypt-derived differentiated epithelial cells have many advantages. Cultured intestinal stem cells are rapidly proliferated and easily differentiated into intestinal absorptive epithelial cells simply by changing the culture media. A unified protocol can be used for the establishment of intestinal stem cell culture from preclinical species and humans. Region-specific gene expression at the collection site of crypts can be reproduced in differentiated cells.
Collapse
Affiliation(s)
- Kazuya Maeda
- Laboratory of Pharmaceutics, School of Pharmacy, Kitasato University, Tokyo, Japan
| |
Collapse
|
9
|
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
|
10
|
Mazzinelli E, Favuzzi I, Arcovito A, Castagnola R, Fratocchi G, Mordente A, Nocca G. Oral Mucosa Models to Evaluate Drug Permeability. Pharmaceutics 2023; 15:pharmaceutics15051559. [PMID: 37242801 DOI: 10.3390/pharmaceutics15051559] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/02/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
Due to its numerous advantages, such as excellent drug accessibility, rapid absorption, and bypass of first-pass metabolism, the route of drug administration that involves crossing the oral mucosa is highly favored. As a result, there is significant interest in investigating the permeability of drugs through this region. The purpose of this review is to describe the various ex vivo and in vitro models used to study the permeability of conveyed and non-conveyed drugs through the oral mucosa, with a focus on the most effective models. Currently, there is a growing need for standardized models of this mucosa that can be used for developing new drug delivery systems. Oral Mucosa Equivalents (OMEs) may provide a promising future perspective as they are capable of overcoming limitations present in many existing models.
Collapse
Affiliation(s)
- Elena Mazzinelli
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Roma, Italy
| | - Ilaria Favuzzi
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Roma, Italy
| | - Alessandro Arcovito
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Roma, Italy
- Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Largo Agostino Gemelli 8, 00168 Roma, Italy
| | - Raffaella Castagnola
- UOC Odontoiatria Generale e Ortodonzia, Dipartimento Scienze dell'Invecchiamento, Neurologiche, Ortopediche e della Testa Collo, Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy
- Dipartimento di Testa-Collo e Organi di Senso, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Giorgia Fratocchi
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Roma, Italy
| | - Alvaro Mordente
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Roma, Italy
- Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Largo Agostino Gemelli 8, 00168 Roma, Italy
| | - Giuseppina Nocca
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Roma, Italy
- Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Largo Agostino Gemelli 8, 00168 Roma, Italy
| |
Collapse
|
11
|
Ekiciler A, Chen WLK, Bo Y, Pugliano A, Donzelli M, Parrott N, Umehara K. Quantitative Cytochrome P450 3A4 Induction Risk Assessment Using Human Hepatocytes Complemented with Pregnane X Receptor-Activating Profiles. Drug Metab Dispos 2023; 51:276-284. [PMID: 36460477 DOI: 10.1124/dmd.122.001132] [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: 09/23/2022] [Revised: 11/14/2022] [Accepted: 11/23/2022] [Indexed: 12/05/2022] Open
Abstract
Reliable in vitro to in vivo translation of cytochrome P450 (CYP) 3A4 induction potential is essential to support risk mitigation for compounds during pharmaceutical discovery and development. In this study, a linear correlation of CYP3A4 mRNA induction potential in human hepatocytes with the respective pregnane-X receptor (PXR) activation in a reporter gene assay using DPX2 cells was successfully demonstrated for 13 clinically used drugs. Based on this correlation, using rifampicin as a positive control, the magnitude of CYP3A4 mRNA induction for 71 internal compounds at several concentrations up to 10 µM (n = 90) was predicted within 2-fold error for 64% of cases with only a few false positives (19%). Furthermore, the in vivo area under the curve reduction of probe CYP substrates was reasonably predicted for eight marketed drugs (carbamazepine, dexamethasone, enzalutamide, nevirapine, phenobarbital, phenytoin, rifampicin, and rufinamide) using the static net effect model using both the PXR activation and CYP3A4 mRNA induction data. The liver exit concentrations were used for the model in place of the inlet concentrations to avoid false positive predictions and the concentration achieving twofold induction (F2) was used to compensate for the lack of full induction kinetics due to cytotoxicity and solubility limitations in vitro. These findings can complement the currently available induction risk mitigation strategy and potentially influence the drug interaction modeling work conducted at clinical stages. SIGNIFICANCE STATEMENT: The established correlation of CYP3A4 mRNA in human hepatocytes to PXR activation provides a clear cut-off to identify a compound showing an in vitro induction risk, complementing current regulatory guidance. Also, the demonstrated in vitro-in vivo translation of induction data strongly supports a clinical development program although limitations remain for drug candidates showing complex disposition pathways, such as involvement of auto-inhibition/induction, active transport and high protein binding.
Collapse
Affiliation(s)
- Aynur Ekiciler
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland (A.E., A.P., M.D., N.P., K.U.) and Roche Pharmaceutical Research and Early Development, China Innovation Center of Roche, Shanghai, China (W.L.K.C., Y.B.)
| | - Wen Li Kelly Chen
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland (A.E., A.P., M.D., N.P., K.U.) and Roche Pharmaceutical Research and Early Development, China Innovation Center of Roche, Shanghai, China (W.L.K.C., Y.B.)
| | - Yan Bo
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland (A.E., A.P., M.D., N.P., K.U.) and Roche Pharmaceutical Research and Early Development, China Innovation Center of Roche, Shanghai, China (W.L.K.C., Y.B.)
| | - Alessandra Pugliano
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland (A.E., A.P., M.D., N.P., K.U.) and Roche Pharmaceutical Research and Early Development, China Innovation Center of Roche, Shanghai, China (W.L.K.C., Y.B.)
| | - Massimiliano Donzelli
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland (A.E., A.P., M.D., N.P., K.U.) and Roche Pharmaceutical Research and Early Development, China Innovation Center of Roche, Shanghai, China (W.L.K.C., Y.B.)
| | - Neil Parrott
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland (A.E., A.P., M.D., N.P., K.U.) and Roche Pharmaceutical Research and Early Development, China Innovation Center of Roche, Shanghai, China (W.L.K.C., Y.B.)
| | - Kenichi Umehara
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland (A.E., A.P., M.D., N.P., K.U.) and Roche Pharmaceutical Research and Early Development, China Innovation Center of Roche, Shanghai, China (W.L.K.C., Y.B.)
| |
Collapse
|
12
|
Chen J, Yuan Z, Tu Y, Hu W, Xie C, Ye L. Experimental and computational models to investigate intestinal drug permeability and metabolism. Xenobiotica 2023; 53:25-45. [PMID: 36779684 DOI: 10.1080/00498254.2023.2180454] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
Oral administration is the preferred route for drug administration that leads to better therapy compliance. The intestine plays a key role in the absorption and metabolism of oral drugs, therefore, new intestinal models are being continuously proposed, which contribute to the study of intestinal physiology, drug screening, drug side effects, and drug-drug interactions.Advances in pharmaceutical processes have produced more drug formulations, causing challenges for intestinal models. To adapt to the rapid evolution of pharmaceuticals, more intestinal models have been created. However, because of the complexity of the intestine, few models can take all aspects of the intestine into account, and some functions must be sacrificed to investigate other areas. Therefore, investigators need to choose appropriate models according to the experimental stage and other requirements to obtain the desired results.To help researchers achieve this goal, this review summarised the advantages and disadvantages of current commonly used intestinal models and discusses possible future directions, providing a better understanding of intestinal models.
Collapse
Affiliation(s)
- Jinyuan Chen
- Institute of Scientific Research, Southern Medical University, Guangzhou, P.R. China.,TCM-Integrated Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Ziyun Yuan
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, P.R. China
| | - Yifan Tu
- Boehringer-Ingelheim, Connecticut, P.R. USA
| | - Wanyu Hu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, P.R. China
| | - Cong Xie
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Ling Ye
- TCM-Integrated Hospital, Southern Medical University, Guangzhou, P.R. China
| |
Collapse
|
13
|
Vu NAT, Song YM, Tran QT, Yun HY, Kim SK, Chae JW, Kim JK. Beyond the Michaelis-Menten: Accurate Prediction of Drug Interactions through Cytochrome P450 3A4 Induction. Clin Pharmacol Ther 2022; 113:1048-1057. [PMID: 36519932 DOI: 10.1002/cpt.2824] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022]
Abstract
The US Food and Drug Administration (FDA) guidance has recommended several model-based predictions to determine potential drug-drug interactions (DDIs) mediated by cytochrome P450 (CYP) induction. In particular, the ratio of substrate area under the plasma concentration-time curve (AUCR) under and not under the effect of inducers is predicted by the Michaelis-Menten (MM) model, where the MM constant ( K m $$ {K}_{\mathrm{m}} $$ ) of a drug is implicitly assumed to be sufficiently higher than the concentration of CYP enzymes that metabolize the drug ( E T $$ {E}_{\mathrm{T}} $$ ) in both the liver and small intestine. Furthermore, the fraction absorbed from gut lumen ( F a $$ {F}_{\mathrm{a}} $$ ) is also assumed to be one because F a $$ {F}_{\mathrm{a}} $$ is usually unknown. Here, we found that such assumptions lead to serious errors in predictions of AUCR. To resolve this, we propose a new framework to predict AUCR. Specifically, F a $$ {F}_{\mathrm{a}} $$ was re-estimated from experimental permeability values rather than assuming it to be one. Importantly, we used the total quasi-steady-state approximation to derive a new equation, which is valid regardless of the relationship between K m $$ {K}_{\mathrm{m}} $$ and E T $$ {E}_{\mathrm{T}} $$ , unlike the MM model. Thus, our framework becomes much more accurate than the original FDA equation, especially for drugs with high affinities, such as midazolam or strong inducers, such as rifampicin, so that the ratio between K m $$ {K}_{\mathrm{m}} $$ and E T $$ {E}_{\mathrm{T}} $$ becomes low (i.e., the MM model is invalid). Our work greatly improves the prediction of clinical DDIs, which is critical to preventing drug toxicity and failure.
Collapse
Affiliation(s)
- Ngoc-Anh Thi Vu
- College of Pharmacy, Chungnam National University, Daejeon, Korea
| | - Yun Min Song
- Department of Mathematical Sciences, KAIST, Daejeon, Korea.,Biomedical Mathematics Group, Institute for Basic Science, Daejeon, Korea
| | - Quyen Thi Tran
- College of Pharmacy, Chungnam National University, Daejeon, Korea
| | - Hwi-Yeol Yun
- College of Pharmacy, Chungnam National University, Daejeon, Korea.,Department of Bio-AI convergence, Chungnam National University, Daejeon, Korea
| | - Sang Kyum Kim
- College of Pharmacy, Chungnam National University, Daejeon, Korea
| | - Jung-Woo Chae
- College of Pharmacy, Chungnam National University, Daejeon, Korea.,Department of Bio-AI convergence, Chungnam National University, Daejeon, Korea
| | - Jae Kyoung Kim
- Department of Mathematical Sciences, KAIST, Daejeon, Korea.,Biomedical Mathematics Group, Institute for Basic Science, Daejeon, Korea
| |
Collapse
|
14
|
Durcik M, Gramec Skledar D, Tomašič T, Trontelj J, Peterlin Mašič L. Last piece in the puzzle of bisphenols BPA, BPS and BPF metabolism: Kinetics of the in vitro sulfation reaction. CHEMOSPHERE 2022; 303:135133. [PMID: 35636595 DOI: 10.1016/j.chemosphere.2022.135133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/10/2022] [Accepted: 05/24/2022] [Indexed: 06/15/2023]
Abstract
Bisphenols are endocrine-disrupting chemicals ubiquitously present in the environment, with the consequent exposure to humans. In humans, bisphenols are metabolized to glucuronide and sulfate conjugates. Recent studies indicate that sulfation represents an important bisphenol metabolic pathway for the most vulnerable humans, such as the growing fetus. Our aim was to evaluate sulfation kinetics of commonly detected bisphenols in biological samples: bisphenol A (BPA), bisphenol S (BPS), and bisphenol F (BPF). Furthermore, we evaluated estrogenic agonist potencies and long-term stability of these bisphenol sulfates. BPS and BPF sulfates were prepared by chemical synthesis. Sulfation kinetics of the selected bisphenols were tested in pooled human liver cytosol, as a source for soluble phase II enzymes, including liver sulfotransferases, with quantification by LC-MS/MS. A validated transactivation assay using the hERα-Hela 9903 cell line was used to determine estrogenic agonist potencies. Moreover, BPA, BPS, and BPF sulfate stabilities were examined under various conditions and during storage. In vitro sulfation of BPA and BPS followed Michaelis-Menten kinetics; BPF sulfation followed a substrate inhibition model. Sulfation rates were comparable for these bisphenols, although their KM values indicated some large differences in affinities. BPA and BPS sulfates are not hERα agonists. The bisphenol sulfates can be considered stable for at least 2 days under these tested media conditions. These data indicate that bisphenol sulfation is an important route in their biotransformation. Compared to glucuronidation, these bisphenols show slower sulfation rates but similar KM values. BPA and BPS metabolic biotransformation by sulfation provides their detoxification as they are without estrogenic activities.
Collapse
Affiliation(s)
- Martina Durcik
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | | | - Tihomir Tomašič
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Jurij Trontelj
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia.
| | | |
Collapse
|
15
|
Atypical kinetics of cytochrome P450 enzymes in pharmacology and toxicology. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 95:131-176. [PMID: 35953154 DOI: 10.1016/bs.apha.2022.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Atypical kinetics are observed in metabolic reactions catalyzed by cytochrome P450 enzymes (P450). Yet, this phenomenon is regarded as experimental artifacts in some instances despite increasing evidence challenging the assumptions of typical Michaelis-Menten kinetics. As P450 play a major role in the metabolism of a wide range of substrates including drugs and endogenous compounds, it becomes critical to consider the impact of atypical kinetics on the accuracy of estimated kinetic and inhibitory parameters which could affect extrapolation of pharmacological and toxicological implications. The first half of this book chapter will focus on atypical non-Michaelis-Menten kinetics (e.g. substrate inhibition, biphasic and sigmoidal kinetics) as well as proposed underlying mechanisms supported by recent insights in mechanistic enzymology. In particular, substrate inhibition kinetics in P450 as well as concurrent drug inhibition of P450 in the presence of substrate inhibition will be further discussed. Moreover, mounting evidence has revealed that despite the high degree of sequence homology between CYP3A isoforms (i.e. CYP3A4 and CYP3A5), they have the propensities to exhibit vastly different susceptibilities and potencies of mechanism-based inactivation (MBI) with a common drug inhibitor. These experimental observations pertaining to the presence of these atypical isoform- and probe substrate-specific complexities in CYP3A isoforms by several clinically-relevant drugs will therefore be expounded and elaborated upon in the second half of this book chapter.
Collapse
|
16
|
Kumar AR, Patilea-Vrana GI, Anoshchenko O, Unadkat JD. Characterizing and Quantifying Extrahepatic Metabolism of (-)-Δ 9-Tetrahydrocannabinol (THC) and Its Psychoactive Metabolite, (±)-11-Hydroxy-Δ 9-THC (11-OH-THC). Drug Metab Dispos 2022; 50:734-740. [PMID: 35370140 PMCID: PMC9199115 DOI: 10.1124/dmd.122.000868] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/31/2022] [Indexed: 11/12/2023] Open
Abstract
(-)-Δ9-Tetrahydrocannabinol (THC) is the psychoactive constituent of cannabis, a drug recreationally consumed orally or by inhalation. Physiologically based pharmacokinetic (PBPK) modeling can be used to predict systemic and tissue exposure to THC and its psychoactive metabolite, (±)-11-hydroxy-Δ9-THC (11-OH-THC). To populate a THC/11-OH-THC PBPK model, we previously characterized the depletion clearance of THC (by CYP2C9) and 11-OH-THC (by UDP-glucuronosyltransferase (UGT), CYP3A, and CYP2C9) in adult human liver microsomes. Here we focused on quantifying extrahepatic depletion clearance of THC/11-OH-THC, important after oral (intestine) and inhalational (lung) consumption of THC as well as prenatal THC use (placenta and fetal liver). THC (500 nM) was metabolized in adult human intestinal microsomes (n = 3-5) by CYP2C9 [Vmax: 1.1 ± 0.38 nmol/min/mg; Michaelis-Menten constant (Km): 70 nM; intrinsic clearance (CLint): 15 ± 5.4 ml/min/mg; fraction metabolized (fm): 0.89 ± 0.31 at concentration ≪ 70 nM] and CYP3A (CLint: 2.0 ± 0.86 ml/min/mg; fm: 0.11 ± 0.050). 11-OH-THC (50 nM) was metabolized by CYP3A (CLint: 0.26 ± 0.058 ml/min/mg; fm: 0.51 ± 0.11) and UGT2B7 (CLint: 0.13 ± 0.027 ml/min/mg; fm: 0.25 ± 0.053). THC at 500 nM (CLint: 4.7 ± 0.22 ml/min/mg) and 11-OH-THC at 50 nM (CLint: 2.4 ± 0.13 ml/min/mg) were predominately (fm: 0.99 and 0.80, respectively) metabolized by CYP3A in human fetal liver microsomes (n = 3). However, we did not observe significant depletion of THC/11-OH-THC in adult lung, first trimester, second trimester, or term placentae microsomes. Using PBPK modeling and simulation, these data could be used in the future to predict systemic and tissue THC/11-OH-THC exposure in healthy and special populations. SIGNIFICANCE STATEMENT: This is the first characterization and quantification of (-)-Δ9-tetrahydrocannabinol (THC) and (±)-11-hydroxy-Δ9-THC (11-OH-THC) depletion clearance by cytochrome P450 and UDP-glucuronosyltransferase enzymes in extrahepatic human tissues: intestine, fetal liver, lung, and placenta. These data can be used to predict, through physiologically based pharmacokinetic modeling and simulation, systemic and tissue THC/11-OH-THC exposure after inhalational and oral THC use in both healthy and special populations (e.g., pregnant women).
Collapse
Affiliation(s)
- Aditya R Kumar
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| | | | - Olena Anoshchenko
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| | - Jashvant D Unadkat
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| |
Collapse
|
17
|
Arian CM, Imaoka T, Yang J, Kelly EJ, Thummel KE. Gutsy science: In vitro systems of the human intestine to model oral drug disposition. Pharmacol Ther 2022; 230:107962. [PMID: 34478775 PMCID: PMC8821120 DOI: 10.1016/j.pharmthera.2021.107962] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 07/07/2021] [Accepted: 07/13/2021] [Indexed: 02/03/2023]
Abstract
The intestine has important gate-keeping functions that can profoundly affect the systemic blood exposure of orally administered drugs. Thus, characterizing a new molecular entity's (NME) disposition within the intestine is of utmost importance in drug development. While currently used in vitro systems, such as Ussing chamber, precision-cut intestinal slices, immortalized cell lines, and primary enterocytes provide substantial knowledge about drug absorption and the intestinal first-pass effect, they remain sub-optimal for quantitatively predicting this process and the oral bioavailability of many drugs. Use of novel in vitro systems such as intestinal organoids and intestinal microphysiological systems have provided substantial advances over the past decade, expanding our understanding of intestinal physiology, pathology, and development. However, application of these emerging in vitro systems in the pharmaceutical science is in its infancy. Preliminary work has demonstrated that these systems more accurately recapitulate the physiology and biochemistry of the intact intestine, as it relates to oral drug disposition, and thus they hold considerable promise as preclinical testing platforms of the future. Here we review currently used and emerging in vitro models of the human intestine employed in pharmaceutical science research. We also highlight aspects of these emerging tools that require further study.
Collapse
Affiliation(s)
- Christopher M Arian
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Tomoki Imaoka
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Jade Yang
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Edward J Kelly
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Kenneth E Thummel
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
18
|
Järvinen E, Deng F, Kiander W, Sinokki A, Kidron H, Sjöstedt N. The Role of Uptake and Efflux Transporters in the Disposition of Glucuronide and Sulfate Conjugates. Front Pharmacol 2022; 12:802539. [PMID: 35095509 PMCID: PMC8793843 DOI: 10.3389/fphar.2021.802539] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022] Open
Abstract
Glucuronidation and sulfation are the most typical phase II metabolic reactions of drugs. The resulting glucuronide and sulfate conjugates are generally considered inactive and safe. They may, however, be the most prominent drug-related material in the circulation and excreta of humans. The glucuronide and sulfate metabolites of drugs typically have limited cell membrane permeability and subsequently, their distribution and excretion from the human body requires transport proteins. Uptake transporters, such as organic anion transporters (OATs and OATPs), mediate the uptake of conjugates into the liver and kidney, while efflux transporters, such as multidrug resistance proteins (MRPs) and breast cancer resistance protein (BCRP), mediate expulsion of conjugates into bile, urine and the intestinal lumen. Understanding the active transport of conjugated drug metabolites is important for predicting the fate of a drug in the body and its safety and efficacy. The aim of this review is to compile the understanding of transporter-mediated disposition of phase II conjugates. We review the literature on hepatic, intestinal and renal uptake transporters participating in the transport of glucuronide and sulfate metabolites of drugs, other xenobiotics and endobiotics. In addition, we provide an update on the involvement of efflux transporters in the disposition of glucuronide and sulfate metabolites. Finally, we discuss the interplay between uptake and efflux transport in the intestine, liver and kidneys as well as the role of transporters in glucuronide and sulfate conjugate toxicity, drug interactions, pharmacogenetics and species differences.
Collapse
Affiliation(s)
- Erkka Järvinen
- Clinical Pharmacology, Pharmacy, and Environmental Medicine, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Feng Deng
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Wilma Kiander
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Alli Sinokki
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Heidi Kidron
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Noora Sjöstedt
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| |
Collapse
|
19
|
Wang H, Brown PC, Chow EC, Ewart L, Ferguson SS, Fitzpatrick S, Freedman BS, Guo GL, Hedrich W, Heyward S, Hickman J, Isoherranen N, Li AP, Liu Q, Mumenthaler SM, Polli J, Proctor WR, Ribeiro A, Wang J, Wange RL, Huang S. 3D cell culture models: Drug pharmacokinetics, safety assessment, and regulatory consideration. Clin Transl Sci 2021; 14:1659-1680. [PMID: 33982436 PMCID: PMC8504835 DOI: 10.1111/cts.13066] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/12/2022] Open
Abstract
Nonclinical testing has served as a foundation for evaluating potential risks and effectiveness of investigational new drugs in humans. However, the current two-dimensional (2D) in vitro cell culture systems cannot accurately depict and simulate the rich environment and complex processes observed in vivo, whereas animal studies present significant drawbacks with inherited species-specific differences and low throughput for increased demands. To improve the nonclinical prediction of drug safety and efficacy, researchers continue to develop novel models to evaluate and promote the use of improved cell- and organ-based assays for more accurate representation of human susceptibility to drug response. Among others, the three-dimensional (3D) cell culture models present physiologically relevant cellular microenvironment and offer great promise for assessing drug disposition and pharmacokinetics (PKs) that influence drug safety and efficacy from an early stage of drug development. Currently, there are numerous different types of 3D culture systems, from simple spheroids to more complicated organoids and organs-on-chips, and from single-cell type static 3D models to cell co-culture 3D models equipped with microfluidic flow control as well as hybrid 3D systems that combine 2D culture with biomedical microelectromechanical systems. This article reviews the current application and challenges of 3D culture systems in drug PKs, safety, and efficacy assessment, and provides a focused discussion and regulatory perspectives on the liver-, intestine-, kidney-, and neuron-based 3D cellular models.
Collapse
Affiliation(s)
- Hongbing Wang
- Department of Pharmaceutical SciencesUniversity of Maryland School of PharmacyBaltimoreMarylandUSA
| | - Paul C. Brown
- Center for Drug Evaluation and ResearchUS Food and Drug Administration (FDA)Silver SpringMarylandUSA
| | - Edwin C.Y. Chow
- Office of Clinical PharmacologyOffice of Translational SciencesCenter for Drug Evaluation and ResearchUS Food and Drug Administration (FDA)Silver SpringMarylandUSA
| | | | - Stephen S. Ferguson
- Division of the National Toxicology ProgramNational Institute of Environmental Health SciencesResearch Triangle ParkNorth CarolinaUSA
| | - Suzanne Fitzpatrick
- Office of the Center DirectorCenter for Food Safety and Applied NutritionUS Food and Drug Administration (FDA)Silver SpringMarylandUSA
| | - Benjamin S. Freedman
- Division of NephrologyDepartment of PathologyKidney Research Institute, and Institute for Stem Cell and Regenerative MedicineUniversity of WashingtonSeattleWashingtonUSA
- Department of MedicineUniversity of WashingtonSeattleWashingtonUSA
| | - Grace L. Guo
- Department of Pharmacology and ToxicologyErnest Mario School of PharmacyRutgers UniversityPiscatawayNew JerseyUSA
| | - William Hedrich
- Pharmaceutical Candidate Optimization, Metabolism and PharmacokineticsBristol‐Myers Squibb CompanyPrincetonNew JerseyUSA
| | | | - James Hickman
- NanoScience Technology CenterUniversity of Central FloridaOrlandoFloridaUSA
| | - Nina Isoherranen
- Department of PharmaceuticsSchool of PharmacyUniversity of WashingtonSeattleWashingtonUSA
| | - Albert P. Li
- In Vitro ADMET LaboratoriesColumbiaMarylandUSA
- In Vitro ADMET LaboratoriesMaldenMassachusettsUSA
| | - Qi Liu
- Office of Clinical PharmacologyOffice of Translational SciencesCenter for Drug Evaluation and ResearchUS Food and Drug Administration (FDA)Silver SpringMarylandUSA
| | - Shannon M. Mumenthaler
- Lawrence J. Ellison Institute for Transformative MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - James Polli
- Department of Pharmaceutical SciencesUniversity of Maryland School of PharmacyBaltimoreMarylandUSA
| | - William R. Proctor
- Predictive Toxicology, Safety AssessmentGenentech, IncSouth San FranciscoCaliforniaUSA
| | - Alexandre Ribeiro
- Office of Clinical PharmacologyOffice of Translational SciencesCenter for Drug Evaluation and ResearchUS Food and Drug Administration (FDA)Silver SpringMarylandUSA
| | - Jian‐Ying Wang
- Department of SurgeryCell Biology GroupUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Ronald L. Wange
- Center for Drug Evaluation and ResearchUS Food and Drug Administration (FDA)Silver SpringMarylandUSA
| | - Shiew‐Mei Huang
- Office of Clinical PharmacologyOffice of Translational SciencesCenter for Drug Evaluation and ResearchUS Food and Drug Administration (FDA)Silver SpringMarylandUSA
| |
Collapse
|
20
|
Drug Disposition in the Lower Gastrointestinal Tract: Targeting and Monitoring. Pharmaceutics 2021; 13:pharmaceutics13020161. [PMID: 33530468 PMCID: PMC7912393 DOI: 10.3390/pharmaceutics13020161] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 12/20/2022] Open
Abstract
The increasing prevalence of colonic diseases calls for a better understanding of the various colonic drug absorption barriers of colon-targeted formulations, and for reliable in vitro tools that accurately predict local drug disposition. In vivo relevant incubation conditions have been shown to better capture the composition of the limited colonic fluid and have resulted in relevant degradation and dissolution kinetics of drugs and formulations. Furthermore, drug hurdles such as efflux transporters and metabolising enzymes, and the presence of mucus and microbiome are slowly integrated into drug stability- and permeation assays. Traditionally, the well characterized Caco-2 cell line and the Ussing chamber technique are used to assess the absorption characteristics of small drug molecules. Recently, various stem cell-derived intestinal systems have emerged, closely mimicking epithelial physiology. Models that can assess microbiome-mediated drug metabolism or enable coculturing of gut microbiome with epithelial cells are also increasingly explored. Here we provide a comprehensive overview of the colonic physiology in relation to drug absorption, and review colon-targeting formulation strategies and in vitro tools to characterize colonic drug disposition.
Collapse
|